Desk Encyclopedia of Microbiology [PDF] | Online Book Share (2024)

1,169 Pages • 619,477 Words • PDF • 29.2 MB

+ ENCYCLOPEDIA+ Microbiology+ Desk

Uploaded at 2021-09-23 13:31

Report DMCA

PREVIEW PDF

Dem-fm.qxd 10/9/03 6:40 PM Page i

The Desk Encyclopedia of Microbiology

Dem-fm.qxd 10/9/03 6:40 PM Page ii

Dem-fm.qxd 10/9/03 6:40 PM Page iii

The Desk Encyclopedia of Microbiology Editor Moselio Schaechter Consulting Editor Joshua Lederberg

Amsterdam Boston Heidelberg London New York Oxford Paris San Diego San Francisco Singapore Sydney Tokyo

Dem-fm.qxd 10/9/03 6:40 PM Page iv

This book is printed on acid-free paper Copyright © 2004, Elsevier Ltd. All rights reserved No part of this publication may be reproduced, stored in a retrieval system, or transmitted in any form or by any means electronic, mechanical, photocopying, recording or otherwise, without the prior written permission of the publisher Permissions may be sought directly from Elsevier’s Science & Technology Rights Department in Oxford, UK: phone: (44) 1865 843830, fax: (44) 1865 853333, e-mail: [emailprotected]. You may also complete your request on-line via the Elsevier homepage (http://www.elsevier.com), by selecting ‘Customer Support’ and then ‘Obtaining Permissions’ Elsevier Academic Press 525 B Street, Suite 1900, San Diego, California 92101-4495, USA http://www.elsevier.com Elsevier Academic Press 84 Theobald’s Road, London WC1X 8RR, UK http://www.elsevier.com

British Library Cataloguing in Publication Data A catalogue record for this book is available from the British Library Library of Congress Catalog Number: 2002114100 ISBN 0-12-621361-5

Printed and bound in China 03 04 05 06 07 08 9 8 7 6 5 4 3 2 1

Dem-fm.qxd 10/9/03 6:40 PM Page v

Contents Contributors

1. 2. 3. 4. 5. 6. 7. 8. 9. 10. 11. 12. 13. 14.

x

Preface

xvii

General websites

xviii

Adhesion, Bacterial Matthew A. Mulvey and Scott J. Hultgren

1

Agrobacterium and plant cell transformation Peter J. Christie

10

Antibiotic resistance in bacteria Julian Davies and Vera Webb

25

Antifungal agents Ana Espinel-Ingroff

47

Antisense RNAs Andrea Denise Branch

68

Antiviral agents Richard J. Whitley

84

Archaea Paul Blum and Vidula Dikshit

108

Attenuation, Transcriptional Charles Yanofsky

117

Bacillus subtilis, Genetics Kevin M. Devine

126

Bacteriophages Hans-Wolfgang Ackermann

135

Biocides (Nonpublic health, Nonagricultural antimicrobials) Mohammad Sondossi

147

Biofilms and biofouling Karen T. Elvers and Hilary M. Lappin-Scott

161

Biological warfare James A. Poupard and Linda A. Miller

168

Bioluminescence, Microbial J. Woodland Hastings

180

v

Dem-fm.qxd 10/9/03 6:40 PM Page vi

vi 15. 16. 17. 18. 19. 20. 21. 22. 23. 24. 25. 26. 27. 28. 29. 30. 31. 32. 33. 34. 35.

contents Bioreactors Larry E. Erickson

189

Bioremediation Joseph B. Hughes, C. Nelson Neale, and C. Herb Ward

196

Biosensors Yoko Nomura and Isao Karube

216

Cell membrane: structure and function Robert J. Kadner

222

Cell Walls, Bacterial Joachim-Volker Höltje

239

Chemotaxis Jeff Stock and Sandra Da Re

251

Chromosome, Bacterial Karl Drlica and Arnold J. Bendich

259

Conjugation, Bacterial Laura S. Frost

271

Crystalline bacterial cell surface layers (S layers) Uwe B. Sleytr and Paul Messner

286

Culture collections and their databases Mary K. B. Berlyn

294

Developmental processes in bacteria Yves V. Brun

314

Diversity, Microbial Charles R. Lovell

326

DNA repair Lawrence Grossman

340

DNA replication James A. Hejna and Robb E. Moses

350

DNA restriction and modification Noreen E. Murray

358

DNA sequencing and genomics Brian A. Dougherty

371

Ecology, Microbial Michael J. Klug and David A. Odelson

381

Emerging infections David L. Heymann

387

Energy transduction processes: from respiration to photosynthesis Stuart J. Ferguson

394

Enteropathogenic bacteria Farah K. Bahrani-Mougeot and Michael S. Donnenberg

403

Escherichia coli and Salmonella, Genetics K. Brooks Low

415

Dem-fm.qxd 10/9/03 6:40 PM Page vii

contents 36. 37. 38. 39. 40. 41. 42. 43. 44. 45. 46. 47. 48. 49. 50. 51. 52. 53. 54. 55. 56.

vii

Exotoxins Joseph T. Barbieri

427

Extremophiles Ricardo Cavicchioli and Torsten Thomas

436

Fimbriae, Pili Matthew A. Mulvey, Karen W. Dodson, Gabriel E. Soto, and Scott J. Hultgren

454

Flagella Shin-Ichi Aizawa

470

Food-borne illnesses David W. K. Acheson

480

Fungal infections, Cutaneous Peter G. Sohnle and David K. Wagner

499

Fungal infections, Systemic Arturo Casadevall

507

Gastrointestinal microbiology T. G. Nagaraja

514

Genetically modified organisms: guidelines and regulations for research Sue Tolin and Anne Vidaver

526

Genomes, Mapping of Bacterial J. Guespin-Michel and F. Joset

536

Germ-free animal techniques Bernard S. Wostmann

547

Gram-negative anaerobic pathogens Arthur O. Tzianabos, Laurie E. Comstock, and Dennis L. Kasper

554

Gram-negative cocci, Pathogenic Emil C. Gotschlich

562

Heat stress Christophe Herman and Carol A. Gross

574

Horizontal transfer of genes between microorganisms Jack A. Heinemann

582

Human immunodeficiency virus Luc Montagnier

591

Identification of bacteria, Computerized Trevor N. Bryant

602

Industrial fermentation processes Thomas M. Anderson

613

Insect’s symbiotic microorganisms A. E. Douglas

626

Iron metabolism Charles F. Earhart

637

Lipopolysaccharides Chris Whitfield

645

Dem-fm.qxd 10/9/03 6:40 PM Page viii

viii 57. 58. 59. 60. 61. 62. 63. 64. 65. 66. 67. 68. 69. 70. 71. 72. 73. 74. 75. 76. 77.

contents Methanogenesis Kevin R. Sowers

659

Methylotrophy J. Colin Murrell and Ian R. McDonald

680

Nitrogen cycle Roger Knowles

690

Nitrogen fixation L. David Kuykendall, Fawzy M. Hashem, Robert B. Dadson, and Gerald H. Elkan

702

Nodule formation in legumes Peter H. Graham

715

Nutrition of microorganisms Thomas Egli

725

Oral microbiology Ian R. Hamilton and George H. Bowden

739

Osmotic stress Douglas H. Bartlett and Mary F. Roberts

754

Outer membrane, Gram-negative bacteria Mary J. Osborn

767

Oxidative stress Pablo J. Pomposiello and Bruce Demple

775

pH Stress Joan L. Slonczewski

781

Plant pathogens George N. Agrios

789

Plasmids, Bacterial Christopher M. Thomas

808

Polymerase chain reaction (PCR) Carol J. Palmer and Christine Paszko-Kolva

824

Prions Christine Musahl and Adriano Aguzzi

829

Protein secretion Donald B. Oliver and Jorge Galan

842

Quorum sensing in gram-negative bacteria Clay Fuqua

859

Recombinant DNA, Basic procedures Judith W. Zyskind

870

Sexually transmitted diseases Jane A. Cecil and Thomas C. Quinn

879

Skin microbiology Morton N. Swartz

899

Soil microbiology Kate M. Scow

914

Dem-fm.qxd 10/9/03 6:40 PM Page ix

contents 78. 79. 80. 81. 82. 83. 84. 85. 86. 87. 88. 89. 90. 91. 92. 93.

Index

ix

SOS response Kevin W. Winterling

927

Space flight, effects on microorganisms D. L. Pierson and S. K. Mishra

934

Sporulation Patrick J. Piggot

942

Starvation, Bacterial A. C. Matin

951

Strain improvement Sarad Parekh

960

Sulfur cycle Piet Lens, Marcus Vallero, and Look Hulshoff Pol

974

Transcriptional regulation in prokaryotes Orna Amster-Choder

984

Transduction: host DNA transfer by bacteriophages Millicent Masters

1000

Transformation, Genetic Brian M. Wilkins and Peter A. Meacock

1012

Transposable elements Peter M. Bennett

1025

Two-component systems Alexander J. Ninfa and Mariette R. Atkinson

1042

Vaccines, Bacterial Susan K. Hoiseth

1053

Vaccines, Viral Ann M. Arvin

1063

Viruses Sondra Schlesinger and Milton J. Schlesinger

1071

Viruses, Emerging Stephen S. Morse

1084

Yeasts Graeme M. Walker

1102 1115

Dem-fm.qxd 10/9/03 6:40 PM Page x

Contributors David W. K. Acheson Center for Food Safety and Applied Nutrition, Food and Drug Administration, Rockville, MD 20740, USA

Mariette R. Atkinson Department of Biological Chemistry, University of Michigan Medical School, 4310 Med Sci I, 1301 Catherine, Ann Arbor, MI 48109-0606, USA

H.-W. Ackermann Department of Medical Biology, Laval University, Local 2332, Pav. Ferdinand Vandry, Laval, Quebec, Canada G1K 7P4

Farah K. Bahrani-Mougeot Department of Medicine, University of Maryland School of Medicine, MSTF 90, 656 W Baltimore St., Baltimore, MD 21201-1559, USA

George N. Agrios Department of Plant Pathology, University of Florida, 1453 Fifield Hall, P.O. Box 110680, Gainesville, FL 32611, USA

Joseph T. Barbieri Department of Microbiology, Medical College of Wisconsin, P.O. Box 26509, 8701 Watertown Plank Rd., Milwaukee, WI 53226-0509, USA

Adriano Aguzzi Institute of Neuropathology, University of Zurich, University Hospital of Zürich, Schmelzbergstrasse 12, Zurich CH-8091, Switzerland

Douglas H. Bartlett Center for Marine Biotechnology and Biomedicine, University of California, San Diego, Scripps Institution of Oceanography, 9500 Gilman Drive, Dept 0202, La Jolla, CA 92093-0202, USA

Shin-Ichi Aizawa Soft Nano-Machine Project, CREST, Japan Science and Technology Agency, 1064-18 Takahori, Hirata, Takanezawa, Shioya-gun, Tochigi 329-1206, Japan

Arnold J. Bendich Professor of Botany and Genetics, Department of Biology, 522 Hitchcock Hall, University of Washington, Box 351800, Seattle, WA 98195, USA

Orna Amster-Choder Department of Molecular Biology, Hebrew University School of Medicine, P.O. Box 12272, Bldg. 3, 2nd Floor, Room 34, Jerusalem 91120, Israel

Peter M. Bennett Department of Pathology and Microbiology, University of Bristol, School of Medical Sciences, University Walk, Bristol BS8 1TD, UK

Thomas M. Anderson Microbiology Manager, Archer Daniels Midland BioProducts, P.O. Box 1470, Decatur, IL 62525, USA

Mary K.B. Berlyn Department of Biology, Yale University, 355-OML, 165 Prospect St., New Haven, CT 06520-8104, USA

Ann M. Arvin Department of Pediatrics, Stanford University School of Medicine, Mailcode 5208, 300 Pasteur Drive, G-312A Stanford, CA 94305, USA

Paul Blum George Beadle Center for Genetics, Nebraska University-Lincoln, P.O. Box 880666, Lincoln, NE 68588-066, USA

x

Dem-fm.qxd 10/9/03 6:40 PM Page xi

contributors Andrea D. Branch Division of Liver Diseases, Department of Medicine, Mount Sinai Medical Center, Recanati/Miller Transplantation Institute, One Gustave L. Levy Place, Box 1633, New York, NY 10029-6574, USA Yves V. Brun Department of Biology, Indiana University, Jordan Hall, Bloomington, IN 47405, USA Trevor N. Bryant Medical Statistics and Computing, University of Southampton, Southampton General Hospital, Tremona Rd, Southampton SO16 6YD, UK

xi

Bruce Demple Department of Cancer Cell Biology, Harvard School of Public Health, Bldg. 1 Floor 6, 665 Huntington Avenue, Boston, MA 02115-6021, USA Kevin M. Devine Department of Genetics, Trinity College, Dublin, Lincoln Place Gate, Dublin 2, UK Vidula Dixit George Beadle Center for Genetics, Nebraska University-Lincoln, P.O. Box 880666, Lincoln, NE 68588-066, USA

George H. Bowden Department of Oral Microbiology, University of Manitoba, Faculty of Dentistry, 780 Bannatyne Avenue, Winnipeg, Manitoba, Canada R3E 0W2

Karen W. Dodson Department of Molecular Microbiology, Washington University School of Medicine, Campus Box 8230, 660 S Euclid Avenue, 8230, St. Louis, MO 63110-1010, USA

Arturo Casadevall Department of Medicine, Infectious Diseases, Albert Einstein College of Medicine, Golding Bldg. Rm. 701, 1300 Morris Park Avenue, Bronx, NY 10461, USA

Michael S. Donnenberg Department of Medicine, University of Maryland School of Medicine, MSTF 90, 656 W Baltimore St., Baltimore, MD 21201-1559, USA

Ricardo Cavicchioli Department of Microbiology and Immunology, University of New South Wales, Sydney, NSW 2052, Australia

Brian A. Dougherty Department of Applied Genomics, Bristol-Myers Squibb Company, Pharmaceutical Research Institute, 5 Research Parkway, Wallingford, CT 06492-7660, USA

Jane A. Cecil The Johns Hopkins University, Ross Research Building 1159, 720 Rutland Avenue, Baltimore, MD 21205-2196, USA Peter J. Christie Department of Microbiology and Molecular Genetics, University of Texas Health Science Center, 6431 Fannin St., Houston, TX 77030-1501, USA Laurie E. Comstock Channing Laboratory, Harvard Medical School, 181 Longwood Avenue, Boston, MA 02115-5899, USA Sandra Da Re Department of Molecular Biology, Princeton University, 330 Lewis Thomas Lab, Princeton, NJ 08544, USA Robert B. Dadson University of Maryland, Eastern Shore, Princess Anne, Maryland, USA Julian Davies Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada

Angela E. Douglas Department of Biology, University of York, P.O. Box 373, York YO1 5YW, UK Karl Drlica Public Health Research Institute, 225 Warren Street, Newark, NJ 07103 Charles F. Earhart Department of Microbiology, University of Texas, ESB 226, A5000, 24th St. & Speedway, Austin, TX 78712-1095, USA Thomas Egli Department of Microbiology, Swiss Federal Institute for Environmental Science and Technology, BU-D04, Ueberlandstrasse 133, Dübendorf CH-8600, Switzerland Gerald H. Elkan Department of Microbiology, North Carolina State University, Raleigh, NC 27605, USA Karen T. Elvers Hatherly Labs, University of Exeter, Exeter, Prince of Wales Road, Devon EX4 4PS, UK

Dem-fm.qxd 10/9/03 6:40 PM Page xii

xii

contributors

Larry E. Erickson Department of Chemical Engineering, Kansas State University, 105 Durland Hall, Manhattan, KS 66506-5102, USA

Ian R. Hamilton Department of Oral Microbiology, University of Manitoba, Faculty of Dentistry, 780 Bannatyne Avenue, Winnipeg, Manitoba R3E 0W2, Canada

Ana A. Espinel-Ingroff Department of Medicine, Division of Infectious Diseases, Medical College of Virginia, Sanger Hall, Room 7049, 1101 E. Marshall Street, Richmond, VA 23298, USA

Fawzy Hashem University of Maryland, Eastern Shore, Princess Anne, Maryland, USA

Stuart J. Ferguson Department of Biochemistry, University of Oxford, South Parks Road, Oxford OX1 3QU, UK Laura S. Frost Department of Biological Sciences, University of Alberta, CW 405 Biological Sciences Bldg., Edmonton, Alberta T6G 2E9, Canada Clay Fuqua Department of Biology, 1001 E. 3rd Street, Jordan Hall 418, Indiana University Bloomington, IN 47405 Jorge Galan Department of Microbial Pathogenesis, Yale University, New Haven, CT 06520, USA Emil C. Gotschlich Laboratory of Bacterial Pathogenesis and Immunology, Rockefeller University, 1230 York Avenue, New York, NY 10021-6399, USA Peter H. Graham Department of Soil, Water and Climate, University of Minnesota, 256 Borlaug Hall, 1991 Upper Buford Circle, St. Paul, MN 55108, USA Carol A. Gross Department of Microbiology, University of California, San Francisco, Medical Sciences Rm 534, #0512, 513 Parnassus Avenue, San Francisco, CA 94143-0512, USA

J. Woodland Hastings Department of Biology, Harvard University, Biological Laboratories, 16 Divinity Avenue, Cambridge, MA 02138-2020, USA Jack A. Heinemann Norweigian Institute of Gene Ecology, Tromso, Norway James A. Hejna Department of Molecular and Medical Genetics, Oregon Health Sciences University, M/C L103, 3181 SW Sam Jackson Park Rd., Portland, OR 97201-3098, USA Christophe Herman Department of Microbiology, University of California, San Francisco, Medical Sciences Rm 534, #0512, 513 Parnassus Avenue, San Francisco, CA 94143-0512, USA David L. Heymann Executive Director, Communicable Diseases, World Health Organization, Geneva 27 CH-1211, Switzerland Susan K. Hoiseth Wyeth Vaccines Research, Pearl River, New York, USA Joachim-V. Höltje Max-Planck-Institut für Entwicklungsbiologie, Abteilung Biochemie, Spemannstrasse 35, Tübingen D-72076, Germany

Lawrence Grossman Department of Biochemistry, Johns Hopkins University School of Hygiene and Public Health, 615 N Wolfe Street, Baltimore, MD 21205-2179, USA

Joseph B. Hughes Energy and Environmental Systems Institute, Rice University, 6100 S. Main, MS-316, Houston, TX 77005, USA

Janine Guespin-Michel Laboratoire de Microbiologie du Froid, IFR CNRS – Université de Rouen, Faculté de Sciences et Techniques, Place Emile Blondel, Mont-Saint-Aignan 76821, France

Scott James Hultgren Department of Molecular Microbiology, Washington University School of Medicine, Campus Box 8230, 660 S Euclid Avenue, 8230, St. Louis, MO 63110-1010, USA

Dem-fm.qxd 10/9/03 6:40 PM Page xiii

contributors Francoise Joset Laboratoire de Chimie Bactérienne, CNRS 13412 Marseille, France Robert J. Kadner Department of Microbiology, University of Virginia School of Medicine, Box 441, Health Sciences Center, Charlottesville, VA 22908, USA Isao Karube School of Bionics, Tokyo University of Technology, 1404-1 Katakura-cho, Hachioji, Tokyo 192-0982, Japan Dennis L. Kasper Channing Laboratory, Harvard Medical School, 181 Longwood Avenue, Boston, MA 02115-5899, USA Michael J. Klug Department of Microbiology, Michigan State University, W.K. Kellogg Biological Station, 3700 East Gull Lake Drive, Hickory Corners, MI 49060, USA Roger Knowles Department of Natural Resource Sciences, McGill University, MacDonald Campus, 21,111 Lakeshore Road, Ste-Anne de-Bellevue, Quebec, Canada H9X 3V9 L. David Kuykendall Agricultural Research Service, Beltsville, US Department of Agriculture, Bldg. 011A, Barc West Rm. 252, Plant Molecular Pathology Laboratory, PSI, Beltsville, MD 20705, USA Hilary M. Lappin-Scott Hatherly Labs, University of Exeter, Exeter, Prince of Wales Road, Devon EX4 4PS, UK Piet Lens Department of Environmental Technology, Wageningen Agricultural University, P.O. Box 8129, Wageningen 6700 EV, The Netherlands Charles R. Lovell Department of Biological Sciences, University of South Carolina, Coker Life Sciences 408, Columbia, SC 29208, USA K. Brooks Low Department of Therapeutic Radiology, Yale University, Hunter Radiation Therapy, M353, 333 Cedar Street, New Haven, CT 06520, USA

xiii

Millicent Masters Institute of Cell & Molecular Biology, University of Edinburgh, Darwin Bldg. King’s Buildings, Mayfield Road, Edinburgh EH9 3JR, UK A. C. Matin Department of Microbiology and Immunology, Stanford University, Sherman Fairchild Science Bldg. D317, Stanford, CA 94305-5402, USA Peter A. Meacock Department of Genetics, University of Leicester, Adrian Building, University Road, Leicester LE1 7RH, UK Ian R. McDonald Department of Biological Sciences, University of Warwick, Coventry CV4 7AL, UK Paul Messner Center for Ultrastructure Research and Institute for Molecular Nanotechnology, University of Agricultural Sciences, Vienna, Gregor-Mendel-Str. 33, Vienna A-1180, Austria Linda A. Miller Department of Automicrobial Profiling/ Clinical Microbiology, SmithKline Beecham Pharmaceuticals, P.O. Box 5089, 1250 S. Collegeville Rd., Mail Code UP1340, Collegeville, PA 19426-0989, USA Saroj K. Mishra Division of Life Sciences, NASA/Johnson Space Center, Houston, 3600 Bay Area Blvd., Houston, TX 77058, USA Luc Montagnier Institut Pasteur, 25–28 rue du Dr Roux, 75015 Paris, France and Department of Biology, Queens College, NY 11367, USA Stephen S. Morse DARPA – Defense Advanced Research Project Agency, Columbia University, 3701 N Fairfax Drive, Room 838, Arlington, VA 22203-1714, USA Robb E. Moses Department of Molecular and Medical Genetics, Oregon Health Sciences University, M/C L103, 3181 SW Sam Jackson Park Rd., Portland, OR 97201-3098, USA Matthew A. Mulvey Department of Molecular Microbiology, Washington University School of Medicine, Campus Box 8230, 660 S Euclid Avenue, 8230, St. Louis, MO 63110-1010, USA

Dem-fm.qxd 10/9/03 6:40 PM Page xiv

xiv

contributors

Noreen E. Murray Institute of Cell and Molecular Biology, University of Edinburgh, King’s Buildings, Mayfield Road, Edinburgh, EH9 3JR, UK

D. L. Pierson Division of Life Sciences, NASA/Johnson Space Center, Houston, 3600 Bay Area Blvd., Houston, TX 77058, USA

J. Colin Murrell Department of Biological Sciences, University of Warwick, Coventry CV4 7AL, UK

Patrick J. Piggot Department of Microbiology and Immunology, Temple University School of Medicine, 3400 N Broad Street, Philadelphia, PA 19140-5104, USA

Christine Musahl Institute of Neuropathology, University of Zurich, University Hospital of Zürich, Schmelzbergstrasse 12, Zurich CH-8091, Switzerland C. Nelson Neale Energy and Environmental Systems Institute, Rice University, 6100 S. Main, MS-316, Houston, TX 77005, USA T. G. Nagaraja Department of Diagnostic Medicine/ Pathobiology, Kansas State University, College of Veterinary Medicine, Manhattan, KS 66506-5606, USA Alexander J. Ninfa Department of Biological Chemistry, University of Michigan Medical School, 4310 Med Sci I, 1301 Catherine, Ann Arbor, MI 48109-0606, USA Yoko Nomura School of Bionics, Tokyo University of Technology, 1404-1 Katakura-cho, Hachioji, Tokyo 192-0982, Japan David A. Odelson Invitrogen Corp. Carlsbad, CA, USA Donald B. Oliver Department of Molecular Biology and Biochemistry, Wesleyan University, Hall-Atwater and Shanklin Labs, Middletown, CT 06459-0175, USA Mary J. Osborn Department of Microbiology, University of Connecticut Health Center, Farmington, CT 06032, USA Carol J. Palmer Department of Pathobiology, University of Florida, USA Sarad R. Parekh Dow AgroSciences, 9330 Zionsville Road, Indianapolis, IN 46268-1054, USA Christine Paszko-Kolva Accelerated Technology Laboratories, Inc., Belmont, CA, 496 Holly Grove School Road, West End, NC 27376, USA

Look Hulshoff Pol Department of Environmental Technology, Wageningen Agricultural University, P.O. Box 8129, Wageningen 6700 EV, The Netherlands Pablo J. Pomposiello Department of Microbiology, University of Massachusetts, Amherst, MA 01003, USA James A. Poupard Department of Automicrobial Profiling/Clinical Microbiology, SmithKline Beecham Pharmaceuticals, P.O. Box 5089, 1250 S. Collegeville Rd., Mail Code UP1340, Collegeville, PA 19426-0989, USA Thomas C. Quinn The Johns Hopkins University, Ross Research Building 1159, 720 Rutland Avenue, Baltimore, MD 21205-2196, USA Mary F. Roberts Chemistry Department, 140 Commonwealth Avenue, Boston College, Chestnut Hill, MA 02467, USA Milton J. Schlesinger Department of Molecular Microbiology, Washington University School of Medicine, Box 8230, 4566 Scott Avenue, St. Louis, MO 63110-1093, USA Sondra Schlesinger Department of Molecular Microbiology, Washington University School of Medicine, Box 8230, 4566 Scott Avenue, St. Louis, MO 63110-1093, USA Kate M. Scow Department of Land, Air, Water Resources, University of California, Hoagland Hall, Davis, CA 95616-5224, USA Uwe B. Sleytr Center for Ultrastructure Research and Institute for Molecular Nanotechnology, University of Agricultural Sciences, Vienna, Gregor-Mendel-Str. 33, Vienna A-1180, Austria

Dem-fm.qxd 10/9/03 6:40 PM Page xv

contributors

xv

Joan L. Slonczewski Department of Biology, Kenyon College, 100 College Road, Gambier, OH 43022, USA

Marcus Vallero Department of Environmental Technology, Wageningen Agricultural University, P.O. Box 8129, Wageningen 6700 EV, The Netherlands

Peter G. Sohnle Department of Infectious Diseases, Medical College of Wisconsin, Research Service/151, VA Medical Center, Milwaukee, WI 53295, USA

Costantino Vetriani Department of Environmental Biology, Portland State University, Science Building 2, 1719 S.W. 10th Avenue, Portland, OR 97201, USA

Mohammad Sondossi Department of Microbiology, Weber State University, 2506 University Circle, Ogden, UT 84408-2506, USA Gabriel E. Soto Department of Molecular Microbiology, Washington University School of Medicine, Campus Box 8230, 660 S Euclid Avenue, 8230, St. Louis, MO 63110-1010, USA Kevin R. Sowers Center of Marine Biotechnology, University of Maryland Biotechnology Institute, Suite 236, Columbus Center, 701 E Pratt Street, Baltimore, MD 21202-4031, USA Jeff B. Stock Department of Molecular Biology, Princeton University, 330 Lewis Thomas Lab, Princeton, NJ 08544, USA Morton N. Swartz Infectious Disease Unit, Massachusetts General Hospital and Harvard Medical School, 70 Blossom Street, Boston, MA 02114-2696, USA Christopher M. Thomas School of Biological Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK Torsten Thomas Department of Microbiology and Immunology, University of New South Wales, Sydney, NSW 2052, Australia Sue Tolin Department of Plant Pathology, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA Arthur O. Tzianabos Channing Laboratory, Harvard Medical School, 181 Longwood Avenue, Boston, MA 02115-5899, USA

Anne Vidaver Department of Plant Pathology, University of Nebraska-Lincoln, Lincoln, NE 68588, USA David K. Wagner Department of Infectious Diseases, Medical College of Wisconsin, Research Service/151, VA Medical Center, Milwaukee, WI 53295, USA Graeme M. Walker School of Molecular and Life Sciences, University of Albertay Dundee, Kydd Building, Bell Street, Dundee, DD1 1HG, UK C. Herb Ward Energy and Environmental Systems Institute, Rice University, 6100 S. Main, MS-316, Houston, TX 77005, USA Vera Webb Lookfar Solutions Inc., P.O. Box 811, Tofino V0R2Z0, British Columbia, Canada Chris Whitfield Department of Microbiology, University of Guelph, 172 Chemistry-Microbiology, Guelph, Canada ON N1G 2W1 Richard J. Whitley Department of Pediatrics, University of Alabama School of Medicine, Ambulatory Care Center 616, 1600 7th Avenue S, Birmingham, AL 35294-0011, USA Brian M. Wilkins† Department of Genetics, University of Leicester, Adrian Building, University Road, Leicester LE1 7RH, UK †

Deceased.

Dem-fm.qxd 10/9/03 6:40 PM Page xvi

xvi

contributors

Kevin W. Winterling Department of Biology, Emory and Henry College, P.O. Box 75, One Garnand Drive, Emory, VA 24327, USA

Charles Yanofsky Department of Biological Science, Stanford University, Gilbert Bldg., 371 Serra Street, Stanford, CA 94305, USA

Bernard S. Wostmann Lobund Laboratory, University of Notre Dame, 16977 Adams Road, Granger, IN 46530, USA

Judith W. Zyskind San Diego State University, Elitra Pharmaceuticals, 3510 Dunhill St, Suite A, San Diego, CA 92121, USA

Dem-fm.qxd 10/9/03 6:40 PM Page xvii

Preface The field of Microbiology encompasses highly diverse life forms—bacteria, archaea, fungi, protists, and viruses. They have a profound influence on all life on Earth: they play an essential role in the cycles of matter in nature, affect all biological environments, interact in countless ways with other living beings, and play a crucial role in agriculture and industry. The literature associated with Microbiology, of necessity, tends to be specialized and focused. For that reason, it is difficult to find sources that provide a broad perspective on a wide range of microbiological topics. That is the aim of The Desk Encyclopedia of Microbiology. The concept behind this venture is to provide a single reference volume with appeal to microbiologists on all levels and fields, including those working in research, teaching, industry, and government. We believe that this book will be helpful, especially for accessing material in areas in which the reader is not a specialist. It is intended to facilitate preparing lectures,

grant applications and reports, and to satisfy curiosity regarding microbiological topics. The Desk Encyclopedia of Microbiology is principally a synthesis from the comprehensive and multivolumed Encyclopedia of Microbiology. Our intention is to provide affordable and ready access to a large variety of topics within one set of covers. To this end we have chosen subjects that, in our opinion, will be of greatest interest to the largest number of readers. Included are the most general chapters from The Encyclopedia of Microbiology, brought up to date and augmented with current references and related URLs. We have emphasized topics that are currently “hot” in the field of Microbiology, including additional chapters from other sources. The result is a volume where coverage is extensive but not overly long in specific details. We believe this will be a most appropriate reference for anyone with an interest in the intriguing field of Microbiology. Moselio Schaechter, 2003

xvii

Dem-fm.qxd 10/9/03 6:40 PM Page xviii

General websites American Society for Microbiology. An extensive list of links is in “Search Microbiology Sites” (members only) http://www.asm.org/ Society for General Microbiology links page http://www.socgenmicrobiol.org.uk/links.htm Links to microbiology courses at various universities http://www.geocities.com/CapeCanaveral/3504/courses. htm

Microbiology clinical cases http://www.medinfo.ufl.edu/year2/mmid/bms5300/case s/index.html List of bacterial names with standing in nomenclature (J. P. Euzéby) http://www.bacterio.cict.fr/index.html Access to online resources on Bacterial Infections and Mycoses. Karolinska Institutet http://www.mic.ki.se/Diseases/c1.html

xviii

ch01.qxd 10/9/03 6:42 PM Page 1

1 Adhesion, Bacterial Matthew A. Mulvey and Scott J. Hultgren Washington University School of Medicine

GLOSSARY

I. MECHANISMS OF BACTERIAL ADHESION

adhesin A molecule, typically a protein, that mediates bacterial attachment by interacting with specific receptors. extracellular matrix A complex network of proteins and polysaccharides secreted by eukaryotic cells. Functions as a structural element in tissues, in addition to modulating tissue development and physiology. invasin An adhesin that can mediate bacterial invasion into host eukaryotic cells. isoreceptors Eukaryotic cell membrane components which contain identical receptor determinants recognized by a bacterial adhesin. lectins Proteins that bind carbohydrate motifs.

Bacterial adhesion to living cells and to inanimate surfaces is governed by nonspecific electrostatic and hydrophobic interactions and by more specific adhesin–receptor binding events. Studies of bacterial adherence indicate that initial bacterial interactions with a surface are governed by long-range forces, primarily van der Waals and electrostatic interactions. The surface of most gram-negative and many grampositive bacteria is negatively charged. Thus, bacteria will often readily adhere nonspecifically to positively charged surfaces. In some cases, bacterial proteins possessing hydrophobic surfaces, including many adhesins, can also mediate nonspecific bacterial interactions with exposed host cell membrane lipids and with other hydrophobic surfaces encountered in nature. If the approach of bacteria to a surface, such as a negatively charged host cell membrane, is unfavorable, bacteria must overcome an energy barrier to establish contact. Protein–ligand binding events mediated by bacterial adhesins can often overcome or bypass repulsive forces and promote specific and intimate microbial interactions with host tissues and other surfaces. Bacteria can produce a multitude of different adhesins, usually proteins, with varying specificities for a wide range of receptor molecules. Adhesins are presented on bacterial surfaces as components of

Adhesion is a principal step in the colonization of inanimate surfaces and living tissues by bacteria. It is estimated that the majority of bacterial populations in nature live and multiply attached to a substratum. Bacteria have evolved numerous, and often redundant, mechanisms to facilitate their adherence to other organisms and surfaces within their environment. A vast number of structurally and functionally diverse bacterial adhesive molecules, called adhesins, have been identified. The adhesins expressed by different bacterial species can directly influence bacterial tropism and mediate molecular crosstalk among organisms. The Desk Encyclopedia of Microbiology ISBN: 0-12-621361-5

1

Copyright © 2003 Elsevier Ltd All rights of reproduction in any form reserved

ch01.qxd 10/9/03 6:42 PM Page 2

2

the desk encyclopedia of microbiology

filamentous, nonflagellar structures, known as pili or fimbriae, or as afimbrial (or nonfimbrial) monomeric or multimeric proteins anchored within the bacterial membrane. Other nonprotein components of bacterial membranes, including lipopolysaccharides (LPS) synthesized by gram-negative bacteria, and lipoteichoic acid in some gram-positive bacteria, can also function as adhesive molecules. Adhesins are often only minor subunits intercalated within pilus rods or located at the distal tips of pili, but they can also constitute the major structural subunits of adhesive pili. The molecular machinery required for the synthesis of many different adhesive pili and afimbrial adhesins is conserved, although the receptor specificities of the different adhesins can vary widely. Many bacterial adhesins function as lectins, mediating bacterial interactions with carbohydrate moieties on glycoproteins or glycolipids. Other adhesins mediate direct contact with specific amino acid motifs present in receptor proteins. Plant and animal cell surfaces present a large array of membrane proteins, glycoproteins, glycolipids, and other components that can potentially serve as receptors for bacterial adhesins. Protein constituents of the extracellular matrix (ECM) are also often used as bacterial receptors. In some cases, ECM proteins can function as bridges, linking bacterial and host eukaryotic cells. In addition, organic and inorganic material that coats inanimate surfaces, such as medical implants, pipes, and rocks, can act as receptors for bacterial adhesins, allowing for the establishment of microbial communities or biofilms. Adhesins also mediate interbacterial associations, facilitating the transfer of genetic material between bacteria and promoting the coaggregation of bacterial species in sites such as the oral cavity. A single bacterium can often express multiple adhesins with varying receptor specificities. These adhesins can function synergistically and, thus, enhance bacterial adherence. Alternately, adhesins may be regulated and expressed differentially, allowing bacteria to alter their adhesive repertoire as they enter different environmental situations. To date, a large number of bacterial adhesins have been described, but relatively few receptors have been conclusively identified. Bacterial adhesins can show exquisite specificity and are able to distinguish between very closely related receptor structures. The ability of bacterial adhesins to recognize specific receptor molecules is dependent upon the threedimensional architecture of the receptor in addition to its accessibility and spatial orientation. Most studies to date of bacterial adhesion have focused on host–pathogen interactions. Numerous investigations have indicated that bacterial adhesion is an essential

step in the successful colonization of host tissues and the production of disease by bacterial pathogens. Examples of adhesins expressed by bacterial pathogens and their known receptors are presented in Table 1.1. To illustrate some of the key concepts of bacterial adhesion, the modes of adhesion of a few well-characterized pathogens are discussed in the following sections.

A. Adhesins of uropathogenic Escherichia coli Uropathogenic strains of E. coli are the primary causative agents of urinary tract infections among humans. These bacteria can express two of the best characterized adhesive structures, P and type 1 pili. These pili are composite organelles, consisting of a thin fibrillar tip structure joined end-to-end to a righthanded helical rod. Chromosomally located gene clusters, that are organizationally as well as functionally homologous, encode P and type 1 pili. The P pilus tip fibrillum contains a distally located adhesin, PapG, in association with three other tip subunits, PapE, PapF, and PapK. The adhesive tip fibrillum is attached to the distal end of a thicker pilus rod composed of repeating PapA subunits. An additional subunit, PapH, anchors the PapA rod to the outer membrane. The P pilus PapG adhesin binds to the -D-galactopyranosyl-(1–4)--D-galactopyranoside (Gal(1–4) Gal) moiety present in the globoseries of glycolipids, which are expressed by erythrocytes and host cells present in the kidney. Consistent with this binding specificity, P pili have been shown to be major virulence factors associated with pyelonephritis caused by uropathogenic E. coli. Three distinct variants of the PapG adhesin (G-I, G-II, and G-III) have been identified that recognize three different Gal(1–4)Galcontaining isoreceptors: globotriaosylceramide, globotetraosylceramide (globoside), and globopentaosylceramide (the Forssman antigen). The different PapG adhesins significantly affect the tropism of pyelonephritic E. coli. For example, urinary tract E. coli isolates from dogs often encode the G-III adhesin that recognizes the Forssman antigen, the dominant Gal(1–4)Gal-containing isoreceptor in the dog kidney. In contrast, the majority of urinary tract isolates from humans express the G-II adhesin that preferentially recognizes globoside, the primary Gal(1–4)Gal-containing isoreceptor in the human kidney. In comparison with P pili, type 1 pili are more widely distributed and are encoded by more than 95% of all E. coli isolates, including uropathogenic and

ch01.qxd 10/9/03 6:42 PM Page 3

TABLE 1.1 Selected examples of bacterial adhesins and their receptors Organism

Adhesin

Receptor

Escherichia coli

P pili (PapG) Type 1 pili (FimH)

Gal(1–4)Gal D-mannose (uroplakin 1a and 1b, CD11, CD18, uromodulin) Fibronectin/laminin/ plasminogen Gal(1–4)Gal -sialyl-2,3--galactose IGLad (nLc4Cer) NeuGc(2–3)Gal4Glc

Curli (CsgA) Prs pili S pili K88 pili (K88ad) K99 pili (FanC)

Form of receptor a

Associated disease(s)

GL GP

Pyelonephritis/cystitis Cystitis

ECM

Sepsis

GL GP GL GL

Cystitis UTI, newborn meningitis Diarrhea in piglets Neonatal diarrhea in piglets, calves, and lambs

DR family DR DR-II AFA-I AFA-III F1845 Nonfimbrial adhesions 1–6 M hemagglutinin Intimin

Decay accelerating factor (SCR-3 domain)

P

Glycophorin A AM determinant of glycophorin A Tir (EPEC encoded phosphoprotein)

GP GP P

Type 4a pili Opa proteins

CD46 CD66 receptor family/HSPG

Opa50 Opc

Vitonectin/fibronectin HSPG/Vitronectin

LOS Inducible adhesin

ASGP-R Lutropin receptor

GP P GL ECM GL ECM GP GP

Listeria monocytogenes

Internalin

E-cadherin

GP

Listeriosis (meningitis, septicemia, abortions, gastroenteritis)

Haemophilus influenzae

Hemagglutinating pili

AnWj antigen/lactosylceramide

Respiratory tract infections

Hsp-70-related proteins HMW1, HMW2

Sulfoglycolipids Negatively charged glycoconjugates

GP GL GL GP

Campylobacter jejuni

CadF

Fibronectin

ECM

Gastroenteritis

Yersinia

Invasin YadA

1 integrins P Cellular fibronectin/collagen/laminin ECM

Plague, Enterocolitis

Bordetella pertussis

FHA Pertactin, BrkA Pertussis toxin

CR3 integrin Integrins Lactosylceramides/gangliosides

P P GP/GL

Whooping cough

Mycobacterium Streptococcus

BCG85 complex, FAP proteins Protein F family Polysaccharide capsule ZOP, FBP4, GAPDH Lipoteichoic acid (LTA)

Fibronectin Fibronectin CD44 Fibronectin Fibronectin/macrophage scavenger receptor CD46/fucosylated glycoconjugates/fibronectin

ECM ECM GP ECM ECM/GP

Tuberculosis, leprosy Pharyngitis, scarlet fever, erysipelas, impetigo, rheumatic fever, UTI, dental caries, neonatal sepsis, glomerulonephritis, endocarditis, pneumonia, meningitis

Neisseria

M protein

Staphylococcus

a

FnbA, FnbB Can Protein A (Spa) ClfA EbpS

Fibronectin Collagen von Willebrand factor Fibrinogen Elastin

UTI UTI UTI UTI, diarrhea diarrhea UTI, newborn meningitis Pyelonephritis Diarrhea

GP/ECM

ECM ECM GP ECM ECM

Gonorrhea/meningitis

Skin lesions, pharyngitis, pneumonia, endocarditis, toxic shock syndrome, food poisoning

P, protein–protein interactions; GP, interaction with glycoproteins; GL, glycolipids; ECM, extracellular matrix proteins.

ch01.qxd 10/9/03 6:42 PM Page 4

4

the desk encyclopedia of microbiology

commensal intestinal strains. The type 1 pilus tip fibrillum is comprised of two subunits, FimF and FimG, in addition to the adhesin, FimH. The adhesive tip is connected to the distal end of a thicker pilus rod composed of repeating FimA subunits. In addition to its localization within the pilus tip, the FimH adhesin also appears to be occasionally intercalated along the length of the type 1 pilus rod. FimH binds to mannose containing host receptors expressed by a wide variety of host cell types and has been shown to be a significant virulence determinant for the development of bladder infections. Natural phenotypic variants of the FimH adhesin have been identified by Sokurenko et al. (1998), which differentially bind to mono-mannose structures. Interestingly, most uropathogenic isolates express FimH variants that bind well to monomannose residues, whereas most isolates from the large intestine of healthy humans express FimH variants that interact poorly with mono-mannose structures. Mono-mannose residues are abundant in the oligosaccharide moieties of host proteins, known as uroplakins, that coat the luminal surface of the bladder epithelium. In vitro binding assays by Wu et al. (1996) have demonstrated that type 1-piliated E. coli can specifically bind two of the uroplakins, UP1a and UP1b. Scanning and high-resolution electron microscopy have shown that type 1 pili can mediate direct and intimate bacterial contact with the uroplakin-coated bladder epithelium (Fig. 1.1). The assembly of P pili and type 1 pili requires two specialized assembly proteins: a periplasmic chaperone and an outer membrane usher. Periplasmic chaperones facilitate the import of pilus subunits across the inner membrane and mediate their delivery to outer membrane usher complexes, where subunits are assembled into pili. Homologous chaperone/usher pathways modulate the assembly of over 30 different adhesive organelles, expressed by uropathogenic E. coli and many other gram-negative pathogens. Among the adhesive structures assembled via a chaperone/usher pathway by uropathogenic E. coli are S pili, nonfimbrial adhesin I, and members of the Dr adhesin family. This family includes the uropathogenic-associated afimbrial adhesins AFA-I and AFA-III and the fimbrial adhesin Dr, in addition to the diarrhea-associated fimbrial adhesin F1845. These adhesins recognize the Dra blood group antigen present on decay accelerating factor (DAF), a complement regulatory factor expressed on erythrocytes and other tissues, including the uroepithelium. These four members of the Dr adhesin family appear to recognize different epitopes of the Dra antigen. The Dr adhesin, but not the other three, also recognizes type IV collagen. Members of the Dr adhesin family

are proposed to facilitate ascending colonization and chronic interstitial infection of the urinary tract. It is unclear why the Dr and F1845 adhesins assemble into fimbria while AFA-I and AFA-III are assembled as nonfimbrial adhesins on the bacterial surface. It has been suggested that afimbrial adhesins, such as AFA-I and AFA-III, are derived from related fimbrial adhesins, but have been altered such that the structural attributes required for polymerization into a pilus are missing while the adhesin domain remains functional and anchored on the bacterial surface.

B. Neisserial adhesins Neisseria gonorrhoeae and N. meningitidis are exclusively human pathogens that have developed several adhesive mechanisms to colonize mucosal surfaces. Initial contact with mucosal epithelia by Neisseria species is mediated by type 4a pili. These adhesive organelles are related to a group of multifunctional structures expressed by a wide diversity of bacterial species, including Pseudomonas aeruginosa, Moraxella species, Dichelobacter nodus, and others. Type-4a pili are assembled by a type II secretion system that is distinct from the chaperone/usher pathway. They are comprised primarily of a small subunit, pilin, that is packaged into a helical arrangement within pili. The type 4a pilin can mediate bacterial adherence, but in Neisseria species, a separate, minor tip protein, PilC, has also been implicated as an adhesin. A eukaryotic membrane protein, CD46, is proposed to be a host receptor for type 4a pili expressed by N. gonorrhoeae, although it is currently unclear which pilus component binds this host molecule. Following primary attachment mediated by type-4a pili, more intimate contact with mucosal surfaces is apparently established by the colony opacity-associated (Opa) proteins of Neisseria species. These proteins constitute a family of closely related but size-variable outer membrane proteins that are expressed in a phase variable fashion. Opa proteins mediate not only adherence, but they also modulate bacterial invasion into host cells. A single neisserial strain can encode from 3 to 11 distinct Opa variants, with each Opa protein being expressed alternately of the others. The differential expression of Opa variants can alter bacterial antigenicity and possibly modify bacterial tropism for different receptors and host cell types. Some Opa variants recognize carbohydrate moieties of cell surface-associated heparin sulfate proteoglycans (HSPGs), which are common constituents of mammalian cell membranes. The majority of Opa variants, however, bind via protein–protein interactions to CD66 transmembrane glycoproteins, which comprise

ch01.qxd 10/9/03 6:42 PM Page 5

adhesion, bacterial

5

FIGURE 1.1 Type 1 pilus-mediated bacterial adherence to the mouse bladder epithelium was visualized by (A and B) scanning and (C–H) high-resolution freeze–fracture, deep-etch electron microscopy. Mice were infected via transurethral inoculation with type 1-piliated uropathogenic E. coli. Bladders were collected and processed for microscopy at 2 h. after infection. Bacteria adhered randomly across the bladder lumenal surface, both singly and in large, biofilmlike microcolonies, some of which contained several hundred bacteria (A and B). The type 1 pili-mediating bacterial adherence were resolved by high-resolution electron microscopy techniques. The adhesive tips of type 1 pili make direct contact with the uroplakin-coated surface of the bladder epithelium (D–G). Hexagonal arrays of uroplakin complexes are visible. The boxed areas in (C) and (D) are shown magnified, respectively, in (D) and (E). In (H), type 1 pili span from the host cell membrane on the right to the bacterium on the left. These images demonstrate that type 1 pili can mediate intimate bacterial attachment to host bladder epithelial cells. Scale bars indicate 5 m (A and B), 0.5 m (C and F), and 0.1 m (D, E, G, H) (Plate 1). (Reprinted with permission from Mulvey, M. A., et al. (1998). Induction and evasion of host defenses by type 1-piliated uropathogenic Escherichia coli. Science 282, 1494–1497. Copyright 1998 American Association for the Advancement of Science.).

a subset of the carcinoembryonic antigen (CEA) receptor family of the immunoglobulin super-family. Individual Opa variants specifically recognize distinct CD66 receptors and this likely influences both the tissue tropism of Neisseria and the host cell responses to neisserial attachment. In addition to pili and Opa proteins, the lipopolysaccharide (lipooligosaccharide, LOS) and a distinct outer membrane protein, Opc, expressed by Neisseria can also influence bacterial adhesion and invasion. Deconvoluting the various roles of the different adhesive components of Neisseria during the infection process remains a major challenge.

C. Adhesins of Haemophilus influenzae Haemophilus influenzae is a common pathogen of the human respiratory tract. Isolates of H. influenzae can be divided into encapsulated and nonencapsulated, or nontypable, forms. Prior to the use of H. influenzae conjugate vaccines, capsulated strains of H. influenzae were the primary cause of childhood bacterial meningitis and a major cause of other bacteremic diseases in children. Vaccines effective against nontypable strains have not yet been developed and these strains remain important human pathogens, causing pneumonia, otitis media, sinusitis, and bronchitis. Several

ch01.qxd 10/9/03 6:42 PM Page 6

6

the desk encyclopedia of microbiology

adhesins have been identified which facilitate the colonization of the respiratory epithelium by both encapsulated and nontypable H. influenzae. During the initial stages of the infection process, nontypable H. influenzae associates with respiratory mucus, apparently through interactions between bacterial outer membrane proteins (OMPs P2 and P5) and sialic acid-containing oligosaccharides within the mucus. Both nontypable and encapsulated strains of H. influenzae can initiate direct contact with the respiratory epithelium via adhesive pili. Over 14 serological types of adhesive pili have been indentified in H. influenzae. These pili are composite structures assembled by chaperone/usher pathways similar to those used by uropathogenic E. coli to assemble P and type 1 pili. Piliated strains of H. influenzae preferentially bind to nonciliated cells or damaged epithelium. The pili of H. influenzae can recognize the AnWj antigen, in addition to gangliosides and other compounds containing siallyllactoceramide. Following initial attachment mediated by pili, the polysaccharide capsule of encapsulated strains is reduced, enabling a second adhesin, Hsf, to establish more intimate bacterial contact with host epithelial cells. Hsf assembles into short, thin fibrils on the bacterial surface. While Hsf expression is restricted to encapsulated strains of H. influenzae, a subpopulation of nontypable strains expresses a Hsf homolog called Hia. Both Hsf and Hia share homology with other bacterial adhesins including AIDA-1, an adherence factor produced by diarrheagenic E. coli. Instead of adhesive pili and Hia, the majority of nontypable H. influenzae isolates produce two alternate adhesins: high molecular weight surface-exposed proteins called HMW1 and HMW2. These two adhesins share significant sequence identity with each other and are similar to filamentous hemagglutinin (FHA), an adhesin and colonization factor expressed by Bordetella pertussis. HMW1 and HMW2 have distinct adhesive specificities and may function at different steps in the infection process. The receptors for the HMW adhesins appear to be negatively charged glycoconjugates that have not yet been completely defined. Nontypable H. influenzae encodes several other adhesive factors, including two Hsp-70-related proteins, which can mediate bacterial binding to sulfoglycolipids. Interestingly, other heat shock proteins have been implicated in the adherence of other microbial pathogens including Helicobacter pylori, Mycoplasma, and Chlamydia trachomatis. Work by St. Geme and coworkers (1998) has highlighted an additional adhesin, Hap, which is expressed by virtually all nontypable H. influenzae isolates. Hap mediates low-level adherence to epithelial

cells, complementing the binding activities of pili and Hia or HMW1 and HMW2. Hap also promotes interbacterial associations leading to bacterial aggregation and microcolony formation on the epithelial surface. The mature Hap adhesin consists of a C-terminal outer membrane protein domain, designated Hap, and a larger extracellular domain designated Haps. The Haps domain, which is responsible for mediating adherence, has serine protease activity and can be autoproteolytically cleaved, releasing itself from the bacterial surface. Interestingly, secretory leukocyte protease inhibitor (SLPI), a natural host component of respiratory-tract secretions, which possibly protects the respiratory epithelium from proteolytic damage during acute inflammation, has been shown to inhibit Hap autoproteolysis and enhance bacterial adherence. Despite the presence of SLPI, Haps-mediated adherence in vivo is likely transient. Over time, the eventual autoproteolysis and release of the Haps adhesin domain from the bacterial surface may allow bacterial spread from microcolonies on the respiratory epithelium and aid the bacteria in evading the host immune response. Identification of the receptor molecules recognized by Hap awaits further studies.

D. Adherence to components of the extracellular matrix One of the principal functions of the ECM is to serve as substrate for the adherence of eukaryotic cells within animal tissues. The ECM is composed of polysaccharides and numerous proteins including fibronectin, vitronectin, laminin. elastin, collagen, fibrinogen, tenascin, entactin, and others. Thin flexible mats of specialized ECM, known as basal laminae or basement membranes, underlie all epithelial cells and surround individual fat cells, muscle cells, and Schwann cells. Binding of ECM proteins is one of the primary mechanisms used by many pathogenic bacteria to adhere to host tissues. Bacterial adhesins have been identified which recognize specific components of the ECM and a few adhesins, such as the Opa50 protein of Neisseria and the YadA adhesin of Yersinia enterolitica, are able to recognize multiple ECM components. Some bacterial adhesins preferentially recognize immobilized, cell-bound ECM components over soluble forms. The YadA adhesin expressed by Y. enterolitica, for example, mediates adherence to cellbound fibronectin, but not to soluble fibronectin within plasma. This may allow Y. enterolitica to more efficiently bind tissue rather than circulating molecules. The tissue distribution of ECM components can directly influence the tropism of a bacterial pathogen.

ch01.qxd 10/9/03 6:42 PM Page 7

7

adhesion, bacterial For example, Mycobacterium leprae, the causative agent of leprosy, binds LN-2, an isoform of the ECM component laminin. This ECM component recognizes a host cell-surface receptor, -dystroglycan, and serves as a bridge linking host and bacterial cells. M. leprae targets the Schwann cells of the peripheral nervous system and can also invade the placenta and striated muscle of leprosy patients. The tissue distribution of LN-2, which is restricted to the basal laminae of Schwann cells, striated muscles, and trophoblasts of the placenta, directly correlates with sites of natural infection by M. leprae. In contrast to the restricted tissue distribution of LN-2, most components of the ECM are more widely apportioned and can interact with receptor molecules expressed by a broad range of cell types present within a variety of different tissues. By interacting with widely distributed components of the ECM, bacteria greatly enhance their adhesive potential. Numerous bacteria are able to bind fibronectin, an ECM component present in most tissues and body fluids and a prominent constituent of wounds. The bacterial adhesins that bind fibronectin are diverse. For example, E. coli and Salmonella species express thin, irregular, and highly aggregated surface fibers, known as curli, that bind fibronectin in addition to other receptor molecules. Mycobacterium species produce at least five fibronectin-binding molecules, three of which are related and collectively known as the BCG85 complex. Streptococcus expresses an even larger number of different fibronectin-binding adhesins, including ZOP, lipoteichoic acid, GAPDH, FBP54, M protein, and several related molecules represented by Protein F. Binding of Protein F and related adhesins to fibronectin is specific and essentially irreversible. Members of the Protein F family of adhesins have similar domain architectures, although they appear to interact with fibronectin differently. Protein F possesses two distinct domains, composed of repeated sequence motifs, which bind independently of each other to different sites at the N-terminus of fibronectin. Additional fibronectin-binding proteins related to the Protein F family of adhesins have also been identified in Staphylococcus. These gram-positive bacteria, in addition to producing fibronectin-binding proteins, can also express an array of other adhesive molecules, which bind other widely distributed ECM components, including collagen, fibrinogen, and elastin. By encoding a large repertoire of adhesins able to recognize ECM components, Streptococcus, Staphylococcus, and other pathogens, presumably, increase their capacity to effectively bind and colonize sites within host tissues.

II. CONSEQUENCES OF BACTERIAL ADHESION Research in recent years has demonstrated that interactions between bacterial adhesins and receptor molecules can act as trigger mechanisms, activating signal transduction cascades and altering gene expression in both bacterial and host cells. Zhang and Normark showed in 1996 that the binding of host cell receptors by P pili activated the transcription of a sensor– regulator protein, AirS, which regulates the bacterial iron acquisition system of uropathogenic E. coli. This response may enable uropathogens to more efficiently obtain iron and survive in the iron-poor environment of the urinary tract. Around the same time, Wolf-Watz and colleagues showed, using Y. pseudotuberculosis, that bacterial contact with host cells could increase the rate of transcription of virulence determinants called Yop effector proteins. More recently, Taha and coworkers (1998) demonstrated that transcription of the PilC1 adhesin of N. meningitidis was transiently induced by bacterial contact with host epithelial cells. The PilC1 adhesin can be incorporated into the tips of type-4a pili, but it can also remain associated with the bacterial outer membrane, where it can, presumably, facilitate pilus assembly. The up-regulation of the PilC1 adhesin may enhance bacterial adherence to host cells by promoting the localization of PilC1 into the tips of type 4a pili. Signal transduction pathways are activated within host eukaryotic cells in response to attachment mediated by many different bacterial adhesins. For example, the binding of type-4a pili expressed by Neisseria to host cell receptors (presumably, CD46) can stimulate the release of Ca stores within target epithelial cells. Fluxes in intracellular Ca concentrations are known to modulate a multitude of eukaryotic cellular responses. Similarly, the binding of P pili to Gal(1–4)Gal-containing host receptors on uroepithelial cells can induce the release of ceramides, important second messenger molecules that can influence a number of signal transduction processes. Signals induced within urepithelial cells upon binding P-piliated bacteria result in the up-regulation and eventual secretion of several immunoregulatory cytokines. The binding of type 1-piliated and other adherent bacteria to a variety of host epithelial and immune cells has also been shown to induce the release of cytokines, although the signaling pathways involved have not yet been well defined. In some cases, bacteria may co-opt host signal transduction pathways to enhance their own attachment. For example, binding of the FHA adhesin of B. pertussis to a monocyte integrin receptor complex activates host signal pathways that lead to the

ch01.qxd 10/9/03 6:42 PM Page 8

8

the desk encyclopedia of microbiology

up-regulation of another integrin, complement receptor 3 (CR3). FHA can bind CR3 through a separate domain and, thus, enhance the adhesion of B. pertussis. The activation of host signal pathways following bacterial attachment can result in dramatic rearrangements of the eukaryotic cytoskeleton, which can lead to the internalization of adherent bacteria. Many pathogenic bacteria invade host eukaryotic cells to evade immune responses or to pass through cellular barriers, such as the intestinal epithelium. In some cases, bacteria introduce effector molecules into their target host cells to trigger cytoskeletal rearrangements and intense ruffling of the host cell membrane that results in bacterial uptake. In other situations, bacterial adhesins (which are sometimes referred to as invasins) more directly mediate bacterial invasion by interacting with host cell membrane receptors that sequentially encircle and envelope the attached bacterium. This type of invasion is referred to as the “zipper” mechanism and requires the stimulation of host signaling cascades, including the activation of protein tyrosine kinases. The invasin protein of Yersinia and internalin expressed by Listeria can both mediate bacterial internalization into host cells by such a zipper mechanism by interacting with 1-integrin and E-cadherin, respectively. The Opa proteins of Neisseria can also mediate bacterial internalization into host cells by a zipperlike mechanism. Recent work by several labs has indicated that fimbrial adhesins, such as FimH within type 1 pili, can also function as invasins.

III. TARGETING ADHESINS FOR ANTIMICROBIAL THERAPY Bacterial adhesin–receptor binding events are critical in the pathogenesis of virtually every bacterial disease. In some cases, the knockout of a specific adhesin can greatly attenuate bacterial virulence. Uropathogenic E. coli strains, for example, which have been engineered to express type 1 pili lacking the FimH adhesin, are unable to effectively colonize the bladder. Similarly, a P-piliated pyelonephritic strain of E. coli lacking a functional PapG adhesin is unable to infect the kidney. For many other bacteria, attachment is a multifaceted process involving several adhesins that may have complementing and overlapping functions and receptor specificities. In these cases, it has been more difficult to discern the roles of individual adhesins in disease processes. The construction of mutants with knockouts in more than one adhesin is beginning to shed light on the interrelationships between multiple bacterial adhesins.

The central role of bacterial adhesins at the host–pathogen interface during the infection process has made them attractive targets for the development of new antimicrobial therapies. Vaccines directed against individual adhesins and adhesive pili have had some success in the past. However, antigenic variation of the major immunodominant domains of some adhesive organelles and the immunorecessive nature of others have frustrated progress in this area. Fortunately, by unraveling the molecular details of adhesin structure and biogenesis, substantial progress is being made. For example, the identification of FimH as the adhesive subunit of type 1 pili and the elucidation of the chaperone/usher pathway used to assemble these adhesive organelles has made it possible to purify large quantities of native FimH and to test its efficacy as a vaccine. Unlike the major type 1 pilus subunit, FimA, there is relatively little heterogeneity among the FimH adhesins expressed by diverse E. coli strains. The use of purified FimH as a vaccine, rather than whole type 1 pili in which FimH is present only in low numbers, has proven to significantly enhance the host immune response against the FimH adhesin. In early trials, FimH-vaccinated animals showed substantial resistance to infection by a wide variety type 1-piliated uropathogenic E. coli strains. In addition to the prophylactic approach of generating vaccines to inhibit bacterial adhesion, other antiadhesin strategies are being explored. With increased knowledge of the mechanisms used to assemble adhesins on the bacterial surface, it may be possible to design specific inhibitors of adhesin biogenesis. For example, synthetic compounds that specifically bind and inactivate periplasmic chaperones could potentially inhibit the biogenesis of a wide range of bacterial adhesive organelles. The use of soluble synthetic receptor analogs that bind bacterial adhesins substantially better than the natural monomeric ligands represents an additional strategy for inhibiting bacterial attachment and colonization. Recent advances in the synthesis of multimeric carbohydrate polymers have highlighted the possibility of creating high affinity receptor analogs that could potentially work at pharmacological concentrations within patients. Such compounds could also be used to competitively remove adherent bacteria from medical implants, industrial pipes, and other surfaces. Furthermore, it may be possible to inhibit multiple bacterial adhesins with a single compound by incorporating several receptor analogs within a single carbohydrate polymer. Continued research into the structure, function, and biogenesis of bacterial adhesins promises not only to enhance our knowledge

ch01.qxd 10/9/03 6:42 PM Page 9

9

adhesion, bacterial of pathogenic processes, but may also help augment our current arsenal of antimicrobial agents.

BIBLIOGRAPHY Dalton, H. M., and March, P. E. (1998). Molecular genetics of bacterial attachment and biofouling. Curr. Op. Biotech. 9, 252–255. Davey, M. E., and O’Toole, G. (2000). Microbial biofilms: from ecology to molecular genetics. Microbiol. Mol. Biol. Rev. 64, 847–867. Dehio, C., Gray-Owen, S. D., and Meyer, T. F. (1998). The role of neisserial Opa proteins in interactions with host cells. Trends Microbiol. 6, 489–495. Finlay, B. B., and Falkow, S. (1997). Common themes in microbial pathogenicity revisited. Microbiol. Mol. Biol. Rev. 61, 136–169. Foster, T. J., and Höök, M. (1998). Surface adhesins of Staphylococcus aureus. Trends Microbiol. 6, 484–488. Goldhar, J. (1996). Nonfimbrial adhesins of Escherichia coli. In “Toward Anti-Adhesion Therapy for Microbial Diseases” (Kahane and Ofek, eds.), pp. 63–72. Plenum Press, New York. Hultgren, S. J., Jones, C. H., and Normark, S. (1996). Bacterial adhesins and their assembly. In “Escherichia coli and Salmonella,” Vol. 2 (F. C. Neidhardt, ed.), pp. 2730–2756. ASM Press, Washington, DC. Jacques, M., and Paradis, S. E. (1998). Adhesin–receptor interactions in Pasteurellaceae. FEMS Microbiol. Rev. 22, 45–59. Jenkinson, H. F., and Lamont, R. J. (1997). Streprococcal adhesion and colonization. Crit. Rev. Oral Biol. Med. 8, 175–200. Kerr, J. R. (1999). Cell adhesion molecules in the pathogenesis of and host defence against microbial infection. Mol. Pathol. 52, 220–230.

Kolenbrander, P. E. (2000). Oral microbial communities: biofilms, interactions, and genetic systems. Annu. Rev. Microbiol. 54, 413–437. Lingwood, C. A. (1998). Oligosaccharide receptors for bacteria: A view to a kill. Curr. Op. Chem. Biol. 2, 695–700. O’Toole, G., Kaplan, H. B., and Kolter, R. (2000). Biofilm formation as microbial development. Annu. Rev. Microbiol. 54, 49–79. Schilling, J. D., Mulvey, M. A., and Hultgren, S. J. (2001). Structure and function of Escherichia coli type 1 pili: new insight into the pathogenesis of urinary tract infections. J. Infect. Dis. 183, (Suppl 1), S36–S40. Sharon, N. (1996). Carbohydrate–lectin interactions in infectious disease. In “Toward Anti-Adhesion Therapy for Microbial Diseases” (Kahane and Ofek, eds.), pp. 1–8. Plenum Press, New York. Soto, G. E., and Hultgren, S. J. (1999). Bacterial adhesins: Common themes and variations in architecture and assembly. J. Bacteriol. 181, 1059–1071. Whittaker, C. J., Klier, C. M., and Kolenbrander, P. E. (1996). Mechanisms of adhesion by oral bacteria. Annu. Rev. Microbiol. 50, 513–552. Wilson, M. (2002). “Bacterial Adhesion to Host Tissues.” Cambridge University Press, Cambridge. Wizemann, T. M., Adamou, J. E., and Langermann, S. (1999). Adhesins as targets for vaccine development. Emerg. Infect. Dis. 5, 395–403.

WEBSITE The E. coli Cell Envelope Protein Data Collection includes many proteins involved in adhesion http://www.cf.ac.uk/biosi/staff/ehrmann/tools/ecce/ecce.htm

ch02.qxd 10/9/03 6:44 PM Page 10

2 Agrobacterium and plant cell transformation Peter J. Christie University of Texas Health Science Center at Houston

Agrobacterium tumefaciens is a gramnegative soil bacterium with the unique ability to infect plants through a process that involves delivery of a specific segment of its genome to the nuclei of susceptible plant cells. The transferred DNA (T-DNA) is a discrete region of the bacterial genome defined by directly repeated border sequences. The T-DNA is important for infection because it codes for genes which, when expressed in the plant cell, disrupt plant cell growth and division events.

GLOSSARY autoinducer An acyl homoserine lactone secreted from bacteria which, under conditions of high cell density, passively diffuses across the bacterial envelope and activates transcription. border sequences 25-bp direct, imperfect repeats that delineate the boundaries of T-DNA. conjugal pilus An extracellular filament encoded by a conjugative plasmid involved in establishing contact between plasmid-carrying donor cells and recipient cells. conjugation Transfer of DNA between bacteria by a process requiring cell-to-cell contact. mobilizable plasmid Conjugal plasmid that carries an origin of transfer (oriT) but lacks genes coding for its own transfer across the bacterial envelope. T-DNA Segment of the Agrobacterium genome transferred to plant cells. transconjugant A cell that has received a plasmid from another cell as a result of conjugation. transfer intermediate A nucleoprotein particle composed of a single strand of the DNA destined for export and one or more proteins that facilitate DNA delivery to recipient cells. type IV transporters A conserved family of macromolecular transporters evolved from ancestral conjugation systems for the purpose of exporting DNA or protein virulence factors between prokaryotic cells or to eukaryotic hosts. The Desk Encyclopedia of Microbiology ISBN: 0-12-621361-5

Approximately 20 years ago, it was discovered that oncogenic DNA could be excised from the T-DNA and in its place virtually any gene of interest could be inserted. Agrobacterium tumefaciens could then efficiently deliver the engineered T-DNA to a wide array of plant species and cell types. Transformed plant cells could be selected by cotransfer of an antibiotic resistance marker and regenerated into fertile, transgenic plants. The discovery that A. tumefaciens is a natural and efficient DNA delivery vector for transforming plants is largely responsible for the burgeoning industry of plant genetic engineering, which today has many diverse goals ranging from crop improvement to the use of plants as “pharmaceutical factories” for highlevel production of biomedically important proteins. Because of the dual importance of Agrobacterium as a plant pathogen and as a DNA delivery system, an extensive literature has emerged describing numerous aspects of the infection process and the myriad of ways this organism has been exploited for plant genetic

10

Copyright © 2003 Elsevier Ltd All rights of reproduction in any form reserved

ch02.qxd 10/9/03 6:44 PM Page 11

agrobacterium and plant cell transformation engineering. The aim of this article is to summarize recent advances in our knowledge of this system, with particular emphasis on chemical signaling events, the T-DNA processing and transport reactions, and exciting novel applications of Agrobacterium-mediated gene delivery to eukaryotic cells.

I. OVERVIEW OF INFECTION PROCESS Agrobacterium species are commonly found in a variety of environments including cultivated and nonagricultural soils, plant roots, and even plant vascular systems. Despite the ubiquity of Agrobacterium species in soil and plant environments, only a small percentage of isolates are pathogenic. Two species are known to infect plants by delivering DNA to susceptible plant cells. Agrobacterium tumefaciens is the causative agent of crown gall disease, a neoplastic disease characterized by uncontrolled cell proliferation and formation of unorganized tumors. Agrobacterium rhizogenes induces formation of hypertrophies with a hairy root appearance referred to as “hairy root” disease. The pathogenic strains of both species possess large plasmids that encode most of the genetic information required for DNA transfer to susceptible plant cells. The basic infection process is similar for both species, although the gene composition of the transferred DNA

11

(T-DNA) differs, and therefore, so does the outcome of the infection. This article focuses on recent advances in our understanding of the A. tumefaciens infection process. The basic infection cycle can be described as follows (Fig. 2.1). Pathogenic A. tumefaciens strains carry large, ~180-kb tumor-inducing (Ti) plasmids. The Ti plasmid harbors the T-DNA and virulence (vir) genes involved in T-DNA delivery to susceptible plant cells. As with many bacterial pathogens of plants and mammals, A. tumefaciens infects only at wound sites. As part of the plant wound response, various plant cell wall precursors, including defined classes of phenolic compounds and monosaccharide sugars, are released into the extracellular milieu. These molecules play an important role in the infection process as inducers of the vir genes. On vir gene activation, T-DNA is processed into a nucleoprotein particle termed the T-complex. The T-complex contains information for (i) export across the A. tumefaciens cell envelope via a dedicated transport system, (ii) movement through the plant plasma membrane and cytosol, (iii) delivery to the plant nuclear pore, and (iv) integration into the plant genome. Once integrated into the plant genome, T-DNA genes are expressed and the resulting gene products ultimately disrupt the balance of two endogenous plant hormones that synergistically coordinate plant cell growth and division events. The imbalance of these hormones contributes to loss of cell growth control

FIGURE 2.1 Overview of the Agrobacterium tumefaciens infection process. Upon activation of the VirA/VirG two-component signal transduction system by signals released from wounded plant cells, a single strand of T-DNA is processed from the Ti plasmid and delivered as a nucleoprotein complex (T-complex) to plant nuclei. Expression of T-DNA genes in the plant results in loss of cell growth control and tumor formation (see text for details).

ch02.qxd 10/9/03 6:44 PM Page 12

12

the desk encyclopedia of microbiology

and, ultimately, the proliferation of crown gall tumors.

II. Ti PLASMID Genetic and molecular analyses have resulted in the identification of two regions of the Ti plasmid that contribute directly to infection (Fig. 2.2). The first is the T-DNA, typically a segment of 20–35 kb in size delimited by 25-bp directly repeated border sequences. The T-DNA harbors genes that are expressed exclusively in the plant cell. Transcription of T-DNA in the plant cell produces 3 polyadenylated RNA typical of eukaryotic RNA message that is translated in the cytoplasm. The translated proteins ultimately disrupt plant cell growth and division processes resulting in the characteristic tumorous phenotype. The second region of the Ti plasmid involved in infection harbors the genes responsible for processing the T-DNA into a transfer-competent nucleoprotein particle and exporting this particle across the bacterial envelope. Two additional regions of the Ti plasmid code for functions that are not essential for the T-DNA transfer process per se but are nevertheless intimately associated with the overall infection process. One of these regions harbors genes involved in catabolism of novel amino acid derivatives termed opines that A. tumefaciens induces plants to synthesize as a result of T-DNA transfer. The second region encodes Ti plasmid transfer functions for distributing copies of the Ti plasmid and its associated virulence factors to other A. tumefaciens cells by a

FIGURE 2.2 Regions of the Ti plasmid that contribute to infection (vir region and T-DNA), cell survival in the tumor environment (opine catabolism), and conjugal transfer of the Ti plasmid to recipient agrobacteria (tra and trb). The various contributions of the vir gene products to T-DNA transfer are listed. T-DNA, delimited by 25-bp border sequences (black arrows), codes for biosynthesis of auxins, cytokinins, and opines in the plant. OD, overdrive sequence that enhances VirD2-dependent processing at the T-DNA border sequences.

process termed conjugation. Intriguing recent work has described a novel regulatory cascade involving chemical signals released both from the transformed plant cells and from the infecting bacterium that activates conjugal transfer of the Ti plasmid among A. tumefaciens cells residing in the vicinity of the plant tumor.

A. T-DNA The T-DNA is delimited by 25-bp direct, imperfect repeats termed border sequences (Fig. 2.2). Flanking one border is a sequence termed overdrive that functions to stimulate the T-DNA processing reaction. All DNA between the border sequences can be excised and replaced with genes of interest, and A. tumefaciens will still efficiently transfer the engineered T-DNA to plant cells. This shows that the border sequences are the only cis elements required for T-DNA transfer to plant cells and that genes encoded on the T-DNA play no role in movement of T-DNA to plant cells. Instead, the T-DNA genes code for synthesis of two main types of enzymes within transformed plant cells. Oncogenes synthesize enzymes involved in the synthesis of two plant growth regulators, auxins and cytokinins. Production of these plant hormones results in a stimulation of cell division and a loss of cell growth control leading to the formation of characteristic crown gall tumors. The second class of enzymes code for the synthesis of novel amino acid derivatives termed opines. For example, the pTiA6 plasmid carries two T-DNAs that code for genes involved in synthesis of octopines, a reductive condensation product of pyruvate and arginine. Other Ti plasmids carry T-DNAs that code for nopalines, derived from -ketoglutarate and arginine, and still others code for different classes of opines. Plants cannot metabolize opines. However, as described later, the Ti plasmid carries opine catabolism genes that are responsible for the active transport of opines and their degradation, thus providing a source of carbon and nitrogen for the bacterium. The “opine concept” was developed to rationalize the finding that A. tumefaciens evolved as a pathogen by acquiring the ability to transfer DNA to plant cells. According to this concept, A. tumefaciens adapted a DNA conjugation system for interkingdom DNA transport to incite opine synthesis in its plant host. The cotransfer of oncogenes ensures that transformed plant cells proliferate, resulting in enhanced opine synthesis. The environment of the tumor, therefore, is a rich chemical environment favorable for growth and propagation of the infecting A. tumefaciens. It is also notable that a given A. tumefaciens strain catabolizes only those opines that it incites plant cells to synthesize. This ensures a selective advantage of the infecting bacterium over other

ch02.qxd 10/9/03 6:44 PM Page 13

agrobacterium and plant cell transformation A. tumefaciens strains that are present in the vicinity of the tumor.

B. Opine catabolism The regions of two Ti plasmids coding for opine catabolism have been sequenced and shown to code for three functions related to opine catabolism (Fig. 2.2). The first is a regulatory function that controls expression of the opine transport and catabolism genes. The regulatory protein is OccR for the octopine catabolism region of plasmid pTiA6. Recent studies have shown that OccR positively regulates expression of the occ genes involved in octopine uptake and catabolism by inducing a bend in the DNA at the OccR binding site. Interestingly, octopine alters both the affinity of OccR for its target site and the angle of the DNA bend, suggesting that octopine modulates OccR regulatory activity. The regulatory protein is AccR for the nopaline catabolism region of plasmid pTiC58. In contrast to OccR, AccR functions as a negative regulator of acc genes involved in nopaline catabolism. The second and third functions, opine transport and catabolism, are encoded by several genes that are transcribed from a single promoter. At the proximal end of the operon is a set of genes that code for one or more transport systems conferring opine-specific binding and uptake. Typically, one or more of these genes encode proteins homologous to energy-coupling proteins found associated with the so-called ATP-binding cassette (ABC) superfamily of transporters. The ABC transporters

13

are ubiquitous among bacterial and eukaryotic cells, and they provide a wide variety of transport functions utilizing the energy of ATP hydrolysis to drive the transport reaction. At the distal end of the operon are genes involved in cleaving the opines to their parent compounds for use as carbon and nitrogen sources for the bacterium.

C. Ti plasmid conjugation The Ti plasmid transfer (tra and trb) functions direct the conjugal transmission of the Ti plasmid to bacterial recipient cells. The transfer genes of conjugative plasmids code for DNA processing and transport system that assembles at the bacterial envelope for the purpose of delivering conjugal DNA transfer intermediates to recipient cells. DNA sequence studies have shown that one set of transfer genes codes for many proteins that are related to components of other plasmid and protein toxin transport systems. As described later in more detail, this evolutionarily conserved family of transporters is referred to as a type IV secretion system. 1. Autoinduction-dependent Ti plasmid transfer Recent work has demonstrated that a regulatory cascade exists to activate Ti plasmid transfer under conditions of high cell density (Fig. 2.3). This regulatory cascade initiates when A. tumefaciens imports opines released from plant cells. For the octopine pTiA6 plasmid, OccR acts in conjunction with octopine to activate transcription of the occ operon. Although the

FIGURE 2.3 A schematic of chemical signaling events between Agrobacterium and the transformed plant cell. Signals released from wounded plant cells initiate the infection process leading to tumor formation. Opines released from wounded plant cells activate opine catabolism functions for growth of infecting bacteria. Opines also activate synthesis of TraR for autoinducer (AAI) synthesis. TraR and AAI at a critical concentration activate the Ti plasmid conjugation functions (see text for details).

ch02.qxd 10/9/03 6:44 PM Page 14

14

the desk encyclopedia of microbiology

majority of the occ operon codes for octopine transport and catabolism functions, the distal end of the occR operon encodes a gene for a transcriptional activator termed TraR. TraR is related to LuxR, an activator shown nearly 20 years ago to regulate synthesis of an acyl homoserine lactone termed autoinducer. Cells that synthesize autoinducer molecules secrete these molecules into the environment. At low cell densities, autoinducer is in low concentration, whereas at high cell densities this substance accumulates in the surrounding environment and passively diffuses back into the bacterial cell to activate transcription of a defined set of genes. In the case of A. tumefaciens, the autoinducer is an N-3-(oxooctonoyl)-L-homoserine lactone termed Agrobacterium autoinducer (AAI). AAI acts in conjunction with TraR to activate transcription of the Ti plasmid tra genes and tral, whose product mediates synthesis of AAI. Therefore, synthesis of TraR under conditions of high cell density creates a positive-feedback loop whereby a TraR–AAI complex induces transcription of TraI, which in turn results in enhanced synthesis of more AAI. It must be noted that this regulatory cascade, involving opinemediated expression of traR and TraR–AAI-mediated expression of Ti plasmid transfer genes under conditions of high cell density, has the net effect of enhancing Ti plasmid transfer in the environment of the plant tumor. Given that the Ti plasmid encodes essential virulence proteins for stimulating T-DNA transfer, A. tumefaciens might have evolved this complex regulatory system to maximize the number of bacterial cells in the vicinity of the plant wound site that are competent for delivery of opine-encoding T-DNA to plant cells.

D. vir genes The Ti plasmid carries an ~35-kb region that harbors at least six operons involved in T-DNA transfer. Two of these operons have a single open reading frame, whereas the remaining operons code for 2–11 open reading frames. The products of the vir region direct events within the bacterium that must precede export of a copy of the T-DNA to plant cells. These events include (i) elaboration of the VirA/VirG sensory transduction system for perception of plant-derived signals and transcriptional activation of the vir genes, (ii) T-DNA processing into a nucleoprotein particle for delivery to plant nuclei by the VirC, VirD, and VirE proteins, and (iii) assembly of a transenvelope transporter composed of VirB proteins for exporting the T-DNA transfer intermediate across the bacterial envelope. 1. vir gene activation Infection is initiated when bacteria sense and respond to an array of signals, including specific classes of

plant phenolic compounds, monosaccharides, and an acidic pH that are present at a plant wound site (Fig. 2.1). Signal perception is mediated by the VirA/VirG signal transduction system together with ChvE, a periplasmic sugar-binding protein, and possibly other phenolic-binding proteins. VirA was one of the first described of what is recognized as a very large family of sensor kinases identified in bacteria and recently in eukaryotic cells. The members of this protein family typically are integral membrane proteins with an N-terminal extracytoplasmic domain. Upon sensory perception, the kinase autophosphorylates at a conserved histidine residue and then transfers the phosphate group to a conserved aspartate residue on the second component of this transduction pathway, the response regulator. The phosphorylated response regulator coordinately activates transcription of several operons, whose products mediate a specific response to the inducing environmental signal. For the A. tumefaciens vir system, the response regulator is VirG, and phosphorylated VirG activates transcription of the six essential vir operons and many other Ti plasmid-encoded operons that are dispensable for virulence. VirA senses all three of the plant-derived signals discussed previously. The most important signal molecules are phenols that carry an ortho-methoxy group. The type of substitution at the para position distinguishes strong inducers such as acetosyringone from weaker inducers such as ferulic acid and acetovanillone. A variety of monosaccharides, including glucose, galactose, arabinose, and the acidic sugars D-galacturonic acid and D-glucuronic acid, strongly enhance vir gene induction. The inducing phenolic compounds and the monosaccharides are secreted intermediates of biosynthetic pathways involved in cell wall repair. Therefore, the presence of these compounds is a general feature of most plant wounds and likely contributes to the extremely broad host range of A. tumefaciens. VirA functions as a homodimer, and recent genetic studies support a model indicating that VirA interacts directly with inducing molecules that diffuse across the outer membrane into the periplasm. Sugar-mediated inducing activity occurs via an interaction between sugars and the periplasmic sugar-binding protein ChvE. In turn, ChvE–sugar interacts with the periplasmic domain of VirA to induce a conformational change that increases the sensitivity of VirA to phenolic inducer molecules. The periplasmic domain of VirA also senses the third environmental signal, acidic pH, required for maximal induction of the vir genes; however, the underlying mechanism responsible for stimulation of VirA activity is unknown. On the basis of recent crystallographic analysis of CheY, a homolog of VirG, phosphorylation of this

ch02.qxd 10/9/03 6:44 PM Page 15

agrobacterium and plant cell transformation family of response regulators is thought to induce a conformational change. Phospho-VirG activates transcription of the vir genes by interacting with a cisacting regulatory sequence (TNCAATTGAAAPy) called the vir box located upstream of each of the vir promoters. Interestingly, both nonphosphorylated and phosphorylated VirG bind to the vir box, indicating that a phosphorylation-dependent conformation is necessary for a productive interaction with components of the transcription machinery.

III. CHROMOSOMALLY ENCODED VIRULENCE GENES Most studies of the A. tumefaciens infection process have focused on the roles of Ti plasmid genes in T-DNA transfer and opine response. Several essential and ancillary chromosomal genes also have been shown to contribute to A. tumefaciens pathogenicity. Although mutations in these genes are often pleiotropic, they generally function to regulate vir gene expression or mediate attachment to plant cells.

A. Regulators of vir gene expression At least three groups of chromosomal genes have been identified that activate or repress vir gene expression. As described previously, the periplasmic sugar-binding protein ChvE complexed with any of a wide variety of monosaccharides induces conformational changes in VirA, allowing it to interact with phenolic inducers. Interestingly, chvE mutants are not only severely compromised for T-DNA transfer but also show defects in chemotaxis toward sugars, suggesting that ChvE interacts both with VirA and with another membrane protein(s) involved in chemotaxis. ChvE therefore plays a dual role in the physiology of A. tumefaciens by promoting chemotaxis toward nutrients and by enhancing the transfer efficiency of opineencoding T-DNA to plant cells. A second locus codes for Ros, a transcriptional repressor of certain vir operons. As described later, the VirC and VirD operons contribute to the T-DNA processing reaction. Although the promoters for these operons are subject to positive regulation by the VirA/VirG transduction system in response to phenolics and sugars, they are also negatively regulated by the Ros repressor. A mutation in ros leads to constitutive expression of virC and virD in the complete absence of VirG protein. Ros binds to a 9-bp inverted repeat, the ros box residing upstream of these promoters. In the absence of plant signals, Ros binding to the virC and virD promoters prevents the T-DNA processing reaction,

15

whereas in the presence of plant signals Ros repression is counteracted by the VirA/VirG induction system. Interestingly, Ros was recently shown to be a novel prokaryotic zinc finger protein that functions to repress not only the expression of T-DNA processing genes in the absence of a suitable plant host but also the expression of the T-DNA oncogenes in the bacterium. A second two-component regulatory system has been identified that, like the VirA/VirG transducer pair, senses environmental signals and mounts a behavioral response by modulating gene expression. ChvG is the sensor kinase and ChvI is the response regulator. Null mutations in genes for these proteins result in cells which cannot induce the vir genes or grow at an acidic pH of 5.5. The molecular basis underlying the effect of the ChvG and ChvI proteins on vir gene expression is unknown.

B. Attachment to plant cells Binding of A. tumefaciens to plant cells is required for T-DNA transfer. Recent evidence indicates there are at least two binding events that may act sequentially or in tandem. The first is encoded by chromosomal loci and occurs even in the absence of the Ti plasmid genes. This binding event directs bacterial binding to many plant cells independently of whether or not the bacterium is competent for exporting T-DNA or the given plant cell is competent for receipt of T-DNA. The second binding event is mediated by a pilus that is elaborated by the virB genes (see Section V.B.1). Binding via the chromosomally encoded attachment loci is a two-step process in which bacteria first attach loosely to the plant cell surface, often in a polar fashion. A series of genes termed att are required for this binding reaction. The second step involves a transition resulting in the tight binding of the bacteria to plant cells. The cel genes that mediate this form of binding direct the synthesis of cellulose fibrils that emanate from the bacterial cell surface. Recent studies indicate that binding due to these chromosomal functions occurs at specific sites on the plant cell surface. Binding is saturable, suggestive of a limited number of attachment sites on the plant cell, and binding of virulent strains can also be prevented by attachment of avirulent strains. Although the identity of a plant cell receptor(s) has not been definitively established, a good candidate is a vitronectin-like protein found in detergent extracts of plant cell walls. Attachment-proficient A. tumefaciens cells bind radioactive vitronection, whereas attachment-deficient cells do not bind this molecule. Intriguingly, human vitronectin and antivitronectin antibodies both inhibit the binding of A. tumefaciens to plant cells.

ch02.qxd 10/9/03 6:44 PM Page 16

16

the desk encyclopedia of microbiology

Efficient attachment of bacteria to plant cells also requires the products of three chromosomal loci: chvA, chvB, and exoC (pscA). All three loci are involved in the synthesis of transport of a cyclic -1,2 glucan molecule. Mutations in these genes are pleiotropic, suggesting that -1,2 glucan synthesis is important for the overall physiology of A. tumefaciens. Periplasmic -1,2 glucan plays a role in equalizing the osmotic pressure between the inside and outside of the cell. It has been proposed that loss of this form of glucan may indirectly disrupt virulence by reducing the activity or function of cell surface proteins. Interestingly, chv mutants accumulate low levels of VirB10, one of the proposed components of the T-complex transport system (see Section V), suggesting that -1,2 glucan might influence T-DNA export across the bacterial envelope by contributing to transporter assembly.

IV. T-DNA PROCESSING One of the early events following attachment to plant cells and activation of vir gene expression in response to plant signals involves the processing of T-DNA into a form which is competent for transfer across the bacterial cell envelope and translocation through the plant plasma membrane, cytosol, and nuclear membrane. The prevailing view, strongly supported by molecular data, is that T-DNA is transferred as a single-stranded molecule that is associated both covalently and noncovalently with Vir proteins. Two proteins identified to date are components of the transfer intermediate: VirD2, an endonuclease that participates in the T-DNA processing reaction, and VirE2, a single-stranded DNA-binding protein which is proposed to associate noncovalently along the length of the singlestranded transfer intermediate (Fig. 2.1). Intriguingly, recent studies have provided strong evidence that A. tumefaciens can export the VirE2 SSB to plant cells independently of T-DNA (see Section IV.B).

A. Formation of the transfer intermediate More than a decade ago, investigators determined that the T-DNA border repeats are cleaved by a strand-specific endonuclease and that the right T-DNA border sequence is essential for and determines the direction of DNA transfer from A. tumefaciens to plant cells. The predominant product of this nicking reaction was shown to be a free single-stranded T-DNA molecule that corresponds to one strand of T-DNA. It was noted that these features of the T-DNA processing reaction are reminiscent of early processing events involved in the conjugative transfer of plasmids

between bacterial cells. In the past 10 years, a large body of evidence has accumulated supporting the notion that DNA processing reactions associated with T-DNA transfer and bacterial conjugation are equivalent. Extensive studies have shown that two systems in particular, the T-DNA transfer system and the conjugation system of the broad host-range plasmid RP4, are highly similar. The substrates for the nicking enzymes of both systems, T-DNA border sequences and the RP4 origin of transfer (oriT), exhibit a high degree of sequence similarity. Furthermore, the nicking enzymes VirD2 of pTi and TraI of RP4 possess conserved active-site motifs that are located within the N-terminal halves of these proteins. Purified forms of both proteins cleave at the nick sites within T-DNA borders and the RP4 oriT, respectively. In the presence of Mg2, purified VirD2 will catalyze cleavage of oligonucleotides bearing a T-DNA nick site. However, VirD1 is essential for nicking when the nick site is present on a supercoiled, double-stranded plasmid. Both VirD2 and TraI remain covalently bound to the 5 phosphoryl end of the nicked DNA via conserved tyrosine residues Tyr-29 and Tyr-22. Finally, both proteins catalyze a joining activity reminiscent of type I topoisomerases. VirD1 was reported to possess a topoisomerase I activity, but recent work suggests instead that VirD1 supplies a function analogous to TraJ of RP4, which is thought to interact with oriT as a prerequisite for TraI binding to an oriT DNA–protein complex. The current model describing the T-DNA and plasmid conjugation processing reactions is that sequence and strand-specific endonucleases initiate processing by cleaving at T-DNA borders and oriT sequences, respectively. This reaction is followed by a strand displacement reaction, which generates a free singlestranded transfer intermediate. Concomitantly, the remaining segment of T-DNA or plasmid serves as a template for replacement synthesis of the displaced strand. It is important to note that the single-stranded transfer intermediates of the T-DNA and RP4 transfer systems remain covalently bound to their cognate endonucleases. Considerable evidence suggests that these protein components play essential roles in delivering the respective transfer intermediates across the bacterial envelope.

B. The role of VirE2 SSB in T-DNA transfer The virE2 gene codes for a single-stranded DNAbinding protein that binds cooperatively to singlestranded DNA (ssDNA). Early studies supplied evidence that VirE2 binds with high affinity to any

ch02.qxd 10/9/03 6:44 PM Page 17

agrobacterium and plant cell transformation ssDNA in vitro and that it binds T-DNA in A. tumefaciens. By analogy to other ssDNA-binding proteins (SSBs) that play important roles in DNA replication, VirE2 was proposed to participate in the T-DNA processing reaction by binding to the liberated T-strand and preventing it from reannealing to its complementary strand on the Ti plasmid. The translocation-competent form of DNA therefore has been depicted as a ssDNA molecule covalently bound at the 5 end by VirD2 and coated along its length with an SSB. The singlestranded form of T-DNA delivered to plants is termed the T-strand, and the VirD2–VirE2-T-strand nucleoprotein particle is termed the T-complex (Fig. 2.1). Considerable evidence indicates that the T-complex represents the biologically active transfer intermediate. The T-complex, composed of a 20-kb T-strand capped at its 5 end with a 60-kDa endonuclease and approximately 600 VirE2 molecules along its length, is a large nucleoprotein complex of an estimated size of 50 106 Da. This size approaches that of some bacteriophages, and it has been questioned whether such a complex could be exported intact across the A. tumefaciens envelope without lysing the bacterial cell. Although this is still unknown, several recent discoveries support an alternative model that assembly of the T-complex initiates within the bacterium but is completed within the plant cell. Approximately 15 years ago, it was discovered that two avirulent A. tumefaciens mutants, one with a deletion of T-DNA and a second with a virE2 mutation, could induce the formation of tumors when inoculated as a mixture on plant wound sites. To explain this observation, it was postulated that A. tumefaciens separately exports VirE2 and VirD2 T-strands to the same plant cell. The virE2 mutant was proposed to export the VirD2 T-strands (T-DNA donor), and the T-DNA deletion mutant could export the VirE2 protein only (VirE2 donor). Once exported, these molecules could then assemble into a nucleoprotein particle, the T-complex, for transmission to the plant nucleus. In strong support of this model, recent genetic analyses have shown that both the proposed T-DNA donor strain and the VirE2 mutant in the mixed infection experiment must possess an intact transport machinery and intact genes mediating bacterial attachment to the plant cell. Furthermore, current genetic data argue against the possible movement of T-DNA or VirE2 between bacterial cells by conjugation as an alternative explanation for complementation by mixed infection. Finally, a virE mutant was shown to incite the formation of wild-type tumors on transgenic plants expressing virE2. This finding indicates that VirE2 participates in A. tumefaciens pathogenesis by supplying essential functions within the plant.

17

C. Role of cotransported proteins in T-DNA transfer and plasmid conjugation As discussed previously, processing of T-DNA and conjugative plasmids results in the formation of a ssDNA transfer intermediate covalently bound at its 5 end to the nicking enzyme. Recent studies have shown that the protein component(s) of these conjugal transfer intermediates participates in the delivery of the DNA to the recipient cell. In the case of T-DNA, the transferred proteins facilitate movement of the T-DNA transfer intermediate to plant nuclei by (i) piloting the T-DNA transfer intermediate across the bacterial envelope and protecting it from nucleases and/or (ii) directing T-DNA movement and integration in plant cells. In the case of the IncP plasmid RP4, TraI relaxase is thought to promote plasmid recircularization, and a primase activity associated with the TraC SSB is considered to be important for second-strand synthesis in the recipient cell. 1. Piloting and protection A piloting function for VirD2 is suggested by the fact that VirD2 is covalently associated at the 5 end of the T-strand and also from the finding that the T-strand is transferred to the plant cell in a 5–3 unidirectional manner. A dedicated transporter functions to export substrates to plant cells (see Section V). VirD2 might guide T-DNA export by providing the molecular basis for recognition of the transfer intermediate by the transport machinery. By analogy to other protein substrates exported across the bacterial envelope by dedicated transport machines, VirD2 might have a linear peptide sequence or a protein motif in its tertiary structure that marks this molecule as a substrate for the T-DNA transporter. Studies of T-DNA integrity in transformed plant cells have shown that the 5 end of the transferred molecule generally is intact, suffering little or no loss of nucleotides as a result of exonuclease attack during transit. By contrast, the 3 end of the transferred molecule typically is often extensively deleted. These findings suggest that a second role of the VirD2 endonuclease is to protect the 5 end of the transfer intermediate from nucleases. Recent molecular studies have also shown that T-DNA transferred to plant cells by an A. tumefaciens virE2 mutant is even more extensively degraded than T-DNA transferred by wild-type cells, suggesting that VirE2 SSB also functions to protect the DNA transfer intermediate from nucleases during transfer. 2. T-DNA movement and integration DNA sequence analyses revealed the presence of a bipartite type of nuclear localization sequence (NLS)

ch02.qxd 10/9/03 6:44 PM Page 18

18

the desk encyclopedia of microbiology

near the C terminus of VirD2. The nuclear localizing function of this NLS was confirmed by fusing the virD2 coding sequence to a reporter gene and demonstrating the nuclear localization of the reporter protein activity in tobacco cells transiently expressing the gene fusion. As predicted, A. tumefaciens strains expressing mutant forms of VirD2 with defects in the NLS sequence are very inefficient in delivering T-DNA to plant nuclei. Similar lines of investigation showed that VirE2 also possesses two NLS sequences that both contribute to its delivery to the nuclear pore. Therefore, both VirD2 and VirE2 are proposed to promote T-DNA delivery to and across the plant nuclear membrane. In this context, VirD2 has been shown to interact with a plant NLS receptor localized at the nuclear pore. Of further interest, VirD2 has also been shown to interact with several members of a family of proteins termed cyclophilins. The postulated role for cyclophilins in this interaction is to supply a chaperone function at some stage during T-complex trafficking to the nucleus. Agrobacterium tumefaciens has been demonstrated to transport DNA to representatives of prokaryotes, yeasts, and plants. Cyclophilins are ubiquitous proteins found in all these cell types and therefore may be of general importance for A. tumefaciens-mediated DNA transfer. T-DNA integrates into the plant nuclear genome by a process termed “illegitimate” recombination. According to this model, T-DNA invades at nicks or gaps in the plant genome possibly generated as a consequence of active DNA replication. The invading ends of the single-stranded T-DNA are proposed to anneal via short regions of homology to the unnicked strand of the plant DNA. Once the ends of T-DNA are ligated to the target ends of plant DNA, the second strand of the T-DNA is replicated and annealed to the opposite strand of the plant DNA. Recent mutational analysis of VirD2 showed that a C-terminal sequence termed appears to play a role in promoting T-DNA integration. A recent study also supports a model that VirE2 also participates in the T-DNA integration step, but the precise functions of VirD2, VirE2, and possible host proteins in this reaction have not been defined.

V. THE T-DNA TRANSPORT SYSTEM A. The essential components of the T-complex transporter Exciting progress has been made during the past 6 years on defining the structure and function of the transporter at the A. tumefaciens cell surface that is dedicated to exporting the T-DNA transfer intermediate to plant cells.

Early genetic studies suggested that products of the ~9.5-kb virB operon are the most likely candidates for assembling into a cell surface structure for translocation of T-DNA across the A. tumefaciens envelope. Sequence analyses of the virB operon have supported this prediction by showing that the deduced products have hydropathy patterns characteristic of membraneassociated proteins. Recently, a systematic approach was taken to delete each of the 11 virB genes from the virB operon without altering expression of the downstream genes. Analyses of this set of nonpolar null mutants showed that virB2–virB11 are essential for T-DNA transfer, whereas virB1 is dispensable. As described in more detail later, the VirB proteins, along with the VirD4 protein, are thought to assemble at the cell envelope as a channel dedicated to the export of T-complexes.

B. The T-complex transporter 1. Type IV transporters: DNA conjugation systems adapted for export of virulence factors DNA sequence studies within the past 4 years have identified extensive similarities between products of the virB genes and components of two types of transporters dedicated to movement of macromolecules from or between cells (Fig. 2.4). The first type, encoded by tra operons of conjugative plasmids, functions to deliver conjugative plasmids to bacterial recipient cells. The IncN plasmid, pKM101, and the IncW plasmid, R388, code for Tra protein homologs of each of the VirB proteins. Furthermore, the genes coding for related proteins are often colinear in these respective virB and tra operons, supporting the view that these DNA transfer systems share a common ancestral origin. Other broad host-range plasmids such as RP4 (IncP) and the narrow host-range plasmid F (IncF) code for proteins homologous to a subset of the VirB proteins. DNA sequence studies also identified a related group of transporters in several bacterial pathogens of humans that function not to export DNA but rather to secrete protein toxins (Fig. 2.4). Bordetella pertussis, the causative agent of whooping cough, uses the Ptl transporter to export the six-subunit pertussis toxin across the bacterial envelope. All nine Ptl proteins have been shown to be related to VirB proteins, and the ptl genes and the corresponding virB genes are colinear in their respective operons. Type I strains of Helicobacter pylori, the causative agent of peptic ulcer disease and a risk factor for development of gastric adenocarcinoma, contain a 40-kb cag pathogenicity island (PAI) that codes for several virulence factors, of which several are related to Vir proteins. These Cag proteins are thought to assemble into a transporter for

ch02.qxd 10/9/03 6:44 PM Page 19

agrobacterium and plant cell transformation

19

FIGURE 2.4 Alignment of genes encoding related components of the type IV transport systems. Of the 11 VirB proteins, those encoded by virB2–virB11, as well as virD4, are essential for T-complex transport to plant cells. The broad host-range (BHR) plasmid pKM101 encodes a conjugation apparatus composed of the products of the tra genes shown. Other BHR plasmids and the narrow host-range (NHR) F plasmid code for Tra proteins related to most or all the VirB genes. A second subfamily of type IV transporters found in bacterial pathogens of humans export toxins or other protein effectors to human cells.

exporting an unidentified protein toxin(s) that induces synthesis of the proinflammatory cytokine IL-8 in gastric epithelial cells. Finally, Legionella pneumophila, the causative agent of Legionnaire’s disease and Pontiac fever, possesses the icm/dot genes, of which dotG and dotB code for proteins related to VirB10 and VirB11 and others code for homologs of transfer proteins encoded by other bacterial conjugation systems. The Icm/Dot proteins are proposed to assemble into a transporter that exports a virulence factor(s) that promotes intracellular survival of L. pneumophila and macrophage killing. The transporters described previously are grouped on the basis of evolutionary relatedness as a distinct transport family. Designated as the type IV secretion family, this classification distinguishes these transporters from other conserved bacterial protein targeting mechanisms that have been identified in bacteria. Although this is a functionally diverse family, the unifying theme of the type IV transporters is that each system has evolved by adapting an ancestral DNA conjugation apparatus or a part of this apparatus for the novel purpose of exporting DNA or proteins that function as virulence factors. 2. Functional similarities among type IV transporters Functional studies have supplied compelling evidence that the type IV transporters are mechanistically related. The non-self-transmissible plasmid RSF1010

of the IncQ incompatibility group possesses an oriT sequence and mobilization (mob) functions for generating a ssDNA transfer intermediate. This transfer intermediate can be delivered to recipient bacteria by the type IV transporters of the IncN, IncW, IncP, and F plasmids. In addition, approximately 10 years ago it was shown using an A. tumefaciens strain harboring a disarmed Ti plasmid (with vir genes but lacking the T-DNA or its borders) and an RSF1010 derivative that the T-complex transporter could deliver the IncQ transfer intermediate to plant cells. This discovery was followed soon afterwards by the demonstration that the T-complex transporter also functions to conjugally deliver the IncQ plasmid to A. tumefaciens recipient cells. Interestingly, A. tumefaciens strains carrying both an IncQ plasmid and an intact T-DNA efficiently deliver the IncQ plasmid to plant cells but do not transfer the T-DNA. Preferential transfer of the IncQ plasmid over the T-DNA transfer intermediate could result from the transporter having a higher affinity for the IncQ plasmid or the IncQ plasmid being more abundant than the T-DNA. Of further interest, the coordinate overexpression of virB9, virB10, and virB11 relieved the IncQ suppression and restored efficient T-DNA transfer to plant cells. These findings suggest that the T-complex and the IncQ transfer intermediate compete for the same transport apparatus. Furthermore, the data suggest that VirB9–VirB11 stoichiometries determine the number of transporters a given cell can assemble or influence the selection of substrates destined for export.

ch02.qxd 10/9/03 6:44 PM Page 20

20

the desk encyclopedia of microbiology

Although the toxin substrates have not been identified for the H. pylori Cag and L. pneumophila Dot/Icm transporters, it is intriguing to note that the Dot/Icm system also has been shown to deliver the non-selftransmissible IncQ plasmid RSF1010 to bacterial recipient cells by a process requiring cell-to-cell contact. Also, as observed for T-complex export, the presence of an IncQ plasmid suppresses export of the natural substrate of the Dot/Icm transporter of L. pneumophila, resulting in inhibition of intracellular multiplication and human macrophage killing. These parallel findings show that the type IV DNA and protein export systems are highly mechanistically related.

TABLE 2.1 Properties of the VirB proteins VirB Localization

Proposed function

B1

Periplasm

Transglycosylase

B1*

Cell exterior

Cell contact/pilin subunit?

B2

Exported/cell exterior Cell contact/pilin subunit

B3

Exported

B4

Transmembrane

ATPase/transport activation

B5

Exported

Cell contact/pilin subunit?

B6

Transmembrane

Candidate pore former

B7

Outer membrane

Lipoprotein/transporter assembly

B8

Periplasmic face of inner membrane

Unknown

C. Architecture of the T-complex transporter

B9

Outer membrane

Lipoprotein/transporter assembly

The T-complex transporter, like other DNA conjugation machines, is proposed to be configured as a transenvelope channel through which the T-DNA transfer intermediate passes and as an extracellular pilus termed the T-pilus for making contact with recipient cells. Most of the VirB proteins fractionate with both membranes, consistent with the view that these proteins assemble as a membrane-spanning protein channel. All the VirB proteins except VirB11 possess periplasmic domains, as shown by protease susceptibility and reporter protein fusion experiments. Although detailed structural information is not available for the T-complex transporter, important progress has been made in the characterization of the VirB proteins, especially in the following areas: (i) characterization of the virB-encoded pilus termed the T-pilus, (ii) structure–function studies of the VirB4 and VirB11 ATPases, and (iii) identification of a nucleation activity of a disulfide cross-linked VirB7/VirB9 heterodimer during transporter assembly (Table 2.1).

B10

Transmembrane

Coupler of inner and outer membrane subcomplexes?

B11

Cytoplasm/ inner membrane

ATPase/transport activation

D4

Transmembrane

ATPase/coupler of DNA processing and transport systems

1. The T-pilus The type IV systems involved in conjugation elaborate pili for establishing contact between plasmidbearing donor cells and recipient cells. Recent studies have demonstrated that VirB proteins direct the assembly of a pilus which is essential for T-DNA transfer. Electron microscopy studies have demonstrated the presence of long filaments (~10 nm in diameter) on the surfaces of A. tumefaciens cells induced for expression of the virB genes. These filaments are absent from the surfaces of mutant strains defective in the expression of one or more of the virB genes. Furthermore, an interesting observation was made that cells grown at room temperature rarely possess pili, whereas cells grown at ~19 C possess these structures in abundance. This finding correlates well with previous findings that low temperature

Unknown

stimulates the virB-dependent transfer of IncQ plasmids to bacterial recipients and T-DNA transfer to plants. Recently, compelling evidence demonstrated that VirB2 is the major pilin subunit. Early studies showed that VirB2 bears both sequence and structural similarity to the TraA pilin subunit of the F plasmid of E. coli. Recent work demonstrated that VirB2, like TraA, is processed from an ~12-kDa propilin to a 7.2-kDa mature protein that accumulates in the inner membrane. During F plasmid conjugation, TraA is mobilized to the surface of the donor cell where it polymerizes to form the pilus. Similarly, the appearance of pili on the surface of A. tumefaciens cells induced for expression of the vir genes is correlated with the presence of VirB2 on the cell exterior. Finally, VirB2 is a major component of pili that have been sheared from the cell surface and purified. Many adhesive and conjugative pili possess one or more minor pilin subunits in addition to the major pilin structural protein. Interestingly, VirB1, a periplasmic protein with transglycosylase activity, is processed such that the C-terminal two-thirds of the protein, termed VirB1*, is secreted to the outer surface of the cell. This localization is consistent with a proposed function for VirB1* as a minor pilus subunit. VirB5 might also assemble as a pilus subunit based on its homology to a possible pilin subunit encoded by the IncN plasmid pKM101 transfer system. 2. Studies of the VirB ATPases Two VirB proteins, VirB4 and VirB11, possess conserved mononucleotide-binding motifs. Mutational

ch02.qxd 10/9/03 6:44 PM Page 21

agrobacterium and plant cell transformation analyses established the importance of these motifs for the function of both proteins. In addition, purified forms of both proteins exhibit weak ATPase activities, suggesting that VirB4 and VirB11 couple the energy of ATP hydrolysis to transport. Both of these putative ATPases appear to contribute functions of general importance for macromolecular transport since homologs have been identified among many DNA and protein transport systems. Of further possible significance, VirB11 and two homologs, TrbB of IncP RP4 and EpsE, of Vibrio cholerae have been reported to autophosphorylate. VirB4 and VirB11 might activate substrate transport by using the energy of ATP hydrolysis or a kinase activity to facilitate assembly of the transport apparatus at the cell envelope. Alternatively, by analogy to the SecA ATPase of E. coli which uses the energy of ATP hydrolysis to drive translocation of exported proteins, one or both of the VirB ATPases may contribute directly to export of the DNA transfer intermediate. Recent studies have shown that both VirB4 and VirB11 assemble as homodimers. Dimerization is postulated to be critical both for protein stability and for catalytic activity. Accumulation of these ATPases to wild-type levels depends on the presence of other VirB proteins, suggesting that complex formation with other components of the T-complex transporter contributes to protein stability. Specific contacts between these ATPases and other transporter components have not been identified. 3. The VirB7 lipoprotein and formation of stabilizing intermolecular disulfide bridges Detailed studies have shown that VirB7 is critical for assembly of a functional T-complex transport system. VirB7 possesses a characteristic signal sequence that ends with a consensus peptidase II cleavage site characteristic of bacterial lipoproteins. Biochemical studies have confirmed that VirB7 is processed as a lipoprotein. Furthermore, maturation of VirB7 as a lipoprotein is critical for its proposed role in T-complex transporter biogenesis. Recent studies have shown that the VirB7 lipoprotein interacts directly with the outer membrane protein VirB9. The first hint of a possible interaction between these proteins was provided by the demonstration that VirB9 accumulation is strongly dependent on co-synthesis of VirB7, suggesting that VirB7 stabilizes VirB9. Interestingly, this stabilizing effect has been shown to be mediated by formation of a disulfide bridge between these two proteins. VirB7 assembles not only as VirB7/VirB9 heterodimers but also as covalently cross-linked homodimers, and there is evidence that VirB9 assembles into higher order multimeric complexes. These dimers and

21

higher order multimers might correspond to stable subcomplexes of the larger transport system. In the case of the VirB7/VirB9 heterodimer, considerable evidence indicates that this heterodimer plays a critical role early during transporter biogenesis by recruiting and stabilizing newly synthesized VirB proteins. The heterodimer has been shown to interact with VirB1*. The heterodimer also interacts with VirB10, a cytoplasmic membrane protein with a large C-terminal periplasmic domain. VirB10 has been postulated to join the VirB7/VirB9 heterodimer at the outer membrane with a VirB protein subcomplex located at the inner membrane. 4. VirB protein stimulation of IncQ plasmid uptake by bacterial recipient cells The T-complex transport system seems designed to function unidirectionally to export substrates to recipient cells. However, a recent discovery indicates that VirB proteins can also assemble as a transenvelope structure that stimulates DNA uptake during conjugation. The fundamental observation is that A. tumefaciens cells harboring an IncQ plasmid conjugally transfer the IncQ plasmid to recipient cells expressing the virB genes at a frequency of ~1000 times that observed for transfer to recipient cells lacking the virB genes. Furthermore, only a subset of virB genes, including virB3, virB4, and virB7–virB10, was required for enhanced DNA uptake by recipient cells. These findings suggest that a subset of the VirB proteins might assemble as a core translocation channel at the bacterial envelope that accommodates the bidirectional transfer of DNA substrates. Such a channel might correspond to an early assembly intermediate that, upon complex formation with additional VirB proteins, is converted to a dedicated T-complex export system.

VI. AGROBACTERIUM HOST RANGE One of the most appealing features of the A. tumefaciens DNA transfer system for genetic engineering is its extremely broad host range. Pathogenic strains of Agrobacterium infect a wide range of gymnosperms and dicotyledonous plant species of agricultural importance. Crown gall disease can cause devastating reductions in yields of woody crops such as apples, peaches, and pears and vine crops such as grapes. Various host range determinants present in different A. tumefaciens strains determine whether a given bacterial strain is virulent for a given plant species.

ch02.qxd 10/9/03 6:44 PM Page 22

22

the desk encyclopedia of microbiology

A. Transformation of monocots In the past 5 years, dramatic progress has been made toward the development of protocols for stably transforming agriculturally important monocotyledonous plant species. The first indication of gene transfer involved the introduction of a plant viral genome into a plant host via A. tumefaciens-mediated transfer of TDNA carrying the viral genome. Once inside the plant host, the viral DNA excises from the T-DNA and infects the host, inciting disease symptoms that are characteristic of the virus. This process, termed agroinfection, supplied compelling evidence that A. tumefaciens transfers T-DNA to monocot plants such as maize. A notable feature of agroinfection is that the introduced viral DNA incites disease without incorporating into the plant genome. Early efforts to obtain stable transformation of monocot species were unsuccessful. The demonstration of agroinfection and the inability to demonstrate T-DNA integration together led to the suggestion that the T-DNA integration step was somehow blocked in monocots. However, protocols have been developed for the efficient and reproducible stable transformation of rice, corn, wheat, and other monocot species. Key to the success of these protocols was the use of actively dividing cells such as immature embryos. In addition, preinduction of A. tumefaciens with phenolic inducers appears to enhance T-DNA transfer efficiencies. Additional factors, such as plant genotype, the type and age of plant tissue, the kinds of vectors and bacterial strains, and the types of selectable genes delivered to plant cells, influence the transformation efficiencies. For rice and corn, most of these parameters have been optimized, so that the delivery of foreign DNA to these crop plants is a routine technique.

shown to efficiently deliver DNA to fungal protoplasts and fungal conidia and hyphal tissue. This discovery extends well beyond academic interest because the simplicity and high efficiency make this gene delivery system an extremely useful tool for the genetic manipulation and characterization of fungi. This DNA transfer system is especially valuable for species such as the mushroom Agaricus bisporus which are recalcitrant to transformation by other methods. It is also of interest to consider that both A. tumefaciens and many fungal species exist in the same soil environment, raising the possibility that A. tumefaciens-mediated gene transfer to fungi may not be restricted solely to the laboratory bench.

VII. GENETIC ENGINEERING OF PLANTS AND OTHER ORGANISMS The extent to which any biological system is understood is reflected by our ability to manipulate that system to achieve novel ends. For A. tumefaciens transformation, the holy grail has been monocot transformation. As described previously, exciting progress has been made toward attaining this objective for several agriculturally important monocot species. Currently, plant genetic engineers are developing the A. tumefaciens gene delivery to achieve equally challenging goals such as (i) designing T-DNA tagging methods for isolating and characterizing novel plant genes, (ii) designing strategies to deliver foreign DNA to specific sites in the plant genome, and (iii) characterizing and genetically engineering other organisms such as agriculturally or medically important fungi.

A. Overcoming barriers to transformation B. Gene transfer to yeast and fungi Intriguing recent work has extended the host range of A. tumefaciens beyond the plant kingdom to include budding and fission yeast and many species of filamentous fungi. The successful transfer of DNA to yeast depends on the presence of stabilizing sequences, such as a yeast origin of replication sequence or a telomere, or regions of homology between the transferred DNA and the yeast genome for integration by homologous recombination. When the T-DNA lacks any extensive regions of homology with the Saccharomyces cerevisiae genome, it integrates at random positions by illegitimate recombination reminiscent of T-DNA integration in plants (see Section IV.C.2). The transformation of filamentous fungi with A. tumefaciens is an exciting advancement. Agrobacterium tumefaciens was

It is remarkable that all progress in A. tumefaciensmediated monocot transformation has been achieved in the intervening period between the publication of the first and second editions of this encyclopedia. In fact, currently A. tumefaciens is the biological DNA delivery system of choice for transformation of most dicot and monocot plant species. The reasons are twofold. First, A. tumefaciens is readily manipulated such that plasmids carrying foreign genes of interest are easily introduced into appropriate bacterial strains for delivery to plants. Typically, strains used for gene delivery are “disarmed,” that is, deleted of oncogenic T-DNA but still harboring intact Ti plasmid and chromosomal vir genes. Foreign genes destined for delivery to plants generally are cloned onto a plasmid that carries a single T-DNA border sequence or

ch02.qxd 10/9/03 6:44 PM Page 23

agrobacterium and plant cell transformation two T-DNA border sequences that flank various restriction sites for cloning as well as an antibiotic resistance gene to select for transformed plant cells. If the plasmid carries a single border sequence, the entire plasmid is delivered to plants, and recent work indicates that A. tumefaciens can deliver as much as 180-kb of DNA to plants. If the plasmid carries two border sequences, only the DNA bounded by T-DNA borders is delivered to plants. Second, the frequency of stable transformation is often very high, far exceeding frequencies achieved by other gene delivery methods. For example, co-cultivation of A. tumefaciens with regenerating protoplasts of certain plant species can result in transformation of up to one-half of the protoplasts. However, with protoplast transformation there is often a significant reduction in the number of transgenic, fertile plants recovered during selective regeneration of transformed protoplasts. For certain species, protoplasts can be transformed but are recalcitrant to regeneration into intact plants. Consequently, other transformation methods have relied on transformation of plant tissues such as excised leaves or root sections. In the case of monocot species such as maize, immature embryos are the preferred starting material for A. tumefaciens-mediated DNA transfer. For rice, success has been achieved with callus tissue induced from immature embryos. In addition to the need to identify transformable and regenerable plant tissues, many varieties of a given species often need to be screened to identify the susceptible varieties. A large variation in transformation efficiencies is often observed depending on which cell line is being tested. This underscores the notion that interkingdom DNA transfer is a complex process dependent on a genetic interplay between A. tumefaciens and host cells. Fortunately, many of the agonomically important species are readily transformable, but additional efforts are needed to overcome the current obstacles impeding efficient transformation of other species of interest.

B. Other applications Agrobacterium tumefaciens is increasingly used to characterize and isolate novel plant genes by an approach termed T-DNA tagging. Several variations to this methodology exist depending on the desired goals. For example, because insertions are generally randomly distributed throughout the plant genome, T-DNA is widely used as a mutagen for isolating plant genes with novel phenotypes. If the mutagenic T-DNA carries a bacterial origin of replication, the mutated gene of interest can easily be recovered in

23

bacteria by suitable molecular techniques. Furthermore, if the T-DNA is engineered to carry a selectable or scorable gene near one of its ends, insertion downstream of a plant promoter will permit characterization of promoter activity. Conversely, if the T-DNA is engineered to carry an outward reading promoter, insertion can result in a modulation of gene expression with potentially interesting phenotypic consequences. Finally, the discovery that A. tumefaciens can transform fungal species of interest means that all approaches developed for plants can be applied to the characterization of fungi. Although random T-DNA insertion is a boon to investigators interested in characterizing plant or fungal genes, it is an undesired event for plant genetic engineering. In addition to the potential result that T-DNA will insert into an essential gene, insertion often is accompanied by rearrangements of flanking sequences, thus further increasing the chances that the insertion will have undesired consequences. Ideally, TDNA could be delivered to a restricted number of sites in the plant genome. Recent progress toward this goal has involved the use of the bacteriophage P1 Cre/lox system for site-specific integration in the plant genome. The Cre site-specific recombinase catalyzes strand exchange between two lox sites which, for P1, results in circularization of the P1 genome upon infection of bacterial cells. For directed T-DNA insertion, both the plant and the T-DNA are engineered to carry lox sequences and the plant is also engineered to express the Cre protein. Upon entry of T-DNA into the plant cell, Cre was shown to catalyze the site-specific integration of T-DNA at the plant lox site. The frequency of directed insertion events is low compared to random insertion events, but additional manipulation of this system should enhance its general applicability.

VIII. CONCLUSIONS The early discovery that oncogenes can be excised from T-DNA and replaced with genes of interest paved the way for the fast growing industry of plant genetic engineering. Currently, much information has been assembled on the A. tumefaciens infection process. This information has been used to successfully manipulate the T-DNA transfer system both to enhance its efficiency and to broaden the range of transformable plants and other organisms. Furthermore, it must be noted that this information has also often established a conceptual framework for initiating or extending the characterization of other pathogenic and symbiotic relationships. The discovery that secreted chemical signals initiate a complex

ch02.qxd 10/9/03 6:44 PM Page 24

24

the desk encyclopedia of microbiology

dialogue between A. tumefaciens and plant cells as well as other A. tumefaciens cells has stimulated a global effort to identify extracellular signals and characterize the cognate signal transduction systems in many bacterial systems. The discovery of T-DNA transport provided a mechanistic explanation for how horizontal gene transfer impacts the evolution of genomes of higher organisms. This discovery also established a precedent for interkingdom transport of virulence factors by bacterial pathogens. Indeed, in only the past 6 years, studies have revealed that numerous pathogens employ interkingdom transport to deliver a wide array of effector proteins to plant and animal hosts. These so-called type III transport systems, like the A. tumefaciens T-complex transporter and related type IV transporters, deliver substrates via a process dependent on cell-to-cell contact and, in some cases, elaboration of an extracellular filament or pilus. It is clear that, in the future, studies of all the various aspects of the A. tumefaciens infection process will continue to spawn new applications for this novel DNA transfer system and yield new insights about the evolution and function of pathogenic mechanisms that are broadly distributed in nature.

ACKNOWLEDGMENTS I thank members of my laboratory for helpful and stimulating discussions. Studies in this laboratory are funded by the National Institutes of Health.

BIBLIOGRAPHY Binns, A. N., and Howitz, V. R. (1994). The genetic and chemical basis of recognition in the Agrobacterium: plant interaction. Curr. Topics Microbiol. Immunol. 192, 119–138. Binns, A. N., Joerger, R. D., and Ward, J. E., Jr. (1992). Agrobacterium and plant cell transformation. In “Encyclopedia of Microbiology” (J. Lederberg, ed.), pp. 37–51. Academic Press, San Diego, CA. Christie, P. J. (1997). The Agrobacterium tumefaciens T-complex transport apparatus: A paradigm for a new family of multifunctional transporters in eubacteria. J. Bacteriol. 179, 3085–3094. Christie, P. J., and Covacci, A. (1998). Bacterial type IV secretion systems: Systems utilizing components of DNA conjugation machines for export of virulence factors. In “Cellular Microbiology” (P. Cossart, P. Boquet, S. Normark, and R. Rappuoli, eds.). ASM Press, Washington, DC. Christie, P. J., and Vogel, J. P. (2000). Bacterial type IV secretion: conjugation systems adapted to deliver effector molecules to host cells. Trends Microbiol. 8, 354–360.

Citovsky, V., and Zambryski, P. (1993). Transport of nucleic acids through membrane channels: Snaking through small holes. Annu. Rev. Microbiol. 47, 167–197. Das, A. (1998). DNA transfer from Agrobacterium to plant cells in crown gall tumor disease. Subcell. Biochem. 29, 343–363. Fernandez, D., Spudich, G. M., Dang, T. A., Zhou, X.-R., Rashkova, S., and Christie, P. J. (1996). Biogenesis of the Agrobacterium tumefaciens T-complex transport apparatus. In “Biology of Plant– Microbe Interactions” (G. Stacey, B. Mullin, and P. Gresshof, eds.), pp. 121–126. ISMPMI, St. Paul, MN. Firth, N., Ippen-Ihler, K., and Skurray, R. A. (1996). Structure and function of the F factor and mechanism of conjugation. In “Escherichia coli and Salmonella typhimurium,” 2nd ed., pp. 2377–2401. American Society for Microbiology, Washington, DC. Fuqua, W. C., Winans, S. C., and Greenberg, E. P. (1996). Census and consensus in bacterial ecosystems: The LuxR–LuxI family of quorum-sensing transcriptional regulators. Annu. Rev. Microbiol. 50, 727–751. Lai, E. M., and Kado, C. I. (2000). The T-pilus of Agrobacterium tumefaciens. Trends Microbiol. 8, 361–369. Moriguchi, K., Maeda, Y., Satou, M., Hardayani, N. S., Kataoka, M., Tanaka, N., and Yoshida, K. (2001). The complete nucleotide sequence of a plant root-inducing (Ri) plasmid indicates its chimeric structure and evolutionary relationship between tumor-inducing (Ti) and symbiotic (Sym) plasmids in Rhizobiaceae. J. Mol. Biol. 307, 771–784. Nester, E. W., Kemner, J., Deng, W., Lee, Y.-W., Fullner, K., Liang, X., Pan, S., and Heath, J. D. (1996). Agrobacterium: A natural genetic engineer exploited for plant biotechnology. In “Biology of Plant–Microbe Interactions” (G. Stacey, B. Mullin, and P. Gresshof, eds.), pp. 111–144. ISMPMI, St. Paul, MN. Ream, W. (1998). Import of Agrobacterium tumefaciens virulence proteins and transferred DNA into plant cell nuclei. Subcell. Biochem. 29, 365–384. Sheng, J., and Citovsky, V. (1996). Agrobacterium–plant cell DNA transport: Have virulence proteins, will travel. Plant Cell 8, 1699–1710. Spudich, G. M., Dang, T. A. T., Fernandez, D., Zhou, X.-R., and Christie, P. J. (1996). Organization and assembly of the Agrobacterium tumefaciens T-complex transport apparatus. In “Crown Gall: Advances in Understanding Interkingdom Gene Transfer” (W. Ream, and S. Gelvin, eds.), pp. 75–98. APS Press, St. Paul, MN. Winans, S. C., Burns, D. L., and Christie, P. J. (1996). Adaptation of a conjugal transfer system for the export of pathogenic macromolecules. Trends Microbiol. 4, 1616–1622. Zupan, J. R., and Zambryski, P. (1995). Transfer of T-DNA from Agrobacterium to the plant cell. Plant Physiol. 107, 1041–1047.

WEBSITES An overview of crow gall and its control http://helios.bto.ed.ac.uk/bto/microbes/crown.htm Website for Comprehensive Microbial Resource of The Institute for Genomic Research. Links to many other microbial genomic sites http://www.tigr.org/tigr-scripts/CMR2/CMRHomePage.spl Website of the University of Washington Crown Hall Group http://depts.washington.edu/agro/

ch03.qxd 10/9/03 6:45 PM Page 25

3 Antibiotic resistance in bacteria Julian Davies University of British Columbia

Vera Webb Lookfar Solutions Inc., British Columbia

I. INTRODUCTION

We must swim with the microbes and study their survival and adaptation to new habitats. Richard M. Krause (1994).

The development of antibiotic resistance can be viewed as a global problem in microbial genetic ecology. It is a very complex problem to contemplate, let alone solve, due to the geographic scale, the variety of environmental factors, and the enormous number and diversity of microbial participants. In addition, the situation can only be viewed retrospectively, and what has been done was uncontrolled and largely unrecorded. Simply put, since the introduction of antibiotics for the treatment of infectious diseases in the late 1940s, human and animal microbial ecology has been drastically disturbed. The response of microbes to the threat of extinction has been to find genetic and biochemical evolutionary routes that led to the development of resistance to every antimicrobial agent used. The result is a large pool of resistance determinants in the environment. The origins, evolution, and dissemination of these resistance genes is the subject of this review.

GLOSSARY Broad-spectrum antibiotic A drug that affects a wide range of bacteria. Cell wall, bacterial A rigid outer layer of bacterial cells containing peptidoglycan. Integron A mobile genetic element that serves as the site of integration for DNA. It encodes a site-specific recombinase called integrase. Multidrug Efflux Pumps Mechanisms for exporting a number of compounds across the cell membrane. Peptidoglycan or Murein A long chain of disaccharides consisting of N-acetylglucosamine and muramic acid with short peptides that are often cross linked to give a mesh-like two-dimensional structure. Its synthetic machinery is the target for -lactam antibiotics. Protein synthesis machinery A large complex of proteins and RNAs, including ribosome, transfer RNAs (tRNA), messenger RNA (mRNA), proteins, and small molecules. Resistance determinants (or R determinants) Genes that confer antibiotic resistance. Target for antibiotics Ideally, a constituent present in the target cell (e.g., bacterial pathogen, cancer cell) and not present in the host. The Desk Encyclopedia of Microbiology ISBN: 0-12-621361-5

II. MECHANISMS OF RESISTANCE: BIOCHEMISTRY AND GENETICS The use of antibiotics should have created a catastrophic situation for microbial populations; however, their genetic flexibility allowed bacteria to survive (and even thrive) in hostile environments. The alternatives for survival for threatened microbial populations were either mutation of target sites or acquisition of novel

25

Copyright © 2003 Elsevier Ltd All rights of reproduction in any form reserved

ch03.qxd 10/9/03 6:45 PM Page 26

26

the desk encyclopedia of microbiology

biochemical functions (also known as resistance determinants or R determinants).1 Table 3.1 lists antibiotic resistance mechanisms that are transferable among bacteria. Mutation and the acquisition of R determinants are not mutually exclusive resistance strategies. Under the selective pressure of the antibiotic, mutation can lead to “protein engineering” of the acquired resistance determinant which may expand its substrate range to include semisynthetic molecules designed to be refractory to the wild-type enzyme (see Section II.A.2.a). Theoretically, the ideal target for a chemotherapeutic agent is a constituent which is present in the target cell (e.g. bacterial pathogen, cancer cell) and not present in the host cell. The first antibiotics to be employed generally, the penicillins, targeted the synthesis of peptidoglycan, a component unique to the bacterial cell wall. Antibiotics have been found that inhibit the synthesis or interfere with the function of essentially all cellular macromolecules. Table 3.2 lists some of the common antimicrobial drugs used clinically and their targets within the bacterial cell. Our discussion of the mechanisms of resistance will be organized according to the targets of antibiotic activity.

TABLE 3.1 Transferable antibiotic resistance mechanisms in bacteria Mechanism

Antibiotic

Reduced uptake into cell Active efflux from cell Modification of target to eliminate or reduce binding of antibiotic

Chloramphenicol Tetracyclines -Lactams Erythromycin Lincomycin Mupirocin

Inactivation of antibiotic by enzymatic modification Hydrolysis Derivatization

Sequestration of antibiotic by protein binding Metabolic bypass of inhibited reaction Binding of specific immunity protein Overproduction of antibiotic target (titration)

-Lactams Erythromycin Aminoglycosides Chloramphenicol Lincomycin -Lactams Fusidic acid Trimethoprim Sulfonamides Bleomycin Trimethoprim Sulfonamides

TABLE 3.2 Antimicrobial drugs: mechanisms of action

A. Targets and specific mechanisms

Target

1. Protein synthesis

Protein synthesis 30 S subunit

Protein synthesis involves a number of components: the ribosbme, transfer RNA (tRNA), messenger RNA (mRNA), numerous ancillary proteins, and other small molecules. When protein synthesis is inhibited by the action of an antibiotic, the ribosome is usually the target. The bacterial ribosome is composed of two riboprotein subunits. The small, 30 S subunit consists of approximately 21 ribosomal proteins (rprotein) and the 16 S ribosomal RNA (rRNA) molecule (about 1500 nucleotides), whereas the large, 50 S subunit contains approximately 34 rproteins and the 23 S and 5 S rRNA molecules (about 3000 and 120 nucleotides, respectively). The complexity of the ribosome structure and the redundancy of many of the genes encoding ribosomal components in most bacterial genera makes resistance due to point mutation an unlikely event. Generally, resistance to antibiotics that inhibit protein synthesis is mediated by R determinants. a. Aminoglycosides Aminoglycosides, which are broad-spectrum antibiotics, are composed of three or more aminocyclitol units.

Drug

50 S subunit

tRNA synthetase Cell wall synthesis Penicillin binding proteins

D-Ala-D-Ala

binding

Muramic acid biosynthesis Nucleic acid synthesis DNA RNA

1

A note concerning terminology: “determinant” refers to the genetic element which encodes a “mechanism” or biochemical activity which confers resistance.

Folic acid metabolism

Tetracyclines Aminoglycosides: streptomycin, amikacin, apramycin, gentamicin, kanamycin, tobramycin, netilmicin, isepamicin Chloramphenicol Fusidic acid Macrolides: erythromycin, streptogramin B Lincosamides Mupirocin -Lactams Penicillins: ampicillin, methicillin, oxacillin Cephalosporins: cefoxitin, cefotaxime Carbapenems: imipenen -Lactamase inhibitors: clavulanic acid, sulbactam Cyclic glycopeptides: vancomycin, teicoplanin, avoparcin Fosfomycin Quinolones and fluoroquinolones: ciprofloxicin, sparfloxacin Rifampicin Trimethoprim Sulfonamides

ch03.qxd 10/9/03 6:45 PM Page 27

antibiotic resistance in bacteria They bind to the 30 S subunit and prevent the transition from the initiated complex to the elongation complex; they also interfere with the decoding process. As noted above, target site mutations of ribosomal components resulting in antibiotic resistance are rare; however, they do occur. For example, in the slow-growing Mycobacterium tuberculosis, mutants resistant to streptomycin appear more frequently than they do in Escherichia coli. Mutations leading to resistance result from an altered S12 rprotein or 16 S rRNA such that the ribosome has reduced affinity for the antibiotic. Most fast-growing bacteria have multiple copies of the rRNA genes, and because resistance is genetically recessive to antibiotic sensitivity, only rare mutations in the gene for protein S12 are isolated under normal situations. However, because the slow-growing mycobacteria possess only single copies of the rRNA genes, streptomycin resistance can arise by mutational alteration of either 16 S rRNA or ribosomal protein S12. Both types of mutations have been identified in M. tuberculosis (Finken et al., 1993). The introduction and therapeutic use of a series of naturally occurring and semisynthetic aminoglycosides over a 20-year period (1968–1988) led to the appearance of multiresistant strains resulting from selection and dissemination of a variety of aminoglycoside resistance determinants. For example, in 1994 the Aminoglycoside Resistance Study Group examined of the occurrence of aminoglycoside-resistance mechanisms in almost 2000 aminoglycoside-resistant Pseudomonas isolates from seven different geographic regions. In their study 37% of the isolates overall had at least two different mechanisms of resistance (Aminoglycoside Resistance Study Group, 1994). A dozen different types of modifications are known to be responsible for resistance to the aminoglycosides. When one considers that each of these enzymes has a number of isozymic forms, there are at least 30 different genes implicated in bacterial resistance to this class of antibiotics. Different proteins in the same functional class may show as little as 44% amino acid similarity. The phylogenic relationships between different aminoglycoside-modifying enzymes were the subject of an excellent review by Shaw and co-workers, who have collated the nucleotide and protein sequences of the known aminoglycoside acetyltransferases, phosphotransferases, and adenylyltransferases responsible for resistance in both pathogenic bacteria and antibiotic-producing strains (Shaw et al., 1993). Rarely do a few point mutations in the aminoglycoside resistance gene arise sufficient to generate a modified enzyme with an altered substrate range that would lead to a significant change in the antibiotic resistance spectrum (Rather et al., 1992; Kocabiyik and Perlin, 1992); in other words, these genes do not

27

appear to undergo facile mutational changes that generate enzymes with altered substrate activity. In vitro mutagenesis studies have failed to generate extendedspectrum resistance to this class of antibiotics, and, so far, such changes have not been identified in clinical isolates, in contrast to the situation with the -lactamases (see Section II.B.2). The bacterial response to the introduction of a new aminoglycoside antibiotic is to acquire a different resistance gene. For example, when the use of kanamycin was superseded by a new but structurally related aminoglycoside (gentamicin), a previously unknown class of antibiotic-inactivating enzymes was detected in the gentamicin-resistant strains that appeared in hospitals. Many gram-positive pathogens possess an unusual bifunctional aminoglycoside-modifying enzyme, the only reported instance of fused resistance genes. The enzyme encodes acetyl- and phosphotransferase activities based on protein domains acquired by the fusion of two independent resistance genes (Ferretti et al., 1986; Rouch et al., 1987). The hybrid gene is widely distributed among hospital isolates of staphylococci and enterococci and can be assumed to have evolved as a fortuitous gene fusion during the process of insertion of the two resistance genes into the “cloning” site of a transposon. The therapeutic use of aminoglycoside antibiotics has decreased in recent years, largely because of the introduction of the less toxic broad-spectrum -lactams. As a result, there has been a relatively limited effort to seek specific inhibitors of the aminoglycosidemodifying enzymes, which might have offered therapeutic potential to extend the effective range of this class of antibiotics. Although some active inhibitors have been identified, none of them has been deemed fit for introduction into clinical practice. The situation may change as a result of structural studies by Puglisi and co-workers (Fourmy et al., 1996). Employing threedimensional (3-D) nuclear magnetic resonance techniques, they were able to model the aminoglycoside paromomycin into its binding site (the decoding site) in a 16 S RNA fragment. The logical extension of this important information, which identifies the key binding functions on the antibiotic, would be to design aminoglycoside molecules that: (a) could bind to a resistant ribosome modified by base substitution or methylation; or (b) might be refractory to modification by one or more of the aminoglycoside-modifying enzymes yet retain high affinity to the rRNA receptor site. This aminoglycoside/rRNA binding is the first to be characterized at this level of resolution and may permit the design and production of not only aminoglycoside analogs refractory to modification but also those which have reduced affinity for mammalian ribosomes; the latter is an important component of aminoglycoside toxicity.

ch03.qxd 10/9/03 6:45 PM Page 28

28

the desk encyclopedia of microbiology

b. Tetracycline Tetracyclines are perhaps the prime example of the development of antibiotic resistance; introduced in the late 1940s, they were the first group of broad-spectrum antibiotics (penicillin use was limited mostly to infections caused by gram-positive bacteria). Tetracyclines act by blocking the binding of aminoacyl-tRNAs to the A-site of the ribosome. They are active against most types of bacteria including gram-positives, gram-negatives, mycoplasmas, chlamydiae, and rickettsiae. This broad spectrum of activity and their relative safety, and low cost, have made tetracyclines the second most widely used group of antimicrobial agents after the lactams. It is probable that larger quantities of the tetracyclines have been produced and used than any other antibiotic. From a point of view of antibiotic resistance, almost everything that could have happened to the tetracyclines has done so! In 1953, just 6 years after their discovery, the first tetracycline-resistant Shigella dysenteriae were isolated in Japan. They were the earliest of the antibiotic substances to be used as feed additives in agriculture; enormous amounts have been dispensed in the last 40 years. In addition, because of their spectrum of activity, the tetracyclines became widely dispensed in both hospitals and the community. In many countries tetracycline has been virtually an over-the-counter drug for many years. As a result resistance to the tetracyclines is widely disseminated in many forms (see Tables 3.3 and 3.4). This class of antibiotics has become a paradigm for antibiotic resistance studies, and a number of useful reviews of tetracycline resistance have appeared (Hillen and Berens, 1994; Roberts, 1994). Because tetracyclines are extensively used, one might expect that resistance is also widespread (Table 3.3). At least 16 different tetracycline-resistance (Tet) determinants from “target organisms” and three different oxytetracycline-resistance (Otr) determinants from the producing organisms, Streptomyces species, have been described and characterized (Table 3.4). The majority of these determinants (13), are plasmid encoded. For example, the genes encoding TetA–E are found on plasmids in gram-negative bacteria, while those for TetK and TetL are found only in gram-positive microbes. The TetM determinant is generally found on the chromosome of both gramnegative and gram-positive bacteria, while TetO in gram-negative and TetK genes in gram-positive organisms have been detected in either location. Three different mechanisms for tetracycline resistance have been described: (a) energy-dependent efflux of tetracycline from the cell; (b) ribosome protection (summarized in Table 3.4); and (c) enzymatic

TABLE 3.3 Distribution of tetracycline-resistance determinants among various bacterial genera Gram-negatives Tet determinant

Gram-positives

Tet determinant

Actinobacillus Aeromonas Bacteroides Campylobacter Citrobacter Edwardsiella Eikenella Enterobacter Escherichia Fusobacterium Haemophilus Kingella Klebsiella Moraxella Neisseria Pasteurella Plesiomonas Prevotella Proteus Pseudomonas Salmonella Serratia Shigella Veillonella Yersinia Vibrio

Actinomyces Aerococcus Bacillus Clostridium Corynebacterium Enterococcus Eubacterium Gardnerella Gemella Lactobacillus Listeria Mobiluncus Mycobacterium Mycoplasma Peptostreptococcus Staphylococcus Streptococcus Streptomyces Ureaplasma

TetL TetM, O TetK, L TetK, L, M, P TetM TetK, L, M, O TetK, M TetM TetM TetO TetK, L, M, S TetO TetK, L, OtrA, B TetM TetK, L, M, O TetK, L, M, O TetK, L, M, O TetK, L, OtrA, B, C TetM

TetB TetA, B, D, E TetM, Q, X TetO TetA, B, C, D TetA, D TetM TetB, C, D TetA, B, C, D, E TetM TetB, M TetM TetA, D TetB TetM TetB, D, H TetA, B, D TetQ TetA, B, C TetA, C TetA, B, C, D, E TetA, B, C TetA, B, C, D TetM TetB TetA, B, C, D, E, G

Based on Roberts, 1994.

TABLE 3.4 Tetracycline-resistance determinants: location and mechanism of resistance Mechanism

Location

Efflux TetA, C, D, E TetB TetG, H TetK TetL TetA(P) OtrB

Plasmid Primarily plasmid Plasmid Chromosome and plasmid Primarily chromosome Plasmid Chromosome

Ribosomal TetM TetO TetS TetQ TetB(P) OtrA

Primarily chromosome Chromosome and plasmid Plasmid Chromosome Plasmid Chromosome

inactivation of tetracycline; only the first two mechanisms have been shown to have clinical significance. The majority of the Tet determinants code for a membrane-bound efflux protein of about 46 kilodaltons (kDa). Nucleotide sequence analysis of the genes encoding these proteins indicate that they are members of the so-called major facilitator (MF) family of efflux proteins (Sheridan and Chopra, 1991; Marger and Saier, 1993) (see Section II.B.2 and Table 3.5). The

ch03.qxd 10/9/03 6:45 PM Page 29

antibiotic resistance in bacteria TABLE 3.5 Multidrug efflux pumpsa Protein

Organism

Substrate

MFS QacA

Staphylococcus aureus

Mono- and divalent organic cations Acriflavin, cetyltrimethylammonium bromide, fluoroquinolones, chloramphenicol, rhodamine 6G CCCP, nalidixic acid, organomercurials, tetrachlorosalicylanilide, thiolactomycin Bicyclomycin, sulfathiazole Similar range as NorA Similar range as NorA

NorA

S. aureus

EmrB

Escherichia coli

Bcr Blt Bmr

E. coli Bacillus subtilis B. subtilis

SMR Smr

S. aureus

EmrE

E. coli

QacE

Klebsiella pneumonia

RND AcrAB TolC E. coli

AcrEF

E. coli

MexAB OprM

Pseudomonas aeruginosa

Monovalent cations, e.g., cetyltrimethylammonium bromide, crystal violet, ethidium bromide Monovalent cations, e.g., cetyltrimethylammonium bromide, crystal violet, ethidium bromide, methyl viologen, tetracycline Similar range to Smr Acriflavin, crystal violet, detergents, decanoate, ethidium bromide, erythromycin, tetracycline, chloramphenicol, -lactams, nalidixic acid Acriflavin, actinomycin D, vancomycin Chloramphenicol, -lactams, fluoroquinolones, tetracycline

a

Summarized from Paulsen et al. (1996).

efflux proteins exchange a proton for a tetracycline– cation complex against a concentration gradient. The other major mechanism for tetracycline resistance is target protection. Ribosome protection is conferred by a 72.5-kDa cytoplasmic protein. Sequence analysis has shown regions with a high degree of homology to elongation factors Tu and G (Taylor and Chau, 1996; Sanchez-Pescador et al., 1988). Every cloud has its silver lining, and the analysis of the tetA operon and its regulation by a modulation of a repressor–operator interaction has provided an exquisitely controllable gene expression system for eukaryotic cells. A number of genes in a variety of cell lines have

29

been placed under the control of the tetracyclineregulated tetA promoter with the result that transcription can be activated on the addition of the drug. c. Chloramphenicol Chloramphenicol acts on the 50 S ribosomal subunit to inhibit the peptidyl transferase reaction in protein synthesis. It is an effective broad-spectrum antibiotic, is inexpensive to produce, and is employed extensively in the Third World for the treatment of a variety of gram-negative pathogens (Salmonella, Vibrio, and Rickettsia). Liberal over-the-counter availability in many countries ensures strong selection pressure for the maintenance of chloramphenicol resistance. The number of indications for which chloramphenicol is the drug of choice has declined rapidly because of chronic toxicity, namely, depression of bone marrow function leading to blood disorders such as aplastic anemia. Thus, though chloramphenicol resistance determinants are widespread, their presence is not of major consequence from a therapeutic standpoint in Europe and North America. Chloramphenicol acetyltransferases (CATs) are widely distributed among bacterial pathogens of all genera. This group of enzymes has been analyzed in great detail by Shaw and collaborators (Shaw, 1984; Murray and Shaw, 1997); at least a dozen breeds of CAT genes encoding similar but not identical acetyltransferases have been identified (Bannam and Rood, 1991; Parent and Roy, 1992). As with the aminoglycoside-modifying enzymes, the CAT genes are (presumably) of independent derivation, because they cannot be linked by a small number of point mutations to a single ancestral gene. Potential origins for the CAT family have yet to be clearly identified. The type I CAT (encoded by transposon Tn9) has two activities: in addition to catalyzing the acylation of chloramphenicol, the protein forms a tight stochiometric complex with the steroidal antibiotic fusidic acid (Bennett and Chopra, 1993); thus sequestered, the latter antibiotic is ineffective. This is the only plasmid-determined resistance to fusidic acid that has been characterized. Notwithstanding their different structures, the two antibiotics bind competitively to the enzyme; interestingly, both chloramphenicol and fusidic acid exert their antibiotic action through inhibition of bacteria protein synthesis (fusidic acid blocks the translocation of the ribosome along the mRNA). Hence, the type I CAT is bifunctional, determining resistance to two structurally related antibiotics by distinct mechanisms. Given our understanding of the basic biochemistry of the drug and knowledge of the mechanism of chloramphenicol resistance (Day and Shaw, 1992), it is

ch03.qxd 10/9/03 6:45 PM Page 30

30

the desk encyclopedia of microbiology

surprising that more effort has not been put into rational drug design of chloramphenicol analogs of lower toxicity that would be active against resistant strains. Shaw’s group has studied the CAT protein extensively; the 3-D structure of the protein is known, and site-directed mutagenesis has been used to establish the kinetics of the reaction mechanism (Murray and Shaw, 1997). Fluorinated analogs of chloramphenicol with reduced substrate activity for CAT have been produced but have not been afforded extensive clinical study (Cannon et al., 1990). The antibiotic has an excellent inhibitory spectrum: might it be possible to use fusidic acid analogs to inhibit chloramphenicol acylation? Although the CAT mechanism is the most common form of chloramphenicol resistance, a second type causing the active efflux of chloramphenicol from Pseudomonas cells has been described (Bissonnette et al., 1991). The cml determinant is encoded on integrons (see Section IV.A) and, like the tetracycline efflux pump, is a member of the MF family of efflux systems (see Section II.B.2, and Table 3.5). The presence of the cloned cml gene in E. coli leads to, in addition to active pumping of the drug from the cell, a reduction in outer membrane permeability to chloramphenicol by repressing the synthesis of a major porin protein (Bissonnette et al., 1991). This effect has also been reported for the homolog of the cml gene inHaemophilus influenzae, a major causative agent of meningitis. d. Macrolides, lincosamides, and streptogramins The macrolide–lincosamide–streptogramin (MLS) group of antibiotics have been used principally for the treatment of infections caused by gram-positive bacteria. The macrolides, especially erythromycin and its derivatives, have been used extensively and may be employed for the treatment of methicillin-resistant Staphylococcus aureus (MRSA), although the multiple drug resistance of the latter often includes the MLS class. Extensive studies on the mechanism of action of erythromycin identify the peptidyltransferase center as the target of the drug although there are clearly some subtleties in mechanism that remain to be resolved. The principal mechanism of resistance to macrolide antibiotics involves methylation of the 23 S rRNA of the host giving the ermR phenotype. In clinical isolates, enzymatic modification of rRNA, rendering the ribosome refractory to inhibition, is the most prevalent mechanism and, worldwide, compromises the use of this class of antibiotic in the treatment of grampositive infections (Leclercq and Courvalin, 1991). Mutation of rRNA has also been shown to be important in some clinical situations: nucleotide sequence comparisons of clarithromycin-resistant clinical isolates

of Helicobacter pylori revealed that all resistant isolates had a single base pair mutation in the 23 S rRNA (Debets-Ossenkopp et al., 1996). In addition, a number of different mechanisms for the covalent modification of the MLS group have been described. For example, O-phosphorylation of erythromycin has been identified in a number of bacterial isolates (O’Hara et al., 1989), and hydrolytic cleavage of the lactone ring of this class of antibiotics has also been described. The lincosamides (lincomycin and clindamycin) have been shown to be inactivated by enzymatic O-nucleotidylation in gram-positive bacteria. Macrolides are also inactivated by esterases and acetyltransferases. For macrolides and lincosamides and the related streptogramins (the MLS group) (Arthur et al., 1987; Brisson-Noël et al., 1988), the latter forms of antibiotic inactivation seem (for the moment) to be a relatively minor mechanism of resistance. The macrolide antibiotics, especially the erythromycin group (14-membered lactones), contain a number of semisynthetic derivatives that have improved pharmacological characteristics. This includes activity against some resistant strains: however, no derivative has been found with effective potency against ermR strains. The 23 S rRNA methyltransferases from pathogenic grampositive cocci remain the most significant problem. To our knowledge, no useful inhibitor of these enzymes has been identified; with the availability of ample quantities of the purified enzymes it would not be surprising if rationally designed, specific inhibitors of some of the erm methyltransferases may eventually be developed. The control of expression of the erm methyltransferases has been studied extensively by Weisblum and by Dubnau and their colleagues (Weisblum, 1995; Monod et al., 1987). The majority of the resistant strains possess inducible resistance which is due to a novel posttranslational process in which the ribosome stalls on a 5-leader sequence in the presence of low concentrations of antibiotic. The biochemistry of this process has been analyzed extensively. Point mutations or deletions that disrupt the secondary structure of the leader protein sequence generate constitutive expression, and such mutants have been identified clinically. These strains are resistant to the majority of MLS antibiotics. The regulation of antibiotic resistance gene expression takes many forms (see tetracycline and vancomycin for other examples); the leadercontrol process seen with the erm methylases is reminiscent of the control of amino acid biosynthesis by attenuation, which has been extensively studied. One cannot help but marvel at the simplicity and “cheapness” of this form of control of gene expression—all that is required is a few extra bases flanking

ch03.qxd 10/9/03 6:45 PM Page 31

antibiotic resistance in bacteria the 5 end of the gene with no requirement of additional regulatory genes. The streptogramins (especially virginiamycins) contain two components, a macrocyclic lactone and a depsipeptide which have synergistic activity (one such combination has been named Synercid®). These compounds have long been used as animal feed additives with the result that bacteria resistant to the MLS class of antibiotics are commonly found in the flora of farm animals. This nontherapeutic application is likely to compromise the newer MLS antibiotics being developed for human therapy. For example, derivatives of the pristinamycins (quinupristin and dalfopristin) will shortly be recommended for the treatment of vancomycin-resistant enterococci, against which they have potent activity. Regrettably, due to animal applications of MLS antibiotics a significant gene pool of resistance determinants is already widely disseminated. e. Mupirocin Mupirocin is an example of how quickly resistance can develop to a new antibiotic. Introduced in 1985, it has been used solely as a topical treatment for staphylococcal skin infections and as a nasal spray against commensal MRSA. Mupirocin, also known as pseudomonic acid A, is produced by the gramnegative bacterium Pseudomonas fluorescens. It acts by inhibiting isoleucyl-tRNA synthetase, which results in the depletion of charged tRNA Ile, amino acid starvation, and ultimately the stringent response. The first reports of mupirocin resistance (MuR) appeared in 1987; while not yet a widespread problem, the MuR phenotype in coagulase-negative staphylococci and MRSA among others is emerging in many hospitals all over the world (Cookson, 1995; Zakrzewska-Bode et al., 1995; Udo et al., 1994). Resistance is due to the plasmid-encoded mupA gene, a mupirocin-resistant isoleucyl-rRNA synthetase (Noble et al., 1988). Transfer has been demonstrated by filter mating and probably accounts for the rapid spread of resistance (Rahman et al., 1993). An analysis of plasmids from clinical isolates of S. aureus has shown that mupirocin resistance can be found on multiple resistance, high copy number plasmids (Needham et al., 1994). 2. Cell wall synthesis The synthesis and integrity of the bacterial cell wall have been the focus of much attention as targets for antimicrobial agents. This is principally because this structure and its biosynthesis is unique to bacteria and also because inhibitors of cell well synthesis are usually bacteriocidal.

31

a. -Lactams In the years following the introduction of the -lactam antibiotics (penicillins and cephalosporins) for the treatment of gram-negative and gram-positive infections, there has been a constant tug-of-war between the pharmaceutical industry and the bacterial population: the one to produce a novel -lactam effective against the current epidemic of resistant bugs in hospitals, and the other to develop resistance to the newest “wonder” drug. The role of mutation is especially important in the evolution or expansion of resistance in the case of -lactams. In a 1992 review Neu showed the “phylogeny” of development of -lactam antibiotics in response to the evolution of bacterial resistance to this class of antibiotics (Neu, 1992). Of the several known mechanisms of resistance to the -lactam antibiotics (Table 3.1), the most elusive target is hydrolytic inactivation by -lactamases. A single base change in the gene for a -lactamase can change the substrate specificity of the enzyme ( Jacoby and Archer, 1991). Such changes occur frequently, especially in the Enterobacteriaceae, and it is frightening to realize that one single base change in a gene encoding a bacterial -lactamase may render useless $100 million worth of pharmaceutical research effort. The cycle of natural protein engineering in response to changing antibiotic-selection pressure has been demonstrated especially for the TEM -lactamase (penicillinase and cephalosporinase) genes. The parental genes appear to originate from a variety of different (and unknown) sources (Couture et al., 1992). The -lactamase families differ by a substantial number of amino acids, as is the case for other antibiotic resistance genes. Sequential expansion of their substrate range to accommodate newly introduced -lactam antibiotics is a special case and occurs by a series of point mutations at different sites within the gene that change the functional interactions between the enzyme and its -lactam substrate. More than 30 of the so-called extendedspectrum -lactamases have been identified (and more will come). The fact that the -lactamase genes so readily undergo mutational alterations in substrate recognition could have several explanations, one being that the -lactamases, like the related proteases, have a single active site that does not require interaction with any cofactors. Other antibiotic-modifying enzymes often have two active binding sites. The pharmacokinetic characteristics of the different classes of antibiotics (e.g. dose regimen, active concentration, and route of excretion) also may favor the pathway of mutational alteration in the development of resistance. One aspect of the mutational variations of the -lactamase genes

ch03.qxd 10/9/03 6:45 PM Page 32

32

the desk encyclopedia of microbiology

might involve the presence of mutator genes on the R plasmids in the bacterial hosts (LeClerc et al., 1996). These genes increase mutation frequency several-fold and could explain the facile evolution of the TEMbased enzymes. This characteristic of bacterial pathogens has received comparatively little study. As one approach to counteracting the destructive activity of -lactamases, a series of effective inhibitors of these enzymes has been employed. The inhibitors are structural analogs of -lactams that are, in most cases, dead-end irreversible inhibitors of the enzyme. Several have been used in combination with a -lactam antibiotic for the treatment of infections from resistant microbes, for example, the successful combination of amoxicillin (antibiotic) and clavulanic acid (inhibitor). However, the wily microbes are gaining the upper hand once again by producing mutant -lactamases that not only are capable of hydrolyzing the antibiotic but concomitantly become refractory to inhibition (Blazquez et al., 1993). In addition to active site mutation, other changes in -lactamase genes have evolved in response to continued -lactam use. In some cases, increased resistance results from increased expression of the gene through an upregulating promoter mutation (Chen and Clowes, 1987; Mabilat et al., 1990); alternatively, chromosomal -lactamase genes can be overexpressed in highly resistant strains as a result of other changes in transcriptional regulation (Honoré et al., 1986). Drug inactivation is not the only mechanism of resistance to the -lactams. In fact, mutations that alter access to the target (penicillin binding proteins, pbp) of the drug through porin channels have been widely reported (Nikaido, 1994). Methicillin resistance in S. aureus (MRSA) is due to an unusual genetic complex which replaces the normal pbp2 with the penicillin refractory pbp2A. The origin of this resistance determinant is unknown, but the consequences of the clonal distribution of MRSA is well documented. It would appear that most MRSA are close relatives of a small number of parental derivatives that have been disseminated by international travel. Alterations of other pbp’s in different bacterial pathogens have been responsible for widespread resistance to -lactam antibiotics; in the case of Streptococcus pneumoniae and Neisseria gonorrhoeae this has occurred by interspecific recombination leading to the formation of mosaic genes that produce pbps with markedly reduced affinity for the drugs.

only come into prominence since the late 1980s, being the only available class of antibiotic effective for the treatment of MRSA and methicillin-resistant enterococci (MRE). However, numerous outbreaks of vancomycin-resistant enterococci (VRE) have been reported in hospitals around the world (VRE are essentially untreatable by any approved antibiotic), and there is great concern that vancomycin-resistance determinants will be transferred to pathogenic staphylococci; the resulting methicillin, vancomycin resistant S. aureus (MVRSA) will be the “Superbug,” the “Andromeda” strain that infectious disease experts fear most (at least according to the newspapers). The glycopeptides block cell wall synthesis by binding to the peptidoglycan precursor dipeptide D-alanyl-D-alanine (D-Ala-D-Ala) and preventing its incorporation into the macromolecular structure of the cell wall. The most common type of resistance is the vanA type found principally in Enterococcus faecalis (Walsh et al., 1996; Arthur et al., 1996). Resistance due to the VanA phenotype results from the substitution of the depsipeptide D-alanyl-D-lactate (D-Ala-D-Lac) for D-Ala-D-Ala residues, thereby reducing the binding of the antibiotic by eliminating a key hydrogen bond in the D-Ala-D-Ala complex. The introduction of D-Lac is encoded by the nine genes of the VanA cluster and is associated with a mechanism to prevent the formation of native D-Ala-D-Ala containing peptide in the same host; thus, the resistance is dominant. Vancomycin resistance is inducible by a two-component regulatory system; the inducers are not the antibiotics, but rather the accumulated peptidoglycan fragments produced by the initial inhibitory action of the glycopeptides. The VanA cluster is normally found on a conjugative transposon related to Tn1546, and it is probably responsible for the widespread dissemination of glycopeptide resistance among the enterococci. A plasmid carrying the VanA resistance determinants has been transferred from enterococci to staphylococci under laboratory conditions (Noble et al., 1992), and there is apprehension that this will occur in clinical circumstances. Regrettably a glycopeptide antibiotic, avoparcin, has been employed extensively as a feed additive for chickens and pigs in certain European countries. This has led to the appearance of a high proportion glycopeptide-resistant enterococci in natural populations. This feeding practice has now been banned by the European Union (EU), but is it too late? Only time will tell. c. Fosfomycin

b. Glycopeptides The glycopeptide antibiotics, vancomycin and teichoplanin, were first discovered in the 1950s but have

A widely used mechanism for the detoxification of cell poisons in eukaryotes is the formation of glutathione adducts; for example, this mechanism is

ch03.qxd 10/9/03 6:45 PM Page 33

antibiotic resistance in bacteria commonly used for herbicide detoxification in plants (Timmermann, 1988). However, in spite of the fact that many microbes generate large quantities of this important thiol, only one example of an antibiotic resistance mechanism of this type has so far been identified in bacteria, namely, that of fosfomycin. Fosfomycin, produced by a streptomycete, is an analog of phosphoenol pyruvate and interferes with bacterial cell wall synthesis by inhibiting the formation of N-acetylmuramic acid, a unique component of bacterial cell walls. It is employed in the treatment of sepsis, both alone and in combination with other antimicrobial agents. In gram-negative bacteria transmissible resistance is due to a plasmid-encoded glutathione S-transferase that catalyzes the formation of an inactive fosfomycin–glutathione adduct (Arca et al., 1990). Two independent genes for fosfomycin resistance have been cloned and sequenced, one from Serratia marcescens (Suárez and Mendoza, 1991) and the other from Staphylococcus aureus (Zilhao and Courvalin, 1990); the two genes are unrelated at the sequence level. It is unlikely that the gram-positive gene encodes an enzyme involved in the production of a glutathione adduct, because S. aureus does not contain glutathione! 3. DNA and RNA synthesis Nucleic acid metabolism has attracted much attention as a potential target for antimicrobial drugs; the strategy of hitting at the “heart” of the microbe seemed the most likely to lead to effective bactericidal agents. Unfortunately, the ubiquity of DNA and RNA and the failure to identify discriminating target differences in the biosynthetic enzymes made this search unproductive until relatively recently when the fluoroquinolones (a class of synthetic drugs with no known natural analogs) were introduced. However, resistance mechanisms were not long in appearing, and the fluoroquinolones instead of being the “superdrugs” needed, are already limited by resistance. a. Fluoroquinolones Nalidixic acid, the prototype quinolone antibiotic discovered in 1962, had limited use, principally for urinary tract infections caused by gram-negative bacteria. Resistance developed by mutation, and the nalR phenotype proved to be the first useful marker for gyrA the gene encoding the A subunit of DNA gyrase (topoisomerase I). The development of resistance to nalidixic acid occurred solely by this type of mutation, and plasmid-determined resistance has never been reliably identified. This is not surprising, given that nalidixic acid is a purely synthetic chemical and no natural analog has been identified.

33

In the late 1970s, the first fluoroquinolone antimicrobials were introduced; these proved vastly superior to nalidixic acid and are among the most potent antimicrobial agents known. A number of fluoroquinolone antibiotics have now been introduced as antiinfectives and most show good, broad-spectrum activity. In laboratory studies, mutations to high-level resistance occurred at relatively low frequency, and genetic studies identified a number of different DNA replication-associated targets. As with nalidixic acid, topoisomerase I is the principal target, but other targets associated with bacterial DNA replication which give the FQR phenotype have been identified in different bacterial species. In clinical use, resistance to the newer fluoroquinolones has been found to develop quite rapidly as a result of one or more mutations. In addition to target mutation, active efflux of the drug is also an important mechanism of resistance; for example, the norA mutation identifies a multiple drug resistance (mdr) system in S. aureus (see Section II.B.2 and Table 3.5). Strains resistant to high levels of the drug have been identified frequently during the course of treatment especially with P. aeruginosa and S. aureus infections. Resistance is due to multiple mutations leading to increased efflux and alteration of components of the DNA synthetic apparatus. Clonal dissemination of FQR strains appears to be quite common in nosocomial P. aeruginosa infections. No plasmidmediated resistance to fluoroquinolones has been identified to date, although possible mechanisms leading to dominant resistance genes can be envisaged. b. Rifampicin Rifampicin is the only inhibitor of bacterial (and mitochondrial) RNA polymerases that has ever been used in the treatment of infectious disease. Mutations in the gene encoding the subunit of RNA polymerase (rpoB) give high level resistance to the drug; the study of these mutations has provided important information on the structure and function of the RNA polymerase proteins in bacteria. Rifampicin is used in the treatment of gram-positive bacteria and is effective against mycobacterial and staphylococcal infections. Because it is lipophilic and thus diffuses rapidly across the hydrophobic cell envelop of mycobacteria, rifampicin is one of the frontline drugs for the treatment of tuberculosis and leprosy. However, the appearance of resistant strains as a result of rpoB mutations is quite common in multiple drug-resistant strains (Cole, 1994). In addition, inactivation of rifampicin in fast growing mycobacterial strains by phosphorylation, glucosylation, and ribosylation has been reported (Dabbs et al., 1995).

ch03.qxd 10/9/03 6:45 PM Page 34

34

the desk encyclopedia of microbiology

4. Folic acid biosynthesis

b. Trimethoprim

As the above discussion illustrates, the biosynthesis and function of cellular macromolecules are the principal targets for the majority of antimicrobial agents. However, interference with the activity of enzymes involved in intermediary metabolism is also an effective strategy. Although many potential antimicrobial targets have been identified and tested, to date only folic acid biosynthesis has been successfully exploited in the development of useful drugs. Folic acid is involved in the transfer of one-carbon groups utilized in the synthesis and metabolism of amino acids such as methionine and glycine and in the nucleotide precursors adenine, guanine, and thymine. Folic acid is converted in two reduction steps to tetrahydrofolate (FH4), which serves as the intermediate carrier of hydroxymethyl, formyl, or methyl groups in a large number of enzymatic reactions in which “one-carbon” groups are transferred from one metabolite to another or are interconverted. The synthetic antibacterial agents sulfonamides and trimethoprim inhibit specific steps in the biosynthesis of FH4. The current state of resistance to sulfonamides and trimethoprim in major bacterial pathogens and the mechanisms of sulfonamide and trimethoprim resistance have been reviewed (Huovinen et al., 1995).

Trimethoprim was first introduced in 1962, and since 1968 it has been used (often in combination with sulfonamides due to a supposed synergistic effect) for numerous clinical indications. Like the sulfonamides, trimethoprim has a wide spectrum of activity and low cost. The target is the enzyme dihydrofolate reductase (DHFR), which is essential in all living cells. Trimethoprim is a structural analog of dihydrofolic acid and acts as a competitive inhibitor of the reductase. The human DHFR is naturally resistant to trimethoprim, which is the basis for its use. Resistance has been reported to both trimethoprim and sulfonamides since their respective introductions into clinical practice. Although the principal form of resistance is plasmid mediated, a clinical isolate of E. coli was described in which the chromosomal DFHR was overproduced several hundredfold, leading to very high trimethoprim resistance (minimum inhibitory concentration 1 g/l). SulR and TmpR strains carry plasmid-encoded dhps and dfhr genes that may be up to 100 times less susceptible to the inhibitors. Extensive genetic and enzymatic studies, principally by Sköld and collaborators, have characterized resistance in the Enterobacteriaceae. The sul gene is part of the 3 conserved region of integron structures and may have been the first resistance determinant acquired (see Section IV.A). Less information is available for resistant grampositive pathogens. Nonetheless, the origins of SulR and TmpR genes remains a mystery.

a. Sulfonamides The sulfonamides were first discovered in 1932 and introduced into clinical practice in the late 1930s. They have a wide spectrum of activity and have been used in urinary tract infections due to the Enterobacteriaceae, in respiratory tract infections due to Streptococcus pneumoniae and Haemophilus influenzae, in skin infections due to S. aureus, and in gastrointestinal tract infections due to E. coli and Shigella spp. The wide range of clinical indications and low production costs maintain the popularity of the sulfonamides in Third World countries. The enzyme dihydropteroate synthase (DHPS) catalyzes the formation of dihydropteric acid, the immediate precursor of dihydrofolic acid. DHPS found in bacteria and some protozoan parasites, but not in human cells, is the target of sulfonamides. These drugs are structural analogs of p-aminobenzoic acid, the normal substrate of DHPS, and act as competitive inhibitors for the enzyme, thus blocking folic acid biosynthesis in bacterial cells. A large number of sulfonamides have been synthesized that show wide variations in therapeutic activity! One class, the dapsones, remains an effective anti-leprosy drug.

B. Broad-spectrum resistance systems In the first part of this section, we examined bacterial targets for antibiotic activity and mechanisms that specifically provide resistance to those antibiotics. The mechanisms included modification of the target (e.g. vancomycin resistance), modification of the antibiotic (e.g. aminoglycoside methyltransferases), overproduction of the target (e.g. trimethoprim resistance), and extrusion of the drug from the cell (e.g. TetA-type tetracycline resistance). However, even as the first antimicrobial agents were being tested, researchers noticed “intrinsic” differences in sensitivity among the target organisms to a wide range of compounds. 1. Membranes and cell surfaces Initially, intrinsic differences in resistance to antibiotics and other chemotherapeutic agents were attributed to structures such as the gram-negative outer membrane and the mycobacterial cell surface. Intrinsic drug resistance in mycobacteria has been reviewed by

ch03.qxd 10/9/03 6:45 PM Page 35

antibiotic resistance in bacteria Nikaido and collaborators (Jarlier and Nikaido, 1994). They suggest that lipophilic molecules are slowed by the low fluidity of the lipid bilayer surrounding the cell wall and that hydrophobic molecules enter the mycobacterial cell slowly because the porins are inefficient and few in number. They note that although these surface features strongly contribute the high level of natural resistance in mycobacteria, other factors are also involved such as pbps with low affinity for penicillin and the presence of -lactamases. 2. Multidrug efflux pumps A cell has a number of different ways to export material across its membrane. All of them involve the expenditure of energy. The most well-characterized multiple drug resistance pumps in mammalian cells are the ABC (ATP binding cassette) transporters. In bacteria, the ABC transporters are primarily seen in the translocation of virulence factors such as hemolysin in E. coli and cyclolysin in Bordetella pertussis. A second type of active export system has been characterized in which the energy to drive the pump comes from the proton motive force (PMF) of the transmembrane electrochemical proton gradient. These multidrug efflux systems have been the subject of a number of reviews (Lewis, 1994; Nikaido, 1996; Paulsen et al., 1996). There are three families of PMF multidrug efflux pumps: the major facilitator superfamily (MFS), the staphylococcal (or small) multidrug resistance (SMR) family, and the resistance/nodulation/cell division (RND) family. Table 3.5 lists examples from each of these families and the types of compounds they pump out of the cell. In addition to these multisubstrate pumps, proton motive force efflux pumps for specific antibiotic resistance, such as the TetK and TetL (MFS-type) pumps, have also been described (see Section II.A.1.b). The Acr multidrug efflux pump found in E. coli is one of the most well characterized of the RND-type pumps. Expression of the Acr efflux pump is controlled by the marA protein. The multiple antibiotic resistance (mar) phenotype was first described in 1983 by Levy and co-workers when they plated E. coli on medium containing either tetracycline or chloramphenicol and obtained mutants that were also resistant to the other antibiotic. Further analysis showed that these mutants had additional resistances to -lactams, puromycin, rifampicin, and nalidixic acid. The most striking aspect of this multiple drug resistance phenotype was the wide range of structurally unrelated compounds with which it was observed. Subsequent studies have found that the mar phenotype is part of a complex stress response system which includes the superoxide response locus soxRS (Miller and Sulavik,

35

1996). Rather than encoding the structural components responsible for the mar phenotype, the mar locus encodes a regulatory system. The marA protein is a positive regulator which controls expression of at least two loci, the acr locus, which encodes the genes for a multidrug efflux pump, and the micF locus, which encodes an antisense repressor of the outer membrane protein ompF. Expression from the mar locus is tightly controlled by the first gene in the operon, marR, which encodes a represser protein (Miller and Sulavik, 1996). As noted above, the mar locus is part of a complex stress response system. Levy and co-workers (Goldman et al., 1996) have reported that mutations of the marR repressor protein protected E. coli from rapid killing by fluoroquinolones. They hypothesize that such protection may allow cells time to develop mutations that lead to higher levels of fluoroquinolone resistance, and may thus explain the increasing frequency of occurrence of resistant clinical isolates. Such mutations have been found among clinical strains of fluoroquinolone-resistant E. coli, suggesting that mutations at the mar locus may be the first step in clinically significant fluoroquinolone resistance. The latter may have been the case for all clinically significant antibiotic resistance: a mutational event leading to a low-level increase in drug efflux, followed by the acquisition of a heterologous resistance determinant, leading to high-level antibiotic resistance. 3. Other types of natural resistance Other types of natural resistance in bacteria will depend on the organism and the drug in question. For example, three species of enterococci, Enterococcus gallinarum, E. casseliflavus, and E. flavescens, produce peptidoglycan precursors which end in D-serine residues and are intrinsically resistant to low levels of vancomycin. Similarly, genera from the lactic acid bacteria Lactobacillus, Leuconostoc, and Pediococcus are resistant to high levels of glycopeptides because their cell wall precursors end with D-lactate. As noted above, mycobacteria species have pbps which have a low affinity for penicillin. In addition, some mycobacteria have been reported to have ribosomes that have a lower affinity for macrolides than the ribosomes of S. aureus (Jarlier and Nikaido, 1994).

III. GENE TRANSFER All available evidence suggests that the acquisition and dissemination of antibiotic resistance genes in bacterial pathogens has occurred since the late 1940s.

ch03.qxd 10/9/03 6:45 PM Page 36

36

the desk encyclopedia of microbiology

The best support for this notion comes from studies of Hughes and Datta (Datta and Hughes, 1983; Hughes and Datta, 1983) who examined the “Murray Collection,” a collection of (mostly) gram-negative pathogens obtained from clinical specimens in the preantibiotic era. None of these strains show evidence of resistance to antibiotics in current use. Given the critical role of gene exchange in bacterial evolution, it is selfevident that extensive interspecific and intergeneric gene transfer must have occurred during the golden age of antibiotics. This subject has been reviewed many times, and a variety of mechanisms of gene transfer have been invoked in the process of resistance determinant acquisition and dispersion (see Table 3.6) and characterized in laboratory studies. It can be assumed that these (and probably other processes) all occur in nature.

A. Transduction Transduction is the exchange of bacterial genes mediated by bacteriophage, or phage. When a phage infects a cell, the phage genes direct the takeover of the host DNA and protein synthesizing machinery so that new phage particles can be made. Transducing particles are formed when plasmid DNA or fragments of host chromosomal DNA are erroneously packaged into phage particles during the replication process. Transducing particles (those carrying nonphage DNA) are included when phage are liberated from the infected cell to encounter another host and begin the next round of infection. Although there are many laboratory studies of transduction of antibiotic resistance, this mechanism has been considered less important in the dissemination of antibiotic resistance genes because phage generally have limited host ranges; they can infect only members of the same or closely related species, and the size of DNA transferred does not usually exceed 50 kb. However, phage of extraordinarily broad host specificity have been described. For example, phage PRR1 and PRD1 will infect any gram-negative bacterium containing the resistance plasmid RP1 (Olsen and Shipley, 1973; Olsen et al., 1974) and could, in principle, transfer genetic information between unrelated bacterial species. Transduction has been documented in at least 60 species of bacteria found in a wide variety of environments (Kokjohn, 1989). Although the actual level of intraspecies and interspecies transduction are unknown, the potential for transductional gene exchange is likely to be universal among the eubacteria. A study of the presence of bacteriophage in aquatic environments has shown that there may be as many as 108 phage particles per milliliter (Bergh et al.,

1989); the authors calculate that at such concentrations one-third of the total bacterial population is subject to phage attack every 24 h. In transduction the DNA is protected from degradation within the phage particle, and Stotzky has suggested that transduction may be as important as conjugation or transformation as a mechanism of gene transfer in natural habitats (Stotzky, 1989).

B. Conjugation Conjugation is the process in which DNA is transferred during cell-to-cell contact. It has long been considered the most important mechanism for the dissemination of antibiotic resistance genes. During an epidemic of dysentery in Japan in the late 1950s increasing numbers of Shigella dysenteriae strains were isolated that were resistant to up to four antibiotics simultaneously. It soon became clear that the emergence of multiply resistant strains could not be attributed to mutation. Furthermore, both sensitive and resistant Shigella could be isolated from a single patient, and the Shigella sp. and E. coli obtained from the same patients often exhibited the same multiple resistance patterns. These finding led to the discovery of resistance transfer factors and were also an early indication of the contribution of conjugative transfer to the natural evolution of new bacterial phenotypes. In addition to plasmid-mediated conjugal transfer, another form of conjugation has been reported to take place in gram-positive organisms. Conjugative transposons were first reported in S. pneumoniae when the transfer of antibiotic resistance determinants occurred in the absence of plasmids (Shoemaker et al., 1980). Salyers has suggested that conjugative transposons may be more important that conjugative plasmids in broad host-range gene transfer between some species of bacteria (Salyers, 1993). Conjugative transposons are not considered typical transposons (Scott, 1992). As of 1993, three different families of conjugative transposons had been found: (a) the Tn916 family (originally found in streptococci but now known also to occur in gram-negative bacteria such as Campylobacter) (Salyers, 1993); (b) the S. pneumoniae family; and (c) Bacteroides family. Conjugative transposons range in size from 15 to 150 kb, and, in addition to other resistance genes, most encode tetracycline-resistance determinants (e.g. Tn916 encodes the TetM determinant, and TetQ (Salyers, 1993) is found on the conjugative transposon from the Bacteroides group). Does conjugation take place in the environment? The ideal site for gene transfer is the warm, wet, nutrientabundant environment of the mammalian intestinal tract with its associated high concentration of bacteria.

ch03.qxd 10/9/03 6:45 PM Page 37

TABLE 3.6 Gene transfer processes Required genetic determinants

Observed in

Process

Components

Donor

Recipient

Laboratory

Nature

DNA formb

DNA size

Conjugation

Cell/cell

Transfer genes

May require receptor

ss

Fusion

Cell/cell

?

?

(rare)

Transduction

Bacteriophage /cell

Phage receptor

Phage receptor

Transformationc

DNA/cell

a.Competence determinants b.Chemical or physical changes

a

a

Host range

Comments

4 Mb

Very broad interspecific, intergeneric

ds

Unlimited

?

ds

50 kb

ss/ds

50 kb

Limited to closely replated species Very broad

Can be of very high efficiency; can reduce problem of restriction in recipient Likely to involve partners with damaged wall or membranes (protoplasts or spheroplasts) Very efficient

Chemically and electrically induced competence required; efficiency variable

All DNA exchange processes are subject to the negative effects of restriction (nuclease action) in the recipient. All processes, in principle, may take place with transfer of intact plasmids, chromosomes, or linear fragments. The requirement for recombination in the recipient depends on the nature of transferred DNA and its properties. ss, Single stranded; ds, double stranded. c Many bacterial species are genetically non-competent, but may be converted to a competent state by laboratory processes. Electrotransformation is a good example. b

ch03.qxd 10/9/03 6:45 PM Page 38

38

the desk encyclopedia of microbiology

The resident microflora is believed to serve as a reservoir for genes encoding antibiotic resistance which could be transferred not only to other members of this diverse bacterial population, but also to transient colonizers of the intestine, such as soil or water microbes or human pathogens. Using oligonucleotide probes having DNA sequence similarity to the hypervariable regions of the TetQ determinant, Salyers and coworkers provided evidence that gene transfer between species of Bacteroides, one of the predominant genera of the human intestine, and Prevotella sp., one of the predominant genera of livestock rumen, has taken place under physiological conditions (Nikolich et al., 1994).

system. Although gene exchange may under normal circumstances be rare in stable microbial microcosms, the intense selective pressure of antibiotic usage is likely to have provoked cascades of antibiotic resistance gene transfer between unrelated microbes. These transfers must involve different biochemical mechanisms during which efficiency is not a critical factor since the survival and multiplication of a small number of resistant progeny suffices to create a clinically problematic situation. It should be apparent that a great deal of additional study using modern molecular and amplification methods with complex microbial communities is necessary before the parameters of natural antibiotic resistance gene transfer can be defined properly.

C. Transformation Natural transformation is a physiological process characteristic of many bacterial species in which the cell takes up and expresses exogenous DNA. Although natural transformation has been reported to occur only in a limited number of genera, these include many pathogenic taxa such as Haemophilus, Mycobacterium, Streptococcus, Neisseria, Pseudomonas, and Vibrio (Stewart, 1989). Initial studies suggested that natural transformation in some of these genera was limited to DNA from that particular species. For example, an 11-bb recognition sequence permits Haemophilus influenzae to take up its own DNA preferentially compared to heterologous DNAs (Kahn and Smith, 1986). Given such specificity one could ask if natural transformation is an important mechanism in the transfer of antibiotic resistance genes. Spratt and co-workers have reported the transfer of penicillin resistance between S. pneumoniae and N. gonorrhoeae by transformation. Further, Roberts reported that when the TetB determinant (which is conferred by conjugation in other gramnegative groups) is present in Haemophilus species and highly tetracycline-resistant Moraxella catarrhalis, it is disseminated by transformation (Roberts et al., 1991).

D. General considerations There is a world of difference between laboratory and environmental studies. While the isolation of pure cultures is an important component of bacterial strain identification, the use of purified bacterial species in gene transfer studies does little to identify the process and probably bears no relationship to the processes of genetic exchange that take place in the complex microbial populations of the gastrointestinal tract (for example). When antibiotic-resistant bacteria are isolated from diseased tissue and identified as the responsible pathogen, this is the identification of the final product of a complex and poorly understood environmental

IV. EVOLUTION OF RESISTANCE DETERMINANT PLASMIDS A. The integron model Studies by Hughes and Datta of plasmids they isolated from the Murray collection (Hughes and Datta, 1983; Datta and Hughes, 1983) suggest that the appearance of resistance genes is a recent event, that is, the multiresistance plasmids found in pathogens must have been created since the 1940s. What really takes place when a new antimicrobial agent is introduced and plasmid-determined resistance develops within a few years? The most significant component of the process of antibiotic resistance flux in the microbial population is gene pickup, which has now been emulated in the laboratory. Largely due to the studies of Hall and co-workers (Stokes and Hall, 1989; Collis et al., 1993; Recchia and Hall, 1995), we have a good idea of the way in which transposable elements carrying multiple antibiotic resistance genes might be formed. From their studies of the organization of transposable elements, these researchers have identified a key structural constituent of one class of transposon that they named an “integron.” The integron is a mobile DNA element with a specific structure consisting of two conserved segments flanking a central region in which “cassettes” that encode functions such antibiotic resistance can be inserted. The 5 conserved region encodes a site-specific recombinase (integrase) and a strong promoter or promoters that ensure expression of the integrated cassettes. The integrase is responsible for the insertion of antibiotic resistance gene cassettes downstream of the promoter; ribosome binding sites are conveniently provided. More than one promoter sequence exists on the element (Lévesque et al., 1994); transcription initiation is very efficient and functions in

ch03.qxd 10/9/03 6:45 PM Page 39

39

antibiotic resistance in bacteria

secondary integration sites could explain how new genes may be inserted into integrons without the necessity for a 59-bp element, as the authors point out.

B. Other multiple resistance plasmids

FIGURE 3.1 The general structure of an integron. Integrons consist of a 5 conserved sequence that encodes an integrase and contains promoter sequences (P) responsible for the transcription of inserted gene cassettes. The 3 conserved sequence encodes resistance to sulfonamide drugs (sul). The insertion site for gene cassettes (GTTRRRY) is indicated.

both gram-negative and gram-positive bacteria. The 3 conserved region carries a gene for sulfonamide resistance (sul) and two open reading frames of unknown function. Probably the ancestral integron, encoded no antibiotic resistance (Fig. 3.1). The ubiquitous presence of sul in an element of this type might be surprising, although sulfonamides (see above) have been employed since the mid-1930s and (apart from mercury salts) are the longest used agents for the treatment of infectious diseases. The resistance gene cassettes are integrated into a specific insertion site in the integron. Typically, in the case of Tn21-related transposons, each antibiotic resistance cassette is associated with one of a functional family of closely related, palindromic 59-b elements (or recombination hot spots) located to the 3 side of the resistance gene (Fig. 3.1). Integrase-catalyzed insertion of resistance gene cassettes into resident integrons has been demonstrated (Collis et al., 1993; Martinez and de la Cruz, 1990). In addition, site-specific deletion and rearrangement of the inserted resistance gene cassettes can result from integrase-catalyzed events (Collis and Hall, 1992). Francia et al. (1993) have expanded our understanding of the role of integrons in gene mobilization by showing that the Tn21 integrase can act on secondary target sites at significant frequencies and so permit the fusion of two R plasmids by interaction between the recombination hot spot of one plasmid and a secondary integrase target site on a second plasmid. The secondary sites are characterized by the degenerate pentanucleotide sequence sequence Ga/tTNa/t. Though the details of the mechanism by which new integrons are then generated from the fusion structure are not established, the use of

Analyses of the integron-type transposons provide a good model for the way in which antibiotic resistance genes from various (unknown) sources may be incorporated into an integron by recombination events into mobile elements and hence into bacterial replicons, providing the R plasmids that we know today (Fig. 3.2) (Bissonnette and Roy, 1992). However, in bacterial pathogens a variety of transposable elements have been found that undergo different processes of recombinational excision and insertion. It is not known what evolutionary mechanisms are implicated or whether some form of integron-related structure is present in all cases. For the type of integron found in the Tn21 family, we have plausible models, supported by in vivo and in vitro studies, to provide a modus operandi by which antibiotic resistance genes were (and are) molecularly cloned in the evolution of R plasmids. A large number of transposable elements carrying virtually all possible combinations of antibiotic resistance genes have been identified (Berg, 1989), and nucleotide sequence analysis of multiresistant integrons shows that the inserted resistance gene cassettes differ markedly in codon usage, indicating that the antibiotic resistance determinants are of diverse origins. Microbes are masters at genetic engineering, and heterologous expression vectors of broad host range in the form of integrons were present in bacteria long before they became vogue for biotechnology companies in the 1980s.

V. ORIGINS We have described how the majority of antibiotic resistance genes found in microbes have been acquired by their hosts. The important question is, From where did they acquire these genetic determinants? The integron model defines a mechanism by which antibiotic genes can be procured by members of the Enterobacteriaceae and pseudomonads. This mechanism requires the participation of extrinsic resistance genes (or cassettes); however, the origins of these open reading frames are a mystery. The same questions can be applied to any of the resistance genes found in pathogenic bacteria. Available evidence concerning some of the origins is discussed below.

ch03.qxd 10/9/03 6:45 PM Page 40

40

the desk encyclopedia of microbiology

FIGURE 3.2 The diversity of antibiotic resistance integrons. The diagram illustrates the insertion of antibiotic resistance gene cassettes into the basic integron structure (Fig. 3.1). It should be noted that the antibiotic resistance gene cassettes are usually inserted in tandem array and more than five genes may be found in a single integron structure.

A. Antibiotic producers Antibiotic-producing microbes are the prime suspects for the maintenance of a pool of resistance genes in nature. Any organism producing a toxic molecule must, by definition, possess a mechanism(s) to survive this potentially suicidal situation. Because the majority of antibiotics are produced by bacteria (principally the actinomycete group), one would expect these organisms to have mechanisms of protection against the antibiotics they make. These mechanisms take various forms, and it is significant that the mechanisms of resistance for the known antibiotics in producing organisms and clinical isolates are biochemically identical (see Table 3.7). The gene clusters for antibiotic biosynthesis in producing organisms almost invariably comprise one or more genes that encode resistance to the antibiotic produced. Many of these genes have been cloned and sequenced. Sequence comparisons of the genes from the producer and the clinical isolate often show very high degrees of similarity. Thus, the homologous biochemical mechanisms and the relatedness of the gene sequences support the hypothesis that producing

TABLE 3.7 Resistance determinants with homologs in antibiotic producing organisms Antibiotic

Mechanism

Penicillins Cephalosporins Aminoglycosides

-Lactamases Penicillin binding proteins Phosphotransferases, acetyltransferases, nucleotidyltransferases Acetyltransferases Ribosomal protection, efflux rRNA methylation Esterases Phosphotransferases, acetyltransferases Phosphorylation, glutathionylation (?) Acetyltransferases, immunity protein

Chloramphenicol Tetracyclines Macrolides Streptogramins Lincosamides Phosphonates Bleomycin

organisms are likely to be the source of most resistance genes. However, other sources of resistance genes are not excluded, and some of these are discussed below. 1. Tetracyclines The first tangible evidence of resistance gene transfer involving antibiotic-producing streptomycetes in a

ch03.qxd 10/9/03 6:45 PM Page 41

antibiotic resistance in bacteria clinical setting has come from Pang et al. (1994). These researchers analyzed human infections of nontuberculous mycobacteria and Streptomyces spp.; the infections did not respond to treatment with tetracyclines. Both microbial species contained resistance genes (tetK and tetL) known to be the basis of tetracycline resistance in gram-positive bacteria (see Section II.A.1.b on tetracycline resistance determinants). These resistance determinants promote efflux of the drug and are typically transposon-associated. Surprisingly, the mycobacteria and the streptomycetes both had the tetracycline resistance genes (otrA and otrB) previously identified in the tetracycline-producing strain Streptomyces rimosis (Davies, 1992; Doyle et al., 1991). Reciprocally, the streptomycetes had acquired the tetK and tetL genes. Because the latter are clearly “foreign” genes (tetK and tetL have a G C content different from those of streptomycete and mycobacterial chromosomal DNA), they must have been acquired as the result of a recent gene transfer. Although this evidence is consistent with resistance gene transfer between the streptomycetes and other bacteria, it is not known which is donor and which is recipient, nor whether the newly acquired tetracycline resistance genes are plasmid or chromosomally encoded. 2. Aminoglycosides Covalent modification of their inhibitory biochemical products is very common in antibiotic-producing bacteria. It was the discovery of antibiotic modification as a means of self-protection in the streptomycetes that led to the proposal that antibiotic-producing microbes were the origins of the antibiotic resistance determinants found in other bacteria (Benveniste and Davies, 1973; Walker and Skorvaga, 1973). Support for this hypothesis has been provided by nucleic acid and protein sequence comparisons of aminoglycoside resistance determinants from producing organisms and clinical isolate sources (Shaw, 1984; Davies, 1992). As mentioned above, producing organisms are not the only potential source of antibiotic resistance mechanisms. The proposal that the enzymes that modify aminoglycosides evolved from such “housekeeping” genes as the sugar kinases and acyltransferases has been made by a number of groups (Udou et al., 1989; Shaw et al., 1992; Rather et al., 1993). 3. Macrolides The principal mechanism of resistance to the macrolide antibiotics involves methylation of the 23 S rRNA of the host. Methylation occurs on a specific adenine residue in the rRNA, and mono- and dimethylation has been described. The N-methyltransferase genes

41

responsible for encoding this protective modification have been studied in detail from both the producing organisms and clinical isolates (Rather et al., 1993). Although no direct transfer of resistance between producer and clinical isolates has been found, the identity of the biochemical mechanism and its regulation make for a compelling evolutionary relationship. 4. Other antibiotics Perhaps the most straightforward path toward the development of a drug resistance mechanism is via the major facilitator superfamily (MFS; see Section II.B.2) of transporters. The MFS is found in all organisms involved and consists of membrane transport systems involved in the symport, antiport, or uniport of various substrates. Other examples of MFS pumps include sugar uptake systems, phosphate ester/phosphate antiport, and oligosaccharide uptake. One might expect that, in addition to methods for the import of nutrients, a cell would have methods for the removal of harmful substances. The -lactamases are an interesting case because they are widely distributed among the bacterial kingdom. The ubiquity of -lactamases suggests that these enzymes may play a part in normal metabolic or synthetic processes, but an essential role has not been shown. Examination of the sequences of -lactamases by Bush et al. (1995) permitted the establishment of extensive phylogenetic relationships, which includes those from microbes employed in the commercial production of -lactam antibiotics. In the case of the glycopeptide antibiotics (vancomycin and teichoplanin), resistance in the enterococci is due to the acquisition of a cluster of genes that encode a novel cell wall precursor (see Section II.A.3). A comparison of D-Ala-D-Ala ligases from different sources has shown that the vanA gene is dissimilar to the other known genes, suggesting a divergent origin (Arthur et al., 1996). The vanA homolog from the producing organism, Streptomyces toyocaensis, has been cloned and sequenced. The predicted amino acid sequence was compared with the D-Ala-D-Ala ligase from Enterococcus and is greater than 60% similar (G. D. Wright et al., 1997).

B. Unknown origin There are gaps in our understanding of the origin of resistance determinants. For example, the aminoglycoside nucleotidyltransferases have been found only in clinical isolates and have no known relatives; the potential origins of the chloramphenicol acetyltransferase genes are still unclear (see Section II.A.1.c); and the sources for sulfonamide and trimethoprim

ch03.qxd 10/9/03 6:45 PM Page 42

42

the desk encyclopedia of microbiology

resistance are still not known (see Section II.A.4). There may be examples of “housekeeping” functions of resistance genes. For example, Providencia stuartii has a chromosomally encoded aminoglycoside acetyltransferase that may play a role in cell wall peptidoglycan formation (Payie et al., 1995).

VI. MAINTENANCE OF ANTIBIOTIC RESISTANCE A variety of surveys have indicated that normal healthy humans (who are not pursuing a course of antibiotic therapy) carry antibiotic-resistant enteric species in their intestinal tract; a substantial proportion are found to contain transmissible antibiotic resistance plasmids. Studies have demonstrated that a lack of antibiotic selective pressure, for example, removing antimicrobials from cattle feed, can lead to a gradual decrease in the percentage of resistance genes and resistance bacteria found in a population (Langlois et al., 1986; Hintone et al., 1985). Although the results of these studies are encouraging, other studies indicate that once resistance cassettes have been developed it is unlikely that they will disappear completely from an environment where antibiotics are routinely used. In a sense “the cat is out of the bag.” Chemostat studies (Chao et al., 1983; Hartl et al., 1983) suggest that insertion elements may themselves provide their hosts with a selective advantage independent of the resistance determinants they carry. Roberts has postulated that commensal bacteria can be reservoirs for tetracycline resistance determinants. When bacteria from the urogenital tracts of females who had not taken antibiotics for 2 weeks previously were examined, 82% of the viridans-type streptococci hybridized with at least one Tet determinant (Roberts et al., 1991).

A. Multiple antibiotic resistance and mercury resistance Although the cooccurrence of antibiotic resistance and resistance to heavy metals such as mercury has long been known, its implications for public health are only now becoming clear. In 1964, the cotransduction of genes encoding resistance to penicillin and mercury by a staphylococcal phage was reported (Richmond and John, 1964). Ten years later it was found that 25% of the antibiotic resistance plasmids isolated from enteric bacteria in Hammersmith Hospital also carried mercury resistance (Schottel et al., 1974). DNA sequence analyses has shown that the Tn21-type transposons carry both a copy of the mer

locus and an integron (see above) (Stokes and Hall, 1989; Grinsted et al., 1990). Summers and co-workers (1993) have observed that resistance to mercury occurs frequently in human fecal flora and is correlated with the occurrence of multiple antibiotic resistance. How does this phenomenon become a public health concern? In the same report Summers’ group found that the mercury released from the amalgam in “silver” dental fillings in monkeys led to the rapid enrichment of many different mercury-resistant bacteria in the oral and fecal flora. They suggest that in humans this chronic and biologically significant exposure to mercury may foster the persistence of multidrug-resistant microbes through selection of linked markers.

B. Other examples There may well be other examples of this phenomenon, where subclinical concentrations of an antibiotic could serve as selection for the maintenance (and propagation) of genetic elements and their resident resistance genes. There is also maintenance by constant selection pressure for other phenotypes, a type of “linked” selection (we have mentioned the role of mercury in this respect). How else does one explain the fact that streptomycin and chloramphenicol resistance can be still found on plasmids in hospitals, even though these antibiotics are no longer used? Are there other positive selective functions carried by plasmids? Resistance to ultraviolet light or other physical or chemical toxins (e.g. detergents, disinfectants) would be a possibility. Is it also conceivable that plasmids improve the fitness of their hosts under “normal” conditions? Evidence for this comes from the work of Lenski (personal communication, 1996).

VII. CONCLUDING REMARKS— FOR NOW AND THE FUTURE It should be apparent from the foregoing discussion of antibiotic modification that there must be a substantial pool of antibiotic resistance genes (or close relatives of these genes) in nature. Gene flux between bacterial replicons and their hosts is likely to be the rule rather than the exception, and it appears to respond quickly to environmental changes (Levy and Novick, 1986; Levy and Miller, 1989; Hughes and Datta, 1983). This gene pool is readily accessible to bacteria when they are exposed to the strong selective pressure of antibiotic usage—in hospitals, for veterinary and agricultural purposes, and as growth promoters in animal and poultry husbandry. It is a life-or-death situation for microbes, and they have survived. A better knowledge

ch03.qxd 10/9/03 6:45 PM Page 43

43

antibiotic resistance in bacteria of the components of this gene pool, particularly with respect to what might happen on the introduction of a new chemical entity such as an antimicrobial agent, might, on the one hand, permit early warning and subsequent chemical modification of antibiotics to permit them to elude potential resistance mechanisms and, on the other, lead to more prudent use of antibiotics under circumstances where the presence of specific resistance determinants can be predicted. The development of resistance to antimicrobial agents is inevitable, in response to the strong selective pressure and extensive use of antibiotics. Resistance may develop as the result of mutation or acquisition, or a combination of the two. The use of antibiotics should be such as to delay the inevitable, and knowledge gained over the past 50 years if correctly interpreted and used to modify current practices appropriately should permit this. Unfortunately, for all of the antimicrobial agents in current use, we have reached a state of no return. The American Society for Microbiology Task Force on Antibiotic Resistance has made a number of recommendations to deal with the current crisis of antibiotic resistant microbes (see Table 3.8; ASM, TABLE 3.8 Recommendations from the American Society for Microbiology Task Force on Antibiotic Resistance Establish a national surveillance system immediately. Lead agency should be the National Center for Infectious Diseases of the U.S. Centers for Disease Control (CDC) and should involve the National Institute of Allergy and Infectious Disease of the National Institutes of Health (NIH), the Environmental Protection Agency, and the Food and Drug Administration. Strengthen professional and public education in the area of infectious disease and antibiotics to reduce inappropriate usage of antibiotics. The curriculum for health professionals should include the appropriate handling, diagnosis, and treatment of infectious disease and antibiotic resistance. Reduce the spread of infectious agents and antibiotic resistance in hospitals, nursing homes, day care facilities, and food production industries. Educate patients and food producers. Improve antimicrobial use for cost-effective treatment and preservation of effectiveness. Increase basic research directed toward development of new antimicrobial compounds, effective vaccines, and other prevention measures. Fund areas directly related to new and emerging infections and antibiotic resistance. Fund basic research in bacterial genetics and metabolic pathways. Establish a culture collection containing representative antibiotic-resistance biotypes of pathogens. Sequence genomes of microbial pathogens. Develop better diagnostic techniques. Develop vaccines and other preventative measures.

1995). It has been suggested that such measures can, at the least, maintain and improve the status quo. Two of the options that would permit continued success of antibiotic therapy in the face of increasing resistance are: (a) the discovery of new antibiotics (by “new” this implies novel chemical structures) and (b) the development of agents, that might be used in combination with existing antibiotics, to interfere with the biochemical resistance mechanisms. Such a strategy has already been partially successful in the development of inhibitors of -lactamases to permit “old” antibiotics to be used. However, there appears to be little success (or effort) in applying this approach to other antibiotic classes. In our discussion of biochemical mechanisms (Section II) we have noted a number of cases where this approach could be taken (e.g. fluorinated analogs of chloramphenicol). With respect to novel antibiotics, several valid approaches exist: (a) natural product screening, especially directed at products of the 99.9% of microbes that cannot be grown in the laboratory; (b) combinatorial chemistry that can be used as a means of discovery of new active molecules or to new substitutions on known ring structures; and (c) rational chemical design based on identification of specific biochemical targets. The success of these methods will depend on the availability of cellbased and biochemical assays that will detect low concentrations of active molecules by high flux screening methods. It seems redundant to insist on the requirement for early identification of natural resistance mechanisms for any compounds of interest (thus permitting the design of analogs) and the study of structure– toxicity relationships at the earliset stage possible. Last, but not least, the introduction of a novel antimicrobial agent into clinical practice must be accompanied by strict limitations on its use. No novel therapeutic should be used for other than human use under prescription, and no structural analog should be employed for “other” purposes.

ACKNOWLEDGMENTS We thank the Canadian Bacterial Diseases Network and the Natural Sciences and Engineering Council of Canada for support.

REFERENCES Aminoglycoside Resistance Study Group. (1994). Resistance to aminoglycosides in Pseudomonas. Trends Microbiol. 2 (10), 347–353. Arca, P., Hardisson, C., and Suárez, J. E. (1990). Purification of a glutathione S-transferase that mediates fosfomycin resistance in bacteria. Antimicrob. Agents Chemother. 34, 844–848.

ch03.qxd 10/9/03 6:45 PM Page 44

44

the desk encyclopedia of microbiology

Arthur, M., Brisson-Noël, A., and Courvalin, P. (1987). Origin and evolution of genes specifying resistance to macrolide, lincosamide and streptogramin antibiotics: Data and hypothesis. J. Antimicrob. Chemother. 20, 783–802. Arthur, M., Reynolds, P., and Courvalin, P. (1996). Glycopeptide resistance in gram-positive bacteria. Trends Microbiol. 4, 401–407. ASM. (1995). Report of the ASM Task Force on antibiotic resistance. Antimicrob. Agents Chemother. (Suppl.), 123. Bannam, T. L., and Rood, J. I. (1991). Relationship between the Clostridium perfringens catQ gene product and chloramphenicol acetyltransferases from other bacteria. Antimicrob. Agents Chemother. 35, 471–476. Bennett, P. M., and Chopra, I. (1993). Molecular basis of -lactamase induction in bacteria. Antimicrob. Agents Chemother. 37, 153–158. Benveniste, R., and Davies, J. (1973). Aminoglycoside antibioticinactivating enzymes in actinomycetes similar to those present in clinical isolates of antibiotic-resistant bacteria. Proc. Natl. Acad. Sci. USA 70, 2276–2280. Berg, D. E. (1989). Transposable elements in prokaryotes. In “Gene Transfer in the Environment” (S. B. Levy and R. V. Miller, Eds.), pp. 99–137. McGraw-Hill, New York. Bergh, O., Borsheim, K. Y., Bratbak, G., and Heldal, M. (1989). High abundance of viruses found in aquatic environments. Nature (London) 340, 467–468. Bissonnette, L., and Roy, P. H. (1992). Characterization of In0 of Pseudomonas aeruginosa plasmid pVS1, an ancestor of integrons of multiresistance plasmids and transposons of gram-negative bacteria. J. Bacteriol. 174, 1248–1257. Bissonnette, L., Champetier, S., Buisson, J.-P., and Roy, P. H. (1991). Characterization of the nonenzymatic chloramphenicol resistance (cmlA) gene of the In4 integron of Tn1696: Similarity of the product to transmembrane transport proteins. J. Bacteriol. 173, 4493–4502. Blazquez, J., Baquero, M.-R., Canton, R., Alos, I., and Baquero, F. (1993). Characterization of a new TEM-type -lactamase resistant to clavulanate, sulbactam, and tazobactam in a clinical isolate of Escherichia coli. Antimicrob. Agents Chemother. 37, 2059–2063. Brisson-Noël, A., Delrieu, P., Samain, D., and Courvalin, P. (1988). Inactivation of lincosaminide antibiotics in Staphylococcus. J. Biol. Chem. 263, 15880–15887. Bush, K., Jacoby, G. A., and Medeiros, A. A. (1995). A functional classification scheme for -lactamases and its correlation with molecular structure. Antimicrob. Agents Chemother. 39, 1211–1233. Cannon, M., Harford, S., and Davies, J. (1990). A comparative study on the inhibitory actions of chloramphenicol, thiamphenicol and some fluorinated derivatives. J. Antimicrob. Chemother. 26, 307–317. Chao, L., Vargas, C., Spear, B. B., and Cox, E. C. (1983). Transposable elements as mutator genes in evolution. Nature (London) 303, 633–635. Chen, S.-T., and Clowes, R. C. (1987). Variations between the nucleotide sequences of Tn1, Tn2, and Tn3 and expression of -lactamase in Pseudomonas aeruginosa and Escherichia coli. J. Bacteriol. 169, 913–916. Cole, S. T. (1994). Mycobacterium tuberculosis: Drug-resistance mechanisms. Trends Microbiol. 10 (2), 411–415. Collis, C. M., and Hall, R. M. (1992). Site-specific deletion and rearrangement of integron insert genes catalysed by the integron DNA integrase. J. Bacteriol. 174, 1574–1585. Collis, C. M., Grammaticopoulos, G., Briton, J., Stokes, H. W., and Hall, R. M. (1993). Site-specific insertion of gene cassettes into integrons. Mol. Microbiol. 9, 41–52. Cookson, B. (1995). Aspects of the epidemiology of MRSA in Europe. J. Chemother. 7 (Suppl. 3), 93–98.

Couture, F., Lachapelle, J., and Levesque, R. C. (1992). Phylogeny of LCR-1 and OXA-5 with class A and class D -lactamases. Mol. Microbiol. 6, 1693–1705. Dabbs, E. R., Yazawa, K., Mikami, Y., Miyaji, M., Morisaki, N., Iwasaki, S., and Furihata, K. (1995). Ribosylation by mycobacterial strains as a new mechanism of rifampin inactivation. Antimicrob. Agents Chemother. 39, 1007–1009. Datta, N., and Hughes, V. M. (1983). Plasmids of the same Inc groups in enterobacteria before and after the medical use of antibiotics. Nature (London) 306, 616–617. Davies, J. (1992). Another look at antibiotic resistance. J. Gen. Microbiol. 138, 1553–1559. Day, P. J., and Shaw, W. V. (1992). Acetyl coenzyme A binding by chloramphenicol acetyltransferase: Hydrophobic determinants of recognition and catalysis. J. Biol. Chem. 267, 5122–5127. Debets-Ossenkopp, Y., Sparrius, M., Kusters, J., Kolkman, J., and Vandenbroucke-Grauls, C. (1996). Mechanism of clarithromycin resistance in clinical isolates of Helicobacter pylori. FEMS Microbiol. Lett. 142, 37–42. Doyle, D., McDowall, K. J., Butler, M. J., and Hunter, I. S. (1991). Characterization of an oxytetracycline-resistance gene, otrA, of Streptomyces rimosus. Mol. Microbiol. 5, 2923–2933. Ferretti, J. J., Gilmore, K. S., and Courvalin, P. (1986). Nucleotide sequence analysis of the gene specifying the bifunctional 6-aminoglycoside acetyltransferase 2 -aminoglycoside phosphotransferase enzyme in Streptococcus faecalis and identification and cloning of gene regions specifying the two activities. J. Bacteriol. 167, 631–638. Finken, M., Kirschner, P., Meier, A., Wrede, A., and Böttger, E. C. (1993). Molecular basis of streptomycin resistance in Mycobacterium tuberculosis: Alterations of the ribosomal protein S12 gene and point mutations within a functional 16S ribosomal RNA pseudoknot. Mol. Microbiol. 9, 1239–1246. Fourmy, D., Recht, M. I., Blanchard, S. C., and Puglisi, J. D. (1996). Structure of the A site of Escherichia coli 16S ribosomal RNA complexed with an aminoglycoside. Science 274, 1367–1371. Francia, M. V., de la Cruz, F., and García Lobo, J. M. (1993). Secondary sites for integration mediated by the Tn21 integrase. Mol. Microbiol. 10, 823–828. George, A., and Levy, S. B. (1983) Amplifiable resistance to tetracycline, chloramphenicol, and other antibiotics in Escherichia coli: involvement of a non-plasmid-determined efflux of tetracycline. J. Bacteriol. 155, 531–540. Goldman, J. D., White, D. G., and Levy, S. B. (1996). Multiple antibiotic resistance (mar) locus protects Escherichia coli from rapid cell killing by fluoroquinolones. Antimicrob. Agents Chemother. 40, 1266–1269. Grinsted, J., de la Cruz, F., and Schmitt R. (1990). The Tn21 subgroup of bacterial transposable elements. Plasmid, 24, 163–189. Hartl, D. L., Dykhuizen, D. E., Miller, R. D., Green, L., and de Framond, J. (1983). Transposable element IS50 improves growth rate of E. coli cells without transposition. Cell (Cambridge, Mass.) 35, 503–510. Hillen, W., and Berens, C. (1994). Mechanisms underlying expression of Tn10 encoded tetracycline resistance. Annu. Rev. Microbiol. 48, 345–369. Hinton, M., Linton, A. H., and Hedges, A. J. (1985). The ecology of Escherichia coli in calves reared as dairy-cow replacements. J. Appl. Bacteriol. 85, 131–138. Honoré, N., Nicolas, M.-H., and Cole, S. T. (1986). Inducible cephalosporinase production in clinical isolates of Enterobacter cloacae is controlled by a regulatory gene that has been deleted from Escherichia coli. EMBO J. 5, 3709–3714.

ch03.qxd 10/9/03 6:45 PM Page 45

antibiotic resistance in bacteria Hughes, V. M., and Datta, N. (1983). Conjugative plasmids in bacteria of the ‘pre-antibiotic’ era. Nature (London) 302, 725–726. Huovinen, P., Sundström, L., Swedberg, G., and Sköld, O. (1995). Trimethoprim and sulfonamide resistance. Antimicrob. Agents Chemother. 39, 279–289. Jacoby, G. A., and Archer, G. L. (1991). New mechanisms of bacterial resistance to antimicrobial agents. N. Engl. J. Med. 324, 601–612. Jarlier, V., and Nikaido, H. (1994). Mycobacterial cell wall: Structure and role in natural resistance to antibiotics. FEMS Microbiol. Lett. 123, 11–18. Kahn, M., and Smith, H. O. (1986). Role of transformazomes in Haemophilus influenzae Rd transformation. In “Antibiotic Resistance Genes: Ecology, Transfer, and Expression” (S. B. Levy and R. P. Novick, Eds.), pp. 143–152. Cold Spring Harbor Laboratory, Cold Spring Harbor, New York. Kocabiyik, S., and Perlin, M. H. (1992). Altered substrate specificity by substitutions at Tyr218 in bacterial aminoglycoside 3-phosphotransferase-II. FEMS Microbiol. Lett. 93, 199–202. Kokjohn, T. A. (1989). Transduction: Mechanism and potential for gene transfer in the environment. In “Gene Transfer in the Environment” (S. B. Levy and R. V. Miller, Eds.), pp. 73–97. McGraw-Hill, New York. Krause, R. M. (1994). Dynamics of emergence. J. Infect. Dis. 170, 265–271. Langlois, B. E., Dawson, K. A., Cromwell, G. L., and Stahly, T. S. (1986). Antibiotic resistance in pigs following a 13 year ban. J. Anim. Sci. 62, 18–32. LeClerc, J. E., Li, B., Payne, W. L., and Cebula, T. A. (1996). High mutation frequencies among Escherichia coli and Salmonella pathogens. Science 274, 1208–1211. Leclercq, R., and Courvalin, P. (1991). Bacterial resistance to macrolide, lincosamide, and streptogramin antibiotics by target modification. Antimicrob. Agents Chemother. 35, 1267–1272. Lévesque, C., Brassard, S., Lapointe, J., and Roy, P. H. (1994). Diversity and relative strength of tandem promoters for the antibiotic-resistance genes of several integrons. Gene 142, 49–54. Levy, S. B., and Miller, R. V., eds. (1989). Gene transfer in the environment. In “Environmental Biotechnology.” McGraw-Hill, New York. Levy, S. B., and Novick, R. P., eds. (1986). “Antibiotic Resistance Genes: Ecology, Transfer, and Expression.” Banbury Report 24. Cold Spring Harbor Laboratory, Cold Spring Harbor, New York. Lewis, K. (1994). Multidrug resistance pumps in bacteria: Variations on a theme. Trends Biochem. Sci. 19, 119–123. Mabilat, C., Goussard, S., Sougakoff, W., Spencer, R. C., and Courvalin, P. (1990). Direct sequencing of the amplified structural gene and promoter for the extended-broad-spectrum lactamase TEM-9 (RHH-1) of Klebsiella pneumoniae. Plasmid 23, 27–34. Marger, M. D., and Saier, M. H. (1993). A major superfamily of transmembrane facilitators that catalyse uniport, symport and antiport. Trends Biochem. Sci. 18, 13–20. Martinez, E., and de la Cruz, F. (1990). Genetic elements involved in Tn21 site-specific integration, a novel mechanism for the dissemination of antibiotic resistance genes. EMBO J. 9, 1275–1281. Miller, P. F., and Sulavik, M. C. (1996). Overlaps and parallels in the regulation of intrinsic multiple-antibiotic resistance in Escherichia coli. Mol. Microbiol. 21, 441–448. Monod, M., Mohan, S., and Dubnau, D. (1987). Cloning and analysis of ermG, a new macrolide-lincosamide-streptogramin B resistance element from Bacillus sphaericus. J. Bacteriol. 169, 340–350.

45

Murray, I. A., and Shaw, W. V. (1997). O-Acetyltransferases for chloramphenicol and other natural products. Antimicrob. Agents Chemother. 41, 1–6. Needham, C., Rahman, M., Dyke, K. G. H., and C., N. W., Noble, W. C. (1994). An investigation of plasmids from Staphylococcus aureus that mediate resistance to mupirocin and tetracycline. Microbiology 140, 2577–2583. Neu, H. C. (1992). The crisis in antibiotic resistance. Science 257, 1064–1073. Nikaido, H. (1994). Prevention of drug access to bacterial targets: Permeability barriers and active efflux. Science 264, 382–387. Nikaido, H. (1996). Multidrug efflux pumps of gram-negative bacteria. J. Bacteriol. 178, 5853–5859. Nikolich, M. P., Hong, G., Shoemaker, N. B., and Salyers, A. A. (1994). Evidence for natural horizontal transfer of tetQ between bacteria that normally colonize humans and bacteria that normally colonize livestock. Appl. Environ. Microbiol. 60, 3255–3260. Noble, W. C., Rahman, M., and Cookson, B. (1988). Transferable mupirocin resistance. J. Antimicrob. Chemother. 22, 771. Noble, W. C., Virani, Z., and Cree, R. G. A. (1992). Co-transfer of vancomycin and other resistance genes from Enterococcus faecalis NCTC 12201 to Staphylococcus aureus. FEMS Microbiol. Lett. 93, 195–198. O’Hara, K., Kanda, T., Ohmiya, K., Ebisu, T., and Kono, M. (1989). Purification and characterization of macrolide 2-phosphotransferase from a strain of Escherichia coli that is highly resistant to erythromycin. Antimicrob. Agents Chemother. 33, 1354–1357. Olsen, R. H., and Shipley, P. (1973). Host range and properties of the Pseudomonas aeruginosa R factor R 1822. J. Bacteriol. 113, 772–780. Olsen, R. H., Siak, J., and Gray, R. H. (1974). Characteristics of PrD1, a plasmid dependent broad host range DNA bacteriophage. J. Virol. 14, 689–699. Pang, Y., Brown, B. A., Steingrube, V. A., Wallace, R. J., Jr., and Roberts, M. C. (1994). Tetracycline resistance determinants in Mycobacterium and Streptomyces species. Antimicrob. Agents Chemother. 38, 1408–1412. Parent, R., and Roy, P. H. (1992). The chloramphenicol acetyltransferase gene of Tn2424: A new breed of cat. J. Bacteriol. 174, 2891–2897. Paulsen, I. T., Brown, M. H., and Skurray, R. A. (1996). Protondependent multidrug efflux systems. Microbiol Rev. 60, 575–608. Payie, K. G., Rather, P. N., and Clarke, A. J. (1995). Contribution of gentamicin 2-N-acetyltransferase to the O-acetylation of peptidoglycan in Providencia stuartii. J. Bacteriol. 177, 4303–4310. Rahman, M., Noble, W. C., and Dyke, K. G. H. (1993). Probes for the study of mupirocin resistance in staphylococci. J. Med. Microbiol. 39, 446–449. Rather, P. N., Munayyer, H., Mann, P. A., Hare, R. S., Miller, G. H., and Shaw, K. J. (1992). Genetic analysis of bacterial acetyltransferases: Identification of amino acids determining the specificities of the aminoglycoside 6-N-acetyltransferase Ib and IIa proteins. J. Bacteriol. 174, 3196–3203. Rather, P. N., Orosz, E., Shaw, K. J., Hare, R., and Miller, G. (1993). Characterization and transcriptional regulation of the 2-Nacetyltransferase gene from Providencia stuartii. J. Bacteriol. 175, 6492–6498. Recchia, G. D., and Hall, R. M. (1995). Gene cassettes: A new class of mobile element. Microbiology 141, 3015–3027. Richmond, M. H., and John, M. (1964). Co-transduction by a staphylococcal phage of the genes responsible for penicillinase synthesis and resistance to mercury salts. Nature (London) 202, 1360–1361.

ch03.qxd 10/9/03 6:45 PM Page 46

46

the desk encyclopedia of microbiology

Roberts, M. C. (1994). Epidemiology of tetracycline-resistance determinants. Trends Microbiol. 2 (10), 353–357. Roberts, M. C., Pang, Y. J., Spencer, R. C., Winstanley, T. G., Brown, B. A., and Wallance, R. J. (1991). Tetracycline resistance in Moraxella (Branhamella) catarrhalis: Demonstration of two clonal out breaks by using pulsed-field gel electrophoresis. Antimicrob. Agents Chemother. 35, 2453–2455. Rouch, D. A., Byrne, M. E., Kong, Y. C., and Skurray, R. A. (1987). The aacA–aphD gentamicin and kanamycin resistance determinant of Tn4001 from Staphylococcus aureus: Expression and nucleotide sequence analysis. J. Gen. Microbiol. 133, 3039–3052. Salyers, A. A. (1993). Gene transfer in the mammalian intestinal tract. Curr. Opin. Biotechnol. 4, 294–298. Sanchez-Pescador, R., Brown, J. T., Roberts, M., and Urdea, M. S. (1988). Homology of the TetM with translational elongation factors: Implications for potential modes of tetM conferred tetracycline resistance. Nucleic Acids. Res. 16, 12–18. Schottel, J., Mandal, A., Clark, D., Silver, S., and Hedges, R. W. (1974). Volatilisation of mercury and organomercurials determined by inducible R-factor systems in enteric bacteria. Nature (London) 251, 335–337. Scott, J. R. (1992). Sex and the single circle: Conjugative transposition. J. Bacteriol. 174, 6005–6010. Shaw, K. J., Rather, P. N., Sabatelli, F. J., Mann, P., Munayyer, H., Mierzwa, R., Petrikkos, G. L., Hare, R. S., Miller, G. H., Bennett, P., and Downey, P. (1992). Characterization of the chromosomal aac(6)-Ic gene from Serratia marcescens. Antimicrob. Agents Chemother. 36, 1447–1455. Shaw, K. J., Rather, P. N., Hare, R. S., and Miller, G. H. (1993). Molecular genetics of aminoglycoside resistance genes and familial relationships of the aminoglycoside-modifying enzymes. Microbiol. Rev. 57, 138–163. Shaw, W. V. (1984). Bacterial resistance to chloramphenicol. Br. Med. Bull. 40, 36–41. Sheridan, R. P., and Chopra, I. (1991). Origin of tetracycline efflux proteins: Conclusions from nucleotide sequence analysis. Mol. Microbiol. 5, 895–900. Shoemaker, N. B., Smith, M. D., and Guild, W. R. (1980). DNase resistant transfer of chromosomal cat and tet insertions by filter mating in pneumococcus. Plasmid 3, 80–87. Stewart, G. J. (1989). The mechanism of natural transformation. In “Gene Transfer in the Environment” (S. B. Levy and R. V. Miller, Eds.), pp. 139–163. McGraw-Hill, New York. Stokes, H. W., and Hall, R. M. (1989). A novel family of potentially mobile DNA elements encoding site-specific gene-integration functions: Integrons. Mol. Microbiol. 3, 1669–1683. Stotzky, G. (1989). Gene transfer among bacteria in soil. In “Gene Transfer in the Environment” (S. B. Levy and R. V. Miller, Eds.), pp. 165–221. McGraw-Hill, New York.

Suárez, J. E., and Mendoza, M. C. (1991). Plasmid-encoded fosfomycin resistance. Antimicrob. Agents Chemother. 35, 791–795. Summers, A. O., Wireman, J., Vimy, M. J., Lorscheider, F. L., Marshall, B., Levy, S. B., Bennet, S., and Billard, L. (1993). Mercury released from dental “silver” fillings provokes an increase in mercury- and antibiotic-resistant bacteria in oral and intestinal floras of primates. Antimicrob. Agents Chemother. 37, 825–834. Taylor, D. E., and Chau, A. (1996). Tetracycline resistance mediated by ribosomal protection. Antimicrob. Agents Chemother. 40, 15. Timmermann, K. P. (1988). Physiol. Plant. 77, 465–471. Udo, E. E., Pearman, J. W., and Grubb, W. B. (1994). Emergence of high-level mupirocin resistance in methicillin-resistant Staphylococcus aureus in western Australia. J. Hospital Infect. 26, 157–165. Udou, T., Mizuguchi, Y., and Wallace, R. J., Jr. (1989). Does aminoglycoside-acetyltransferase in rapidly growing mycobacteria have a metabolic function in addition to aminoglycoside inactivation? FEMS Microbiol. Lett. 57, 227–230. Walker, J. B., and Skorvaga, M. (1973). Phosphorylation of streptomycin and dihydrostreptomycin by Streptomyces. Enzymatic synthesis of different diphosphorylated derivatives. J. Biol. Chem. 248, 2435–2440. Walsh, C. T., Fisher, S. L., Park, I.-S., Prahalad, M., and Wu, Z. (1996). Bacterial resistance to vancomycin: Five genes and one missing hydrogen bond tell the story. Curr. Biol. 3, 21–28. Weisblum, B. (1995). Insights into erythromycin action from studies of its activity as inducer of resistance. Antimicrob. Agents Chemother. 39, 797–805. Zakrzewska-Bode, A., Muytjens, H. L., Liem, K. D., and Hoogkamp-Korstanje, J. A. (1995). Mupirocin resistance in coagulase-negative staphylococci, after topical prophylaxis for the reduction of colonization of central venous catherters. J. Hospital Infect. 31, 189–193. Zilhao, R., and Courvalin, P. (1990). Nucleotide sequence of the fosB gene conferring fosfomycin resistance in Staphylococcus epidermidis. FEMS Microbiol. Lett. 68, 267–272.

WEBSITE Website of the CDC National Center for Infectious diseases http://www.cdc.gov/drugresistance/

ch04.qxd 10/13/03 3:17 PM Page 47

4 Antifungal agents Ana Espinel-Ingroff Medical College of Virginia Commonwealth University

GLOSSARY

humans. Most fungi associated with disease are considered opportunistic pathogens (especially the yeasts) because they live as normal flora in humans, lower animals, and plants and rarely cause disease in otherwise healthy individuals. Many fungi, on the other hand, are important plant and lower animal parasites and can cause damage to crops (wheat rust, corn smut, etc.) and to fruit (banana wilt), forest (Dutch elm disease), and ornamental trees and other plants. Historically, the potato famine, which was the reason for the great migration from Ireland to the Americas, was caused by a fungal infection (potato blight). At the same time, fungi and their products play an important economic role in the production of alcohol, certain acids, steroids, antibiotics, etc. Due to the high incidence of toxicity among antifungal agents and the perception before the 1970s that the number of severe and invasive infections was low, only 12 antifungal agents are currently licensed for the treatment of systemic fungal infections: the polyene amphotericin B and its three lipid formulations, the pyrimidine synthesis inhibitor 5-fluorocytosine (flucytosine), the imidazoles miconazole and ketoconazole, the triazoles fluconazole, itraconazole and voriconazole, and the echinocandin caspofungin. However, the number of fungal diseases caused by both yeasts and molds has significantly increased during the past 20 years, especially among the increased number of immunocompromised patients, who are at high risk for life-threatening mycoses.

emerging fungal infections Fungal infections caused by new or uncommon fungi. granulocytopenia/neutropenia Acquired or chemically induced immunosuppression caused by low white blood cell counts. immunocompromised Having a defect in the immune system. in vitro and in vivo Describing or referring to studies carried out in the test tube and in animals, respectively. mycoses and mycotic infections Diseases caused by yeasts or molds. nephrotoxicity Damage to the kidney cells. opportunistic infections Infections caused by saprophytic fungi or not true parasites. Antifungal Agents are naturally occurring or synthetically produced compounds that have in vitro or in vivo activity against yeasts, molds, or each. Fungi and mammalian cells are eukaryotes, and antifungal agents that inhibit synthesis of proteins, RNA, and DNA are potentially toxic to mammalian cells. Fungi can be unicellular (yeasts) and multicellular or filamentous (molds) microorganisms. Some medically important fungi can exist in both of these morphologic forms and are called dimorphic fungi. Of the estimated 250 000 fungal species described, fewer than 150 are known to be etiologic agents of disease in The Desk Encyclopedia of Microbiology ISBN: 0-12-621361-5

47

Copyright © 2003 Elsevier Ltd All rights of reproduction in any form reserved

ch04.qxd 10/13/03 3:17 PM Page 48

48

the desk encyclopedia of microbiology

There are more antifungal agents for topical treatment and agriculture and veterinary use, and several agents are under investigation for the management of severe and refractory fungal infections in humans (Table 4.1).

This article summarizes the most relevant facts regarding the chemical structure, mechanisms of action and resistance, pharmacokinetics, safety, adverse interactions with other drugs and applications of the established systemic and topical antifungal

TABLE 4.1 Antifungal agents, mechanisms of action, and their usea Antifungal class Polyenes

Antifungal target of action Membranes containing ergosterol

Phenolic benzyfuran cyclo-hexane Natural glutarimide Phenylpyrroles

Microtubule aggregation and DNA inhibition Protein synthesis inhibition Unknown

Synthetic pyrimidines

Fungal cytosine permeae and deaminase Ergosterol inhibition

Anilinopyrimidines

Enzyme secretion

Azoles

Ergosterol biosynthesis inhibition

Agent

Use

Amphotericin B (AMB) Nystatin (NYS) AMB lipid complex AMB colloidal dispersion Liposomal AMB Liposomal NYS Pimaricin Griseofulvin

Systemic mycosesb,c Superficial mycosesb,c Systemic mycoses intolerant or refractory to AMB

Cycloheximide Fenpiclonil Fludioxonil Flucytosine Triarimol Fenarimol Pyrimethanil Cyprodinil Imidazoles Clotrimazole Econazole Isoconazole Oxiconazole Tioconazole Miconazole Ketoconazole Enilconazole Epoxiconazole Fluquinconazole Triticonazole Prochoraz Triazoles Fluconazole Itraconazole Terconazole Voriconazole

Allylamines Benzylamines Thiocarbamates

Dithiocarbamates

Non-specific

Posaconazole Ravuconazole Terbinafine Naftifine Butenafine Tolnaftate Tolciclate Piritetrade Mancozeb Thiram

Under investigation Topical keratitisb,c Dermatophytic infectionsb Laboratory and agriculture Agriculture Systemic (yeasts) in combination with AMBb,c Agriculture

Topical, oral trocheb,c

P. boydii infections only and veterinaryc Secondary alternative to other agents and veterinaryb,c Veterinary Agriculture

Certain systemic and superficial diseasesb,c Intravaginal Treatment of acute aspergillosis and salvage therapy for serious fungal infections Under Investigation Under Investigation Superficial infections Topical Topical Topical

Agriculture

ch04.qxd 10/13/03 3:17 PM Page 49

49

antifungal agents TABLE 4.1 (Continued) Benzimidazoles and methylbenz-imidazole carbamates Morpholines

Nuclear division

Ergosterol biosynthesis inhibition

Pyridines Fungal (1,3)-glucan synthesis inhibition

Echinocandins

Carbendazim Benomyl Thiophanate Amorolfine Fenpropimorph Tridemorph Buthiobate Pyrifenox Papulocandins Caspofungin

Pradimicins

Fungal sacharide (mannoproteins)

Benanomycins Polyoxins

Fungal chitin synthase inhibition

Nikkomycins Sordarins

Protein synthesis inhibition

Cinnamic acid Oomycete fungicide Phthalimides

Cell wall Oxidative phosphorylation Non-specific

Cationic peptides

Lipid bilayer of biological membranes

Amino acid analogs

Amino acid synthesis interference

Anidulafungin Pradamicin FA-2 (BMY 28864) Benanomycin A Polyoxin D Nikkomycin Z GM 222712 GM 237354 GM 211676 GM 193663 Dimethomorph Fluazynam Captan Captafol Folpet Natural peptides Cecropin Indolicidin Synthetic peptides RI 331 Azoxybacillins Cispentacin

Agriculture

Topical Agriculture Agriculture None Treatment of candidemia and refractory aspergillosis Under investigation Under investigation Under investigation None Under investigation Under investigation

Agriculture Agriculture Agriculture

Under investigation Under investigation Under investigation

a

Only licensed, commonly used, and antifungals under clinical investigation are listed; see text for other antifungals. Clinical and veterinary use; other applications for use in humans only. c A human product used in veterinary practice. b

agents currently licensed for clinical, veterinary, or agricultural uses. A shorter description is provided for antifungal compounds that are in the last phases of clinical development, under clinical trials in humans, or that have been discontinued from additional clinical evaluation. The former compounds have potential use as therapeutic agents. More detailed data regarding these agents are found in the references.

I. THE POLYENES The polyenes are macrolide molecules that target membranes containing ergosterol, which is an important

sterol in the fungal cell membranes. Traces of ergosterol are also involved in the overall cell cycle of fungi.

A. Amphotericin B Amphotericin B is the most important of the 200 polyenes. Amphotericin B replaced 2-hydroxystilbamidine in the treatment of blastomycosis in the mid-1960s. Two amphotericins (A and B) were isolated in the 1950s from Streptomyces nodosus, an aerobic bacterium, from a soil sample from Venezuela’s Orinoco River Valley. Amphotericin B (the most active molecule) has seven conjugated double bounds, an internal ester, a free carboxyl group, and a glycoside side-chain with a primary amino group (Fig. 4.1A). It is unstable to

ch04.qxd 10/13/03 3:17 PM Page 50

50

the desk encyclopedia of microbiology

FIGURE 4.1 Chemical structures of some systemic licensed antifungal agents: (A) amphotericin B, (B) 5-fluorocytosine, (C) miconazole, (D) ketoconazole, (E) fluconazole, and (F) itraconazole.

ch04.qxd 10/13/03 3:17 PM Page 51

51

antifungal agents heat, light, and acid pH. The fungistatic (inhibition of fungal growth) and fungicidal (lethal) activity of amphotericin B is due to its ability to combine with ergosterol in the cell membranes of susceptible fungi. Pores or channels are formed causing osmotic instability and loss of membrane integrity. This effect is not specific; it extends to mammalian cells. The drug binds to cholesterol, creating the high toxicity associated with all conventional polyene agents. A second mechanism of antifungal action has been proposed for amphotericin B, which is oxidation dependent. Amphotericin B is highly protein bound (91–95%). Peak serum of 1–3 g/ml and trough concentrations of 0.5–1.1 g/ml are usually measured after the intravenous (i.v.) administration of 0.6 mg/kg doses. Its half-life of elimination is 24–48 h, with a long terminal half-life of up to 15 days. Although resistance to amphotericin B is rare, quantitative and qualitative changes in the cell membrane sterols have been associated with the development of microbiological resistance both in vitro and in vivo. Clinically, resistance to amphotericin B has become an important problem, particularly with certain yeast and mold species, such as Candida lusitaniae, C. krusei, C. glabrata, Aspergillus terreus, Fusarium spp., Malassezia furfur, Pseudallescheria boydii, Scedosporium prolificans, Trichosporon beigelii, and other emerging fungal pathogens. The in vitro spectrum of activity of amphotericin B includes yeasts, dimorphic fungi, and most of the opportunistic filamentous fungi. Clinically, amphotericin B is considered the gold standard antifungal agent for the management of most systemic and disseminated fungal infections caused by both yeasts and molds, including endemic (infections caused by the dimorphic fungi, Cocccidioides immitis, Histoplasma capsulatum, and Blastomyces dermatitidis) and opportunistic mycoses. Although it penetrates poorly into the cerebrospinal fluid (CSF), amphotericin B is effective in the treatment of both Candida and Cryptococcus meningitis alone and/or in combination with 5-fluorocytosine. Current recommendations regarding daily dosage, total dosage, duration, and its use in combination with other antifungal agents are based on the type of infection and the status of the host. Since severe fungal infections in the granulocytopenic host are difficult to diagnose and cause much mortality, empirical antifungal therapy with amphotericin B and other agents has improved patient care. Systemic prophylaxis for patients at high risk for invasive mycoses has also evolved. Toxicity is the limiting factor during amphotericin B therapy and has been classified as acute or delayed (Table 4.2). Nephrotoxicity is the most significant delayed adverse effect. Therefore, close monitoring of renal function tests,

TABLE 4.2

Adverse Side effects of the Licensed Systemic Antifungal Agentsa

Side effect

Drug

Fever, chills Rash Nausea, vomiting Abdominal pain Anorexia Diarrhea Elevation of transaminases Hepatitis (rare) Anemia Leukopenia, thrombocytopenia Decreased renal function (azotemia, acidosis, hypokalemia, etc.) Decreased testosterone synthesis Adrenal insufficiency, menstrual irregularities, female alopecia Syndrome of mineralocorticoid excess, pedal edema Headache Photophobia Dizziness Seizures Confusion Arthralgia, myalgia, thrombophlebitis Abnormal vision Cardiovascular (tachycardia and others) Hypokalemia

A, K, C, V FC, K, I, FL, C, V A, FC, K, I, FL, V FC, K, V A, K FC, V FC, K, I, FL, C, V FC, K, I, FL A, FC FC A, C K (I, rare) K I A, FC, K, I, FL, V K, V I, V FL FC, V A V C, V C, V

a See Groll et al. (1998) for more detailed information. A, amphotericin B; FC, flucytosine; K, ketoconazole; FL, fluconazole; I, itraconazole; C, caspofungin, and V, voriconazole. (% of side effects for C and V are usually lower than those for other licensed agents.)

bicarbonate, electrolytes including magnesium, diuresis, and hydration status is recommended during amphotericin B therapy. Adverse drug interactions can occur with the administration of electrolytes and other concomitant drugs. This drug is also used for the treatment of systemic infections in small animals, especially blastomycosis in dogs, but it is not effective against aspergillosis. Side effects (especially in cats) and drug interactions are similar to those in humans.

B. Nystatin Nystatin was the first of the polyenes to be discovered when it was isolated from S. noursei in the early 1950s. It is an amphoteric tetrane macrolide that has a similar structure (Fig. 4.2A) and identical mechanism of action to those of conventional amphotericin B. Although it has an in vitro spectrum of activity similar to that of amphotericin B, this antifungal is used mostly for the therapy of gastrointestinal (orally) and mucocutaneous candidiasis (topically). This is not only due to its toxicity after parenteral administration to humans and lower animals but also to its lack of effectiveness when given i.v. to experimental animals.

ch04.qxd 10/13/03 3:17 PM Page 52

52

the desk encyclopedia of microbiology

FIGURE 4.2 Chemical structures of the most commonly used topical antifungal agents: (A) nystatin, (B) griseofulvin, (C) clotrimazole, and (D) terbinafine.

It is used for candidiasis in small animals and birds and for otitis caused by Microsporum canis.

C. Lipid formulations 1. Amphotericin B lipid formulations In an attempt to decrease the toxicity and increase the efficacy of amphotericin B in patients with deep-seated fungal infections refractory to conventional therapy, several lipid formulations of this antifungal have been developed since the 1980s. These preparations have selective toxicity or affinity for fungal cell membranes and theoretically promote the delivery of the drug to the site of infection while avoiding the toxicity of supramaximal doses of conventional amphotericin B. Because lysis of human erythrocytes is reduced, higher doses of amphotericin B can be safely used. Three lipid formulations of amphotericin B have been evaluated in

clinical trials: an amphotercin B lipid complex, an amphotericin B colloidal dispersion, and a liposomal amphotericin B. However, despite evidence of nephrotoxicity reduction, a significant improvement in their efficacy compared to conventional amphotericin B has not been clearly demonstrated. Although these three formulations have been approved for the treatment of invasive fungal infections that have failed conventional amphotericin B therapy, enough information is not available regarding their pharmacokinetics, drug interactions, long-term toxicities, and the differences in both efficacy and tolerance among the three formulations. Also, the most cost-effective clinical role of these agents as first-line therapies has not been elucidated. a. Liposomal amphotericin B In the only commercially available liposomal formulation (ambisome), amphotericin B is incorporated

ch04.qxd 10/13/03 3:17 PM Page 53

53

antifungal agents into small unilamellar, spherical vesicles (60–70-nm liposomes). These liposomes contain hydrogenated soy phospatidylcholine and disteaoryl phosphatidylglycerol stabilized by cholesterol and amphotericin B in a 2 : 0.8 : 1 : 0.4 molar ratio. In the first liposomes, amphotericin B was incorporated into large, multilamellar liposomes that contained two phospholipids, dimyristoyl phosphatidylcholine (DMPC) and dimyristoyl phosphatidylglycerol (DMPG), in a 7:3 molar ratio (5–10% mole ratio of amphotericin B to lipid). This formulation is not commercially available, but it led to the development of commercial formulations.

its use was restricted to topical applications for the treatment of vaginal candidiasis (infections by Candida albicans and other Candida spp.).

E. Pimaricin Pimaricin is a tetraene polyene produced by S. natalensis. It has a higher binding specificity for cholesterol than for ergosterol and, therefore, it is highly toxic for mammalian cells. The therapeutic use of pimaricin is limited to the topical treatment of keratitis (eye infections; also in horses) caused by the molds, Fusarium spp., Acremonium spp., and other species.

b. Amphotericin B lipid complex Amphotericin B lipid complex (abelcet) contains a DMPC/DMPG lipid formulation in a 7:3 ratio and a 50% molar ratio of amphotericin B to lipid complexes that form ribbon-like structures. c. Amphotericin B colloidal dispersion Amphotericin B colloidal dispersion (amphotec) contains cholesteryl sulfate and amphotericin B in a 1:1 molar ratio. This formulation is a stable complex of disk-like structures (122-nm in diameter and 4-nm thickness). 2. Liposomal nystatin In order to protect human erythrocytes from nystatin toxicity and thus make this drug available as a systemic therapeutic agent, nystatin has been incorporated into stable, multilamellar liposomes, which contain DMPC and DMPG in a 7:3 ratio. It has been demonstrated that the efficacy of liposomal nystatin is significantly superior to that of conventional nystatin and is well tolerated in experimental murine models of systemic candidiasis and aspergillosis (fungal infections caused by Candida spp. and Apergillus spp.). In patients with hematological malignancies and refractory febrile neutropenia, doselimiting nephrotoxicity has not been observed at high dosages. A 37% response to therapy has been documented in a small group of patients. Ongoing clinical trials would confirm the potential value of liposomal nystatin.

D. Candicidin Candicidin is a conjugated heptaene complex produced by S. griseus that is selectively and highly active in vitro against yeasts. It is more toxic for mammalian cells than either amphotericin B or nystatin; therefore,

II. GRISEOFULVIN Griseofulvin is a phenolic, benzyfuran cyclohexane agent (Fig. 4.2B) that binds to RNA. It is a product of Penicillium janczewskii and was the first antifungal agent to be developed as a systemic plant protectant. It acts as a potent inhibitor of thymidylate synthetase and interferes with the synthesis of DNA. It also inhibits microtubule formation and the synthesis of apical hyphal cell wall material. With the advent of terbinafine and itraconazole, the clinical use of griseofulvin as an oral agent for treatment of dermatophytic infections has become limited. However, it is frequently used for these infections in small animals, horses, and calves (skin only) as well as for equine sporotrichosis. Abdominal adverse side effects have been noted, especially in cats.

III. CYCLOHEXIMIDE This is a glutaramide agent produced by S. griseus. This agent was among the three antifungals that were reported between 1944 and 1947. Although cycloheximide had clinical use in the past, it is currently used as a plant fungicide and in the preparation of laboratory media.

IV. PYRROLNITRIN, FENPICLONIL, AND FLUDIOXONIL Pyrrolnitrin is the fermentation product of Pseudomonas spp. It was used in the past as a topical agent. Fenpiclonil and fludioxonil (related to pyrrolnitrin) were the first of the phenylpyrrols to be introduced as cereal seed fungicides.

ch04.qxd 10/13/03 3:17 PM Page 54

54

the desk encyclopedia of microbiology

V. THE SYNTHETIC PYRIMIDINES A. 5-Fluorocytosine (5-FC, flucytosine) The synthetic 5-fluorocytosine is an antifungal metabolite that was first developed as an antitumor agent, but it is not effective against tumors. It is an oral, low-molecular-weight, fluorinated pyrimidine related to 5-fluorouracil and floxuridine (Fig. 4.1B). It acts as a competitive antimetabolite for uracil in the synthesis of yeast RNA; it also interferes with thymidylate synthetase. Several enzymes are involved in the mode of action of 5-fluorocytosine. The first step is initiated by the uptake of the drug by a cell membranebound permease. Inside the cell, the drug is deaminated to 5-fluorouracil, which is the main active form of the drug. These activities can be antagonized in vitro by a variety of purines and pyrimidine bases and nucleosides. At least two metabolic sites are responsible for resistance to this compound: one involves the enzyme cytosine permease, which is responsible for the uptake of the drug into the fungal cell, and the other involves the enzyme cytosine deaminase, which is responsible for the deamination of the drug to 5-fluorouracil. Alterations of the genetic regions encoding these enzymes may result in fungal resistance to this drug by either decreasing the cell wall permeability or synthesizing molecules that compete with the drug or its metabolites. Development of flucytosine resistance during therapy against Candida spp. and C. neoformans has been documented since the early 1970s. 5-Fluorocytosine has fungistatic but not fungicidal activity mostly against yeasts; its activity against molds is inoculum dependent. Clinically, the major therapeutic role of 5-fluorocytosine is its use in combination with amphotericin B in the treatment of meningitis caused by the yeast C. neoformans. The synergistic antifungal activity of these two agents has been demonstrated in clinical trials in non-HIV-infected and AIDS patients. 5-Fluorocytosine should not be used alone for the treatment of any fungal infections. Therapeutic combinations of 5-fluorocytosine with several azoles are under investigation. The most serious toxicity associated with 5-fluorocytosine therapy is bone marrow suppression (6% of patients), which leads to neutropenia, thrombocytopenia, or pancytopenia (Table 4.2). Therefore, monitoring of the drug concentration in the patient’s serum (serial 2-h levels post-oral administration) is highly recommended to adjust dosage and maintain serum levels between 40 and 60 g/ml. Since the drug is administered in combination with amphotericin B, a decrease in glomerular filtration rate, a side

effect of the latter compound, can induce increased toxicity to 5-fluorocytosine. Adverse drug interactions can occur with other antimicrobial and anticancer drugs, cyclosporine, and other therapeutic agents. Because of its toxic potential, 5-fluorocytosine should not be administered to pregnant women or animals. This drug has been used in combination with ketoconazole for cryptococcosis in small animals (very toxic for cats) and also for respiratory apergillosis and severe candidiasis in birds.

B. Triarimol, fenarimol, pyrimethanil, and cyprodinil Triarimol and fenarimol are pyrimidines with a different mechanism of action than that of 5-fluorocytosine. They inhibit lanosterol demethylase, an enzyme involved in the synthesis of ergosterol, which leads to the inhibition of this biosynthetic pathway. Triarimol and fenarimol are not used in medicine but are used extensively as antifungal agents in agriculture. The anilino-pyrimidines, pyrimethanil and cyprodinil, inhibit the secretion of the fungal enzymes that cause plant cell lysis. Pyrimethanil has activity (without cross-resistance) against Botrytis cinerea (vines, fruits, vegetables, and ornamental plants infections) and Venturia spp. (apples and pears), whereas cyprodinil has systemic activity against Botrytis spp., but only a preventive effect against Venturia spp.

VI. THE AZOLES The azoles are the largest single source of synthetic antifungal agents; the first azole was discovered in 1944. As a group, they are broad-spectrum in nature and mostly fungistatic. The broad spectrum of activity involves fungi (yeasts and molds), bacteria, and parasites. This group includes fused ring and N-substituted imidazoles and the N-substituted triazoles. The mode of action of these compounds is the inhibition of lanosterol demethylase, a cytochrome P-450 enzyme.

A. Fused-ring imidazoles The basic imidazole structure is a cyclic five-member ring containing three carbon and two nitrogen molecules. In the fused-ring imidazoles, two carbon molecules are shared in common with a fused benzene ring. Most of these compounds have parasitic activity (anthelmintic) and two have limited antifungal activity: 1-chlorobenzyl-2-methylbenzimidazole and thiabendazole.

ch04.qxd 10/13/03 3:17 PM Page 55

antifungal agents 1. 1-Chlorobenzyl-2-methylbenzimidazole The azole 1-chlorobenzyl-2-methylbenzimidazole was developed specifically as an anti-Candida agent. It has been used in the past in the treatment of superficial yeast and dermatophytic infections. 2. Thiabendazole Thiabendazole was developed as an anthelmintic agent and has a limited activity against dermatophytes. It was also used in the past in the treatment of superficial yeast and dermatophytic infections. Thiabendazole has been used for aspergillosis and penicillosis in dogs.

B. N-substituted (mono) imidazoles In this group, the imidazole ring is intact and substitutions are made at one of the two nitrogen molecules. At least three series of such compounds have emerged for clinical and agricultural use. In the triphenylmethane series, substitutions are made at the nonsymmetrical carbon atom attached to one nitrogen molecule of the imidazole ring. In the second series, the substitutions are made at a phenethyl configuration attached to the nitrogen molecule. The dioxolane series is based on a 1,3-dioxolane molecule rather than on the 1-phenethyl molecule. These series vary in spectrum, specific level of antifungal activity, routes of administration, and potential uses.

55

agent for superficial infections, despite its broad spectrum of activity. Its limited use is the result of its toxic side effects for mammalian cells. Bifonazole is retained in the dermis for a longer time than clotrimazole. 3. Econazole, isoconazole, oxiconazole, and tioconazole Other frequently used topical imidazoles include econazole (1% cream), isoconazole (1% cream), oxiconazole (1% cream and lotion), and tioconazole (6.5% vaginal ointment) (Table 4.1). As with clotrimazole, a single application of tioconazole is effective in the management of vulvovaginal candidiasis and as a nail lacquer for fungal onychomycosis (nail infections). Mild to moderate vulvovaginal burning has been associated with intravaginal therapy. Oxiconazole and econazole are less effective than terbinafine and itraconazole in the treatment of onychomycosis and other infections caused by the dermatophytes. Although topical agents do not cure onychomycosis as oral drugs do, they may slow down the spread of this infection. However, the recommended drugs for the treatment of onychomycosis are terbinafine (by dermatophytes) and itraconazole. 4. Lanoconazole In recent years, lanoconazole has been introduced for topical treatment of dermatomycoses. It appears to have superior activity in vitro and in experimental infections in guinea pigs than those of earlier compound.

1. Clotrimazole 5. Miconazole

Clotrimazole is the first member of the triphenylmethane series of clinical importance (Fig. 4.2C). It has good in vitro activity at very low concentrations against a large variety of fungi (yeasts and molds). However, hepatic enzymatic inactivation of this compound, after systemic administration, has limited its use to topical applications (1% cream, lotion, solution, tincture, and vaginal cream) for superficial mycoses (nail, scalp, and skin infections) caused by the dermatophytes and M. furfur, for initial and/or mild oropharyngeal candidiasis (OPC; 10-mg oral troche), and for the intravaginal therapy (single application of 500-mg intravaginal tablet) of vulvovaginal candidiasis. Other intravaginal drugs require 3–7-day applications. This drug is also used for candidal stomatitis, dermatophytic infections, and nasal aspergillosis (infused through tubes) in dogs.

Miconazole was the first azole derivative to be administered intravenously for the therapy of systemic fungal infections. Its use is limited, due to toxicity and high relapse rates, to certain cases of invasive infections caused by the opportunistic mold, P. boydii. Since this compound is insoluble in water, it was dissolved in a polyethoxylated castor oil for its systemic administration. This solvent appears to be the cause of the majority of miconazole side effects (pruritus, headache, phlebitis, and hepatitis). On the other hand, miconazole is used for dermatophytic infections in large animals, fungal keratitis and pneumonia in horses, resistant yeast infections to nystatin in birds, and aspergillosis in raptors. However, safety and efficacy data are not available (veterinary use).

2. Bifonazole

6. Ketoconazole

Bifonazole is a halogen-free biphenylphenyl methane derivative. Bifonazole is seldom utilized as a topical

Ketoconazole was the first representative of the dioxolane series (Fig. 4.1D) to be introduced into clinical

ch04.qxd 10/13/03 3:17 PM Page 56

56

the desk encyclopedia of microbiology TABLE 4.3 Adverse interactions of the licensed systemic azoles with other drugs during concomitant therapya

Azole

Concomitant drug

Adverse side effect of interaction

K, Fl, I K, Fl, I, V

Nonsedating antihistamines, cisapride, terfenadine, astemizole Rifampin, isoniazid, phenobarbital, rifabutin, carbamazepine, and phenytoin Phenytoin, benzodiazepines, rifampin Antacids, H2 antagonists, omeprazole, sucralfate, didanosine Lovastin, simvastatin Indinavir, vincristine, quinidine, digoxin, cyclosporine, tacrolimus, methylprednisolone, and ritonavir Warfarin, rifabutin, sulfonylurea Saquinavir, chlordiazepoxide, methylprednisone Protein-binding drugs Cyclosporine A

Fetal arrhythmia Reduce azole plasma concentrations

K, Fl, I, V K, I K, Fl, I I Fl, I, V K K K, C, V C C

Tacrolimus Efavirenz, nevirapine, phenytoin, dexamethasone, carbamazepine, and rifampin

Induces the potential toxicity levels of cocompounds Reduces azole absorption Rhabdomyolysis Induces potential toxicity cocompounds Induces potential toxicity of cocompounds Induces potential toxicity of these compounds Increases the release of fractions of free drug Nephrotoxicity (concomitant use with C is not recommended) Tacrolimus concentration can be decreased Can significantly reduce C concentrations (use of daily dose of 70 mg of C should be considered when C is co-administered with some of these compounds.

a

See Groll et al. (1998) for more detailed information. K, ketoconazole; Fl, fluconazole; I, itraconazole; C, caspofungin; V, voriconazole.

use and was the first orally active azole. Ketoconazole requires a normal intragastric pH for absorption. Its bioavailability is highly dependent on the pH of the gastric contents; an increase in pH will decrease its absorption, for example, in patients with gastric achlorhydria or treated with antacids or H2-receptor antagonists (Table 4.3). This drug should be taken with either orange juice or a carbonated beverage. Ketoconazole pharmocokinetics corresponds to a dual model with an initial half-life of 1–4 h and a terminal half-life of 6–10 h, depending on the dose. This drug highly binds to plasma proteins and penetrates poorly into the CSF, urine, and saliva. Peak plasma concentrations of approximately 2, 8, and 20 g/ml are measured 1–4 h after corresponding oral doses of 200, 400, and 800 mg. The most common and dosedependent adverse effects of ketonazole are nausea, anorexia, and vomiting (Table 4.2). They occur in 10% of the patients receiving a 400-mg dose and in approximately 50% of the patients taking 800-mg or higher doses. Another limiting factor of ketoconazole therapy is its numerous and significant adverse interactions with other concomitant drugs (see Table 4.3 for a summary of the interactions of the azoles with other drugs administered to patients during azole therapy). In vitro, ketoconazole has a broad spectrum of activity comparable to that of miconazole and the triazoles. However, due to its adverse side effects, its adverse interaction with other drugs, and the high rate of relapses, ketoconazole has been replaced by itraconazole as an alternative to amphotericin B for the treatment of immunocompetent individuals with non-life-threatening, non-central nervous system, localized or disseminated histoplasmosis, blastomycosis, mucocutaneous

candidiasis, paracoccidioidomycosis, and selected forms of coccidioidomycosis. In non-cancer patients, this drug can be effective in the treatment of superficial Candida and dermatophytic infections when the latter are refractory to griseofulvin therapy. Therapeutic failure with ketoconazole has been associated with low serum levels; monitoring of these levels is recommended in such failures. Ketoconazole also has been used for a variety of systemic and superficial fungal infections in cats and dogs. 7. Enilconazole This is the azole most widely used in veterinary practice for the intranasal treatment of aspergillosis and penicillosis as well as for dermatophytic infections. The side effects are few. 8. Epoxiconazole, fluquinconazole, triticonazole, and prochloraz Epoxiconazole, fluquinconazole, and triticonazole are important agricultural fungicides which have a wider spectrum of activity than that of the earlier triazoles, triadimefon and propiconazole, and the imidazole, prochloraz, as systemic cereal fungicides. However, development of resistance to these compounds has been documented.

C. The triazoles The triazoles are characterized by a more specific binding to fungal cell cytochromes than to mammalian cells due to the substitution of the imidazole ring by the triazole ring. Other beneficial effects of this substitution

ch04.qxd 10/13/03 3:17 PM Page 57

antifungal agents are: (i) an improved resistance to metabolic degradation; (ii) an increased potency; and (iii) a superior antifungal activity. Although fluconazole, itraconazole and voriconazole are the only three triazoles currently licensed for antifungal systemic therapy, several other triazoles are at different levels of clinical evaluation. Voriconazole has been licensed in Europe (Table 4.1). 1. Fluconazole Fluconazole is a relatively small molecule (Fig. 4.1E) that is partially water soluble, minimally protein bound, and excreted largely as an active drug in the urine. It penetrates well into the CSF and parenchyma of the brain and the eye, and it has a prolonged halflife (up to 25 h in humans). The pharmacokinetics are independent of the route of administration and of the drug formulation and are linear. Fluconazole is well absorbed orally (its total bioavailability exceeds 90%), and its absorption is not affected by food or gastric pH. Plasma concentrations of 2–7 g/ml are usually measured in healthy subjects after corresponding single doses of 100 and 400 mg. After multiple doses, the peak plasma levels are 2.5 times higher than those of single doses. The CSF to serum fluconazole concentrations are between 0.5 and 0.9% in both healthy human subjects and laboratory animals. Fluconazole does not have in vitro or in vivo activity against most molds. Both oral and i.v. formulations of fluconazole are available for the treatment of candidemia in nonneutropenic and other nonimmunosuppressed patients, mucosal candidiasis (oral, vaginal, and esophageal), and chronic mucocutaneous candidiasis in patients of all ages. Fluconazole is the current drug of choice for maintenance therapy of AIDS-associated cryptococcal and coccidioidal meningitis. It is also effective as prophylactic therapy for immunocompromised patients to prevent both superficial and life-threatening fungal infections. However, since the cost of fluconazole is high and resistance to this drug can develop during therapy, fluconazole prophylaxis should be reserved for HIV-infected individuals or AIDS patients, who are refractory and intolerant to topical agents, or for patients with prolonged (2 weeks) and profound neutropenia (1500 cells). Although the recommended dosage of fluconazole for adults is 100–400 mg qd, higher doses (800 mg qd) are required for the treatment of severe invasive infections and for infections caused by a Candida spp. that exhibit a minimum inhibitory concentration (MIC) of 8 g/ml. However, despite the fluconazole MIC obtained when the infecting yeast is either Candida krusei or C. glabrata, intrinsic resistance to these yeasts precludes its use for the treatment of such infections. In contrast to the imidazoles

57

and itraconazole, fluconazole does not exhibit major toxicity side effects (2.8–16%). However, when the dosage is increased above 1200 mg, adverse side effects are more frequent (Table 4.2). Fluconazole interactions with other concomitant drugs are similar to those reported with other azoles, but they are less frequent than those exhibited by ketoconazole and itraconazole (Table 4.3). Fluconazole has been used to treat nasal aspergillosis and penicillosis in small animals and birds when topical enilconazole is not feasible. 2. Itraconazole Itraconazole is another commercially available oral triazole for the treatment of certain systemic mycoses. In contrast to fluconazole, itraconazole is insoluble in aqueous fluids; it penetrates poorly into the CSF and urine but well into skin and soft tissues; and it is highly protein bound (90%). Its structure is closely related to that of ketoconazole (Fig. 4.1F), but itraconazole has a broader spectrum of in vitro and in vivo antifungal activity than those of both ketonazole and fluconazole. Similar to ketoconazole, itraconazole is soluble only at low pH and is better absorbed when the patient is not fasting. Absorption is erratic in cancer patients or when the patient is taking concomitant H2-receptor antagonists, omeprazole, or antacids. Therefore, this drug should be taken with food and/or acidic fluids. Plasma peak (1.5–4 h) and trough concentrations between 1 and 2.2 and 0.4 and 1.8 g/ml, respectively, are usually obtained after 200-mg dosages (capsule) as either single daily dosages (po or bid) or after i.v. administration (bid) for 2 days and qd for more days; these concentrations are also obtained in cancer patients receiving 5 mg/kg divided into two oral solution dosages. Clinically, itraconazole (200–400 mg/day) has supplanted ketoconazole as first-line therapy for endemic, non-life-threatening mycoses caused by B. dermatitidis, C. immitis, and H. capsulatum as well as by Sporothrix schenckii. For more severe mycoses, higher doses are recommended and clinical resistance may emerge. It can also be effective as a second-line agent for refractory or intolerant infections to conventional amphotericin B therapy, for example, infections by the phaeoid (dematiaceous or black molds or yeasts) fungi and Aspergillus spp. Itraconazole is commercially available as oral solution, tablet, and i.v. suspension. The oral solution is better absorbed than the tablet and has become useful for the treatment of HIV-associated oral and esophageal candidiasis, especially for those cases that are resistant to fluconazole. However, monitoring of itraconazole plasma concentrations is recommended during treatment of both superficial and invasive diseases: Drug concentration

ch04.qxd 10/13/03 3:17 PM Page 58

58

the desk encyclopedia of microbiology

0.5 g/ml by high-performance liquid chromatography and 12 g/ml by bioassay appear to be critical for favorable clinical response. Treatment with itraconazole has been associated with less adverse and mostly transient side effects (110%) than that with ketoconazole (Table 4.2), and these effects are usually observed when the patient takes up to 400 mg during several periods of time. Itraconazole has been used for the treatment of endemic mycoses, aspergillosis, and crytococcosis in dogs (especially blastomycosis), equine sporotrichosis, and osteomyelitis caused by C. immitis in large animals, but its use is minimal.

No data are available regarding its side effects or drug interactions in animals. 3. Voriconazole (UK-109496) Voriconazole is a novel fluconazole derivative obtained by replacement of one triazole moiety by fluoropyrimidine and -methylation groups (Fig. 4.3A). In contrast to fluconazole and similar to itraconazole, voriconazole is non-water soluble. As do the other azoles, voriconazole acts by inhibiting fungal cytochrome P450-dependent, 14--sterol

FIGURE 4.3 Chemical structures of three new triazoles: (A) voriconazole, (b) posaconazole, and (C) ravuconazole.

ch04.qxd 10/13/03 3:17 PM Page 59

antifungal agents demethylase-mediated synthesis of ergosterol. Voriconazole pharmacokinetics in humans are nonlinear. Following single oral doses, peak plasma concentrations were achieved after 2 h and multiple doses resulted in a higher (eight times) accumulation. The mean half-life of elimination is about 6 h. Voriconazole binds to proteins (65%), is extensively metabolized in the liver, and is found in the urine (78–88%) practically unchanged after a single dose. Voriconazole has an improved in vitro fungistatic activity and an increased potency against most fungi compared to those of fluconazole. It is fungicidal against some fungi, especially Aspergillus spp. However, less in vitro activity has been demonstrated for the opportunistic molds Fusarium spp., Rhizopus arrhizus, S. schenckii, and other less common emerging fungi. Studies in neutropenic animal models have demonstrated that voriconazole is superior to both amphotericin B and itraconazole for the treatment of certain opportunistic (especially aspergillosis) and endemic mycoses. This compound has undergone phase III evaluation for the treatment of invasive aspergillosis and infections refractory to established antifungal agents in humans. In the United States, voriconazole was approved May 24, 2002 for primary treatment of acute aspergillosis and as salvage therapy for serious fungal infections caused by S. apiospermum and Fusarium spp. However, the European label is for treatment of invasive aspergillosis, fluconazoleresistant serious invasive Candida infections (including C. krusei) and treatment of serious fungal infections caused by Scedosporium spp. and Fusarium spp. The drug has been well tolerated with only reversible side effects. In patients, hepatic (10–15%), transient visual (10–15%) and skin rash (1–5%) side effects have been observed. 4. Terconazole Terconazole was the first triazole marketed for the topical treatment of vaginal candidiasis and superficial dermatophyte infections. Currently, it is only used for vulvovaginal candidiasis (0.4 and 0.8% vaginal creams and 80-mg vaginal suppositories).

D. Investigational triazoles As fungal infections became an important health problem and resistance to established agents began to emerge, new triazoles were developed with a broader spectrum of antifungal activity. Early investigational triazoles, such as R 66905 (saperconazole), BAY R 8783, SCH 39304, and SCH 51048, were discontinued from further development due a variety of adverse

59

side effects. Two triazoles (posaconazole and ravuconazole) are currently under clinical investigation (Table 4.1) and others are at earlier stages of development. 1. SCH 39304, SCH 51048, and SCH 56592 a. SCH 39304 SCH 39304 is an N-substituted difluorophenyl triazole with both in vitro and in vivo (oral and parenteral) activity for both yeasts and molds. Although preliminary clinical trials demonstrated that this compound was well tolerated by humans and had good pharmacokinetic properties, additional clinical development was precluded by the incidence of hepatocellular carcinomas in laboratory animals during prolonged treatment. b. SCH 51048 SCH 51048 is a tetrahydrofurane-based triazole that has superior potency (orally) than that of SCH 39304 toward the target enzyme and good in vitro activity against a variety of fungi. Although animal studies demonstrated that this drug is also orally effective for the treatment of systemic and superficial yeast and mold infections, the slow absorption rate from the intestinal track due to its poor water solubility precluded its further clinical development. c. Posaconazole Posaconazole (SCH 56592) is the product of a modification of the n-alkyl side chain of SCH 51048 which included a variety of chiral substituents (Fig. 4.3b). The in vitro fungistatic and fungicidal activities of posaconazole are similar to those of voriconazole and ravuconazole and superior or comparable to those of the established agents against yeasts, the dimorphic fungi, most opportunistic molds including Aspergillus spp., the Zygomycetes, certain phaeoid fungi, and the dermatophytes. It has been demonstrated that posaconazole is superior to itraconazole for the treatment of experimental invasive aspergillosis in animals infected with strains of Aspergillus fumigatus with high and low itraconazole MICs. Posaconazole has been effective in the treatment of patients with non-meningeal coccidioidomycosis, oropharangeal infections and refractory and invasive mold infections including these caused by Fusarium spp. and the Zygomycetes. Similar results have been obtained for a variety of superficial and invasive infections in other animal models. The pharmacokinetics of

ch04.qxd 10/13/03 3:17 PM Page 60

60

the desk encyclopedia of microbiology

posaconazole have been studied in laboratory animals and although drug concentrations above both MIC and MFC (fungicidal) have been determined after a single po dose at 24 h, it has been demonstrated that plasma concentrations should be 5–10 times higher than the MIC. Also, its absorption from the intestinal tract is slow and peak serum concentrations are achieved 11–24 h after the actual dose. The clinical utility of this compound has yet to be determined in clinical trials in humans. 2. Ravuconazole (BMS-207147; ER-30346) Ravuconazole (BMS-207147) is a novel oral thiazolecontaining triazole (Fig. 4.3c) with a broad spectrum of activity against the majority of opportunistic pathogenic fungi. The antifungal activity of this triazole against A. fumigatus appears to be enhanced by the introduction of one carbon chain between the benzylic tert carbon and thiazole substituents and the cyano group on the aromatic ring attached to the thiazole. Ravuconazole has a similar or superior in vitro activity compared to those of the other investigational and established drugs against most pathogenic yeasts, with the exceptions of C. tropicalis and C. glabrata. Ravuconazole also has good in vivo antifungal activity in murine models for the treatment of invasive aspergillosis, candidiasis, and cryptococcosis. Ravuconazole shows good pharmacokinetics in animals that is similar to that of itraconazole. This indicates that ravuconazole is absorbed at levels comparable to those of itraconazole. However, the half-life of ravuconazole (4 h) is longer than that of itraconazole (1.4 h) and similar to that of fluconazole. The potential use of ravuconazole has yet to be determined in clinical trials in humans. 3. Saperconazole (R 66905) Saperconazole is a lipophilic and poorly watersoluble fluorinated triazole; its chemical structure resembles that of itraconazole. Although both in vitro and in vivo antifungal activities were demonstrated against yeasts and molds and it was well tolerated during three clinical trials, this triazole was discontinued due to the incidence of malignant adrenal tumors in laboratory animals (long-term toxicity experiments). 4. BAY R 3783 This metabolite triazole was also discontinued from further clinical development due to the potential toxic effect during prolonged therapy.

5. SDZ 89-485 The antifungal activity of the D-enantiomer SDZ 89–485 antifungal triazole was demonstrated only in a few laboratory animal studies, and additional studies were not conducted with this compound. 6. D 0870 Although more in vitro and in vivo studies were conducted with D 0870 than with SDZ-89-485, and D 0870 showed good antifungal activity, this drug was also discontinued by its original developers. The in vitro activity of D 0870 is lower than that of itraconazole against Aspergillus spp., but higher for the common Candida spp. Therefore, evaluation of this compound has been continued by another pharmaceutical company for the treatment of OPC in HIV-infected individuals. It has also shown activity against the parasite Trypanosoma cruzei. 7. T-8581 T-8581 is a water-soluble 2-fluorobutanamide triazole derivative. High peak concentrations (7.14–12 g/ml) of T-8581 were determined in the sera of laboratory animals following the administration of single oral doses of 10 mg/kg, and the drug was detected in the animals sera after 24 h. The half-life of T-8581 varies in the different animal models from 3.2 h in mice to 9.9 h in dogs. Animal studies suggest that the absorption of this compound is almost complete after po dosages. The maximum solubility of T-8581 is superior (41.8 mg/ml) to that of fluconazole (2.6 mg/ml), which suggests the potential use of this compound as an alternative to fluconazole for high-dose therapy. T-8581 has shown potent in vitro antifungal activity against Candida spp., C. neoformans, and A. fumigatus. The activity of T-8581 is similar to that of fluconazole for the treatment of murine systemic candidiasis and superior to itraconazole for aspergillosis in rabbits. The safety of T-8581 is under evaluation. 8. UR-9746 and UR-9751 UR-9746 and UR-9751 are similar and recently introduced fluoridated triazoles that contain an N-morpholine ring, but UR-9746 has an extra hydroxyl group. The pharmacokinetics of these two compounds in laboratory animals has demonstrated peak concentrations (biological activity) of 184 (UR-9746) and 34 g/ml (UR-9751) after 8 and 8–24 h, respectively. A slow decline of these levels was seen after 48 h. Chronic (19 days) doses of 100 mg/kg produced higher peak levels than single doses; two peaks were

ch04.qxd 10/13/03 3:17 PM Page 61

61

antifungal agents observed after 1 and 8 h. However, the rate of decline of the drug after 24 h was faster after multiple than after single doses. Superior in vitro and in vivo activity than that of fluconazole has been demonstrated with these compounds against Candida spp., C. neoformans, H. capsulatum, and C. imitis. Both antifungals lacked detectable toxicity in experimental animal infections. Although UR-9751 MICs were fourfold higher than those of UR-9746, the in vivo activity in the animal model of systemic murine coccidioidomycosis was similar. Additional studies will determine the potential use of these compounds as systemic therapeutic agents in humans. 9. TAK 187 and SSY 726 Some in vitro and very little in vivo data are available for these new triazoles. 10. KP-103 KP-103 is another novel triazole that is being developed for the local treatment of dermatomycoses. Because this azole has low affinity for keratin, its antifungal activity is not lost as it penetrates skin tissue. The clinical values of this agent is to be determined in clinical trials.

VII. THE ALLYLAMINES The allylamines are synthetic compounds that were introduced in the 1970s. They act by inhibiting squalene epoxidase, which results in a decrease of the ergosterol content and an accumulation of squalene affecting membrane structure and function (e.g. nutrient uptake).

A. Terbinafine Terbinafine is the most active derivative of this class of antifungals. It has an excellent in vitro activity against the dermatophytes and other filamentous fungi, but its in vitro activity against the yeasts is controversial. It follows linear pharmacokinetics over a dose range of 125–750 mg; drug concentrations of 0.5–2.7 g/ml are detected 1 or 2 h after a single oral dose. Terbinafine has replaced griseofulvin and ketoconazole for the treatment of onychomycosis and other infections caused by dermatophytes (oral and topical). It is also effective for the treatment of vulvovaginal candidiasis. It is usually well tolerated at oral doses of 250 and 500 mg/day and the side effects (~10%) are gastrointestinal and cutaneous. The

metabolism of terbinafine may be decreased by cimetidine and increased by rifampin. Resistance has been reported for Ustilago maydis, a corn pathogen; resistance involved a decreased affinity for the target enzyme as well as a decreased accumulation of drug inside the fungal cell.

B. Naftifine Pharmacokinetics and poor activity have limited the use of naftifine to topical treatment of dermatophytic infections.

VIII. THE BENZYLAMINES, THIOCARBAMATES, AND DITHIOCARBAMATES The benzylamine, butenafine, and the thiocarbamates, tolnaftate, tolciclate, and piritetrade, also inhibit the synthesis of ergosterol at the level of squalene. Their clinical use is limited to the topical treatment of superficial dermatophytic infections. The Bordeaux mixture (reaction product of copper sulfate and lime) was the only fungicide used until the discovery of the dithiocarbamate fungicides in the mid-1930s. Of those, mancozeb and thiram are widely used in agriculture, but because they are only surfaceacting materials frequent spray applications are required. Ferbam, maneb, and zineb are not used as much.

THE BENZIMIDAZOLES AND METHYLBENZIMIDAZOLE CARBAMATES A great impact on crop protection was evident with the introduction of the benzimidazoles and other systemic (penetrate the plant) fungicides. These compounds increased spray intervals to 14 days or more. The methylbenzimidazole carbamates (MBCs; carbendazim, benomyl, and thiophanate) inhibit nuclear division and are also systemic agricultural fungicides. However, since MBC-resistant strains of B. cinerea and Penicillium expansum have been isolated, these compounds should be used in combination with N-phenylcarbamate or agents that have a different mode of action.

THE MORPHOLINES The morpholines interfere with 14 reductase and 7 and 8 isomerase enzymes in the ergosterol

ch04.qxd 10/13/03 3:17 PM Page 62

62

the desk encyclopedia of microbiology

biosynthetic pathway, which leads to an increase of toxic sterols and an increase in the ergosterol content of the fungal cell.

A. Amorolfine Amorolfine, a derivative of fenpropimorph, is the only morpholine that has a clinical application for the topical treatment of dermatophytic infections and candidal vaginitis.

B. Fenpropimorph, tridemorph, and other morpholines Protein binding and side effects have precluded the clinical use of these morpholines, but they are important agricultural fungicides.

The echinocandins have better in vitro and in vivo antifungal activity than the papulocandins. Pharmaceutical development has resulted in several semisynthetic echinocandins with an improved antifungal activity compared to those of the naturally occurring molecules described previously. 1. Cilofungin (LY 121019) Cilofungin is a biosemisynthetic analog of the naturally occurring and toxic (erythrocytes lysis) 4-n-octyloxybenzoyl-echinocandin B. Although it showed good in vitro activity against Candida spp., this drug was discontinued due to the incidence of metabolic acidosis associated with its intravenous carrier, polyethylene glycol. 2. Anidulafungin (V-echinocandin, LY 303366)

XI. THE PYRIDINES The pyridines are another class of antifungal agents that inhibit lanosterol demethylase.

A. Buthiobate and pyrifenox These agents are important agricultural fungicides.

XII. THE ECHINOCANDINS, PNEUMOCANDINS, AND PAPULOCANDINS The echinocandins and papulocandins are naturally occurring metabolites of Aspergillus nidulans var. echinulatus (echinocandin B), A. aculeatus (aculeacin A), and Papularia sphaerosperma (papulocandin). They act specifically by inhibiting the synthesis of fungal (1,3)-glucan synthesis, which results in the depletion of glucan, an essential component of the fungal cell wall.

A. The papulocandins The papulocandins A–D, L687781, BU4794F, and chaetiacandin have in vitro activity only against Candida spp., but poor in vivo activity, which precluded clinical development.

B. The echinocandins The echinocandins include echinocandins, pneumocandins, aculeacins, mulundo- and deoxymulundocandin, sporiofungin, vWF 11899 A–C, and FR 901379.

This is another semisynthetic cyclic lipopeptide, which resulted from an increase of aromatic groups in the cilofungin side-chain (Fig. 4.4A). It has high potency and oral and parenteral bioavailability. In laboratory animals, peak levels in plasma (5 or 6 h) of 0.5–2.9 g/ml have been measured after single doses of 50–250 mg/kg. In humans, peak levels of 105–1624 ng/ml are measured after oral administrations of 100–1000 mg/kg; its pharmacokinetics is linear and the half-life is about 30 h and is dose independent. Tissue concentrations are usually higher than those in plasma in animals. Anidulafungin has good in vitro activity against a variety of yeasts, including isolates resistant to itraconazole and fluconazole, and molds. This compound is not active against C. neoformans, T. beigelii, and B. dermatitidis; its MICs for certain molds are higher than those of the three new investigational azoles. However, its fungicidal activity against some species of Candida is superior to those of the azoles, which are mostly fungistatic drugs. Although the drug is well tolerated up to 700 mg/kg doses, gastrointestinal adverse effects have been observed with 100 mg/kg doses in human subjects. Potentially peak plasma concentrations in excess of MIC values have been demonstrated in rabbits and tissue levels are above MIC endpoints in major organs. In human volunteers, it exhibits linear pharmacokinetics after single oral doses of 100–1000 mg. Anidulafungin peak plasma levels occurred after 6–7 h after ingestion with an elimination half-life of approximately 30 h. Anidulafungin has in vivo activity in experimental murine (normal and immunocompromised animals) aspergillosis and candidiasis and P. carinii pneumonia. Clinical trials are being conducted to assess the efficacy of anidulafungin against Candida infections.

ch04.qxd 10/13/03 3:17 PM Page 63

antifungal agents

FIGURE 4.4 Chemical structures of: (A) anidulafungin (LY-303366), (B) caspofungin (L-743872 or MK-0991), and (C) nikkomycin Z.

63

ch04.qxd 10/13/03 3:17 PM Page 64

64

the desk encyclopedia of microbiology

3. Anidulafungin derivatives Several derivatives of anidulafungin have been synthesized including the phospate derivative LY-307853 and phosphate ester derivatives LY-329960 and LY333006, which had improved water solubility. Good activity has been demonstrated in a murine model of candidiasis with the two latter compounds. Other echinocandin derivative is mulundocandin, which was obtained from a variant of A. sydowii. It has in vitro antifungal activity against itraconazoleresistant Candida spp. and A. fumigatus. 4. Cyclopeptamines A-192411.29 is a novel cyclopeptamine antifungal lipopeptide derived by total synthesis from the structural template of the natural product echinocandin. It has similar in vitro activity to that of amphotericin B against Candida spp. and C. neoformans and partial activity against A. fumigatus. 5. Non-echinocandin macrocyclic lipopeptidolactones (FR-901469) FR-901469 is a water-soluble, non-echinocandin-type lipoprotein fungal derivative. It has good in vitro activity against C. albicans and A. fumigatus.

C. Pneumocandin derivatives The pneumocandins have similar structures to those of the echinocandins, but they possess a hexapeptide core with a -hydroxyglutamine instead of the threonine residue, a branched-chain 14C fatty acid acyl group at the N-terminal, and variable substituents at the C-terminal proline residue. The pneumocandins are fermentation products of the mold Zalerion arbolicola. Of the three naturally occurring pnemocandins (A–C), only A and B have certain antifungal activity in vitro and in vivo against Candida spp. and Pneumocystis carinii (in rodents), but they are non-water-soluble; this group has pneumocandin Ao (L-671329) and pneumocandin Bo (L-688786), which are fermentation products produced by Zalerion arboricola ATCC 20868 and related semysinthetic derivatives. 1. L-639989, L-733560, L-705589, and L-731373 Modification of the original pneumocandin B by phosphorylation of the free phenolic hydroxyl group led to the improved, water-soluble pneumocandin B phosphate (L-639989). Further modifications of pneumocandin B led to the water-soluble semisynthetic

molecules L-733560, L-705589, and L-731373. Although studies were conduced in laboratory animals, these molecules were not evaluated in humans.

D. Caspofungin (MK-0991 or L-743872) Caspofungin acetate (Fig. 4.4B) is the product of a modification of L-733560 and was selected for further evaluation in clinical trials in humans. As are the other semisynthetic pneumocandins, caspofungin is water soluble. Caspofungin is highly protein bound (97%) with a half-life that ranges from 5 to 7.5 h and drug concentrations are usually higher in tissue than in plasma. Caspofungin has fungistatic and fungicidal activities similar to those of anidulafungin against most Candida spp. and lower activity against the dimorphic fungi. It also has fungistatic in vitro activity against some of the other molds, especially Aspergillus spp. However, both anidulafungin and caspofungin pose difficulties regarding their in vitro laboratory evaluation and the data are controversial regarding their MICs for the molds. Animal studies have demonstrated that this compound has good in vivo activity not only against yeast infections but also in murine models of disseminated aspergillosis and pulmonary pneumocystosis and histoplasmosis. The drug is not effective for the treatment of disseminated experimental infections caused by C. neoformans. In laboratory animals, the drug is mostly well tolerated, but histamine release and mild hepatotoxicity have been reported. Caspofungin is generally well tolerated in humans, but side effects include hypokalemia, nephrotoxicity, chills, fever, intestinal, tachycardia, rash, and sweating (Table 4.2). Data from 69 patients with either invasive refractory aspergillosis or intolerant infection to standard therapies demonstrated a 41% favorable response in patients receiving at least one dose of caspofungin and a 50% favorable response in patients receiving more than 7 days of therapy. Caspofungin has been licensed in the United States for candidemia and other Candida infections and for refractory aspergillosis.

E. Micafungin (FK 463) FK 463 is a semisynthetic derivative of naturally occurring lipopeptide that was synthesized by a chemical modification from a product of the mould Coleophoma empedri. As the other related compounds, it has good in vitro activity against Candida and Aspergillus species, but it is inactive against C. neoformans, T. beigelii, and F. solani. It also has good activity

ch04.qxd 10/13/03 3:17 PM Page 65

65

antifungal agents in vivo in experimental invasive candidiasis and pulmonary aspergillosis in neutropenic mice; the activity was similar to that of amphotericin B. The drug was well tolerated (up to 2.5–50 mg single, or 25 mg multiple doses, 7 days) in healthy adult male volunteers and adult bone marrow or peripheral stem cell transplant patients (200 mg/day, 10 days dosing average). The serum concentrations in the latter two groups of patients were higher than the serum levels obtained in experimental candidiasis and aspergillosis. Micafungin is protein binding (99%) and plasma concentrations attain a steady state by day 4 with repeated doses. Clearing of esophageal candidiasis symptoms has been demonstrated among 74 HIV-positive patients treated with micafungin at 50, 25, and 12.5 mg/day as 1-h infusion for a mean of 12 days. Diarrhea was the only side effect reported. Micafungin is undergoing Phase III clinical trials.

F. Other fungal cell inhibitors Various aromatic natural products have been shown to have antifungal activity against S. cerevisiae (xanthofulvin) and C. albicans (Ro-41-0986 and its derivative Ro-09-3024).

XIII. THE PRADIMICINS AND BENANOMYCINS The pradimicins and benanomycins are fungicidal metabolites (benzonaphthacene quinones) of Actinomadura spp., which were introduced in the 1980s. Several semisynthetic molecules have also been produced. They act by disrupting the cell membrane through a calcium-dependent binding with the saccharide component of mannoproteins, which results in disruption of the plasma membrane and leakage.

A. Pradimicin A (BMY 28567) and FA-2 (BMY 28864) The poor solubility of pradimicin A led to the development of BMY 28864, which is a water-soluble derivative of pradimicin FA-2. BMY 28864 appears to have good in vitro and in vivo activity against most common yeasts and A. fumigatus. Clinical trials in humans have not been conducted.

B. BMS 181184 This compound is either a semisynthetic or biosynthetic derivative of BMY 28864. Although it was selected for further clinical evaluation due its promising in vitro

and in vivo data, elevation of liver transaminases in humans led to the discontinuation of this drug.

C. Benanomycin A This compound has shown the best antifungal activity among the various benanomycins. Its great advantage compared to other new antifungals is its good in vivo activity in animals against P. carinii.

XIV. THE POLYOXINS AND NIKKOMYCINS The polyoxins are produced by S. cacaoi and the nikkomycins by S. tendae. The former compounds were discovered during a search for new agricultural fungicides and pesticides. Both polyoxins and nikkomycins are pyrimidine nucleosides that inhibit the enzyme chitin synthase, which leads to the depletion of chitin in the fungal cell wall; they were introduced in the 1960s and 1970s. These molecules are transported into the cell via peptide permeases.

A. Polyoxin D Although polyoxin D has in vitro antifungal activity against C. immitis (parasitic phase), C. albicans, and C. neoformans, it was not effective in the treatment of systemic candidiasis in mice.

B. Nikkomycin Z This compound appears to have both in vitro and in vivo activity against C. immitis, B. dermatitidis, and H. capsulatum, which are highly chitinous fungi. It also has in vitro modest activity against C. albicans and C. neoformans. Studies to evaluate its safety have been conducted and clinical trials have been designed for the treatment of human coccidioidomycosis. These studies will determine its role as a therapeutic agent in humans.

XV. THE SORDARINS The natural sordarin GR 135402 is an antifungal fermentation product of Graphium putredinis. The compounds GM 103663, GM 211676, GM 222712, and GM 237354 are synthetic derivatives of GR 135402. In vitro, GM 222712 and GM 237354 have shown broad-spectrum antifungal activity for a variety of yeasts and molds. Development of these two sordarines has been discontinued.

ch04.qxd 10/13/03 3:17 PM Page 66

66

the desk encyclopedia of microbiology

XVI. DIMETHOMORPH AND FLUAZINAM Dimethomorph is a cinnamic acid derivative for use against Plasmopara viticola on vines and Phytophthora infestans on tomatoes and potatoes; it is not crossresistant to phenylamides (systemic controllers of Phycomycetes plant infections). Fluazinam is used in vines and potatoes but also acts against B. cinera as an uncoupler of oxidative phosphorylation.

XIX. OTHER ANTIFUNGAL APPROACHES A. Natural and synthetic cationic peptides Cationic peptides provide a novel approach to antifungal therapy that warrants further investigation. 1. Cecropin Cecropin is a natural lytic peptide that is not lethal to mammalian cells and binds to ergosterol. Its antifungal activity varies according to the fungal species being challenged.

XVII. THE PHTHALIMIDES The discovery of captan in 1952 and later of the related captafol and folpet initiated the proper protection of crops by the application of specific fungicides. Captan is also used to treat dermatophytic infections in horses and cattle, but it causes skin sensitization in horses.

2. Indolicidin Indolicidin is a tridecapeptide that has good in vitro antifungal activity and when incorporated into liposomes has activity against experimental aspergillosis in animals. 3. Synthetic peptides

XVIII. THE SPHINGOLIPID SYNTHESIS INHIBITORS The sphingofungins, lipoxamycins, myriocin (ISP-1) and viridiofungins selectively inhibit the fatty acidlike natural products. Fumonisins (produced by the corn and human pathogen F. moniliforme) and the structually related Alternaria toxin are inhibitors of ceramide synthase; resemble the structurally unrelated australifungins. Because these compounds inhibit the mammalian sphingolipid pathway and cause accumulation of sphinganine (sphingolipid depletion of both mammalian and fungal cells), they are toxic. The fumonisins have been associated with cancer in humans and also could cause disease in animals. Aureobasidin A is an inhibitor of the IPC (inositolphosphorylceramide) synthase that is produced by the black yeast Aureobasidium pullulans; it is the less toxic of this class of compouds. The oral fungicidal activity of aureobasidin has been demonstrated in experimental murine candidiasis. Khafrefungin and rustmicin (galbonolide A) also inhibit IPC synthase and have fungicidal activity against some yeasts and moulds by causing ceramide accumulation in the cell membrane. Due to their toxic effects, only aureobasidin A had both preclinical and Phase I clinical trials. However, its development was discontinued owing to its limited activity against Candida spp.

Synthetic peptides have been derived from the natural bactericidal-permeability increasing factor. They appear to have in vitro activity against C. albicans, C. neoformans, and A. fumigatus and also show synergistic activity with fluconazole in vitro.

B. Amino acid analogs RI 331, the azoxybacillins, and cispentacin are amino acid analogs with good in vitro antifungal activity against Aspergillus spp. and the dermatophytes (RI 331 and azoxybacillins) and also good in vivo activity (cispentacin). RI 331 and the azoxybacillins inhibit homoserine dehydrogenase and the biosynthesis of sulfur-containing amino acids, respectively. The derivative of histatin 5 called P-113 has antifungal in vitro activity against Candida species. See also the following articles: ANTIVIRAL AGENTS, BACTERIOCINS, FUNGAL INFECTIONS, FUNGI, FILAMENTOUS

BIBLIOGRAPHY Allen, D. G., Pringle, J. K., Smith, D. A., Conlon, P. D., and Burgmann, P. M. (1993). Handbook of Veterinary Drugs. Lippincott, Philadelphia, PA. Chiou, C.C., Groll, A. H., and Walsh, T. J. (2000). New drugs and novel targets for treatment of invasive fungal infections in patients with cancer. The Oncologist 5, 120–135.

ch04.qxd 10/13/03 3:17 PM Page 67

67

antifungal agents Clemons, K. V. and Stevens, D. A. (1997). Efficacies of two novel azole derivatives each containing a morpholine ring, UR-9746 and UR-9751, against systemic murine coccidioidomycosis. Antimicrob. Agents Chemother. 41, 200–203. Espinel-Ingroff, A. (1996). History of medical mycology in the United States. Clin. Microbiol. Rev. 9, 235–272. Espinel-Ingroff, A. (1998). Comparison of in vitro activity of the new triazole SCH 56592 and the echinocandins MK-0991 (L-743,872) and LY303366 against opportunistic filamentous and dimorphic fungi. J. Clin. Microbiol. 36, 2950–2956. Espinel-Ingroff, A., and Shadomy, S. (1989). In vitro and in vivo evaluation of antifungal agents. Eur. Clin. Microbiol. Infect. Dis. 8, 352–361. Espinel-Ingroff, and Pfaller, M. A. (2003). Susceptibility methods: yeasts and filmentous fungi. In Manual of Clinical Microbiology (P. R. Murray, E. J. Baron, M. A. Pfaller, F. C. Tenover, and R. H. Yolken, Eds.), 8th edn. ASM, Washington, DC. Espinel-Ingroff, A., Boyle, K., and Sheehan, D. J. (2001). In vitro antifungal activities of voriconazole and reference agenst as determined by NCLLS methods: review of the literature. Mycopathologia 150, 101–115. Georgopapadakou, N. H. (2000). Antifungals targeted to sphingolipid synthesis: focus on inositol phosphorylceramide synthase. Exp. Opin. Invest. Drugs 9, 1787–1796. Georgopapadakou, N. H. (2001). Update on antifungals targeted to the cell wall: focus on -1,3-glucan synthase inhibitors. Exp. Opin. Invest. Drugs 10, 269–280.

Groll, A. H., Piscitelli, S. C., and Walsh, T. J. (1998). Clinical pharmacology of systemic antifungal agents: A comprehensive review of agents in clinical use, current investigational compounds, and putative targets for antifungal drug development. Adv. Pharmacol. 44, 343–500. Russell, P. E., Milling, R. J., and Wright, K. (1995). Control of fungi pathogenic to plants. In Fifty Years of Antimicrobials: Past Perspectives and Future Trends (P. A. Hunter, G. K. Darby, and N. J. Russell, Eds.). Cambridge University Press, New York. Sheehan, D. J., Hitchcock, C. A., and Sibley, C. M. (1999). Current and emerging azole antifungal agents. Clin. Microbiol. Rev. 12, 40–79. St Georgiev, V. (2000). Membrane transporters and antifungal drug resistance. Curr. Drug Targets 1, 261–268. Yang, Y. L. and Lo, H. J. (2001). Mechanisms of antifungal agent resistance. J Microbiol Immunol. Infect. 34, 79–86.

WEBSITES Practice guidelines of the Infectious Diseases Society of America http://www.journals.uchicago.edu/IDSA/guidelines/ Dr. Fungus: Antifungal drugs. With links. (Merck & Co.) http://www.doctorfungus.org/

ch05.qxd 10/9/03 6:47 PM Page 68

5 Antisense RNAs Andrea Denise Branch The Mount Sinai School of Medicine

GLOSSARY

posttranscriptional gene silencing A process through which specific RNAs are degraded posttranscriptionally, resulting in loss of expression of associated genes. RNA interference (RNAi) An efficient process that allows small (21–23 nucleotide-long) RNAs derived from double-stranded RNAs to inhibit (silence) specific target genes. The small interfering RNAs (siRNAs) that mediate RNAi may be produced through the cleavage of longer double-stranded RNAs, by an enzyme called Dicer, or they may be derived from artificial siRNA duplexes.

artificial RNAs RNA molecules expressed from genes that have been introduced into cells (transgenes) or RNA molecules synthesized in cell-free systems. The mode of action of artificial antisense RNAs is under active investigation. In some biological systems, artificial RNAs may themselves form double-stranded RNAs that mediate targetgene inhibition through novel mechanisms. complementarity A measure of the percentage of nucleotides in two sequences that are theoretically able to form Watson–Crick base pairs. cosuppression A type of posttranscriptional gene silencing in which transcripts of both an endogenous gene and an homologous transgene are synthesized and then degraded. homology-dependent viral resistance A form of posttranscriptional gene silencing in which viral RNAs are degraded in transgenic plants expressing RNAs homologous to viral RNAs, resulting in inhibition of viral replication and attenuation of virus symptoms. perfect double-stranded RNA (dsRNA) duplex A helical structure in which two segments from a single RNA molecule (an intramolecular duplex), or segments of two separate RNA molecules (an intermolecular duplex) in an anti-parallel orientation to each other form an uninterrupted series of Watson–Crick base pairs (C pairing with G; A pairing with U).

The Desk Encyclopedia of Microbiology ISBN: 0-12-621361-5

Antisense RNAs are RNA molecules that bind to a second, sense, RNA through complementary Watson–Crick base pairing of anti-parallel strands; RNA molecules that are at least 70% complementary to a second RNA for at least 30 nucleotides and thus have the potential for binding; or RNA molecules that are transcribed from the DNA strand opposite that of a second RNA. Antisense RNAs in gene therapy are complementary to target RNAs and are intended to eliminate the expression of specific genes; target RNAs may be either associated with diseases or with normal cellular functions. Naturally occurring antisense RNAs comprise a structurally and functionally diverse group that includes RNAs known to bind to their target RNAs and RNAs that simply contain sequences complementary to other previously identified RNAs. In 1984, Izant and Weintraub thrust antisense RNA into the center stage of molecular research by proposing

68

Copyright © 2003 Elsevier Ltd All rights of reproduction in any form reserved

ch05.qxd 10/9/03 6:47 PM Page 69

antisense rnas that artificial antisense RNAs could be used to eliminate the expression of specific target genes, offering an alternative to the labors of classical mutational analysis. Rather than producing random mutations and then screening for those affecting genes of interest, they suggested that mutants could be created at will by introducing antisense RNAs complementary to sense transcripts of selected genes. They envisioned antisense RNAs binding to messenger RNAs (mRNAs) or their precursors, forming duplexes, and thereby inhibiting gene expression. The promise of streamlined genetic analysis, and improved pharmaceutical agents, livestock, and crops stimulated tremendous interest in antisense technology in members of the research community and on Wall Street. However, artificial antisense RNAs have not always performed as intended. The molecular events responsible for their unexpected behavior are not yet known, but enough information has emerged to indicate that these events merit thorough investigation. To understand the properties of artificial antisense RNA and to gain a more complete understanding of RNA’s regulatory functions, it is essential to study both naturally occurring and artificial antisense RNA. This collection of molecules includes RNAs known to alter the expression of their sense RNA counterparts and RNAs whose sequences appear to equip them to interact with their sense counterparts (i.e., RNAs that are at least 70% complementary to a second RNA for at least 30 nucleotides). Many natural and artificial antisense RNAs exist—far too many for each to be discussed here. Therefore, this article focuses on the principles governing their behavior. (Information about antisense oligomers composed of DNA is not included, but has been reviewed by the author.)

I. INTRODUCTION A. Naturally occurring antisense RNAs are extremely versatile Antisense RNAs are best known for their ability to eliminate the expression of target RNAs by binding to complementary sequences. However, antisense RNAs do much more than turn off other genes. For example, in virus-infected mammalian cells, antisense RNA combines with sense RNA to form biologically active double-stranded RNA (dsRNA), which triggers the interferon (IFN) response. Other antisense RNAs are involved in RNA maturation. These molecules are often omitted from lists of antisense RNAs because they promote expression of their target RNAs, rather than inhibit it. However, there has never been a requirement

69

for antisense RNAs to function as negative regulators of gene expression. The antisense RNAs involved in RNA maturation illustrate how complementary RNA sequences contribute to essential cellular functions. The guide RNAs of certain parasites bind to mitochondrial mRNA precursors through short complementary regions and direct upstream editing of the pre-mRNA. (RNA editing is any process leading to an alteration in the coding capacity of an mRNA, other than splicing or 3-end processing). Similarly, small nucleolar RNAs (snoRNAs) bind to complementary regions of ribosomal RNA (rRNA) precursors, leading to methylasemediated site-specific modification of the precursor. Of the antisense RNAs on the frontiers of research, those transcribed from mammalian genes are among the most intriguing and in greatest need of further investigation. Based on evidence showing that certain of these RNAs down-regulate their targets—diminishing synthesis of sense RNA, interfering with pre-mRNA processing, and inhibiting sense RNA translation—it has generally been assumed that any newly discovered antisense RNA would also function as a negative regulator. However, recent data indicate that each RNA must be individually investigated. An antisense transcript of the Wilms’s tumor gene (a gene imprinted under certain circumstances) appears to enhance expression of the sense RNA (Moorwood et al., 1998). Concerning the range of possible antisense RNA functions, it is interesting to note that an antisense RNA to basic fibroblast growth factor mRNA is thought to serve in two capacities: to act as the mRNA for a highly conserved protein of its own and to down-regulate growth factor expression. Several additional antisense RNAs contain open reading frames and may specify proteins. When interpreting a report of a newly discovered antisense RNA, particularly one detected in eukaryotic cells, it is important to remember that terminology in this part of the field permits a molecule to be designated an “antisense RNA” on the basis of sequence information alone. There is no requirement that the RNA bind to its sense counterpart or alter expression of the sense RNA in any way. Furthermore, throughout the entire antisense field, there is no requirement that sense and antisense RNAs be transcribed from opposite strands of the same DNA, and thus they are not necessarily exact complements of each other. The looseness in antisense terminology could be problematic. However, it serves a useful purpose, increasing the chances that meaningful similarities will be recognized. Such similarities illustrate the principles governing the behavior of antisense RNAs. Examples are selected from three areas: prokaryotic systems, virus-infected mammalian cells, and artificial

ch05.qxd 10/9/03 6:47 PM Page 70

70

the desk encyclopedia of microbiology

inhibitory RNAs. Antisense RNAs involved in RNA maturation, such as guide RNAs and snoRNAs, are not discussed further due to space limitations. However, the ability of guide RNAs to transfer genetic information is reflected in the function of the minus-strand viral RNAs, which are included; and the ability of snoRNAs to induce site-specific methylation is echoed in the gene-specific DNA methylation associated with some of the artificial inhibitory RNAs.

B. Artificial RNAs expanded the antisense field in unexpected directions Some naturally occurring antisense RNAs are highly effective gene regulators. Their efficacy, and the conceptual simplicity of antisense-mediated gene ablation stimulated efforts to develop artificial antisense RNAs that could be used to inhibit specific genes in higher organisms and to confer resistance to micro-organisms. These efforts have already yielded commercial agricultural products, such as the transgenic Flavr Savr tomato. They have also revealed that it is sometimes possible to substitute a sense transcript for an antisense transcript and achieve the same level of target-gene inhibition. Because it is usually impossible for sense transcripts and their targets to form a perfect duplex containing more than about 7–12 bp, sense inhibition appears to be a manifestation of a novel regulatory pathway. It is important to learn the details of this pathway in order to gain insight into RNA function and to facilitate the development of more effective artificial RNAs for use in biotechnology and basic research.

C. Despite the diversity of antisense RNAs, four general principles account for most antisense effects The first principle is that, above all else, antisense RNAs are ribonucleic acids. As such, they are endowed with a unique combination of properties. RNAs can store and transmit genetic information, just as DNA can. Moreover, naturally occurring RNAs readily form intricate three-dimensional structures and can produce catalytic active sites. RNAs are directly involved in protein synthesis at a variety of levels. Most RNAs are transcribed from DNA through a complex process involving cis-active promoter elements and many proteins. Nascent transcripts are converted into mature RNAs through an equally complex set of reactions. Regulation can occur at any of a number of points during transcription and subsequent processing. RNAs can be stable, or they can turn over rapidly. RNAs can readily move from the nucleus to the cytoplasm, and shuttle back and forth. They can form structural signals recognized by

proteins, and they can interact with other nucleic acids through complementary base paring. These properties allow antisense RNAs to weave their way in and out of an enormous variety of cellular processes. The second principle is that complementarity between an antisense RNA and a second nucleic acid is no guarantee that the two molecules will bind to each other. The tendency of antisense and target RNAs to form complexes, or to remain as separate molecules, is strongly influenced by their individual intramolecular structures. Potential nucleation sites can be prominently displayed, or virtually inaccessible. Complex formation is a bimolecular reaction whose rate is sensitive to concentration; the rate increases with increasing RNA concentration. The relationship between antisense RNA structure and function is illustrated most clearly by the antisense RNAs of prokaryotic systems, which are described in Sections II.A and II.B. Subcellular location also affects the probability that two RNAs will interact. Sense and antisense RNAs transcribed from the same genetic locus are more likely to encounter each other than RNAs transcribed from distant sites in the DNA. Similarly, two RNAs that accumulate in the same membrane-bound compartment are more likely to interact than those in separate compartments. The third principle is that antisense activity is often mediated by proteins; these proteins must be identified and their modes of action characterized for antisense RNA function to be understood. Many different proteins bind to antisense RNAs and to the RNA–RNA duplexes they create. Depending on the protein and the nature of the duplex, binding can have a variety of effects. The same protein may catalyze a range of reactions, with the outcome determined by information encoded in the structure of the RNA–RNA duplex. Interactions between antisense RNAs and proteins are described in Sections II.D and III.C. The fourth principle is that dsRNA can act as a signal. Mammalian cells recognize dsRNA as a sign that they, or their neighbors, are infected by a virus. Double-stranded RNA causes mammalian cells to enter an antiviral state. This response is mediated by a group of dsRNA-binding proteins, which make up a very sensitive dsRNA biosensor. There is growing evidence that dsRNA has symbolic value to cells from a variety of plants and animals. The potential of dsRNA to act as a signal is described in Sections III.A and IV.B–D and should be kept in mind when considering the possible biological effects of an antisense RNA.

D. Summary Antisense RNAs have many roles. They can act as negative regulators of gene expression, induce interferon,

ch05.qxd 10/9/03 6:47 PM Page 71

antisense rnas or direct RNA maturation. Antisense effects are strongly influenced by internal RNA structure and are often mediated by proteins. Artificial antisense RNAs have allowed new agricultural products to be developed and revealed unexpected roles of RNA in gene regulation.

II. ANTISENSE RNAS IN PROKARYOTIC SYSTEMS: INHIBITION BY DIRECT BINDING TO TARGET RNAS A. The copy number of plasmid ColE1 is regulated by RNA I, an antisense transcript RNA I of the Escherichia coli plasmid ColE1 was the first regulatory antisense RNA to be discovered. In 1981, Lacatena and Cesareni reported that base pairing between complementary regions of RNA I and RNA II inhibits plasmid replication. Because this system illustrates many principles of antisense RNA action it is discussed in detail. As is typical of antisense reactions, binding between RNA I and RNA II is a bimolecular process. Its rate is concentration dependent. The concentration dependence of the RNA I–RNA II binding reaction is harnessed to achieve the desired biological effect— maintenance of plasmid copy number at a stable 10–20 copies per cell. Formation of the RNA I and RNA II complex inhibits plasmid DNA replication by preventing RNA II from maturing into the RNA primer required for DNA synthesis (for an excellent review of this system by a leading research group, see Eguchi et al., 1991). RNA I is constitutively synthesized at a high rate and has a short half-life. Its concentration

71

reflects the number of template DNA molecules. When copies of the plasmid are numerous, RNA I concentration is high, binding to RNA II is favored, and plasmid DNA synthesis is inhibited. Conversely, when the plasmid DNA concentration is low, RNA I concentration falls, and plasmid replication is stimulated. Because RNA I contains regions complementary to the RNA II molecules produced by related plasmids, it provides the basis for plasmid compatibility and incompatibility. RNA I has no coding capacity. Synthesis of RNA II is initiated 555 bases upstream from the origin of DNA replication. RNA I is perfectly complementary to 108 bases at the 5-end of RNA II and is transcribed from the same region of the genome, but in the opposite direction. RNA I must bind RNA II shortly after RNA II synthesis is initiated. If binding is delayed, the nascent RNA II transcript forms structures that render it resistant to inhibition by RNA I. This competition between formation of the RNA I–RNA II complex and the RNA II self-structure means that antisense activity requires rapid association. RNA I and RNA II interact through an intricate process whose individual steps are predetermined by the structures of the two RNAs. As illustrated in Fig. 5.1. RNA I has three stem–loop structures and a short tail at its 5-end. The loops contain seven bases. Figure 5.1 also depicts RNA II, in a conformation that may exist in nascent RNA II transcripts. The secondary structures of RNA I and RNA II maximize the chances that they will form a bimolecular complex. Three sets of complementary bases are exposed in single-stranded loops. Bases making up these potential nucleation sites are displayed in structures somewhat similar to those that project the bases of tRNA anti-codons toward the

FIGURE 5.1 Binding of ColEI RNA I to RNA II is a stepwise process. RNA I and RNA II interact through complementary sequences present in loops to form C**. Pairing between the 5-end of RNA I and RNA II is followed by a series of structural changes that culminate in the formation of a stable complex, Cs. Finally, RNA I hybridizes to RNA II throughout its entire length. Adapted from Eguchi et al. (1991).

ch05.qxd 10/9/03 6:47 PM Page 72

72

the desk encyclopedia of microbiology

mRNA codons. During nucleation, bases in corresponding loops of RNA I and RNA II interact weakly, evidently forming a limited number of bonds between bases in only one or two loops. The very unstable early intermediates can dissociate rapidly, or establish a “kissing complex,” which can, in turn, produce the C** complex, a structure in which all three corresponding loops (reversibly) interact with each other. The formation of C** can be facilitated by the plasmid-encoded protein, Rom (RNA-one-modulator), which binds to the kissing complex and reduces its equilibrium dissociation constant. The solution structure of a helix modeled after the RNA I–RNA II loop–loop helix was recently solved by nuclear magnetic resonance spectroscopy (Lee and Crothers, 1998). As expected from previous biochemical studies, all seven bases in the loops form complementary base pairs. The loop–loop helix partially stacks on the stem helices, producing a nearly linear structure. The loop–loop helix is bent toward the major groove, which is thereby narrowed. This bend, as well as phosphate clusters flanking the major groove, distinguish this helix from standard A-form RNA, perhaps accounting for the ability of Rom to recognize it. Conversion of C** to stable structures, such as Cs, begins with events occurring at the 5-end of RNA I. If enough time elapses, the stable complex convert into full-length dsRNA molecules. However, inhibition can result from stable complex formation itself and does not require the generation of a complete duplex. Any lengthy dsRNA regions that form are likely to be rapidly degraded by the endonuclease RNase III.

B. Antisense RNAs in prokaryotic systems have many features in common 1. Antisense RNA structure RNA I is only the first example of the many wellcharacterized antisense RNAs in prokaryotic systems. Four other representative antisense RNAs are depicted in Table 5.1 and their modes of action are presented. Prokaryotic antisense RNAs are typically small (65–100 bases), stable, noncoding RNAs, whose secondary structures contain one or more stem-loops (see Table 5.1). The loops usually contain 5–8 bases and often have sequences that are similar to each other. The stems of antisense RNA stemloop structures often contain unpaired nucleotides at precise locations. These “imperfections” protect the RNAs from cleavage by dsRNA-specific ribonucleases and also reduce the stability of the stems, allowing them to open up during binding to their target RNAs (Hjalt and Wagner, 1995).

2. Antisense and target RNAs associate through a stepwise binding process Antisense and target RNAs interact through a stepwise process that proceeds from nucleation to stable complex formation. This process has been studied extensively in three systems: RNA I and RNA II of the plasmid ColE1, CopA and CopT of the plasmid R1 (CopT occurs in repA mRNA; see Table 5.1), and RNAOUT and RNA-IN from the mobile genetic element IS10. The apparent second-order rate constants for pairing between these RNAs are in the range of 0.3–1.0 106 M1 s1. In the binding reactions between RNA I and RNA II and between CopA and CopT, loop–loop interactions between antisense and target RNAs nucleate binding and formation of the kissing complex. In the binding reaction between RNA-OUT and RNAIN, the first bonds form between bases in the loop of RNA-OUT and the 5-end of RNA-IN, an RNA thought to have a relatively open structure. Rapid association appears to be a general requirement for efficient antisense activity. Because they mediate this early and rapid association, the initial base pairs are far more important to the overall binding process than their thermodynamic contribution to the final complex would suggest. Early intermediates are rapidly replaced by complexes containing more intermolecular base pairs. The final outcome of antisense binding depends on the system. Effects include termination of transcription, destabilization, and inhibition of translation. All known prokaryotic antisense RNAs are negative regulators of gene expression, although according to Wagner and Simons, “mechanisms for positive control are quite plausible” (Wagner and Simons, 1994). Most, but not all, antisense RNAs are transcribed from overlapping gene sequences. As indicated in Table 5.1, mRNA-interfering complementary (micF) RNA is not closely linked to its target, ompF RNA. The duplex they form contains looped-out regions and noncanonical base pairs, in addition to conventional bonds (Delihas et al., 1997). This duplex helps to establish the lower limit of complementarity for an antisense–target RNA pair. Within the duplex, only 24 of 33 bases (73%) of micF RNA are Watson–Crick base-paired to nucleotides in ompF RNA.

C. Bioengineers hope to use the special features of naturally occurring antisense RNAs to develop effective artificial antisense RNAs Engdahl and colleagues attempted to apply their knowledge of naturally occurring antisense RNAs to develop bioengineered antisense RNAs capable of

ch05.qxd 10/9/03 6:47 PM Page 73

antisense rnas

73

TABLE 5.1 Representative antisense RNAs of prokaryotic accessory DNA elements and bacterial DNA System

Antisense RNA structure

Plasmid Plasmid R1 Antisense: CopA (91 bases) Target: CopT

Phage Bacteriophage Antisense: oop RNA (77 bases) Target: cII mRNA

Transposable element Insertion sequence IS10 Antisense: RNA OUT (70 bases) Target: RNA-IN

Bacterial chromosome Escherichia coli Antisense: micF RNA (93 bases) Target: ompF mRNA

inhibiting selected target genes (Engdahl et al., 1997). They tested antisense RNAs containing a recognition element resembling the major stem-loop of CopA and either a segment complementary to the ribosome binding site of the target RNA or a ribozyme. None of their antisense RNAs inhibited the target genes by more than 50%. They concluded, “we still have too

Mode of action CopA indirectly prevents synthesis of RepA, a protein required for plasmid replication, by pairing with CopT sequences in the polycistronic RNA that encodes RepA. Binding blocks the ribosome binding site of the tap gene, and prevents its translation, which is coupled to translation of the repA gene. Binding also yields duplexes which are RNase III substrates.

oop RNA binds to (55) bases at the 3-end of the cII portion of the cII-O mRNA, creating an RNase III cleavage site, destabilizing the cII message, and thereby enhancing the burst size during induction.

RNA-OUT binds to (35) nucleotides at the 5-end of RNA-IN, transposase mRNA, preventing translation by blocking the ribosome binding site or by creating an RNase III cleavage site. Antisense inhibition increases as IS10 copy number increases, producing multicopy inhibition. IS10 is the mobile element of Tn10 (a tetracycline-resistance transposon).

micF RNA is about 70% complementary to the 5-end of ompF mRNA in the region of the ribosome binding site. Binding modulates production of OmpF, a major component of the E. coli outer membrane.

little insight into the factors that determine this property [the ability to rapidly associate] and, hence cannot yet tailor such structures to any chosen target sequence.” However, their study and similar studies by other investigators are helping to identify the structural features needed to produce effective artificial antisense RNAs for use in bacterial systems.

ch05.qxd 10/9/03 6:47 PM Page 74

74

the desk encyclopedia of microbiology

D. Proteins often mediate antisense RNA activity: RNase III is a prototypic dsRNA-binding protein Important steps in several antisense systems are carried out by RNase III. Purified in 1968, RNase III was the first enzyme to be discovered that recognizes RNA–RNA duplexes as substrates. Like many other dsRNAbinding proteins, RNase III does not act exclusively on perfect duplexes. The varied interactions between RNase III and its substrates illustrate the range of functions that can be included in the repertoire of a single enzyme. As illustrated in Fig. 5.2A, RNase III cleaves perfect RNA–RNA duplexes into short fragments averaging about 15 bases in length. Cuts are made across both strands of the duplex, at sites that are usually offset by one or two bases. This reaction shows no sequence specificity. Certain complexes of antisense and target RNAs are degraded by such randomly placed doublecleavages. In contrast, the 30S rRNA precursor is cut at four precise bonds. The rRNA precursor folds into a structure with two large stems, one topped by the

sequence of 16S rRNA, the other by 23S rRNA. RNase III cleaves each of these stems exactly twice at specific nucleotides, releasing rRNA intermediates. The stem flanking 23S rRNA is shown in Fig. 5.2B. The cleavage reactions carried out on the nearly perfect stems in the rRNA precursor resemble those carried out on perfect duplexes in certain respects, but not others. They are double-cleavages, but at predetermined locations. Surprisingly, RNase III also makes a series of precise cuts in the early mRNA precursor of bacteriophage T7 (Fig. 5.2C), even though this molecule contains no structures that bear an obvious similarity to perfect duplexes. Although the bacteriophage T7 cleavage sites lack the hallmark feature of dsRNA—a consecutive series of Watson–Crick bonds—they almost certainly contain noncanonical bonds that confer stability and allow them to fold into three-dimensional structures with features similar to dsRNA. As a group, the RNase III cleavage sites demonstrate the subtlety of the interactions between dsRNA-binding proteins and RNA molecules. RNase III acts as a rampant random nuclease on dsRNA substrates. Duplexes are cleaved at multiple sites regardless of

FIGURE 5.2 Structural features of dsRNA regions direct RNase III cleavage. RNase III cleaves perfect RNA–RNA duplexes into fragments averaging 15 base pairs in length (A). In addition, it cuts the 30S ribosomal RNA precursor at four positions, releasing intermediates that will become the mature ribosomal RNAs. The double-cleavage site in the region 23S region (Bram et al., 1980) is shown (B). Finally, RNase III cleaves certain complex RNA structures, such as those in the bacteriophage T7 early mRNA precursor at single sites. These complex structures are typically depicted as “bubbles” because they are devoid of Watson–Crick pairs; however, it is likely that they contain a precise array of non-canonical bonds and are not open loops (C).

ch05.qxd 10/9/03 6:47 PM Page 75

antisense rnas their sequence; yet RNase III makes precise cleavages at sequence-specific sites in certain RNA precursor molecules. The structure of the RNA dictates the outcome of encounters with RNase III. The ability of singlestranded RNAs to exploit and sometimes to thwart dsRNA-specific enzymes is a significant survival factor for viruses, as illustrated by bacteriophage T7, and by the mammalian viruses discussed in the next section.

E. Summary Naturally occurring antisense RNAs in prokaryotic systems are small, noncoding molecules whose structures facilitate rapid association with target RNAs. These antisense RNAs are much more than RNA molecules that happen to have sequences complementary to those of other RNAs. They are highly evolved machines designed to snare and entwine their targets. Inhibition can be a direct consequence of binding, or it can involve cellular proteins, especially endonucleases. RNase III is an endonuclease that cleaves RNA– RNA duplexes, such as those produced by certain antisense RNAs and their targets. Degradation is not the inevitable fate of an RNase III substrate. Depending on the structure of the duplex, RNase III can also introduce specific cleavages and promote RNA maturation.

III. ANTISENSE RNAS IN VIRUS-INFECTED MAMMALIAN CELLS: SIGNALS OF DANGER A. dsRNAs are potent inducers of interferon and activators of antiviral defenses Because the interferon response is central to mammalian antiviral defense, few antisense molecules can compete for significance with those producing the dsRNAs that induce interferon and otherwise contribute to the antiviral state. Despite the technical obstacles that make these dsRNAs difficult to study, they are analyzed here because of their importance to mammalian survival and because they illustrate the ability of dsRNA to act as a signal. The protective power of an intact interferon response is demonstrated by studies of transgenic mice deficient in either the type I or the type II interferon receptor. These mice rapidly succumb to viral infections that they would otherwise readily clear. For example, while the median lethal dose of intravenously administered vesicular stomatitis virus is normally in the range of 108 plaque-forming units, mutant mice lacking the type I IFN receptor die within 3–6 days after

75

receiving 30–50 units (Muller et al., 1994). A functional interferon system raises the lethal dose of this virus 2 million-fold. Mammalian cells have an extraordinarily sensitive mechanism for detecting dsRNA and for responding to dsRNA by synthesizing interferons. In 1977, Marcus and Sekellick reported that primary chick embryo cells are capable of responding to a single molecule of dsRNA. They exposed cells to defective interfering particles of vesicular stomatitis virus containing a covalently linked antisense–sense RNA molecule in a ribonucleoprotein complex. A single dsRNA molecule was presumed to form upon entry into the cell. Peak interferon titers were obtained in cultures incubated with 0.3 particles per cell. Both higher and lower doses of particles resulted in very marked reductions in interferon production, producing a bell-shaped dose– response curve. The data indicated that cells that attached two or more particles produced little or no interferon. Cells in the cultures exposed to the defective interfering particles entered a general antiviral state, as indicated by their resistance to a subsequent challenge with Sindbis virus. Control experiments ruled out the possibility that interferon induction was due to proteins in the particles. In these experiments, cells responded to intracellular dsRNA. Cells also respond to exogenously applied dsRNA. This ability may allow cells to respond to dsRNA released from their moribund neighbors. A pharmaceutical form of dsRNA has been developed for intravenous delivery to virus-infected patients. When dsRNA acts as a danger signal, it is performing a function similar to that of unmethylated CpGcontaining DNA. Such DNA is a strong immune stimulant and is interpreted by B-cells and cells of the innate immune system as a sign of microbial attack. Thus, two types of nucleic acid, dsRNA and unmethylated CpG-containing DNA, are central to mammalian defenses against infectious agents.

B. Viruses are thought to be the source of interferon-inducing antisense RNA and dsRNA, but hard evidence is very difficult to obtain Conventional wisdom holds that the dsRNAs responsible for interferon induction are entirely of viral origin. However, as stated by Jacobs and Langland, “Actually identifying the potential sources of dsRNA in infected cells has in fact been problematic over the years” ( Jacobs and Langland, 1996). The replication intermediates of RNA viruses contain both plus and minus RNAs, and are obvious potential sources of dsRNA. DNA viruses could generate dsRNA by aberrant

ch05.qxd 10/9/03 6:47 PM Page 76

76

the desk encyclopedia of microbiology

transcription or by transcription of overlapping genes encoded on opposite strands. The lack of direct evidence concerning the identity of the interferon-inducing dsRNA reflects technical difficulties that make it nearly impossible to analyze the critical dsRNA molecules. Ironically, the cell’s extreme sensitivity is a major problem; it sets a standard for dsRNA detection that existing molecular techniques cannot equal. RNA viruses generate relatively large quantities of complementary (minus) strands during the course of replication. Minus strands create a potential background problem because they can associate with sense strands during the extraction process. Particularly if the extraction is carried out in the presence of phenol, which catalyzes nucleic acid hybridization, the presence of viral dsRNA in extracts does not prove that it existed in cells. Even when dsRNA can be shown to be present in cells, for example by the use of dsRNA-specific antibodies, questions remain about whether this dsRNA was accessible to the cell’s dsRNA-sensing machinery. Magliano and colleagues demonstrated that the membrane-bound cytoplasmic vacuoles, called rubella virus “replication complexes,” are virus-modified lysosomes (Magliano et al., 1998). This compartmentalization may effectively hide the rubella virus dsRNA. Similar membranous structures have been described in other virusinfected cells. The sequestration of dsRNA may be a common viral defense strategy. Vaccinia virus, a DNA virus that produces large quantities of complementary transcripts, also produces proteins encoded by the E3L gene that bind dsRNA. This countermeasure is effective. Although vaccinia virus produces dsRNA, it is relatively resistant to interferon unless the E3L gene is mutated. Moreover, interferon resistance can be restored to mutant vaccinia by supplying RNase III (Shors and Jacobs, 1997), the bacterial endonuclease which destroys dsRNA. The ability of vaccinia to replicate in cells expressing RNase III suggests that the viral dsRNA is not required by the virus, but rather is a side product. It would be interesting to know whether RNase IIIproducing cells support the replication of mammalian RNA viruses, or if the RNA to RNA replication cycle of such viruses render them sensitive to this endonuclease. The replication intermediates of RNA bacteriophage contain little if any exposed dsRNA and are not sensitive to RNase III in vivo. The studies of rubella virus and vaccinia virus illustrate the general point that the stronger the evidence that viral dsRNA exists inside infected mammalian cells, the stronger the evidence that the dsRNA is obscured in some way. These experiments indicate that viruses produce very little dsRNA that is exposed to the interferon sensing machinery, and suggest that

the RNAs reaching the dsRNA biosensor may not be mainstream viral RNAs required for replication. They also raise the possibility that viral dsRNA may not be the only sign cells use to detect viral infection. The high concentration of viral mRNA is a potential additional stigma. It has been proposed that higher plants have a surveillance system that recognizes and eliminates RNAs whose concentrations rise above threshold limits. This system has been associated with posttranscriptional gene silencing (see Section IV).

C. Double-stranded RNA-binding proteins contribute to the antiviral state Double-stranded RNAs play two different roles in the interferon response. First, they stimulate interferon production and thus induce expression of at least 30 genes. Second, they interact with interferoninduced enzymes and thereby promote the antiviral state of the cell. Despite technical difficulties that impede direct analysis, it is possible to deduce some characteristics of these dsRNAs by studying the properties—length preferences, affinities, and concentrations—of the interferon-induced enzymes. Like dsRNA itself, at least one of these proteins, the interferon-induced RNA-dependent protein kinase (PKR), plays a dual role in the interferon response. PKR transduces the dsRNA signal, communicating to the nucleus that a virus is present. In addition, PKR and two other dsRNA-binding enzymes catalyze reactions that inhibit virus production. 1. The interferon-induced RNA-dependent protein kinase, PKR PKR levels have been measured in human Daudi cells. Each cell contains about 5 105 molecules in the cytoplasm (mostly associated with ribosomes) and 1 105 in the nucleus (mostly in the nucleolus). Following interferon treatment, the PKR concentration rises three- to fourfold, with almost all of the increase occurring in the cytoplasm. IFN-treated cells contain approximately one molecule of PKR for each ribosome ( Jeffrey et al., 1995). Ribosomes compete with dsRNA for binding to PKR (Raine et al., 1998). Ribosome-bound PKR may constitute a reserve supply that can be rapidly deployed without the need for new RNA or protein synthesis. PKR binds to and is activated by long dsRNA molecules. As is true for many interactions between dsRNAspecific enzymes and perfect duplexes, there is no discernible sequence specificity for the reactions between PKR and dsRNA. Duplexes shorter than 30 base pairs do not bind stably to PKR and do not activate the

ch05.qxd 10/9/03 6:47 PM Page 77

antisense rnas enzyme. Those longer than 30 base pairs bind with increasing efficiency, reaching a maximum at about 85 base pairs. The lack of sequence specificity allows the PKR to recognize dsRNAs regardless of their origins, satisfying a prerequisite for any broadbased antiviral response. The rather long optimal length of dsRNA makes PKR resistant to activation by the short and imperfect duplexes present in many cellular RNAs, including rRNAs. However, PKR’s requirement for perfect duplexes is not absolute. Certain RNAs lacking extensive perfect duplexes activate PKR, including a cellular RNA recently identified by Petryshyn et al. (1997). Binding to long dsRNA causes PKR to undergo an autophosphorylation reaction and become activated. This process displays a marked concentration dependence: PKR is activated by low concentrations of dsRNA (in the range of 10–100 ng/ml), but higher concentrations are less and less effective, giving rise to a bell-shaped activation curve. The shape of this curve indicates that PKR is optimally activated by a particular ratio of dsRNA to PKR. It also suggests that virus-infected cells do not contain high concentrations of accessible dsRNA. If they did, the PKR defense system would not function efficiently. Once it has been activated, PKR phosphorylates a number of proteins, most notably the translation initiation factor eIF-2. This phosphorylation inhibits protein synthesis, thereby diminishing virus production. In addition, PKR appears to transduce the dsRNA signal, at least in part, by phosphorylating I-B, releasing and activating the transcription factor NF-B. Analysis of the interferon response is progressing rapidly, producing a picture of an intricate combinatorial cascade in which PKR activation by dsRNA is an early event in a series of reactions that culminates in the antiviral state. PKR is a highly effective antiviral agent. Accordingly, several viruses have evolved strategies for neutralizing it. One of the best characterized of the anti-PKR viral products, and the one with the closest ties to dsRNA, is VAI RNA of adenovirus. In cells infected with mutant viruses deficient in VAI RNA synthesis, PKR is activated and protein synthesis comes to a halt. VAI RNA is an abundant RNA (108 copies per infected cell) that is about 160 bases long. It has enough similarity to bona fide dsRNA to bind to PKR, but lacks the structural features required for activation. Thus, it competitively inhibits activation by dsRNA. 2. The 2,5-oligo(A) synthetases The 2,5-oligo(A) synthetases are activated upon binding to dsRNA molecules. They then polymerize

77

ATP into 2,5-oligo(A), which in turn activates RNase L (a normally latent endonuclease), leading to mRNA degradation. Constituitive expression of 2,5-oligo(A) synthetase has been shown experimentally to confer resistance to picorna virus infection. In an attempt to determine the form of picorna virus RNA physically associated with the enzyme, Gribaudo and colleagues analyzed RNAs coprecipitating with 2,5-oligo(A) synthetase extracted from encephalomyocarditis virus (EMCV)-infected HeLa cells (Gribaudo et al., 1991). Precipitates contained both plus and minus EMCV RNAs. About 10% of the viral RNA was resistant to single-stranded RNA-specific ribonucleases, and thus potentially representative of preexisting dsRNA. However, the authors commented that this value might be “an overestimate resulting from the annealing of regions of complementary strands upon removing proteins which blocked the annealing.” Providing further evidence that only a small percentage of the viral RNA was doublestranded in vivo, the synthetase prepared from these cells was not fully activated. Activation could be enhanced 20-fold by incubation with artificial dsRNA [poly(I)-poly(C)]. Because both adenovirus VAI RNA (Desai et al., 1995) and heterogeneous nuclear RNA (Nilsen et al., 1982) activate 2,5-oligo(A) synthetases in vitro, extensive regions of perfect duplex structure are clearly not required for synthetase activation. Plus and minus EMCV RNAs may have stable structural elements capable of partially activating the synthetase, particularly in cells primed to respond. Further study is needed to determine whether both duplexes (composed of plus and minus viral RNAs) and free viral RNAs contribute to synthetase activation in picorna virus-infected cells.

3. The dsRNA adenosine deaminase, dsRAD The dsRNA adenosine deaminase, dsRAD, catalyzes the C-6 deamination of adenosine to yield inosine. Deamination reduces the stability of dsRNAs. In conjunction with a ribonuclease specific for inosinecontaining RNA (Scadden and Smith, 1997), dsRAD may play a role in viral defense. However, its contribution to viral defense is not as clearly established as that of the PKR kinase and the 2,5-oligo(A) synthetases. Primarily a nuclear enzyme that is expressed in virtually all mammalian cells, dsRAD contributes to normal metabolism. It has also been implicated in the production of hypermutated measles virus RNAs during chronic infection of the central nervous system. Such chronic infection can lead to a fatal degenerative neurological disease, subacute sclerosing panencephalitis.

ch05.qxd 10/9/03 6:47 PM Page 78

78

the desk encyclopedia of microbiology

The substrate preferences of dsRAD are rapidly coming to light, and will clarify its biological functions when they are fully known. The deaminase has no clear in vitro sequence specificity for action on perfect dsRNA; however, some adenosines may be preferred. For maximum modification, intermolecular or intramolecular duplexes need to contain a minimum of 100 base pairs. From the standpoint of molecular structure, it is remarkable that this enzyme acts upon continuous duplex RNA. It is able to deaminate adenosines even though the C-6 amino group of adenosine lies in the deep and narrow major groove of standard A-form RNA, a space that is usually inaccessible to amino acid side-chains. In addition to perfect duplexes, dsRAD acts on RNAs that lack extensive duplex structure. For example, it edits certain cellular pre-mRNAs, such as that of the glutamate-gated ion channel GluR. Like PKR and the 2,5-oligo(A) synthetases, dsRAD binds to adenovirus VAI RNA in vitro (Lei et al., 1998). In a manner similar to the assistance RNase III lends to bacteriophage T7, dsRAD aids the human hepatitis delta agent by deaminating a specific adenosine residue and carrying out an RNA editing event essential for the survival of this viroid-like pathogen. Exhibiting an additional similarity to RNase III, dsRAD behaves like one type of enzyme when interacting with long dsRNA molecules, in this case acting as a very robust and vigorous deaminase while it functions as a highly selective editing enzyme when interacting with single-stranded RNAs that have specific structural elements. The dsRNA adenosine deaminase is clearly an enzyme with important cellular functions, and it is sometimes exploited by pathogens. Its role in antiviral defense is less clearly established, but the fact that its level rises following interferon treatment suggests that dsRAD contributes to the antiviral state.

D. Summary Antisense RNAs play an important role in antiviral defenses by forming dsRNA. dsRNA is recognized by mammalian cells as a danger signal indicating that viral infection has occurred. A single molecule of dsRNA can induce interferon. Viruses often produce detectable amounts of dsRNA. However, in many cases, this dsRNA is obscured by other viral products. As a result, it has been difficult to pinpoint the actual dsRNA responsible for inducing the interferon response. Several cellular dsRNA-specific enzymes are induced by interferon and are thought to contribute to the antiviral state. Two of these enzymes degrade RNA, a third blocks protein synthesis. This

enzyme, the PKR, also contributes to the antiviral state by setting off a cascade that activates interferon synthesis.

IV. ARTIFICIAL RNAS, DSRNA, AND POSTTRANSCRIPTIONAL GENE SILENCING A. Artificial sense, antisense, and dsRNAs place a spotlight on gene silencing Just as Rutherford was unprepared for what happened when he shot alpha particles at a thin sheet of gold, biologists were unprepared for what occurred when they engineered plants to express sense transcripts of the chalcone synthetase A (chA) gene (van der Krol et al., 1990; Napoli et al., 1990). In many plants, both the transgene and the endogenous homolog of the transgene were silent, or co-suppressed. Thus, an attempt to overexpress genes involved in pigment formation resulted in plants with white flowers. These plants, and many studied subsequently, exhibit posttranscriptional gene silencing (PTGS), a condition in which specific RNA molecules are degraded. The examples of PTGS in plants involve many different genes, plant species, and DNA constructs. It is believed that PTGS could be produced in all plant species with most endogenous genes. Furthermore, PTGS can be used to confer virus resistance to transgenic plants expressing sense transcripts of viral genes. The RNA affected by PTGS may be the product of a transgene, an endogenous plant mRNA, or a viral RNA. According to Balcombe, who reviewed hypotheses concerning the pathway leading to PTGS, all mechanisms require the production of an antisense RNA (Baulcombe, 1996). Evidence reveals that PTGS also occurs in nematodes (Fire et al., 1998). At least in some cases, PTGS involves dsRNA molecules functioning as signals. In order to gain a full understanding of gene regulatory pathways and the biological role of RNA, it is critical to identify the molecular events leading to PTGS. To this end, PTGS is described here in three experimental settings.

B. Homology-dependent virus resistance occurs in transgenic plants Viral genes have been transformed into a wide range of plant species to obtain viral protection. Results of these experiments support a two-phase model of homologydependent resistance. In the first phase, viral and transgene RNA concentrations rise. After their combined concentration exceeds a threshold (or for some other

ch05.qxd 10/9/03 6:47 PM Page 79

antisense rnas reason), a switch is triggered in a surveillance system and a factor capable of causing gene-specific RNA turnover is produced. Appearance of this factor— which is probably either a perfect or an imperfect RNA duplex—initiates the second (maintenance) phase. During the second phase, the factor moves from the inoculated leaf to other portions of the plant, causing systemic resistance (Voinnet and Baulcombe, 1997). Like other forms of PTGS in plants, virus resistance is usually seen in only a fraction of the plants carrying a particular transgene. This variability suggests that PTGS results from a rare event. The level of virus resistance is also variable. In the most extreme cases, there is no detectable accumulation of the virus anywhere in the inoculated plant. In others, virus accumulates at least in the inoculated leaves. A plant showing intermediate resistance to tobacco etch virus (TEV) allowed the sequential events that take place during PTGS to be analyzed. This plant was initially susceptible to TEV and the transgene expressed high levels of viral sense transcripts. However, symptoms were attenuated in the (upper) leaves, which developed after inoculation. These new leaves contained little TEV RNA or RNA of the transgene. Such plants are said to have “recovered” (although the tissues showing “recovery” never developed symptoms in the first place) (Lindbo et al., 1993; Goodwin et al., 1996). This plant was resistant to challenge inoculation with a related virus, but was susceptible to infection by unrelated viruses, indicating that resistance was homology dependent. Very similar events take place during certain natural viral infections, suggesting that homologydependent virus resistance and natural resistance have common features. Ratcliff and colleagues demonstrated that Nicotiana clevelandii inoculated with tomato black ring nepovirus (strain W22) initially showed symptoms, and then recovered. Inoculated plants were resistant to challenge with a second inoculation of the same strain and were partially resistant to challenge with a related strain, BUK, which is 68% identical to W22. However, they were sensitive to an unrelated virus, potato virus X (Ratcliff et al., 1997). Thus, in both homology-dependent virus resistance in transgenic plants and in certain natural viral infections, a factor is generated in highly infected tissues that moves to other parts of the plant, rendering them resistant. The specificity of these events strongly implicates a nucleic acid, almost certainly an RNA. PTGS can be a two-way street, as illustrated by RNA viruses that have been engineered to contain inserts of nuclear genes. When such viruses replicate, they produce RNAs with sequences of the nuclear

79

genes and they inhibit expression of the nuclear gene. For example, the upper leaves of N. benthamiana inoculated with a recombinant tobacco mosiac virus containing part of the phytoene desaturase gene displayed a photobleaching effect, indicating that the desaturate gene had been inhibited (resulting in the loss of carotenoid-mediated protection) (Kumagai et al., 1995). Because the RNAs of plant viruses are thought to have a strictly cytoplasmic location, these results indicate that either all steps of PTGS can take place in the cytoplasm or that the RNAs interact with DNA during cell division following breakdown of the nuclear envelope. Waterhouse and colleagues produced strong evidence that dsRNA can trigger PTGS. They showed that N. tabaccum cv W38 engineered to express dsRNAs of the potato virus Y (PVY) protease gene were much more likely to acquire virus immunity than plants expressing either sense or antisense transcripts of this gene (Waterhouse et al., 1998). As they pointed out, the efficacy of their constructs, which were designed to produce dsRNA in which both strands of the duplex originated from transgenes (rather than duplexes in which one strand originated from the transgene and the other strand was of viral origin), argues against conventional models of antisense action. Conventional models require pairing to take place between transcripts of transgenes and their targets, while the data of Waterhouse and colleagues indicate that constructs were much more effective when their transcripts were capable of forming dsRNA on their own. It will be interesting to learn what makes such constructs more effective than those expressing antisense RNA and to identify the cellular factors interacting with the dsRNA. Proteins will almost certainly be involved in this process. Changes at the DNA level, such as altered methylation, may also occur. There are many reports of DNA methylation of transgenes associated with PTGS. For example, in tobacco plants displaying homology-dependent resistance and PTGS of a PVY transgene, the DNA of the transgene was methylated (Smith et al., 1995). At the moment, the significance of DNA methylation is not clear. It may be a cause of homology-dependent PTGS, or it may be a consequence of it. When one considers the impact RNA may have on DNA methylation patterns, it is interesting to consider the results of studies carried out on transgenic plants carrying defective viroid cDNAs. Viroids are circular RNAs that replicate through an RNA-to-RNA rolling-circle process in the nucleus. Inoculation of the transgenic plants with infectious viroid led to viroid replication and to specific methylation of the viroid cDNA sequences, even though this DNA was not the

ch05.qxd 10/9/03 6:47 PM Page 80

80

the desk encyclopedia of microbiology

template for the viroid RNA (Wassenegger et al., 1994). Unfortunately, because viroid-infected nuclei contain both free viroid RNA and viroid replication intermediates (which may have dsRNA segments of unknown length), these experiments do not reveal whether single-stranded viroid RNA or doublestranded viroid RNA induced the DNA methylation.

C. Co-suppression of nuclear Genes in plants involves RNA–RNA duplexes In studies paralleling those described above, Waterhouse and colleagues also compared the ability of various constructs to inhibit a -glucuronidase (GUS) reporter gene in rice. Some constructs were designed to produce single-stranded RNA transcripts, others to produce dsRNA. Based on their results, they concluded that co-suppression, like homology-dependent virus resistance, is triggered by dsRNA (Waterhouse et al., 1998). Their conclusion is consistent with observations made by many earlier investigators who found that PTGS is more likely to occur in plants containing two tandem copies of a transgene arranged as inverted repeats, an organization favoring the production of transcripts capable of forming dsRNA. Despite the strength of the evidence implicating a perfect duplex as the PTGS trigger, Metzlaff and colleagues have developed a model that involves an imperfect duplex. Their model is based on the profiles of calcone synthetase-specific RNAs present in wildtype Petunia and in transgenic plants manifesting PTGS. They reported that white flowers of transgenic plants had little full-length poly(A) chsA RNA, but instead had characteristic mRNA fragments. They proposed that a self-sustaining degradation cycle is set in motion when sequences from the 3-portion of an “aberrant” chsA transcript bind to partially complementary sequences in a second chsA RNA molecule, causing it to be cleaved and to release a new 3 fragment (Metzlaff et al., 1997). Significantly, fragments similar to those in the white-flowering transgenic plants accumulate in the white portions of a nontransgenic Petunia (Red Star) whose flowers have purple-white patterned flowers. This result shows that co-suppression uses steps of a preexisting control pathway. Eventually, the model proposed by Metzlaff and colleagues will need to be reconciled with the evidence that perfect dsRNA can trigger PTGS. A consistent model may emerge when the events leading to the production of the “aberrant” transcript are understood in greater detail. Further studies are also needed to define the role of DNA methylation in gene silencing. DNA methylation has been studied in Petunia and Arabidopsis manifesting

PTGS of the calcone synthetase gene. In purple Petunia flowers, an EcoRII site in the 3-end of the endogenous genes only rarely contains a methylated cytosine, whereas in leaf DNA, these sites are frequently methylated. There is, therefore, a developmentally regulated loss of methylation at these sites. In transgenic plants with white flowers, this developmental change does not occur, and these sites are frequently methylated (Flavell et al., 1998). To determine whether methylation is required for PTGS, Furner and colleagues studied transgenic Arabidposis plants that carry a mutation in a gene required for DNA methylation. PTGS was reversed and expression of the chs transgene was restored in plants defective in DNA methylation, leading these investigators to conclude that “methylation is absolutely necessary” for PTGS (Furner et al., 1998). Confirmation of this observation in another system will shed further light on the significance of DNA methylation.

D. dsRNA-induced homology-dependent posttranscriptional gene silencing takes place in Caenorhabditis elegans RNA interference in nematodes was discovered by Guo and Kemphues, who found that antisense and sense transcripts yielded identical results when used to block gene expression in the maternal germ line (Guo and Kemphues, 1995). Fire and colleagues later demonstrated that dsRNA mediates PTGS in nematodes and that these worms have a transport system that allows dsRNA-mediated interference to move across cell membranes. On a mole-per-mole basis, they found that dsRNA transcripts were about 100 times more potent than either antisense or sense transcripts. Their initial experiments involved the unc-22 gene, which encodes an abundant but nonessential myofilament protein. They injected a mixture of sense and antisense transcripts covering a 742-nucleotide segment of unc-22 into the body cavity of adults and observed robust interference in the somatic tissues of the recipients and in their progeny broods. Only a few molecules of dsRNA were required per affected cell, suggesting that the dsRNA signal may have been amplified. Several genes have been tested for susceptibility to dsRNA-mediated interference in addition to unc-22. The following observations have been made (Montgomery et al., 1998; Tabara et al., 1998; Fire et al., 1998). First, PTGS effects are usually similar to those of null mutants, indicating that the target gene has been fully and selectively inhibited. However, some expression of the target gene can occur in dsRNAtreated worms. In some cases, suppression occurs in

ch05.qxd 10/9/03 6:47 PM Page 81

antisense rnas only some cells. Second, mRNAs appear to be the targets of PTGS, rather than mRNA precursors. dsRNA segments corresponding to introns and promoter segments are not effective, as might be expected if precursors or the transcription process were the target. Furthermore, dsRNA covering upstream genes in polar operons have no effect on downstream genes, underscoring the conclusion that mRNAs, rather than precursors, are the target. In addition, cytoplasmic levels of target RNAs drop precipitously. Third, dsRNA-mediated interference is able to cross cellular boundaries. In fact, the dsRNA crosses cellular membranes so readily that it is possible to induce PTGS by feeding worms transgenic Escherichia coli expressing dsRNA covering the target gene (Timmons and Fire, 1998) or by soaking them in dsRNA—although neither of these approaches is as effective as microinjection. Fourth, PTGS passes into the F1 generation; remarkably, Tabara and colleagues report that for certain genes, “interference can be observed to transmit in the germ line apparently indefinitely” (Tabara et al., 1998).

E. The discovery of the importance of RNA interference is a major breakthrough Growing evidence indicates that dsRNA is involved in gene regulation in higher organisms. In mammalian cells, dsRNA (whether applied exogenously or synthesized endogenously) induces interferon; in plants, dsRNA has been linked to homologydependent virus resistance and to PTGS of both endogenous and transgenes; and in nematodes, dsRNA moves across cell boundaries and selectively inhibits gene expression. It was once thought that bioengineered antisense RNA would function by establishing Watson–Crick base pairs with target RNAs, thereby eliminating their function. It now appears that dsRNA mediates the effects of artificial RNAs in nematodes and in some cases of PTGS in higher plants. This dsRNA is evidently recognized by cellular factors that work in conjunction with it to destroy other RNAs of similar sequence. Most of the molecular intermediates of PTGS remain a mystery. However, DNA methylation frequently occurs during PTGS in plants and is thought to play an essential role by some investigators. When one considers the possible significance of PTGS for organisms other than vascular plants and worms, it is interesting to note that some of the mammalian genes that are subject to imprinting, a process associated with DNA methylation, express antisense transcripts (Ward and Dutton, 1998; Moorwood et al., 1998; Rougeulle et al., 1998; Reik and Constancia, 1997). As more studies are carried out on mammalian genes producing natural antisense

81

RNA and on mammalian cells synthesizing artificial antisense RNAs, mechanistic ties to PTGS may become apparent. Continued exploration of phenomena such as PTGS, homology-dependent virus resistance, and co-suppression in plants; and RNA interference in nematodes have yielded important insights into the molecular biology of eukaryotes at the most basic level. It is now clear that many eukaryotic organisms—including humans, fruit flies, plants, and fungi—have the capacity to use short dsRNAs to (down) regulate expression of homologous target sequences. Several genes and gene products that contribute to this process have been identified. Their roles in a variety of biological processes, such as embryonic development, inhibition of transposon movement, and transcriptional silencing of chromosomal genes, remain areas of very active investigation. Gene regulation that is mediated by short dsRNA molecules is now most commonly referred to as “RNA silencing”. The short dsRNAs involved in RNA silencing are part of a much larger group of noncoding RNAs that perform both structural and regulatory functions—often through complementary base pair (antisense) interactions. A five-part “Special Section”—RNA Silencing and Noncoding RNA, Science (296, 1259–1273, 2002) provides an up-to-date and wellreferenced review of RNA silencing, non-coding RNAs, and the biological functions they perform. Several proteins involved in RNA silencing have been identified. In the “classical” RNA silencing degradative pathway, long dsRNA molecules are cleaved to short duplexes (21–23 nucleotides) by an RNase III-like enzyme called the Dicer, which leaves 2- to 3-nucleotide long 3 overhangs. These short dsRNAs may bind to 250–500 kD nuclease complex called RISC (RNA-induced silencing complex). The individual strands of the short dsRNAs associate with target RNAs through complementary base pairing, perhaps using an RNA helicase to separate from each other and thus gain assess to sequences in the target mRNAs. The target RNAs are then cleaved by RISC. Should one of the individual strands of the short dsRNA bind to the target RNA in the absence of RISC, a structure results that may be amplified by an RNA dependent RNA polymerase, yielding additional long dsRNA that is a substrate for Dicer. In the fields of genomics and biotechnology, short artificial dsRNAs are being introduced into cells to knock out target genes and thereby determine its function. Short dsRNAs can be more effective inhibitors than single-standard antisense oligonucleotides, and are being considered as potential therapeutic agents, as well as research tools. While initial experiments have produced impressive results and

ch05.qxd 10/9/03 6:47 PM Page 82

82

the desk encyclopedia of microbiology

demonstrate the RNA silencing can be obtained in mammalian tissue culture cells, the extent of target gene inhibition is variable. If RNA silencing molecules can be optimized, they may provide a shortcut for deletion mutagenesis and for large scale efforts to assign functions to individual genes. RNA silencing is one intriguing component of the larger and perennially eye-opening field of RNA biochemistry. Many more exciting discoveries can be expected as the full range of RNA’s role in biology comes to light.

ACKNOWLEDGMENTS I thank Dr. Jose Walewski, Mr. Decherd Stump, and Ms. Toby Keller for help with the manuscript. This work was supported in part by NIDDK (grants R01DK52071 and P01-DK50795, project 2), the Liver Transplantation Research Fund, and the Division of Liver Diseases research funds.

BIBLIOGRAPHY Altuvia, S., and Wagner, E. G. (2000). Switching on and off with RNA. Proc. Natl. Acad. Sci. USA. 97, 9824–9826. Bass, B. L. (1997). RNA editing and hypermutation by adenosine deamination. Trends Biochem. Sci. 22, 157–162. Baulcombe, D. C. (1996). RNA as a target and an initiator of posttranscriptional gene silencing in transgenic plants. Plant Mol. Biol. 32, 79–88. Bram, R. J., Young, R. A., and Steitz, J. A. (1980). The ribonuclease III site flanking 23S sequences in the 30S ribosomal precursor RNA of E. coli. Cell 19, 393–401. Branch, A. D. (1998). A good antisense molecule is hard to find. Trends Biochem. Sci. 23, 45–50. Delihas, N., Rokita, S. E., and Zheng, P. (1997). Natural antisense RNA/target RNA interactions: Possible models for antisense oligonucleotide drug design. Nat. Biotechnol. 15, 751–753. Desai, S. Y., Patel, R. C., Sen, G. C., Malhotra, P., Ghadge, G. D., and Thimmapaya, B. (1995). Activation of interferon-inducible 2–5 oligoadenylate synthetase by adenoviral VAI RNA. J. Biol. Chem. 270, 3454–3461. Eguchi, Y., Itoh, T., and Tomizawa, J. (1991). Antisense RNA. Annu. Rev. Biochem. 60, 631–652. Engdahl, H. M., Hjalt, T. A., and Wagner, E. G. (1997). A two unit antisense RNA cassette test system for silencing of target genes. Nucleic Acids. Res. 25, 3218–3227. Fire, A., Xu, S., Montgomery, M. K., Kostas, S. A., Driver, S. E., and Mello, C. C. (1998). Potent and specific genetic interference by double-stranded RNA in Caenorhabditis elegans. Nature 391, 806–811. Flavell, R. B., O’Dell, M., and Metzlaff, M. (1998). Transgenepromoted epigenetic switches of chalcone synthase activity in petunia plants. Novartis Found. Symp. 214, 144–154. Franch, T., and Gerdes, K. (2000). U-turns and regulatory RNAs. Curr. Opin. Microbiol., 3, 159–164. Furner, I. J., Sheikh, M. A., and Collett, C. E. (1998). Gene silencing and homology-dependent gene silencing in arabidopsis: Genetic modifiers and DNA methylation. Genetics 149, 651–662.

Goodwin, J., Chapman, K., Swaney, S., Parks, T. D., Wernsman, E. A., and Dougherty, W. G. (1996). Genetic and biochemical dissection of transgenic RNA-mediated virus resistance. Plant Cell 8, 95–105. Gribaudo, G., Lembo, D., Cavallo, G., Landolfo, S., and Lengyel, P. (1991). Interferon action: Binding of viral RNA to the 40-kilodalton 2-5-oligoadenylate synthetase in interferon-treated HeLa cells infected with encephalomyocarditis virus. J. Virol. 65, 1748–1757. Guo, S., and Kemphues, K. J. (1995). par-1, a gene required for establishing polarity in C. elegans embryos, encodes a putative Ser/Thr kinase that is asymmetrically distributed. Cell 81, 611–620. Hjalt, T. A., and Wagner, E. G. (1995). Bulged-out nucleotides in an antisense RNA are required for rapid target RNA binding in vitro and inhibition in vivo. Nucleic. Acids Res. 23, 580–587. Izant, J. G., and Weintraub, H. (1984). Inhibition of thymidine kinase gene expression by anti-sense RNA: A molecular approach to genetic analysis. Cell 36, 1007–1015. Jacobs, B. L., and Langland, J. O. (1996). When two strands are better than one: The mediators and modulators of the cellular responses to double-stranded RNA. Virology 219, 339–349. Jeffrey, I. W., Kadereit, S., Meurs, E. F., Metzger, T., Bachmann, M., Schwemmle, M., Hovanessian, A. G., and Clemens, M. J. (1995). Nuclear localization of the interferoninducible protein kinase PKR in human cells and transfected mouse cells. Exp. Cell Res. 218, 17–27. Katze, M. G. (1992). The war against the interferon-induced dsRNA-activated protein kinase: Can viruses win? J. Interferon Res. 12, 241–248. Kumagai, M. H., Donson, J., della Cioppa, G., Harvey, D., Hanley, K., and Grill, L. K. (1995). Cytoplasmic inhibition of carotenoid biosynthesis with virus-derived RNA. Proc. Natl. Acad. Sci. USA 92, 1679–1683. Lee, A. J., and Crothers, D. M. (1998). The solution structure on an RNA loop-loop complex: The ColE1 inverted loop sequence. Structure 6, 993–1005. Lei, M., Liu, Y., and Samuel, C. E. (1998). Adenovirus VAI RNA antagonizes the RNA-editing activity of the ADAR adenosine deaminase. Virology 245, 188–196. Lindbo, J., Silva-Rosales, L., Proebsting, W. M., and Dougherty, W. G. (1993). Induction of a highly specific antiviral state in transgenic plants: Implications for regulation of gene expression and virus resistance. Plant Cell 5, 1749–1759. Magliano, D., Marshall, J. A., Bowden, D. S., Vardaxis, N., Meanger, J., and Lee, J. Y. (1998). Rubella virus replication complexes are virus-modified lysosomes. Virology 240, 57–63. Malmgren, C., Wagner, E. G. H., Ehresmann, C., Ehresmann, B., and Romby, P. (1997). Antisense RNA control of plasmid R1 replication. The dominant product of the antisense RNAmRNA binding is not a full RNA duplex. J. Biol. Chem. 272, 12508–12512. Marcus, P. I., and Sekellick, M. J. (1977). Defective interfering particles with covalently linked [/] RNA induce interferon. Nature 266, 815–819. Mathews, M. B., and Shenk, T. (1991). Adenovirus virus-associated RNA and translation control. J. Virol. 65, 5657–5662. Matzke, M. A., and Matzke, A. J. (1995). Homology-dependent gene silencing in transgenic plants: What does it really tell us? Trends Genet. 11, 1–3. Metzlaff, M., O’Dell, M., Cluster, P. D., and Flavell, R. B. (1997). RNA-mediated RNA degradation and chalcone synthase A silencing in petunia. Cell 88, 845–854. Mizuno, K., Chou, M. Y., and Inouye, M. (1984). A unique mechanism regulating gene expression: Translational inhibition by a complementary RNA transcript (micRNA). Proc. Natl. Acad. Sci. USA 81, 1966–1970.

ch05.qxd 10/9/03 6:47 PM Page 83

83

antisense rnas Montgomery, M. K., Xu, S., and Fire, A. (1998). RNA as a target of double-stranded RNA-mediated genetic interference in Caenorhabditis elegans. Proc. Natl. Acad. Sci. USA 95, 15502–15507. Moorwood, K., Charles, A. K., Salpekar, A., Wallace, J. I., Brown, K. W., and Malik, K. (1998). Antisense WT1 transcription parallels sense mRNA and protein expression in fetal kidney and can elevate protein levels in vitro. J Pathol 185, 352–359. Muller, U., Steinhoff, U., Reis, L. F., Hemmi, S., Pavlovic, J., Zinkernagel, R. M., and Aguet, M. (1994). Functional role of type I and type II interferons in antiviral defense. Science 264, 1918–1921. Napoli, C., Lemieux, C., and Jorgensen, R. (1990). Introduction of a chimeric chalcone synthase gene into petunia results in reversible co-suppression of homologous genes in trans. Plant Cell 2, 279–289. Nilsen, T. W., Maroney, P. A., Robertson, H. D., and Baglioni, C. (1982). Heterogeneous nuclear RNA promotes synthesis of (2,5)oligoadenylate and is cleaved by the (2,5)oligoadenlateactivated endoribonuclease. Mol. Cell Biol. 2, 154–160. Petryshyn, R. A., Ferrenz, A. G., and Li, J. (1997). Characterization and mapping of the double-stranded regions involved in activation of PKR within a cellular RNA from 3T3-F442A cells. Nucleic Acids Res. 25, 2672–2678. Proud, C. G. (1995). PKR: A new name and new roles. Trends Biochem. Sci. 20, 241–246. Raine, D. A., Jeffrey, I. W., and Clemens, M. J. (1998). Inhibition of the double-stranded RNA-dependent protein kinase PKR by mammalian ribosomes. FEBS Lett. 436, 343–348. Ratcliff, F., Harrison, B. D., and Baulcombe, D. C. (1997). A similarity between viral defense and gene silencing in plants. Science 276, 1558–1560. Reik, W., and Constancia, M. (1997). Genomic imprinting. Making sense or antisense? Nature 389, 669–671. Robertson, H. D. (1982). Escherichia coli ribonuclease III cleavage sites. Cell 30, 669–672. Rougeulle, C., Cardoso, C., Fontes, M., Colleaux, L., and Lalande, M. (1998). An imprinted antisense RNA overlaps UBE3A and a second maternally expressed transcript. Nat. Genet. 19, 15–16. Samuel, C. E. (1994). Interferon-induced proteins and their mechanisms of action. Hokkaido Igaku. Zasshi. 69, 1339–1347. Scadden, A. D., and Smith, C. W. (1997). A ribonuclease specific for inosine-containing RNA: A potential role in antiviral defence? EMBO J. 16, 2140–2149. Shors, T., and Jacobs, B. L. (1997). Complementation of deletion of the vaccinia virus E3L gene by the Escherichia coli RNase III gene. Virology 227, 77–87. Simons, R. W. (1988). Naturally occurring antisense RNA control— a brief review. Gene 72, 35–44.

Smith, H. A., Powers, H., Swaney, S., Brown, C., and Dougherty, W. G. (1995). Transgenic potato virus Y resistance in potato: Evidence for an RNA-mediated cellular response. Phytopathol 85, 864–870. Smith, H. A., Swaney, S. L., Parks, T. D., Wernsman, E. A., and Dougherty, W. G. (1994). Transgenic plant virus resistance mediated by untranslatable sense RNAs: Expression, regulation, and fate of nonessential RNAs. Plant Cell 6, 1441–1453. Tabara, H., Grishok, A., and Mello, C. C. (1998). RNAi in C. elegans: Soaking in the genome sequencing. Science 282, 430. Timmons, L., and Fire, A. (1998). Specific interference by ingested dsRNA. Nature 395, 854. van der Krol, A. R., Mur, L. A., Beld, M., Mol, J. N., and Stuitje, A. R. (1990). Flavonoid genes in petunia: Addition of a limited number of gene copies may lead to a suppression of gene expression. Plant Cell 2, 291–299. Vanhée-Brossollet, C., and Vaquero, C (1998). Do antisense transcripts make sense in eukaryotes? Gene 211, 1–9. Voinnet, O., and Baulcombe, D. C. (1997). Systemic signalling in gene silencing. Nature 389, 553. Wagner, E. G., Blomberg, P., and Nordstrom, K. (1992). Replication control in plasmid R1: Duplex formation between the antisense RNA, CopA, and its target, CopT, is not required for inhibition of RepA synthesis. EMBO J. 11, 1195–1203. Wagner, E. G., and Simons, R. W. (1994). Antisense RNA control in bacteria, phages, and plasmids. Annu. Rev. Microbiol. 48, 713–742. Ward, A., and Dutton, J. R. (1998). Regulation of the Wilm’s Tumour suppressor (WT1) gene by an antisense RNA: A link with genomic imprinting? J. Pathol. 185, 342–344. Wassenegger, M., Heimes, S., Riedel, L., and Sänger, H. L. (1994). RNA-directed de novo methylation of genomic sequences in plants. Cell 76, 567–576. Waterhouse, P. M., Graham, M. W., and Wang, M. B. (1998). Virus resistance and gene silencing in plants can be induced by simultaneous expression of sense and antisense RNA. Proc. Natl. Acad. Sci. USA 95, 13959–13964.

WEBSITES List of websites giving information about antisense RNA http://www.ambion.com/techlib/hottopics/rnai http://www.imb-jena.de/RNA.html

ch06.qxd 10/9/03 6:47 PM Page 84

6 Antiviral agents Richard J. Whitley The University of Alabama at Birmingham

GLOSSARY

hyperemia Literally excess blood; flushed, reddened, and engorged with blood. interferon Any group of glycoproteins with antiviral activity. interstitial nephritis An inflammation of the substance of kidney exclusive of the structure called the glomerulus. leukopenia Deficiency in circulating white blood cells. monotherapy Treatment with a single drug, contrasted with combination therapies with more than one drug at the same time. mucocutaneous Refers to the skin where there is both exterior skin and mucus membranes, such as the borders of the mouth. nephrotoxicity Kidney toxicity. neuraminidase An enzyme, present on the surface of some viruses, which catalyzes the cleavage of a sugar derivative called neuraminic acid. neuropathy Pathological changes in the nervous system. papillomavirus A group of viruses causing warts of various kinds. peptidomimetic A molecule having properties similar to those of a peptide or short protein. pharmacokinetic Refers to the rates and efficiency of uptake, distribution, and disposition of a drug in the body. phase III The final stage in testing of a new drug, after determination of its safety and effectiveness, in which it is tested on a broad range,

acyclic purine nucleoside analog A molecule with the structure of the normal purine components of DNA or RNA but with the sugar ring cleaved open (acyclic). alanine amino transferase An enzyme found in the liver and blood serum, the concentration of which is often elevated in cases of liver damage. antiretroviral agent Any drug used in treating patients with human immunodeficiency virus (HIV) infection. bioavailability The property of a drug to be absorbed and distributed within the body in a way that preserves its useful characteristics; for example, it is not broken down, inactivated, or made insoluble. condyloma acuminatum Venereal warts. conjunctivitis Inflammation of the conjuctiva or white of the eye. EC50 Concentration of a drug which produces a 50% effect, e.g., in virus yield. enterovirus One of a group of viruses which infect the intestinal track. Epstein–Barr virus A member of the herpesvirus family. hantavirus pulmonary syndrome A pneumonia-like illness resulting from infection with hantavirus, a virus normally carried in rodents. hepatotoxicity Liver toxicity. The Desk Encyclopedia of Microbiology ISBN: 0-12-621361-5

84

Copyright © 2003 Elsevier Ltd All rights of reproduction in any form reserved

ch06.qxd 10/9/03 6:47 PM Page 85

antiviral agents and large population of patients for comparison to existing treatments and to test for rare complications. picornavirus A group of viruses with small RNA genomes, such as poliovirus. prodrug A drug that is given in a form that is inactive and must be metabolized in the body to the active form. stromal keratitis Inflammation of the deep layers of the cornea of the eye. superficial punctate keratopathy Fine, spot-like pathological changes in the superficial layer of the cornea of the eye. t1/2 The time for reduction of some observed quantity, for example, the blood concentration of a drug, by 50%. thrombocytopenia Deficiency of platelets, the blood-clotting agents, in the blood. tubular necrosis Death of the tubule cells in the kidney. uveitis Inflammation of the iris or related structures in the eye. zoster Infection with varicella-zoster virus which leads to skin lesions on the trunk (usually) following the distribution of the sensory nerves; commonly called shingles. Antiviral Agents are drugs that are administered for therapeutic purposes to humans with viral diseases. Importantly, many people are infected by viruses but only some develop disease attributed to these microbes. Antiviral agents used to treat these diseases are currently limited and only exist for the management of herpes simplex virus, varicella-zoster virus, cytomegalovirus, hepatitis B, hepatitis C, human immunodeficiency virus, respiratory syncytial virus, human papillomavirus, and influenza virusrelated diseases. Only a few antiviral agents of proven value are available for a limited number of clinical indications. Unique problems are associated with the development of antiviral agents. First, viruses are obligate intracellular parasites that utilize biochemical pathways of the infected host cell. Second, early diagnosis of viral infection is crucial for effective antiviral therapy because by the time symptoms appear, several cycles of viral multiplication usually have occurred and replication is waning. Third, since many of the disease syndromes caused by viruses are relatively benign and self-limiting, the therapeutic index, or ratio of efficacy to toxicity, must be extremely high in order for therapy to be acceptable.

85

Fortunately, molecular biology research is helping solve two of these problems. Enzymes unique to viral replication have been identified and, therefore, distinguish between virus and host cell functions. Unique events in viral replication are sites which serve as ideal targets for antiviral agents; examples include the thymidine kinase (TK) of herpes simplex virus (HSV) or protease of human immunodeficiency virus (HIV). Second, several sensitive and specific viral diagnostic methods are possible because of recombinant DNA technology [e.g., monoclonal antibodies, DNA hybridization techniques, and polymerase chain reaction (PCR)]. This article will synthesize knowledge of the existing antiviral agents as it relates to both pharmacologic and clinical properties.

I. THERAPEUTICS FOR HERPESVIRUS INFECTIONS

A. Acyclovir and valaciclovir Acyclovir has become the most widely prescribed and clinically effective antiviral drug available to date. Valaciclovir, the L-valine ester oral prodrug of acyclovir, was developed to improve the oral bioavailability of acyclovir. Valaciclovir is cleaved to acyclovir by valine hydrolase which then is metabolized in infected cells to the active triphosphate of acyclovir.

1. Chemistry, mechanism of action, and antiviral activity Acyclovir [9-(2-hydroxyethoxymethyl) guanine], a synthetic acyclic purine nucleoside analog, is a selective inhibitor of HSV-1 and -2 and varicellazoster virus (VZV) replication. Acyclovir is converted by

ch06.qxd 10/9/03 6:47 PM Page 86

86

the desk encyclopedia of microbiology

FIGURE 6.1 The mechanism of action of acyclovir. (A) activation and (B) Inhibition of DNA synthesis and chain termination.

virus-encoded TK to its monophosphate derivative, an event that does not occur to any significant extent in uninfected cells. Subsequent di- and triphosphorylation is catalyzed by cellular enzymes, resulting in acyclovir triphosphate concentrations 40–100 times higher in HSV-infected than in uninfected cells. Acyclovir triphosphate inhibits viral DNA synthesis by competing with deoxyguanosine triphosphate as a substrate for viral DNA polymerase, as illustrated in

Fig. 6.1. Because acyclovir triphosphate lacks the 3 hydroxyl group required for DNA chain elongation, viral DNA synthesis is terminated. Viral DNA polymerase is tightly associated with the terminated DNA chain and is functionally inactivated. Also, the viral polymerase has greater affinity for acyclovir triphosphate than does cellular DNA polymerase, resulting in little incorporation of acyclovir into cellular DNA. In vitro, acyclovir is most active against

ch06.qxd 10/9/03 6:47 PM Page 87

antiviral agents

87

HSV-1 (average EC50 0.04 mg/ml), HSV-2 (0.10 g/ml), and VZV (0.50 g/ml). Epstein–Barr virus (EBV) requires higher acyclovir concentrations for inhibition, and cytomegalovirus (CMV), which lacks a virus-specific TK, is relatively resistant. Acyclovir is available in topical, oral, and intravenous preparations. Oral formulations include a 200-mg capsule, a 800-mg tablet, and suspension (200 mg/5 ml) and absorption of acyclovir results in 15–30% bioavailability. After multidose oral administration of 200 or 800 mg of acyclovir, the mean steadystate peak levels are approximately 0.57 and 1.57 g/ml, respectively. Higher plasma acyclovir levels are achieved with intravenous administration. Steady-state peak acyclovir concentrations following intravenous doses of 5 or 10 mg/kg every 8 hr are approximately 9.9 and 20.0 g/ml, respectively. The terminal plasma time for a 50% decrease in drug concentration (t1/2) is 2 or 3 hr in adults with normal renal function. Acyclovir is minimally metabolized, and approximately 85% is excreted unchanged in the urine via renal tubular secretion and glomerular filtration. Valaciclovir is only available as a tablet formulation and is metabolized nearly completely to acyclovir within minutes after absorption. Plasma levels of acyclovir, following 2 g of valaciclovir given three times a day by mouth, approximate 5 mg/kg administered every 8 h intravenously. Both acyclovir and valaciclovir must be dose adjusted if renal impairment exists.

chemotherapy. Topically applied acyclovir has no clinically beneficial effect. Orally administered acyclovir (200 mg five times daily or 400 mg three times daily) shortens the duration of virus shedding and time to healing (6 versus 7 days) when initiated within 24 h of onset, but the duration of pain and itching is not affected. Oral acyclovir administration effectively suppresses frequently recurring genital herpes. Daily administration of acyclovir reduces the frequency of recurrences by up to 80%, and 25–30% of patients have no further recurrences while taking the drug. Successful suppression for as long as 3 years has been reported, with no evidence of significant adverse effects. Titration of acyclovir (400 mg twice daily or 200 mg two to five times daily) may be required to establish the minimum dose that is most effective and economical. Asymptomatic virus shedding can continue despite clinically effective acyclovir suppression, resulting in the possibility of person-to person transmission. Valaciclovir therapy of recurrent genital herpes (either 1 g or 500 mg twice a day) is clinically equivalent to acyclovir administered at either 200 mg three times daily or five times daily. It is also effective for suppression of recurrences when 1 g per day is administered.

2. Clinical indications

Topical therapy for HSV-1 mouth or lip infections is of no clinical benefit. Orally administered acyclovir (200 or 400 mg five times daily for 5 days) reduces the time to loss of crust by approximately 1 day (7 versus 8 days) but does not alter the duration of pain or time to complete healing. Oral acyclovir therapy has modest clinical benefit but only if initiated very early after a recurrence. Valaciclovir is easier to administer and is given at 500 mg 2 day every 12 hours.

a. Genital herpes First episode genital HSV infection can be treated with topical, oral, or intravenous acyclovir. Topical application is less effective than oral or intravenous therapy. Intravenous acyclovir is the most effective treatment for first-episode genital herpes and results in a significant reduction in the median duration of viral shedding, pain, and time to complete healing (8 versus 14 days) but is reserved for patients with systemic complications. Oral therapy (200 mg five times daily) is the standard treatment. Neither intravenous nor oral acyclovir treatment alter the frequency of recurrences. While neither valaciclovir nor famciclovir have been evaluated in patients with primary genital herpes, their pharmacokinetic properties would predict efficacy. Many experienced physicians would preferentially use these drugs over acyclovir. The dose of valaciclovir is one gram t.i.d. for 7–10 days. Recurrent genital herpes is less severe and resolves more rapidly than primary infection, offering a shorter time interval for successfully antiviral

b. Herpes labialis

c. Immunocompromised host HSV infections of the lip, mouth, skin, perianal area, or genitals may be more severe in immunocompromised patients. Clinical benefit from intravenous or oral acyclovir therapy is documented as evidenced by a significantly shorter duration of viral shedding and accelerated lesion healing. Acyclovir prophylaxis of HSV infections is of significant clinical value in severely immunocompromised patients, especially those undergoing induction chemotherapy or organ transplantation. Intravenous or oral acyclovir administration reduces the incidence of symptomatic HSV infection from 70 to 5–20%. A variety of oral dosing

ch06.qxd 10/9/03 6:47 PM Page 88

88

the desk encyclopedia of microbiology

regimens, ranging from 200 mg three times daily to 800 mg twice daily, have been used successfully. d. Herpes simplex encephalitis Acyclovir therapy (10 mg/kg every 8 h for 14–21 days) reduces mortality overall from 70 to 19%. Furthermore, 38% of acyclovir recipients returned to normal neurologic function. e. Neonatal HSV infections Acyclovir treatment of babies with disease localized to the skin, eye, or mouth yielded 100% survival, whereas 18 and 55% of babies with central nervous system (CNS) or disseminated infection died, respectively. For babies with HSV localized to the skin, eye, and mouth, 98% of acyclovir recipients developed normally 2 years after infection. For babies surviving encephalitis and disseminated disease, 43 and 57% of acyclovir recipients developed normally. The currently recommended intravenous dose is 20 mg/kg every 8 h for 14–21 days. f. Varicella Oral acyclovir therapy in normal children and adolescents with chicken pox shortens the duration of new lesion formation by about 1 day, reduces total lesion count, and improves constitutional symptoms. Therapy of older patients with chicken pox (who may have more severe manifestations) is indicated, whereas treatment of younger children must be decided on a case-by-case basis. The oral dose of acyclovir is 20 mg/kg/t.i.d. upto 800 mg p.o. t.i.d. Acyclovir therapy of chicken pox in immunocompromised children substantially reduces morbidity and mortality. Intravenous acyclovir treatment (500 mg/m2 of body surface area every 8 h for 7–10 days) improved the outcome, as evidenced by a reduction of VZV pneumonitis from 45 to 5%. Oral acyclovir therapy is not indicated for immunocompromised children with chicken pox; instead, treatment is with intravenous drug. g. Herpes zoster Intravenous acyclovir therapy of herpes zoster in the normal host produces some acceleration of cutaneous healing and resolution of pain—both acute neuritis and zoster-associated pain. Oral acyclovir (800 mg five times a day) administration results in accelerated cutaneous healing and reduction in the severity of acute neuritis. Oral acyclovir treatment of zoster

ophthalmicus reduces the incidence of serious ocular complications such as keratitis and uveitis. Valaciclovir (1 g three times daily for 7–10 days) is superior to acyclovir for the reduction of pain associated with shingles. Similar data established efficacy for famciclovir as shown below. The increased frequency of significant morbidity in immunocompromised patients with herpes zoster highlights the need for effective antiviral chemotherapy. Intravenous acyclovir therapy significantly reduces the frequency of cutaneous dissemination and visceral complications of herpes zoster in immunocompromised adults. Acyclovir is the standard therapy at a dose of 10 mg/kg (body weight) or 500 mg/m2 (body surface area) every 8 h for 7–10 days. Oral acyclovir therapy in immunocompromised patients with herpes zoster likely is effective, but valaciclovir is presumably superior.

3. Antiviral resistance Resistance of HSV to acyclovir develops through mutations in the viral gene encoding TK via generation of TK-deficient mutants or the selection of mutants possessing a TK which is unable to phosphorylate acyclovir. DNA polymerase mutants also have been recovered from HSV-infected patients. Acyclovir-resistant HSV isolates have been identified as the cause of pneumonia, encephalitis, esophagitis, and mucocutaneous infections, all occurring in immunompromised patients. Acyclovir-resistant mutants have been described in the normal host but are uncommon. Acyclovirresistant isolates of VZV have been identified much less frequently than acyclovir-resistant HSV but have been recovered from marrow transplant recipients and AIDS patients. The acyclovir-resistant VZV isolates all had altered or absent TK function but remained susceptible to vidarabine and foscarnet.

4. Adverse effects Acyclovir and valaciclovir therapies are associated with few adverse effects. Renal dysfunction can occur but is relatively uncommon and usually reversible. A few reports have linked intravenous acyclovir use with CNS disturbances, including agitation, hallucinations, disorientation, tremors, and myoclonus. An Acyclovir in Pregnancy Registry has gathered data on prenatal acyclovir exposures. Though no

ch06.qxd 10/9/03 6:47 PM Page 89

antiviral agents

89

significant risk to the mother or fetus has been documented, the total number of monitored pregnancies remains too small to detect any low-frequency teratogenic events.

provides superior benefit over lower maintenance doses. Probenecid and liberal intravenous hydration have been added to prevent significant nephrotoxicity. Because of nephrotoxicity, this regimen is less attractive than oral valganciclovir therapy.

B. Cidofovir

3. Resistance The development of resistance with clinical use is uncommon; however, mutations in CMV DNA polymerase can mediate altered susceptibility. 4. Adverse events Nephrotoxicity is associated with the cidofovir administration, occurring in up to 30% of patients. Oral probenecid administration accompanies intravenously administered HPMPC in order to prevent significant nephrotoxicity.

1. Chemistry, mechanism of action, and antiviral activity Cidofovir, (S)-1-(3-hydroxy-2-phosphonomethoxypropyl) cytosine (HPMPC), is a novel acyclic phosphonate nucleoside analog and is used to reat acyclovir- and foscarnet-resistant HSV infections as well as CMV retinitis. The drug has a similar mechanism of action as the other nucleoside analog but employs cellular kinases to produce the active triphosphate metabolite. Activated HPMPC has a higher affinity for viral DNA polymerase, and therefore it selectively inhibits viral replication. The drug is less potent than ACV in vitro; however, in vivo HPMPC persists in cells for prolonged periods, increasing drug activity. In addition, HPMPC produces active metabolites with long half-lives (17–48 h), permitting once-weekly dosing. Unfortunately, HPMPC concentrates in kidney cells 100 times greater than in other tissues and produces severe proximal convoluted tubule nephrotoxicity when administered systemically. Attempts to limit nephrotoxicity include coadministration of probenecid with intravenous hydration, synthesis of cyclic congener prodrugs of HPMPC, and use of topical formulations. HPMPC has limited and variable oral bioavailability (2–26%) when tested in rats and, therefore, is administered intravenously.

C. Fomivirsen Fomivirsen is the first antisense oligonucleotide licensed for the treatment of a viral disease. 1. Chemistry, mechanism of action, and antiviral activity Fomivirsen (5-GCG TTT GCT CTT CTT-30) is approved for the treatment of CMV retinitis. The IC50 against laboratory strains of CMV is about 0.37 M. Drug binds to the mrRNA of the immediate early 2 gene of CMV. Fomivirsen can only be administered by intravitreal injection. The pharmacokinetics of drug administration to the rabbit eye indicates a half-life of 62 hours. 2. Clinical indications Fomivirsen delays progression of CMV retinitis when administered at a dosage of 330 g every other weeks on three occasions, followed by the same dose monthly. Drug is approved for patients intolerant to other medications. 3. Resistance No isolates from humans have been reported as resistant to fomivirsen.

2. Clinical indications Cidofovir is licensed for treatment of CMV retinitis and has been used to treat acyclovir-resistant HSV infection. A treatment regimen of 5 mg/kg per week for 2 weeks followed by the same dose once weekly

4. Adverse events Increased intraocular pressure and inflammation have been reported as a major side effect in as many as 20% of patients.

ch06.qxd 10/9/03 6:47 PM Page 90

90

the desk encyclopedia of microbiology

D. Foscarnet

laboratory and in the clinical setting. Isolates of HSV which are resistant to foscarnet have EC50 100 g/ml. These isolates are all DNA polymerase mutants. 4. Adverse effects

1. Chemistry, mechanism of action, and antiviral activity Foscarnet, a pyrophosphate analog of phosphonoacetic acid has potent in vitro and in vivo activity against all herpesviruses and inhibits the DNA polymerase by blocking the pyrophosphate binding site, inhibiting the formation of the 3,5 phosphodiester bond between primer and substrate and preventing chain elongation. Unlike acyclovir, which requires activation by a virusspecific TK, foscarnet acts directly on the virus DNA polymerase. Thus, TK-defi- cient, acyclovir-resistant herpesviruses remain sensitive to foscarnet. The oral bioavailability of foscarnet is poor; thus, administration is by the intravenous route. An intravenous infusion of 60 mg/kg every 8 h results in peak and trough plasma concentrations which are approximately 450–575 and 80–150 M, respectively. The cerebrospinal fluid concentration of foscarnet is approximately two-thirds of the plasma level. Renal excretion is the primary route of clearance of foscarnet, with 80% of the dose appearing in the urine. Bone sequestration also occurs, resulting in complex plasma elimination.

Although foscarnet has significant activity in the management of herpesvirus infections, nephrotoxicity, including acute tubular necrosis and interstitial nephritis, can occur. Metabolic aberrations of calcium, magnesium, phosphate, and other electrolytes are associated with foscarnet administration and warrant careful monitoring. Symptomatic hypocalcemia and resultant seizures are the most common metabolic abnormality. Increases in serum creatinine will develop in one-half of patients who receive medication but usually are reversible after cessation. Other CNS side effects include headache (25%), tremor, irritability, and hallucinations.

E. Ganciclovir and valganciclovir

2. Clinical indications Foscarnet is licensed for the treatment of CMV retinitis as well as HSV and VZV disease caused by acycloviror penciclovir-resistant viruses. Administration of foscarnet at 60 mg/kg every 8 h for 14–21 days followed by maintenance therapy at 90–120 mg/kg per day is associated with stabilization of retinal disease in approximately 90% of patients. However, as is with the case with ganciclovir therapy of CMV retinitis, relapse occurs. Mucocutaneous HSV infections and those caused by VZV in immunocompromised hosts can be treated with foscarnet at dosages lower than that for the management of CMV retinitis. Foscarnet dosages of 40mg/kg administered every 8hr for 7 days or longer will result in cessation of viral shedding and healing of lesions in the majority of patients. However, relapses will occur which may or may not be amenable to acyclovir therapy. 3. Resistance Isolates of HSV, CMV, and VZV have all been demonstrated to develop resistance to foscarnet both in the

1. Chemistry, mechanism of action, and antiviral activity Ganciclovir [9-(1,3-dihydroxy-2-propoxymethyl) guanine] (Cytovene) has enhanced in vitro activity against all herpesviruses as compared to acyclovir, including an 8–20 times greater antiviral activity against CMV. Like acyclovir, the activity of ganciclovir in herpesvirus-infected cells depends on phosphorylation by virus-induced TK. Also like acyclovir, ganciclovir monophosphate is further converted to its di- and triphosphate derivatives by cellular kinases. In cells infected by HSV-1 or -2, ganciclovir triphosphate competitively inhibits the incorporation of guanosinetriphosphate into viral DNA. Ganciclovir triphosphate is incorporated at internal and terminal sites of viral DNA, inhibiting DNA synthesis. The mode of action of ganciclovir against CMV is mediated by a protein kinase, UL-97, that efficiently promotes the obligatory initial phosphorylation of ganciclovir to its monophosphate.

ch06.qxd 10/9/03 6:47 PM Page 91

antiviral agents The oral bioavailability of ganciclovir is poor (5–7%). Peak plasma levels are approximately 1.0 g/ml after administration of 1 g every 6 h. Intravenous administration of a standard dose of 5 mg/kg will result in peak and trough plasma concentrations of 8–11 and 0.5–1.2 g/ml, respectively. Concentrations of ganciclovir in biologic fluids, including aqueous humor and cerebrospinal fluid (CSF), are less than plasma levels. The plasma elimination t1/2 is 2–4 h for individuals with normal renal function. The kidney is the major route of clearance of the drug, and therefore, impaired renal function requires adjustment of dosage. Valganciclovir is orally bioavailable (approximately 60%) and is rapidly converted to ganciclovir after absorption. It is currently in clinical development. Valganciclovir, L-valine, 2-[2-amino-1,6-dihydro-6oxo-9H-purin-9-yl)methoxy]-3-hydroxypropyl ester, is metabolized completely to ganciclovir; thus, it has the same spectrum of activity and mechanism of activity as the parent compound.

2. Clinical indications a. HIV-infected patients Ganciclovir has been administered to large numbers of patients with AIDS having CMV retinitis. Most patients (78%) experience either improvement or stabilization of their retinitis based on fundoscopic exams compared to historical controls. Induction therapy is usually at a dosage of 5.0 mg/kg twice a day given intravenously for 14–21 days. Maintenance therapy is essential. Median time to relapse for patients receiving no maintenance therapy averages 47 days. Maintenance therapy of 25–35 mg/kg per week significantly lengthens median time to relapse to 105 days. Virtually every patient treated will experience either a cessation or reduction of plasma viremia. Visual acuity usually stabilizes at pretreatment levels but rarely improves dramatically. Relapse occurs quickly in the absence of maintenance therapy but usually occurs eventually, even in patients receiving maintenance therapy (5 mg/kg for 5–7 days per week). The significance of bone marrow toxicity must be taken into consideration since 30–40% of patients develop neutropenia. Benefit has been reported with the use of ganciclovir for the treatment of other CMV infections, particularly in those involving the gastrointestinal tract. Ganciclovir can be administered orally for prevention of CMV disease and retinitis in patients with AIDS. The utilization of ganciclovir at dosages of 1 g three to six times daily, following intravenous

91

induction therapy, provides a sustained period prior to the next episode of reactivated retinitis at similar, albeit less (but not significantly less) intervals as when drug is given intravenously. Valganciclovir is comparable to ganciclovir for the treatment of CMV retinitis. The dose is 900 mg twice daily for 3 weeks followed by 900 mg once daily. b. Transplant recipients Prophylaxis and preemptive therapy of CMV infections in high-risk transplant recipients is common. Both prevention and therapy of CMV infection of the lung are amenable to ganciclovir therapy. Ganciclovir of CMV pneumonia in conjunction with CMV immune globulin is therapeutically beneficial. Ganciclovir has been administered in anticipation of CMV disease to bone marrow transplant recipients (preemptive therapy). Several clinical trials utilizing different designs (e.g., initiation of ganciclovir after engraftment versus at the time of documentation of infection by bronchial alveolar lavage but in the absence of clinical symptomatology) have established the effectiveness of ganciclovir in preventing CMV pneumonia and reducing mortality during the treatment period. The utilization of ganciclovir in these circumstances has support among transplant physicians; however, long-term survival benefit (120 days) is not apparent. Valganciclovir is under investigation in organ transplant recipients. 3. Resistance Resistance to CMV is associated with a deteriorating clinical course. Two mechanisms of resistance to ganciclovir have been documented: (i) the alteration of protein kinase gene, UL-97, reduces intracellular phosphorylation of ganciclovir, and (ii) point mutations in the viral DNA polymerase gene. Resistance is associated with decreased sensitivity up to 20-fold. 4. Adverse effects The most important side effects of ganciclovir therapy are the development of neutropenia and that of thrombocytopenia. Neutropenia occurs in approximately 24–38% of patients. The neutropenia is usually reversible with dosage adjustment of ganciclovir, including withholding of treatment. Thrombocytopenia occurs in 6–19% of patients. Ganciclovir has gonadal toxicity in animal models, most notably as a potent inhibitor of spermatogenesis. It causes an increased incidence of tumors in the preputial gland of male mice, a finding of unknown

ch06.qxd 10/9/03 6:47 PM Page 92

92

the desk encyclopedia of microbiology

significance. As an agent affecting DNA synthesis, ganciclovir has carcinogenic potential.

edema of the eyelids, and superficial punctate keratopathy.

F. Idoxuridine and trifluorothymidine

G. Penciclovir and famciclovir

1. Chemistry, mechanism of action, and antiviral activity 1. Chemistry, mechanism of action, and antiviral activity Idoxuridine (5-iodo-2-deoxyuridine) and trifluorothymidine (trifluridine, Viroptic) are analogs of thymidine. When administered systemically, these nucleosides are phosphorylated by both viral and cellular TK to active triphosphate derivatives which inhibit both viral and cellular DNA synthesis. The result is antiviral activity but also sufficient host cytotoxicity to prevent the systemic use of these drugs. The toxicity of these compounds is not significant when applied topically to the eye in the treatment of HSV keratitis. Both idoxuridine and trifluorothymidine are effective and licensed for treatment of HSV keratitis. Topically applied idoxuridine or trifluorothymidine will penetrate cells of the cornea. Low levels of drugs can be detected in the aqueous humor. 2. Clinical indications Trifluorothymidine is the most efficacious of these compounds. These agents are not of proven value in the treatment of stromal keratitis or uveitis, although trifluridine is more likely to penetrate the cornea and, ultimately, may prove beneficial for these conditions. 3. Resistance Little effort has been directed to evaluating HSV isolates obtained from the eye, in large part because of the difficulty in accomplishing this task. 4. Adverse effects The ophthalmic preparation of idoxuridine and trifluridine causes local irritation, photophobia,

A new member of the guanine nucleoside family of drugs is famciclovir [9-(4-hydroxy-3-hydroxymethylbut-1-yl) guanine; Famvir], the prodrug of penciclovir. Penciclovir does not have significant oral bioavailability (5%), but famciclovir is orally bioavailable (approximately 77%) and has a good therapeutic index for the therapy of both HSV and VZV infections. Famciclovir is the diacetyl ester of 6-deoxy penciclovir. When administered orally, it is rapidly converted to penciclovir. The spectrum of activity of penciclovir is similar to that of acyclovir. Penciclovir is phosphorylated more efficiently than acyclovir in HSV- and VZVinfected cells. Host cell kinases phosphorylate both penciclovir and acyclovir to a small but comparable extent. The preferential metabolism in HSV and VZVinfected cells is the major determinant of its antiviral activity. Penciclovir triphosphate has, on average, a 10-fold longer intracellular half-life than acyclovir triphosphate in HSV-1, HSV-2, and VZV-infected cells after drug removal. Penciclovir triphosphate is formed at sufficient concentrations to be an effective inhibitor of viral DNA polymerase, albeit at a lower Ki than that of acyclovir triphosphate. Both compounds have good activity against HSV-1, HSV-2, and VZV. The activity of penciclovir in vitro, like acyclovir, is dependent on both the host cell and the assay (plaque reduction, virus yield, and viral DNA inhibition). The mean penciclovir EC50 standard deviation for HSV-1 in MRC-5, HEL, WISH, and W138 cells is 0.4 0.2, 0.6 0.4, 0.2 0.2, and 1.8 0.8 g/ml, respectively. For HSV-2, similar levels of activity in the identical cell lines are 1.8 0.6, 2.4 2.5, 0.8 0.1, and 0.3 0.2 g/ml, respectively. These assays utilize a plaque reduction procedure. In virus yield reduction assays, inhibition of VZV replication in MRC-5 cells is between 3.0 and 5.1 g/ml, values virtually identical to those of acyclovir. Penciclovir, like acyclovir, is

ch06.qxd 10/9/03 6:47 PM Page 93

antiviral agents relatively inactive against CMV and EBV. Penciclovir is also active against hepatitis B. Conversion of famciclovir to penciclovir occurs at two levels. The major metabolic route of famciclovir is de-acetylation of one ester group as the prodrug crosses the duodenal barrier of the gastrointestinal tract. The drug is transported to the liver via the portal vein where the remaining ester group is removed and oxidation occurs at the sixth position of the side chain, resulting in penciclovir, the active drug. The first metabolite which appears in the plasma is almost entirely the de-acetylated compound, with little or no parent drug detected. Thus, the major metabolite of famciclovir is penciclovir. Pharmacokinetic parameters for penciclovir are linear over famciclovir oral dose ranges of 125–750 mg. Penciclovir is eliminated rapidly and almost unchanged by active tubular secretion and glomerular filtration by the kidneys. The elimination t1/2 in healthy subjects is approximately 2 h.

93

those of acyclovir. Namely, resistance variants can be attributed to alterations or deficiencies of TK and DNA polymerase. 4. Adverse effects Therapy with oral famciclovir is well tolerated, being associated only with headache, diarrhea, and nausea—common findings with other orally bioavailable antiviral agents. Preclinical studies of famciclovir indicated that chronic administration was tumorigenic (murine mammary tumors) and causes testicular toxicity in other rodents.

H. Vidarabine

2. Clinical indications Famciclovir is available in an oral preparation. Penciclovir is available for topical therapy (Denavir). a. Herpes zoster Famciclovir (250, 500, or 750 mg three times a day) therapy is equivalent to the standard acyclovir treatment and superior to no therapy of herpes zoster for cutaneous healing, and in a subgroup analysis it accelerated resolution of pain (zoster-associated pain). b. Genital HSV infection Studies of patients with recurrent gential HSV infection (either intravenous penciclovir or oral famciclovir therapy) indicate beneficial effects in acceleration of all clinical parameter (e.g., pain, virus shedding, and duration). Famciclovir is given twice daily (125, 250 or 500 mg twice daily for 5 days). Famciclovir therapy on recurring HSV infections of immunocompromised hosts also is effective as suppressive therapy. c. Herpes labialis Topical application of penciclovir (Denavir) accelerates lesion healing (1 day) and resolution of pain. It is available over-the-counter in many countries. 3. Resistance Herpes simplex virus and VZV isolates resistant to penciclovir have been identified in the laboratory. These isolates have similar patterns of resistance as

1. Chemistry, mechanism of action, and antiviral activity Vidarabine (vira-A, adenine arabinoside, and 9Darabinofuranosyl adenine) is active against HSV, VZV, and CMV. Vidarabine is a purine nucleoside analog that is phosphorylated intracellularly to its mono-, di, and triphosphate derivatives. The triphosphate derivative competitively inhibits DNA dependent DNA polymerases of some DNA viruses approximately 40 times more than those of host cells. In addition, vira-A is incorporated into terminal positions of both cellular and viral DNA, thus inhibiting elongation. Viral DNA synthesis is blocked at lower doses of drug than is host cell DNA synthesis, resulting in a relatively selective antiviral effect. However, large doses of vira-A are cytotoxic to dividing host cells. The benefit demonstrated in initial placebocontrolled clinical trials of this drug was a major impetus for the development of antiviral therapies. However, because of poor solubility and some toxicity, it was quickly replaced by acyclovir in the physician’s armamentarium. Today, it is no longer available as an intravenous formulation. Vidarabine should be recognized historically as the first drug licensed for systemic use in the treatment of a viral infection.

ch06.qxd 10/9/03 6:47 PM Page 94

94

the desk encyclopedia of microbiology

2. Clinical indications Vidarabine is only available as a topical formulation for ophthalmic administration. 3. Resistance Studies of resistance to vidarabine have not been pursued. 4. Adverse effects Ocular toxicity consists of occasional hyperemia and increased tearing, both of low incidence.

II. THERAPEUTICS FOR RESPIRATORY VIRUS INFECTIONS A. Amantadine and rimantadine

1. Chemistry, mechanism of action, and antiviral activity Amantadine (1-adamantane amine hydrochloride; Symmetrel) is a tricyclic amine which is effective against all influenza A variants. Amantadine has a narrow spectrum of activity, being useful only against influenza A infections. Rimantidine is the -methyl derivative of amantadine (-methyl-1-adamantane methylamine hydrochloride). Rimantidine is 5- to 10fold more active than amantadine and has the same spectrum of activity, mechanism of action, and clinical indications. Rimantadine is slightly more effective against type A viruses at equal concentrations. The mechanism of action of these drugs relates to the influenza A virus M2 protein, a membrane protein which is the ion channel for this virus. By interfering with the function of the M2 protein, amantadine and rimantidine inhibit the acid-mediated association of the matrix protein from the ribonuclear protein complex within endosomes. This event occurs early in the viral replicate cycle. The consequences of this drug are the potentiation of acidic pH-induced conformational changes in the viral hemagglutinin during its intracellular transport.

Absorption of rimantadine is delayed compared to that of amantadine, and equivalent doses of rimantadine produce lower plasma levels compared to amantadine, presumably because of a larger volume of distribution. Both amantadine and rimantadine are absorbed after oral administration. Amantadine is excreted in the urine by glomerular filtration and, likely, tubular secretion. It is unmetabolized. The plasma elimination, t1/2, is approximately 12–18 h in individuals with normal renal function. However, the elimination, t1/2, increases in the elderly with impaired creatinine clearance. Rimantadine is extensively metabolized following oral administration, with an elimination t1/2 which averages 24–36 h. Approximately 15% of the dose is excreted unchanged in the urine. 2. Clinical indications Amantadine and rimantadine are licensed both for the chemoprophylaxis and treatment of influenza A infections. The efficacy of amantadine and rimantadine when used prophylactically for influenza A infections averages 70–80% (range, 0–100%), which is approximately the same as with influenza vaccines. Effectiveness has been demonstrated for prevention of both experimental (i.e., artificial challenge) and naturally occurring infections for all three major subtypes of influenza A. Because of a lower incidence of side effects associated with rimantadine, it is used preferentially. Rimantadine can be given to any unimmunized member of the general population who wishes to avoid influenza A, but prophylaxis is especially recommended for control of presumed influenza outbreaks in institutions housing high-risk persons. High-risk individuals include adults and children with chronic disorders of the cardiovascular or pulmonary system requiring regular follow-up or hospitalization during the preceding year as well as residents of nursing homes and other chronic-care facilities housing patients of any age with chronic medical conditions. These drugs are also effective for the treatment of influenza A. All studies showed a beneficial effect on the signs and symptoms of acute influenza as well as a significant reduction in the quantity of virus in respiratory secretions at some time during the course of infection. Because of the short duration of disease, therapy must be administered within 48 h of symptom onset to show benefit. 3. Resistance Rimantadine-resistant strains of influenza have been isolated from children treated for 5 days. There have

ch06.qxd 10/9/03 6:47 PM Page 95

antiviral agents been subsequent reports of rimantadine-resistant strains being transmitted from person to person and producing clinical influenza. Development of resistance of influenza A viruses is mediated by single nucleotide changes in RNA segment 7, which results in amino acid substitutions in the transmembrane of the M2 protein. Obviously, amantadine and rimantadine share cross-resistance. 4. Adverse effects Amantadine is reported to cause side effects in 5–10% of healthy young adults taking the standard adult dose of 200 mg/day. These side effects are usually mild and cease soon after amantadine is discontinued, although they often disappear with continued use of the drug. Central nervous system side effects, which occur in 5–33% of patients, are most common and include difficulty in thinking, confusion, lightheadedness, hallucinations, anxiety, and insomnia. More severe adverse effects (e.g., mental depression and psychosis) are usually associated with doses exceeding 200 mg daily. About 5% of patients complain of nausea, vomiting, or anorexia. Rimantadine appears better tolerated. Side effects associated with rimantadine administration are significantly less than those encountered with amantadine, particularly of the CNS. Rimantadine has been associated with exacerbations of underlying seizure disorders.

B. Oseltamivir 1. Chemistry, mechanism of action, and antiviral activity

95

usage by 30% to 40%. Prophylactic efficacy is reported to be 75% to 85%. 3. Resistance Mutations in the neuraminidase have been detected rarely in patients exposed to medication. In clinical studies, 1.3% to 8.6% of posttreatment isolates have altered susceptibility to oseltamivir. In vitro, the emergence of a resistant variant occurs with the substitution of a lysine for the conserved arginine at amino acid 292 of the neuraminidase. 4. Adverse events Oseltamivir is generally well tolerated. Nausea with or without vomiting occurs in about 10% of the patients. Food alleviates this side effect.

C. Zanamivir 1. Chemistry, mechanism of action, and antiviral activity Zanamivir (5-acetylamino-4-[aminoiminomethylamino]-2,6-anhydro-3,4,5-trideoxy-D-glycero-D-galactonon-2-enonic acid) is a neuraminidase inhibitor. It binds competitively to influenza neuraminidase, habiting both influenza A and B. Influenza neuraminidase catalyzes the cleavage of the terminal sialic acid attached to glycolipids and glycoproteins. The oral bioavailability of zanamivir is poor, about 2%, thus it is only available as an inhaled medication.

Oseltamivir [ethyl (3R,a4R,5S)-4-acetomido-5-amino3-(1-ethylpropoxy)-1cyclohexene-1-carboxylate] is a selective neuroaminidase inhibitor. It inhibits both influenza A and B virus at concentrations of 2 nM. Drug inhibits viral replication by targeting the neuraminidase protein via binding in a competitive fashion to the enzyme, rendering the virus incapable of reproducing. Because it has activity against influenza B, it has an advantage over the adamantadines. It has no activity against any other virus.

2. Clinical indications

2. Clinical indications

Resistance is an uncommon occurrence in clinical trials, occurring no more frequently than in 1% of exposed patients. The site of mutation is that where drug binds the neuraminidase.

Oseltamivir is licensed for the treatment and prevention of influenza A and B infections for individuals 2 years of age and older. Clinical trials indicate 30% acceleration in resolution of clinical symptoms. In pediatric studies, treatment accelerates disease resolution and is associated with a significantly decreased incidence of otitis media and antibiotic

Zanamivir is licensed for the treatment and prevention of influenza A and B infections in patients over 7 years of age. In clinical trials, treatment reduced the duration of symptoms from 6 to 5 days and symptom scores by about 44%. The prophylactic efficacy of zanamivir is about 80%. 3. Resistance

4. Adverse events Most adverse effects are related to the respiratory tree. These include rhinorrhea and, rarely, bronchospasm.

ch06.qxd 10/9/03 6:47 PM Page 96

96

the desk encyclopedia of microbiology

Nausea and vomiting have been reported in less than 3%.

D. Ribavirin

exposure. Although respiratory secretions will contain milligram quantities of drug, only microgram quantities (0.5–3.5 g/ml) can be detected in the plasma. The kidney is the major route of clearance of drug, accounting for approximately 40%. Hepatic metabolism also contributes to the clearance of ribavirin. Notably, ribavirin triphosphate concentrates in erythrocytes and persists for a month or longer. Likely, the persistence of ribavirin in erythrocytes contributes to its hematopoietic toxicity. 2. Clinical indications a. Respiratory syncytial virus

1. Chemistry, mechanism of action, and antiviral activity Ribavirin ( -methyl-1-adamantane methylamine hydrochloride) has antiviral activity against a variety of RNA and DNA viruses. Ribavirin is a nucleoside analog whose mechanisms of action are poorly understood and probably not the same for all viruses; however, its ability to alter nucleotide pools and the packaging of mRNA appears important. This process is not virus specific, but there is a certain selectivity in that infected cells produce more mRNA than noninfected cells. A major action is the inhibition by ribavirin-5-monophosphate of inosine monophosphate dehydrogenase, an enzyme essential for DNA synthesis. This inhibition may have direct effects on the intracellular level of GMP; other nucleotide levels may be altered, but the mechanisms are unknown. The 5-triphosphate of ribavirin inhibits the formation of the 5-guanylation capping on the mRNA of vaccinia and Venezuelan equine encephalitis viruses. In addition, the triphosphate is a potent inhibitor of viral mRNA (guanine-7) methyltransferase of vaccinia virus. The capacity of viral mRNA to support protein synthesis is markedly reduced by ribavirin. Of note, high concentrations of ribavirin also inhibit cellular protein synthesis. Ribavirin may inhibit influenza A RNA-dependent RNA polymerase. Ribavirin can be administered orally (bioavailability of approximately 40–45%) or intravenously. Aerosol administration has become standard for the treatment of respiratory synctial virus (RSV) infections in children. Oral doses of 600 and 1200 mg result in peak plasma concentrations of 1.3 and 2.5 g/ml, respectively. Intravenous dosages of 500 and 1000 mg result in 17 and 24 g/ml plasma concentrations, respectively. Aerosol administration of ribavirin results in plasma levels which are a function of the duration of

While ribavirin is licensed for the treatment of carefully selected, hospitalized infants and young children with severe lower respiratory tract infections caused by RSV, it is no longer used. Use of aerosolized ribavirin in adults and children with RSV infections reduced the severity of illness and virus shedding. In patients receiving 8 or more hours of continuous therapy, the mean peak level in tracheal secretions may be 100 times greater than the minimum inhibitory concentration preventing RSV replication in vitro. The use of ribavirin for the treatment of RSV infections is controversial and remains discretionary. It is under study for prevention of RSV pneumonia in bone marrow transplant recipients. Combination ribavirin and pegylated IFN therapy is licensed for the treatment of hepatitis C. 3. Resistance Emergence of viruses resistant to ribavirin has not been documented. 4. Adverse effects Adverse effects attributable to aerosol therapy with ribavirin of infants with RSV include bronchospasm, pneumothorax in ventilated patients, apnea, cardiac arrest, hypotension, and concomitant digitalis toxicity. Pulmonary function test changes after ribavirin therapy in adults with chronic obstructive pulmonary disease have been noted. Reticulocytosis, rash, and conjunctivitis have been associated with the use of ribavirin aerosol. When given orally or intravenously, transient elevations of serum bilirubin and the occurrence of mild anemia have been reported. Ribavirin has been found to be teratogenic and mutagenic in preclinical testings. This drug is therefore contraindicated in women who are or may become pregnant during exposure to the drug. Concern has been expressed about the risk to persons in the room of infants being treated with ribavirin aerosol, particularly females of childbearing age. Although this

ch06.qxd 10/9/03 6:47 PM Page 97

antiviral agents risk seems to be minimal with limited exposure, awareness and caution are warranted. Furthermore, the use of a “drug salvage” hood is mandatory. 5. Hepatitis C With interferon- , ribavirin is approved for combination therapy of chronic hepatitis C (see Section III.A).

III. HEPATITIS AND PAPILLOMAVIRUS A. Interferons 1. Chemistry, mechanism of action, and antiviral activity Interferons (IFNs) are glycoprotein cytokines (intracellular messengers) with a complex array of immunomodulating, antineoplastic, and antiviral properties. Interferons are currently classified as , , or , the natural sources of which, in general, are leukocytes, fibroblasts, and lymphocytes, respectively. Each type of IFN can be produced via recombinant DNA technology. Binding of IFN to the intact cell membrane is the first step in establishing an antiviral effect. Interferon binds to specific cell surface receptors; IFN- appears to have a different receptor from those of IFN- and - which may explain the purported synergistic antiviral and antitumor effects sometimes observed when IFN- is given with either of the other two IFN species. A prevalent view of IFN action is that, following binding, there is synthesis of new cellular RNAs and proteins, particularly protein kinase R, which mediate the antiviral effect. Chromosome 21 is required for this antiviral state in humans, no matter which species of IFN is employed. At least three of the newly synthesized proteins in IFN-treated cells appear to be associated with the development of an antiviral state: (i) 25-oligoadenylate synthetase, (ii) a protein kinase, and (iii) an endonuclease. The antiviral state is not fully expressed until these primed cells are infected with virus. Interferon must be administered intramuscularly or subcutaneously (including into a lesion such as a wart). Plasma levels are dose dependent, peaking 4–8 h after intramuscular administration and returning to baseline between 18 and 36 h. There appears to be some variability in absorption between each of the three classes of IFN and, importantly, resultant plasma levels. Leukocyte and IFN- appear to have elimination t1/2 values of 2–4 h. Interferon is inactivated by various organs of the body in an as yet undefined method.

97

2. Clinical indications a. Condyloma acuminatum Several large, controlled trials have demonstrated the clinical benefit of IFN- therapy of condyloma acuminatum which was refractory to cytodestructive therapies. Administration of 1.0 106 International Units (IU) of recombinant IFN- led to significant benefit as evidenced by enhancing clearing of treated lesions (36 vs 17% placebo recipients) and by reduction in mean wart area (40% reduction vs 46% increase). In other well-controlled studies, either a similar rate (46%) or higher rates (62%) of clearance were reported. Notably, clearing responses of placebo recipients averaged 21 or 22%. b. Hepatitis B Hepatitis B DNA polymerase level, a marker of replication, is reduced with IFN therapy. Treatment with IFN- in chronic hepatitis B has subsequently been investigated in several large, randomized, controlled trials. Clearance serum HBeAg and hepatitis B virus (HBV)–DNA polymerase occurs with treatment (30–40%). c. Hepatitis C The activity of IFN as a treatment of hepatitis C has undergone extensive evaluation. Interferon dosages have ranged from 1 106 to 10 106 IU three times weekly for 1–18 months. Of the placebo controls, only 2.6% normalized serum alanine amino transferase (ALT). In contrast, treatment led to serum ALT normalization in 33–45% of patients. Unfortunately, 50–80% of patients relapsed. Recently, IFN- has been administered with ribavirin. Concomitant therapy for 40 weeks resulted in sustained responses in more than 60% of patients. 3. Resistance Resistance to administered interferon has not been documented although neutralizing antibodies to recombinant interferons have been reported. The clinical importance of this latter observation is unknown. 4. Adverse effects Side effects are frequent with IFN administration and are usually dose limiting. Influenza-like symptoms (i.e., fever, chills, headache, and malaise) commonly occur, but these symptoms usually become less severe with repeated treatments. At doses used in the treatment of condyloma acuminatum, these side effects rarely cause termination of treatment. For local

ch06.qxd 10/9/03 6:47 PM Page 98

98

the desk encyclopedia of microbiology

treatment (intralesional administration), pain at the injection site does not differ significantly from that for placebo-treated patients and is short-lived. Leukopenia is the most common hematologic abnormality, occurring in up to 26% of treated patients. Leukopenia is usually modest, not clinically relevant, and reversible upon discontinuation of therapy. Increased alanine aminotransferase levels may also occur as well as nausea, vomiting, and diarrhea. At higher doses of IFN, neurotoxicity is encountered, as manifested by personality changes, confusion, loss of attention, disorientation, and paranoid ideation. Early studies with IFN- show similar side effects as those of treatment with and IFN- and - but with the additional side effects of dose-limiting hypotension and a marked increase in triglyceride levels.

B. Adefovir dipivoxil 1. Chemistry, mechanism of action, and antiviral activity Adefovir dipivoxil, bis-pivaloyloxymethyl-9-(2-phosphonyl-methoxyethyl)adenine, is the orally bioavailable prodrug of adefovir.

treatment of chronic hepatitis B. Phase III trials are in progress.

IV. PROSPECTS FOR ENTEROVIRAL THERAPIES Pleconaril, a compound with activity against many rhinoviruses and enteroviruses, is the first compound for which data exist to define anti-viral drug interaction with a virion at the atomic level. This compound is one of a class of compounds which resembles arildone, a drug known to inhibit uncoating of poliovirus. X-ray diffraction studies of bound to rhinovirus 14 show that the compound adheres tightly to a hydropic pocket formed by VP1, one of the structural proteins of rhinovirus 14. These hydrophobic pockets were found in the VP1 proteins of poliovirus and meningovirus and may be common to all picomaviruses. These compounds may lock into the conformation of the VP1 so that the virus cannot disassemble. Pleconaril is under investigation for chronic enterovirus infections of the CNS in the immune deficient patients. It was shown to have an inadequate therapeutic index for therapy of rhinovirus cold.

2. Clinical indications Adefovir has activity against both herpes and hepadnavirus. It is in the nucleotide class of medications. Treatment of chronic hepatitis B at 10 mg daily significantly decreases HBV DNA polymerase (3.56 logs compared with 0.55 logs in placebo recipients), improves hepatitic hitopathology scores, and induces loss of HBeAg.

V. ANTIRETROVIRAL AGENTS A. Reverse transcriptase inhibitors 1. Zidovudine

3. Resistance Mutations within the HBV DNA polymerase which confer resistance to adefovir have not been identified in clinical trials. HBV isolates resistant to lamivudine or hepatitis B hyperimmune globulin retain susceptibility to adefovir. 4. Adverse events Severe acute exacerbation of hepatitis has been reported who have discontinued anti-HBV therapy. Lactic acidosis and severe hepatomegaly with steatosis have also been reported.

C. Entecavir [1S-(1,3,4)]-2-amino-1,9-dihydro-9[4-hydroxymethyl-2-methyllenecyclopentyl]-6H-purin-6-one, is a nucleoside analog that is orally bioavailable for the

a. Chemistry, mechanism of action, and antiviral activity Zidovudine (3-azido-2,3-dideoxythymidine; azidothymidine and Retrovir) is a pyrimidine analog with an azido group substituting for the 3 hydroxyl group on the ribose ring. The drug is initially phosphorylated by cellular TK and then to its diphosphate by cellular thymidylate kinase. The triphosphate derivative competitively inhibits HIV reverse transcriptase and also functions as a chain terminator. Zidovudine inhibits HIV-1 at concentrations of approximately 0.013 g/ml.

ch06.qxd 10/9/03 6:47 PM Page 99

antiviral agents In addition, it inhibits a variety of other retroviruses. Synergy has been demonstrated against HIV-1 when zidovudine is combined with didanosine, zalcitabine, lamiviudine, nevirapine, delavirdine, saquinavir, indinavir, ritonavir, and other compounds. It was the first drug to be licensed for the treatment of HIV infection and still is used in combination with other drugs as initial therapy for some patients. Zidovudine is available in capsule, syrup, and intravenous formulations. Oral bioavailability is approximately 65%. Peak plasma levels are achieved approximately 0.5–1.5 h after treatment. Zidovudine is extensively distributed, with a steady-state volume of distribution of approximately of 1.6 liters/kg. The drug penetrates cerebrospinal fluid, saliva, semen, and breast milk, and it crosses the placenta. The drug is predominately metabolized by the liver through the enzyme uridine diphosphoglucuronosyltransferase to its major inactive metabolite 3-azido- 3-deoxy-5-OB-D-glucopyranuronosylthymidine. The elimination t1/2 is approximately 1 h; however, it is extended in individuals who have altered hepatic function.

99

identified, with the degree of resistance being proportional to the number of mutations. The development of resistant HIV strains correlates with disease progression. The utilization of combination drug therapies delays the onset of resistance. d. Adverse events The predominant adverse effect of zidovudine is myelosuppression, as evidenced by neutropenia and anemia, occurring in 16 and 24% of patients, respectively. Zidovudine has been associated with skeletal and cardiac muscle toxicity, including polymyositis. Nausea, headache, malaise, insomnia, and fatigue are common side effects. 2. Didanosine

b. Clinical applications Zidovudine was the first approved antiretroviral agent, and as a consequence, has been the most widely used antiretroviral drug in clinical practice. In monotherapy studies, zidovudine improves survival and decreases the incidence of opportunistic infections in patients with advanced HIV disease. Importantly, zidovudine decreased the incidence of transmission of HIV infection from pregnant women to their fetuses. However, its usefulness as monotherapy has been outlived. Recently, zidovudine has been incorporated into multidrug regimens, including combinations with didanosine or zalcitabine which demonstrate a delay in disease progression and improved survival compared to zidovudine monotherapy; zidovudine plus didanosine and zidovudine plus lamivudine have also been shown to improve both outcome and important markers of disease, including CD4 counts and plasma HIV RNA levels. Currently, three-drug combinations include the use of zidovudine with other reverse transcriptase inhibitors and nonnucleoside reverse transcriptase inhibitors and protease inhibitors. Triple-drug combinations offer enhanced therapeutic benefits, particularly as noted by survival and restoration of normal immune function.

a. Chemistry, mechanism of action, and antiviral activity Didanosine (2,3-dideoxyinosine; ddl and Videx) is a purine nuceloside with inhibitory activity against both HIV-1 and HIV-2. Didanosine is activated by intracellular phosphorylation. The conversion of 2,3dideoxyinsine-5-monophosphate to its triphosphate derivative is more complicated than that with other nucleoside analogs because it requires additional enzymes, including a 5 nucleotidase and subsequently, adenylosuccinate synthetase and adenylosccinate lyase. The triphosphate metabolite is a competitive inhibitor of HIV reverse transcriptase and is also a chain terminator. The spectrum of activity of didanosine is enhanced by synergism with zidovudine and stavudine as well as the protease inhibitors. Didanosine is available in an oral formulation; however, it is acid labile and has poor solubility. A buffered tablet results in 20–25% bioavailability. A 300-mg oral dose achieves peak plasma concentrations of 0.5–2.6 g/ml with a t1/2 of approximately 1.5 h. Drug is metabolized to hypoxanthine and is cleared primarily by the kidney.

c. Resistance

b. Clinical indications

Zidovudine resistance occurs rapidly after the onset of therapy. Numerous sites of resistance have been

Didanosine is used in combination with other nucleoside analogs and protease inhibitors. In combination

ch06.qxd 10/9/03 6:47 PM Page 100

100

the desk encyclopedia of microbiology

with zidovudine, improvement in both clinical outcome and immunologic markers of disease has been reported (CD4 lymphocyte counts).

c. Resistance As with zidovudine, mutations and reverse transcriptase appear promptly after administration of didanosine therapy, resulting in a 3- to 10-fold decrease in susceptibility to therapy.

b. Clinical indications Zalcitabine is used in combination with other reverse transcriptase and protease inhibitors. As with other nucleoside combinations, zidovudine and zalcitabine do not benefit patients to the same extent as combinations of zidovudine and didanosine. Currently, it is used as part of a two- or three-drug regimen in combination with zidovudine and saquinavir.

c. Resistance d. Adverse effects The most significant adverse effect associated with didanosine therapy is the development of peripheral neuropathy (30%) and pancreatitis (10%). Adverse effects of note include diarrhea (likely attributed to the phosphate buffer), headache, rash, nausea, vomiting, and hepatotoxicty. Myelosuppression is not a component of toxicity associated with didanosine administration. 3. Zalcitabine

Zalcitabine-resistant HIV-1 variance has been documented both in vitro and in vivo.

d. Adverse effects Peripheral neuropathy is the major toxicity associated with zalcitabine administration, occurring in approximately 35% of individuals. Pancreatitis can occur but does so infrequently. Thrombocytopenia and neutropenia are uncommon (5 and 10%, respectively). Other zalcitabine-related side effects include nausea, vomiting, headache, hepatotoxicity, and cardiomyopathy.

4. Stavudine a. Chemistry, mechanism of action, and antiviral activity

a. Chemistry, mechanism of action, and antiviral activity Zalcitabine (2,3-dideoxycytidine; ddC and Hivid) is a pyrimidine analog which is activated by cellular enzymes to its triphosphate derivative. The enzymes responsible for activation of zalcitabine are cell cycle independent, and therefore this offers a theoretical advantage for nondividing cells, specifically dendritic and monocyte/macrophage cells. Zalcitabine inhibits both HIV-1 and HIV-2 at concentrations of approximately 0.03 M. Synergy has been described between zidovudine and zalcitabine as well as with saquinavir. The oral bioavailability following zalcitabine administration is more than 80%. The peak plasma concentrations following an oral dose of 0.03 mg/kg range from 0.1 to 0.2 M, and the t1/2 is short (approximately 20 min). The drug is cleared mainly by the kidney, and therefore, in the presence of renal insufficiency a prolonged plasma t1/2 is documented.

Stavudine (2,3-didehydro, 3-deoxythymidine; d4T and Zerit) is a thymidine analog with significant activity against HIV-1, having inhibitory concentrations which range from 0.01 to 4.1 M. Its mechanism of action is similar to that of zidovudine. It is either additive or synergistic in vitro with other combinations of both nucleoside and nonnucleoside reverse-transcriptase inhibitors. The oral bioavailability of stavudine is more than 85%. Peak plasma concentrations of approximately 1.2 g/ml are reached within 1 h of dosing at 0.67 mg/kg per dose. The drug penetrates CSF and breast milk. The drug is excreted by the kidney unchanged and, in part, by renal tubular secretion.

b. Clinical indications Stavudine has been studied both as monotherapy and in combination with other antiretroviral drugs. It is gaining increasing use as front-line therapy for HIV infection. Stavudine’s clinical benefit is superior to that of zidovudine, particularly as it relates to

ch06.qxd 10/9/03 6:47 PM Page 101

antiviral agents increasing CD4 cell counts, slowing progression to AIDS or mortality.

c. Resistance The development of resistance on serial passage in the laboratory can be achieved. Cross-resistance with didanosine and zalcitabine has been identified by specific mutations for stavudine. The development of resistance in clinical trials has not been identified.

d. Adverse effects The principal adverse effect of stavudine therapy is the development of peripheral neuropathy. The development of this complication is related to both dose and duration of therapy. Neuropathy tends to appear after 3 months of therapy and resolves slowly with medication discontinuation. Other side effects are uncommon.

5. Lamivudine

101

b. Clinical indications Lamivudine is used in combination with other reverse transcriptase inhibitors and protease inhibitors. In combination with zidovudine, enhanced CD4 responses and suppression of HIV RNA levels occur to a greater extent than with zidovudine monotherapy. The combination of zidovudine and lamivudine is without significant adverse event. Because of this degree of tolerability, it is widely used in clinical practice. In addition, lamivudine is licensed for the treatment of chronic hepatitis B. However, resistance appears soon after administration in many patients.

c. Resistance With clinical therapy, resistance to lamivudine monotherapy develops rapidly. In large part, resistance is mediated by amino acid change at codon 184, resulting in a 100- to 1000-fold decrease in susceptibility. The 184 mutation site, which is of importance, also occurs with didanosine and zalcitabine and appears to increase sensitivity to zidovudine, providing a logical basis for its combination with this agent.

d. Adverse effects Lamivudine has an extremely favorable toxicity profile. This may largely be attributed to the low affinity of lamivudine for DNA polymerase. At the highest doses of 20 mg/kg/day, neutropenia is encountered but at a low frequency. In pediatric studies, pancreatitis and peripheral neuropathies have been reported. a. Chemistry, mechanism of action, and antiviral activity Lamivudine is the ( ) enantiomer of a cytidine analog, with sulfur substituted for the 3 carbon atom in the furanose ring [( ) 2,3-dideoxy, 3-thiacytidine; 3TC, Epivir]. It has significant activity in vitro against both HIV-1 and HIV-2 as well as HBV. Lamivudine is phosphorylated to the triphosphate metabolite by cellular kinases. The triphosphate derivat-ive is a competitive inhibitor of the viral reverse transcriptase. Lamivudine’s oral bioavailability in adults is in excess of 80% for doses between 0.25 and 8.0 mg/kg. Peak serum concentrations of 1.5 g/ml are achieved in 1–1.5 h and the plasma t1/2 is approximately 2–4 h. The drug is cleared by the kidney unchanged by both glomerular filtration and tubular excretion.

6. Abacavir 1. Chemistry, mechanism of action, and antiviral activity Abacavir is a carbocyclic synthetic nucleoside analogue. Intracellularly, abacavir is phosphorylated by cellular enzymes to its active metabolite, carbovir triphosphate, which is an analogue of deoxyguanosine-5-TP. Carbovir TP then inhibits the activity of HIV reverse transcriptase both by competing with the natural substrate dGTP and by its incorporation into viral DNA. The lack of a 3-OH group in the incorporated nucleoside analogue prevents the formation of the 5 to 3 phosphodiester linkage essential for DNA chain elongation, producing chain termination.

ch06.qxd 10/9/03 6:47 PM Page 102

102

the desk encyclopedia of microbiology

2. Clinical indications

b. Clinical indications

Abacavir often is used in combination with lamivudine and zidovudine, as well as with either a nonnucleoside reverse transcriptase inhibitor or protease inhibitor. It is licensed for the treatment of HIV infections of humans. The proposed dosage is 300 mg daily.

Nevirapine monotherapy is associated with a nonsustained antiviral effects at a dosage of 200 mg/day. Concomitant with this minimal effect is the rapid emergence of resistant virus, such that by 8 weeks all patients had evidence of viral resistance. Thus, drug can only be administered in combination with other antiretroviral agents. In combination with nucleoside reverse-transcriptase inhibitors, there is evidence of reduction in viral HIV RNA load as well as increasing CD4 counts.

3. Resistance Abacavir resistance is conferred by mutations in the HIV reverse transcriptase gene that resulted in amino acid substitutions at positions K65R, L74V, Y115F, and M184V. M184V and L74V are the most frequently observed mutations among clinical isolates. Multiple reverse transcriptase mutations conferring abacavir resistance exhibit cross-resistance to lamivudine, didanosine, and zalcitabine in vitro. 4. Adverse events Adverse reactions associated with abacavir therapy include nausea, headache, stomach pain, diarrhea, insomnia, rash, fever, and dizziness. Importantly, fatal hypersensitivity reactions have been associated with abacavir use.

B. Non-nucleoside reverse transcriptase inhibitors

c. Adverse effects The most common adverse effects include the development of a nonpruritic rash in as many as 50% of patients who received 400 mg/day. In addition, fever, myalgias, headache, nausea, vomiting, fatigue, and diarrhea have also been associated with administration of drug. d. Resistance Nevirapine resistance has been identified according to its binding site on viral polymerase. Specifically, two sets of amino acid residues (100–110 and 180–190) represent sites at which resistant mutations have occurred. Nevirapine monotherapy is associated with resistance, most frequently appearing at codon 181. Because of the rapid appearance of resistance, nevirapine must be administered with other antiretroviral agents.

1. Nevirapine 2. Delavirdine

a. Chemistry, mechanism of action, and antiviral activity Nevirapine (11-cyclopropyl-5,11-dihydro-4-methyl6H-dipyrido[3,2-b:2, 3-e]; [1,4]diazepin-6-one and Viramune) is a reverse transcriptase inhibitor of HIV1. Nevirapine is rapidly absorbed with a bioavailability of approximately 65%. Peak serum concentration is achieved approximately 4 h after a 400-mg oral dose of 3.4 g/ml. Nevirapine is metabolized by liver microsomes to hydroxymethyl-nevirapine. In vitro, synergy has been demonstrated when administered with nucleoside reverse transcriptase inhibitors.

a. Chemistry, mechanism of action, and antiviral activity Delavirdine (1-[5-methanesulfonamido-1H-indol-2yl-carbonyl]-4-[3-(1-methylethylamino) pyridinyl] piperazine; Rescriptor) is a second-generation bis (heteroaryl) piperazine licensed for the treatment of HIV infection. It is absorbed rapidly when given orally to 60%. Delavirdine is metabolized by the liver with an elimination t1/2 of approximately 1.4 h. It has an inhibitory concentration against

ch06.qxd 10/9/03 6:47 PM Page 103

antiviral agents HIV-1 of approximately 0.25 M. Inhibitory concentrations for human DNA polymerases are significantly higher. b. Clinical indications Reductions in plasma HIV RNA of more than 90% have been documented when delavirdine is administered such that trough levels exceed 50 M. However, there is a rapid return to baseline over 8 weeks as resistance develops. As a consequence, delavirdine must be administered with either zidovudine or didanosine to have a more protracted effect. c. Adverse effects Delavirdine administration is associated with a maculopapular rash. Other side effects are less common. d. Resistance Delavirdine resistance can be generated rapidly both in vitro and in vivo with the codon change identified at 236, resulting in an increase and susceptibility to 60 M. Delavirdine resistance can also occur at codons 181 and 188, as noted for nevirapine administration.

3. Efavirenz

103

b. Clinical indications Efavirenz is employed in combination with other antiretroviral agents indicated in the treatment of HIV-1 infection. Efficacy has been documented in the demonstration of plasma HIV negativity (400 HIV RNA copies/ml) in approximately 80% of patients. Combination therapy has resulted in a 150-fold or greater decrease in HIV-1 RNA levels. Importantly, data have shown efficacy in children for both virologic and immunologic end points. c. Adverse effects The most common adverse events are skin rash (25%), which is associated with blistering, moist desquamation, or ulceration (1%). In addition, delusions and inappropriate behavior have been reported in 1 or 2 patients per 1000. d. Resistance As with other non-nucleoside reverse-transcriptase inhibitors, resistance appears rapidly and is mediated by similar enzymes. 4. Future Prospect Capravirine Capravirine is a non-nucleoside reverse transcriptase inhibitor currently under investigation. Need more It has potent in vitro activity against HIV variants with RT substitutions, including K103N that confer broad cross resistance to the other drugs in this class.

C. Protease inhibitors 1. Saquinavir a. Chemistry, mechanism of action, and antiviral activity Efavirenz Sustiva [(S)-6-chloro-4-(cyclopropylethynyl)1,4-dihydro-4-(trifluoromethyl)-2H-3,1-benzoxazin2-one; Sustiva and DMP266] is a nonnucleoside reverse-transcriptase inhibitor which can be administered once daily. Activity is mediated predominately by noncompetitive inhibition of HIV-1 reverse transcriptase. HIV-2 reverse transcriptase in human cellular DNA polymerases , , , and are not inhibited by efavirenz. The 90–95% inhibitory concentration of efavirenz is approximately 1.7–25 nM. In combination with other anti-HIV agents, particularly zidovudine, didanosine, and indinaver, synergy is demonstrated.

a. Chemistry, mechanism of action, and antiviral activity Saquinavir (cis-N-tert-butyl-decahydro-2[2(R)-hydroxy4-phenyl-3-(S)-([N-(2-quinolycarbonyl)-L-asparginyl] amino butyl)-4aS, 8aS]-isoquinoline-3[S]-carboxyamide methanesulfonate; Invirase) is a hydroxyethylaminederived peptidomimetic HIV protease inhibitor.

ch06.qxd 10/9/03 6:47 PM Page 104

104

the desk encyclopedia of microbiology

Saquinavir inhibits HIV-1 and HIV-2 at concentrations at 10 nM and is synergistic with other nucleoside analogs as well as selected protease inhibitors. Oral bioavailability is approximately 30% with extensive hepatic metabolism. Peak plasma concentrations of 35 mg/l are obtained following a 600-mg dose. b. Clinical indications The clinical efficacy of saquinavir is limited by poor oral bioavailability, but improved formulation (soft-gel capsule) will likely enhance efficacy. Currently, it is used in combination therapy with other nucleoside analogs, particularly zidovudine, lamivudine, zalcitabine, and stavudine. c. Adverse effects Adverse effects are minimal, with no dose-limiting toxicities. Abdominal discomfort, including diarrhea, nausea, and photo sensitization has been reported infrequently. d. Resistance Resistance to saquinavir develops rapidly when it is administered as monotherapy. By 1 year, 45% of patients develop resistance at codon sites 90 and 48, resulting in, an approximately 30-fold decrease in susceptibility.

b. Clinical indications Indinavir has been established as effective therapy for the treatment of HIV infection, particularly in combination with nucleoside analogs. At a dose of 800mg per 8h, 80% of patients experience at least a 100-fold reduction in HIV-RNA levels, and in 50% of patients there is up to a 2 log reduction. In approximately 30% of patients plasma HIV RNA levels are reduced below 500 copies/ml, with an associated increase in CD4 cell counts over baseline. In combination with zidovudine and lamivudine, a 2 log decrease in plasma RNA levels can be achieved for a majority of patients (more than 80%). c. Adverse effects Although indinavir is well tolerated, commonly encountered adverse effects include indirect hyperbilirubinemia (10%) and nephrolithiasis (5%). d. Resistance Indinavir resistance develops rapidly with monotherapy and occurs at multiple sites. The extent of resistance is directly related to the number of codon changes in the HIV protease gene. Codon 82 is a common mutation in indinavir-resistant HIV isolates. 3. Ritonavir

2. Indinavir a. Chemistry, mechanism of action, and antiviral activity

a. Chemistry, mechanism of action, and antiviral activity {N-[2(R)-hydroxy-1(S)-indanyl]-5-[2(S)-(1,1-dimethylethlaminocarbonyl)-4-(pyridin-3-yl) methylpiperazin-1-yl]-4[s]-hydroxy-2[2]-phenylmethyl entanamide; Crixivan} is a peptidomimetic HIV-1 and HIV-2 protease inhibitor. At concentrations of 100 nM, indinavir inhibits 90% of HIV isolates. Indinavir is rapidly absorbed with a bioavailability of 60% and achieves peak plasma concentrations of 12 M after an 800-mg oral dose.

Ritonavir (10-hydroxy-2-methyl-5-[1-methylethyl]1[2-(1-methylethyl)-4-thiazo lyl]-3,6,dioxo-8,11-bis [phenylmethyl]-2,4,7,12-tetra azatridecan-13-oic-acid, 5-thiazolylmethylester, [5S-(5R, 8R, 10R, 11R)]; Norvir) is a symmetric HIV protease inhibitor which has exquisite activity in vitro against HIV-1 laboratory strains (0.02–0.15 M). It is synergistic when administered with nucleoside analogs. Oral bioavailability is approximately 80%, with peak plasma levels of approximately 1.8 M after 400 mg administered every 12 h. The plasma halflife is approximately 3 h. b. Clinical indications Ritonavir is used for treatment of HIV infection in combination with nucleoside analogs. As monotherapy, a 10- to 100-fold decrease in plasma HIV RNA is achieved with a concomitant increase in CD4

ch06.qxd 10/9/03 6:47 PM Page 105

antiviral agents cell counts of approximately 100 cells/mm3. Combination therapy results in a more significant decrease in HIV RNA plasma levels. c. Adverse effects

105

d. Resistance Cross-resistance to other protease inhibitors, particularly saquinavir, indinavir, or ritonavir, is not common. The most frequently demonstrated site of mutation is at codon 30.

Adverse effects include nausea, diarrhea, and headache, but all occur at a low frequency. 5. Amprenavir d. Resistance Resistance to ritonavir resembles that to indinavir. Mutations at codon 82 are the most common.

4. Nelfinavir

a. Chemistry, mechanism of action, and antiviral activity

a. Chemistry, mechanism of action, and antiviral activity Nelfinavir [3S-(3R, 4aR, 8aR, 22S, 3S)]-2- [2hydroxy-3-phenylthiomethyl-4-aza-5-ox-o-5-(2 methyl-3-hydroxyphenyl)pentyl]-decahydroisoquinoline-3-N-(tert-butyl-carboxamide methanesulfonic acid salt) is another peptidomimetic HIV protease inhibitor. Inhibitory concentrations of HIV-1 are in the range of 20–50 nM. Nalfinavir is orally bioavailable at approximately 40%, achieving peak plasma concentrations of 2 or 3 mg following a 800-mg dose every 24 h. The drug is metabolized by hepatic microsomes.

Amprenavir is a hydroxyethylamine sulfonamide peptidomimetric with a structure identified as (3S)tetrahydro- 3-furyl N-(1S,2R)-3-(4-amino-N-isobutylbenzenesulfonamido)-1-benzyl-2-hydroxypropyl carbamate. It is active at a concentration of 10–20 nM. The oral bioavailability is 70%, and peak plasma concentrations of 6.2–10 g/ml are achieved after dosages of 600–1200 mg. The plasma half-life is 7–10 h. Cerebrospinal fluid concentrations are significant. Amprenavir acts by binding to the active site of HIV-1 protease, preventing the processing of viral gag and gag-pol polyprotein precursors and resulting in the formation of immature non-infectious viral particles. In vitro, amprenavir has synergistic anti-HIV-1 activity in combination with abacavir, zidovudine, didanosine, or saquinavir, and additive anti-HIV-1 activity in combination with indinavir, nelfinavir, and ritonavir.

b. Clinical indications Nalfinavir is utilized in combination with nucleoside analogs. Monotherapy will achieve significant decreases in HIV RNA plasma levels up to 100-fold. Currently, the drug is used in combination with nuceloside analogs, particularly zidovudine, lamivudine, or stavudine, which results in 100- to 1000-fold reductions of HIV plasma RNA levels.

2. Clinical indications Amprenavir is licensed for the treatment of HIV infections. The recommended dosage for adults is 1200 mg twice daily.

3. Adverse effects c. Adverse effects Nelfinavir is well tolerated, with mild gastrointestinal complication reported.

The most serious adverse effect is a rash. Other side effects include nausea, vomiting, diarrhea, abdominal pain, and perioral paresthesias.

ch06.qxd 10/9/03 6:47 PM Page 106

106

the desk encyclopedia of microbiology

4. Resistance Resistance to amprenavir is conferred by amino acid substitutions primarily at positions M46I/L, I47V, I50V, I54L/V, and I84V, as well as mutations in the viral protease p1/p6 cleavage site. Cross-resistance between amprenavir and the other protease inhibitors is possible. 6. Lopinavir/Ritonavir Lopinavir/ritonavir combination (marketed as Kaletra) interferes with processing of viral polyprotein precursors, resulting in non-infectious progeny virions. The addition of ritonavir enhances the concentrations of lopinavir that which can be achieved following oral administration. It is given in combination with nucleoside and/or non-nucleoside reverse transcriptase inhibitors. Side effects of lopinavir/ ritonavir include diarrhea, nausea, abdominal pain, and headache. As lopinavir/ritonavir is a new addition to the protease inhibitors, a complete understanding of resistance profiles will await its widespread utilization.

D. Nucleotide Analogues Viread; Tenofovir (tenofovir disoproxil fumarate) Tenofovir or disproxil fumarate salt is an acyclic nucleoside phosphonate diester analog of adenosine monophosphate with an in vitro the 50% inhibitor concentration for HIV is 0.04–8.5 mol. After diester hydrolysis, tenofovir is phosphorylated to the DP that then inhibits HIV reverse transcriptase by competing with the natural substrate deoxyadenosine 5-TP and, after incorporation into DNA, by DNA chain termination. Tenofovir DP is a weak inhibitor of mammalian DNA polymerases alpha, beta and mitochondrial DNA polymerase gamma. Additive or synergic antiHIV activity with nucleoside analog, non-nucleoside analog and protease inhibitors has been demonstrated in vitro. Side effects include lactic acidosis, hepatomegaly with steatosis, and diarrhea. Resistance is uncommon and occurs at codon 65.

E. HIV Fusion Inhibitors Fuseon; Enfuvirtide The recent licensure of a fusion inhibitor introduces a new class of antiviral compounds for the treatment of HTV. T-20 is an inhibitor of fusion of HIV-1 with CD4 cells that consists of a 36 amino acid synthetic peptide with the N-terminus acetylated and the

C-terminus is a carboxamide. administered subcutaneously.

Medication

is

VI. SUMMARY It is anticipated that many new compounds will be licensed for the treatment of viral disease because many are currently under development.

ACKNOWLEDGMENTS Work performed and reported by the author was supported by Contracts NO1-Al-15113, NO1-Al-62554, NO1-Al-12667, and NO1-A1–65306 from the Antiviral Research Branch of the National Institute of Allergy and Infectious Diseases, a grant from the Division of Research Resources (RR-032) from the National Institutes of Health, and a grant from the state of Alabama.

BIBLIOGRAPHY Abu-ata, O., Slim, J., Perez, G., and Smith, S. M. (2000). HIV therapeutics: past, present, and future. Adv. Pharmacol. 49, 1–40. Balfour, H. H., Jr. (1999). Antivirals (non-AIDS). N. Engl. J. Med. 340, 1255–1268. Beutner, K. R., Friedman, D. J., Forszpaniak, C., et al. (1995). Valaciclovir compared with acyclovir for improved therapy for herpes zoster in immunocompetent adults. Antimicrob. Agents Chemother. 39, 1547–1553. Crumpacker, C. S. (1996). Ganciclovir. N. Engl. J. Med. 335, 721–728. DeClercq E. (2002). New anti-HIV agents and targets. Med. Res. Rev. 22, 531–565. Douglas, J. M., Critchlow, C., Benedetti, J., et al. (1984). Doubleblind study of oral acyclovir for suppression of recurrences of genital herpes simplex virus infection. N. Engl. J. Med. 310, 1551–1556. Dunkle, L. M., Arvin, A. M., Whitley, R. J., et al. (1991). A controlled trial of acyclovir for chicken pox in normal children. N. Engl. J. Med. 325, 1539–1555. Emery, S., and Cooper, D. A. (2003). Antiviral agents. In “Antibiotic and Chemotherapy”, R. G. Finch, D. Greenwood, S. R. Norrby, and R. J. Whtley (Eds.), pp. 473–493, Churchill Livingstone, Philadelphia. Galasso, G., Whitley, R. J., and Merigan, T. C. (Eds.) (1997). “Antiviral Agents and Viral Diseases of Man.” LippincottRaven, New York. Inouye, R. T., Panther, L. A., Hay, M. H., and Hammer, S. M. (2002). Antiviral Agents. In “Clinical Virology”, D. D. Richman, R. J. Whitley, and F. G. Hayden (Eds.), 2nd edn., pp. 171–242, ASM Press, Washington, DC. Lalezard, J. P., Henry, K., O’Hearn, M., et al. (2003). N. Engl. J. Med. 348, 2175–2185.

ch06.qxd 10/9/03 6:47 PM Page 107

107

antiviral agents Richman, R., Whitley, F., and Hayden. (Eds.) (2002). “Clinical Virology.” ASM Press, Washinton. Tyring, S., Barbarash, R. A., Nahlik, J. E., et al. (1995). Famciclovir for the treatment of acute herpes zoster. Effects on acute disease and postherpetic neuralgia: A randomized, double-blind, placebo-controlled trial. Ann. Intern. Med. 123, 89–96. Whitley, R. J. (in press). Antiviral therapy. In: “Infections Diseases”, S. L. Gorbach, J. G. Bartlett, and N. R. Blacklow (Eds.), 3rd edn., pp. 330–350, Saunders, Philadelphia. Whitley, R. J. (2003). Other antiviral agents. In: “Antibiotic and Chemotherapy”, R. G. Finch, D. Greenwood, S. R. Norrby, and R. J. Whtley (Eds.), pp. 495–509, Churchill Livingstone, Philadelphia. Whitley, R. J., and Gnann, J. (1992). Acyclovir: A decade later. N. Engl. J. Med. 327, 782–789.

Whitley, R. J., Alford, C. A., Jr., Hirsch, M. S., et al. (1986). Vidarabine versus acyclovir therapy in herpes simplex encephalitis. N. Engl. J. Med. 314, 144–149.

WEBSITES Center for Disease Control (USA) website on antiviral drugs for influenza http://www.cdc.gov/ncidod/diseases/flu/fluviral.htm Antiviral Agents FactFile by International Medical Press (with search capabilities) http://www.mediscover.net/antiviral.cfm

ch07.qxd 10/13/03 3:15 PM Page 108

7 Archaea Paul Blum and Vidula Dixit George Beadle Center for Genetics, University of Nebraska-Lincoln

GLOSSARY

hyperthermophiles From the Greek hyper- (over), therme- (heat), and philos (loving). Includes organisms that grow optimally at temperatures warmer than 80C. mesophiles From the Greek mesos- (middle) and philos (loving). Includes organisms that grow optimally at temperatures between 20 and 50C. methanogens Strictly anaerobic Archaea that produce (Greek gen, to produce) methane. psychrophiles From the Greek psychros- (cold) and philos (loving). Includes organisms that grow optimally at temperatures between 0 and 20C. thermophiles From the Greek therme- (heat) and philos (loving). Includes organisms that grow optimally at temperatures between 50 and 80C.

Archaea One of three domains of life. From the Greek archaios (ancient, primitive). Prokaryotic cells; membrane lipids predominantly isoprenoid glycerol diethers or diglycerols tetraethers. Formerly called archaebacteria. Bacteria One of three domains of life. From the Greek bacterion (staff, rod). Prokaryotic cells; membrane lipids predominantly diacyl glycerol diesters. Formerly called eubacteria. Crenarchaeota One of two kingdoms of organisms of the domain Archaea. From the Greek crene- (spring, fountain) for the resemblance of these organisms to the ancestor of the Archaea, and archaios (ancient). Include sulfur-metabolizing, extreme thermophiles. Eukarya One of three domains of life. From the Greek eu- (good, true) and karion, (nut; refers to the nucleus). Eukaryotic cells; cell membrane lipids predominantly glycerol fatty acyl diesters. Euryarchaeota One of two kingdoms within the domain Archaea. From the Greek eurys- (broad, wide), for the relatively broad patterns of metabolism of these organisms, and archaios (ancient). Include halophiles, methanogens, and some anaerobic, sulfur-metabolizing, extreme thermophiles. halophiles From the Greek halos- (salt) and philos (loving). Includes organisms that grow optimally at high salt concentrations.

The Desk Encyclopedia of Microbiology ISBN: 0-12-621361-5

I. INTRODUCTION In an effort to accommodate molecular signatures evident in ribosomal small subunit RNAs, Woese and Fox (1977) proposed that prokaryotes are not a monophyletic group (single root). Instead, they argued for two distinct evolutionary lineages of organisms represented by the Bacteria and those now called Archaea (Fig. 7.1). Archaea, Bacteria and Eukarya are placed in separate taxonomic groups called Domains. The distinction between Archaea and Bacteria has since received impressive support from many sources.

108

Copyright © 2003 Elsevier Ltd All rights of reproduction in any form reserved

ch07.qxd 10/9/03 6:50 PM Page 109

109

archaea

Cyanobacteria

Meth

ter

cteria

ac

s

Archaea

gale

lob

to mo

Ha

r The

ta chaeo Korar us cocc s u rmo The ococc s Pyr ccu co no tha Me ax er of

l Ha

Euk

Flavoba

ta

aeo

ch arya

ium

anob

Bacteria es tiv

i os

m ra

p

ha

Alp

acte

rium

ria

cte

ba teo

G

pro

Gamma proteobacteria

ap

oba

cter

ia

er

P m yro op di ro cti te um us

rote

Th

Cre

Bet

phaera s obu lfol u S

Metallos

cha nar

eot

a

Acidian us

Microsporidia Animals Yeast 0.1

Plants

Fungi

Eukarya FIGURE 7.1 Universal (distance) phylogenetic tree based on ribosomal small subunit RNA sequences.

Perhaps the most compelling data comes from whole genome sequencing studies which reveal extensive gene and protein sequence conservation between members of the Archaea but not Bacteria. Comparative genomics also suggest that Archaea are a distinct group and share a common origin with Eukarya. This is further supported by the finding that Archaea employ a broad range of eukaryotic-like genes for conducting subcellular processes including the synthesis and processing of DNA, RNA, and protein (Blum, 2001). In contrast, Archaea use bacterial-like mechanisms for much of central metabolism including key biosynthetic (anabolic) and degradative (catabolic) pathways. Archaea derived from extreme environments are studied intensively leading to the incorrect interpretation that all Archaea are extremophiles. Since extreme environments comprise only a small portion of inhabitable earth, Archaea are often thought to comprise only a small proportion of total prokaryotes. In contrast to this notion, studies on marine prokaryotic abundance indicate that global oceans harbor approximately equal numbers of archaeal and bacterial cells (Karner et al., 2001). The major fraction of these Archaea are assigned to the subdivision called Crenarchaeota and these represent one of the ocean’s single most abundant cell types. Archaea are also evident in soil and

fresh water, and are associated with plant roots. It may be that Archaea are as abundant a life form as Bacteria. Archaea are readily distinguished from Bacteria by several universal and unique chemical features. In contrast, the ecology and physiology of Archaea often overlaps that of Bacteria with the notable absence of differentiated cell forms or developmental cycles. Investigations of archaeal genomics and their molecular biology, however, reveals the true magnitude of the similarities between Archaea and Eukarya and differences from Bacteria. Perhaps this latter finding explains the utter lack of pathogenic Archaea despite the occurrence of host-adpated species such as the methanogens.

II. CELL STRUCTURE OF THE ARCHAEA Archaea, like Bacteria, are prokaryotes (Table 7.1). They lack membrane-bound organelles such as a nucleus, or mitochondria, and are devoid of a cytoskeleton. Their chromosomal DNA is typically a single circular molecule and their ribosomes are of the 70S type. Their cell membranes and surface layers

ch07.qxd 10/9/03 6:50 PM Page 110

110

the desk encyclopedia of microbiology

TABLE 7.1 Major features distinguishing archaea and bacteria Characteristic Cell envelope (a) Wall Membrane (a) Chirality of glycerol (b) Hydrocarbon glycerol linkage (c) Side-chains (d) Side-chain branching Bacterial antibiotic sensitivity (a) Cell wall inhibitors (b) Protein synthesis inhibitors (c) Transcription inhibitors

Archaeaa

Bacteria

Mostly S-layer

Peptidoglycan

L-glycerol Ether-linkage

D-glycerol Ester-linkages

Isoprene Fatty acids Highly branched Linear No Noa No

Yes Yes Yes

a Some methanogenic Archaea are sensitive to puromycin and chloramphenicol.

(envelope) are structurally and chemically distinct from those of Bacteria. Archaeal cell envelope. Bacterial envelopes usually comprise a peptidoglycan wall with a single lipid bilayer internal to the wall (gram-positive Bacteria) or two lipid bilayers one internal and the other external to the wall (gram-negative Bacteria). In contrast, Archaea have neither peptidoglycan nor a wall. Instead the most common outer layer of their envelope consists of a paracrystalline S-layer, composed of noncovalently linked hexagonally or tetragonally arranged protein or glycoprotein subunits. Methanogens show particular diversity in envelope composition. Methanobacteria have a structure referred to as pseudomurein which resembles peptidoglycan, Methanosarcina have a structure referred to as methanochondroitin and Methanococcus and Methanoplanus have protein or glycoprotein layers. Pseudomurein is distinguished from peptidoglycan by its use of L-isomeric amino sugars with -1,3 linkages rather than D-isomeric amino sugars using -1,4 linkages. Haloarchaea have a protein layer containing a great excess of acidic amino acids whose negative charges counterbalance the high concentration of positively charged sodium ions in their salty environment. The envelope of Natronobacteria contains a novel layer composed of a glutamine polymer with N-acetylglucosamine, glucose and other sugars linked via the amide group of glutamine. Archaeal cell membranes. Archaeal cell membranes are chemically unique from those of Bacteria or Eukarya. Differences include: (a) chirality of glycerol; (b) hydrocarbon–glycerol linkage; (c) isoprenoid chains; and (d) side-chain branching. The basic unit from which cell membranes are built is the phospholipid. The glycerol in archaeal phospholipids is a

stereoisomer (L-glycerol) of that found in bacterial and eukaryal membranes (D-glycerol). The side chains in the phospholipids of Bacteria and Eukarya are fatty acids usually 16–18 carbon atoms in length and are coupled to glycerol via an ester linkage. In Archaea, side chains are coupled via an ether linkage. Archaea have side chains built from isoprene. Isoprene is the simplest member of a class of chemicals called terpenes. The ester linked fatty acids of Bacteria are linear, whereas the ether linked hydrocarbons of Archaea are highly branched. Archaeal glycerol diether lipids form a true bilayer membrane, whereas archaeal glycerol tetraether lipids form lipid monolayers. Lipid monolayer membranes occur in certain methanogens, are widespread among hyperthermophilic Archaea and are thought to confer additional thermal stability. Archaeol (diphytanylglycerol diether) is the predominant membrane core lipid in most methanogens and all extreme halophiles. In contrast, cell membrane of hyperthermophilic archaea and a few methanogens contain caldarchaeol, a dibiphytanyldiglycerol tetraether. The production of ether-linked lipids is so distinctive in Archaea that it is used as a biomarker for detecting fossilized Archaea in micropaleontological studies of rocks, sediment cores and other ancient materials. The chemical difference between archaeal and bacterial lipids provides additional support for the evolutionary distance between the Archaea and Bacteria.

III. ECOLOGY, PHYSIOLOGY AND SYSTEMATICS OF THE ARCHAEA The cultivated Archaea are distributed into two kingdoms, the Euryarchaeota and the Crenarchaeota (Table 7.2). A third kingdom called Korarchaeota consists of uncultivated members. Cultivated Archaea are divided into 12 orders, 20 families, and 69 genera (Boone et al., 2001). Cultivated Euryarchaeota include all of the known methanogens and extreme halophiles as well as some extreme thermophiles. Cultivated members of the Crenarchaeota include other hyperthermophiles and most thermoacidophiles. In addition, many other archaeal taxa have been detected in a variety of environments using molecular phylogenetic methods. While as yet uncultivated, these Archaea also are distributed across both the euryarchaeotal and the crenarchaeotal kingdoms. Cultivated Archaea include terrestrial and aquatic members, and occur in diverse locations from anaerobic sediments to hypersaline pools and geothermally heated environments. Some also occur as symbionts in animal digestive tracts. Archaea

ch07.qxd 10/9/03 6:50 PM Page 111

111

archaea TABLE 7.2 Systematics of the Archaea Orde

Family

Habitat

Features

Kingdom: Euryarchaeota Archaeoglobales

Archaeaglobaceae

Geothermally heated sites

Strict anaerobes, facultative chemolithoautotrophs, hyperthermophiles, neutrophiles

Halobacteriales

Halobacteriaceae

Ubiquitous in areas of high salt concentration like salt lakes, salterns

Aerobes or facultative anaerobes, require 3.5–4.5 M NaCl, chemoorganotrophs, usually mesophiles

Methanobacteriales

Methanobacteriaceae

Strict anaerobes, chemolithoautotrophs, mesophiles to thermophiles

Methanothermaceae

Aquatic sediment, sewage digestor, GI tract of animals Hot solfataric fields

Methanococcales

Methanococcaceae

Marine environments

Methanocaldococcaceae

Deep sea hydrothermal vents

Methanocorpusculaceae

Lake sediments, digestors Marine sediments, sewage digestors Sewage sludge, waste digestors

Strict anaerobes, chemoorganotrophs, mesophiles

Methanomicrobiales

Methanomicrobiaceae Methanospirillaceae

Strict anaerobes, chemolithotrophs, hyperthermophiles Strict anaerobes, chemolithoautotrophs, mesophiles to thermophiles, selenium required Strict anaerobes, chemolithoautotrophs, hyperthermophiles

Strict anaerobes, acetate required, chemolithoautotrophs, mesophiles to thermophiles Strict anaerobes, fix N2, autotrophs or chemoorganotrophs, mesophiles

Methanopyrales

Methanopyraceae

Hot vents

Strictly anaerobic, chemolithoautotrophs, hyperthermophiles

Methanosarcinales

Methanosaetaceae

Sewage sludge and sediments Aquatic sediment, sewage digestors, GI tract of animals

Strictly anaerobic, chemolithotrophs, mesophiles to thermophiles Strictly anaerobic, N2 may be fixed, chemoautotophs, mesophiles to thermophiles

Methanosarcinaceae

Thermococcales

Thermococcaceae

Marine and terrestrial thermal environments

Strict anaerobes, heterotrophs, hyperthermophiles

Thermoplasmatales

Thermoplasmataceae

Self heating coal refuse piles, acidic solfatara fields Hot geothermal solfotara soils and springs

Wall-less, facultative aerobes, heterotrophs, thermoacidophiles

Marine environments, hot solfataric areas Hot sea floors, sediments, black smokers Hot acidic solfataric springs Solfataric hot springs Acidic hot springs

Mostly anaerobic, chemolithotrophs or heteroptrophs, hyperthermophiles Anaerobic, facultative chemolithoautotrophs, hyperthermophiles Aerobic or anaerobic, chemolithoautotrophs, S0 metabolizers, extreme thermoacidophiles Strict anaerobes, organotrophs, thermoacidophiles Anaerobes to facultative anaerobes, chemolithoautotrophs or organotrophs, hyperthermophiles

Picrophilaceae Kingdom: Crenarchaeota Desulfurococcales

Desulfurococcaceae Pyrodictiaceae

Sulfolobales

Sulfolobaceae

Thermoproteales

Thermofilaceae Thermoproteaceae

Obligate aerobes, heterotrophs, hyperacidophiles

Kingdom: Korarchaeota Uncultivated Archaea delineated on the basis of 16 rRNA sequences Key: M, Molar; NaCl, sodium chloride; GI, gastrointestinal; N2, nitrogen; S0, sulfur.

include aerobes, anaerobes and facultative anaerobes, chemolithotrophs, organotrophs and facultative organotrophs. Archaea can be mesophilic or thermophilic with some species growing at temperatures up to 110 C. Psychrophilic members have also been detected but not cultured. The Archaea often are

divided into three key biotypes; methanoarchaea, haloarchaea and hyperthermophilic Archaea (Madigan et al., 2000; also see http://www. ncbi.nlm. nih.gov:80/ entrez/query. fegi? dbTaxonomy). The methanogenic Archaea. The methanoarchaea belong to the Euryarchaeota and constitute

ch07.qxd 10/9/03 6:50 PM Page 112

112

the desk encyclopedia of microbiology

a phylogenetically unique biotype. Methane (CH4) production is an integral part of their energy metabolism and this form of metabolism is still unique to the Archaea. Such organisms are called methanogens and the process of methane formation is called methanogenesis, which is the terminal step in biodegradation of organic matter in many anaerobic environments. Habitats of methanogens range from anoxic sediments such as swamps, and wastewater treatment facilities to ruminant digestive tracts. They can also be found as endosymbionts of various anaerobic protozoa. Most known methanogens are mesophilic although “extremophilic” species growing optimally at very high or very low temperatures or at very high salt concentrations have also been isolated. These are strict anaerobes that are able to form methane as the principal metabolic end product using various oxidized forms of one- and two-carbon compounds as terminal electron acceptors. Key genera of this group include; Methanobacterium, Methanococcus, and Methanosarcina. The halophilic Archaea. This diverse group of Euryarchaeota inhabit highly saline environments such as solar evaporation ponds, natural salt lakes and surfaces of salted foods. Moderately halophilic Archaea require 2–4 M sodium chloride (12–23%) for optimal growth. Virtually all extremely halophilic Archaea grow at 5.5 M sodium chloride (32%) which is the limit of saturation of sodium chloride. Haloarchaea can be aerobic or facultatively anaerobic and are chemoorganotrophic. They are generally mesophilic but can be moderately thermophilic (growth up to 55C). Some species contain a protein called bacteriorhodopsin and the carotenoid pigments, bacterioruberins, which are used for the light mediated synthesis of ATP. Presence of these proteins results in a distinct pigmentation leading to their redpurple coloration. Halobacterium employs potassium as a compatible solute to withstand high external sodium concentrations to ensure a positive water balance. Extreme halophilicity was thought to occur only in Archaea; recently, however, molecular phylogenetic evidence indicates Bacteria may also have this property. Key genera of the Haloarchaea include; Halobacterium, Haloferax, and Natronobacterium. The thermophilic Archaea. A number of Archaea thrive in thermal environments. Those with growth optima at or above 80oC are called hyperthermophiles. Thermophilic Archaea inhabit terrestrial and marine regions with geothermal or hydrothermal intrusion. A growth requirement for exterme acid (acidophily) can accompany the thermophilic lifestyle. The sulfur- and sulfate-reducing thermophiles are split between members of the Euryarcheota and the

Crenarchaeota. These are strict anaerobes using elemental sulfur as a terminal electron acceptor, which is reduced to hydrogen sulfide. These organisms are extremely thermophilic with growth up to 100C. Members of the Thermococci and Pyrococci are Euryarchaeota. Methanopyrus, another Euryarchaeote, is a hyperthermophilic methanogen. Only Archaeoglobus is a true sulfate reducer, capable of reducing sulfate to hydrogen sulfide. Sulfur reducing Crenarchaeota include Thermoproteus, Pyrodictium, and Pyrolobus. Pyrolobus fumarii holds the current record for the most thermophilic of all known organisms, its growth temperature maximum is 113C. Crenarchaeotal sulfur oxidizers include the Sulfolobales, which are found in acidic sulfur containing geothermal pools such as those found in Yellowstone National Park, USA (Fig. 7.2). These are obligate aerobes that use oxygen as a terminal electron acceptor and grow in the temperature range 70–90C and at pH values of between 2 and 5. Acidophily is also found among members of two euryarchaeotal genera, Thermoplasma and Picrophilus, which are among the most acidophilic of all known prokaryotes. Most species of Thermoplasma have been obtained from self-heating coal refuse piles. Thermoplasma resemble the mycoplasma in having an envelope comprising of only a cytoplasmic membrane. Picrophilus is capable of growth even below pH 0. These organisms are both chemoorganotrophic. Again, members of the Bacteria share at least the thermophilic and hyperthermophilic biotypes of Archaea while extreme acidophily remains unique to the Archaea. Key genera of the thermophilic Archaea include; Thermococcus, Pyrococcus, Pyrobaculum, and Sulfolobus. Archaea as non-extremophiles. The known phenotypes of cultivated Archaea are still largely represented by extreme halophiles, sulfur metabolizing thermophiles, and methanogens. This picture has altered as new methods have been applied to the study of uncultivated microbes. The presence of new types of uncultivated Archaea was first suggested during surveys of marine plankton. Surveys of polymerase chain reaction (PCR) amplified small subunit rRNA genes revealed archaeal rRNA sequences in deep ocean water samples. The discovery of high numbers of Archaea in a wide number of oceans and the association of a novel crenarchaeotal isolate with a marine sponge living at 10C, provide further evidence of new archaeal biotypes native to cold seawater. Since their initial detection, evidence for widespread distribution of new uncultivated Archaea has been extended to include forest and agricultural soils, deep subsurface paleosols, freshwater lakes and various sediments.

ch07.qxd 10/9/03 6:50 PM Page 113

113

archaea

(A)

(B)

FIGURE 7.2 Electron micrographs of Sulfolobus solfataricus. (A) transmission electron micrograph; (B) scanning electron micrograph. Both images show the irregular cell shape of the organism.

TABLE 7.3 Ecology and physiology of the model archaeal species Genus

Biotype

Habitat

Energy metabolism

Energy source

Methanococcus maripaludis Methanosarcina acetivorans Haloferax volcanii

Methanogen

Salt marsh sediment

H2 CO2, pyruvate CO2, formate

Methanogen

Marine sediment

Halophile

Dead Sea

Halobacterium salinarium Sulfolobus solfataricus

Halophile Hyperhermophile

Hypersaline lakes and salted foods Sulfur-rich hot springs

Sulfolobus acidocaldarius

Hyperhermophile

Sulfur-rich hot springs

Obligate anaerobes chemolithotrophs/ Obligate anaerobes chemolithotrophs/ Usually obligate aerobe chemoorganotroph, Usually obligate aerobe chemoorganotroph, Aerobic chemolithotrophs/ chemoorganotrophs Aerobic chemolithotrophs/ chemoorganotrophs

H2 CO2, methanol, methylamines, acetate Amino acids Amino acids, organic acids S0, H2S, sugars, amino acids S0, H2S, sugars, amino acids

Key: H2, hydrogen; CO2, carbon dioxide; S0, sulfur; H2S, hydrogen sulfide.

Archaeal model systems. As is the case with Bacteria, particular archaeal taxa are employed as model experimental systems to address mechanistic questions about this group of prokaryotes. The key feature distinguishing these species is their genetic systems. Among the Haloarchaea Haloferax volcanii and Halobacterium salinarum (including NRC-1) are employed. Methanococcus maripaludis and Methanosarcina acetivorans, are popular among the methanogenic Archaea, while among the thermophilic Archaea Sulfolobus solfataricus and Sulfolobus acidocaldarius are best studied (Table 7.3).

IV. MOLECULAR BIOLOGY OF THE ARCHAEA The distinguishing feature of Archaea is their use of eukaryotic-like genes rather than bacterial-like genes for the synthesis, repair, and turnover of DNA, RNA, and protein (Table 7.4). Key aspects of this relationship have been recently described in greater detail (Blum, 2001). This striking example of gene conservation supports the idea of a shared or common evolutionary origin for Archaea and Eukarya. An understanding of these subcellular processes is therefore a prerequisite for appreciating their place in

ch07.qxd 10/9/03 6:50 PM Page 114

114

the desk encyclopedia of microbiology TABLE 7.4 Comparative molecular biology of archaea

Characteristic

Archaea

Eukarya

Bacteria

Chromosomal DNA Introns in genes Operons DNA recombination and repair

Covalently closed circle Rare Present Eukaryal homologs of Rad proteins and others Euryarchaeotes: Histones Crenarchaeotes: Nonhistone proteins One (10–12 subunits, all eukaryal homologs) TATA box

Linear DNA in nucleus Present Absent Rad proteins

Covalently closed circle a Rare Present RecA protein

Histones

Nonhistone proteins

Three (12–14 subunits)

One (4 subunits)

TATA box

Eukaryal homologs of TBP, TFIIB, TFIIS No

TBP, TFIIB, TFIIS and others Yes

10 and 35 hexamers (Pribnow box) Sigma factor

Eukaryal homologs of eIF2, , and , eIF4A, eIF5 Methionine 70 S Eukaryal homologs of Prefoldin, NAC, TCP1 Present 7.5 S SRP RNA complex

eIF2, , and , eIF4A, eIF5 Methionine 80 S Prefoldin, TCP1, NAC

Chromosome packaging

RNA polymerase Promoter structure Transcription factors mRNA Caps and Long Poly-A tails Translation initiation Initiator tRNA Ribosomes Protein folding Proteosome Protein secretion

Present 7.5 S SRP RNA complex

No IF-1, IF-2, and IF-3 Formylmethionine 70 S Hsp70 system (DnaK), GroELS, TF Rare 4.5 S SRP RNA and Sec system

a Few exceptions. Key: TBP, TATA box binding protein; TFIIB, transcription factor B; TFIIS, transcription factor S, eIF, eukaryotic Initiation factors; IF, initiation factors; NAC, nascent polypeptide-associated complex; TF, trigger factor; SRP, signal recognition particle.

biology and evolution and their relationship to bacterial prokaryotes. DNA replication and packing. Two features distinguish archaeal DNA replication, initiation of DNA synthesis, and packing and condensation of DNA into eukaryallike nucelosomes. Though the functions necessary for initiation of DNA replication are common between members of all domains, the relevant archaeal proteins bear strong sequence conservation to those found in Eukarya and not Bacteria. These include; origin recognition complex protein 1 (ORC1), single strand binding protein (RPA), primase, minichromosome maintenance protein (MCM), clamp loader (PCNA), ATP-dependent ligase, and primer removal protein (FEN1). In Eukarya, ORC and MCM proteins are used to coordinate DNA replication with cell cycle, perhaps this is true in Archaea. Archaeal DNA replication like that in Bacteria initiates at a single origin and proceeds in a bidirectional manner. In most Euryarchaeota, DNA is packaged into nucleosomes resembling eukaryotic tetrasomes called half nucleosomes (for information on archaeal histones, see: http://www.biosci.ohiostate.edu/~microbio/ Archaealhistones/). These structures are evident in electron micrographs and consist of DNA complexed with archaeal histone proteins. These structures have the same histone-DNA stoicheometry,

DNA topology, DNA recognition, and nucleosome positioning features as those of Eukarya. In Eukarya nucleosomes provide an important means of regulating gene expression, their presence in many Archaea implicates the existence of similar processes. In contrast to the Euryarchaeota, DNA packaging in members of the Crenarchaeota mirrors that of Bacteria. Small positively charged and highly abundant DNA binding proteins are present with no evidence of ordered packing structure. DNA recombination and repair. Processing of the DNA ends at a double strand break (DSB) in Bacteria employs the RecBCD enzyme. In contrast, it appears RecBCD is absent in Archaea, which instead employ homologs of Eukaryal proteins Mre11/Rad50 and Spo11. In Bacteria, homologous recombination involving DNA strand pairing and exchange is catalyzed by RecA. In Archaea RecA is absent and this critical step is mediated instead by a eukaryotic Rad51 homolog termed RadA. Subsequent resolution of holiday junctions to form recombinant DNA molecules appears to be mediated by archaeal-specific enzymes. Recombinational repair in Archaea is generally more similar to eukaryal repair largely based on the presence of RAD52-like proteins. Other types of repair in Archaea such as alkylation repair, and nucleotide excision repair, employ proteins

ch07.qxd 10/9/03 6:50 PM Page 115

archaea with homology to both Bacteria and Eukarya. Mismatch and error-prone repair are less studied, however, and the lack of archaeal MutS homologs suggests Archaea employ mechanisms unlike those of Bacteria for overcoming this type of DNA damage. RNA synthesis, modification, and degradation. The general transcription apparatus of Archaea exhibits extensive overlap in both function and gene sequence with that of Eukarya. Transcription in Bacteria employs a fundamentally different system. Early studies on this topic supported the separation of Archaea from Bacteria. In Bacteria, the association of RNA polymerase (RNAP) with sigma factor confers on RNAP the ability to bind specifically to promoters. In Eukarya and Archaea, RNAP never has the ability to bind to promoters, but instead is recruited to the DNA by proteins that pre-associate at such sequences. The overlap between the archaeal and eukaryotic transcription systems include: homologous promoter structure, and homologs of TATA binding protein (TBP), TFIIB, TFIIS, and the complex RNAP enzyme itself consisting of more than 10 different subunits. Despite such intriguing evolutionary overlaps, the identity of the accessory regulatory components of Archaea, which must control rates of transcription initiation in response to metabolic and environmental stimuli is poorly understood. rRNA methylation in Archaea occurs in an analogous fashion to Eukarya and not like Bacteria (see http://rna.wustl.edu/snoRNAdb/). Ribosomal RNA of Eukarya undergoes extensive posttranscriptional modification including the addition of between 50 and 100 2-O-ribose methylations depending on the species. Such modifications are thought important in rRNA folding, stability, ribosomal protein binding and rRNA activity within the ribosome. Methylation is mediated by a ribonucleoprotein complex consisting of the protein fibrallarin and NOP56/58 and, small antisense RNAs called snoRNAs. Each methylated position is targeted by a distinct snoRNA. The rRNA of Bacteria such as E. coli, in contrast, contain only four ribose methylations with no evidence of snoRNAs. It appears that all Archaea examined to date encode fibrallarin and NOP56/58 homologs. Extensive arrays of snoRNAs have also been identified in thermophilic Archaea. A hallmark distinguishing Bacteria from Eukarya is the extremely short lifetime of bacterial mRNA. Direct measurements of archaeal mRNA stability indicate they have long halflives and therefore more closely resemble Eukarya (Bini et al., 2002). In addition, Archaea appear to lack most of the key enzymes employed by Bacteria to degrade mRNA particularly those required for initial endonucleolytic cleavage and

115

3 to 5 exonuclease activity. They do encode however, enzymes for the processing, maturation, and degradation of stable RNA that are co-situated in a highly conserved locus distributed throughout the Archaea. Protein synthesis, folding, and turnover. Like Bacteria, archaeal mRNAs are neither capped nor contain polyA tails of significant length. Initiation of bacterial translation depends on the interaction of sequences located 5 to the start codon with the 3 end of the 16S rRNA and initiation factors IF1, 2 and 3. In Eukarya, mRNAs are capped and have long polyA tails, and localization of the start codon by a specialized ribosomal preinitiation complex occurs via a scanning-type mechanism without involvement of rRNA. Surprisingly, comparative analysis of protein factors in Archaea involved in translation indicate a high degree of conservation with those of Eukarya. For translation initiation this includes homologs of eukaryal proteins; eIF1A, eIF2, , and suggesting formation of a preinitiation complex analogous to that of Eukarya, as well as eIF4A and eIF5 implicating the use of a scanning-type mechanism to locate initiation codons. In Archaea, monocistronic genes and those located in promoter-proximal positions within polycistronic units, often lack rRNA complementary sequences to guide incoming ribosomes. They also lack untranslated RNA leaders such that the transcription and translation start sites coincide. In contrast, genes located within polycistronic units do have rRNA complementary sequences. These observations suggest Archaea may employ distinct mechanisms for the initiation of translation of different gene categories. Protein folding in Archaea employs enzymes that are distinct from those found in Bacteria and homologous to those in Eukarya. In Bacteria the key protein folding enzymes include the chaperone system consisting of heat shock protein 70 (HSP70; DnaK), DnaJ and HtpG, and the chaperonin system consisting of HSP60 (GroEL), and GroES. The latter forms double ring toroidal structures readily observed by electron microscopy. Additional activities include enzymes involved in protein secretion, catalysts for protein folding, the small HSPs, and trigger factor (TF). In Bacteria, nascent proteins proceed from the ribosome to TF to the HSP70 system and thence to the HSP60 system. In Archaea, TF is not present, the HSP70 system is very rare and the HSP60 system is absent. In Archaea nascent polypeptides interact possibly with a homolog of the eukaryotic protein, NAC, and subsequently with a eukaryotic homolog of Prefoldin (GimC), which functionally replaces the action of the HSP70 system. Subsequently there is an interaction with an alternate chaperonin system homologous to the eukaryotic TCP1 protein.

ch07.qxd 10/9/03 6:50 PM Page 116

116

the desk encyclopedia of microbiology

The key mechanism employed in Archaea for protein degradation involves a multisubunit hollow barrel-like structure called the 20S proteasome. The same structure is universally present in Eukarya but only rarely present in Bacteria. The proteasome is readily observed by electron microscopy, being 15 nm in length and containing 28 subunits. When present in Bacteria, proteasomes are thought to have been acquired by horizontal transmission. The proteasome associates with ATPase regulatory components in the form of a cap increasing the size of the structure considerably. The eukaryotic ubiquitin protein targeting system is not evident in Archaea and though other energy dependent proteases exist in Archaea additional studies are necessary to determine their distribution and relationship to bacterial proteases. Protein secretion. The translocation (secretion) of proteins through membranes in all organisms employs a ribonucleoprotein complex called signal recognition particle (SRP). To date, this appears to be the sole mechanism in Eukarya and Archaea, while Bacteria have the additional (Sec) system. SRP comprises RNA and associated proteins, which bind nascent polypeptides to promote their interaction and transfer across the cytoplasmic membrane. Archaea like Eukarya have a 7.5S SRP RNA while Bacteria have a smaller 4.5S RNA. The SRP protein components of Archaea (SRP19 and SRP54) are more similar to those of Eukarya though present in fewer numbers. Other components of the archaeal protein secretion system including the leader peptidase that removes a portion of the N-terminal end of secreted proteins, exhibit greater similarity to eukaryotic enzymes.

V. CONCLUSIONS Archaea like Bacteria are prokaryotes with simple cell structures. They can be readily distinguished from Bacteria by differences in the structure and chemistry of their envelopes. With some exceptions they cannot generally be distinguished by biotype, physiology, or metabolism. The most striking difference which appears to unite the Archaea is their use of eukaryal-type genes often with unique functions, to make and process DNA, RNA, and protein. This latter feature has major

implications for the origin of life and the relationship between Archaea and the last common ancestral cell.

BIBLIOGRAPHY Amend, J. P., and Shock, E. L. (2001). Energetics of overall metabolic reactions of thermophilic and hyperthermophilic Archaea and bacteria. FEMS Microbiol. Rev. 25, 175–243. Bell, S. D., and Jackson, S. P. (2001). Mechanism and regulation of transcription in archaea. Curr. Opin. Microbiol. 4, 208–213. Bini, E., Dikshit, V., Dirksen, K., Drozda, M., and Blum, P. (2002). Stability of mRNA in hyperthermophilic archaea. RNA 8, 1129–1136. Blum, P. (2001). Ancient microbes, extreme environments, and the origin of life. Adv. Appl. Microbiol. 50. Boone, D. R., Castenholz, R. W., and Garrity, G. M. (2001). Procaryotic domains. In “Bergey’s Manual of Systematic Bacteriology”, Vol. 1, Springer-Verlag, Berlin. Karner, M. B., DeLong, E. F., and Karl, D. M. (2001). Archaeal dominance in the mesopelagic zone of the Pacific Ocean. Nature 409, 507–510. Madigan, M. T., Martinko, J. M., and Parker, J. (2000). Prokaryotic diversity: the Archaea In “Brock Biology of Microbiology”, Chap. 14, pp. 545–572, Prentice-Hall, Englewood Cliffs, NJ. Reysenbach, A. L., and Cady, S. L. (2001). Microbiology of ancient and modern hydrothermal systems. Trends Microbiol. 9, 79–86. Sandman, K., Soares, D., and Reeve, J. N. (2001). Molecular components of the archaeal nucleosome. Biochimie 83, 277–281. Thomas, N. A., Bardy, S. L., and Jarrell, K. F. (2001). The archaeal flagellum: a different kind of prokaryotic motility structure. FEMS Microbiol Rev. 25, 147–174. Woese, C. R., and Fox, G. E. (1977). Phylogenetic structure of the prokaryotic domain: the primary kingdoms. Proc. Natl. Acad. Sci. USA 74, 5088–5090.

WEBSITES For more information on the systematics of Archaea see: http://www.ncbi.nlm.nih.gov:80/entrez/query.fcgi?dbTaxonomy For more information on archaeal histones see: http://www.biosci.ohio-state.edu/~microbio/Archaealhistones/ For more information on RNA modification in Archaea see: http://rna.wustl.edu/snoRNAdb/ Archaea browser of the NCBI (National Center for Biotechnology Information, USA) http://www.ncbi.nlm.nih.gov/htbin-post/Taxonomy/wgetorg?name Archaea Website for Comprehensive Microbial Resource of The Institute for Genomic Research and links to many other microbial genomic sites http://www.tigr.org/tigr-scripts/CMR2/CMRHomePage.spl List of bacterial names with standing in nomenclature (J. P. Euzéby) http://www.bacterio.cict.fr/index.html

ch08.qxd 10/9/03 6:51 PM Page 117

8 Attenuation, Transcriptional Charles Yanofsky Stanford University

GLOSSARY

transcription pausing A temporary pause or delay in RNA polymerase movement on its DNA template. transcription pause structure An RNA hairpin that causes RNA polymerase to pause or stall during transcription. transcription termination Cessation of RNA synthesis and release of transcript and DNA template from RNA polymerase. transcriptional attenuation A mechanism used to regulate continuation vs termination of transcription.

antiterminator An RNA hairpin structure that generally contains several paired nucleotides that are essential for terminator formation. When these nucleotides are paired in the antiterminator they cannot participate in terminator formation. attenuator A short DNA region that functions as a site of regulated transcription termination. charged tRNA A transfer RNA bearing its cognate amino acid (e.g., Trp-tRNATrp). leader peptide A peptide encoded by the leader segment of a transcript. leader peptide coding region A short peptide coding region in the leader segment of a transcript. RNA-binding attenuation regulatory protein An RNA-binding protein that binds to a specific RNA sequence and, by so doing, either promotes or prevents formation of a transcription terminator. RNA hairpin structure A base-paired stem and loop structure that has sufficient stability to remain in the base-paired, hairpin configuration. terminator ( factor-dependent) An RNA sequence usually causing transcription pausing that serves as a site of factor-dependent transcription termination. terminator (intrinsic) An RNA hairpin followed immediately by a sequence rich in U’s. The terminator serves as a signal to RNA polymerase to terminate transcription. The Desk Encyclopedia of Microbiology ISBN: 0-12-621361-5

Transcriptional Attenuation is the term used to describe a general transcription regulatory strategy that exploits various sensing events and molecular signals to alter the rate of transcription termination at a site or a site preceding one or more genes of an operon. Many mechanisms of transcriptional attenuation exist. Each regulates operon expression by responding to an appropriate molecule or event and determining whether transcription will or will not be terminated.

I. OBJECTIVES AND FEATURES OF REGULATION BY TRANSCRIPTIONAL ATTENUATION It is evident that an appreciable fraction of the genetic material of each organism is dedicated to regulating gene expression. The ability to alter expression provides

117

Copyright © 2003 Elsevier Ltd All rights of reproduction in any form reserved

ch08.qxd 10/9/03 6:51 PM Page 118

118

the desk encyclopedia of microbiology

the variability that an organism needs in order to initiate or respond to the many changes that are associated with or responsible for each physiological and/or developmental event. Initiation of transcription is perhaps the single biological act that is most often subject to regulation. There are numerous examples of “negativeacting” repressor proteins—proteins that inhibit transcription initiation by binding to their respective DNA operator site(s) within or in the vicinity of the regulated promoter. Similarly, there are many examples of “positive-acting” regulatory proteins that activate transcription by binding at specific DNA elements in the vicinity of the affected promoter. Both negativeand positive-acting regulatory proteins are commonly activated or inactivated by small or large molecules as well as by reversible processes, i.e., phosphorylation and dephosphorylation. However, transcription initiation is only one of several common metabolic events that may be modulated to alter gene expression. The two subsequent stages in transcription, transcript elongation and transcription termination, are also common targets for regulatory change. The principal advantages achieved by regulating these events is that different classes of molecules and different metabolic processes can participate in regulatory decisions. Thus, once transcription has begun, the nascent transcript is a potential target for a regulatory event. In addition, in prokaryotes, in which most transcripts are initially translated as they are being synthesized, components of the translation machinery may participate in regulatory decisions. By exploiting these additional targets, organisms have greatly increased their regulatory options. A separate objective may have been to devote as little unique genetic information as possible to a regulatory process. Accordingly, some of the transcriptional attenuation regulatory mechanisms that will be described use less than 150 bp of DNA to achieve gene- or operon-specific control. Often attenuation regulation is achieved using only the common cell components that participate in RNA and protein synthesis. In this article, I review the features of several examples of regulation by transcriptional attenuation.

II. MECHANISMS OF TRANSCRIPTIONAL ATTENUATION A. Regulation of termination at an intrinsic terminator Many operons regulated by transcriptional attenuation contain a DNA region that specifies a RNA

sequence that can fold to form a hairpin structure followed by a run of U’s, a structure called an intrinsic terminator. Intrinsic terminators instruct RNA polymerase to terminate transcription. The region encoding the intrinsic terminator is located immediately preceding the gene or genes that are being regulated. The transcript segment before and including part of the terminator often contains a nucleotide sequence that can fold to form a competing, alternative hairpin structure called the antiterminator. The existence of this structure prevents formation of the terminator. Antiterminator and terminator structures generally share a short nucleotide sequence, which explains why prior formation of the antiterminator prevents formation of the terminator. Additional features of the nucleotide sequence preceding or following a terminator or antiterminator can influence whether these structures will form or act. The transcript segment preceding the terminator often contains sequences that allow the organism to sense a relevant metabolic signal and to respond to that signal by allowing or preventing antiterminator or terminator formation. A variety of mechanisms are used to sense specific cell signals. In operons concerned with amino acid biosynthesis, ribosome translation of a peptide coding region rich in codons for a crucial amino acid is often used to sense the presence or absence of the corresponding charged tRNA. Depending on the location of the translating ribosome on the transcript, an antiterminator will or will not form. In another example, in an operon concerned with pyrimidine biosynthesis, coupling of RNA synthesis with translation is employed to sense the availability of a nucleotide needed for RNA synthesis. In some mechanisms, RNA-binding proteins regulate termination. These proteins bind to specific transcript sequences or structures and allow or prevent antiterminator or terminator formation. One common regulatory mechanism is designed to sense the relative concentrations of a charged and uncharged tRNA and, depending on which is in excess, induce formation of an antiterminator or terminator. It is evident from these and other examples that regulation of the formation of an intrinsic terminator is a common strategy used to alter operon expression. 1. Ribosome-mediated attenuation Synthesis of most proteins requires not only the availability of all 20 amino acids but also these amino acids must be in their activated state, covalently attached to their respective tRNAs. As such, they are primed for participation in polypeptide synthesis. The intracellular concentration of each amino acid reflects a balance

ch08.qxd 10/9/03 6:51 PM Page 119

attenuation, transcriptional

119

of several events, including rates of synthesis, utilization, import from the environment, and release from proteins by degradation. Occasionally, induction of a degradative pathway also affects the cellular level of an amino acid. The concentration of a specific charged tRNA also reflects several events, including the presence of the corresponding amino acid, its rate of charging onto tRNA, the availability of that tRNA, and use of that charged tRNA in protein synthesis. Other factors also affect the rate of protein synthesis, such as whether there are rare codons in the coding region being translated and whether all needed species of charged tRNA are available. The availability of free ribosomes and accessory molecules required for protein synthesis also has an impact on the rate of protein synthesis. Given these many variables, it is not surprising that so many attenuation mechanisms are used to sense and respond to specific cellular needs. a. The trp operon of Escherichia coli Transcription of the trp operon of E. coli is regulated by both repression and transcriptional attenuation. The initial event in regulation by attenuation is the formation of a RNA hairpin structure that directs the transcribing RNA polymerase molecule to pause after initiating transcription (Fig. 8.1, stage 1). This transcription pause provides sufficient time for a ribosome to bind to the ribosome binding site of a peptide coding region in the leader transcript and initiate translation (Fig. 8.1, stage 2). The moving ribosome in fact releases the paused polymerase, permitting resumption of transcription (Fig. 8.1, stage 3). Thereafter, transcription and translation proceed in unison. As the polymerase molecule transcribes the leader region, the translating ribosome moves along the transcript and reaches a segment that is capable of folding to form an antiterminator structure. However, whether or not this structure forms depends on the location of the translating ribosome. In a bacterium deficient in charged tRNATrp, the translating ribosome would stall over either of two adjacent Trp codons in the leader peptide coding region (Fig. 8.1, stage 4). Stalling would allow a downstream RNA segment to fold and form an antiterminator hairpin structure. As transcription proceeds, persistence of the antiterminator would prevent formation of the terminator since paired nucleotides at the base of the antiterminator must be free for terminator formation to occur. Under these conditions, transcription would continue into the structural genes of the operon. In a cell with adequate levels of charged tRNATrp (Fig. 8.1, stage 4 alternate), the tandem Trp codons would be translated and the

FIGURE 8.1 Stages in ribosome-mediated transcriptional attenuation regulation of the trp operon of E. coli. Transcription initiation and pausing (stage 1), ribosome loading (stage 2), and initiation of translation and release of the pause transcription complex (stage 3) occur under all conditions. When a cell is deficient in tryptophancharged tRNATrp (stage 4), the translating ribosome stalls at either of the two Trp codons in the leader peptide coding region. Stalling permits the antitterminator to form; this prevents terminator formation, allowing transcription to continue into the structural genes of the operon. When a cell has sufficient charged tRNATrp to support ongoing protein synthesis (stage 4 alternate), translation proceeds to the leader peptide stop codon. A ribosome at this position blocks formation of the antiterminator structure and permits the terminator to form and cause termination.

translating ribosome would proceed to the leader peptide stop codon. At this position, the ribosome would block formation of the antiterminator and allow the terminator to form; hence, transcription would be terminated.

ch08.qxd 10/9/03 6:51 PM Page 120

120

the desk encyclopedia of microbiology

The leader regions of many bacterial operons, such as the his, phe, leu, thr, ilvGMEDA, and ilvBN operons, are organized much like that of the trp operon of E. coli. These operons appear to be regulated by the same mechanism, with only minor variations tailored to each operon’s needs. Generally, the leader region sequence and organization reflects differences in regulatory requirements. For example, transcription of the his operon of S. typhimurium is regulated only by attenuation, unlike transcription of the trp operon of E. coli which is regulated by both repression and attenuation. The his operon’s leader peptide coding region contains seven consecutive His codons. This organization allows greater sensitivity to changes in the cellular level of charged tRNAHis; a slight reduction is sufficient to delay ribosome movement through the His codon region. Any delay promotes antiterminator formation. An operon with a leader region that is organized differently is the ilvGMEDA operon of E. coli. Here, attenuation is regulated in response to the availability of three charged tRNAs those for tRNAIle, tRNAVal, and tRNAThr. Codons for these tRNAs are arranged in the leader peptide coding region so that a deficiency of any of these charged species would promote antiterminator formation. Another operon regulated similarly is pheST of E. coli. This operon specifies the two polypeptides of phenylalanyl-tRNA synthetase. Translation of its leader peptide coding region containing five Phe codons is used to regulate termination/antitermination. An interesting consideration that bears on pheST operon regulation is that the product of this operon, phenylalanyl-tRNA synthetase, is needed under all growth conditions. b. The pyrBI operon of E. coli Another well-studied example in which ribosomemediated attenuation regulates transcription of an operon concerns the pyrBI operon of E. coli. When a cell has inadequate levels of UTP for RNA synthesis, the UTP deficiency triggers transcription antitermination in the leader region of this operon (Fig. 8.2). Continued transcription of the operon allows the cell to increase its rate of pyrimidine nucleotide synthesis. The pyrBI leader transcript has several features that explain its role in transcription regulation. It can fold to form alternative antiterminator and terminator structures. In addition, the leader segment contains the coding region for a leader peptide; this coding segment overlaps the antiterminator and terminator. The leader transcript also has several U-rich sequences which play a role in transcription pausing. When the UTP level is insufficient to sustain continued RNA

FIGURE 8.2 Stages in transcriptional attenuation in the pyrBl operon of E. coli. When a cell is deficient in UTP, the RNA polymerase molecule that is transcribing the pyrBl operon leader region pauses at one or more UTP deficiency-dependent pause sites (stage 1). While the polymerase is paused a ribosome binds to the leader transcript and initiates translation (stage 2). When the polymerase is released, the translating ribosome moves closely behind the transcribing polymerase. Continued translation by this ribosome prevents formation of the terminator structure; thus, transcription continues into the structural genes of the operon (stage 3). When there are adequate levels of UTP to support rapid RNA synthesis (bottom) the transcribing polymerase pauses very briefly in the leader region and then continues transcription. The terminator sequence is formed well before the translating ribosome can approach this segment of the transcript. Terminator formation results in termination.

synthesis, the polymerase transcribing the pyrBI operon stalls at these U-rich pause sites (Fig. 8.2, stage 1). Reduced polymerase migration allows sufficient time for a ribosome to bind to the transcript and move closely behind the polymerase (Fig. 8.2, stage 2). A translating ribosome at this position could prevent formation of the terminator structure; thus, transcription of the operon would continue (Fig. 8.2, stage 3). When a cell has an adequate level of UTP, the transcribing polymerase molecule moves through the pause sites rapidly and is positioned well ahead of the

ch08.qxd 10/9/03 6:51 PM Page 121

attenuation, transcriptional translating ribosome. This separation permits the terminator to form and cause transcription termination. Transcription of this operon is also regulated by an unrelated UTP-dependent mechanism. c. Other examples A related although different mechanism of ribosomemediated transcription attenuation is used to regulate expression of the ampC operon of E. coli. The regulatory region of this operon, preceding ampC, encodes a leader transcript segment containing a ribosome binding site, adjacent start and stop codons, and a sequence that can form an intrinsic terminator. Expression of this operon is subject to growth rate regulation. During rapid growth, when the ribosome content per cell is high, a ribosome is likely to bind at the ribosome binding site in the leader segment and interfere with terminator formation. Under these conditions, transcription of the operon will continue. When the ribosome content per cell is low, the leader segment of the transcript is likely to be ribosome free for a period sufficiently long to allow the terminator to form and promote termination. An antiterminator is not used in this attenuation mechanism.

121

(RNA-binding) species (Fig. 8.3). BglF is a membranebound phosphoenolpyruvatedependent phosphotransferase. When BglF senses a -glucoside, it phosphorylates the sugar and transports it into the cell. Substrate-activated BglF also dephosphorylates BglG, converting it into the active, RNA-binding dimeric form. In the absence of a -glucoside, BglF phosphorylates BglG, rendering it monomeric and inactive. The transcript segment preceding bglG and blgF can fold to form either an antiterminator or a terminator structure. When BglG is dephosphorylated and active, it binds to and stabilizes the antiterminator (Fig. 8.3). Since the stem of the antiterminator contains bases that are part of the terminator, the terminator does not form. Nucleotides in the single-stranded loop region of each antiterminator as well as paired bases in the antiterminator stem appear to be the sites of BglG binding. BglG is believed to act similarly at the two antiterminators. When BglG is inactive and the bgl operon is being transcribed, terminator structures form in the transcript and terminate transcription (Fig. 8.3). The sacB and sacPA genes of B. subtilis, genes concerned with sucrose utilization, appear to be regulated by a very similar antitermination/termination mechanism. The protein products of genes sacY and

2. Binding protein-mediated attenuation In several operons regulated by transcriptional attenuation, specific RNA-binding proteins determine whether or not transcription will be terminated. These proteins recognize specific sites or sequences in a transcript and, by binding, regulate formation of an antiterminator or terminator. Well-studied examples include the bgl operon of E. coli and the sac operon of Bacillus subtilis, which are regulated similarly, and the trp and pyr operons of B. subtilis, which are regulated differently. The RNA-binding regulatory proteins that regulate transcription of these operons function much like the stalled ribosome in amino acid biosynthetic operons, as described previously. a. The bgl operon of E. coli The bgl operon of E. coli, bglG–bglF–bglB, is a threegene operon encoding proteins required for the utilization of -glucosides as carbon sources. The operon contains two independent sites of regulated transcription termination, the first before bglG and the second between bglG and bglF. The products of the first two genes of the operon, BglG and BglF, are necessary for regulation of this operon by attenuation. BglG exists in two forms: a phosphorylated, monomeric, inactive species and a dephosphorylated, dimeric, active

FIGURE 8.3 Protein-mediated transcriptional attenuation in the bgl operon of E. coli. In the presence of a -glucoside carbon source the BglF protein phosphorylates the sugar and transports it into the cell (top). -Glucoside-activated BglF also dephosphorylates the BglG protein. Dephosphorylated BglG dimerizes, and the dimer binds at one or both of the antiterminators in the transcript of the bgl operon, stabilizing the antiterminator structure. The existence of the antiterminator prevents formation of the terminator; thus, transcription proceeds. In the absence of a -glucoside BglF phosphorylates BglG and the phosphorylated form remains as a monomer, incapable of binding to RNA (bottom). Under these conditions, the antiterminator is not stabilized, and the terminator forms, terminating transcription.

ch08.qxd 10/9/03 6:51 PM Page 122

122

the desk encyclopedia of microbiology

sacT regulate sacB and sacPA expression, respectively. The leader regions preceding sacB and sacPA specify RNA antiterminator structures that closely resemble those of the bgl operon. Dephosphorylation of SacY by SacX, in response to the presence of sucrose, leads to antitermination of transcription in the leader region preceding sacB. The proteins and sites involved in attenuation control in the sac and bgl systems are homologous. b. The trp operon of B. subtilis The leader segment of the transcript of the trp operon of B. subtilis can fold to form mutually exclusive antiterminator and terminator structures (Fig. 8.4). When a cell is deficient in tryptophan and the leader region of the operon is being transcribed, the antiterminator forms, preventing terminator formation and termination (Fig. 8.4). In the presence of excess tryptophan, an RNA-binding protein, TRAP (trp RNAbinding attenuation protein), encoded by the mtrB gene, binds tryptophan and becomes activated. Activated TRAP can bind to the trp operon leader transcript while it is being synthesized. The TRAP binding site consists of a series of U/GAG repeats located immediately preceding and including part of the antiterminator structure (Fig. 8.4). TRAP binding to the

FIGURE 8.4 Protein-mediated transcriptional attenuation in the trp operon of B. subtilis. When a cell is deficient in tryptophan the TRAP protein is not active, the leader region of the trp operon is transcribed, the antiterminator forms, and transcription continues into the operon (top). When a cell has sufficient tryptophan to support rapid growth, the TRAP protein is activated by bound tryptophan (bottom). Activated TRAP binds at U/GAG repeat sequences (small boxes) in the transcript segments located before and within the antiterminator. Bound TRAP essentially melts the antiterminator, allowing a sequence at the base of the antiterminator to exist in an unpaired form. This unpaired sequence participates in the formation of the terminator hairpin structure, which promotes termination.

transcript prevents formation of the antiterminator, thereby promoting formation of the terminator. The 3D structure of TRAP has been described, and the residues in the protein principally responsible for RNA binding have been identified. The protein is doughnut shaped and consists of 11 identical subunits, each of which associates with a U/GAG sequence in the transcript. TRAP is believed to wrap the single-stranded leader transcript around its periphery and, by so doing, prevent formation of the antiterminator. Tryptophan-activated TRAP also binds to a similar sequence of U/GAG repeats that precede the trpG coding region. trpG is the sole trp gene of B. subtilis that is not in the trp operon. trpG is located in a folate operon and specifies a bifunctional polypeptide that functions both in tryptophan and in folate biosynthesis. A TRAP binding site overlaps the trpG ribosome binding site; thus, TRAP binding inhibits translation of trpG. TRAP action therefore coordinates trp gene expression in the folate and tryptophan operons. c. The pyr operon of B. subtilis Another example of transcriptional attenuation mediated by a RNA-binding protein concerns regulated expression of the pyr operon of B. subtilis. This organism produces a novel uracil phosphoribosyltransferase, PyrR, that also functions as a RNA-binding transcription regulator. PyrR can bind at similar sites in three ~150-nt untranslated segments of the pyr transcript, each preceding a polypeptide coding segment and each containing a terminator. The first terminator precedes the first gene in the operon, pyrR, which in fact encodes this RNA-binding regulatory protein/ enzyme. The second terminator is located between pyrR and pyrP; pyrP encodes a uracil permease. The third terminator is located between pyrP and pyrB. pyrB specifies aspartate transcarbamylase. Each of the three untranslated segments of the transcript can fold to form an alternative antiterminator structure that can prevent formation of an intrinsic terminator. In addition, each untranslated transcript segment can form a third structure, earlier in the transcript, termed an anti-antiterminator. This structure includes part of the antiterminator; thus, its formation prevents formation of the antiterminator. When pyrimidines are plentiful PyrR binds to the nascent pyr operon transcript and stabilizes the anti-antiterminator stem-loop structure. Stabilization of this structure blocks formation of the antiterminator structure, promoting formation of the terminator and thereby causing termination. When cells are deficient in pyrimidines and PyrR is inactive the antiterminator prevents formation of the terminator, allowing transcription to continue.

ch08.qxd 10/9/03 6:51 PM Page 123

attenuation, transcriptional

123

The three antiterminators are predicted to be the most stable of the several RNA structures. PyrR’s RNA binding ability is responsive to the relative concentrations of UMP and PRPP, with bound UMP favoring RNA binding and bound PRPP preventing UMP binding and activation of the protein. The 3D structure of PyrR of B. subtilis has been determined. The RNA sequences that are recognized have also been identified. Several bacterial species appear to produce homologs of PyrR and to regulate their pyr operons by the same or a similar mechanism. The organization of the leader region of the pur operon of B. subtilis suggests that this operon is regulated by transcription termination/antitermination in response to changes in the availability of guanine nucleotides. d. The S10 operon of E. coli The 11-gene S10 ribosomal protein operon of E. coli contains a 172-base pair leader regulatory region which is used to achieve protein-mediated transcriptional attenuation. The S10 operon is regulated autogenously; that is, the product of one of its structural genes, protein L4, binds to the S10 leader transcript and regulates transcription termination. L4 binding also inhibits translation of coding regions of the operon. The transcript of the leader region forms multiple hairpin structures, two of which are essential for L4 activity. During transcription of the leader region RNA polymerase pauses after synthesizing one of these hairpins, a potential intrinsic terminator. Pausing at this site is enhanced in vitro by bound NusA protein and, most important, the pause complex is further stabilized by bound L4 protein. Enhanced stabilization of the terminator hairpin is believed to be responsible for efficient transcription termination. The leader RNA terminator structure, hairpin HE, participates in these events. An additional hairpin, HD, just preceding hairpin HE, also influences termination. How the structure HD is involved is not understood. 3. tRNA-mediated attenuation a. tRNA synthetase operons of B. subtilis In B. subtilis and other gram-positive bacteria, many operons encoding aminoacyl-tRNA synthetases, and some operons encoding amino acid biosynthetic enzymes, are regulated by tRNA-mediated transcriptional attenuation (Fig. 8.5). Each of these operons contains a leader region that specifies a transcript segment that can fold to form a complex set of structures, two of which are mutually exclusive and function as

FIGURE 8.5 Uncharged tRNA-mediated transcriptional attenuation in tRNA synthetase and amino acid biosynthetic operons of B. subtilis and other gram-positive bacteria. When a bacterial cell is defective in charging a tRNA with the corresponding amino acid, the uncharged tRNA pairs with the leader transcript of the operon specifying the tRNA synthetase that charges that amino acid. Two segments of the tRNA are believed to be involved in RNA–RNA pairing. One segment, the anticodon of the tRNA, is thought to pair with a complementary sequence in a side bulge in the leader transcript, called the specifier. The acceptor end of the tRNA is also believed to pair with the leader transcript. Its target is a single-stranded bulge sequence in the antiterminator, called a T box. Pairing of the uncharged tRNA at these two sites is proposed to stabilize an antiterminator structure, thereby preventing formation of the terminator (top). When the relevant tRNA is mostly charged, it cannot pair with the T box sequence. The leader RNA then folds to form the terminator structure, which terminates transcription (bottom).

antiterminator and terminator. Translation is not used to choose between these alternative RNA structures. Rather, each leader RNA is designed to recognize the accumulation of an uncharged tRNA species as the signal to prevent termination. The crucial recognition sequence in leader RNA includes a single-stranded segment with a triplet codon, designated the specifier sequence (Fig. 8.5). The triplet specifier is located in a side bulge of a RNA hairpin structure. The specifier sequence is complementary to the anticodon of the tRNA that is a substrate of the tRNA synthetase that is being regulated. In amino acid biosynthetic operons regulated by this mechanism, this triplet codes for the amino acid that is synthesized by the proteins specified by the operon. A second tRNA binding site, termed a T box, located within a side bulge in the antiterminator, is complementary to nucleotides preceding the acceptor end of the tRNA (Fig. 8.3). The current regulatory model (Fig. 8.5) predicts that when an uncharged tRNA is plentiful, it binds to the specifier and T box of an appropriate leader RNA, stabilizing the antiterminator

ch08.qxd 10/9/03 6:51 PM Page 124

124

the desk encyclopedia of microbiology

and thereby preventing terminator formation. When the tRNA is charged, its acceptor end is blocked by an amino acid and thus it cannot pair with the T box. Under these conditions, the terminator will form, resulting in transcription termination. The charged tRNA apparently is still recognized because it competes with uncharged tRNA. Switching the codon in a leader RNA can change the specificity of the response. Although the events described only concern interactions between tRNA and leader RNA, unidentified factors may also participate.

There are other examples, particularly in bacteriophage, in which specific viral proteins mediate antitermination events. In these instances, the mechanisms of antitermination vary somewhat from the mechanism attributed to the N protein. In addition, the ribosomal RNA operons of E. coli are known to be regulated by an antitermination mechanism that prevents Rho-dependent termination. This system has several features in common with N-mediated antitermination, including use of some of the same proteins and similar RNA binding sites.

B. Regulation of termination at a factor-dependent terminator

2. Translation-mediated antitermination in the tna operon

In many bacterial species there is a second class of transcription termination sites—factor-dependent sites—at which a specific protein, Rho, interacts with RNA polymerase and causes termination. Rhodependent termination requires an unstructured RNA segment as a site of Rho binding and a downstream RNA segment as a site of RNA polymerase pausing and termination. Accessory proteins that interact with RNA polymerase or with Rho also influence the termination process. Generally, once Rho binds to a transcript it migrates in the 3 direction until it contacts a stalled polymerase. When it does, it can trigger the act of termination. Rho-dependent termination sites are not intrinsic terminators.

Escherichia coli and other bacteria contain operons that encode enzymes that can degrade specific amino acids, making them available as carbon and/or nitrogen sources. The tryptophanase (tna) operon of E. coli is one example. This operon encodes two polypeptides— one that degrades tryptophan and another that transports tryptophan into the cell. Transcription of the structural genes of this operon is subject to regulation by transcriptional attenuation. Transcription initiation in the operon is regulated by catabolite repression. Attenuation is mediated by a mechanism that involves tryptophan-induced transcription antitermination. The anititermination process prevents Rho from terminating transcription at specific sites in the leader region of the operon. The transcript of the leader region contains a short peptide coding region, tnaC, which has a single Trp codon. Synthesis of the 24-residue TnaC peptide, with its crucial Trp residue and certain other key residues, is essential for antitermination. In the presence of inducing levels of tryptophan the nascent TnaC peptide is believed to act in cis on the ribosome engaged in synthesizing the peptide, inhibiting its release at the leader peptide stop codon. Ribosome release at this stop codon is thought to be essential for termination since release is required to expose a presumed Rho entry/binding site in the vicinity of the tnaC stop codon. How the inducer tryptophan is recognized is not known, nor is it known how the leader peptide interacts with the translating ribosome to block its release at the tnaC stop codon.

1. N protein-mediated antitermination in phage The earliest studied and most thoroughly analyzed example of regulation by transcription termination/ antitermination involves the action of the N protein of bacteriophage in mediating antitermination at sites of Rho-dependent termination in the phage genome. Regulation at these sites controls expression from major leftward and rightward phage promoters. N protein functions by interacting with RNA polymerase, forming an antitermination complex. This requires cisacting transcript sites and sequences, called nut sites. These sites are composed of two elements, a BoxA sequence and a BoxB sequence. Box-B folds to form a hairpin loop structure. N associates with BoxB, and several host proteins associate with N and RNA polymerase in the formation of the antitermination complex. Other proteins in the complex either recognize the BoxA sequence or associate with N and the transcribing RNA polymerase complex. The fully formed N protein–RNA polymerase antitermination complex is resistant to the action of Rho. This antitermination complex can transcribe through intrinsic terminators as well as sites of Rho-dependent termination.

III. CONCLUSIONS The transcriptional attenuation mechanisms described previously achieve operon-specific regulation by modifying one or more biological events that influence transcription termination. Use of these mechanisms greatly expands the regulatory capacity

ch08.qxd 10/9/03 6:51 PM Page 125

125

attenuation, transcriptional of each organism. An additional advantage is that these mechanisms permit a facile adjustment of the basal level of expression of an operon—expression in the absence of signals that regulate termination. Thus, variations in RNA structure, stability, or arrangement can establish an appropriate basal level of operon expression. In addition, as mentioned previously, some transcriptional attenuation mechanisms are economical because they require little unique genetic information. In eukaryotes there are several examples of regulated transcription delay with features resembling those of some of the attenuation mechanisms that were described.

BIBLIOGRAPHY Grunberg-Manago, M. (1996). Regulation of the expression of aminoacyl-tRNA synthetases and translation factors. In “Transcription Attenuation in Escherichia coli and Salmonella: Cellular and Molecular Biology” (F. Neidhardt et al., Eds.), pp. 1432–1457. ASM Press, Washington, DC. Hatfield, G. W. (1996). Codon context, translational step—Times and attenuation. In “Regulation of Gene Expression in E. coli” (E. C. C. Lin and A. S. Lynch, Eds.), pp. 47–65. Landes/ Chapman & Hall, Austin, TX.

Henkin, T. M. (1996). Control of transcription termination in prokaryotes. Annu. Rev. Genet. 30, 35–57. Landick, R., Turnbough, C. L., Jr., and Yanofsky, C. (1996). Transcription attenuation. In “Escherichia coli and Salmonella: Cellular and Molecular Biology” (F. Neidhardt et al., Eds.), pp. 1263–1286. ASM Press, Washington, DC. Platt, T. (1998). RNA structure in transcription elongation, termination, and antitermination. In “RNA Structure and Function,” pp. 541–574. Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY. Roberts, J. W. (1996). Transcription termination and its control. In “Regulation of Gene Expression in E. coli” (E. C. C. Lin and A. S. Lynch, Eds.), pp. 27–45. Landes/Chapman & Hall, Austin, TX. Switzer, R. L., Turner, R. J., and Lu, Y. (1999). Regulation of the Bacillus subtilis pyrimidine biosynthetic operon by transcriptional attenuation: control of gene expression by an mRNAbinding protein. Proc. Nucleic Acid Res. Mol. Biol. 62, 329–67. Yanofsky, C. (2000). Transcription attenuation: once viewed as a novel regulatory strategy. J. Bacteriol. 182(1), 1–8.

WEBSITE Website for Comprehensive Microbial Resource of The Institute for Genomic Research and links to many other microbial genomic sites http://www.tigr.org/tigr-scripts/CMR2/CMRHomePage.spl.

ch09.qxd 10/9/03 6:52 PM Page 126

9 Bacillus subtilis, Genetics Kevin M. Devine Trinity College, Dublin

GLOSSARY

regulator. The kinase is activated when it senses some environmental or nutritional parameter. It then activates the response regulator, which alters gene expression in a manner that allows the bacterium to respond to the prevailing conditions.

competence Development of the ability to bind and internalize DNA from the medium. endospore A metabolically quiescent cell that is resistant to desiccation, ultraviolet light, and other environmental insults. forespore The cell compartment of the sporangium destined to become the spore. integrating plasmid A plasmid that cannot replicate autonomously in a host bacterium. It can, however, establish itself by integration into the chromosome through recombination between homologous plasmid and chromosomal sequences. mother cell The compartment of the sporangium which engulfs the forespore, synthesizes spore coat proteins, and lyses when the mature endospore is formed. polymerase chain reaction Amplification of specific DNA sequences in vitro using oligonucleotide primers and thermostable DNA polymerase. sigma factor A transcription factor which recognizes specific DNA sequences and directs RNA polymerase to initiate transcription at these sites. SOS response A regulon that is induced to protect cells against DNA damage. sporangium The developing bacterial cell. sporulation The developmental process whereby the bacterial cell forms a quiescent spore. two-component system A signal transduction system composed of a sensor kinase and a response The Desk Encyclopedia of Microbiology ISBN: 0-12-621361-5

Bacillus subtilis is an endospore-forming, gram-positive, rod-shaped bacterium. Several characteristics of B. subtilis have attracted intense interest and therefore it has become a model system for bacterial research. It produces enzymes that are widely used in the brewing, baking, and washing powder industries. Because its products have traditionally been used in the food industry, B. subtilis is classified as a GRAS organism (generally regarded as safe) and is therefore a natural choice of host for the production of heterologous proteins using recombinant DNA methodology. Bacillus subtilis cells become naturally competent during the transition between exponential growth and the stationary phase of the growth cycle. Competent cells have the ability to bind and internalize DNA present in the medium. Therefore, although the regulation of competence development has attracted research interest, competence development has provided the means through which B. subtilis can be readily genetically manipulated. This has led to the development of sophisticated molecular techniques, primarily based on integrating plasmids and transposons, for genetic analysis. Spore formation is a developmental process whereby a vegetative cell undergoes a series of morphological

126

Copyright © 2003 Elsevier Ltd All rights of reproduction in any form reserved

ch09.qxd 10/9/03 6:52 PM Page 127

bacillus subtilis, genetics events to become a metabolically quiescent spore. This process involves temporal and spatial regulation of gene expression and communication between the forespore and mother cell of the sporangium. These features make spore formation in B. subtilis an attractive model system to study development. The complete genome sequence of B. subtilis was published in November 1997. This knowledge has greatly expedited research efforts in this bacterium. It has also revealed that the genome encodes many genes that cannot be assigned a function. The challenge now is to determine how these genes contribute to the cellular metabolism and physiology.

I. CHARACTERISTICS OF BACILLUS SUBTILIS A. Taxonomy and habitat The genus Bacillus consists of gram-positive, endospore-forming, rod-shaped bacteria. There are more than 70 species, which display wide morphological and physiological diversity. Only 2 (B. anthracis and B. cereus) are known to be human pathogens. The defining feature of the genus is endospore formation. The genus is subdivided into six groups using a variety of morphological (particularly sporangial) and metabolic criteria. Bacillus subtilis belongs to group II, whose distinguishing features are (i) the formation of an ellipsoidal spore which does not swell the mother cell, (ii) the ability of cells to grow anaerobically with glucose as the carbon source in the presence of nitrate, and (iii) the production of acid from a variety of sugars. The natural habitat of B. subtilis is the soil, but it is also found in fresh water, coastal waters, and oceans. The ubiquity of the bacterium is probably a consequence of endospore formation, which allows survival after exposure to even the most hostile environments. Bacillus subtilis is also associated with plants, animals, and foods and is found in animal feces. The significance of these associations is not clear. It is thought that its presence in feces is merely the result of ingestion and passage through the gut, whereas a synergistic relationship may exist with plants in which the bacterium enhances the supply of nutrients.

B. Development of competence Bacillus subtilis cells become competent naturally. Competence is the ability to bind and internalize exogenous DNA from the medium. This capability develops during nutrient limitation when cells are in

127

transition between exponential growth and the stationary phases of the growth cycle. Only 10% of the cell population becomes competent. The competent and non-competent fractions can be separated using renograffin gradients indicating that they are morphologically distinguishable. Competent cells can also be distinguished because they do not engage in either macromolecule or nucleotide synthesis and the SOS response is induced. The mechanics of DNA binding and internalization have been established. DNA fragments of heterogeneous size adhere noncovalently to approximately 50 binding sites on the cell surface. DNA binding is not sequence specific. The DNA is then fragmented randomly. During internalization, one strand (chosen randomly) is degraded while the other is transported into the cytoplasm. Internalized DNA fragments are approximately 10 kilobases in size. The nature of the transforming DNA determines its fate: DNA that is homologous to the bacterial chromosome will form a heteroduplex with the chromosome leading to homologous recombination. Plasmid DNA will be established as autonomously replicating molecules.

C. Enzyme and antibiotic production Bacillus species produce a range of enzymes and antibiotics in response to nutrient limitation. The enzymes include proteases, amylases, cellulases and lipases. Production is maximal when cells are in the stationary phase of the growth cycle. Production of these enzymes is presumably a survival strategy to scavenge macromolecular energy sources when nutrient levels are low. Many of these enzymes are widely used in the food, brewing, and biological washing powder industries. Enzymes with useful properties, such as thermostability, activity over a wide pH range, activity in detergents and oxidizing environments, have been identified in many Bacillus species. The role of B. subtilis in the enzyme industry is twofold: (i) Many Bacillus species are refractory to genetic analysis and B. subtilis is therefore the organism of choice to study the regulation of enzyme production and (ii) heterologous genes encoding enzymes with desirable properties can be cloned into B. subtilis strains which have been manipulated to give high product yields. Bacillus species also produce antibiotics when cells enter the stationary phase of the growth cycle. This is probably a strategy to limit bacterial competition for the energy sources liberated through macromolecular degradation by the scavenging enzymes. Bacillus subtilis produces a range of peptide antibiotics, including subtilin, surfactin, bacillomycin, bacilysin,

ch09.qxd 10/9/03 6:52 PM Page 128

128

the desk encyclopedia of microbiology

and fengycin, that display a range of antibacterial and antifungal activities. Although the synthesis of these antibiotics and their role(s) in bacterial cell physiology and survival are academically interesting, they are not of great medical importance. They are synthesized by a variety of mechanisms: For example, subtilin is a lantibiotic (contains the modified amino acid lanthionine) which is produced ribosomally, whereas surfactin is produced by the multienzyme thiotemplate mechanism. The complete genome sequence (see Section II) has revealed many of the loci encoding enzymes for antibiotic synthesis: for example, pks encodes a polyketide synthase, srf encodes surfactin synthetase, and pps encodes a peptide synthetase. These three loci comprise 4% of the total genome length.

D. Sporulation Bacillus subtilis undergoes spore formation in response to carbon, nitrogen, or phosphate limitation. This process results in the formation of a metabolically quiescent cell that is resistant to desiccation, ultraviolet (UV) light, and other environmental insults. The

process of sporulation involves temporal and cell typespecific regulation of gene expression, intercellular communication (between mother cell and forespore), morphological differentiation and programmed cell death (bacterial apoptosis). Such features are characteristic of more complex developmental systems. Sporulation in B. subtilis is therefore a simple developmental system amenable to genetic and biochemical analysis. The process requires 6–8 hr for completion and can be divided into seven stages (Fig. 9.1). At stage 0, the cell senses its environment and makes the decision to initiate sporulation. At stage II an asymmetric cell division has occurred, with the larger cell becoming the mother cell and the smaller cell the forespore. At stage III, the mother cell has completely engulfed the forespore to produce a cell within a cell. A cell type-specific program of gene expression has been established in each compartment at this stage. A series of morphological changes occur between stages IV and VI that lead to the formation of the spore cortex and spore coat. At stage VI, the developing endospore becomes resistant to heat, UV light, and desiccation, and at stage VII the mother cell lyses and releases the mature dormant spore.

FIGURE 9.1 The morphological stages of sporulation in Bacillus subtilis. The decision to sporulate has occurred (stages 0 and 1) with two chromosomes (wavy circles) located at opposite poles of the cell. An asymmetric cell division occurs (stage II) with a single chromosome positioned in each compartment. At this stage, SigmaF is activated only in the forespore (smaller) compartment. During engulfment of the forespore (stages II and III), SigmaE becomes active in the mother cell (larger) compartment. SigmaG becomes active on completion of engulfment (stage III). At stage IV, SigmaK is activated in the mother cell and a layer of cortex (stippled ellipse) surrounds the developing spore. Further morphological changes occur during stages IV–VI that include deposition of a coat (dark ellipse) outside the cortex. The mother cell lyses (stage VII), releasing the mature ellipsoid spore [from Stragier and Losick (1996) with permission, from the Annual Review of Genetics, Volume 30, © 1996, by Annual Reviews].

ch09.qxd 10/9/03 6:52 PM Page 129

bacillus subtilis, genetics

II. THE COMPLETE GENOME SEQUENCE OF B. SUBTILIS A. Genome organization The complete nucleotide sequence of the B. subtilis genome was published in 1997. The circular genome is 4,214 kilobases in size and has an average G C content of 43.5%. There are 10 regions which have a G C content significantly lower than average and that correspond to known bacteriophage and bacteriophage-like elements. The origin and terminus of replication are almost perfectly diametrically opposed on the genome. The B. subtilis genome displays significant GT skew at third codon positions in common with many other bacteria. The leading strands have an excess of G (9%) and T (4%) over the lagging strands, and the position at which skew reversal occurs corresponds to the positions of the origin and terminus of replication. Approximately 87% of the genome is coding. More than 74% of all open reading frames and 94% of ribosomal genes are transcribed co-directionally with replication.

B. Gene composition Fifty-three percent of genes are present in single copy. The remainder are present in multigene families, which range in size from those with 2 gene copies (568 genes are duplicated) to the ABC family of transporters that has 77 members. Multigene families present the opportunity for individual member genes to diverge and fulfill different functions and roles within the cell. In addition, individual members can have different regulatory signals so that they can be expressed under different environmental and nutritional conditions. Approximately 220 transcriptional regulators have been identified, including a family of 18 sigma factors (18 different types of promoter), a family of 34 two-component systems, 20 members of the GntR family, 19 members of the LysR family, and 12 members of the Lacl family. It is evident, therefore, that B. subtilis has the potential to sense and respond to nutritional and environmental signals in a complex manner. This may be a reflection of the varied habitats in which B. subtilis can survive.

C. Gene identity Approximately 58% of genes can be assigned an identity based either on functional analysis or extensive homology to a gene of known function. Therefore, the function of 42% of genes is unknown. This is a feature

129

common to all genomes sequenced to date. Twelve percent of the unknown genes have homologs in other organisms. The large number of genes with unknown functions represents a formidable challenge to understanding the metabolism and physiology of B. subtilis. It is not clear why such a large number of genes were refractory to discovery by classical genetic analysis. It is probable that among this group are essential genes, redundant genes, and genes which participate in metabolic and physiological processes not yet discovered. Some of these questions will be resolved during the ongoing joint European– Japanese functional analysis project, the objective of which is to examine the expression of all genes of unknown function in B. subtilis by systematically inactivating each gene and testing the resultant mutant strain for a wide variety of phenotypes. The intermediary metabolic pathways and the metabolic potential of a bacterium can be constructed from knowledge of the complete genome sequence. Analysis of this type shows that both the glycolytic and TCA cycles are complete and functional in B. subtilis, and the enzymes and regulator genes required for anaerobic growth with glucose as carbon source and nitrate as electron acceptor are also present. Anaerobic growth under these conditions has been experimentally verified.

III. GENETIC METHODOLOGY IN B. SUBTILIS The knowledge of the complete genomic sequence has had a profound effect on research on B. subtilis. This is manifest most clearly in the accelerated pace at which research is now done. The information in the complete genomic sequence is enhanced by three additional features: (i) Polymerase chain reaction techniques allow any chromosomal fragment to be rapidly amplified, (ii) the transformation frequency of B. subtilis is high, and (iii) there is a sophisticated range of integrating plasmids and transposons available for use in B. subtilis. Integrating plasmids are the predominant and most versatile tool for genetic manipulation of the B. subtilis chromosome. The essential features of an integrating plasmid are (i) the inability to replicate autonomously in B. subtilis, (ii) the presence of a gene for selecting plasmid establishment in B. subtilis, and (iii) a segment of B. subtilis chromosomal DNA through which the plasmid can integrate into the chromosome by homologous recombination. There are basically two types of integration events. When the transforming plasmid is circular, integration occurs by a single

ch09.qxd 10/9/03 6:52 PM Page 130

130

the desk encyclopedia of microbiology

crossover event. When the transforming plasmid is linear, and contains two regions of homology with the chromosome, integration occurs by a double-crossover event that results in gene replacement. Incorporating additional genetic functions into the integrating plasmid can extend the repertoire of genetic manipulation. Such functions include reporter genes to generate transcriptional and translational fusions (e.g., -galactosidase and chloramphenicol acetyl transferase to detect protein accumulation and green fluorescence protein to determine intracellular location), inducible promoters such as Pspac (an IPTG-inducible system based on the lac operon of Escherichia coli), and site-specific recombination functions. The details of these systems and the mechanisms through which specific genetic manipulations can be achieved using integrating plasmids are beyond the scope of this article. However, it is useful to illustrate the range of genetic analysis that can be performed using integrating plasmids. Any gene can be mutated through either insertional inactivation or deletion. Complementation analysis and the dominance or recessivity of specific mutations can be tested. Genetic loci can be inserted into heterologous sites to test whether they function in cis or in trans. The phenotype caused by overproduction of a gene product can be assessed by gene amplification. Similarly, amplification of a control region can be used to test for titration of repressors. Large chromosomal fragments can be deleted or inverted. Strains can be constructed with multiple deletions in non-contiguous chromosomal regions. Genes and/or their control regions can be mutated in vitro and reinserted into homologous or heterologous sites of the chromosome in single or multiple copy. The expression profile of a gene/operon can be established by generating transcriptional and translational fusions to reporter genes. Regulation at the transcriptional and posttranscriptional levels can be distinguished. Conditional expression of any gene can be effected by placing it under the control of an inducible promoter. This is particularly useful for analysis of essential genes.

IV. GENETIC ANALYSIS OF B. SUBTILIS An objective of bacterial research is to understand how individual processes are regulated and integrated within the cell. Two themes have emerged from the study of how post-exponential phenomena, such as competence development, enzyme production, and sporulation, are regulated: (i) Multiple signals detected by the cell are integrated by a signal

transduction cascade which converges on a central regulator and (ii) the regulation of these processes overlaps so that entering one of these physiological states precludes activation of the other states.

A. Two-component signal transduction systems It is imperative that bacteria adapt their gene expression and metabolism to the prevailing conditions. Two-component systems comprise a family of proteins, found ubiquitously in bacteria, which sense environmental and nutritional conditions and effect appropriate metabolic and physiological responses. They are generally (but not always) composed of two proteins: a sensor kinase and a response regulator. The kinase detects a parameter(s) of the environment that results in enzyme activation. The active kinase autophosphorylates and then transfers the phosphate to the response regulator. Phosphorylation of the response regulator activates (or alters) its transcriptional activity. Thirty-four two-component systems have been identified in B. subtilis, suggesting great versatility and flexibility in its response to changing environmental and nutritional conditions. Three such systems, ComP–ComA, DegS–DegU, and the unusual phosphorelay KinABC–Spo0F–Spo0B–Spo0A, are involved in regulating the post-exponential phase phenomena of competence development, enzyme synthesis, and sporulation, respectively, in B. subtilis.

B. Competence development The regulation of competence development can be divided into three stages: (i) sensing the environmental and nutritional conditions which trigger the process, (ii) the signal transduction pathway that integrates the signals, and (iii) activation of the transcription factor ComK. The composition of the growth medium is an important parameter in competence development. Cells do not become competent in rich medium. In defined medium supplemented with amino acids, competence develops when cells enter the stationary phase of the growth cycle. In defined glucose-minimal medium, cells become competent during exponential growth. Cell density is a second parameter to which competence development responds. This signal is mediated by peptide factors that accumulate in the medium as cells grow to high density. Two such peptides have been identified. Competence stimulating factor (CSF) is a small peptide that is secreted from the cell after signal sequence cleavage. The secreted peptide is further proteolytically processed and a pentapeptide is reimported into

ch09.qxd 10/9/03 6:52 PM Page 131

bacillus subtilis, genetics the cell through the oligopeptide transport system. The second peptide, ComX, is secreted from the cell by an unknown mechanism. Accumulation of both these peptides causes an increase in the level of phosphorylated ComA (ComA~P). ComX does this by stimulating the ComP kinase that specifically phosphorylates ComA, whereas CSF is thought to inhibit the activity of a phosphatase which dephosphorylates ComA~P. Phosphorylated ComA then activates expression of srf, the surfactin synthetase operon, leading to increased levels of ComS, the next regulator in the signal transduction cascade. ComS is encoded by a small gene (46 codons) located entirely within the much larger srfA gene. The reason for this unusual gene organization is not known, but it provides a link between the postexponential growth phase phenomena of competence development and antibiotic production. ComS destabilizes a ternary protein complex composed of MecA, ClpC, and ComK leading to release of free ComK, which can then function as a transcription factor. ComK also activates its own expression leading to very high levels of the protein, thereby further committing cells to the competent state. The ComK regulon comprises the group of genes and operons encoding the proteins required for binding, fragmentation, and uptake of DNA. The comF operon encodes a helicase that is involved in unwinding transforming DNA. The comG operon encodes proteins homologous to (i) the pilin protein and proteins involved in pilin assembly, (ii) proteins involved in pullulanase secretion in Klebsiella pneumoniae, and (iii) proteins encoded by the virB operon of Agrobacterium tumefaciens which function to transfer T-DNA from the bacterium to the plant. It is interesting that the transfer of DNA into B. subtilis cells shares features with other systems designed to transfer both DNA and proteins across cell walls and membranes.

C. Regulation of enzyme production Production of extracellular enzymes occurs in response to nutrient limitation, and accumulation is observed when cells enter the stationary phase of the growth cycle. This is approximately the same growth period during which the cells become competent (see Sections I,B and IV,B). Although the regulatory pathways of these two physiological states overlap, it appears that enzyme production and competence development are alternate physiological states. The signals which trigger enzyme production (the nature of these signals is not precisely known) are sensed by the DegS kinase. Activation of the kinase leads to accumulation of phosphorylated DegU (DegU~P). DegU~P is a transcriptional activator which stimulates transcription of

131

genes encoding the amylases, proteases, and glucanases produced when cells enter the stationary phase of the growth cycle. Two additional regulators, DegQ and DegR, are also required for enzyme production. DegU~P has an additional role in that it inhibits production of ComS, the regulator required for activation of the competence transcription factor ComK. Therefore, accumulation of Deg~P leads to stimulation of enzyme production and inhibition of competence development. In contrast, the nonphosphorylated form of DegU stimulates competence development. Phosphorylation of DegU therefore acts as a switch mechanism allowing cells to become competent (high levels of DegU) or to produce extracellular enzymes (high levels of DegU~P). The equilibrium between the phosphorylated and nonphosphorylated states will depend on the extent to which the kinase (which is responsive to nutritional and environmental conditions) is activated.

D. Regulation of sporulation 1. Initiation of sporulation The conditions that trigger sporulation include limitation of carbon, nitrogen, and phosphorous and high cell density. These signals, and perhaps others, are sensed and integrated by a signal transduction pathway which converges on the transcriptional regulator Spo0A. The critical parameter in the decision to sporulate is the level of phosphorylated Spo0A, the form of the protein required for transcriptional activation. The nonphosphorylated form of Spo0A has no known transcriptional activity, whereas high Spo0A~P levels are required for initiation of sporulation. The commitment to sporulate is reinforced by a positive autoregulatory loop whereby Spo0A~P activates transcription of the spo0A gene. Spo0A is unusual among two-component transcriptional activators in that it is phosphorylated indirectly by a so-called phosphorelay (Fig. 9.2). Spo0F is phosphorylated by sensor kinases in response to nutritional and environmental conditions. The phosphate is then transferred from Spo0F to Spo0A via the Spo0B phosphotransferase. The relative cellular levels of Spo0A and Spo0A~P are the result of competing kinase and phosphatase activities. There are at least three kinases which phosphorylate Spo0F that lead to a buildup of Spo0A~P in the cell. The precise nature of the nutritional and/or environmental signals that activate the kinases is not firmly established. There are also four phosphatases that function to lower the cellular level of Spo0A~P: RapA, RapB, and RapE specifically dephosphorylate Spo0F~P, whereas the Spo0E phosphatase

ch09.qxd 10/9/03 6:52 PM Page 132

132

the desk encyclopedia of microbiology 2. Regulation of endospore development a. Activation of SigmaF in the forespore

FIGURE 9.2 The phosphorelay leading to formation of Spo0A~P. A variety of environmental and nutritional conditions (stippled arrows) are sensed by sensor kinases leading to their autophosphorylation. The phosphate group is then transferred from the kinase to Spo0F (to give Spo0F~P). It is subsequently transferred from Spo0F~P to Spo0A (to give Spo0A~P) by the phosphotransferase Spo0B. Phosphate groups can be drained from the phosphorelay at two points: (i) by dephosphorylation of Spo0F~P by any of three response regulator aspartate phosphatases (Rap A/B/E) and (ii) by dephosphorylation of Spo0A~P by Spo0E phosphatase. The phosphatase activities are also responsive to a distinct group of environmental and nutritional conditions (stippled arrows). The competing actions of the kinase and phosphatase activities determine the relative cellular levels of Spo0A and Spo0A~P.

specifically dephosphorylates Spo0A~P. It appears that cell density signals can be detected through these phosphatases by a quorum-sensing mechanism. There is a gene encoding a small peptide juxtaposed to the RapA and RapE phosphatase genes called phrA and phrE, respectively. The PhrA and PhrE peptides are secreted from the medium, processed, and reimported into the cell. This results in inhibition of Spo0F~P dephosphorylation by RapA and RapE and leads to an increase in the cellular level of Spo0A~P. The genetic evidence indicates that those conditions which favor competence development signal an inhibition of sporulation. For example, the high levels of Com~P that direct competence development also lead to increased levels of RapA, which results in dephosphorylation of Spo0F. This leads to a decrease in cellular levels of Spo0A~P, thereby inhibiting sporulation. The AbrB regulator also provides a link between competence development, enzyme and antibiotic production, and sporulation. The level of AbrB varies throughout the growth cycle to ensure that cells can become competent, produce enzymes, or sporulate but cannot enter all three physiological states at the same time.

Asymmetric septum formation in the sporangium is one of the first morphological events of endospore development (Fig. 9.1). At this stage (stage II) there are two complete chromosomes in the sporangium, each having been directed into one of the two compartments by a chromosome partitioning mechanism. When septum formation is complete, the fates of the two cells differ. The smaller compartment becomes the spore and the larger becomes the mother cell. Therefore, it is necessary to establish a separate program of gene expression in each compartment. The first step in this process is activation of expression of the operon encoding the transcription factor SigmaF by high levels of Spo0A~P. This operon is expressed before completion of septum formation and the SigmaF protein is therefore present in both compartments. However, it becomes active only in the forespore compartment. Three additional proteins, SpoIIAA, SpoIIAB, and SpoIIE, effect asymmetric activation of SigmaF. SpoIIAB is an anti-sigma factor that can bind either to SigmaF (making SigmaF inactive) or to SpoIIAA (allowing SigmaF to be transcriptionally active). The phosphorylation state of SpoIIAA determines whether SpoIIAB binds to SigmaF or to SpoIIAA. When SpoIIAA is phosphorylated, SpoIIAB binds to SigmaF preventing it from engaging in transcription; when SpoIIAA is not phosphorylated, it binds to SpoIIAB and SigmaF can now engage in transcription. The dephosphorylation of SpoIIAA is effected by SpoIIE, a phosphatase that is located in the asymmetric septum and dephosphorylates SpoIIAA only in the forespore (Fig. 9.3). This is a very clear example of morphological differentiation coupled with regulation of gene expression. b. Activation of SigmaE in the mother cell A cell-type-specific pattern of gene expression, mediated by the SigmaE transcription factor, is established in the mother cell after SigmaF has been activated in the forespore (Fig. 9.1). SigmaE protein is also synthesized in the predivisional sporangium and is therefore present in both the forespore and the mother cell compartments. However, it is activated only in the mother cell. SigmaE is activated by cleavage of a small peptide from the amino terminus of the protein. The proteolytic cleavage is effected by SpoIIGA, a membrane-localized protease (Fig. 9.3). Genetic analysis has revealed that SigmaF must be activated in the forespore before SigmaE can be activated in the mother cell. The basis of this requirement is that

ch09.qxd 10/9/03 6:52 PM Page 133

133

bacillus subtilis, genetics

c. Activation of SigmaG and SigmaK in the forespore and mother cell, respectively Separate programs of gene expression are first established in the two compartments by activation of SigmaE and SigmaF. These sigma factors are then replaced by two new compartment-specific sigma factors, SigmaG in the forespore and SigmaK in the mother cell. Production of the SigmaK protein is directed by the mother cell-specific SigmaE, whereas production of SigmaG is directed by the foresporespecific SigmaF. However, activation of SigmaG and SigmaK requires a signal from the other compartment (Fig. 9.3). Activation of SigmaG in the forespore requires gene products encoded by the spoIIIA operon which is transcribed by SigmaE in the mother cell. Activation of SigmaK in the mother cell is similar to activation of SigmaE. SigmaK must be proteolytically processed to become active. The protease is produced in the mother cell. However, the forespore produces a product (under SigmaG control) which is secreted into the space between the forespore and mother cell that is required for activation of the protease in the mother cell. d. Features of the regulation of endospore formation FIGURE 9.3. Crisscross regulation of compartmental gene expression during sporulation. Transcriptional dependency is indicated by thin arrows: transcription of both the sigmaE- and sigmaF- encoding genes requires SigmaA, SigmaH, and Spo0A~P. The formation of the septum (shaded rectangle) between the forespore and mother cell compartments is also dependent on these three transcription factors. SigmaF is required for transcription of the sigmaG-encoding gene in the forespore and SigmaG positively autoregulates its own expression (arrowed circle). Likewise, SigmaE is required for transcription of the sigmaK-encoding gene, which also positively regulates its own expression. A second level of control operates at the level of the activities of these factors (thick arrows). The activity of SigmaF in the forespore is dependent on septum formation and the septum-linked SpoIIE protein. The activity of SigmaE is dependent on the activity of SigmaF through the septumlinked SpoIIGA and SpoIIR proteins. The activity of SigmaG is dependent on the activities of SigmaE in the mother cell and the activity of SpoIIIA, whereas the activity of SigmaK is dependent on the activity of SigmaG in the forespore and the activities of SpoIVF and SpoIVB [from Stragier and Losick (1996) with permission, from the Annual Review of Genetics, Volume 30, © 1996, by Annual Reviews].

SigmaF is required to produce SpoIIR in the forespore (Fig. 9.3). SpoIIR is then secreted from the forespore into the intercompartmental space between forespore and mother cell where it binds to, and activates, the membrane-localized protease SpoIIGA. This protease then activates SigmaE, which effects the mother cell-specific program of gene expression.

The establishment of temporal and cell-type-specific programs of gene expression in the forespore and mother cell compartments of the sporangium displays many interesting features. Activation of both SigmaE and SigmaF is coupled to the morphological event of asymmetric septum formation by locating SpoIIE and SpoIIGA in the septum. Temporal regulation of gene expression is effected by sequential activation of sigma factors. The timing of sigma factor activation and the coordination of gene expression in the forespore and mother-cell compartments are controlled by so-called crisscross regulation (Fig. 9.3). Activation of SigmaF in the forespore is required before SigmaE can be activated in the mother cell; SigmaE must be activated in the mother cell before SigmaG can be activated in the forespore, and SigmaG must be activated in the forespore before SigmaK can be activated in the mother cell (Fig. 9.3). Both SigmaE and SigmaK are activated in the mother cell by two similar (but not identical) signal transduction systems. In both cases, a signal is produced and secreted from the forespore to effect sigma factor activation in the mother cell.

V. CONCLUSION Bacillus subtilis is a very useful model organism for bacterial research. The complete genome sequence is

ch09.qxd 10/9/03 6:52 PM Page 134

134

the desk encyclopedia of microbiology

known, it is amenable to genetic manipulation, and it exhibits many fundamental biological processes. Whole bacterial genomes can be sequenced with relative ease. However, only a small number of the bacteria are amenable to genetic manipulation. Therefore, the metabolic and physiological capabilities of these bacteria will have to be deduced from knowledge of their gene content coupled with research performed in model organisms such as B. subtilis. The large number of genes to which we cannot assign a function suggests that there is still much to be discovered in B. subtilis. It is likely, therefore, that B. subtilis will remain a primary focus of bacterial research.

BIBLIOGRAPHY Fabret, C., Feher, V. A., and Hoch, J. A. (1999). Two-component signal transduction in Bacillus subtilis: how one organism sees its world. J. Bacteriol. 181(7), 1975–1983. Fisher, S. H. (1999). Regulation of nitrogen metabolism in Bacillus subtilis: vive la difference! Mol. Microbiol. 32(2), 223–232. Harwood, C., and Cutting, S. M. (Eds.) (1990). “Molecular Biological Methods for Bacillus.” Wiley, Chichester, UK.

Kunst, F., et al. (1997). The complete genome sequence of Bacillus subtilis. Nature 390, 249–256. Sonenshein, A. L. (2000). Control of sporulation initiation in Bacillus subtilis. Curr. Opin. Microbiol. Sonenshein, A. L., Hoch, J. A., and Losick, R. (Eds.) (2001). “Bacillus subtilis and Its Closest Relatives: From Genes to Cells.” ASM Press. Stragier, P., and Losick, R. (1996). Sporulation in Bacillus subtilis. Annu. Rev. Genet. 30, 297–341.

WEBSITES SubtiList World-Wide Web Server http://genolist.pasteur.fr/SubtiList/ The Non-redundant Bacillus subtilis database http://pbil.univ-lyon1.fr/nrsub/nrsub.html Bacillus subtilis Japan Functional Analysis Network http://bacillus.genome.ad.jp/ Bacillus subtilis Genetics at the University of London http://web.rhul.ac.uk/Biological-Sciences/cutting/index.html Micado (formerly Mad Base). A relational database on B. subtilis genetics (V. Biaudet, F. Samson & Ph. Bessieres) http://locus.jouy.inra.fr/cgi-bin/genmic/madbase_home.pl Website for Comprehensive Microbial Resource of the Institute for Genomic Research and links to many other microbial genomic sites http://www.tigr.org/tigr-scripts/CMR2/CMRHomePage.spl.

ch10.qxd 10/9/03 6:52 PM Page 135

10 Bacteriophages Hans-Wolfgang Ackermann Laval University

groups. Phages are tailed, cubic, filamentous, or pleomorphic and contain single-stranded or double-stranded DNA or RNA. They are classified into 13 families. Tailed phages are far more numerous than other types, are enormously diversified, and seem to be the oldest of all phage groups.

GLOSSARY bacteriophage Virus that replicates in a bacterium; literally “eater of bacteria”. capsid Protein coat surrounding the nucleic acid of a virus. envelope Lipoprotein membrane surrounding a virus capsid. genome Complete set of genes in a virus or a cell; in viruses, it consists of either DNA or RNA. host range Number and nature of organisms in which a virus or group of viruses replicate. integrase Viral enzyme mediating the integration of viral DNA into host DNA. prokaryote Type of cell whose DNA is not enclosed in a membrane. restriction endonuclease Enzyme that recognizes a specific base sequence in double-stranded DNA and cuts the DNA strand at this site. superinfection Infection of a virus-infected host by a second virus. virion Complete infectious virus particle.

Bacteriophages occur in over 140 bacterial genera and many different habitats. Infection results in phage multiplication or the establishment of lysogenic or carrier states. Bacterial genes may be transmitted in the process. Some phages (e.g., T4, T7, , MS2, fd, and X174) are famous experimental models. Phage research has led to major advances in virology, genetics, and molecular biology (concepts of lysogeny, provirus, induction, transduction, eclipse; DNA and RNA as carriers of genetic information; discovery of restriction endonucleases). Phages are used in phage typing and genetic engineering, but the high hopes set on phage therapy have generally been disappointed. In destroying valuable bacterial cultures, some phages are nuisances in the fermentation industry.

I. ISOLATION AND IDENTIFICATION OF PHAGES

Bacteriophages, or “phages”, are viruses of prokaryotes including eubacteria and archaebacteria. They were discovered and described twice, first in 1915 by the British pathologist Frederick William Twort and then in 1917 by the Canadian bacteriologist Félix Hubert d’Herelle working at the Pasteur Institute of Paris. With about 5150 isolates of known morphology, phages constitute the largest of all virus The Desk Encyclopedia of Microbiology ISBN: 0-12-621361-5

A. Propagation and maintenance 1. Propagation On solid media, phages produce clear, lysed areas in bacterial lawns or, if sufficiently diluted, small holes

135

Copyright © 2003 Elsevier Ltd All rights of reproduction in any form reserved

ch10.qxd 10/9/03 6:52 PM Page 136

136

the desk encyclopedia of microbiology

called “plaques,” each of them corresponding to a single viable phage. In liquid media, phages sometimes cause complete clearing of bacterial cultures. Phages are grown on young bacteria in their logarithmic phase of growth, usually in conditions that are optimal for their host. Some phases require divalent cations (Ca2, Mg2) or other cofactors. Phages are propagated by three types of techniques: (i) in liquid media inoculated with host bacteria; (ii) on agar surfaces with a monolayer of bacteria; and (iii) in agar double layers consisting of normal bottom agar covered with a mixture of soft agar (0.3–0.9%), phages, and bacteria. Phages are harvested after a suitable incubation time, generally 3 h for liquid cultures and 18 h for solid media. Phages from agar cultures are extracted with buffer or nutrient broth. Phage suspensions, or lysates, are sterilized, best by filtration through membrane filters (0.45 m pore size) and then titrated. Sterilization by chloroform or other chemicals is of questionable value. 2. Storage No single technique is suitable for all phages. Many phages can be kept as lystes at 4C or in lyophile, but others are quickly inactivated under these conditions. Lystes should be kept without additives such as thymol or chloroform. The best procedures seems to be preservation at 70C in 50% glycerol. Phages may also be preserved in liquid nitrogen, by drying on filter paper, and, in the case of endospore-forming bacteria, by trapping phage genomes in spores. Ideally, any phage should be preserved by several techniques.

B. Isolation of phages 1. Isolation from nature All samples must be liquid. Soil and other solid material are homogenized and suspended in an appropriate medium. Solids and bacteria are removed, usually by filtration preceded or not by centrifugation. Very rich samples can be assayed directly on indicator bacteria. In most cases, phages must be enriched by incubating the sample in a liquid medium inoculated with indicator bacteria. The culture is then filtered and titrated and phages are purified by repeated cloning of single plaques. Large samples must be concentrated before enrichment. This is done by centrifugation, filter adsorption and elution, flocculation, or precipitation by polyethylene glycol 6000. Adsorption–elution techniques may involve strongly acidic or alkaline conditions that inactivate phages. 2. Isolation from lysogenic bacteria Many bacteria produce phages spontaneously. These phages may be detected by testing culture filtrates on

indicator bacteria. It is generally preferable to induce phage production by mitomycin C, ultraviolet (UV) light, or other agents. A suspension of growing bacteria is exposed to the agent (e.g., 1 g/ml of mitomycin C for 10 min or UV light for 1 min), incubated again, and then filtered. After mitomycin C induction, the bacteria should be separated from the agent by centrifugation and transferred into a fresh medium. Bacteriocins (see Section II.C), which are a source of error, are easily identified because they cannot be propagated and do not produce plaques when diluted.

C. Concentration and purification Small samples of 100 ml are usually concentrated by ultracentrifugation (about 60 000g in swinging-bucket rotors), followed by several washes in buffer. Fixedangle rotors allow considerable reduction of the g force because large phages sediment at as little as 10 000g for 1 h. Further purification may be achieved by centrifugation in a CsCl or sucrose density gradient. Large samples raise problems of contamination, aeration, and foaming. Preparation schedules are often complex: (a) pretreatment by low-speed centrifugation and/or filtration; (b) concentration, mostly by precipitation with polyethylene glycol; and (c) final purification in a density gradient or by ultracentrifugation.

D. Identification Phase identification relies greatly on the observation that most phages are specific for their host genus; however, enterobacteria, in which polyvalent phages are common, are considered in this context as a single “genus.” Phages are first examined in the electron microscope. This usually provides the family diagnosis and often indicates relationships on the species level. If no phages are known for a given host genus or only phages of different morphology, the new isolate may be considered as a new phage. If the same host genus has phages of identical morphology, they must be compared to the isolate by DNA–DNA hybridization and/or serology. Further identification may be achieved by determining restriction endonuclease cleavage patterns or constitutive proteins.

II. PHAGE TAXONOMY A. General D’Herelle thought that there was only one phage with many races, the Bacteriophagum intestinale. Early attempts at classification by serology, host range, and inactivation tests showed that phages were highly

ch10.qxd 10/9/03 6:52 PM Page 137

137

bacteriophages diversified, but these attempts proved premature. Modern taxonomy started in 1962 when a system of viruses based on the properties of the virion and its nucleic acid was introduced by Lwoff, Horne, and Tournier. In 1967, phages were grouped into six basic types on the basis of morphology and nature of nucleic acid. Other types were later established, and this process is likely to continue if more archaebacteria and other “unusual” microbes are investigated for the presence of phages. The International Committee on Taxonomy of Viruses presently recognizes one order, 13 families, and 31 genera in phages. Their morphology is illustrated in Fig. 10.1 and their basic characteristics and hosts are listed in Tables 10.1–10.5. The most important family criteria are type of nucleic acid, particle shape, and presence or absence of an envelope. As in other viruses, family names end in -viridae and genus names in -virus. Species are designated by the vernacular names of their best-known (or only) members (e.g., T4 or ).

B. Phage families and genera 1. Tailed phages With approximately 4950 observations, tailed phages comprise 96% of phages and are the largest virus group known. They contain a single molecule of dsDNA and are characterized by a tubular protein

FIGURE 10.1 Morphology of phage families. C, Corticoviridae; Cy, Cystoviridae; F, Fuselloviridae; Ii, Inoviridae, Inovirus genus; Ip, Inoviridae, Plectrovirus genus; L, Leviviridae; Li, Lipothrixviridae; M, Myoviridae; Mi, Microviridae; P, Podoviridae; Pl, Plasmaviridae; R, Rudiviridae; S, Siphoviridae; T, Tectiviridae. [Modified from Ackermann, H.-W. (1987). Microbiol. Sci. 4, 241–218. With permission of Blackwell Scientific Publications Ltd., Oxford, England]

TABLE 10.1 Main properties and frequency of phage familiesa No. of Membersb

Shape

Nucleic acid

Family

Genera

Particulars

Example

Tailed

DNA, ds, L

Myoviridae Siphoviridae Podoviridae

6, see text 6, see text 3, see text

Tail contractile Tail long, noncontractile Tail short

T4 T7

Cubic

DNA, ss, C

Microviridae

Conspicuous capsomers

X174

40

DNA, ds, C, S DNA, ds, L RNA, ss, L

Corticoviridae Tectiviridae Leviviridae

Complex capsid, lipids Double capsid, pseudo-tail, lipids

PM2 PRD1 MS2

3? 18 39

RNA, ds, L, M

Cystoviridae

Microvirus Bdellomicrovirus Chlamydiomicrovirus Spiromicrovirus Corticovirus Tectivirus Levivirus Allolevirirus Cystovirus

Envelope, lipids

6

DNA, ss, C

Inoviridae

DNA, ds, L DNA, ds, L

Inovirus Plectrovirus Lipothrixviridae Lipothrixvirus Rudiviridae Rudivirus

Long filaments Short rods Envelope, lipids Stiff rods, no envelope, no lipids

fd L51 TTV1 SIRV1

DNA, ds, C, S DNA, ds, C, S

Plasmaviridae Fuselloviridae

Envelope, no capsid, lipids Lemon-shaped, envelope, lipids

MVL2 SSV1

Filamentous

Pleomorphic

Plasmavirus Fusellovirus

1143 3011 698

1 57 6? 2 6? 8?

a Modified from Ackermann (1987) with permission of Blackwell Scientific Publications Ltd. C, circular; L, linear; M, multipartite; S, supercoiled; ss, single-stranded; ds, double-stranded. b Exluding phage-like bacteriocins and known defective phages. Computed October 31, 2000.

ch10.qxd 10/9/03 6:52 PM Page 138

138

the desk encyclopedia of microbiology TABLE 10.2 Dimensions and physicochemical propertiesa Virion

Nucleic acid

Particle size (nm)

Phage group or family Tailed phages Average Range Microviridae Corticoviridae Tectiviridae Leviviridae Cystoviridae Inoviridae Inovirus Plectrovirus Lipothrixviridae Rudiviridae Plasmaviridae Fuselloviridae

Tail length (nm)

Weight (MDa)

Buoyant density

Lipids (%)

Content (%)

63b 38–160b 27 60 63 23 75–80

153 3–825 — — — — —

100 29–470 7 49 70 4 99

1.49 1.4–1.54 1.39 1.28 1.29 1.46 1.27

— — — 13 15

760–1950 7 85–250 15 400–2400 38 780–950 20–40 80 85 60

— — — — — —

12–34

1.30 1.37 1.25 1.36

— — 22 — 11 10

33

1.24

20

Size (kbp or kb)

GC (%)

46 30–62 26 14 14 30 10

79 17–745 4.4–6.1 9.0 15.2 3.5–4.3 13.4

48 27–72 44 43 51 51 56

6?–21

5.8–7.3 4.4–8.3 16–42 33–36 11.7 15.5

40–60

3

32

Modified from Ackermann (1987) with permission of Blackwell Scientific Publications Ltd. Buoyant density is g/ml in Cscl; G C, guanine–cytosine content; —, absent. b Isometric heads only. a

TABLE 10.3 Comparative biological properties Infection

Adsorption

Assembly

Shape

Phage group

Nature

By

By

To

Tailed

Caudovirales

V or T

DNA

Tail end

Isometric

Microviridaea Corticoviridae Tectiviridae Leviviridae Cystoviridae

V V V V V

DNA DNA DNA RNA Capsid

Filamentous

Inoviridae Inovirus Plectrovirus Lipothrixviridae Rudiviridae

S or T S V or T S

Virion Virion?

Plasmaviridae Fuselloviridae

T T

DNA?

Pleomorphic

Virion

Site

Start

Release

Cell wall, pili, Nucleoplasm, capsule, flagella cell periphery

Capsid

Lysis

Spikes Spikes Pseudo-tail A protein Envelope

Cell wall Pili Pili, cell wall Pili Pili

Nucleoplasm PM Nuceloplasm Cytoplasm Nuceloplasm

Capsid Capsid Capsid RNA Capsid

Lysis Lysis Lysis Lysis Lysis

Virus tip Virus tip Virus tip Virus tip

Pili PM Pili Pili

PM PM

DNA DNA?

Extrusion Extrusion Lysis

Envelope Spikes

PM

PM

DNA

Budding Extrusion

a Data are for Microvirus genus only. Abbreviations used: PM, plasma membrane; S, steady state; T, temperate; V, virulent.

tail, a specialized structure for the transfer of phage DNA into host bacteria. Tailed phages have recently been given order rank and the name Caudovirales. They fall into three families: 1. Myoviridae: phages with long complex tails consisting of a core and a contractile sheath (25% of tailed phages, six genera named after phages T4, P1, P2, Mu, SPO1, and H); 2. Siphoviridae: phages with long noncontractile, more or less flexible tails (61%, six genera named after phages , T1, T5, L1, c2, M); and

3. Podoviridae: phages with short tails (15%, three genera named after T7, P22, and 29). Classification of tailed phages into genera is still in its infancy. Phage capsids, usually named heads, are icosahedra or derivatives thereof. Capsomers are rarely visible. Elongated heads are relatively rare but occur in all three families. Heads and tails vary enormously in size and may have facultative structures such as head or tail fibers, collars, base plates, or terminal spikes (Fig. 10.2). The DNA is coiled inside the head. Its composition generally reflects that of the

ch10.qxd 10/9/03 6:52 PM Page 139

139

bacteriophages TABLE 10.4 Occurrence and frequency of tailed and cubic, filamentous, and pleomorphic (CFP) phagesa

Volume and section

Bacterial group according to Bergey’s Manual

Phages

(Holt, 1990)

Tailed

CFP

Total

I 1 2 4 5 6 7 8 9 10 11

Spirochetes Spirilla and vibrioids Gram-negative aerobic rods and cocci Gram-negative facultatively anerobic rods and cocci Gram-negative anaerobic rods Gram-negative sulfate and sulfur reducers Gram-negative anaerobic cocci Rickettsias and chlamydias Mycoplasmas Endosymbionts

11 40 856 1080 30 2 4 2 17 2

II 12 13 14 15 16 17, 26

Gram-positive cocci Endospore formers Gram-positive nonsporing regular rods Gram-positive nonsporing pleomorphic rods Mycobacteria Nocardioforms

1217 625 286 196 78 97

III 18 19 20 21 22 23 24 25

Anoxygenic phototrophs Cyanobacteria Chemolithotrophs Budding and/or appendaged bacteria Sheathed bacteria Nonfruiting gliding bacteria Myxobacteria Archaebacteria

IV 28 29 30 31 32 33 Total

Actinoplanetes Streptomycetes Maduromycetes Thermomonosporae Thermoactinomycetes Other actinomycete genera

12 44 2 112 1 32 16 14 5 131 3 27 4 6 4950

9 22 93

2 21?

10 1

11 49 878 1173 30 2 4 4 38 2 1217 635 286 197 78 97

18

12 44 2 120 1 34 16 32

186

5 131 3 27 4 6 5139

8 2

a Excluding phage-like baceriocins and known defective phages; computed October 31, 2000. Based on a detailed computation published in 2001 (Ackermann, 2001).

host bacterium, but it may contain unusual bases such as 5-hydroxymethylcytosine. Genetic maps are complex and include 271 genes in phage T4 (possibly more in larger phages). Genes for related functions cluster together. Up to 40 structural proteins have been found in some phages (T4). Lipids are generally absent, but have been reported in a few exceptional cases. Response to inactivating agents is variable and no generalization is possible here. Despite the absence of lipids, about one-third of tailed phages are chloroform-sensitive, making chloroform use in phage isolation a dangerous procedure. Most properties of tailed phages appear as individual or species characteristics. Accordingly, genera have not yet been established, but about 250 species are currently recognizable,

mostly on the basis of morphology, DNA–DNA hybridization, and serology.

2. Cubic, filamentous, and pleomorphic phages This group includes 10 small phage families that correspond to approximately 4% of phages, differ greatly in nucleic acid nature and particle structure, and sometimes have a single member. Host ranges are mostly narrow (Table 10.4). Capsids with cubic symmetry are, with one exception, icosahedra or related bodies. Filamentous phages have, according to present knowledge, helical symmetry. Particles may or may not be enveloped. As in other viruses, the

ch10.qxd 10/9/03 6:52 PM Page 140

140

the desk encyclopedia of microbiology TABLE 10.5 Host range

Bacterial division Eubacteria

Phage group

Bacterial group or genus

Caudovirales Microviridae

Any Enterobacteria, Bdellovibrio, Chlamydia, Spiroplasma Alteromonas a. Enterics, Acinetobacter, Pseudomonas, Thermus, Vibrio b. Bacillus, Alicyclobacillus Enterics, Acinetobacter, Caulobacter, Pseudomonas Pseudomonas

Corticoviridae Tectiviridae

Leviviridae Cystoviridae Inoviridae: Inovirus

Archaea

Plectrovirus Plasmaviridae Caudovirales Lipothrixviridae Rudiviridae Fuselloviridae

Enterics, Pseudomonas, Thermus, Vibrio, Xanthomonas Acholeplasma, Spiroplasma Acholeplasma, Spiroplasma Extreme halophiles and methanogens Acidianus, Sulfolobus, Thermoproteus Sulfolobus Acidianus, Sulfolobus (Methanococcus, Pyrococcus?)

capsid consists of two protein shells and a lipid bilayer sandwiched in between. Two similar, littleknown phages were isolated from seawater. iii. Tectiviridae Phages are characterized by a double capsid and a unique mode of infection. The outer capsid, which is rigid and apparently proteinic, surrounds a thick, flexible lipoprotein membrane. Upon adsorption to bacteria or chloroform treatment, this inner coat becomes a tail-like tube of about 60 nm in length, obviously a nucleic acid ejection device. Tectiviruses of bacilli have apical spikes. Despite their small number, tectiviruses are found in widely different bacteria.

b. Cubic RNA phages i. Leviviridae Leviviruses resemble enteroviruses and have no morphological features. Most of them are plasmid-specific coliphages that adsorb to F or sex pili and have been divided, by serology and other criteria, into two genera. Several not yet classified leviviruses are specific for other plasmid types (C, H, M, etc.) or occur outside of the enterobacteria family. ii. Cystoviridae The single officially recognized member of the family Cystoviridae is unique in several ways. It is the only phage to contain dsRNA and RNA polymerase. The RNA is multipartite and consists of three molecules.

c. Filamentous phages

FIGURE 10.2 Schematic representation of phage T4 with extended tail and folded tail fibers (left) and sectioned with contracted tail (right). [Modified from Ackermann, H.-W. (1985). Les virus des bactéries. In “Virlogie médicale” (J. Maurin, ed.), p. 200. With permission of Flammarion Médecine-Sciences, Paris.]

presence of lipids is accompanied by low buoyant density and high sensitivity to chloroform and ether. a. Cubic DNA phages i. Microviridae The genus Microvirus includes the phage X174 and related phages of enterobacteria and is characterized by large capsomers. Similar phages occur in so taxonomically distinct bacteria as Bdellovibrio, Chlamydia, and Spiroplasma. ii. Corticoviridae The only certain member of the family Corticoviridae is a maritime phage, PM2. Its

i. Inoviridae The Inoviridae family includes two genera with very different host ranges and similarities in replication and morphogenesis that seem to derive from the single-stranded nature of phage DNA rather than a common origin of these phages. Despite the absence of lipids, viruses are chloroform-sensitive. The Inovirus genus includes 42 phages that are long, rigid, or flexible filaments of variable length. They are restricted to a few related Gram-negative bacteria, sensitive to sonication, and resistant to heat. Many of them are plasmid-specific. The Plectrovirus genus includes 15 isolates. Phages are short, straight rods and occur in mycoplasmas only. ii. Lipothrixviridae This family includes four viruses of the archaebacterial genus Thermoproteus. Particles are characterized by the combination of a lipoprotein envelope and rodlike shape. iii. Rudiviridae This family includes two viruses of different length, isolated from the archaebacteria Acidianus, Sulfolobus, and Thermoproteus. Particles are straight rods without envelopes and closely resemble the tobacco mosaic virus.

ch10.qxd 10/9/03 6:52 PM Page 141

bacteriophages d. Pleomorphic phages i. Plasmaviridae Only one certain member is known, Acholeplasma virus MVL2 or L2. It contains dsDNA, has no capsid, and may be called a nucleprotein condensation with a lipoprotein envelope. Four similar isolates are known, but one them has been described as containing single-stranded DNA and their taxonomic status is uncertain. ii. Fuselloviridae This family has only one certain member, SSV1, which is produced upon induction by the archaebacterium Sulfolobus shibatae. Particles are lemon-shaped with short spikes at one end. The coat consists of two hydrophobic proteins and host lipids and is disrupted by chloroform. SSV1 has not been propagated for absence of a suitable host. It persists in bacterial cells as a plasmid and as an integrated prophage (see Section IV.B.1). Possibly related spindleor droplet-shaped viruses have been found in Acidianus and Sulfolobus.

C. Plasmids, episomes, and bacteriocins Plasmids are extrachromosomal genetic elements that consist of circular or linear dsDNA and replicate independently of the host chromosome. Certain prophages behave as plasmids, but phages and plasmids are sharply differentiated: contrary to plasmids, phages have a coat and genomes of uniform size, occur free in nature, and generally lyse their hosts. The term episome designates both plasmids and prophages that can integrate reversibly into host DNA. Bacteriocins are antibacterial agents that are produced by bacteria, require specific receptors, and kill other bacteria. Highmolecular weight or “particulate” bacteriocins are defective phages (e.g., contractile or non-contractile tails without heads). Low-molecular weight or “true” bacteriocins are a mixed group of entities, including enzymes and phage tail spikes.

D. Origin and evolution of Bacteriophages Phages are probably polyphyletic in nature and originated at different times. This is indicated by seemingly unbridgeable fundamental differences between most phage families and by their host ranges. Phages may have derived from cell constituents that acquired a coat and became independent (e.g., leviviruses from messenger RNA (mRNA) and filamentous inoviruses from plasmids). Tailed phages are obviously phylogenetically related and may be the oldest phage group of all. Their occurrence in eubacteria and archaebacteria suggests that they appeared before their hosts diverged, thus at least 3 billion years ago. Phage

141

groups linked to aerobic bacteria may have emerged at the same time or after the atmosphere became oxygenated by the activity of cyanobacteria. In some cases, nature repeated itself. Convergent evolution is evident in the pseudo-tails of tectiviruses and perhaps in the general resemblance of Inovirus and Plectrovirus phages. Microviruses, tectiviruses, and leviviruses show little or no morphological differentiation, possibly because of constraints imposed by capsid size or of the relatively young geological age of these phages. Inoviruses differentiated by elongation. By contrast, tailed phages are extremely diversified and must have an eventful evolutionary history. In terms of structural simplicity and present-day frequency, the archetypal tailed phage from which the other types evolved is a Siphovirus with an isometric head. The diversification of tailed phages is attributed to point mutation and uniparental reproduction, which are found in all viruses, and two principal factors: modular evolution by exchange of genes or gene blocks, and the frequency of lysogeny (see Section IV.B.1), which perpetuates prophages and makes them available for recombination with superinfecting phages. Other avenues are gene rearrangement (deletions, duplications, inversions, and transpositions) and recombination with plasmids or the host genome. On the other hand, morphological properties may be highly conserved and some phages appear as living fossils, indicating phylogenetic relationships of their hosts.

III. PHAGE OCCURRENCE AND ECOLOGY A. Distribution of phages in bacteria Phages have been found in over 140 bacterial genera distributed all over the bacterial world: in aerobes and anaerobes, actinomycetes, archaebacteria, cyanobacteria and other phototrophs, endospore formers, appendaged, budding, gliding, and sheathed bacteria, spirochetes, mycoplasmas, and chlamydias (Table 10.4). Phage-like particles of the podovirus type have even been found in endosymbionts of paramecia. However, tailed phages reported in cultures of green algae and filamentous fungi are probably contaminants. Most phages have been found in a few bacterial groups: enterobacteria (approximately 900 phages), bacilli, clostridia, lactococci, pseudomonads, staphylococci, and streptococci. This largely reflects the availability and ease of cultivation of these bacteria and the amount of work invested. About half of phages have been found in cultures of lysogenic bacteria. Tailed phages predominate everywhere except

ch10.qxd 10/9/03 6:52 PM Page 142

142

the desk encyclopedia of microbiology

in mycoplasmas. In archaebacteria, they have been found in halobacteria and methanogens, but not yet in extreme thermophiles. Siphoviridae are particularly frequent in actinomycetes, coryneforms, lactococci, and streptococci. Myoviruses and podoviruses are relatively frequent in enterobacteria, pseudomonads, bacilli, and clostridia. There must be phylogenetic reasons for this particular distribution.

B. Phage ecology 1. Habitats Phages have essentially the same habitats as their hosts; indeed, their most important habitat is the lysogenic bacterium because it protects prophages from the environment and frees them from the need to find new bacteria for propagation. In nature, phages occur in an extraordinary variety of habitats ranging from Icelandic solfataras to fish sauce, fetal calf serum, and cooling towers of thermal power stations. They are found on the surfaces and in normal and pathological products of humans and animals, on plants, and in food, soil, air, and water, especially sewage. Body cavities with large bacterial populations, such as intestines and rumen, are extremely rich in phages. According to their habitat, phages may be acido-, alkali-, halo-, psychro-, or thermophilic. These properties are not linked to particular phage groups but, rather, appear as individual adaptations. Psychrophilic phages are often temperature-sensitive and occur frequently in spoiled, refrigerated meat or fish. 2. Geographical distribution Except for phages from extreme environments, phage species generally seem to be distributed throughout the whole earth. This is suggested by: (a) electron microscopical observations of rare and characteristical phage morphotypes in different countries; and (b) worldwide distribution of certain lactococcal phage species in dairy plants and of RNA coliphages in sewage. Unfortunately, most data are from developed countries.

between 0 and 107 per milliliter in domestic sewage. Titers of actinophages in soil vary between 0 and 105 per gram. Purely electron microscopic phage counts, which do not allow phage identification, indicate that total phage titers are between 104 and 107 per milliliter in seawater and may attain 1010 per milliliter in sewage and 109 per milliliter in the rumen. 4. Persistence of phages in the environment Phage survival in nature is frequently studied with the aim of using phages as indicators of contamination. The principal experimental models are cubic RNA phages (MS2, f2) because of their resemblance to enteroviruses, other coliphages (X174, T4), and cyanophages. The indicator value of phages has not been conclusively proven and still lacks a solid statistical basis, but considerable data on phage ecology have been obtained. Phages appear as parts of complex ecosystems including various competing bacteria. Their numbers are affected by factors governing bacterial growth, notably nutrient supply and, in cyanobacteria, sunlight. The lowest bacterial concentration compatible with phage multiplication seems to be 104 cells/ml. In addition, phage counts are affected by association of phages with solids and colloids (e.g., clay), presence of organic matter, concentration and type of ions, pH, temperature, UV and visible light, the type of water (e.g., seawater), and nature and phage sensitivity of bacteria. Finally, phage titers depend on intrinsic phage properties such as burst size (see Section IV.A.4) and host range. No generalization is possible and each phage seems to have its own ecology.

IV. PHAGE PHYSIOLOGY A. The lytic cycle The lytic cycle, also called vegetative or productive, results in the production of new phages. Phages undergoing lytic cycles only are virulent. Lytic cycles consist of several steps and show considerable variation according to the type of phage (Table 10.3).

3. Frequency of phages in nature Sizes of phage populations are difficult to estimate because plaque assays and enrichment and (most) concentration techniques depend on bacterial hosts; they therefore only detect phages for specific bacteria and environmental conditions. Consequently, phage titers vary considerably—for example, for coliphages between 0 and 109 per gram in human feces and

1. Adsorption Phages encounter bacteria by chance and adsorb to specific receptors, generally located on the cell wall, but also on flagella, pili, capsules, or the plasma membrane. Adsorption sometimes consists of a reversible and an irreversible stage and may require cofactors (see Section I.A.1).

ch10.qxd 10/9/03 6:52 PM Page 143

bacteriophages 2. Infection In most phage groups, only the viral nucleic acid enters the host and the shell remains outside. The mechanism of this step is generally poorly understood. In the Inovirus genus and in cystovirus 6, the capsid penetrates the cell wall but not the plasma membrane. In phages with contractile tails, the cell wall is degraded by phage enzymes located on the tail tip. The sheath then contracts (Fig. 10.2) and the tail core is brought in contact with the plasma membrane.

3. Multiplication The interval from infection to the release of new phages is called the latent period. It depends largely on the nature and physiological state of the host and varies between 20 min and 30–50 h. After infection, normal bacterial syntheses are shut off or modified. Phage nucleic acid is transcribed into mRNA using host and/or phage RNA polymerases. The RNA of leviviruses acts as mRNA and needs no transcription. In tailed phages, gene expression is largely sequential. Host syntheses are shut off first and structural genes are expressed last. According to present knowledge, replication of phage DNA and RNA is semiconservative, each strand of a double helix acting as a template for the synthesis of a complementary strand. In phages with single-stranded nucleid acid, doublestranded replicative forms are produced. In tailed phages, replication generally starts at fixed sites of the DNA molecule, is bidirectional, and generates giant DNA molecules, or concatemers, which are then cut to fit into phage heads. Translation is generally poorly know in phages. Microviridae, Leviviridae, the Inovirus genus, and Fuselloviridae have overlapping genes that are translated in different reading frames, allowing the synthesis of different proteins from the same DNA or RNA segment. Lipids, if present at all, are of variable origin. Phospholipids are specified or regulated by phages and fatty acids seem to derive from the host. The assembly of new phages is called maturation. Phage constituents assemble spontaneously or with the help of specific enzymes. In most phage families, the nucleic acid enters a preformed capsid; in others, the capsid is constructed around or co-assembled with the nucleic acid. In tailed phages, assembly is a highly regulated process with sequentially acting proteins and separate pathways for heads and tails, which are finally joined together. The envelope of plasmaviruses is acquired by budding, but that of cystovirus 6 is of cellular origin. The assembly of tailed phages often results in aberrant particles including

143

giant or multi-tailed phages and structures consisting of polymerized head or tail protein, called polyheads, polytails, or polysheaths. Inoviruses produce particles of abnormal length. Leviviruses and some tailed phages produce intracellular crystalline inclusion bodies. 4. Release Phages are liberated by lysis, extrusion, or budding. Lysis occurs in tailed and cubic phages and in the Lipothrixviridae. Bacterial cells are weakened from the inside and burst, liberating some 20–1000 phages (often 50–100). Exceptional burst sizes (up to 20 000) have been recorded in leviviruses. Extrusion, with phages being secreted through the membranes of their surviving hosts, is observed in inoviruses and fuselloviruses. Budding is found in plasmaviruses. Cells are not lysed and produce phages for hours. Progeny sizes for budding and extruded phages have been estimated at 130–1000 per cell.

B. The temperate cycle 1. Lysogeny In phages called temperate, infection results in a special equilibrium between phage and host. The phage genome persists in a latent state in the host cell, replicates more or less in synchrony with it, and may be perpetuated indefinitely in this way. It behaves as a part of the bacterium. If this equilibrium breaks down, either spontaneously or after induction, phages are produced as in a lytic cycle. A bacterium harboring a latent phage genome or prophage is called lysogenic because it has acquired the ability to produce phages. Polylysogenic bacteria may carry up to five different prophages. Defective lysogeny is the perpetuation of temperate phages that are unable to replicate and often consist of single heads or tails. Most temperate phages are tailed, but some members of the Inovirus genus and the Lipothrixviridae, Plasmaviridae, and Fuselloviridae can also lysogenize (Table 10.3). Lysogeny is nearly ubiquitous and occurs in eubacteria, including cyanobacteria, and in archaebacteria. Its frequency in a given bacterial species varies between 0 and 100% (often approximately 40%) according to the species, induction techniques, and number of indicator strains. Mitomycin C and UV light are the principal inducing agents (see Section I.B.2). Many others are known, notably antitumor agents, carcinogens, and mutagens. They often act by damaging host DNA or inhibiting its synthesis. The type of lysogeny is particularly well understood. After infection, the genome of coliphage

ch10.qxd 10/9/03 6:52 PM Page 144

144

the desk encyclopedia of microbiology

forms a circle and some proteins are immediately synthesized. They direct the bacterial cell to make a choice between the lytic and the temperate cycle. If a certain protein prevails, the genome integrates via a cross-over, mediated by an enzyme called “integrase,” at a specific site of the host DNA. It is then replicated at every bacterial division and makes the bacterium immune against superinfection by related phages. Spontaneous or induced excision of the prophage leads to normal phage replication. Some phages have several integration sites. In the P1 type of lysogeny, the phage genome, though able to integrate, usually persists as a plasmid, perhaps in association with the plasma membrane. In the Mu type, the infecting DNA does not form circles and integrates at random at any site of the bacterial genome. The core domain of Mu integrase (transposase) resembles retrovirus integrase.

2. Pseudolysogeny and steady-state infections In pseudolysogenic bacteria, only part of a culture is infected with phages and an equilibrium exists between free phages and noninfected, phage-sensitive bacteria. Phage-free strains can be obtained by simple cloning or by cultivating the bacteria in antiphage serum. In steady-state infections, the whole culture is infected, but cells are not lysed and produce phages continuously (see Section IV.A.4).

3. Transduction and conversion Transduction is transfer of host DNA by viruses and is normally a rare event. In generalized transduction, fragments of bacterial DNA are packaged by accident into phage heads and transferred to a new bacterium. Any host gene may be transferred and the implicated phages may be virulent or temperate. Specialized transduction is carried out by temperate phages that can integrate into host DNA (e.g., ). If the phage DNA is not properly excised, bacterial genes adjacent to the prophage site may be packaged into phage heads along with normal genes. The resulting particle has a defective genome and may be nonviable. In conversion, bacteria acquire new properties through lysogenization by normal temperate phages. Conversion is a frequent event, affecting the whole bacterial population that has been lysogenized. The new properties are specified by phage genes and include new antigens, antibiotic resistance, colony characteristics, or toxin production (e.g., of diphtheria or botulinus toxin). They will disappear if the bacterium loses its

prophage. Transduction and conversion are common in tailed phages; conversion to cholera toxin production has recently been found in the Inovirus genus.

V. PHAGES IN APPLIED MICROBIOLOGY A. Therapeutic agents, reagents, and tools 1. Therapy and prophylaxis of infectious diseases Phage therapy started with high hopes and was strongly advocated by d’Herelle. Phages were, enthusiastically and uncritically, applied in many human and animal diseases and spectacular results were reported as well as failures. When antibiotics became available, phage therapy was practically abandoned. The main reasons were host specificity of phages and rapid appearance of resistant bacteria. However, unbeknownst to the West, phage therapy was widely practiced in the former USSR until 1990 and surprisingly good results were reported in the 1980s from Poland. They suggest that phage therapy is a viable therapeutic alternative in antibiotic-resistant pyogenic infections (wounds, abscesses, furunculosis, osteomyelitis, septicemia). More basic research is needed on, for example, inactivation of phages by body fluids. Phage prophylaxis of infectious diseases was also attempted. Despite encouraging results in cholera prevention, it is of historic interest only. Phage control of plant diseases has not been recommended.

2. Identification and classification of bacteria Early attempts to use phages for bacterial identification were abandoned because no phage lyses all strains of a bacterial species and no others. A few diagnostic phages are still used as screening agents in specialized laboratories, for example, for rapid identification of Bacillus anthracis, members of the genus Salmonella, or the biotype E1 of Vibrio cholerae. By contrast, phage typing is an important technique in epidemiology. In analogy with the antibiogram, bacteria are tested against a set of phages and subdivided into resistance patterns or phage types. Briefly, a continuous layer of bacteria is created on an agar surface, phage suspensions are deposited on it, and results are read the next day. Phage typing is invaluable for subdividing biochemically and serologically homogeneous bacterial species. Besides international typing schemes for Salmonella typhi and Salmonella paratyphi

ch10.qxd 10/9/03 6:53 PM Page 145

145

bacteriophages B, there are typing sets for most human, animal, and plant pathogenic bacteria. Because of their host specificity, phages are also valuable tools in bacterial taxonomy. Phage host ranges were a major argument in reclassifying Pasteurella pestis as an enterobacterium of the genus Yersinia. 3. Genetic engineering Phages have made many contributions to recombinant DNA technology. Restriction endonucleases were first identified in a phage-host system and the DNA ligase of phage T4 is used to insert foreign DNA into viral or plasmid vectors. In addition, phages have several major applications: 1. Phage , derivatives of it, cosmids (hybrids between DNA and plasmids), phage P1, and “mini-Mu” phages (derivatives of Mu containing the left and and right ends of the Mu genome) are used as cloning vectors. Recombinant DNA (vector plus foreign DNA) is introduced into phage proheads. After completion of phage assembly, it can be injected into bacteria. 2. Filamentous coliphages of the Inovirus genus are used for DNA sequencing. Foreign DNA is introduced into the double-stranded replicative form of these phages. The same coliphages are also used to express proteins or peptides at their surface. The technique, called “phage display,” is a powerful tool for the selection and cloning of antibody fragments. 3. Phage Mu DNA, able to integrate at random into any gene, is used to create mutations and to displace genes to other locations. 4. Other applications 1. Destruction of unwanted bacteria in bacterial and cell cultures, milk, meat, and freshwater (e.g., of cyanobacteria in “algal blooms”). 2. Assay of antivirals, disinfectants, air filters, and aerosol samplers. 3. Detection of fecal pollution in water and of carcinogens, mutagens, and antitumor agents. 4. Detection of Listeria bacteria in food using luciferase-marked phages. 5. Tracing of water movements (surface water and aquifers).

B. Phages as pests In industrial microbiology, phages may destroy valuable starter cultures or disrupt fermentation processes.

Phage interference has been reported in various branches of the fermentation industry, notably in the production of antibiotics, organic solvents, and cheese. In the dairy industry, phage infection is considered as the largest single cause of abnormal fermentations and a great source of economic losses. Phages derive from raw material, plant environment, or phagecarrying starter cultures. They are disseminated mechanically by air and may persist for months in a plant. Phage control is attempted by: (i) preventing contamination by cleanliness, sterilization of raw material, sterile maintenance of starter cultures, and use of phage-free starters; (ii) disinfection by heat, hypochlorites, UV light, and other agents; or (iii) impeding phage development by starter rotation, use of genetically heterogeneous starters, and phageinhibiting media. A recently developed approach is to construct phage-resistant starters by genetic engineering.

BIBLIOGRAPHY Ackermann, H.-W. (1985). Les virus des bactéries. In “Virologie Médicale” (J. Maurin, ed.). Flammarion Médecine-Sciences, Paris). Ackermann, H.-W. (1987). Bacteriophage taxonomy in 1987. Microbiol. Sci. 4, 214–218. Ackermann, H.-W. (1998). Tailed bacteriophages—The order Caudovirales. Adv. Virus Res. 51, 135–201. Ackermann, H.-W. (2001). Frequency of morphological phage descriptions in the year 2000. Arch. Virol. 146, 843–857. Ackermann, H.-W., and DuBow, M. S. (1987). “Viruses of Prokaryotes,” Vols 1 and 2. CRC Press, Boca Raton, FL. Calendar, R. (ed.) (1988). “The Bacteriophages,” Vols 1 and 2, Plenum Press, New York. Casjens, S., Hatfull, G., and Hendrix, R. (1992). Evolution of the dsDNA tailed-bacteriophage genomes. Sem. Virol. 3, 383–397. Friedman, D. I., and Court, D. L. (2001). Bacteriophage Lambda: alive and well and still doing its thing. Curr. Opin. Microbiol. 4, 201–207. Goyal, S. M., Gerba, C. P., and Bitton, G. (eds.) (1987). “Phage Ecology.” John Wiley & Sons, New York. Holt, J. G. (editor-in-chief) (1984, 1986, 1989, 1990). “Bergey’s Manual of Systematic Bacteriology,” Vols 1–4. Williams & Wilkins, Baltimore, MD. Klaus, S., Krüger, D., and Meyer, J. (1992). “Bakterienviren.” Gustav Fischer, Jena, 1992. Maniloff, J., and Ackermann, H.-W. (1998). Taxonomy of bacterial viruses: establishment of tailed virus genera and the order Caudovirales. Arch. Virol. 143, 2051–2063. Smith, M. C. M., and Rees, C. E. D. (1999). Exploitation of bacteriophages and their components. In “Methods in Microbiology” (M.C.M. Smith, and R.E. Sockett, eds.) Academic Press, San Diego, Vol. 29, 97–132.

ch10.qxd 10/9/03 6:53 PM Page 146

146

the desk encyclopedia of microbiology

Tidona, C. A., and Darai, G. (eds.) (2001). “The Springer Index of Viruses.” Springer Verlag, Berlin. Van Regenmortel, M. H. V., Fauquet, C. M., Bishop, D. H. L., Carstens, E., Estes, M. K., Lemon, S., Maniloff, J., Mayo, M. A., McGeoch, D. J., Pringle, C. R., and Wickner, R. B. (eds.) (2000). “Virus Taxonomy. Classification and Nomenclature of Viruses.” Seventh Report of the International Committee on Taxonomy of Viruses. Academic Press, San Diego, CA. Wilson, D. R., and Finlay, B. B. (1998). Phage display: applications, innovations, and issues in phage and host biology. Can. J. Microbiol. 44, 313–329. Zillig, W., Arnold, H. P., Holz, I., Prangishvili, D., Schweier, A., Stedman, K., She, Q., Phan, H., Garrett, R., and Kristjansson, J. K. (1998). Genetic elements in the extremely thermophilic archeon Sulfolobus. Extremophiles 2, 131–140.

WEBSITES ATCC Bacteriophage Collection Catalog http://www.atcc.org/SearchCatalogs/bacteriophages.cfm Bacteriophage Division of the American Society for Microbiology http://www.asmusa.org/division/m/M.html Phage Ecology and Phage Catalog http://www.phage.org/ Phage Therapy http://www.evergreen.edu/phage/ The Institute for Genomic Research (TIGR) and links to many other microbial genomic sites http://www.tigr.org/tigr-scripts/CMR2/CMRHomePage.spl

ch11.qxd 10/9/03 6:53 PM Page 147

11 Biocides (Nonpublic health, Nonagricultural antimicrobials) Mohammad Sondossi Weber State University

suffix cide would be classified under the category of biocides. This broad literal meaning encompasses many other topics in this and other encyclopedias. The terms herbicide and insecticide indeed have biocidal activities. Therefore, the topic biocides has to be defined more narrowly in relation to specific subjects and applications.

GLOSSARY biocidal agent An agent that kills all living organisms. biocide Primarily a chemical substance or composition used to kill microorganisms considered to be undesirable (i.e. pest organisms). biodegradation A chemical breakdown of a substance into smaller molecules caused by microorganisms or their enzymes. biodeterioration A physical or chemical alteration of a product, directly or indirectly caused by living organisms, their enzymes, or by-products, thereby making the product less suitable for its intended use. biostatic agent An agent that inhibits or halts growth and multiplication of organisms. This means that when the agent is removed, the organism resumes growth and multiplication. deteriogenic organisms The organisms that cause biodeterioration. minimal inhibitory concentration The concentration of a particular biocide/antimicrobial agent necessary to inhibit the growth of a particular microorganism. selective toxicity The ability of a chemotherapeutic agent (antibiotic, etc.) to kill or inhibit a microbial pathogen with minimal damage to the host at concentrations used.

Perhaps the most appropriate terminology for this topic drawn from common usage is “industrial biocides.” It should be accepted that scientific terms can acquire new or multiple meanings according to common usage. It is of particular importance that terminologies used are consistent and descriptive of the activity of the agents involved. It is imperative to recognize that the terminology implies a specific use and the language used for labeling biocides may have legal consequences. However, the common usage changes the meaning of the terms and even the perception of the spectrum of their activities. Table 11.1 represents some of the common terms related to the control or suppression of microbial growth. According to the Environmental Protection Agency’s (EPA) Office of Pesticide Programs, antimicrobial pesticide (a term used by the EPA) products contain approximately 300 different active ingredients and are marketed in several formulations: sprays, liquids, concentrated powders, and gases. Currently, more than 8000 antimicrobial pesticide products that are registered with the EPA are sold, constituting an approximately $1 billion market. The EPA estimates that the total amount of antimicrobial pesticide active ingredients used in 1995 was 3.3 billion

The Simplest Definition of a Biocide is evident from the terminology: bio, meaning life, and cide, referring to killing–an agent that destroys life. Therefore, any word with the The Desk Encyclopedia of Microbiology ISBN: 0-12-621361-5

147

Copyright © 2003 Elsevier Ltd All rights of reproduction in any form reserved

ch11.qxd 10/9/03 6:53 PM Page 148

148

the desk encyclopedia of microbiology TABLE 11.1 Terminology related to control of microbial growth

Term

Definition

Comments

Microbicide

An agent that kills microbes but not necessarily their spores An agent that kills pathogens and many nonpathogens but not necessarily their spores An agent that kills bacteria but not necessarily bacterial endospores An agent that kills fungi (mold and yeast) but not necessarily their spores An agent that kills algae An agent that inactivates viruses An agent that kills bacterial endospores and fungal spores

Usually a general term

Germicide Bactericide Fungicide Algicide Virucide Sporocide

Sanitizer

An agent that reduces the microbial contaminants to safe levels as determined by public health requirements

Disinfectant

An agent, usually chemical, commonly used on inanimate objects to destroy pathogenic and harmful organisms but not necessarily their spores Usually a chemical agent commonly applied to living tissue, skin, or mucous membrane to kill or inhibit microorganisms An agent that prevents or preempts biodeterioration and spoilage of a product or material under storage conditions A chemical agent that destroys all forms of life, including spores, and inactivates viruses An agent that inhibits growth and multiplication of bacteria but does not kill them An agent that inhibits growth and multiplication of fungi but does not kill them Substance or mixture of substances used to destroy or suppress the growth of undesirable (pest) microorganisms on inanimate objects and surfaces A substance, usually produced by microorganisms, which in low concentrations inhibits the growth or kills disease-causing microorganisms

Antiseptic

Preservative

Chemical sterilizer Bacteriostatic agent Fungistatic agent Antimicrobial pesticide

Antibiotic

lbs, accounting for 75% of all pesticides’ active ingredients used. Antimicrobial pesticides, from a regulatory standpoint, are divided into public health products and nonpublic health products. The latter products are used to control microorganisms which cause spoilage, deterioration, or fouling of materials. Industrial biocides are classified under this category. They are chemical compositions used to control and prevent microbial biodeterioration and contamination of industrial/commercial material, systems, and products and/or to improve the efficiency of operation. The application of industrial biocides varies greatly depending on major use categories. Although biocides used in the preservation of cosmetics and personal care products are relevant to this article, they

A general term A general term A general term A general term A general term Sporocidal action is not to be equated to sporistatic action which results in inhibition of spore germination Usually used on inanimate objects and places where no specific pathogens are present or suspected and complete killing of all forms of microorganisms is not necessary Widely used term, legal definition includes more details (relative to factors of time, temperature, percentage kill, concentration, etc.); may inactivate viruses Not considered safe for internal use

A chemical or physical agent or process resulting in the act of preservation Sterilization is an absolute term and there are no degrees of sterilization If the bacteriostatic agent is removed bacterial growth may resume If the fungistatic agent is removed fungal growth may resume This is the terminology used by the EPA and it encompasses all the antimicrobial agents fitting the definition under public health and nonpublic health categories Synthetic and semisynthetic antibiotics are also available, usually used in treatment of disease; has selective toxicity

will not be discussed. The emphasis will be on nonpublic health-related topics. In the United States, cosmetics and toiletries are regulated under the Federal Food, Drug, and Cosmetic Act.

I. HISTORICAL PERSPECTIVE Current uses of biocides are aimed at the inhibition and control of undesirable microorganisms based on their antimicrobial action and the microbes’ potential roles in biodeterioration, spoilage, and disease. However, even before the discovery of the microbial world by Antoni van Leeuwenhoek in 1674, many practices were used to preserve material, food, and

ch11.qxd 10/9/03 6:53 PM Page 149

149

biocides animal and human bodies (mummification). Although early methods were effective, the scientific foundation of these practices was not understood until almost two centuries after van Leeuwenhoek’s discovery. Drying and salting fish and meat was one of the earliest food preservation techniques developed. In cold climates, food could also be buried in snow or underground. An early version of surface sterilization was the practice of passing metal objects through fire to clean them. One of the first documented cases of chemical sanitation was practiced as early as 450 BC in the Persian Empire, where boiled water was stored in copper and silver containers. This allowed for a portable water supply that helped the Persian army in many military conquests. Medical applications of chemical sanitation include the use of mercuric chloride as a wound dressing by Arab physicians in the Middle Ages, the use of bleaching powder as a disinfectant by Alcock in 1827, the use of iodine by Davies in 1839, the use of chlorinated water for hand washing in hospitals by Semmelweis in the 1840s, and the use of phenol on surgical wounds by Lister in the 1860s. It was not until after the mid-nineteenth century that quantitative and comparative antimicrobial efficacies of some compounds were established. For example, in 1875 Bucholtz determined the minimal inhibitory concentrations of phenol, creosote, salicylic acid, and benzoic acid against bacterial growth. These and other findings were followed by the introduction of hydrogen peroxide as a disinfectant by Traugott in 1893 and chlorine-releasing compounds in 1915 by Dakin. It is obvious that most of these findings and their applications were in the areas of medicine and public health. Here, the description of biocides is not in the context of the general definition based on antimicrobial activity but instead covers applications in more specific areas dealing with biodeterioration. The latter term has been in use for the past 30 years and is basically not present in most dictionaries, traditional printed material, or even recent electronic media. Hueck (1968) defined biodeterioration as any undesirable change in the properties of a material used by humans caused by the vital activities of organisms in which the material is any form of matter, with the exception of living organisms. The term biodeterioration is distinguished from biodegradation in having a more negative or harmful connotation. Where the material is known to be at risk, preventative measures can be taken. These measures could be based on many physical and chemical parameters affecting microbial growth and include the use of biocides. It should be mentioned that the term biocide is also a new term introduced in the past few decades. The range of materials used by man has changed

dramatically in comparison to days when raw materials were used with minimal processing. Currently, complex and heavily processed materials, composites, and synthetic and semisynthetic materials are everywhere. Complex man-made environments and the materials used, combined with a wide range of biotic and abiotic parameters, provide abundant and ideal environments for the growth of microorganisms. The total cost of losses due to biodeterioration and spoilage is approximately $100 billion per year. When the costs of replacement of deteriorated material, remedial measures, and lost productivity are considered, the importance of preventative measures and the role of biocides is clear.

II. CURRENT APPLICATIONS There are many arbitrary categories of biocide applications in published articles and reference books. They have been grouped from an application aspect, with some differences. It should be noted that the use of a particular biocide is not restricted to any one group and may be used in different areas. One such summary of application categories could be assembled from articles, reviews, and reference materials as presented in Table 11.2. Another grouping based on areas of application is used by regulatory agencies (the EPA and the Food and Drug Administration). These groups can be divided into two categories based on the type of microorganism (pest) against which the biocide (antimicrobial pesticide) is used. First, public health antimicrobials are intended to control infectious microorganisms (to humans) in any inanimate environment. Disinfectants, sterilizers, sanitizers, and antiseptics are included in TABLE 11.2 Selected application areas of biocides Human drinking water dis-infectants and purifiers Freshwater algicide Swimming pools Animal husbandry Animal feed preservatives Food and beverage processing hygiene Food preservation Crop protection Hospital disinfectants Hospital and medical antiseptics Pharmaceuticals Cosmetics Personal care disinfectants Process cooling water

Paper and pulp Metalworking lubricants and hydraulic fluids Oil field operations Fuels Textiles Paint and paint film preservation Wood preservation Plastics Resins Polymer emulsion, latex, adhesives, slurries Tannery Museum specimens Construction

ch11.qxd 10/9/03 6:53 PM Page 150

150

the desk encyclopedia of microbiology

TABLE 11.3 Antimicrobial product use sites and categories under consideration by the EPAa

Agricultural premises and equipmentb Food handling/storage establishment premises and equipmentb Commercial, institutional, and industrial premises and equipment Residential and public access premisesb Medical premises and equipment Human drinking water systemsb Materials preservativesb Industrial processes and water systems Antifouling coatings Wood preservatives Swimming pools Aquatic areas a Major use categories are subdivided further based on exposure scenarios. b Use of a biocide product on some sites in this category with direct or indirect food contact will be considered a food use and registration must be supported by data sufficient to support the establishment of a tolerance or exemption from the requirement of a tolerance under the Federal Food, Drug, and Cosmetic Act.

this category. Second, the nonpublic health antimicrobial products are the products used to control growth of microorganisms causing deterioration, spoilage, and fouling of material, including growth of algae and odorcausing bacteria. Human exposure, product chemistry, and toxicology are considered in assigning antimicrobial agents into these main application categories. In order to meet registration and data requirements, the EPA is currently considering classifying the antimicrobial products into 12 major use categories which are further subdivided based on exposure scenarios. The major categories are shown in Table 11.3.

III. EXAMPLES OF INDUSTRIAL BIOCIDES Providing a complete list of registered industrial biocides is beyond the scope of this article and is not practically possible for many reasons. For example, in the United States, most states require biocide registration after registration with the EPA. In addition to Canada, European countries, and Japan, many other countries have laws regulating biocide registration and use. Toxicological and environmental impact regulations vary worldwide and are reevaluated constantly. An extensive list of biocides with current and past use in North America and Europe is given in Table 11.4, although some have been discontinued, have use limitations, and may not be registered in some countries. Therefore, for updated information on industrial biocides, contacting biocide manufacturers and regulatory agencies is strongly recommended.

Databases are available that indicate the name and location of the basic manufacturers of compounds as listed by CAS registration number. Most of the regulatory statuses can also be obtained from on-line databases and search engines.

IV. CLASSIFICATION OF BIOCIDES Classifying industrial biocides based on their chemical structures is not an easy task. Many review articles present group classifications and include a miscellaneous group whose members do not fit in any major class. Table 11.5 is a representation of this type of classification. Biocides are sometimes also grouped based on their mode of action. This can be organized based on the target region of the microorganism affected by biocide action. Terms and categories such as membraneactive biocides and permeabilizers, cell wall inhibitors, cytotoxic agents (affecting targets in cytoplasm and interfering with metabolism and total cell function), and genotoxic agents (affecting DNA biosynthesis and reacting with DNA) have occasionally been used. The chemical reactivity of biocides provides another, less frequently used, classification. Terms such as oxidizing, non-oxidizing, and electrophilic biocides have been used to separate industrial biocides into smaller groups. Some have used terms such as chlorine-yielding, bromine-yielding, and formaldehydereleasing compounds to designate specific groups of biocides based on the active moiety/mechanism of action. It is therefore understandable that, depending on the subject, audience, users, and presenters (regulatory, academic, and industry) of information pertaining to industrial biocides, any of these classifications could be used.

V. EVALUATION There are numerous methods that have been and can be employed to evaluate biocide efficacy, including a variety of basic microbiological tests, simulation tests in the laboratory, practical tests, and field tests to demonstrate the effectiveness of a biocide. First, it has to be demonstrated that the chemical or preparation being evaluated has antimicrobial activity. In this stage, the spectrum of activity is determined against bacteria (gram-positive, gram-negative, Mycobacteria, etc.), fungi (mold and yeast), and spores (bacterial and fungal) and dose-response relationships are established.

ch11.qxd 10/9/03 6:53 PM Page 151

151

biocides TABLE 11.4 Selected industrial biocidesa,b Active chemical

Applications

6-Acetoxy-2,4-dimethyl-m-dioxane

Metalworking fluids, textile lubricants, polymer emulsions, other aqueous emulsions Paper and pulp Paper and pulp, process cooling waters Wood, process cooling waters Wood Metalworking fluids Adhesive, latex, paper coatings, aqueous emulsions Paint raw materials (cellulose and casein) Adhesives Paper and pulp

Acrolein (acrylaldehyde) Alkenyl (C12–C18)dimethylethyl ammonium bromide Alkyldimethylbenzyl ammonium chloride Arsenic pentoxide 1-Aza-3,7-dioxa-5-ethylbicyclo-[3.3-0]octane 1,2-Benzisothiazolin-3-one Benzyl bromoacetate Benzyl-hemiformals mixture Bis(1,4-bromoacetoxy)-2-butene 5,5-Bis(bromoacetoxymethyl)-m-dioxane 2,6-Bis(dimethylaminomethyl) cyclohexanone 1,2-Bis(monobromoacetoxy) ethane Bis(tributyltin)oxide Bis(trichloromethyl)sulphone Boric oxide Brominated salicylanilides 5,4-Dibromosalicylanilide 3,5,6-Tribromosalicylanilide Other brominated salicylanilides Bromine-yielding chemicals Sodium bromide, NaBr (must be activated by oxidizing agent, e.g. NaOCl, Cl2, and potassium peroxymono-sulfate) 1-Bromo-3-chloro-5,5-dimethylhydantoin 4-Bromoacetoxymethyl-m-dioxolane 2-Bromo-4-hydroxyacetophenone 2-Bromo-2-nitro-1,3-propanediol Bromo-nitrostyrene 1,1-(2-Butenylene)bis(3,5,7-triaza-1-azoniaadamantane chloride) Chlorethylene bisthiocyanate Chlorinated levulinic acids Chlorine/chlorine-yielding chemicals: Chlorine (gas), Cl NaOCl Ca(OCl)2•4H2O Na dichloro-s-triazinetrione/trichloro-s-tri azinetrione 1-Bromo-3-chloro-5,5-dimethylhydantoin 1-Bromo-3-chloro-5-methyl-5-ethylhydantoin 1,3-Dichloro-5,5-dimethylhydantoin Chlorine dioxide, ClO2 1-(3-Chloroallyl)-3,5,7-triaza-1-azoniaadamantane chloride sodium bicarbonate Chloromethyl butanethiolsulfonate 5-Chloro-2-methyl-4-isothiazolin-3-one 2-methyl-4isothiazolin-3-one p-Chlorophenyl diiodomethyl sulfone Chromic oxide Coal tar creosote Copper naphthenate Copper sulfate Cupric oxide Copper-8-quinolinolate Cresylic acids Cupric nitrate Dialkyl methylbenzyl ammonium chloride 1,2-Dibromo-2,4-dicyanobutane

Wood, process cooling water Paper and pulp, process cooling water, adhesives, wet state protection concrete additives Wood Water-based latex pains and emulsions, joint cement, PVC plastic, acrylic and PVA water-based paints, polyvinyl acetate latex, adhesives Process cooling waters

Paper and pulp Pulp and paper mills, paper making chemicals, felt Metalworking fluids, textile, Process cooling waters Pulp and paper mills, water systems, lignosulphonates Latex paints, resin emulsions, adhesives, dispersed colors Water systems, emulsions Paper and pulp Process cooling waters

Adhesives, metalworking fluids, latex paints, textile, emulsions, water-based coating formulations Paper and pulp Wood veneer, cutting fluids and coolants, paste, slimes, cooling towers, paper, and paperboard Paint Wood Wood Wood Wood Wood Wood and wood products, glues and adhesives, paper products Rubbers (synthetic and natural) Paper and pulp Paper and pulp, wood, process cooling waters Metalworking fluids, aqueous paints, latex emulsions, joint cement adhesive (Continued overleaf)

ch11.qxd 10/9/03 6:53 PM Page 152

152

the desk encyclopedia of microbiology TABLE 11.4 (Continued)

Active chemical

Applications

2,2-Dibromo-3-nitrilopropionamide (20, 10, or 5%) in polyethylene glycol 2,3-Dibromopropionaldehyde 2,4 Dichlorobenzyl alcohol 2,3-Dichloro-1,4-naphthoquinon Didecyl dimethyl ammonium chloride (2,2-Dihydroxy-5,5-dichloro)-diphenyl methane (2,2-Dihydroxy-5,5-dichloro-diphenyl monosulfide Di-iodomethyl-p-tolyl sulfone Di-isocyanate Ditmethyl aminomethyl phenol Dimethylbenzyl ammonium chloride 4,4-Dimethyloxazoldine 3,4,4-trimethyloxazolidine 3,5-Dimethyl-tetrahydro-1,3,5,-2H-thiadiazine-2-thion Dioctyl dimethyl ammonium chloride and ethanol Diquat 1,1-ethylene-2,2-dipyridiylium Disodium cyanodithioimidocarbamate Disodium ethylenebis(dithiocarbamate) Dithio-2,2-bis-benzmethylamide Dithiocarbamates benzimidazole derivatives

Water cooling towers, pulp and paper mills, metalworking fluids, oil recovery Paper and pulp Textile Toxic wash of construction material (interior use) Wood Textile, cement additive, toxic wash (exterior use) Textile Paint Toxic wash construction material (interior use) Rubbers (synthetic and natural) Construction toxic wash (exterior and interior use) Metalworking fluids, in-can paint Leather, paint, glue, casein, starch, paper mill Cooling water systems Construction toxic wash (exterior and interior use) Paper and pulp, process cooling waters Paper and pulp, process cooling waters Adhesives Adhesives, filters, stoppers, groutings, jointing compounds, sealants, putty Construction toxic wash (exterior and interior use) Paper and pulp, process cooling waters

Dodecylamine salicylate Dodecylguanidine hydrochloride, dodecylguanidine hydro-chloride Fatty acids of quaternary compounds Fluorinated sulfonamide Formaldehyde Glutaraldehyde (1,5-pentanedial) 1,2,3,4,5,6-Hexachlorocyclohexane (lindane) Hexachloro dimethyl sulfone Hexahydro-1,3,5-triethyl-s-triazine Hexahydro-1,3,5-tris(2-hydroxyethyl)-s-triazine Hydrogen peroxide p-Hydroxybenzoic acid esters Ethyl p-hydroxybenzoate Methyl p-hydroxybenzoate Propyl p-hydroxybenzoate Butyl p-hydroxybenzoate 5-Hydroxymethoxymethyl-1-aza-3,7dioxabicyclo(3.3.0)octane 5-Hydroxymethyl-L-aza-3,7-dioxabicyclo(3.3.0)octane 5-Hydroxypoly[methyleneoxy (74% C2, 21% C3, 4% C4, 1% C5)]methyl-L-aza-3,7-dioxabicyclo(3.3.0)octane 2-[(Hydroxymethyl) amino] ethanol 2-[(Hydroxymethyl) amino]-2-methylpropanol 2-(p-Hydroxyphenyl) glyoxylohydroximoyl chloride 2-Hydroxypropyl methanethiol sulphonate 3-Iodo-2-propynyl butyl carbamate 3-Methyl-4-chlorophenol Methyl-2,3-dibromopropionate 2,2-Methylenebis(4-chlorophenol) Methylene bis thiocyanate N-[alpha-(nitroethyl)benzy] ethylenediamine N-dimethyl-N-fluorodichloromethylthio) sulfamide N-(fluordichloromethylthio) phthalmide N-trichloromethylthio-4-cyclohexene-1,2-dicarboximide N-(trichloromethylthio) phthalimide

Textile Filters, stoppers, groutings Toxic wash (exterior and interior use) Metalworking fluids, Process cooling waters Wood Industrial emulsions Cutting oils, synthetic rubber latexes, adhesives, latex emulsions Cutting oils and diluted coolants Process cooling waters Adhesives, starch and gum solutions, inks, polishes, latexes, other emulsions

Latex paints

Paints, resin emulsions, in-can paint Latex paints, resin emulsions Paper and pulp Paper and pulp, paint films Interior and exterior coatings Adhesives, filters, stoppers, groutings Process cooling waters Textiles, rubber products, hoser Paper slimes, recirculating cooling water systems Paper and pulp Construction toxic wash (exterior use) Construction toxic wash (exterior and interior use), jointing compounds, sealants, putty, plastic products Paper and pulp, polyethylene, paint, paste, rubber and rubber-coated products Nonaqueous paints and caulking compounds

ch11.qxd 10/9/03 6:53 PM Page 153

biocides

153

TABLE 11.4 (Continued) Active chemical

Applications

N-(trimethylthio) phthalimide 2-Nitrobutyl bromoacetate 4-(2-Nitrobutyl) morpholine 4.4-(2-ethyl-2-nitrotrimethylene) dimorpholine 2-n-Octyl-4-isothiazolin-3-one

Paint film Paper and pulp Metalworking fluids, pulp and paper industry, petroleum production, jet fuels Latex and oil-based paints, in-can paint preservative, fabrics, wet processing of hides Paints Cooling towers, air washer systems Cooling water systems PVC, polyurethane, other polymeric compositions Film and sheeting, extruded plastics, plastisols, molded goods, organosols, fabric coatings, etc. Hydrocarbon fuels, boat and ship fuel and marine storage, home heating fuel Plastic products Plastic products Process cooling waters Metalworking fluids Metalworking fluids Wood preservation, adhesives, cement additive, toxic wash (exterior use), rubbers (synthetic and natural) Adhesives, wet state protection concrete additives, plastic products, rubbers (synthetic and natural), textile Bitumen products, jointing compounds, sealants, putty Adhesives Adhesives, wet state protection concrete additives Adhesives, wet state protection concrete additives Protein-based paints, metalworking fluids, polishes, adhesives, gums, latexes, textiles Cooling towers and evaporative condensers Cooling towers Industrial water systems, process cooling waters

Organic mercurials Organosulfur compound blends Organotin, quartemaries and amines 10,10-Oxybisphenoxarsine 5% in a polymeric resin carrier 10,10-Oxybisphenoxarsine in various nonvolatile plasticizer carriers 2,2-Oxybis-(4.4,6-trimethyl-1,3,2-dioxaborinane)-2,2(1-methyltrimethylenedioxy)-bis-(4-methyl-1,3,2-dioxaborinane) Oxyquinofine Oxyquinoline sulphate Ozone Para-chloro-meta-cresol Para-chloro-meta xylenol Pentachlorophenol Pentachlorophenyl laurate Phenoxy fatty acid polyester Phenyl mercury acetate Phenyl mercury nonane Phenyl mercury oleate o-Phenylphenol(sodium-o-phenylphenate tetrahydrate) Polychlorophenates, alcohol, and amines Polychlorophenates, organosulfurs Poly[hydroxyethylene (dimethyliminio)-ethylene (dimethyliminio)]methylene dichloride Poly[oxyethylene (dimethyliminio)-ethylene (dimethyliminio)]ethylene dichloride Potassium dichromate Potassium dimethyl thiocarbamate Potassium N-hydroxymethyl-N-methyldithiocarbamate Potassium N-methyldithiocarbamate Quaternary phosphonium salt surfactant Rosin amine D-pentachlorophenate Salicylamide Silver fluoride, silver nitrate Sodium dimethyldithiocarbamate Sodium fluoride Sodium 2-mercaptobenzothiazole

Sodium pentachlorophenate Sodium 2-pyridinethiol-1-oxide 1,3,6,8-Tetraazatricyclo[6.2.1.1]dodecane 2,4,5,6-Tetrachloroisophthalonitrile

Cooling water systems, cutting fluids Wood Metalworking fluids, process cooling water Water-thinned colloids, emulsion reins, emulsion paints, waxes, cutting oils, adhesives Paper and pulp, process cooling waters Textile Paper, textiles, rope, emulsion systems (Cable insulation) jointing compounds, sealants, putty, plastic products Paper and pulp Paper mills, cooling towers, paper and paperboard, cotton fabrics, paste, wood, veneer, cutting oils Wood Adhesives, wet state protection concrete additives, paper mills, cooling towers, paper and paperboard, cotton fabrics, paste, wood, veneer, cutting oil, water-thinned colloids, emulsion reins, emulsion paints, waxes, adhesives, textiles, rug backings Paper making, pulp, paper and paper products, leather, hides, drilling muds Aqueous-based metalworking fluid systems, vinyl, latex emulsions for short-term, in-can inhibition of bacterial growth Paper and pulp Adhesives, jointing compounds, sealants, putty, plastic products, latex paints (Continued overleaf)

ch11.qxd 10/9/03 6:53 PM Page 154

154

the desk encyclopedia of microbiology TABLE 11.4 (Continued)

Active chemical

Applications

2,3,4,6-Tetrachlorophenol 3.3,4.4-Tetrachlorotetrahydrothiophene-1,1-dioxide Tetrahydro-3,5-dimethyl-2H-1,3,5-thiadiazine-2-thione and blends 2-(4-Thiazolyl)benzimidazole 2-(Thiocyanomethylthio)benzothiazole Tributyltin acetate Tributyltin fluoride Tributyltin maleate Tributyltin oxide Tributyltin oxide nonionic emulsifier Trifluoromethyl thiophthalmide

Wood preservation Paper and pulp Slimicide for coatings, clay slurries, adhesives, glues, latex emulsions, casein, titanium slurries, cooling towers Paint Wood, paint films Cement additive, paints Wood, antifouling paint Textile Construction toxic wash (exterior and interior use), wet state protection concrete additives, antifouling paints, adhesives, wood Construction toxic wash (exterior and interior use), wet state protection concrete additives Construction toxic wash (exterior use)

3-(Trimethoxysilyl)-propyl-dimethyloctadecyl ammonium chloride Tris(hydroxymethyl) nitromethane Zinc dimethylditbiocarbamate Zinc 2-mercaptobenzothiazole Zinc naphthenate Zinc 2-pyridinethiol-1-oxide

Oil in water emulsions, pulp and paper industry, water treatment, in-can paint Adhesives, cooling water, paper mill, paper and paperboard, textiles Textile, wood Aqueous-based metalworking fluids, PVC plastics

a Not necessarily currently registered and some have been discontinued. This list was collected from many sources, including Sharpell (1980), Allsopp and Allsopp (1983), Bravery (1992), Rossmoore (1995a,b), Lutey (1995), Eagon (1995), McCarthy (1995), Downey (1995), and Leightley (1995). b Current registration eligibility decision (RED) and fact sheets on biocides can be found on: http://www.epa.gov/pesticides/ reregistration/status.htm.

TABLE 11.5 Biocides by chemical classa Phenols Organic and inorganic acids: esters and salts Aromatic diamidines Biguanides organometallic compounds Surface-active agents: cationic and anionic agents Aldehydes: formaldehyde, glutaraldehyde, and others Dyes Halogen compounds Quinolines and isoquinoline derivatives

Alcohols Perooxygens Chelating agents Heavy metals and Anilides Formaldehyde adducts Isothiazolones Organosulfur compounds Essential oils Miscellaneous

a

From Hugo and Russell (1992), Rossmoore (1995a), and others.

The nature of antimicrobial activity, biocidal or biostatic, may also be determined. The second stage includes suspension tests to determine MIC, establishing kill curves by plating, capacity tests (several reinoculations), and carrier tests (effects on organisms on the carrier). These may be followed by practical tests that, although done in the laboratory, demonstrate the efficacy under real-life conditions. Third, and most important, is evaluation in the field and under actual use conditions. For the regulatory

agencies and registration purposes the tests should satisfy the label claims for specific applications. There are numerous test methods issued by federal and state governments or government-sanctioned publications, standards societies, and trade organizations as well as test methods developed by biocide manufacturers, users, and testing laboratories to demonstrate efficacy. Among governmental-sanctioned publications are those by the Association of Official Analytical Chemists, the American Public Health Association, and the United States Pharmacopeia. Voluntary consensus standards societies and groups include the American National Standards Institute, the American Society for Testing and Materials, and the International Standards Organization.

VI. MODE OF ACTION Considering the heterogeneity of chemicals used as biocides, and the fact that they have been considered general cell poisons for a long time, one can understand the lack of detailed information on mode of action of industrial biocides. For most of the biocides, mode of action seems to be a concentration-dependent phenomenon by which individual effects can be

ch11.qxd 10/9/03 6:53 PM Page 155

biocides identified and studied. Since biocides will act on organisms in an outside to inside direction, many have classified the target regions based on this directional impact. In other words, the cell wall, cell membrane and membrane-associated components, and the cytoplasmic regions will be sequentially affected by the biocide as it interacts with the intended target organism. Unlike antibiotics that have very specific targets, biocides may have more than one potential target. These could be located at any or all areas of the affected cell. The chemical structure of a biocide determines its affinity to specific targets and is the key to understanding its mode of action. Furthermore, the accumulated effects of sequentially affected regions of the cell may ultimately manifest as antimicrobial activity. Considering the structural and physiological differences of organisms and extrinsic parameters affecting the activity of biocides, the knowledge about mode of action is far from comprehensive. However, it is becoming increasingly evident that biocides indeed have a specific target(s) and cannot be labeled as general cell poisons. Abnormal morphology of organisms exposed to biocides, studied by light and electron microscopy, has long been considered evidence of damage to the cell wall or its construction process. Lysis of cells due to initiation of autolysis has also been included in this category of mode of action. Early reports classified phenol, formaldehyde, mercurials, alcohols, and some quaternary ammonium compounds in this category. It should be noted that any of these events could also be a consequence of damage(s) exerted on cytoplasmic targets or initiated by trans-membrane signaling events. Since the cell wall composition of microorganisms (e.g., gram-positive and gram-negative bacteria, mycobacteria, fungi, and algae) differs significantly, one biocide may cause damage to the cell wall of one organism and may have no effect on the cell wall of another. Interaction with the cytoplasmic membrane, membrane-bound enzymes, electron transport, and substrate transport systems are the next group to be affected by the action of biocides. Among other biocides, chlorhexidine, 2-bromo-2nitro-1,3-propanediol, and 1,2-benzisothiazolin-3-one have been reported to affect targets in the cytoplasmic membrane. Some membrane-active biocides may cause leakage of the intracellular material, whereas others have been reported to produce an increased permeability to ions acting as uncouplers of oxidative phosphorylation and inhibitors of active transport. Although there are many early reports that describe the mode of action of certain biocides in terms such as coagulation of cytoplasmic proteins and precipitation of cytoplasmic constituents, there are recent reports of

155

more specific actions on selective inhibition of cytoplasmic enzymes and reactions with essential biomolecules. These more specific interactions result in inhibition of selected biosynthetic and energyproducing processes in the affected organisms. Some recent reports describing more specific aspects of industrial biocides’ modes of action include the following examples: 2-pyridinethiol-1-oxide has been suggested to act on cell membranes to eliminate important ion gradients used to store energy. In fungi, it eliminates the membrane charge gradient and interferes with nutrient transport. The collapse of delta, the pH component of the proton motive force, affects bacterial cells. It has been suggested that 2-pyridinethiol-1-oxide is not accumulated in cells and is not destroyed during action on cells but rather acts catalytically. 2-Hydroxybiphenyl ethers effectively inhibit fatty acid synthesis in vivo and the key enzyme of the fatty acid synthase system in vitro. This contradicts early reports on mode of action due to direct disruption of cell membranes. 5-Chloro-2-methyl-4isothiazolin-3-one has been claimed to have multiple modes of action in the inhibition of microorganisms. These include lethal loss of protein thiols by covalent modification of protein molecules through direct electrophilic attack, generation of secondary electrophiles by disulfide exchange and tautomerization to a thioacyl chloride, and intracellular generation of free radicals as a result of the severe metabolic disruption. Early studies on the mode of action of 2-bromo-2nitro-1,3-propanediol using Escherichia coli, Staphylococcus aureus, and Pseudomonas aeruginosa indicated effects on cell membrane integrity and also on aerobic glucose metabolism. The major finding was the inhibition of thiol-containing enzymes such as glyceraldehyde-3-phosphate dehygrogenase. Studies on cysteine and glutathione showed the ability of 2-bromo-2-nitro1,3-propanediol to oxidize the thiol group to form a disulfide bond and this was postulated as the inhibition mechanism. Later work confirmed this property of 2-bromo-2-nitro-1,3-propanediol, showing that it acted catalytically to oxidize thiol groups under aerobic conditions. In addition, there was evidence that 2-bromo-2-nitro-1,3-propanediol led to the formation of active oxygen species such as superoxide, suggesting interference with the electron transport mechanism within the cell. There have also been reports which do not directly describe mode of action but rather clarify active moiety(s) involved in mode of action of certain groups of biocides. There are many industrial biocides that are synthesized with formaldehyde as one of the starting materials. The question about the role of formaldehyde in mode of action of formaldehyde-adduct

ch11.qxd 10/9/03 6:53 PM Page 156

156

the desk encyclopedia of microbiology

TABLE 11.6 Formaldehyde–adduct biocides with

TABLE 11.7 Biocide mixtures used in metalworking and

formaldehyde as active moiety in mode of actiona

Hexahydro-1,3,5-tris(2-hydroxyethyl)-s-triazine Hexahydro-1,3,5-triethyl-s-triazine 2-[(Hydroxymethyl) amino]-2-methylpropanol 2-[(Hydroxymethyl) amino] ethanol 4,4-Dimethyloxazolidine 3,4,4-trimethyloxazolidine 1,3-Dihydroxymethyl)-5,5-dimethylhydantoin 5-Hydroxymethoxymethyl-L-aza-3,7-dioxabicyclo(3.3.0)octane 5-Hydroxymethyl-L-aza-3,7-dioxabicyclo(3.3.0)octane 5-Hydroxypoly[methyleneoxy(74% C2, 21% C3, 4% C4, 1% C5)] methyl-L-aza-3,7-dioxabicyclo(3.3.0)octane a

From Rossmoore and Sondossi (1988).

hydraulic fluidsa

Hexahydro-1,3,5-tris(2-hydroxyethyl) triazine 2-sodium2-pyridinethiol-1-oxide Sodium dimethyldithiocarbarnate sodium 2-mercaptobenzothiazole 5-Chloro-2-methyl-4-isothiazolin-3-one 2-methyl-4isothiazolin-3-one CuSO4 1,3,5-Hexahydro-tris-(2-hydroxyethyl)-triazine 5-chloro2-methyl-4-isothiazolin-3-one Bisoxazolidine 5-chloro-2-methyl-4-isothiazolin-3-one Formols 5-chloro-2-methyl-4-isothiazolin-3-one Dimethylolurea formols 5-chloro-2-methyl-4-isothiazolin-3-one 1,2-Dibromo-2,4-dicyanobutane 5-chloro-2-methyl-4isothiazolin-3-one a

From Rossmoore (1995b).

biocides was not always clearly addressed. Studies with bacterial strains resistant to formaldehyde and formaldehyde-adduct biocides derived separately from sensitive wild types and concurrent development of cross-resistance among all resistant strains established formaldehyde as the active moiety in mechanism of action of many formaldehyde-adduct biocides. Subsequently, it was shown that high levels of resistance were coupled to high levels of formaldehyde dehydrogenase in resistant cells. Although this does not resolve the problem of mode of action of formaldehyde, it consolidates the mechanism of action question and clearly demonstrates the involvement of formaldehyde as the active moiety in mode of action of many formaldehyde-adduct biocides. Table 11.6 shows some of these formaldehyde-adduct industrial biocides. There are many other adducts that are not included, although partial formaldehyde involvement in mode of action has been suggested.

VII. COMBINATION BIOCIDES (MIXTURES AND FORMULATIONS) Biocides are used in combination for many reasons: (i) to broaden the antimicrobial spectrum; (ii) to minimize physical and chemical incompatibilities; (iii) to minimize toxicity; and (iv) to produce biochemical synergism. In its broadest sense, the subject of interactions among industrial biocides that alter their biological activities would include interactions in the extracellular environment and those within the target organisms. Gathering data on combined modes of action of biocide mixtures is not an easy task. Although many biocide mixtures have been used and are commercially available, a considerable amount of

research will be required to define the biochemical nature of these interactions and their physiological effects on microorganisms. A select number of biocide mixtures primarily used in metalworking and hydraulic fluids are listed in Table 11.7. It should be noted that there are many more mixtures with a variety of applications and different active ingredients. Traditionally, a toxicological interaction has been described as “a condition in which exposure to two or more chemicals results in a quantitatively or qualitatively altered biological response relative to that predicted from the action of a single chemical. Such multiple-chemical exposures may occur simultaneously or sequentially in time and the altered response may be greater or lesser in magnitude” (Murphy, 1980). It has been common practice to classify the quantitative joint action of chemicals including biocides using three general terms: Addition: when the toxic effect produced by two or more biocides in combination is equivalent to that expected by simple summation of their individual effects. Antagonism: when the effect of a combination is less than the sum of the individual effects. Synergism: when the effect of the combination is greater than would be predicted by summation of the individual effects. For biocides, there must be a quantitatively definable effect for each compound involved. Minimal inhibitory concentrations and other dose–response information could be used to construct a graphic representation (isobologram) to show additive, synergistic, and antagonistic interactions in biocide mixtures. This could easily be applied to combinations of any number of agents. For a combination of more than three agents, no graphic construction is possible; however, the interaction index

ch11.qxd 10/9/03 6:53 PM Page 157

157

biocides could be calculated mathematically to describe additivity, antagonism, and synergism. Because development costs of new biocides are estimated between $10 and $15 million, the introduction of new biocidal compounds to the market is difficult. This cost escalation is due to increasing legislative requirements and concerns regarding environmental impact. Therefore, the use of registered biocides in combinations that yield synergistic activity is an attractive alternative. There is an extensive range of biocides from which to formulate mixtures, with substantially reduced initial screening costs and possibly an easier registration process if the active ingredients are well known and individually registered for particular end use(s). However, there are concerns regarding the toxicity and environmental impact of biocide mixtures.

VIII. RESTRICTIONS ON USE AND REGULATION The fundamental requirements for industrial biocides suitable for protection of material in the early days of use were effective and aggressive antimicrobial activities, broad spectrum of activity, stability and persistence, and economical feasibility. With the constant expansion of biocide use and number, there has been increasing concern about their impact on human and environmental health. In approximately the past two decades, regulatory agencies have put in place restrictions for the application and selection of industrial biocides. The new requirements include spectrum of activity and effectiveness according to the category of application, stability relevant to application, very low human toxicity according to required toxicological data, very low environmental impact (ecotoxicity), and economical feasibility of use. This has produced a stream of new regulations. The EPA registers and regulates antimicrobial pesticides including industrial biocides under the Federal Insecticide, Fungicide and Rodenticide Act (FIFRA). To register an industrial biocide, manufacturers of such products must meet EPA requirements to show that: (i) the product will not cause unreasonable adverse effects on human health and the environment; and (ii) the product labeling and composition comply with requirements of FIFRA. Since 1996, the antimicrobials division within the Office of Pesticide Programs (OPP) has been responsible for all activities related to regulating antimicrobial pesticides. The OPP reviews submitted detailed and specific information on the chemical composition of the product,

efficacy data against specific intended microorganisms, support of directions for use on the label, appropriate labeling for safe and effective use, and extensive toxicological data and hazards associated with the product use. Title 40 CFR, Part 158, explicitly outlines the data requirements for antimicrobial pesticides. Further amendments are being considered to Part 158. These data requirements are for tiered human health and exposure data requirements for nonfood uses, product chemistry, and toxicology. Explicit tiered testing approaches for environmental fate and effects have been developed for antimicrobials, wood preservatives, antifoulants, and algicides. Specifically, data are required for end-use antimicrobial products, including data on end-use formulation, active ingredient, product chemistry information, residue chemistry, efficacy, toxicity, environmental fate, and ecotoxicity. Data are required to assess acute toxicity, chronic and subchronic toxicity, developmental toxicity, reproductive toxicity, mutagenicity, neurotoxicity, metabolic effects, and immunological effects. Toxicology test requirements are set out in tiers based on general requirements and risk assessment. It should be noted that the EPA may require additional data on a caseby-case basis in order to conduct a risk assessment for the product. To ensure that biocides meet current scientific and regulatory standards, EPA is reviewing older pesticides, including biocides, under FIFRA. This process is called reregistration and considers human health and ecological effects of pesticides. Biocides registered prior to November 1984 are being processed first. Numerous documents containing information on biocides could be found on EPA web sites.

IX. PROBLEMS ASSOCIATED WITH BIOCIDE USE It should be kept in mind that biocides are all toxic by definition and most are also corrosive. For decades there have been concerns regarding toxicity issues of biocides, even though biocides must be registered for specific use and require a battery of data submissions which include human and environmental toxicological profiles. Recognition for the need to control risks from biocides has come from scientists in academia, regulatory agencies, and industry, and there is a need for comprehensive information and new data especially on the biocide mixtures and biocide-containing formulations. One of the most important problems based on toxicity concerns is the international

ch11.qxd 10/9/03 6:53 PM Page 158

158

the desk encyclopedia of microbiology

recognition of the registration of biocides. Acceptance of toxicity data, including ecotoxicity of biocides, by the regulatory bodies in different countries has been of great concern to multinational producers of biocides. In addition to the regulatory concerns associated with biocide use, there are problems beyond the obvious toxicity of biocides that have to be considered. Like antibiotics, when biocides interact with mixed populations of microorganisms they kill all susceptible organisms and promote the selection of resistant populations. The level of resistance may be intrinsic, developed by mutation, or could even be acquired by gene exchange. It should be very clear from the previous discussion that the ultimate result of biocide use is selection of resistant populations. Although resistance is a relative term, a resistant organism is one that is not affected by biocide concentrations used to control microorganisms regularly found in a system. There are many review articles on the development of resistance to antimicrobial agents (including antibiotics). These reviews and numerous other research articles have classified organisms according to their intrinsic levels of resistance to antimicrobials, mechanism(s) of resistance development, and concepts such as phenotypic and genotypic resistance to antimicrobial agents. Regarding intrinsic resistance to biocides, in general, it can be stated that gram-negative bacteria are more resistant to biocides than are gram-positive bacteria. In addition to this generalization, there are specific examples of intrinsic resistance of microorganisms, such as mycobacterial, peudomonad, and fungal species, to biocides. Microorganisms may also gain the capacity to resist the biocide by the acquisition of gene function(s). These gene functions are mostly concerned with inactivation or modification of the biocide, efflux systems, specification of a new target, or enzymatic modification of the target. Microorganisms could also simply persist in the presence of the biocide. This phenomenon may result from mutation or temporary resistance due to gene regulatory events or phenotypic changes. It is accepted that the general resistance mechanisms producing biocide resistance in microorganisms are the same mechanisms found in antibiotic resistance. Excessive use of biocides may produce organisms with a non-specific mechanism(s) of cross-resistance to other biocides and, most important, to antibiotics (double resistance to biocides and to antibiotics). Resistance development to industrial biocides has received much attention, and numerous published research articles and reviews have been devoted to this subject. There should be no doubt that the inappropriate and excessive use of biocides often results

in selection of resistant populations. This usually includes unintentional under-dosing and, more important, misunderstanding of the kinetics of biocide effects and the dynamics of the system treated with biocides. With regard to the resistance categories mentioned previously, there are some points worth noting. When the mechanism of resistance involves inactivation or modification of a biocide and the organism(s) becomes the dominant microbial population in the biocidetreated system, other less susceptible populations may also be protected. There have been reports indicating survival of biocide-sensitive organisms in the presence of resistant populations in metalworking fluids. The addition of biocides (intermittent slug dosing or continuous addition with pumps) to a system may result in an unintended biocide buildup in which the extremely hostile toxic environment kills virtually all the microbial populations usually found in systems treated with recommended doses of biocide (gramnegative bacteria, especially Pseudomonas species). This results in an environment, although hostile, with little or no competition. Microbial populations which could tolerate these conditions will eventually colonize this environment and flourish. There are reports indicating the isolation of unusual organisms in the presence of biocide several times in excess of the recommended dose. The evidence strongly suggests that these organisms have an extremely low permeability to hydrophilic biocides used in the system. This scenario may be involved in recent outbreaks of hypersensitivity pneumonitis associated with exposure to metalworking fluid aerosols. The evidence suggests that hypersensitivity pneumonitis has occurred where “atypical” flora have predominated in metalworking fluids. Although the microbiological origin of hypersensitivity pneumonitis is strongly suspected, the involvement of biocides and other constituents of the fluids has not been excluded. There have been many reports on allergic contact dermatitis among workers exposed to biocide-treated components of industrial systems and contact sensitization to products containing biocides at the consumer end. These types of reports include most of the frequently used industrial biocides.

X. RECENT DEVELOPMENTS AND CONCERNS OVER RESISTANCE DEVELOPMENT In July 2001, the OPP of the EPA announced purely voluntary pesticide resistance management labeling guidelines based on mode/target site of action for

ch11.qxd 10/9/03 6:53 PM Page 159

159

biocides agriculturally used herbicides, fungicides, bactericides, and insecticides. This, Pesticide Registration Notice to registrants, concerns pesticide products intended for general agricultural use at this time. One could envision similar notices for all biocides in the future. This possibility becomes even more realistic if new studies establish the existence of cross-resistance between biocides and antibiotics. This aspect of cross-resistance could be approached considering two scenarios: (a) antibiotic resistant organism is also resistant to biocides; and (b) biocide resistant organism also exhibits cross-resistance to antibiotics. At this time, there is no evidence that antibiotic resistant microorganisms are more resistant to biocides, excluding the intrinsic resistance characteristics of some organisms. There have been reports studying the susceptibility of antibiotic resistant bacteria to disinfectants that suggest the above conclusion. With the ever increasing use of antimicrobials in consumer products and biocides to control biodeterioration, there is a recent but growing and legitimate concern over cross-resistance of biocide resistant organisms to antibiotics. It is possible that selection of microorganisms with extremely low permeability as a result of biocide treatment may indeed produce populations less accessible to antibiotics. Although similarities between the mechanisms of resistance development to biocides and antibiotics exist, efflux pumps, thickening of the cell wall, outer membrane alterations in gram-negative bacteria, it has not yet been established that biocide use will result in selection of an antibiotic resistance trait in the same organisms.

XI. HANDLING OF BIOCIDES It should be clear that “biocides” are poisons and are designed to kill living cells. Therefore, direct contact with concentrated biocides (undiluted product) should be avoided. The Material Safety Data Sheet (MSDS) for a biocide should be studied carefully and appropriate personal protective equipment (protective clothing, glasses or face shield, gloves, and other protective material) should be used when handling biocides. If there is any likelihood of exposure through inhalation, then respiratory protection should be included as protective equipment. An emergency action plan should be in place to deal with accidental spill. This should include the decontamination and deactivation of small spills based on recommendations of the biocide manufacturer and in case of major spills, local authorities should be contacted immediately.

Since biocides are considered hazardous materials, disposal of biocides should be done according to regulations and through licensed disposal contractors. Biocide concentrates should not be allowed to enter surface waters, ground waters, wastewater treatment systems or the environment in general.

BIBLIOGRAPHY Allsopp, C., and Allsopp, D. (1983). An updated survey of commercial products used to protect material against biodeterioration. Int. Biodeterioration Bull. 19, 99–145. Block, S. (ed.) (1991). “Disinfection, Sterilization, and Preservation,” 4th ed. Leo & Feiger, Philadelphia, PA. Bravery, A. F. (1992). Preservation in construction industry. In “Principles and Practice of Disinfection, Preservation and Sterilization” (A. D. Russell, W. B. Hugo, and G. A. J. Ayliffe, eds.), 2nd ed., pp. 437–458. Blackwell, London. Denyer, S. P. (1990). Mechanisms of action of biocides. Int. Biodeterioration 29, 89–100. Downey, A. (1995). The use of biocides in paint preservation. In “Handbook of Biocide and Preservative Use” (H. W. Rossmoore, ed.), pp. 254–266. Chapman & Hall, New York. Eagon, R. G. (1995). Paper, pulp and food grade paper. In “Handbook of Biocide and Preservative Use” (H. W. Rossmoore, ed.), pp. 83–95. Chapman & Hall, New York. Hueck, H. J. (1968). The biodeterioration of materials—an appraisal. In “Biodeterioration of Materials 6–12.” Elsevier, London. Hugo, W. B. (1992). Historical introduction. In “Principles and Practice of Disinfection, Preservation and Sterilization” (A. D. Russell, W. B. Hugo, and G. A. J. Ayliffe, eds.), 2nd ed., pp. 3–6. Blackwell, London. Hugo, H. W., and Russell, A. D. (1992). Types of antimicrobial agents. In “Principles and Practice of Disinfection, Preservation and Sterilization” (A. D. Russell, W. B. Hugo, and G. A. J. Ayliffe, eds.), 2nd ed., pp. 7–88. Blackwell, London. Leightley, L. (1995). Biocide use in wood preservation. In “Handbook of Biocide and Preservative Use” (H. W. Rossmoore, ed.), pp. 283–301. Chapman & Hall, New York. Lutey, R. W. (1995). Process cooling water. In “Handbook of Biocide and Preservative Use” (H. W. Rossmoore, ed.), pp. 50–76. Chapman & Hall, New York. McCarthy, B. J. (1995). Biocides for use in the textile industry. In “Handbook of Biocide and Preservative Use” (H. W. Rossmoore, ed.), pp. 238–253. Chapman & Hall, New York. Murphy, S. D. (1980). Assessment of the potential for toxic interactions among environmental pollutants. In “The Principles and Methods in Modern Toxicology” (C. L. Galli, S. D. Murphy, and R. Paoletti, eds.), p. 277. Elsevier/North-Holland Biomedical Press. Murtough, S. M., Hiom, S. J., Palmer, M., and Russell, A. D. (2001). Biocide rotation in the healthcare setting: is there a case for policy implementation? J. Hosp. Infect. 48, 1–6 Rossmoore, H. W. (1995a). Introduction to biocide use. In “Handbook of Biocide and Preservative Use” (H. W. Rossmoore, ed.), pp. 1–17. Chapman & Hall, New York. Rossmoore, H. W. (1995b). Biocides for metalworking lubricants and hydraulic fluids. In “Handbook of Biocide and Preservative

ch11.qxd 10/9/03 6:53 PM Page 160

160

the desk encyclopedia of microbiology

Use” (H. W. Rossmoore, ed.), pp. 133–156. Chapman & Hall, New York. Rossmoore, H. W., and Sondossi, M. (1988). Application and mode of action of formaldehyde condensate biocides. Adv. Appl. Microbiol. 33, 233–277. Russell, A. D., Hugo, W. B., and Ayliffe, G. A. J. (eds.) (1992). “Principles and Practice of Disinfection, Preservation and Sterilization,” 2nd ed. Blackwell, London. Rutala, W. A., Stiegel, M. M., Sarabi, F. A., and Wber, D. J. (1997). Susceptibility of antibiotic-susceptible and antibiotic-resistant hospital bacteria to disinfectants. Infect. Control Hosp. Epidemiol. 18, 417–421. Sharpell, F. (1980). Industrial use of biocides in processes and products. Dev. Ind. Microbiol. 21, 133–140.

WEBSITES U.S. Environmental Protection Agency. Tolerance Reassessment and Registration http://www.epa.gov/pesticides/reregistration/ U.S. Environmental Protection Agency, Office of Pesticide Programs http://www.epa.gov/pesticides/about/index.htm European Chemicals Bureau, Links to regulations, assessment procedures, documents http://ecb.jrc.it/ OECD (Organisation for Economic Co-operation and Development) website, Links to regulations in various countries and international organizations on biocides http://www1.oecd.org/ehs/biocides/

ch12.qxd 10/9/03 6:54 PM Page 161

12 Biofilms and biofouling Karen T. Elvers1 and Hilary M. Lappin-Scott University of Exeter

heterogeneous structure and composition. Although bacteria may attach to surfaces in minutes, biofilms can take hours or days to develop. Biofouling refers to the damage caused by biofilms to surfaces. The combination of growth processes, the production of metabolites, and the physical presence of the biofilm can damage the surface and reduce its efficiency or effectiveness. Early studies on the significance of bacterial adhesion to surfaces emerged from the work of Claude ZoBell in the 1930s. Since this initial research, it is now known that bacterial adhesion is widespread in the environment and the subject of biofilms constitutes an extensive field within microbiology. In addition, modern biofilm studies have shown that biofouling affects a surprisingly wide range of materials. This article contains examples of biofilms in medical and industrial situations and describes how particular biofilms cause damage and resist treatment.

GLOSSARY biofilm Complex association or matrix of microorganisms and microbial products attached to a surface. biofouling Damage caused to a surface by microorganisms attached to a surface. consortia Spatial grouping of bacterial cells within a biofilm in which different species are physiologically coordinated with each other, often to produce phenomenally efficient chemical transformations. planktonic Free-floating bacteria living in the aqueous phase and not associated with a biofilm. sessile Bacteria living within a biofilm. Biofilms are generally described as consisting of the cells of microorganisms immobilized at a substratum, attached to a surface, and frequently embedded in an extracellular polymer matrix of microbial origin. In this context, studies have concentrated on bacterial cells rather than on other microorganisms. Bacteria attach firmly to almost any surface submerged in an aquatic environment or bulk liquid. Immobilized bacterial cells within a biofilm are called sessile, whereas those free floating in the aquatic environment are called planktonic. The immobilized cells grow and reproduce, with the newly formed cells attaching to each other as well as to the surface. They also produce extracellular polymers, which extend from the cells to form a matrix of fibers. This matrix entraps debris, nutrients, and other microorganisms establishing a biofilm that has a very

I. BIOFILM FORMATION AND DETACHMENT Many physical, chemical, and biological processes determine biofilm formation. A general description of biofilm formation on a surface begins with the transportation of molecules and small particles to the surface by molecular diffusion to form a conditioning film. This occurs very rapidly or almost instantaneously on exposure of a surface to an aqueous environment. Its effect is to cause changes in the surface properties,

1

Present address: University of Wales Institute.

The Desk Encyclopedia of Microbiology ISBN: 0-12-621361-5

161

Copyright © 2003 Elsevier Ltd All rights of reproduction in any form reserved

ch12.qxd 10/9/03 6:54 PM Page 162

162

the desk encyclopedia of microbiology

including the acquisition of a small negative surface charge and a decrease in hydrophobicity. The composition of the conditioning film varies depending on the surface type but apparently it contains polysaccharides, glycoproteins, and proteins. It is generally uniform in composition and coverage and is an important influence on the subsequent adsorption of bacteria. The growth phase follows the initial phase of biofilm formation. Fluid dynamics within the aquatic environment play an important role in determining the transport of bacteria to the surface during the growth phase. Under quiescent flow conditions, bacterial transport is affected by gravitational forces, Brownian motion, or motility. Under laminar flow conditions, bacteria are transported to the surface by diffusion with a significant increase in transport rate if the cells are motile. Under turbulent flow conditions, bacteria are transported to the surface by fluid dynamic forces (inertia, lift, drag, drainage, and downsweeps) which can be enhanced by further increasing turbulence and surface roughness. Furthermore, eddies that develop in turbulent flow are able to propel bacteria to the surface. The Reynolds number, which describes the relative magnitude of inertia to viscous forces, can be used to describe whether a system is laminar or turbulent. Bacteria approaching the surface are subjected to repulsion forces which must be overcome if they are to adsorb to the surface. The outcome of the forces is described by the Derjaguin and Landau; Verwey and Overbeek theory of colloidal stability. The theory postulates two separation distances from the surface where adhesion can occur. Two- and three-step mechanisms have been proposed for the adhesion process, which results in irreversible adsorption usually by the production of exopolysaccharide (EPS). Development of the biofilm includes further attachment, cell growth, cell division, and EPS production resulting in the formation of distinct microcolonies. Mature biofilms then develop by the attraction of more planktonic bacteria and entrapment of inorganic and organic molecules and microbial products, developing a complex consortia within which there is physiological cooperation between different species. This results in increased heterogeneity and the development of chemical microgradients within the biofilm. With time, portions of the biofilm detach and biofilm development reaches a plateau or steady state of development with accumulation equaling loss by detachment. Detachment is defined as the loss of components (biomass) from the biofilm matrix to the bulk liquid and is a means of interaction and cell turnover between the planktonic organisms in the liquid phase and the sessile organisms within the biofilm. This interaction can affect the overall species

distribution. Detachment occurs by erosion, sloughing, and abrasion. It can be caused by several factors: the action of polymerases from the biofilm organisms, the result of grazing or predator harvesting by protozoa, the effects of substratum texture and surface chemistry, the production of unattached daughter cells through attached cell replication, and the availability of nutrients. Fluid dynamics is also thought to significantly influence detachment, in which an increase in fluid velocity causes an increase in detachment. There are also artificial methods of detachment which aim to control biofilm growth and biofouling, including chemical treatment (chelants, surfactants, and oxidants) and physical treatments (increased fluid velocity, ultrasound, and scrubbing).

II. EPS AND THE GLYCOCALYX EPS and glycocalyx are terms used to describe the polysaccharide produced by bacterial cells. EPS refers to one of the major components of biofilms, and glycocalyx refers to the polysaccharide matrix surrounding individual cells. EPS has an important role in biofilm structure and function and has a complex physical and chemical nature. Its functions are mostly protective in nature and this is one of the benefits for bacteria in the sessile state. Because the glycocalyx is the outermost component of bacterial cells, this layer mediates virtually all bacterial associations with surfaces and other cells: it dictates location, juxtaposition, and the eventual success in the ecosystem. EPS production may be a direct response to selective pressures in the environment and may protect against desiccation (by binding water molecules) and predation by feeding protozoa. It also provides protection against antimicrobial agents, including antibiotics, biocides, and host defense mechanisms. This defense may occur by means of a physical barrier or through aiding the bacteria to evade phagocytosis. Other advantages for bacteria of EPS derive from its polyanionic nature, which confers on the biofilm some ion-exchange properties that assist entrapment and the concentration of nutrients, the removal of toxins, and the exchange of metabolites within the consortia. Finally, the close proximity of cells within the biofilm allows plasmid transfer and an alteration in phenotypical characteristics as a response to changes in the environment.

III. BIOFILM STRUCTURE Biofilm structure has been studied using many techniques, including transmission electron microscopy,

ch12.qxd 10/9/03 6:54 PM Page 163

biofilms and biofouling scanning electron microscopy (SEM), and confocal scanning laser microscopy (CSLM). SEM has been used extensively to study the surface architecture of biofilms. The resulting images reveal an uneven outer surface topography, with the high resolution achieved by this method allowing individual cells to be clearly distinguished among a condensed matrix. Although this technique provides valuable information regarding the nature of biofilms, it is not entirely useful because it is well-known that the dehydration stages of sample preparation for SEM can destroy the EPS matrix. CSLM allows nondestructive in situ analysis of hydrated biofilms in combination with a wide range of fluorescent compounds. This technique can be used to form 3-D computer reconstructions of biofilms. These show a variable distribution of biomass with bacteria aggregating at different horizontal and vertical sites, with the highest cell densities at the biofilm base or at the top of the biofilm, forming “mushroom,” “cone,” or “stacks” shapes. Where biofilms have developed under turbulent conditions, they form additional structures termed “streamers.” CSLM has also shown that biofilms are highly hydrated and that the total biofilm volume is made up of cell clusters, horizontal and vertical interstitial voids, and conduits beneath the clusters. These clusters and channels produce biofilms of varying depth and structure. Species composition has been shown to be an important determining factor in biofilm structure. Recently, it was suggested that the structural complexity of biofilms is determined by the organisms through signaling molecules. It has been established that a family of diffusible chemical signals (N-acyl homoserine lactones) can regulate the production of virulence determinants and secondary metabolites, in suspended cultures, in a cell density-dependent manner. Also known as quorum sensing, it is thought that this may be important for the formation of biofilms which also contain densely packed cells. Evidence based on a pure culture Pseudomonas aeruginosa biofilm growing in laminar flow by Davies and coworkers (1998) supports this theory. They reported that N-(3oxododecanoyl)-L-homoserine lactone (OdDHL) was required for the biofilm to develop a complex structure by comparing wild-type biofilms with a Lasl defective mutant (Lasl directs the synthesis of OdDHL). This work illustrates the interest in this field, which has a huge potential for biofilm control. However, since quorum sensing is a concentration-dependent phenomenon, it will be strongly influenced by mass transfer processes. It may be expected that quorum sensing will have a greater significance in diffusion-dominated regions such as those found in large cell clusters or channels when bulk liquid flow is very low.

163

IV. MIXED-SPECIES BIOFILMS Much of the understanding of biofilm development, activity, and physiology is derived from studying single cultures of bacteria, with relatively few studies having been done on mixed cultures. There has been little discussion of the significance of other species within biofilms (e.g., bacterial and algal interactions and fungi), despite the fact that they are excellent colonizers of surfaces. Fungi are able to respond by growth at a surface and fungal hyphal slimes may have many of the functions attributed to bacterial EPS. These functions include the anchorage of mycelium to the substrate, retardation of desiccation, and service as a source of support and nutrition. Biofilms of filamentous fungi have been involved in industrial processes, such as the degradation of aromatic pollutants, biofouling of cooling tower timbers, voice prostheses, and photoprocessors.

V. TECHNIQUES FOR BIOFILM ANALYSIS Techniques available to cultivate and study biofilms can be broadly categorized as disruptive and nondisruptive to the biofilm. These include fermentors and sampling devices such as the modified Robbins device (MRD). The MRD contains replaceable surfaces that can be examined (viewed by epifluorescence microscopy and SEM) for viable counts, total carbohydrate, total protein, and metabolic activity. The MRD is versatile in that colonization of different surfaces can be investigated, surface roughness can be controlled, and biocides and antibiotics can be tested. Chemostats are widely used for studying microorganisms under constant environmental conditions over long periods of time. They can be used to cultivate well-defined two- or three-member mixed cultures or those with even more members. Other reports demonstrate the use of a two-stage chemostat system, with the inoculum grown in the first vessel before being passed to the second. This second or test vessel allows parameters to be changed, e.g., addition of biocide and insertion of coupons of differing materials on which the biofilm develops. Chemostats have also been used in combination with the MRD. Other biofilm fermentors include the constant depth film fermentor, which allows the biofilm to accumulate to a preset depth which can be maintained, and the continuous perfused biofilm fermentor, which allows establishment of a biofilm on the underside of a cellulose membrane perfused with sterile fresh medium.

ch12.qxd 10/9/03 6:54 PM Page 164

164

the desk encyclopedia of microbiology

Microscopic techniques include light, epifluorescence and electron microscopy, CSLM and computerenhanced microscopy with image analysis, and Nomarski differential contrast microscopy. Epifluorescence is particularly useful when cells are stained with fluorescent dyes such as acridine orange and propidium iodide, which mark nucleic acids, or those that determine metabolic activity, such as rhodamine and 5-cyano-2,3-ditolyl tetrazolium chloride. CSLM can be used in combination with microelectrodes which, depending on their construction, can measure oxygen, pH, and sulfide gradients within cell clusters. Voids can be visualized by following the movement of fluorescent latex beads. Other techniques include the use of continuous-flow cell cultures with image analysis, Fourier transform infrared spectroscopy, nuclear magnetic resonance, atomic force microscopy, and cryosectioning. The data generated from all these techniques have greatly altered the understanding of biofilms in both pure and mixed cultures. They have shown biofilms as being spatially and temporally heterogeneous systems with microscale variations in architecture, chemistry, microgradients, and reactions to antimicrobial agents.

VI. BIOFILMS AND BIOFOULING IN DIFFERENT ENVIRONMENTS: THEIR CONTROL AND RESISTANCE The phenotypic plasticity of bacteria allows colonization in a wide variety of environments. Adhering bacterial species are inherently different from their planktonic equivalents. In particular, biofilm bacteria are more resistant to medical and industrial control strategies than their planktonic counterparts. Biofilms may be either beneficial or detrimental for their host systems. The following sections discuss the variety and importance of biofilms and biofouling in the context of medical and industrial systems.

A. Biofilms in medical systems Biofilms affecting human health can be divided into the following categories: 1. Biofilms formed on human tissue. These biofilms occur in the healthy body, for example, on teeth, in the digestive tract, and in the female genital tract. They may have a role in prevention of certain infections but can be overgrown by pathogenic microorganisms. 2. Biofilms formed on medical implants within the body. 3. Biofilms formed on surfaces outside the body that may harbor harmful pathogens. Examples of

these surfaces include those in water systems that may harbor potentially pathogenic bacteria such as Legionella sp. and that consequently may be protected from chlorination. Biofilms have also been found on contact lenses and contact lens storage cases, for which bacteria induce severe eye irritation and inflammation and may play a role in persistence of the organisms. In all cases, once established, the biofilm’s resistance to phagocytosis and antibiotics allows the organism within it to continue living after planktonic organisms in the same environment have been killed by treatment. Artificial implants are used for the replacement of diseased or damage body parts, e.g., joint or vascular prostheses. Many temporary devices, (e.g., urinary catheters, intravascular catheters, and endotracheal tubes) are inserted into patients for various lengths of time. Many inert materials are used for such devices, including vitallium, titanium, stainless steel, polyethylene, polymethyl methacrylate, silicone rubber polyttrafluorethylene (Teflon), and polyvinyl chloride. All of these can serve as substrata for bacterial biofilms. A variety of bacteria are involved in the colonization of implants. These include gram-negative organisms (e.g., Pseudomonas aeruginosa and Escherichia coli) and gram-positive bacteria (e.g., Staphylococcus aureus and S. epidermidis). The latter, which are normally found on the skin, possess a high degree of adhesiveness to the prosthetic device surface. In the biofilm these species are protected from the effects of antibiotics and they can act as the disseminating center for infection. In addition, biofilm formation can also lead to malfunction of the device and destruction of adjacent tissue. Biofilms have been the cause of significant problems for patients receiving artificial hearts (Jarvik hearts). In cystic fibrosis, patients’ lungs become chronically infected by EPS-producing strains of P. aeruginosa. This bacterium has also been found frequently in catheterized patients. The production of large amounts of EPS and copious quantities of mucous by P. aeruginosa allows it to cause persistent infections. Isolation of mucoid strains and subsequent subculture results in reversion to non-mucoid colonies. This suggests that the host defense mechanisms must have a selective effect in favor of the mucoid strains. Treatment with antibiotics further selects for the mucoid strains. The mucous and EPS protects P. aeruginosa from attack by antibiotics, surfactants, and macrophages. It has been shown that EPS is a large anionic hydrated matrix that can partition charged molecules, preventing them from reaching the bacterial cell. Silicone is a material that is widely used for tubing, catheters, mammary and testicular implants, and

ch12.qxd 10/9/03 6:54 PM Page 165

biofilms and biofouling voice prostheses. Voice prostheses become colonized rapidly by mixed biofilms of bacteria and yeasts and these devices must be removed and replaced frequently before infection can be eradicated. It has been shown by SEM that voice prostheses become damaged by the yeast cells, which grow under the silicone surface. Treatment of infections for short-term devices, such as urinary and intravenous catheters, consists of their immediate removal followed by administration of antibiotics to the patient. Urinary tract infections are most commonly caused by E. coli, Proteus mirabilis, Enterococcus, and Streptococcus spp., found in the gastrointestinal tract, and by pathogens directly transmitted through sexual activity. These infections include acute and chronic cystitis, struvite urolithiasis, chronic prostatitis, and catheter-associated infections. Once the microorganisms are established, they adopt the biofilm mode of growth. The bladder resists infection by the periodic passing of urine, which washes out unattached pathogens, and by sloughing of colonized uroepithelial cells on the glycosaminoglycan (GAG) mucous layer. The GAG layer is a very thin cover on the cell epithelium of the bladder that physically shields the bladder from surface pathogens. Catheter-associated infections increase by approximately 10% each day the catheter is in place. The organisms initially colonize the external surfaces, form a biofilm, and ascend into the bladder where the biofilm can act as a source of infection for the bladder and kidneys. Mineralization can also occur, which can reduce the diameter or block the catheter. Frequent replacement of catheters would reduce infections but is not always practical. Methods to control biofilm growth on catheters are being investigated and include the development of materials that block or kill adherent organisms. These methods include altering the hydrophobicity of polymers and the incorporation of disinfectants and antibiotics in the design of the implant. Surfaces within the mouth become readily colonized with bacterial deposits, forming a biofilm, usually called dental plaque. By attaching to the teeth or dental implants, the biofilm helps to prevent colonization of the mouth by pathogenic bacteria. Although dental plaque forms naturally without good oral hygiene, it can be a source of dental caries or periodontal disease. The attached organisms obtain nutrients from the ingested food, saliva, and gingival crevice fluid found between the teeth and gums: It is thought that most of the nutrients are derived from the host rather than the from the host’s diet. Environmental factors that contribute to plaque formation are an optimum temperature of 35 or 36 C and a neutral pH. The pH can become more acidic

165

when carbohydrates are metabolized or more alkaline during an inflammatory host response. These local changes in pH can lead to shifts in the colonized species. The resident microflora of dental plaque is extremely diverse and consists of gram-positive and -negative bacteria. Few are truly aerobic; most are facultatively or obligately anaerobic. These include Streptococcus, Neisseria, Actinomyces, Lactobacillus, Corynebacterium, and Fusobacterium species, but there are many others and not all are culturable in the laboratory. Sometimes, a particular species will colonize a preferred habitat in the oral cavity, e.g., Streptococcus mutans colonizes the occlusal fissures. Formation of dental plaque begins by the adsorption of a proteinaceous conditioning film or acquired pellicle which is composed of albumin, lysozyme, glycoproteins, and lipids from saliva and gingival cervicular fluid. The first colonizers are streptococci and actinomycetes, which rapidly divide to form microcolonies that quickly change into a confluent film of varying thickness. Species diversity increases with the attachment of rods and filaments and layering that is attributable to bacterial succession. Unusual combinations of bacteria such as “corn-cobs” (gram-positive filaments covered by cocci), “rosettes” (cocci covered by small rods), or “bristle brushes” (large filaments surrounded by rods) are seen under SEM. If plaque formation continues undisturbed for weeks, its composition will vary with location on the teeth. Different environmental conditions exist on and between the teeth, resulting in different chemical gradients and shear forces. The bacterial communities are thought to form as a result of short-range specific molecular interactions between the bacterial cell adhesions of primary colonizers and host receptors in the conditioning film, the attachment of secondary colonizers to primary colonizers (coaggregation) and EPS synthesis and growth. Coaggregation has a major role in forming the distinct patterns in plaque. Dental caries are formed as a result of the localized dissolution of the tooth enamel by acids produced by metabolism of carbohydrates, lowering the pH and favoring the growth of mutans streptococci and lactobacilli. Periodontal diseases occur when the supporting tissues of the teeth are attacked by obligately anaerobic gram-negative rods, filaments, or spiral-shaped bacteria. Prevention of dental plaque is by efficient oral hygiene (brushing and flossing can almost completely prevent plaque-mediated diseases), fluoridization of drinking water, and the addition of antiplaque or antimicrobial agents to toothpastes and mouthwashes. Biofilms are central to the survival of bacteria when they are attacked by the normal host immune system or antibiotics. Gram-positive and -negative bacteria

ch12.qxd 10/9/03 6:54 PM Page 166

166

the desk encyclopedia of microbiology

activate the complement system, with the major components causing this reaction being peptidoglycan and lipopolysaccharide, respectively. Activation of complement would eradicate the serum-sensitive bacteria but may also react to live or dead bacteria or bacterial fragments. Continual production of complement may destroy the tissues. Studies on the immune response to biofilms have concentrated on P. aeruginosa and infection in cystic fibrosis patients. It was shown that biofilm-grown P. aeruginosa was able to resist complement action. The biofilm bacteria activated the complement system to a lesser extent than did the planktonic bacteria. However, some fragments of the activated complement were deposited on the biofilm contributing to chronic inflammation.

B. Biofilms in industrial systems Biofilm formation in industries involves the ability of the biofilm to act as a reservoir for potential pathogens and in instances in which the biofilm causes surface damage. The degree of contamination in a system is often measured by planktonic counts that fail to detect the presence of sessile bacteria and this leads to incorrect conclusions regarding the level of pathogens in the system. In the water industry, biofilms form on the pipe surfaces that connect the consumer to the supply. The level of biofilm formation is difficult to monitor in these situations, and levels of coliforms, pseudomonads, and Flavobacterium sp., detaching from biofilms, have been reported as being higher than permitted the levels. Treatment with chlorine does not control the problem because the biofilm bacteria are protected from the disinfectant. There is also evidence of accelerated material deterioration (corrosion) due to biofilm accumulation in water distribution pipes. Excessive biofilm accumulation in porous media, on heat exchanger surfaces, and in storage tanks is responsible for reduced efficiency of heat transfer and reduction of flow rates. Transfer of heat is reduced because the thick surface growth physically prevents an efficient heat exchange between the liquid phase and the cooling surface. Biofilms on ship hulls consist of diatoms, singlecelled algae, and bacteria. This biofilm growth reduces speed in the water and increases fuel consumption. As the biofilm develops, the hull must be physically cleaned, which results in further expense. In an attempt to control this growth, antifouling paints have been used. These are not always effective; although good at preventing colonization of small animals, they do not stop bacterial growth. Corrosion in marine environments on structures such as oil rigs is a result of biofilms that contain

sulfate-reducing or acid-producing bacteria. These microorganisms create anodes and cathodes on metal surfaces. This unequal distribution of ions causes electrical currents, resulting in metal loss. Ideal anaerobic conditions for the biofouling action of sulfatereducing bacteria are found around oil rig legs. Biofilms in the food processing industry may form on food contact and non-contact surfaces. These biofilms may contain food spoilage and pathogenic micro-organisms which affect the quality and safety of the food product by reducing shelf life and increasing the probability of food poisoning. Stainless steel is commonly used as a food contact surface because it is chemically and physiologically stable at a variety of processing temperatures, easy to clean, and has a high resistance to corrosion. Food processing environments provide a variety of conditions that favor the formation of biofilms, e.g., flowing water, suitable attachment surfaces, ample nutrients (although possibly sporadic), and the raw materials or the natural flora providing the inocula; however, these conditions may be extremely varied. Time available for biofilm development is relatively short; for example, the production line may run for a few hours before cleaning. Various preventative and control strategies, such as hygienic plant lay-out and design of equipment, choice of materials, correct use and selection of detergents and disinfectants, coupled with physical methods, can be applied for controlling biofilm formation on food contact surfaces. Biofilms have been shown to occur in many food environments. In the dairy industry, pasteurization ensures the destruction of pathogens and most vegetative organisms within raw milk. However, heatresistant organisms and spores survive and may form biofilms that could result in post-pasteurization contamination. Biofilms have been found on gaskets and “O” rings from the pipes within the dairy industry. Pathogens, Listeria and Bacillus sp., have also been isolated from food contact and environmental surfaces in the dairy industry. Biofilms have been found in pipes of breweries, on rubber seals, conveyor belts, and in waste-water pipes, and in flour mills and malt houses. There is also evidence of microbial adherence to environmental surfaces during poultry processing. This could result in crosscontamination during the slaughter process, which may play an important role in product contamination with Listeria, Campylobacter, and Staphylococcus aureus.

BIBLIOGRAPHY Costerton, J. W., Lewandowski, Z., Caldwell, D. E., Korber, D., and Lappin-Scott, H. M. (1995). Microbial biofilms. Annu. Rev. Microbiol. 49, 711–745.

ch12.qxd 10/9/03 6:54 PM Page 167

167

biofilms and biofouling Costerton, J. W., Stewart, P. S., and Greenberg, E. P. (1999). Bacterial biofilms: a common cause of persistent infections. Science 284(5418), 1318–1322. Davey, M. E., and O’Toole, G. A. (2000). Microbial biofilms: from ecology to molecular genetics. Microbiol. Mol. Biol. Rev. 64, 847–867. Davies, D., Parsek, M. R., Pearson, J. P., Iglewski, B. H., Costerton, J. W., and Greenburg, E. P. (1998). The involvement of cell-to-cell signals in the development of a bacterial biofilm. Science 280, 295–298. Lappin-Scott, H. M., and Costerton, J. W. (eds.) (1995). “Microbial Biofilms.” Cambridge Univ. Press, Cambridge, UK. O’Toole, G., Kaplan, H. B., and Kolter, R. (2000). Biofilm formation as microbial development. Annu. Rev. Microbiol. 54, 49–79. Stewart, P. S. (2001). Multicellular resistance: biofilms. Trends Microbiol. 9(5), 204.

Stewart, P. S. and Costerton, J. W. (2001). Antibiotic resistance of bacteria in biofilms. Lancet 358(9276), 135–138. Wackett, L. P. (2001). Microbioal biofilms. Environ. Microbiol. 3(2), 144. ZoBell, C. E. (1937). The influence of solid surface upon the physiological activities of bacteria in sea water. J. Bacteriol. 33, 86.

WEBSITES The website for the Center for Biofilm Engineering, U. of Montana http://www.erc.montana.edu/ American Society for Microbiology, Biofilm image collection http://www.asmusa.org/edusrc/biofilms/index.html

ch13.qxd 10/9/03 6:57 PM Page 168

13 Biological warfare James A. Poupard and Linda A. Miller SmithKline Beecham Pharmaceuticals

The Most General Concept of Biological Warfare involves the use of any biological agent as a weapon directed against humans, animals, or crops with the intent to kill, injure, or create a sense of havoc against a target population. This agent could be in the form of a viable organism or a metabolic product of that organism, such as a toxin. This article will focus on the use of viable biological agents because many of the concepts relating to the use of toxins are associated more with chemical warfare. The use of viable organisms or viruses involves complex issues that relate to containment. Once such agents are released, even in relatively small numbers, the focus of release has the potential to enlarge to a wider population due to the ability of the viable agent to proliferate while spreading from one susceptible host to another.

GLOSSARY biological warfare Use of microorganisms, such as bacteria, fungi, viruses, and rickettsiae, to produce death or disease in humans, animals, or plants. The use of toxins to produce death or disease is often included under the heading of BWR (U.S. Army definition, included in U.S. Army report to the Senate Committee on Human Resources, 1977). biological weapons Living organisms, whatever their nature, which are intended to cause disease or death in man, animals, or plants and which depend for their effects on their ability to multiply in the person, animal, or plant attacked [United Nations definition, included in the report of the secretary general titled “Chemical and Bacteriological (Biological) Weapons and the Effects of Their Possible Use,” 1969]. genetic engineering Methods by which the genomes of plants, animals, and microorganisms are manipulated: includes but is not limited to recombinant DNA technology. recombinant DNA technology Techniques in which different pieces of DNA are spliced together and inserted into vectors such as bacteria or yeast. toxin weapon(s) Any poisonous substance, whatever its origin or method of production, which can be produced by a living organism, or any poisonous isomer, homolog, or derivative of such a substance (U.S. Arms Control and Disarmament Agency definition, proposed on August 20, 1980). The Desk Encyclopedia of Microbiology ISBN: 0-12-621361-5

I. INTRODUCTION As the twentieth century draws to a close three events mark significant alterations in the concept of biological warfare (BW): the end of the Cold War, the open threat of using BW agents in the Gulf War, and the realization that the developed world is quite susceptible to attack by radical terrorists employing BW agents. These events mark major changes in the concept of BW and transform the subject from one which was once limited to the realm of political and military policy makers to one that must be considered by a wide range of urban disaster planners, public health officials, and the general public. BW is a complex

168

Copyright © 2003 Elsevier Ltd All rights of reproduction in any form reserved

ch13.qxd 10/9/03 6:57 PM Page 169

biological warfare subject that is difficult to understand without a basic knowledge of a long and convoluted history. BW can be traced to ancient times and has evolved into more sophisticated forms with the maturation of the science of bacteriology and microbiology. It is important to understand the history of the subject because often there are preconceived notions of BW that are not based on facts or involve concepts related more to chemical rather than biological warfare. Many of the contemporary issues relating to BW deal with Third World conflicts, terrorist groups, or nonconventional warfare. An understanding of these issues is important because many of the long-standing international treaties and conventions on BW were formulated either in an atmosphere of international conflict or during the Cold War period of international relations. Many of the classic issues have undergone significant alteration by recent events. The issue of BW is intimately bound to such concepts as offensive versus defensive research or the need for secrecy and national security. It is obvious that BW will continue to demand the attention of contemporary students of microbiology and a wide range of specialists during the twenty-first century.

II. HISTORICAL REVIEW A. 300

BC

to 1925

Many early civilizations employed a crude method of warfare that could be considered BW as early as 300 BC when the Greeks polluted the wells and drinking water supplies of their enemies with the corpses of animals. Later, the Romans and Persians used these same tactics. All armies and centers of civilization need palatable water to function, and it is clear that well pollution was an effective and calculated method for gaining advantage in warfare. In 1155 at a battle in Tortona, Italy, Barbarossa broadened the scope of BW by using the bodies of dead soldiers as well as animals to pollute wells. Evidence indicates that well poisoning was a common tactic throughout the classical, medieval, and Renaissance periods. In modern times, this method has been employed as late as 1863 during the Civil War by General Johnson, who used the bodies of sheep and pigs to pollute drinking water at Vicksburg. Catapults and siege machines in medieval warfare were a new technology for delivering biological entities. In 1422 at the siege of Carolstein, catapults were used to project diseased bodies over walled fortifications, creating fear and confusion among the people under siege. The use of catapults as weapons was well established by the medieval period, and projecting diseased bodies over walls was an effective strategy employed by

169

besieging armies. The siege of a well-fortified position could last for months or years, and it was necessary for those outside the walls to use whatever means available to cause disease and chaos within the fortification. This technique became commonplace, and numerous classical tapestries and works of art depict diseased bodies or the heads of captured soldiers being catapulted over fortified structures. In 1763, BW took a significant turn from the crude use of diseased corpses to the introduction of a specific disease, smallpox, as a weapon in the North American Indian wars. It was common knowledge at the time that the Native American population was particularly susceptible to smallpox, and the disease may have beeen used as a weapon in earlier conflicts between European settlers and Native Americans. In the spring of 1763, Sir Jeffrey Amherst, the British commander-in-chief in North America, believed the western frontier, which ran from Pennsylvania to Detroit, was secure, but the situation deteriorated rapidly during the next several months. The Indians in western Pennsylvania were becoming particularly aggressive in the area near Fort Pitt (Pittsburgh). It became apparent that unless the situation was resolved, western Pennsylvania would be deserted and Fort Pitt isolated. On June 23, 1763, Colonel Henry Bouquet, the ranking officer for the Pennsylvania frontier, wrote to Amherst, describing the difficulties Captain Ecuyer was having holding the besieged Fort Pitt. These difficulties included an outbreak of smallpox among Ecuyer’s troops. In his reply to Bouquet, Amherst suggested that smallpox be sent among the Indians to reduce their numbers. This welldocumented suggestion is significant because it clearly implies the intentional use of smallpox as a weapon. Bouquet responded to Amherst’s suggestion by stating that he would use blankets to spread the disease. Evidence indicates that Amherst and Bouquet were not alone in their plan to use BW against the Indians. While they were developing a plan of action, Captain Ecuyer reported in his journal that he had given two blankets and handkerchiefs from the garrison smallpox hospital to hostile chiefs with the hope that the disease would spread. It appears that Ecuyer was acting on his own and did not need persuasion to use whatever means necessary to preserve the Pennsylvania frontier. Evidence also shows that the French used smallpox as a weapon in their conflicts with the native population. Smallpox also played a role in the American Revolutionary War, but the tactics were defensive rather than offensive: British troops were vaccinated against smallpox, but the rebelling American colonists were not. This protection from disease gave the British an advantage for several years, until Washington ordered vaccination of all American troops.

ch13.qxd 10/9/03 6:57 PM Page 170

170

the desk encyclopedia of microbiology

It is clear that by the eighteenth century BW had become disease oriented, even though the causative agents and mechanisms for preventing the spread of diseases were largely unknown. The development of the science of bacteriology in the nineteenth and early twentieth centuries considerably expanded the scope of potential BW agents. In 1915, Germany was accused of using cholera in Italy and plague in St. Petersburg. Evidence shows that Germany used glanders and anthrax to infect horses and cattle, respectively, in Bucharest in 1916 and employed similar tactics to infect 4500 mules in Mesopotamia the next year. Germany issued official denials of these accusations. Although there apparently was no large-scale battlefield use of BW in World War I, numerous allegations of German use of BW were made in the years following the war. Britain accused Germany of dropping plague bombs, and the French claimed the Germans had dropped disease-laden toys and candy in Romania. Germany denied the accusations. Although chemical warfare was far more important than BW in World War I, the general awareness of the potential of biological weapons led the delegates to the Geneva Convention to include BW agents in the 1925 Protocol for the Prohibition of the Use in War of Asphyxiating, Poisonous or Other Gases, and of Bacteriological Methods of Warfare. The significance of the treaty will be discussed in Section III.

B. 1925–1990 The tense political atmosphere of the period following the 1925 Geneva Protocol and the lack of provisions to deter biological weapons research had the effect of undermining the treaty. The Soviet Union opened a BW research facility north of the Caspian Sea in 1929; the United Kingdom and Japan initiated BW research programs in 1934. The Japanese program was particularly ambitious and included experiments on human subjects prior to and during World War II. Two factors were significant in mobilizing governments to initiate BW research programs: (i) continuing accusations regarding BW and (ii) the commitment of resources for BW research by several national adversaries, thus creating insecurity among governments. The presence of BW research laboratories in nations that were traditional or potential adversaries reinforced this insecurity. Thus, despite the Geneva Protocol, it was politically unwise for governments to ignore the threat of BW, and the result was increasingly sophisticated biological weapons. In 1941, the United States and Canada joined other nations and formed national programs of BW research and development. Camp Detrick (now Fort

Detrick) became operational as the center for U.S. BW research in 1943, and in 1947 President Truman withdrew the Geneva Protocol from Senate consideration, citing current issues such as the lack of verification mechanisms that invalidated the underlying principles of the treaty. However, there was no widespread use of BW in a battlefield setting during World War II. BW research, however, continued at an intense pace during and after the war. By the end of the decade, the United States, the United Kingdom, and Canada were conducting collaborative experiments involving the release of microorganisms from ships in the Caribbean. In 1950, the U.S. Navy conducted open-air experiments in Norfolk, Virginia, and the U.S. Army conducted a series of airborne microbial dispersals over San Francisco using Bacillus globigii, Serratia marcescens, and inert particles. Not surprisingly, the intense pace of BW research led to new accusations of BW use, most notably by China and North Korea against the United States during the Korean War. In 1956, the United States changed its policy of “defensive use only” to include possible deployment of biological weapons in situations other than retaliation. During the 1960s, all branches of the U.S. military had active BW programs, and additional open-air dissemination experiments with stimulants were conducted in the New York City subway system. By 1969, however, the U.S. military concluded that BW had little tactical value in battlefield situations, and since it was believed that nuclear weapons dominated the strategic equation the United States would be unlikely to need or use BW. Thus, President Nixon announced that the United States would unilaterally renounce BW and eliminate stockpiles of biological weapons. This decision marked a turning point in the history of BW: Once the U.S. government made it clear it did not consider biological weapons a critical weapon system, the door was opened for negotiation of a strong international treaty against BW. Once military strategists had discounted the value of BW, an attitude of openness and compromise on BW issues took hold, leading to the 1972 Convention on the Prohibition of the Development, Production and Stockpiling of Bacteriological (Biological) and Toxin Weapons and on Their Destruction (see Section III). The parties to the 1972 convention agreed to destroy or convert to peaceful use all organisms, toxins, equipment, and delivery systems. Following the signing of the 1972 treaty, the U.S. government generated much publicity about its compliance activities, inviting journalists to witness destruction of biological weapons stockpiles. The problem of treaty verification beleaguered the 1972 convention. Press reports accusing the Soviet

ch13.qxd 10/9/03 6:57 PM Page 171

171

biological warfare Union of violating the treaty appeared as early as 1975. When an outbreak of anthrax was reported in Sverdlovsk, Soviet Union, in 1979, the United States claimed it was caused by an incident at a nearby Soviet biological defense laboratory that had released anthrax spores into the surrounding community. The Soviet government denied this allegation, claiming the outbreak was caused by contaminated black market meat. BW continued to be discussed in the public media throughout the 1980s. In 1981, reports describing the American “cover-up” of Japanese BW experiments on prisoners of war began to surface in the public and scientific literature. In 1982, The Wall Street Journal published a series of articles on Soviet genetic engineering programs that raised many questions about the scope of Soviet BW activities. The environmental effects of testing biological agents at Dugway Proving Grounds in Utah received considerable press attention in 1988, leading to a debate over the need for such a facility. The 1980s also were characterized by debate over larger issues relating to BW. A public debate in 1986 considered the possible role of biological weapons in terrorism. Scientific and professional societies, which had avoided discussing BW for many years, began considering both specific issues, such as Department of Defense support for biological research, and more general issues, such as adopting ethical codes or guidelines for their members.

better prepared to protect its troops against biological attack. Fortunately, BW was not used during the Gulf War, but the threat of its use provided several significant lessons. Although there was considerable concern that genetic engineering would produce new, specialized biological weapons, most experts predicted that “classical” BW agents, such as anthrax and botulism, would pose the most serious threats to combat troops in Operation Desert Storm. Efforts by the United Nations after the war to initiate inspection programs demonstrated the difficulty of verifying the presence of production facilities for BW agents; these difficulties highlight the need for verification protocols for the BW convention. Verification and treaty compliance are major contemporary BW issues. Following the Gulf War the extent of the intense Iraq BW research programs demonstrated the inadequacy of all estimates and post war verification procedures of Iraq BW capacity. The third significant contemporary development is the realization that urban centers and public facilities are vulnerable to attack by terrorists employing BW agents. Local and national governments are now realizing the extent of this vulnerability and are taking early measures to formulate policies to address these issues. Much work remains to be accomplished in this area.

III. INTERNATIONAL TREATIES

C. 1990 and contemporary developments The last decade of the twentieth century witnessed three significant events that will have long-term effects on developing policies relating to BW. The first event was the demise of the Soviet Union. Most U.S. defensive research was directed to counter potential use by the Soviet Union. As the wall of Soviet secrecy eroded during the 1990s the extent of the Soviet BW program became apparent. There is international concern that many unemployed BW researchers will find work as advisors for developing countries that view BW as a rational defense strategy, especially those countries without nuclear capability or those without restrictive laws against radical terrorist groups. This is an ongoing issue without readily apparent solutions. The second major event was the Gulf War. The open threat by the Iraqi military to use BW agents raised serious concerns and changed attitudes about BW. The plans for Operation Desert Storm included provisions for protective equipment and prophylactic administration of antibiotics or vaccines to protect against potential biological weapons. Many of the critics of the U.S. Biological Defense Research Program (BDRP) were now asking why the country was not

A. The 1925 Geneva protocol The 1925 Geneva Protocol was the first international treaty to place restrictions on BW. The Geneva Protocol followed a series of international agreements that were designed to prohibit the use in war of weapons that inflict or prolong unnecessary suffering of combatants or civilians. The St. Petersburg Declaration of 1868 and the International Declarations Concerning the Laws and Customs of War, which was signed in Brussels in 1874, condemned the use of weapons that caused useless suffering. Two major international conferences were held at the Hague in 1899 and 1907. These conferences resulted in declarations regarding the humanitarian conduct of war. The conference regulations forbid nations from using poison, treacherously wounding enemies, or using munitions that would cause unnecessary suffering. The so-called Hague Conventions also prohibited the use of projectiles to diffuse asphyxiating or deleterious gases. The Hague Conventions still provide much of the definitive law of war as it exists today. The Hague Conventions did not specifically mention BW, due in part to the lack of scientific understanding of

ch13.qxd 10/9/03 6:57 PM Page 172

172

the desk encyclopedia of microbiology

the cause of infectious diseases at that time. The conventions have, however, been cited as an initial source of the customary international laws that prohibit unnecessary suffering of combatants and civilians in war. Although biological weapons have been defended as humanitarian weapons on the grounds that many biological weapons are incapacitating but not lethal, there are also biological weapons that cause a slow and painful death. It can be argued, therefore, that the Hague Conventions helped to set the tone of international agreements on laws of war that led to the 1925 Geneva Protocol. The 1925 Geneva Protocol, formally called the Prohibition of the Use in War of Asphyxiating, Poisonous or Other Gases, and of Bacteriological Methods of Warfare, was opened for signature on June 17, 1925 in Geneva. More than 100 nations signed and ratified the protocol, including all members of the Warsaw Pact and North Atlantic Treaty Organization (NATO). The 1925 Geneva Protocol was initially designed to prevent the use in war of chemical weapons; however, the protocol was extended to include a prohibition on the use of bacteriological methods of warfare. The Geneva Protocol distinguishes between parties and nonparties by explicitly stating that the terms of the treaty apply only to confrontations in which all combatants are parties and when a given situation constitutes a “war.” In addition, many nations ratified the Geneva Protocol with the reservation that they would use biological weapons in retaliation against a biological weapons attack. This resulted in the recognition of the Geneva Protocol as a “no first-use” treaty.

B. The 1972 biological warfare convention International agreements governing BW have been strengthened by the 1972 BW convention, which is officially called the 1972 Convention on the Prohibition of the Development, Production and Stockpiling of Bacteriological (Biological) and Toxin Weapons and on Their Destruction. The convention was signed simultaneously in 1972 in Washington, London, and Moscow and entered into force in 1975. The preamble to the 1972 BW convention states the determination of the parties to the treaty to progress toward general and complete disarmament, including the prohibition and elimination of all types of weapons of mass destruction. This statement places the convention in the wider setting of international goals of complete disarmament. The 1972 BW convention is also seen as a first step toward chemical weapons disarmament. The 1972 BW convention explicitly builds on the Geneva Protocol by reaffirming the prohibition of the use of BW in war. The preamble, although not legally binding, asserts that the goal of the convention is to

completely exclude the possibility of biological agents and toxins being used as weapons and states that such use would be repugnant to the conscience of humankind. The authors of the 1972 convention, therefore, invoked societal attitudes as justification for the existence of the treaty. The 1972 BW convention evolved, in part, from a process of constant reevaluation of the Geneva Protocol. From 1954 to the present, the United Nations has periodically considered the prohibition of chemical and biological weapons. The Eighteen-Nation Conference of the Committee on Disarmament, which in 1978 became the Forty-Nation Committee on Disarmament, began talks in 1968 to ban chemical weapons. At that time, chemical, toxin, and biological weapons were being considered together in an attempt to develop a comprehensive disarmament agreement. However, difficulties in reaching agreements on chemical warfare led to a series of separate negotiations that covered only BW and toxin weapons. The negotiations resulted in the drafting of the 1972 BW convention. The 1972 BW convention consists of a preamble followed by 15 articles. Article I forms the basic treaty obligation. Parties agree never in any circumstance to develop, produce, stockpile, or otherwise acquire or retain the following: 1. Microbial or other biological agents, or toxins whatever their origin or method of production, of types and in quantities that have no justification for prophylactic, protective, or other peaceful purposes. 2. Weapons, equipment, or means of delivery designed to use such agents or toxins for hostile purposes or in armed conflict. Article II requires each party to destroy, or divert to peaceful purposes, all agents, toxins, equipment, and delivery systems that are prohibited in Article I and are under the jurisdiction or control of the party. It also forbids nations from transferring, directly or indirectly, materials specified in Article I and prohibits nations from encouraging, assisting, or inducing any state, group of states, or international organizations from manufacturing or acquiring the material listed in Article I. There is no specific mention of subnational groups, such as terrorist organizations, in the treaty. Articles IV requires each party to the convention to take any measures to ensure compliance with the terms of the treaty. Article IV has been interpreted by some states as the formulation of civil legislation or regulations to ensure adherence to the convention. This civil legislation could regulate activities by individuals, government agencies, universities, or corporate groups.

ch13.qxd 10/9/03 6:57 PM Page 173

173

biological warfare Articles V–VII specify procedures for pursuing allegations of noncompliance with the 1972 BW convention. The United Nations plays an integral part in all the procedures for investigating allegations of noncompliance. According to Article VI, parties may lodge a complaint with the Security Council of the United Nations if a breach of the treaty is suspected. All parties must cooperate with investigations that may be initiated by the Security Council. Article VII requires all parties to provide assistance or support to any party that the Security Council determines has been exposed to danger as a result of violation of the convention. Articles VII–IX are general statements for obligations of the parties signing the protocol. Article X gives the parties the right to participate in the fullest possible exchange of equipment, materials, and scientific or technological information of the use of bacteriological (biological) agents and toxins for peaceful purposes. Article XI allows parties to propose amendments to the convention. The amendments only apply to those states that accept them and enter into force after a majority of the states’ parties to the convention have agreed to accept and be governed by the amendment. Article XII requires that a conference be held 5 years after the entry into force of the BW convention. Article XIV states that the 1972 BW convention is of unlimited duration. A state party to the treaty is given the right to withdraw from the treaty if it decides that extraordinary events, related to the subject matter of the convention, have jeopardized the supreme interests of the country. This article also opens the convention to all nations for signature. Nations that did not sign the convention before its entry into force may accede to it at any time.

TABLE 13.1 Representative organisms regulated by the CDC Bacteria Bacillus anthracis Brucella abortus, melitensis, suis Burkholderia mallei, pseudomallei Clostridium botulinum Francisella tularensis Yersinia pestis

Rickettsiae Coxiella burnetii Rickettsia prowazekii Rickettsia rickettsii

D. Additional U.S. laws and acts The following U.S. laws have been enacted since 1989 that impact on BW: ●

C. Review conferences ●

The 1972 convention contained a stipulation that a conference be held in Geneva 5 years after the terms of the convention entered into force. The purpose of the conference was to review the operation of the convention and to ensure that the purposes of the convention were being realized. The review was to take into account any new scientific and technological developments that were relevant to the convention. The first review conference was held in Geneva in 1980. Several points contained in the original convention were clarified at this conference. The second review conference was held in 1986, and a third was held in 1991. There is general agreement that these conferences and the one that followed serve a definite function in solving contemporary problems that need clarification based on changing events and have made significant contributions in keeping the 1972 convention relevant to the needs of a changing world situation.

Viruses Crimean–Congo hemorrhagic fever virus Eastern equine encephalitis virus Ebola virus Equine morbillivirus Lassa fever virus Marburg virus Rift Valley fever virus South American hemorrhagic fever viruses Tick-borne encephalitis complex virus Variola (smallpox) major virus Venezuelan equine encephalitis virus Hantavirus Yellow fever virus

Biological Weapons and Anti-Terrorist Act (1989): established as a federal crime the development, manufacture, transfer, or possession of any biological agent, toxin, or delivery system for use as a weapon. Chemical and Biological Weapons Control Act (1991): places sanctions on companies that knowingly export goods or technologies relating to biological weapons to designated prohibited nations. The Defense Against Weapons of Mass Destruction Act (1996): designed to enhance federal, state and local emergency response capabilities to deal with terrorist incidents. Antiterrorism and Effective Death Penalty Act (1996): established as a criminal act any threat or attempt to develop BW or DNA technology to create new pathogens or make more virulent forms of existing organisms. Centers for Disease Control (CDC) Hazardous Biological Agent Regulation (1997): identification of infective agents that pose a significant risk to public health. Some of the organisms regulated by the CDC are listed in Table 13.1.

IV. CURRENT RESEARCH PROGRAMS Biological weapons research in the United States is under the direction of the BDRP, headquartered at Fort Detrick, Maryland. In accordance with official U.S. policy, the BDRP is solely defensive in nature,

ch13.qxd 10/9/03 6:57 PM Page 174

174

the desk encyclopedia of microbiology

with the goal of providing methods of detection for, and protective measures against, biological agents that could be used as weapons against U.S. forces by hostile states or individuals. Current U.S. policy stems from the 1969 declaration made by President Nixon that confined the U.S. BW program to research on biological defense such as immunization and measures of controlling and preventing the spread of disease. Henry Kissinger further clarified the U.S. BW policy in 1970 by stating that the United States biological program will be confined to research and development for defensive purposes only. This did not preclude research into those offensive aspects of biological agents necessary to determine what defensive measures are required. The BDRP expanded significantly in the 1980s in an apparent response to alleged treaty violations and perceived offensive BW capabilities of the Soviet Union. These perceptions were espoused primarily by representatives of the Reagan administration and the Department of State. At congressional hearings in May 1988, the U.S. government reported that at least 10 nations, including the Soviet Union, Libya, Iran, Cuba, Southern Yemen, Syria, and North Korea, were developing biological weapons. Critics of the U.S. program refuted the need for program expansion. The BDRP is administered through two separate government organizations—the army and the CIA. Details of the program are described in the April 1989 Environmental Impact Statement published by the Department of the Army, U.S. Army Medical Research and Development Command. The BDRP is located at three sites: the U.S. Army Medical Research Institute of Infectious Diseases (USAMRIID) at Fort Detrick, Maryland; the Aberdeen Proving Ground in Maryland; and the Dugway Proving Ground in Utah. USAMRIID is designated as the lead laboratory in medical defense against BW threats. Research conducted at the USAMRIID focuses on medical defense such as the development of vaccines and treatments for both natural diseases and potential BW agents. Work on the rapid detection of microorganisms and the diagnosis of infectious diseases is also conducted. The primary mission at the Aberdeen Proving Ground is nonmedical defense against BW threats including detection research, such as the development of sensors and chemiluminescent instruments to detect and identify bacteria and viruses, and development of methods for material and equipment decontamination. The U.S. Army Dugway Proving Ground is a Department of Defense major range and test facility responsible for development, testing, evaluation, and operation of chemical warfare equipment, obscurants and smoke munitions,

and biological defense equipment. Its principle mission with respect to the BDRP is to perform developmental and operational testing for biological defense material, including the development and testing of sensors, equipment, and clothing needed for defense against a BW attack. One hundred secondary sites have received contracts for biological defense research. Secondary sites include the Swiftwater Lab, operated by the Salk Institute in Swiftwater, Pennsylvania; the Naval Medical Research Institute in California; medical centers; universities; and private biotechnology firms in the United States, Scotland, and Israel. The CIA also participates in the administration of the BDRP. In 1982, Thomas Dashiell of the office of the secretary of defense reported on a classified technology watch program related to BW that was operated by the intelligence community. The program was designed to monitor worldwide developments related to BW that could affect the vulnerability of U.S. and NATO forces to biological attack. BDRP research focuses on five main areas: 1. Development of vaccines. 2. Development of protective clothing and decontamination methods. 3. Analysis of the mode of action of toxins and the development of antidotes. 4. Development of broad-spectrum antiviral drugs for detecting and diagnosing BW agents and toxins. 5. Utilization of genetic engineering methods to study and prepare defenses against BW and toxins. The BDRP has often been a center of controversy in the United States. One BDRP facility, the Dugway Proving Ground, was the target of a lawsuit that resulted in the preparation of the environmental impact statement for the facility. A proposal for a highlevel containment laboratory (designated P-4) was ultimately changed to a plan for a lower-level (P-3) facility. The use of genetic engineering techniques in BDRP facilities has also been a focus of controversy. The BDRP takes the position that genetic engineering will be utilized if deemed necessary. The Department of Defense stated that testing of aerosols of pathogens derived from recombinant DNA methodology is not precluded if a need should arise in the interest of national defense. One specific program requires special note. The Defense Advanced Research Project Agency is a Pentagon program that invests significantly in pathogen research through grants to qualified institutions. This project initially focused on engineering and electronics (computer) projects; however, starting in 1995 biology became a key focus, and several BW

ch13.qxd 10/9/03 6:57 PM Page 175

biological warfare defensive research grants are now in operation at several academic and private institutions. Very little is written in the unclassified literature on BW research conducted in countries other than the United States. Great Britain has maintained the Microbiological Research Establishment at Porton Down; however, military research is highly classified in Great Britain and details regarding the research conducted at Porton are unavailable. During the 1970s and 1980s, much of the U.S. BW policy was based on the assumption of Soviet offensive BW capabilities. Most U.S. accounts of Soviet BW activities were unconfirmed accusations or claims about treaty violations. The Soviet Union was a party to both the 1925 Geneva Protocol and the 1972 BW convention. According to Pentagon sources, the Soviet Union operated at least seven top-security BW centers. These centers were reported to be under strict military control. Although the former Soviet Union proclaimed that their BW program was purely defensive, the United States consistently asserted that the Soviet Union was conducting offensive BW research.

V. CONTEMPORARY ISSUES A. Genetic engineering There has been considerable controversy regarding the potential for genetically engineered organisms to serve as effective BW agents. Recombinant DNA technology has been cited as a method for creating novel, pathogenic microorganisms. Theoretically, organisms could be developed that would possess predictable characteristics, including antibiotic resistance, altered modes of transmission, and altered pathogenic and immunogenic capabilities. This potential for genetic engineering to significantly affect the military usefulness of BW has been contested. It has been suggested that because many genes must work together to endow an organism with pathogenic characteristics, the alteration of a few genes with recombinant DNA technology is unlikely to yield a novel pathogen that is significantly more effective or usable than conventional BW agents. The question of predictability of the behavior of genetically engineered organisms was addressed at an American Society for Microbiology symposium held in June 1985. Some symposium participants believed that the use of recombinant DNA increases predictability because the genetic change can be precisely characterized. Other participants, however, believed that the use of recombinant DNA decreases predictability because it widens the potential range of DNA sources. Other evidence supports the view that genetically engineered

175

organisms do not offer substantial military advantage over conventional BW. Some studies have shown that in general, genetically engineered organisms do not survive well in the environment. This fact has been cited as evidence that these organisms would not make effective BW agents. Despite the contentions that genetic engineering does not enhance the military usefulness of BW, a significant number of arguments support the contrary view. At the 1986 review conference of the BW convention, it was noted that genetic engineering advances since the convention entered into force may have made biological weapons a more attractive military option. Several authors have contended that the question of the potential of genetic engineering to enhance the military usefulness of BW is rhetorical because the 1972 BW convention prohibits development of such organisms despite their origin or method of production. Nations participating in both the 1980 and 1986 review conferences of the BW convention accepted the view that the treaty prohibitions apply to genetically engineered BW agents. An amendment to the treaty, specifically mentioning genetically engineered organisms, was deemed to be unnecessary. In addition, the United States, Great Britain, and the Soviet Union concluded in a 1980 briefing paper that the 1972 BW convention fully covered all BW agents that could result from genetic manipulation. Although the utility of genetic engineering for enhancing the military usefulness of BW agents has been questioned, the role of genetic engineering for strengthening defensive measures against BW has been clear. Genetic engineering has the potential to improve defenses against BW in two ways: (i) vaccine production and (ii) sensitive identification and detection systems. The issues of the new technologies in defensive research have been evident in the U.S. BW program. Since 1982, U.S. Army scientists have used genetic engineering to study and prepare defenses against BW agents. Military research utilizing recombinant DNA and hybridoma technology includes the development of vaccines against a variety of bacteria and viruses, methods of rapid detection and identification of BW agents, and basic research on protein structure and gene control. By improving defenses against BW, it is possible that genetic engineering may potentially reduce the risk of using BW. The primary effect of BW on government regulations on genetic engineering is the tendency toward more stringent control of the technologies. The fear of genetically engineered BW agents has prompted proposals for government regulation of BW research utilizing genetic engineering research. The Department of Defense released a statement indicating that all

ch13.qxd 10/9/03 6:57 PM Page 176

176

the desk encyclopedia of microbiology

government research was in compliance with the 1972 BW convention. The government has also prepared an environmental impact statement of research conducted at Fort Detrick. Government regulations on genetic engineering also affect BW research through limitations on exports of biotechnology information, research products, and equipment. In addition to controls of exports due to competitive concerns of biotechnology companies, a substantial amount of information and equipment related to genetic engineering are prohibited from being exported from the United States. The Commerce Department maintains a “militarily critical technology” list, which serves as an overall guide to restricted exports. Included on the list are containment and decontamination equipment for large production facilities, high-capacity biological reactors, separators, extractors, dryers, and nozzles capable of disseminating biological agents in a fine mist. Genetic engineering has altered the concept of BW. A current, comprehensive discussion of BW would include both naturally occurring and potential genetically engineered agents. Many current defenses against BW are developed with genetic engineering techniques. Government regulations on biotechnology have limited BW research, while fears of virulent genetically engineered BW agents have strengthened public support for stronger regulations. Future policies related to BW will need to be addressed in light of its altered status.

conventional war, including terrorism and guerrilla warfare. In the 1980s, the low-intensity conflict doctrine, which was espoused by the Reagan administration, was a plan for U.S. aid to anti-Communist forces throughout the world as a way of confronting the Soviet Union without using U.S. combat troops. Despite the significant changes in the world since the inception of the low-intensity conflict doctrine, the probability of increasing numbers of small conflicts still exists. Although no evidence indicates that the United States would consider violating the 1972 BW convention and support biological warfare, the overall increase in low-level conflicts in the future may help create an environment conducive to the use of BW. Although BW may not be assessed as an effective weapon in a full-scale conventional war, limited use of BW agents may be perceived as advantageous in a small-scale conflict. Although strong deterrents exist for nuclear weapons, including unavailability and, most formidably, the threat of uncontrolled worldwide “nuclear winter,” BW may be perceived as less dangerous. In addition, the participants of low-level conflicts may not possess the finances for nuclear or conventional weapons. BW agents, like chemical weapons, are relatively inexpensive compared to other weapon systems and may be seen as an attractive alternative to the participants and leaders of lowlevel conflicts. Low-level conflict, therefore, increases the potential number of forums for the use of BW.

B. Mathematical epidemiology models

D. Terrorism

Although genetic engineering may potentially alter characteristics of BW agents, mathematical models of epidemiology may provide military planners with techniques for predicting the spread of a released BW agent. One of the hindrances that has prevented BW from being utilized or even seriously considered by military leaders has been the inability to predict the spread of a BW agent once it has been released into the environment. Without the capability to predict the spread of the released organisms, military planners would risk the accidental exposure of their own troops and civilians to their own weapons. The development of advanced epidemiology models may provide the necessary mechanisms for predicting the spread of organisms that would substantially decrease the deterrent factor of unpredictability.

A final factor that could significantly affect BW is the worldwide increase in terrorism or the violent activities of subnational groups. Although there has not been an incident to date of the successful use of BW by a terrorist group, the possibility of such an event has increased in many forums. The relationship of terrorism and BW can be divided into two possible events. The first is terrorist acts against laboratories conducting BW-related research. The level of security at Fort Detrick is high, the possibility of a terrorist attack has been anticipated, and contingency plans have been made. Complicating the problem of providing security against terrorist attack in the United States is the fact that although most BW research projects are conducted with the BW research program of the Army, an increasing number of projects are supported by the government that are conducted outside of the military establishment. These outside laboratories could be potential targets. The second type of terrorist event related to BW is the potential use of BW by terrorists against urban areas or major public facilities. Biological weapons are

C. Low-level conflict Another important factor that has affected the current status of BW is the increase in low-level conflict or the spectrum of violent action below the level of small-scale

ch13.qxd 10/9/03 6:57 PM Page 177

biological warfare relatively inexpensive and easy to develop and produce compared to conventional, nuclear, or chemical weapons. BW agents can be concealed and easily transported across borders or within countries. In addition, terrorists are not hampered by a fear of an uncontrolled spread of the BW agent into innocent civilian populations. On the contrary, innocent civilians are often the intended targets of terrorist activity and the greater chance for spread of the BW agent may be considered to be a positive characteristic (see Section II.C).

E. Offensive versus defensive biological warfare research The distinctions between “offensive” and “defensive” BW research have been an issue since 1969, when the United States unilaterally pledged to conduct only defensive research. The stated purpose of the U.S. BDRP is to maintain and promote national defense from BW threats. Although neither the Geneva Convention nor the 1972 convention prohibits any type of research, the only research that nations have admitted to conducting is defensive. The problem is whether or not the two types of research can be differentiated by any observable elements. Although production of large quantities of a virulent organism and testing of delivery systems have been cited as distinguishing characteristics of an offensive program, a substantial amount of research leading up to these activities, including isolating an organism and then using animal models to determine pathogenicity, could be conducted in the name of defense. Vaccine research is usually considered defensive, whereas increasing the virulence of a pathogen and producing large quantities are deemed offensive. However, a critical component of a strategic plan to use biological weapons would be the production of vaccines to protect the antagonist’s own personnel (unless self-annihilation was also a goal). This means that the intent of a vaccine program could be offensive BW use. Furthermore, research that increases the virulence of an organism is not necessarily part of an offensive strategy because one can argue that virulence needs to be studied in order to develop adequate defense. The key element distinguishing offensive from defensive research is intent. If the intent of the researcher or the goals of the research program are the capability to develop and produce BW, then the research is offensive BW research. If the intent is to have the capability to develop and produce defenses against BW use, then the research is defensive BW research. Although it is true that nations may have policies of open disclosures (i.e., no secret research), “intent” is not observable.

177

Although the terms offensive BW research and defensive BW research may have some use in describing intent, it is more a philosophical than a practical distinction—one that is based on trust rather than fact.

F. Secrecy in biological warfare-related research Neither the Geneva Protocol nor the 1972 BW convention prohibits any type of research, secret or nonsecret. Although the BDRP does not conduct secret or classified research, it is possible that secret BW research is being conducted in the United States outside of the structure of the BDRP. The classified nature of the resource material for this work makes it impossible to effectively determine if secret research is being conducted in the United States or any other nation. It is not, however, unreasonable to assume that other nations conduct significant secret BW research. Therefore, regardless of the facts, one cannot deny the perception that such research exists in a variety of countries and that this perception will exist for the foreseeable future. Secrecy has been cited as a cause of decreased quality of BW research. If secret research, whether offensive or defensive, is being conducted in the United States or other nations, it is unclear if the process of secrecy affects the quality of the research. If the secret research process consists of a core of highly trained, creative, and motivated individuals sharing information, the quality of the research may not suffer significantly. It must be stated, however that secrecy by its very nature will limit input from a variety of diverse observers. Secrecy may increase the potential for violations of the 1972 BW convention; however, violations would probably occur regardless of the secrecy of the research. Secrecy in research can certainly lead to infractions against arbitrary rules established by individuals outside of the research group. The secret nature of the research may lure a researcher into forbidden areas. In addition, those outside of the research group, such as policy-makers, may push for prohibited activities if the sense of secrecy prevails. Secrecy also tends to bind those within the secret arena together and tends to enhance their perception of themselves as being above the law and knowing what is “right.” As in the case of Oliver North and the Iran-Contra Affair, those within the group may believe fervently that the rules must be broken for a justified purpose and a mechanism of secrecy allows violations to occur without penalty. The distrust between nations exacerbates the perceived need for secret research. The animosity between

ch13.qxd 10/9/03 6:57 PM Page 178

178

the desk encyclopedia of microbiology

the United States and the Soviet Union during the 1980s fueled the beliefs that secret research leading to violations of the 1972 BW convention was being conducted in the Soviet Union. As the belligerence of the 1980s faded into the new world order of the 1990s, the questions focus less on the Soviet Union and more on the Middle East and Third-World countries. There are factions in the United States that believe strongly that other countries are conducting secret research that will lead to violations of the convention. There is also a tendency to believe that the secrecy in one’s own country will not lead to treaty violations, whereas the same secret measures in an enemy nation will result in activities forbidden by international law. The importance of the concept of secrecy in BW research is related to the perception of secrecy and arms control agreements. Regardless of the degree of secrecy in research, if an enemy believes that a nation is pursuing secret research, arms control measures are jeopardized. The reduction of secrecy has been suggested as a tool to decrease the potential for BW treaty violations. A trend toward reducing secrecy in BW research was exemplified by the 1986 review conference of the 1972 BW convention, which resulted in agreements to exchange more information and to publish more of the results of BW research. Whether or not these measures have any effect on strengthening the 1972 BW convention remains to be seen. Organizations and individuals have urged a renunciation by scientists of all secret research and all security controls over microbiological, toxicological, and pharmacological research. This action has been suggested as a means of strengthening the 1972 BW convention. The belief that microbiologists should avoid secret research is based on the assumption that (i) secret research is of poor quality due to lack of peer review and (ii) secrecy perpetuates treaty violations. Although it may be reasonable to expect microbiologists to avoid secret research, it is not realistic. Secrecy is practiced in almost every type of research including academic, military, and especially industrial. Furthermore, there will always be those within the military and intelligence structures who believe that at least some degree of secrecy is required for national security. Secrecy in BW research is a complex issue. The degree to which it exists is unclear. Individuals are generally opposed to secrecy in BW research although other examples of secrecy in different types of research exist. The effect of secrecy on the quality of research, the need for the secrecy, and the choice of microbiologists to participate in secret BW research remain unanswered questions.

G. Problems relating to verification One of the major weaknesses of the 1972 BW convention has been the lack of verification protocols. Problems with effectively monitoring compliance include the ease of developing BW agents in laboratories designed for other purposes and the futility of inspecting all technical facilities of all nations. Measures that have been implemented with the goal of monitoring compliance included (i) open-inspections, (ii) intelligence gathering, (iii) monitor-research, (iv) use of sampling stations to detect the presence of biological agents, and (v) international cooperation. The progress achieved with the Chemical Weapons Convention has renewed interest in strengthening mechanisms for verification of compliance with the 1972 BW convention. Although this renewed interest in verification along with the emergence of the Commonwealth of Independent States from the old Soviet Union has brought an optimism to the verification issue, the reticence of countries such as Iraq to cooperate with United Nations inspection teams is a reminder of the complexities of international agreements. The examples discussed in this article are typical of the many issues attached to the concept of BW.

BIBLIOGRAPHY Atlas, R. M. (1998). Biological weapons pose challenge for microbiology community. ASM News 64, 383–389. Buckingham, W. A., Jr. (Ed.) (1984). “Defense Planning for the 1990s.” National Defense Univ. Press, Washington, DC. Cole, L. (1996, December). The specter of biological weapons. Sci. Am. 60–65. Frisna, M. E. (1990). The offensive–defensive distinction in military biological research. Hastings Cent. Rep. 20(3), 19–22. Gravett, C. (1990). “Medieval Siege Warfare.” Osprey, London. Harris, R., and Paxman, J. (1982). “A Higher Form of Killing.” Hill & Wang, New York. Khan, A. S., Morse, S., Lillibridge, S. (2000) Public-health preparedness for biological terrorism in the USA. Lancet 356(9236), 1179–1182. Livingstone, N. C. (1984). Fighting terrorism and “dirty little wars.” In “Defense Planning for the 1990s” (W. A. Buckingham, Jr., Ed.), pp. 165–196. National Defense Univ. Press, Washington, DC. Livingstone, N. C., and Douglass, J., Jr. (1984). “CBW: The Poor Man’s Atomic Bomb.” Tufts University, Institute of Foreign Policy Analysis, Medford, MA. Louria, D. B. (1986). Recombinant DNA technology and biological warfare. N. J. Med. 83(6), 399–400. Meselson, M., Guillemin, J., Hugh-Jones, M., Langmuir, A., Popova, I., Shelokov, A., and Yampolskaya, O. (1994). The Sverdlovsk anthrax outbreak of 1979. Science 266, 1202–1208. Milewski, E. (1985). Discussion on a proposal to form a RAC working group on biological weapons. Recombinant DNA Technol. Bull. 8(4), 173–175. Miller, L. A. (1987). The use of philosophical analysis and Delphi survey to clarify subject matter for a future curriculum for

ch13.qxd 10/9/03 6:57 PM Page 179

179

biological warfare microbiologists on the topic of biological weapons. Unpublished thesis, University of Pennsylvania, Philadelphia. (University Micro-films International, Ann Arbor, MI. 8714902). Murphy, S., Hay, A., and Rose, S. (1984). “No Fire, No Thunder.” Monthly Review Press, New York. Poupard, J. A., Miller, L. A., and Granshaw, L. (1989). The use of smallpox as a biological weapon in the French and Indian War of 1763. ASM News 55, 122–124. Smith, R. J. (1984). The dark side of biotechnology. Science 224, 1215–1216. Stockholm International Peace Research Institute (1973). “The Problem of Chemical and Biological Warfare,” Vol. 2. Humanities Press, New York. Taubes, G. (1995). The defense initiative of the 1990s. Science 267, 1096–1100. Wright, S. (1985). The military and the new biology. Bull. Atomic Sci. 42(5), 73.

Wright, S., and Sinsheimer, R. L. (1983). Recombinant DNA and biological warfare. Bull. Atomic Sci. 39(9), 20–26. Zilinskas, R., (Ed.) (1992). “The Microbiologist and Biological Defense Research: Ethics, Politics and Intermediate Security.” New York Academy of Sciences, New York.

WEBSITES A website of the Stockholm International Peace Research Institute, http://projects.sipri.se/cbw/bw-mainpage.html DefenseLINK. A website of the U.S. Department of Defense http://www.defenselink.mil/sites/

ch14.qxd 10/9/03 6:59 PM Page 180

14 Bioluminescence, Microbial J. Woodland Hastings Harvard University

GLOSSARY

scintillons Bioluminescent organelles unique to dinoflagellates which emit brief bright flashes of light following stimulation.

autoinducer A homoserine lactone produced by bacteria which, after accumulating in the medium to a critical concentration, initiates transcription of specific genes by a mechanism referred to as autoinduction, recently dubbed quorum sensing. bioluminescence Emission of light by living organisms that is visible to other organisms. It derives from an enzyme-catalyzed chemiluminescence, a highly exergonic reaction in which chemical energy is transformed into light energy. bioluminescence quantum yield The number of photons produced per luciferin (substrate) molecule oxidized in a bioluminescent reaction. blue and yellow fluorescent proteins Accessory proteins in the bioluminescence system in some bacteria, carrying lumazine and flavin chromophores, respectively, and serving as secondary emitters under some conditions. luciferase The generic name for enzymes that catalyze bioluminescent reactions. Luciferases from different major groups of organisms are not homologous (e.g. firefly and jellyfish luciferases are unrelated to bacterial luciferase) so the organism must be specified in referring to a specific luciferase. luciferin (light bearing) The generic name for a substrate that is oxidized to give light in a bioluminescent reaction; identified as a flavin in bacteria and a tetrapyrrole in dinoflagellates. The Desk Encyclopedia of Microbiology ISBN: 0-12-621361-5

Bioluminescence is defined as an enzyme-catalyzed chemiluminescence, a chemical reaction in which the energy released is used to produce an intermediate or product in an electronically excited state, which then emits a photon. It does not come from or depend on light absorbed, as in fluorescence or phosphorescence. However, the excited state produced in such a chemical reaction is indistinguishable from that produced in fluorescence after the absorption of a photon by the ground state of the molecule concerned. All bioluminescent reactions involve the oxidation by molecular oxygen of a substrate by an enzyme, generically referred to as luciferin and luciferase, respectively, with the production of an electronically excited state, typically luciferase bound (Fig. 14.1A). The energy released from the oxidation of a luciferin in such reactions is about 10 times greater than that obtained from the hydrolysis of ATP. There are numerous (20–30) extant bioluminescent systems, which mostly bear no evolutionary relationships with one another. The many different luciferases are thus considered to have arisen de novo and evolved independently, and the luciferins are likewise different. Thus, genes coding for luciferases from fireflies and jellyfish, for example, have no sequence similarities to bacterial or dinoflagellate luciferase, which themselves are unrelated. The luciferases discussed in this article

180

Copyright © 2003 Elsevier Ltd All rights of reproduction in any form reserved

ch14.qxd 10/9/03 6:59 PM Page 181

181

bioluminescence, microbial

FIGURE 14.1 (A) Generalized reaction scheme for bioluminescent reactions. (B) The luciferase reaction showing the components involved in the bacterial system. Reduced flavin derived from the electron transport pathway reacts with luciferase and molecular oxygen to form an intermediate peroxide. In a mixed function oxidation with long-chain aldehyde (RCHO), hydroxy-FMN is formed in its excited state (*), from which emission of a photon (h) occurs. The FMN and long-chain acid products are recycled.

should therefore be called bacterial luciferase and dinoflagellate luciferase.

I. BACTERIA A. Occurrence, habitats, species, and functions In the ocean luminous bacteria occur ubiquitously and can be isolated from most seawater samples from the surface to depths of ~1000 m, and they appear as bright colonies on plates (Fig. 14.2). They are very often found in some kind of symbiotic association with higher organisms (e.g. fish or squid), in which the light emission is evidently of functional importance to the host. In parasitic or saprophytic associations the advantage of light emission accrues more to the bacteria: The light attracts animals to feed, enhancing the

FIGURE 14.2 Colonies of luminous bacteria photographed by their own light (right) and in room light (left). Light is emitted continuously but is controlled by a quorum-sensing mechanism (autoinducer) and is thus not proportional to growth or cell density (Plate 2).

ch14.qxd 10/9/03 6:59 PM Page 182

182

the desk encyclopedia of microbiology

FIGURE 14.3 The flashlight fish (Photoblepharon) showing the exposed light organ, which harbors luminous bacteria and is located just below the eye. A special lid allows the fish to turn the light on and off (photograph by Dr. James Morin) (Plate 3).

dispersal of the bacteria. The maximum light emission of a bacterial cell is about 104 q s1, meaning that to be seen, the cell density must be high—about 109–1010 cells/ml. Most luminous bacteria are classed under three major genera—Vibrio, Photobacterium, and Photorhabdus, the first two of which are almost exclusively marine, whereas the last is terrestrial. All are characterized as enteric bacteria and are notable for the symbioses in which they participate, most commonly in light organs in which the light is used by the host for some purpose. The flashlight fish, Photoblepharon (Fig. 14.3), maintains cultures of such bacteria in special organs located beneath the eyes. For all luminous bacterial species the primary habitat can be assumed to be in some association, either as a light organ or gut symbiont, or in a parasitic or saprophytic association. If such associations are viewed as primary habitats, planktonic or “free-living” bacteria in the ocean may be considered secondary or reservoir habitats, produced as overflows or escapees into an environment in which luminescence may not be advantageous and thus not selected for. Thus, the failure, for whatever reason, of the luminescence system to be expressed under these conditions may be advantageous for the survival of the bacteria possessing the genes for luminescence and thus for their ability to compete favorably with heterotrophs not carrying the genes. Different species occupy different specific habitats. Vibrio harveyi is the most cosmopolitan species and is not known to be involved in a light organ symbiosis. It occurs as a gut symbiont in many marine animals, and it is known to parasitize and/or infest saprophytically crustaceans and other species. Fish or squid having specific associations with Vibrio fischeri,

Photobacterium phosphoreum, and Photobacterium leigonathi as symbionts have been identified, and all of these species have more restricted requirements for growth. Still other symbionts have not been cultured successfully, but affinities and relationships are known from their luciferase DNA sequences. Photorhabdus luminescens is symbiotic with nematodes, which parasitize caterpillars, where they release the bacteria as an inoculum into the body cavity along with their own fertilized eggs. The bacteria grow, providing nutrient for the developing nematode larvae. The caterpillar does not survive but becomes brightly luminous, possibly to attract animals to feed on it and thereby disperse both nematodes and bacteria. Each young nematode then carries a fresh inoculum, estimated to be about 50–100 bacteria.

B. Biochemistry 1. Light-emitting reaction Biochemically, light emission results from the luciferase-catalyzed mixed function oxidation of reduced flavin mononucleotide and long-chain aldehyde by molecular oxygen, populating the excited state of a luciferase-hydroxyflavin intermediate, which emits a blue-green light (max ~ 490 nm). Bacterial luciferase is an – heterodimer lacking metals, prosthetic groups, and non-amino acid residues. To date, no sequences in data bases exhibit any similarities to it, so the origin of the gene for the enzyme remains unknown. Although the two subunits are homologous, the active site and the detailed kinetics features of the reaction are properties of the subunit (singular kinetic). The reaction represents a biochemical shunt of the respiratory electron transport system, carrying electrons from the level of reduced flavin (FMNH2) directly to oxygen (Fig. 14.1B). FMNH2 reacts first with oxygen to form a linear hydroperoxide, which then reacts with long-chain fatty aldehyde to give the postulated peroxyhemiacetal intermediate. This breaks down to give long-chain acid and the intermediate hydroxyflavin in a high-energy electronically excited state. Although aldehydes with chain lengths from 7 to 18 carbon atoms give light in the reaction with isolated luciferase, tetradecanal (14C) has been identified as the naturally occurring molecule in the species studied. While its oxidation provides energy, the aldehyde is not the emitter and is thus not a luciferin, which means “light-bearing.” The flavin is the luciferin in the bacterial system. One photon is produced for about every four molecules of FMNH2 oxidized; thus, the bioluminescence

ch14.qxd 10/9/03 6:59 PM Page 183

bioluminescence, microbial quantum yield is 25%. However, since the fluorescence quantum yield of FMN is about 30%, and the excited state produced as in Fig. 14.1B is equivalent to that which would be produced from light absorption by the hydroxyflavin, it may be concluded that the luciferase reaction is highly efficient. In the living cell light is produced continuously; the oxidized FMN formed in the reaction is reduced again as indicated in Fig. 14.1B by pyridine nucleotide. Similarly, the myristic acid product is converted back to the corresponding aldehyde by enzymes of a specific fatty acid reductase complex with ATP and NADPH as cofactors. 2. Luciferase structure The tertiary structure of luciferase (Fig. 14.4) was correctly predicted based on the X-ray crystal structure of a related and homologous protein expressed from lux F. The structures of the and subunits are similar, differing in a region in which substrate is presumed to bind to the , although the structure of the site is not known because a determination of the structure with flavin bound has not been made. Both subunits exhibit the so-called [/]8 barrel form (/ do not refer to subunits), in which stretches of beta sheet alternate with alpha-helical strands, all of which are parallel to one another, together forming a closed barrel with the eight helices on the outside. The region of the subunit where substrate is likely to

183

bind was not seen in the electron density map, including a region known to be highly sensitive to protease attack that is absent in the subunit. 3. Antenna proteins: blue and yellow fluorescent proteins as emitters The basic structure of the luciferase and the biochemistry of the reaction are the same in all luminous bacteria, which typically emit light peaking at about 490 nm. However, in some bacterial strains, the color of the light emitted by the living cell is blue or red shifted, even though the isolated luciferases still peaks at ~490 nm. In a strain of P. phosphoreum, the emission is blue shifted, peaking at about 480 nm, whereas in a strain of V. fischeri the light is yellow in color (max ~ 540 nm). In both cases a second (“antenna”) protein with its own chromophore is responsible (lumazine and flavin, respectively, for the two cases). The mechanisms involved in these cases have not been fully resolved, but they may be mechanistically similar because the proteins are homologous. Nonradiative energy transfer has been suggested, but evidence indicates that this alone cannot be responsible since the antenna protein appears to actually enter into the light-emitting reaction in the case of the yellow-emitting system. The functional importance for such spectral shifts has not been elucidated, although strains with a blue-shifted emission occur at depths of ~600 m in the ocean. 4. Molecular biology: genes of the lux operon

FIGURE 14.4 Ribbon representation of the structure of bacterial luciferase showing the and subunits and how they may associate (from Fisher et al., 1995).

Lux genes cloned from several different species exhibit sequence similarities indicative of evolutionary relatedness and conservation. In V. fischeri, the species most extensively studied, the lux operon has five structural and two regulatory genes with established functions. As shown in Fig. 14.5, these include two that code for the and subunits of luciferase and three that code for the reductase, transferase, and synthetase components of the fatty acid reductase complex responsible for aldehyde synthesis and reduction and recycling of the acid product. Upstream, in the same operon, is the regulatory gene lux I, and immediately adjacent but transcribed in the opposite direction is lux R, whose product is responsible for the transcriptional activation of lux A–E and others. The latter include lux F and G, found in some species and located in the same region; these code for proteins whose functions are not well established. However, genes coding for the antenna proteins responsible for color shifting are located elsewhere on the genome but still subject to regulation by the autoinduction mechanism. In luminous Vibrios the

ch14.qxd 10/9/03 6:59 PM Page 184

184

the desk encyclopedia of microbiology

FIGURE 14.5 Organization of the lux genes in Vibrio fischeri. The operon on the right, transcribed from the 5 to the 3 end, carries genes

for synthesis of autoinducer (lux I), for luciferase and subunits (lux A and B), and for aldehyde synthesis (lux C–E). The operon on the left carries the lux R gene, which encodes for a receptor molecule that binds autoinducer; the complex controls the transcription of the right operon. Other genes, lux F (N). G, and H (right), are associated with the operon but with uncertain functions; genes for accessory emitter proteins also occur (left).

regulatory genes (lux I and R or their counterparts) are also located remotely from the lux A–E operon.

certain specific genes is functionally important only at higher cell densities. The phenomenon was then appropriately dubbed “quorum sensing.”

5. Physiology: regulation of light emission a. Autoinduction and quorum sensing Quorum sensing, which was first discovered and referred to as autoinduction in luminous bacteria, refers to a mechanism causing the lux genes to be transcribed only at higher cell densities. It is mediated by a homoserine lactone molecule produced by the cells that has been dubbed the autoinducer. In luminous bacteria in a confined environment, such as a light organ, autoinducer can accumulate and act, whereas in free-living bacteria, in which the light could not be seen at the low cell densities, luciferase is of no value and is not produced. Autoinduction was discovered in the early 1970s and was proposed is as an explanation for the fact that luciferase and other components of the light-emitting system are not produced in cells growing at low cell densities but are produced, and rapidly so, above a critical cell concentration (Fig. 14.6). In laboratory cultures subjected to continuous (or repetitive) dilution (maintaining densities lower than ~107 cells/ml), such that autoinducer accumulation is not possible, no synthesis of luciferase or its messenger RNA occurs. The same is true of planktonic populations in the ocean, which are typically at densities of no greater than ~102 cells/ml. The autoinducer in V. fischeri is the product of the lux I gene and acts as a positive regulator of the lux operon in the presence of a functional lux R gene. In the early 1990s it was found that a similar mechanism, also utilizing specific homoserine lactones, occurs in many other diverse groups of bacteria in which expression of

FIGURE 14.6 An experiment demonstrating autoinduction in luminous bacteria. Growth of the cells (measured by optical density at 660 nm) is exponential for the first few hours, during which time there is no change in the bioluminescence or the luciferase content of the culture (arbitrary units). After about 2.5h, at a cell density of 1 or greater, the lux genes are rapidly transcribed and luciferase and other related proteins are synthesized, determined also by reaction with antiluciferase (CRM).

ch14.qxd 10/9/03 6:59 PM Page 185

bioluminescence, microbial b. Glucose, iron, and oxygen In both V. harveyi and V. fischeri luminescence is repressed by glucose, reversible by cyclic AMP. All species except Photorhabdus luminescens exhibit induction of luminescence by the addition of iron after growth under conditions of iron limitation, suggesting that eukaryotic hosts might use iron limitation to limit growth while maximizing luminescence of bacterial symbionts. As a reactant in the luciferase reaction, oxygen can control light emission directly but only at extremely low oxygen concentrations (lower than ~0.1%), above which the luminescence is independent of oxygen. Growth, however, is reduced at concentrations lower than that of air (21%), so growth could be strongly inhibited without affecting the luciferase reaction. Indeed, in some species and strains, transcription of the lux operon is greatly favored over growth under microaerophilic conditions, where bright light emission may occur at low cell densities. Regulation by control of oxygen has thus been proposed as a mechanism whereby eukaryotic hosts might control growth of bacterial symbionts in light organs while maximizing luminescence. In P. luminescens grown in pure (100%) oxygen, which is lethal for many bacteria including other luminous species, both luciferase synthesis and luminescence are enhanced in relation to cell mass. It has been proposed that in this case the luciferase system serves to detoxify damaging oxygen radicals.

c. Dark variants In culture collections it has often been reported that bacteria may lose their luminosity over time in cases in which subculturing care has not been taken to reisolate single bright colonies each time. This can be attributed to the spontaneous occurrence and overgrowth of dark (e.g. very dim) variants, which is some ways appear to be similar to phase variants reported in other groups of bacteria. These dark variants do not produce luciferase or other luminescence components and are pleiotropic, being altered in several other properties such as cell morphology and phase sensitivity. Dark variants could presumably provide a genetic mechanism whereby cells could respond to environmental conditions that select for or against the property of luminescence. Although such conditions have not been established, this would allow cells to compete better with other heterotrophic bacteria under conditions where luminescence is of no use and thereby become more widely dispersed but prepared to populate a niche where luminescence is functionally important.

185

II. DINOFLAGELLATES A. Occurrence, habitats, species, and functions Ocean “phosphorescence,” commonly seen at night (especially in summer) when the water is disturbed, is due in large part to the bioluminescence of dinoflagellates. The organisms occur ubiquitously in the oceans as planktonic forms and respond to mechanical stimulation when the water is disturbed, such as by waves or fish swimming, by emitting brief (~0.1 s) bright flashes (~109 photons each). The wake of a large ship may be evident from such light emission for approximately 20 miles. Luminescent dinoflagellates occur primarily in surface waters and many species are photosynthetic. Only approximately 20–30% of marine species are bioluminescent. The so-called red tides are transient blooms (usually for weeks) of individual dinoflagellate species. Cells typically migrate vertically during the night to deeper water where available nutrients are taken up, returning to the surface to photosynthesize during the day. Phosphorescent bays (e.g. in Puerto Rico and Jamaica) are persistent blooms of this type; in Puerto Rico, the dominant species is Pyrodinium bahamense. As a group, dinoflagellates are important as symbionts, notably for contributing photosynthesis and carbon fixation in certain animals. Unlike bacteria, however, luminous dinoflagellates are not known to be harbored as symbionts on the basis of their light emission. Since dinoflagellates are stimulated to emit light when predators (e.g. crustaceans) are active, predators might thereby be alerted to feed on crustaceans, resulting in a reduced predation on dinoflagellates generally. Predation on dinoflagellates may also be impeded more directly and help individual cells. The flash could startle or divert a predator, allowing that cell to escape predation. The response time to stimulation (milliseconds) is certainly fast enough to have this effect. This latter explanation, though supported by experiment, does not easily account for the fact that not all species are bioluminescent.

B. Cell biology Luminescence in dinoflagellates is emitted from many small (~0.5 m) cortical locations (Fig. 14.7). The structures have been identified as a new type of organelle termed scintillons (flashing units). They occur as outpocketings of the cytoplasm into the cell vacuole, like a balloon, with the neck remaining connected. Scintillons contain only dinoflagellate luciferase and

ch14.qxd 10/9/03 6:59 PM Page 186

186

the desk encyclopedia of microbiology

luciferin and a protein that binds luciferin and keeps it from reacting with luciferase in between flashes. Other cytoplasmic components are somehow excluded from scintillons, which can be identified ultrastructurally by immunolabeling with antibodies raised against the luminescence proteins. They can also be visualized using image intensification by their bioluminescent flashing following stimulation as well as by the fluorescence of luciferin, which is present nowhere else in the cell.

C. Biochemistry Dinoflagellate luciferin is a highly reduced novel tetrapyrrole related to chlorophyll (Fig. 14.8) and in extracts remains tightly bound to a ~75-kDa specialized protein at cytoplasmic pH (~8). The luciferase is also inactive at pH 8; it is a large single polypeptide chain of about 136 kDa with three contiguous and intramolecularly homologous domains, each having luciferase activity. Activity can be obtained in soluble extracts made at pH 8 simply by shifting the pH to 6; the luciferin is released from its binding protein and the luciferase assumes an active conformation. The pK for both proteins is at pH 6.7. A similar activity can be found in the particulate (scintillon) fractions. Together, these results suggest that during extraction some scintillons are lysed with the proteins released into the soluble fraction, whereas others seal off at the neck and form closed vesicles. With the scintillon fraction, the in vitro activity is also triggered by a pH change and occurs as a flash (~100 ms), very close to that of the living cell, and the kinetics are independent of the dilution of the suspension. For the soluble fraction, the kinetics are dependent on dilution, as in an enzyme reaction.

FIGURE 14.7 Gonyaulax cells viewed by fluorescence microscopy showing scintillons (bioluminescent organelles) visualized by the fluorescence of dinoflagellate luciferin (max of emission, 475 nm), with chlorophyll fluorescence as the red background. Scintillons are structurally formed and destroyed on a daily basis, controlled by the circadian clock. (Right) Night phase cell with many scintillons; (left) day phase cell with few scintillons (Plate 4).

D. Cellular flashing The flashing of dinoflagellates in vivo is typically initiated by mechanical shear or cell stimulation, which has been shown to result in the generation of a conducted action potential in the vacuolar membrane. It is postulated that as this action potential traverses the vacuolar membrane it sweeps over the scintillons, opening voltage-gated ion channels, thus allowing protons from the acidic vacuole to enter, causing a transient pH change in the scintillons and thus a flash. Spontaneous flashes also occur (Fig. 14.9B).

E. Circadian clock control of dinoflagellate luminescence Unlike bacteria, cell density and growth conditions have no effect on the development and expression of

FIGURE 14.8 (A) Dinoflagellate luciferin, a tetrapyrrole, showing the location (132 ) of the oxygen addition and (B) the steps in the bioluminescent reaction.

bioluminescence in dinoflagellates. However, in G. polyedra and some other dinoflagellates, luminescence is regulated by day–night light–dark cycles and an internal circadian biological clock mechanism. Spontaneous flashing (and also flashing in response to mechanical stimulation) is far greater during the night than during the day (and therefore flashes are more frequent), and a steady low-level emission (glow) exhibits a peak toward the end of the night phase. The regulation is attributed to an endogenous mechanism; cultures maintained under constant conditions (light and temperature)

ch14.qxd 10/9/03 6:59 PM Page 187

187

bioluminescence, microbial

FIGURE 14.9 Bioluminescent flashes and glow of Gonyaulax. (A) Oscilloscope trace (ordinate, light intensity; abscissa, time, 8 s) recording from a vial with 32 000 cells. Six flashes having durations of about 100 ms are superimposed on a background glow. (B) Recording for 5 days from a similar vial kept in constant conditions showing the circadian rhythm of the background glow. The frequency of flashing also exhibits a circadian rhythm (not shown).

continue to exhibit rhythmicity (Fig. 14.9B), but with a period that is not exactly 24 h—it is only about (circa) 1 day (diem) (thus the origin of the term). Genes coding for molecular components of the circadian clock have been identified and studied in several systems, and a mechanism involving negative feedback on the transcription of a gene by its protein product is postulated to be responsible for the rhythm. How such a mechanism might exert physiological control is not understood. In humans and other higher animals, in which it regulates the sleep–wake cycle and many other physiological processes, the mechanism involves the nervous system. However, it also occurs in plants and unicellular organisms, such as G. polyedra, in which daily changes in the cellular concentrations of luciferase, luciferin, and its binding protein occur. The two proteins are synthesized and destroyed each day,

as are the scintillons in which they are located. Hence, the biological clock exerts control at a very basic level by controlling gene expression. This might explain the greater amount of luminescence at night, but a biochemical basis for the increased sensitivity to mechanical stimulation is not evident.

III. FUNGI A. Occurrence, habitats, species, and functions Light emission now known to be due to fungi has been observed since ancient times and was noted by both Aristotle and Pliny. Robert Boyle placed “shining wood” in his vacuum apparatus and showed that light emission was reversibly extinguished by the removal of

ch14.qxd 10/9/03 6:59 PM Page 188

188

the desk encyclopedia of microbiology

air, anticipating the requirement for molecular oxygen by all bioluminescent systems. Definitive knowledge of the fungal origin of luminous wood emerged during the nineteenth century from extensive studies of timbers used for support in mines, and by the mid-twentieth century about 80 luminous species had been inventoried. As in bacteria, the emission is continuous and not affected by mechanical stimulation, but it is really quite dim. There is no indication that luminescence is regulated in relation to cell growth or density, but there is some evidence that nutrition may play a role. There have been reports that the luminescence is circadian regulated, as in dinoflagellates, but such results have not been confirmed or extended. With one possible exception, all luminous fungi are basidiomycetes, and most are in the mushroom family; both the mycelium and fruiting body are luminous (Fig. 14.10). Such fungi occur in the many diverse habitats in which fungi occur, with the luminescence being visible most readily in dark forests—both tropical and temperate. The most striking reports describe luminescence from the interior or an infested tree split open by lightning. The function of bioluminescence in fungi is not well understood. It has been suggested that the light serves as an attractant, which is consistent with the generalization that a continuous light emission acts in this way. If so, insects or other invertebrates might be attracted and enhance spore dispersal. However, this leaves the function of emission in the mycelium unexplained. The system might have evolved biochemically without constraints regarding its localization, and since it is probably not an energy-intensive function its value in any part of the lifecycle could be adequate to justify its retention.

B. Biochemistry The spectrum of the light emitted has been determined from several species, all of which peak at about 525 nm, consistent with a flavin emitter. However, no biochemical evidence indicates this, and indeed no satisfactory understanding of the chemical basis for light emission has been obtained. Many years ago, a luciferin– luciferase-type system was reported with a link to reduced pyridine nucleotide, comparable to the bacterial system. However, this has not been confirmed in more recent studies, which suggest that the reaction may be a nonenzymatic chemiluminescence.

BIBLIOGRAPHY Case, J. F., Herring, P. J., Robison, B. H., Haddock, S. H. D., Kricka, L., and Stanley, P. E. (2002). “Bioluminescence and Chemiluminescence: Proceedings of the 11th International lymporium”. World Scientific, Singapore, 517 pp. Fisher, A. J., Raushel, F. M., Baldwin, T. O., and Rayment, I. (1995). Three dimensional structure of bacterial luciferase from Vibrio harveyi at 2.4 Å resolution. Biochemistry 34, 6581–6586. Hastings, J. W. (1994). The bacterial luciferase reaction: model or maverick in flavin biochemistry? In “Flavins and Flavoproteins” (K. Yagi, ed.), pp. 813–822, De Gruyter, Amsterdam. Hastings, J. W. (1996). Chemistries and colors of bioluminescent reactions—a review. Gene 173, 5–11. Hastings, J. W. (2001). Bioluminescence. In “Cell Physiology” (N. Sperelakis, ed.), 3rd edn., pp. 1115–1131, Academic Press, New York. Hastings, J. W., and Johnson, C. (2003). Bioluminescence and chemi luminescence. Biophotonics. Meth. Enz. 360, 76–104. Hastings, J. W., and Wood, K. V. (2001). Luciferases did not all evolve from precursors having similar enzymatic properties. In “Photobiology 2002” (D. Valenzeno and T. Coohill, eds.). pp. 199–210, Valdenmar Publ. Co., Overland Park, KS. Hastings, J. W., Kricka, L. J., and Stanley, P. E. (eds.) (1997). “Bioluminescence and Chemiluminescence: Molecular Reporting with Photons.” Wiley, Chichester, UK. Roda, A., Kricka, L. J., and Stanley, P. E. (eds.) (1999). “Bioluminescence and Chemiluminescence: Perspectives for the 21st Century.” Wiley, Chichester, UK. Taylor, F. J. R. (ed.) (1987). “The Biology of Dinoflagellates.” Blackwell, Oxford. Wilson, T., and Hastings, J. W. (1998). Bioluminescence. Annu. Rev. Cell. Dev. Biol. 14, 197–230.

WEBSITES

FIGURE 14.10 Bioluminescent mushroom photographed by its own light (photograph by Dr. Dan Perlman) (Plate 5).

These websites provide material and images on bioluminescence. http://siobiolum.ucsd.edu/Biolum_intro.html http://lifesci.ucsb.edu/~biolum/ http://mcb.harvard.edu/hastings/Images/bioluminescence.html http://www.hboi.edu/marinesci/biolum.html http://www.herper.com/Bioluminescence.html Journal of Bioluminescence and Chemiluminescence; WebSite, Wiley Publ. http://www3.interscience.wiley.com/cgi-bin/jtoc? ID=4087

ch15.qxd 10/9/03 7:00 PM Page 189

15 Bioreactors Larry E. Erickson Kansas State University

cell is a miniature bioreactor; other examples include shake flasks, petri dishes, and industrial fermentors. Diagnostic products based on enzymatic reactions, farm silos for silage fermentations, bread pans with fermenting yeast, and the soil in a Kansas wheat field may also be viewed as bioreactors. Although the bioreactor may be simple or highly instrumented, the important consideration is the ability to produce the desired product or result. The bioreactor is designed and operated to provide the environment for product formation selected by the scientist, baker, or winemaker. It is the heart of many biotechnological systems that are used for agricultural, environmental, industrial, and medical applications.

GLOSSARY airlift reactor Column with defined volumes for upflow and downflow of the culture broth; vertical circulation occurs because air is bubbled into the upflow volume. batch bioreactor Culture broth is fed into the reactor at the start of the process; air may flow continuously. bubble reactor Aerated column without mechanical agitation. fed batch Liquid media is fed to the reactor continuously; the broth accumulates in the reactor because there is no outflow of liquid. heterotrophs Microorganisms growing on an organic compound that provides carbon and energy. insect cell culture Cultivation of insect cells in a bioreactor to produce a protein or other product. photoautotrophs Microorganisms that use light for energy and carbon dioxide for their carbon source. plant cell culture Production of plant cells in a bioreactor to produce useful products. protein engineering The design, development, and production of new protein products with properties of commercial value. tissue engineering The design, development, and production of tissue cells (biomaterials) for use on or in humans.

I. INTRODUCTION The importance of the bioreactor is recorded in early history. The Babylonians apparently made beer before 5000 BC. Wine was produced in wineskins, which were carefully selected for their ability to produce a beverage that met the approval of the king and other members of his sensory analysis taste panel. Food and beverage product quality depended on art and craftsmanship rather than on science and engineering during the early years of bioreactor selection and utilization. Early recorded history shows that some understood the importance of the reactants and the environmental or operating conditions of the reactor. This allowed leavened bread and cheese to be produced in Egypt more than 3000 years ago.

Bioreactors are vessels or tanks in which whole cells or cell-free enzymes transform raw materials into biochemical products and/or less undesirable byproducts. The microbial The Desk Encyclopedia of Microbiology ISBN: 0-12-621361-5

189

Copyright © 2003 Elsevier Ltd All rights of reproduction in any form reserved

ch15.qxd 10/9/03 7:00 PM Page 190

190

the desk encyclopedia of microbiology

The process of cooking food to render it microbiologically safe for human consumption and to improve its sensory qualities is also an ancient tradition. The process of thermal inactivation of microorganisms through the canning of food to allow safe storage was an important early achievement in bioreactor design and operation. As humans learned to live in cities, waste management including wastewater treatment became a necessity for control of disease. One of the first process engineering achievements was the biological treatment of wastes in bioreactors designed and built by humans for that purpose. Because a significant fraction of the population of a city could die from disease spread by unsanitary conditions, these early bioreactors represented important advancements. After microorganisms were discovered, microbiologists and engineers increased their understanding of the biochemical transformations in bioreactors. Simple anaerobic fermentations for the production of ethyl alcohol, acetone, and butanol were developed. Aerobic and anaerobic treatment of waste-water became widely used. Sanitary engineering became a part of civil engineering education. In the 1940s, the field of biochemical engineering emerged because of developments in the pharmaceutical industry that required large-scale bioreactors for the production of streptomycin and penicillin. Progress in bioreactor design and control resulted from research on oxygen transfer, air and media sterilization, and pH control. The central concern of the early biochemical engineers was the development of bioreactors that could achieve and maintain the chemical and physical environment for the organism that the biochemist/microbiologist recommended. The ability to scale-up from laboratory bioreactors to large fermentors required the development of instrumentation such as the sterilizable oxygen electrode. Early courses in biochemical engineering were concerned with the analysis, design, operation, and control of bioreactors. Although the field of biochemical engineering is less than 60 years old and some of the pioneers are still available to provide a first-person account of those exciting days, great progress has been made in bioreactor engineering. Some of the significant developments in bioreactor technology and their approximate dates are listed in Table 15.1.

II. CLASSIFICATIONS OF BIOREACTORS Several methods have been used to classify bioreactors, including the feeding of media and gases and the

TABLE 15.1 Significant developments in bioreactor technology Development

Yeara

Fermented beverages Pasteur’s discovery of yeast First medium designed for culturing bacteria Trickling filter for wastewater Anaerobic digester Production of citric acid using mold Production of penicillin in a petri dish Production of penicillin in small flasks Hixon and Gaden paper on oxygen transfer Air sterilization in fermentors Continuous media sterilization Aiba, Humphrey, and Millis biochemical engineering textbook on bioreactor design Continuous airlift reactor for production of yeast Advances in instrumentation and computer control Progress in airlift bioreactor design Recombinant DNA technology Insect cells grown in suspension culture Large-scale cell culture to produce interferon Insulin produced using bacteria Bioreactors for fragile cell cultures Textbook on plant cell biotechnology Textbook on protein engineering Textbook on tissue engineering

5000 BC 1857 1860 1868 1881 1923 1928 1942 1950 1950 1952 1965 1969 1970 1973 1973 1975 1980 1982 1988 1994 1996 1997

a The dates are approximate and are indicative of periods of time when advances were progressing from initial studies to published works or commercial use.

withdrawal of products; the mode of operation may be batch, fed batch, or continuous. The classification may be based on the electron acceptor; the design may be for aerobic, anaerobic, or microaerobic conditions. In aerobic processes, the methods of providing oxygen have resulted in mechanically agitated bioreactors, airlift columns, bubble columns, and membrane reactors. The sterility requirements of pure culture processes with developed strains differ from those of environmental mixed-culture processes, which are based on natural selection. In some bioreactors the vessel is made by humans and there are also natural bioreactors, such as the microbial cell, the flowing river, and the field of native grass. In this article, the classification of bioreactors is based on the physical form of the reactants and products.

A. Gas phase reactants or products Oxygen and carbon dioxide are the most common gas phase reactants and products; others include hydrogen, hydrogen sulfide, carbon monoxide, and methane. Oxygen is a reactant in aerobic heterotrophic growth processes, whereas it is a product in photoautotrophic growth. Generally, the concentration of the

ch15.qxd 10/9/03 7:00 PM Page 191

bioreactors

191

reactants and products in the liquid phase in the microenvironment of the cell influences the kinetics of the cellular reaction. Mass transfer to and from the gas phase affects bioreactor performance in most processes with gas phase reactants or products. The anaerobic reactor is designed to exclude oxygen. In some cases, inert gases are bubbled into the anaerobic reactor to provide a gas–liquid interfacial area to remove the product gases. Because the solubility of oxygen in water is very low, the dissolved oxygen in the broth is rapidly depleted if oxygen transfer from the gas to the liquid phase is disrupted in aerobic processes. The distribution of dissolved oxygen throughout the reactor volume and the transient variation affect reactor performance. When mold pellets or biofilms are present, the diffusion of oxygen into the interior should be considered. A significant portion of the bioreactor literature is devoted to oxygen transfer and the methods recommended for the design and operation of aerobic bioreactors. The phase equilibrium relationship is based on thermodynamic data, whereas the rate of oxygen transfer depends on the gas–liquid interfacial area and the concentration driving force. Mechanical agitation increases the gas–liquid interfacial area. Aeration provides the supply of oxygen, and it affects the gas–liquid interfacial area. Oxygen has been supplied by permeation through membranes in cultures in which bubbles may damage shear-sensitive cells. The membrane area and concentration driving force determine the oxygen transfer rate in these bioreactors. Most large-scale bioreactors have either oxygen or carbon dioxide among the reactants or products. In many anaerobic fermentations the formation of carbon dioxide results in bubbling, and often no additional mixing is required for either mass transfer or suspension of the microbial cells. Methane is produced through anaerobic digestion of waste products. It is also a product of microbial action in landfills, bogs, and the stomach of the cow. Packed-bed bioreactors are used to biodegrade volatile organic compounds in air pollution control applications. The rhizosphere provides a natural environment in which many volatile compounds in soil are transformed by microbial and plant enzymes.

and, in some cases, the products. The Monod kinetic model and the Michaelis–Menten kinetic model show that many biochemical reactions have first-order dependence on reactant (substrate) concentration at low concentrations and zero-order dependence at higher concentrations. Rates are directly proportional to concentrations lower than 10 mg/liter for many reactants under natural environmental conditions. At very high concentrations, inhibition may be observed. Hydrocarbons that are relatively insoluble in the water phase, such as hexadecane, may also be reactants or substrates for biochemical reactions. Microbial growth on hydrocarbons has been observed to occur at the liquid–liquid interface and in the water phase. The oxygen requirements are greater when hydrocarbon substrates are used in place of carbohydrates. In the past, there was great interest in the production of microbial protein from petroleum hydrocarbons. The commercialization of the technology was most extensive in the former Soviet Union. The airlift bioreactor is uniquely suited for this four-phase process because of the tendency of the hydrocarbon phase to migrate to the top of the fermentor. The hydrocarbons are found suspended as drops in the water phase, adsorbed to cells, and at the gas–liquid interface. The cells are found adsorbed to hydrocarbon drops, suspended in the water phase, and at the gas–liquid interface. In the airlift fermentor, the vertical circulation mixes the hydrocarbons and cells that have migrated to the top of the fermentor with the broth that enters the downflow side of the column. One of the oldest and most widely practiced fermentations is the microbial production of ethanol and alcoholic beverages such as beer and wine. Because ethanol inhibits fermentation at high concentrations, the process of inhibition has been extensively studied for this fermentation. Ethanol affects the cell membrane and the activities of enzymes. This inhibition limits the concentration of ethanol that can be obtained in a fermentor. Because ethanol is also produced for use as a motor fuel, there is still considerable research on ethanol production. Because the cost of the substrate is a major expense, inexpensive raw materials such as wastes containing cellulose have been investigated.

B. Liquid phase reactants or products

C. Solid phase reactants or products

Many bioreactors have liquid phase reactants and products. Ethanol, acetone, butanol, and lactic acid are liquid products that can be produced by fermentation. The kinetics of biochemical reactions depends on the liquid phase concentrations of the reactants

There are many examples of bioreactors with solid phase reactants. The cow may be viewed as a mobile bioreactor system that converts solid substrates to methane, carbon dioxide, milk, and body protein. Although the cow is a commercial success, many

ch15.qxd 10/9/03 7:00 PM Page 192

192

the desk encyclopedia of microbiology

efforts to transform low-cost cellulosic solid waste to commercial products in human-made bioreactors have not achieved the same level of success. Solid substrates such as soybean meal are commonly fed into commercial fermentations. Through the action of enzymes in the fermentation broth, the biopolymers are hydrolyzed and more soluble reactants are obtained. Many food fermentations involve the preservation of solid or semisolid foods such as in the conversion of cabbage to sauerkraut and meats to sausage products. Cereals, legumes, vegetables, tubers, fruits, meats, and fish products have been fermented. Some fermented milk processes result in solid products such as cheeses and yogurts. Other examples include the composting of yard wastes, leaching of metals from ores, silage production, biodegradation of crop residues in soil, microbial action in landfills, and the remediation of contaminated soil. In many of these fermentations, mixing is difficult or expensive. Transport of essential reactants may depend on diffusion; the concentrations of reactants and products vary with position. Rates may be limited by the transport of essential reactants to the microorganisms. Most compounds that are present as solids in bioreactors are somewhat soluble in the water phase. For reactants that are relatively insoluble, biochemical reaction rates may be directly proportional to the available interfacial area. The surface of the solid may be the location of the biochemical transformation. An example of microorganisms growing on the surface of a solid substrate is mold on bread. To design bioreactors for solid substrates and solid products, the solubility and the transport processes should be considered as well as the kinetics of the process. Recently, there has been considerable progress in tissue engineering. The rational design of living tissues and the production of these tissues by living cells in bioreactors are advancing rapidly because of progress in systems design and control for both in vitro flow reactors and in vivo maintenance of cell mass.

In the activated sludge process, this is done by allowing the biomass to flocculate and settle; it is then recycled. The trickling filter retains biomass by allowing growth on the surfaces of the packing within the bioreactor. A variety of immobilized cell reactors and immobilized enzyme reactors have been designed and operated because of the economy associated with reuse of cells and enzymes. In the anaerobic production of ethanol, lactic acid, and the other fermentation products, the product yield is greatest when the organisms are not growing and all the substrate is being converted to products. Continuous processes can be designed in which most of the cells are retained and the limiting maximum product yield is approached. Ultra-filtration membrane bioreactors have been used to retain cells, enzymes, and insoluble substrates. In nature, cells are retained when biofilms form along flow pathways. The biofilms allow microorganisms to grow and survive in environments in which washout would be expected. The excellent quality of groundwater is the result of microbial biodegradation and purification under conditions in which microbial survival is enhanced by biofilm formation and cell retention on soil and rock surfaces. The ability of microorganisms to survive even after their food supply appears to be depleted is well established; this is the reason that there are microorganisms almost everywhere in nature. When spills occur, organic substances will often be degraded by microorganisms, if the nutritional environment is balanced. Nitrogen, phosphorous, and other inorganic nutrients often must be added. The concentration of cells adsorbed to the surface and the concentration in the water phase depends on an adsorption phase equilibrium relationship and the operating conditions. In many environmental applications, most of the cells are adsorbed to surfaces. However, in large-scale fermentors with high cell concentrations and rich media feeds, only a small fraction of the cells are found on surfaces.

E. Photobioreactors D. Microorganisms in bioreactors The rate of reaction in bioreactors is often directly proportional to the concentration of microbial biomass. In biological waste treatment, the influent concentration of the organic substrate (waste) is relatively low, and the quantity of microbial biomass that can be produced from the waste is limited. The economy of the operation and the rate of biodegradation are enhanced by retaining the biomass in the bioreactor.

Light is the energy source that drives photoautotrophic growth processes. Because light is absorbed by the growing culture, the intensity decreases rapidly as the distance from the surface increases. Photobioreactors are designed to produce the quantity of product that is desired. Heat transfer is an important design aspect because any absorbed light energy that is not converted to chemical energy must be dissipated as heat.

ch15.qxd 10/9/03 7:00 PM Page 193

193

bioreactors

III. PRINCIPLES OF BIOREACTOR ANALYSIS AND DESIGN

IV. SENSORS, INSTRUMENTATION, AND CONTROL

The basic principles of bioreactor analysis and design are similar to those for chemical reactors; however, many biochemical processes have very complex biochemistry. The chemical balance equations or stoichiometry of the process must be known or investigated. The yield of microbial biomass and products depends on the genetics of the strain and the operating conditions. The consistency of data from experimental measurements can be evaluated using mass balances such as the carbon balance and the available electron balance. Microorganisms obey the laws of chemical thermodynamics; some heat is produced in heterotrophic growth processes. The free energy change is negative for the complete system of biochemical reactions associated with heterotrophic growth and product formation. Thus, the chemical energy available for growth and product formation decreases as a result of microbial assimilation of the reactants. The rate of growth and product formation depends on the number of microorganisms and the concentrations of the nutrients. The kinetics of growth and product formation is often written in terms of the concentration of one rate-limiting substrate; however, in some cases, more than one nutrient may be rate limiting. The kinetics must be known for rational design of the bioreactor. Heat is evolved in microbial bioreactors. For aerobic processes, the quantity of heat generated (heat of fermentation) is directly proportional to the oxygen utilized. Thus, the heat transfer and oxygen transfer requirements are linked by the energy regularity of approximately 450 kJ of heat evolved per mole of oxygen utilized by the microorganisms. Transport phenomena is widely applied in bioreactor analysis and design. Many fermentation processes are designed to be transport limited. For example, the oxygen transfer rate may limit the rate of an aerobic process. Bioreactor design depends on the type of organism and the nutritional and environmental requirements. For example, in very viscous mycelial fermentations, mechanical agitation is often selected to provide the interfacial area for oxygen transfer. Likewise, animal cells that grow only on surfaces must be cultured in special bioreactors, which provide the necessary surface area and nutritional environment. In other cases, animals are used as the bioreactors because the desired biochemical transformations can best be achieved by competitively utilizing animals; cost and quality control are both important when food and pharmaceutical products are produced.

The ability to measure the physical and chemical environment in the fermentor is essential for control of the process. In the past 50 years, there has been significant progress in the development of sensors and computer control. Physical variables that can be measured include temperature, pressure, power input to mechanical agitators, rheological properties of the broth, gas and liquid flow rates, and interfacial tension. The chemical environment is characterized by means of electrodes for hydrogen ion concentration (pH), redox potential, carbon dioxide partial pressure, and oxygen partial pressure. Gas phase concentrations are measured with the mass spectrometer. Broth concentrations are measured with gas and liquid chromatography; mass spectrometers can be used as detectors with either gas or liquid chromatography. Enzyme thermistors have been developed to measure the concentrations of a variety of biochemicals. Microbial mass is commonly measured with the spectrophotometer (optical density) and cell numbers through plate counts and direct microscopic observation. Instruments are available to measure components of cells such as reduced pyridine nucleotides and cell nitrogen. Online biomass measurements can be made using a flow cell and a laser by making multi-angle light scattering measurements. Multivariate calibration methods and neural network technology allow the data to be processed rapidly and continuously such that a predicted biomass concentration can be obtained every few seconds. The basic objective of bioreactor design is to create and maintain the environment that is needed to enable the cells to make the desired biochemical transformations. Advances in instrumentation and control allow this to be done reliably.

V. METABOLIC AND PROTEIN ENGINEERING Genetic modification has allowed many products to be produced economically. With the use of recombinant DNA technology and metabolic engineering, improved cellular activities may be obtained through manipulation of enzymatic, regulatory, and transport functions of the microorganism. The cellular modifications of metabolic engineering are carried out in bioreactors. Successful manipulation requires an understanding of the genetics, biochemistry, and physiology of the cell. Knowledge of the biochemical

ch15.qxd 10/9/03 7:00 PM Page 194

194

the desk encyclopedia of microbiology

pathways involved, their regulation, and their kinetics is essential. Living systems are bioreactors. Through metabolic engineering, man can modify these living bioreactors and alter their performance. Metabolic engineering is a field of reaction engineering that utilizes the concepts that provide the foundation for reactor design, including kinetics, thermodynamics, physical chemistry, process control, stability, catalysis, and transport phenomena. These concepts must be combined with an understanding of the biochemistry of the living system. Through metabolic engineering, improved versions of living bioreactors are designed and synthesized. Although many products are produced in microbial cells, other cell lines, including insect cells, mammalian cells, and plant cells, are utilized for selected applications. The science to support these various living bioreactors is growing rapidly and the number of different applications is increasing steadily. The choice of which organism to select for a specific product must be made carefully, with consideration of biochemistry, biochemical engineering, safety, reliability, and cost. Both production and separation processes affect the cost of the product; however, the costs of product development, testing, regulatory approval, and marketing are also substantial. Proteins with specific functional properties are being designed, developed, and produced through applications of protein engineering. Through molecular modeling and computer simulation, proteins with specific properties are designed. Protein production may involve applications of recombinant DNA technology in host cell expression bioreactors. An alternative is the production of a protein with the desired amino acid sequence through direct chemical synthesis.

VI. STABILITY AND STERILIZATION Although beneficial genetic modification has led to many successful industrial products, contamination and genetic mutations during production operations have resulted in many batches of useless broth. Batch processes are common in bioreactors because of the need to maintain the desired genetic properties of a strain during storage and propagation. Continuous operation is selected for mixed-culture processes such as wastewater treatment, in which there is natural selection of effective organisms. Bioreactors that are to operate with pure cultures or mixed cultures from selected strains must be free of contamination; i.e., the reactor and associated instrumentation must be sterilizable. The vessels that

are to be used for propagation of the inoculum for the large-scale vessel must also be sterilizable. Methods to sterilize large vessels, instrumentation, and connecting pipes are well developed; however, there is a continuous need to implement a wide variety of good manufacturing practice principles to avoid contamination problems. Steam sterilization has been widely applied to reduce the number of viable microorganisms in food and in fermentation media. As temperature increases, the rates of biochemical reactions increase exponentially until the temperature affects the stability of the enzyme or the viability of the cell. The Arrhenius activation energies, which have been reported for enzymatic reactions and rates of cell growth, are usually in the range of 20–80 kJ/g/mol, whereas activation energies for the thermal inactivation of microorganisms range from 200 to 400 kJ/g/mol. Many of the preceding principles also apply to the thermal inactivation of microorganisms in bioreactors. When solids are present in foods or fermentation media, heat transfer to the interior of the solids occurs by conduction. This must be considered in the design of the process because of the increase in the required sterilization time.

VII. CONCLUSIONS Bioreactors are used for a variety of purposes. The knowledge base for their application has increased significantly because of the advances in chemical, biochemical, and environmental engineering during the past 60 years.

BIBLIOGRAPHY Alleman, B. C., and Leeson, A. (Eds.) (1999). “Bioreactor and Ex Situ Biological Treatment Technologies: The Fifth International In Situ and On-Site Bioremediation Symposium: San Diego, California.”Battelle Press: Columbus OH. Asenjo, J. A., and Merchuk, J. C. (Eds.) (1995). “Bioreactor System Design.” Dekker, New York. Bailey, J. E., and Ollis, D. F. (1986). “Biochemical Engineering Fundamentals,” 2nd ed. McGraw-Hill, New York. Barford, J. P., Harbour, C., Phillips, P. J., Marquis, C. P., Mahler, S., and Malik, R. (1995). “Fundamental and Applied Aspects of Animal Cell Cultivation.” Singapore Univ. Press, Singapore. Blanch, H. W., and Clark, D. S. (1996). “Biochemical Engineering.” Dekker, New York. Carberry, J. J., and Varma, A. (Eds.) (1987). “Chemical Reaction and Reactor Engineering.” Dekker, New York. Characklis, W. G., and Marshall, K. C. (Eds.) (1990). “Biofilms.” Wiley–Interscience, New York. Christi, M. Y. (1989). “Airlift Bioreactors.” Elsevier, New York.

ch15.qxd 10/9/03 7:00 PM Page 195

195

bioreactors Cleland, J. L., and Craik, C. S. (Eds.) (1996). “Protein Engineering: Principles and Practice.” Wiley, New York. Cookson, J. T. (1995). “Bioremediation Engineering.” McGraw-Hill, New York. Drioli, E., and Giorni, L. (1999). “Biocatalytic Membrane Reactors.” Taylor & Francis, London, UK. Endress, R. (1994). “Plant Cell Biotechnology.” Springer-Verlag, Berlin. Erickson, L. E., and Fung, D. Y. (Eds.) (1988). “Handbook on Anaerobic Fermentations.” Dekker, New York. Fan, L. T., Gharpuray, M. M., and Lee, Y. H. (1987). “Cellulose Hydrolysis.” Springer-Verlag, Heidelberg. Goosen, M. F. A., Daugulis, A. J., and Faulkner, P. (Eds.) (1993). “Insect Cell Culture Engineering.” Dekker, New York. Lanza, R. P., Langer, R. S., and Chick, W. L. (Eds.) (1997). “Principles of Tissue Engineering.” Academic Press, San Diego. Lubiniecki, A. S. (Ed.) (1990). “Large-Scale Mammalian Cell Culture Technology.” Dekker, New York. Mitchell, D. A., Berovic, M., and Krieger, N. (2000). Biochemical engineering aspects of solid state bioprocessing. Adv. Biochem. Eng. Biotechnol. 68, 61–138. Moo-Young, M. (Ed.) (1988). “Bioreactor Immobilized Enzymes and Cells: Fundamentals and Applications.” Elsevier, New York.

Nielsen, J. H., and Villadsen, J. (1994). “Bioreaction Engineering Principles.” Plenum, New York. Schugerl, K. (2000). Development of bioreaction engineering. Adv. Biochem. Eng. Biotechnol. 70, 41–76. Shuler, M. L., and Kargi, F. (2002). “Bioprocess Engineering,” 2nd edn. Prentice Hall, Englewood Cliffs, NJ. Sikdar, S. K., and Irvine, R. L. (Eds.) (1998). “Bioremediation: Principles and Practice.” Technomic, Lancaster, PA. Twork, J. V., and Yacynych, A. M. (Eds.) (1990). “Sensors in Bioprocess Control.” Dekker, New York. Van’t Riet, K., and Tramper, J. (1991). “Basic Bioreactor Design.” Dekker, New York.

WEBSITES The Electronic Journal of Biotechnology http://www.ejbiotechnology.info/ List of biotechnology organizations and research institutes http://www.ejb.org/feedback/borganizations.html

ch16.qxd 10/9/03 7:01 PM Page 196

16 Bioremediation Joseph B. Hughes, C. Nelson Neale, and C. Herb Ward Rice University

GLOSSARY

ex situ bioremediation Biological treatment of excavated or removed contaminated media. immobilization Chemical and/or physical processes by which contaminants become strongly associated or sorbed with a soil matrix or sludge and desorption is limited. in situ bioremediation Treatment without physical disruption or removal of contaminated media. land treatment Accelerated biodegradation of contaminated media through application to surface soils to enhance aeration and, in some cases, to allow for nutrient amendment. microcosm Highly controlled laboratory-scale apparatus used to model or simulate the fate or transport of compounds under the biological, chemical, and physical conditions found in the natural environment. plume Dissolved contaminants emanating from a source region due to groundwater transport processes. unsaturated zone Region which spans the area located just beneath the surface and directly above the water table.

bioattenuation The nonengineered, natural decomposition of organic contaminants in soil and groundwater systems. biochemical markers Easily monitored (e.g., substrate-specific microbial population) or chemical (e.g., metabolic intermediates and end products) indicators of biodegradation or biotransformation. biodegradation Metabolism of a substance by microorganisms that yields mineralized end products. bioslurry treatment Accelerated biodegradation of contaminants by the suspension of contaminated soil or sediment in water through mixing energy. biotransformation Microbially mediated process in which the original compound is converted to secondary or intermediate products. bioventing Accelerated biodegradation of contaminants in contaminated subsurface materials by forcing and/or drawing air through the unsaturated zone. cometabolism Fortuitous metabolism of a compound by a microorganism that neither yields energy directly nor produces a metabolic product that can subsequently be involved in energy metabolism. composting Accelerated biodegradation of contaminants at high temperatures by aerating and adding bulking agents and possibly nutrients to waste in a compost pile. The Desk Encyclopedia of Microbiology ISBN: 0-12-621361-5

Bioremediation is defined in this article as the process by which microorganisms are stimulated to rapidly degrade hazardous organic contaminants to environmentally safe levels in soils, sediments, subsurface materials, and groundwater. Biological remediation processes have also recently been devised to either precipitate or effectively immobilize

196

Copyright © 2003 Elsevier Ltd All rights of reproduction in any form reserved

ch16.qxd 10/9/03 7:01 PM Page 197

197

bioremediation inorganic contaminants such as heavy metals; however, treatment of inorganic contaminants will not be included in this definition of bioremediation. Stimulation is achieved by the addition of growth substrates, nutrients, terminal electron acceptor, electron donors, or some combination therein, resulting in an increase in contaminant biodegradation and biotransformation. The microbes involved in bioremediation processes may obtain both energy and carbon through the metabolism of organic contaminants. In some cases, metabolism occurs via a cometabolic process or by a terminal electron-accepting process. Independent of the metabolic pathways, bioremediation systems are designed to degrade hazardous organic contaminants sorbed to soils and sediments or dissolved in water. Bioremediation of contaminants may occur in situ or within the contaminated soil, sediments, or groundwater. Alternatively, the contaminated media may be removed and treated using ex situ techniques.

I. INTRODUCTION With the advent of petroleum refining and manufacture of synthetic chemicals, many potentially hazardous organic compounds have been introduced into the air, water, and soil. One method for removing these undesirable compounds from the environment is bioremediation, an extension of carbon cycling. Given the appropriate organism(s), time, and growth conditions, a variety of organic compounds, such as oil and petroleum products, creosote wastes, and a variety of synthetic organic chemicals, can be metabolized to innocuous materials, usually carbon dioxide (CO2), water, inorganic salts, and biomass (mass of bacterial cells); however, metabolic byproducts, some of which are undesirable, may accumulate when biodegradation of compounds is incomplete. Bioremediation is normally achieved by stimulating the indigenous microflora (naturally occurring microorganisms) present in or associated with the material to be treated. In instances in which the indigenous microflora fails to degrade the target compounds or has been inhibited by the presence of toxicants, microorganisms with specialized metabolic capabilities may be added (bioaugmentation). The technical basis for modern bioremediation technology has a very long history (e.g., composting of organic wastes into mulch and soil conditioners). Bioremediation technology has grown to include the biological treatment of sewage and wastewater, food processing wastes, agricultural wastes, and, recently, contaminants in soils and groundwater. In this article, bioremediation is defined and limited to the biological treatment of organic contaminants. First, we present

important background information on the metabolic processes that drive bioremediation, the requirements for the stimulation of specific metabolic processes, and the influence of contaminant behavior and distribution on contaminant availability for microbial uptake. This discussion is followed by sections outlining favorable growth conditions for microorganisms that are capable of degrading common classes of organic contaminants found in soil, sediments, and groundwater. Engineered systems used for the treatment of contaminated media are then presented.

II. BACKGROUND SCIENCES The following two sections describe the fundamental metabolic processes that govern bioremediation as well as biodegradation and biotransformation characteristics of selected classes of organic contaminants. A more thorough treatment of the subject matter may be found in many texts and monographs, and the reader is encouraged to consult these materials for further information. Some of the more notable and recent references include Microbial Transformation and Degradation of Toxic Organic Chemicals (Young and Cerniglia, 1994), Biological Degradation and Bioremediation of Toxic Chemicals (Chaudry, 1994), Biodegradation and Bioremediation (Alexander, 1994), Biology of Microorganisms (Madigan et al., 1997), and Biodegradation of Nitroaromatic Compounds (Spain, 1995).

A. Metabolic processes The metabolism of organic contaminants can be broadly differentiated by the ability of the organisms to gain energy for cell growth from the process. If the metabolism of a compound provides energy for cell maintenance and division, the contaminant is referred to as a primary substrate. In some cases, a compound is metabolized and provides the cell with energy but does not support growth. Contaminants of this type are referred to as secondary substrates. If a compound is transformed without benefit of the cell (no energy or carbon provided for use by the organism) while the cell is obtaining energy from another transformable compound, the biotransformation is referred to as cometabolic. Finally, an additional classification has been recently identified in which some contaminants are capable of serving as the terminal electron acceptor in the respiratory chain of certain anaerobic (without oxygen) bacteria. In this case, energy is not obtained from the contaminant itself, but its transformation is a component of metabolic processes that provide energy to the cell for growth.

ch16.qxd 10/9/03 7:01 PM Page 198

198

the desk encyclopedia of microbiology

B. Growth requirements

C6H6 in water is approximately 1800 mg/liter, the availability of oxygen limits the extent to which hydrocarbons may be biodegraded.

An essential element of bioremediation processes is the ability to sustain enhanced levels of metabolic activity for extended periods of time. To accomplish this objective, an assessment of conditions at contaminated sites is conducted to determine limiting factors that will be manipulated in an engineered process. A comprehensive list of considerations in this assessment is provided in Table 16.1. For naturally occurring organic compounds (i.e., petroleum hydrocarbons), the availability of oxygen as an electron acceptor is often the primary limiting factor. This can be demonstrated through an evaluation of the stoichiometry of hydrocarbon mineralization, as is shown here for benzene or C6H6 (a common contaminant of concern at sites at which gasoline has been spilled):

C. Bioaugmentation For contaminant metabolism to occur in a bioremediation system, organisms with the genetic capacity to transform compounds of interest must be present. Experience has demonstrated that this is often the case in media in which contamination has been present for even short time periods. In certain cases, the addition of organisms acclimated to specific contaminants, or bioaugmentation, may decrease the duration of lag phases. The ability to effectively bioaugment bioremediation systems is a function of the process used. Bioaugmentation is best suited for processes in which contaminated soil or sediments have been excavated and can be mixed or tilled. The bioaugmentation of in situ processes is more difficult because of difficulties in uniformly distributing cells throughout a porous medium. Few cases exist in which bioagumentation of contaminated groundwater aquifers has proven beneficial.

C6H6 7.5O2→6CO2 3H2O For the complete mineralization of 1 mol of C6H6, 7.5 mol of oxygen will be consumed. Water, in equilibrium with the atmosphere, contains approximately 8 mg/liter dissolved oxygen, which can support the oxidation of 2.6 mg/liter C6H6. Since the solubility of

TABLE 16.1 Requirements for microbial growth in bioremediation processes Requirement

Description

Carbon source

Carbon contained in many organic contaminants may serve as a carbon source for cell growth. If the organism involved is an autotroph, CO2 or HCO–3 in solution is required. In some cases, contaminant levels may be too low to supply adequate levels of cell carbon, or the contaminant is metabolized via cometabolism. In these cases the addition of carbon sources may be required. Energy source In the case of primary metabolism, the organic contaminant supplies energy required for growth. This is not the case when the contaminant is metabolized via secondary metabolism or cometabolism or as a terminal electron acceptor. If the contaminant does not serve as a source of energy, the addition of a primary substrate(s) is required. Electron acceptor All respiring bacteria require a terminal electron acceptor. In some cases, the organic contaminant may serve in this capacity. Dissolved oxygen is a common electron acceptor in aerobic bioremediation processes. Under anaerobic conditions, NO3 , SO24 , SO2 and CO2 may serve as terminal electron acceptors. Certain cometabolic transformations are carried out by fermentative and other anaerobic organisms, in which terminal electron acceptors are not required. Nutrients Nitrogen (ammonia, nitrate, or organic nitrogen) and phosphorus (ortho-phosphate or organic phosphorus) are generally the limiting nutrients. In certain anaerobic systems, the availability of trace metals (e.g., Fe, Ni, Co, Mo, and Zn) can be of concern. Temperature Rates of growth and metabolic activity are strongly influenced by temperature. Surface soils are particularly prone to wide fluctuations in temperature. Mesophilic conditions are generally best suited for most applications (with composting being a notable exception). pH A pH ranging between 6.5 and 7.5 is generally considered optimal. The pH of most ground-water (8.0–8.5) is not considered inhibitory. Absence of toxic Many contaminated sites contain a mixture of chemicals, organic and inorganic, which may materials be inhibitory or toxic to microorganisms. Heavy metals and phenolic compounds are particular concerns. Adequate contact For a contaminant to be available for microbial uptake it must be present in the aqueous between microorganisms phase. Thus, contaminants that exist as nonaqueous phase liquids or are sequestered and substrates within a solid phase may not be readily metabolized. Time This is an important factor in the start-up of bioremediation systems. Even when the first eight considerations in this table are met, lag phases are often observed prior to the onset of activity. In some cases, the dramatic bacterial population shifts that are required for bioremediation will lengthen periods of slow activity. —

ch16.qxd 10/9/03 7:01 PM Page 199

bioremediation

III. ENHANCEMENT OF CONTAMINANT METABOLISM The method by which the rate or extent of contaminant metabolism can be increased in a bioremediation system is governed largely by the substrate-specific metabolic processes that result in its transformation. An understanding of contaminant metabolism is essential. In the following sections, an overview of specific metabolic pathways for common contaminant classes is presented.

A. Monoaromatic hydrocarbons As constituents of gasoline, diesel, and jet fuels, monoaromatic hydrocarbons enter the subsurface environment due to accidental spills and leaking underground storage tanks (UST). These contaminants are commonly found in the environment in the form of free product entrapped or sorbed to porous media or dissolved in water. Monoaromatics are typically referred to as light nonaqueous phase liquids because their specific gravity is less than that of water. Of particular interest in this class of pollutants are benzene, toluene, ethylbenzene, and xylene isomers (BTEX). The biodegradation of these compounds has been and continues to be studied extensively. Under aerobic (containing oxygen) conditions, all the constituents are rapidly biodegraded as primary substrates. Oxygen is important in this process in two ways. First, it is a substrate in the initial attack of the aromatic ring catalyzed by oxygenase enzymes. Second, oxygen serves as the terminal electron acceptor for respiratory chains. Figure 16. 1 illustrates the dual functionality of oxygen in the metabolism of benzene. The biodegradation of BTEX compounds is not as well characterized under anaerobic conditions as it is under aerobic conditions. Certainly, the biodegradation of all BTEX compounds has been observed under a range of anaerobic electron acceptor conditions, but it does not occur in all cases. In particular, benzene can be recalcitrant under anaerobic conditions. In some cases, however, the metabolism of benzene has been observed in the absence of oxygen. Little is known about the pathways of these processes or the enzymes that may be involved in these reactions. In any case, the anaerobic degradation of BTEX compounds is generally slower than aerobic processes. Thus, bioremediation systems targeted for BTEX remediation are typically operated under aerobic conditions. The basis of most BTEX bioremediation systems is the enhancement of the rate of aerobic metabolism by increasing the availabililty of oxygen in contaminated areas. Several methods for doing so are presented later.

199

B. Polynuclear aromatic hydrocabons Polynuclear aromatic hydrocarbons (PAHs) typically result from activities including combustion of fossil fuels and coal gasification processes, and they may also be found in creosote wastes used in wood preservation. PAHs are generally found sorbed to soils and sediments in the natural environment. This class of chemicals contains many compounds with varying biodegradation and physicochemical characteristics. In general, PAH biodegradation is limited to aerobic metabolism and is initiated by oxygenase attack (similar to that depicted in Fig. 16.1). PAHs of three or fewer rings, including naphthalene, fluorene, and phenanthrene, are known to be primary substrates for bacterial growth. Larger PAHs (i.e., four rings and larger) tend to behave as secondary substrates in the presence of the smaller, more water-soluble PAHs.

C. Phenolic compounds Phenol and chlorinated phenols have historically been used in the treatment or preservation of wood products and have served as bacterial disinfectants. These compounds are biodegraded as primary substrates under aerobic and anaerobic conditions. These compounds are often recalcitrant in the environment due to their toxicity and the low water solubility of certain chlorinated forms (e.g., pentachlorophenol). When present at concentrations lower than toxic thresholds, phenols can be rapidly mineralized by a wide range of microorganisms. As the degree of chlorine substituents increases, the rate of degradation often decreases, especially under aerobic conditions.

D. Chlorinated hydrocarbons Chlorinated methanes, ethanes, and ethenes comprise a group of compounds commonly referred to as chlorinated hydrocarbons (also referred to as chlorinated solvents). These compounds have been used extensively as degreasers, dry cleaning agents, and paint removers, and they are widely present and persistent in the environment. They are common contaminants of subsurface soils and groundwater, and contamination has resulted from leaking storage facilities or improper disposal practices. Due to their high specific gravity and density, chlorinated hydrocarbon compounds may often be referred to as dense nonaqueous phase liquids (DNAPLs). DNAPLs will typically be found near the lower confining unit of an acquifer since their densities are greater than the density of water. Common chlorinated hydrocarbon contaminant compounds include trichloroethane, perchloroethene (PCE),

ch16.qxd 10/9/03 7:01 PM Page 200

200

the desk encyclopedia of microbiology

FIGURE 16.1 The metabolic pathways of benzene biodegradation under aerobic conditions. TABLE 16.2 Summary of the biotransformation processes of chlorinated hydrocarbons Compound

1 Substrate

2 Substrate

Cometabolic substrate

Terminal electron acceptor

Dichloromethane Chloroform Carbon tetrachloride Perchloroethene Trichloroethene Dichloroethenesa Vinyl chloride Hexachloroethane 1,1,1-Trichloroethane

Yes No No No No Nob Yes No No

Yes No No No No No Possible No No

Yes Yes Yes Yes Yes Yes Yes Yes Yes

No No No Yes Yes Yesc Yesc No No

a

Three isomers of dichloroethenes exist: 1,1-dichloroethene, cis-dichloroethene, and trans-dichloroethene. Recent studies have identified oxidative pathways for cis-dichloroethene that may yield energy for growth. c Vinyl chloride and cis-dichloroethene are intermediates during the respiration of perchloroethene and trichloroethene to ethene. b

trichloroethene (TCE), dichloroethene (DCE), carbon tetrachloride, chloroform, and vinyl chloride. The metabolism of chlorinated hydrocarbons is perhaps more diverse than that of any other group of environmental contaminants. Depending on the compound of interest, the electron acceptor condition, and the presence of inducing substrates, the metabolism of chlorinated hydrocarbons may occur through primary metabolism, secondary metabolism, cometabolism, or terminal electron acceptor processes.

Table 16.2 lists common chlorinated hydrocarbon contaminants and the processes by which individual compounds are known to be transformed.

E. Nitroaromatic compounds Nitroaromatics are common pollutants of water and soils as a result of their use in plastics, dyes, and explosives. Typical nitroaromatic contaminants used in explosives manufacture include trinitrotoluene

ch16.qxd 10/9/03 7:01 PM Page 201

201

bioremediation (TNT), hexahydro-1,3,5-trinitro-1,3,5-triazine (RDX), and octahydro-1,3,5,7-tetranitro-1,3,5,7,-tetrazoncine (HMX) (Spain, 1995). The nitro group has a strong electron withdrawing functionality, which previously was thought to reduce the potential for oxygenase attack toward the aromatic ring. Recent studies have demonstrated that certain organisms are capable of oxidizing selected nitroaromatic compounds to obtain energy for growth (i.e., primary metabolism). The activity is generally limited to nitroaromatics containing two or fewer nitro groups. Under anaerobic conditions, nitroaromatic transformation generally yields reduced aromatic products. For example, the product of the complete reduction of nitrobenzene is aniline (aminobenzene). The formation of an aryl amine from an aryl nitro group requires that two intermediate forms be produced; the first is any aryl nitroso intermediate followed by the second intermediate, an aryl hydroxylamine. Recent work has demonstrated the importance of the aryl hydroxylamine intermediate in the ultimate fate of nitroaromatics under anaerobic conditions. The hydroxylamine can be reduced to the amine or undergo more complex reactions that can result in binding with natural organic matter or the formation of an aminophenol through rearrangement reactions.

IV. APPLICATION OF CONTAMINANT METABOLISM IN ENGINEERED SYSTEMS The process of transforming an individual contaminant molecule into a nontoxic form occurs at the enzymatic level. Potential remediation sites contain kilograms to tons of contaminants distributed over large areas. Reconciling the difference in scale between molecular processes and the cleanup of tremendous volumes of contaminated media is a significant engineering challenge. Fundamentally, the application of microscale phenomena to the field-scale bioremediation of large, complex contaminated sites begins with a thorough analysis of site conditions. Key steps in site characterization may include (i) determination of the contaminants present and their concentration and distribution, (ii) delineation of the volume of material undergoing treatment, (iii) evaluation of the physical and chemical state of contaminants, (iv) analysis of the redox conditions at the site, and (v) establishment of site hydrogeologic conditions. Upon completion of this phase of the investigation, an analysis would be conducted to determine whether in situ or ex situ treatment or some other technology would be most appropriate given the site conditions. Regardless of the selected

mode of treatment, systems would be designed to create the appropriate ecological conditions to select for organisms that possess the ability to degrade target contaminants. Furthermore, considerations outlined in Table 16.1 would be evaluated to identify potential limiting factors to bioremediation so that modifications or additions to the treatment scheme could be made to enhance the rates of contaminant biodegradation and biotransformation. Specific bioremediation technologies are discussed in detail later. In all cases, these technologies are predicated on the stimulation of specific metabolic activities. The selection of a bioremediation process begins with the understanding of how specific contaminants may be metabolized. In some cases, metabolism may already be occurring, and application of a bioremediation system to those sites would focus on accelerating the rate of the naturally occurring processes. In other cases, contaminant metabolism may be negligible and conditions may require significant alteration through an engineered process. In all cases, bioremediation technologies intended to distribute metabolic activity throughout a region of contamination that is vastly larger than that of a bacterial cell. Thus, the coupling of microscale metabolic processes with macroscale mass transfer processes is one of the most significant challenges in the development of efficient bioremediation technologies. It should be noted that more detailed information on these technologies may be found in many texts as well as in various collections of monographs. Some of the more pertinent references include In Situ Bioremediation: When Does It Work? (Rittman et al., 1993), Handbook of Bioremediation (Norris et al., 1994), Bioremediation: Field Experience (Flathman et al., 1994), Innovative Site Remediation Technology: Bioremediation (Ward et al., 1995), Bioremediation Engineering: Design and Application (Cookson, 1995), Innovations in Ground Water and Soil Cleanup (Rao et al., 1997), Soil Bioventing: Principles and Practice (Leeson and Hinchee, 1997), Subsurface Restoration (Ward et al., 1997), and Innovative Site Remediation Technology Design & Application: Bioremediation (Dupont et al., 1998).

V. NATURAL BIOATTENUATION A. Overview Natural bioattenuation, sometimes termed intrinsic bioremediation or natural bioremediation, refers to the biodegradation or biotransformation of both subsurface soil and groundwater contaminants through

ch16.qxd 10/9/03 7:01 PM Page 202

202

the desk encyclopedia of microbiology TABLE 16.3 Application of bioremediation treatment options to various classes of contaminantsa Contaminant class Monoaromatic hydrocarbons

In situ

Ex situ

Natural bioattenuation Biostimulation Electron donor delivery Bioventing Permeable reactive barriers Land treatment Composting Bioslurry processes

Chlorinated solvents

Yes Yes No Yes Yes Yes Yes Yes

Yes Yes Yes No Yes No No Yes

Nitroaromatics b

? ? ? No ? Yes Yes Yes

Phenols

PAHs

? Yes ? No ? Yes Yes Yes

? ? No Yes ? Yes Yes Yes

a

Adapted in part from Rao et al. (1997). ?, Undetermined or in developmental stages.

b

microbially mediated processes. This mechanism for contaminant mass reduction represents a key component of the broader remediation process of natural attenuation which focuses on the reduction of contaminant concentration, mass, mobility, and/or toxicity through natural processes, including dilution, dispersion, volatilization, adsorption to solid surfaces, and chemical and biological transformation reactions. Bioattenuation is a nonintrusive process (i.e., does not require a mechanical or engineered system for remediation) and is generally more cost-effective than other in situ and ex situ cleanup strategies. This remediation process has been successfully demonstrated in the mitigation of BTEX contamination resulting from leaking underground storage tanks and may also be applicable to the remediation of other compounds, including chlorinated solvents. The applicability of this treatment scheme has not been determined for PAHs, PCBs, explosives, and pesticides (as shown in Table 16.3; Rao et al., 1997). For many UST sites (47%), natural attenuation is the chief mechanism for groundwater remediation, whereas nearly 67% of U.S. states recognize and implement natural attenuation as a viable alternative for soil and groundwater cleanup (USEPA, 1997). Table 16.4 compares the use of natural attenuation with other remediation technologies (some of which are described later) for both soil and groundwater cleanup. It is important to recognize that natural bioattenuation may not necessarily replace active remediation processes, but it does offer an attractive option to complement techniques that may be very costly to implement. For example, at UST sites, large pools of nonaqueous phase liquids may serve as a continual source for groundwater contamination. Typically, a more intensive strategy is required to remove most of the free-phase contaminant before natural bioattenuation is implemented as a cleanup method. Natural bioremediation should also not be

TABLE 16.4 Use of remediation technologies at UST sitesa Remediation technology Soil remediation Soil washing Bioventing Incineration Thermal desorption Landfarming Soil vapor extraction Biopiles Natural attenuation Landfilling Groundwater remediation Biosparging Dual-phase extraction In situ bioremediation Air sparging Pump and treat Natural attenuation

Use at UST sites (% of sites) 0.2 0.8 2 3 7 9 16 28 34 2 5 5 13 29 47

a

Source: USEPA (1997).

characterized as a “no action” approach to cleanup; rather, it requires both long-term monitoring of the parent contaminant compound and secondary metabolites and monitoring of other chemical and biological markers that are indicative of the attenuation process (Ward et al., 1997). However, the efficacy of natural attenuation for meeting remedial objectives and managing risk of groundwater contamination is controversial in research and regulatory communities. Its role in soil and groundwater cleanup continues to develop as we learn more about quantifying the processes involved. The rate and extent of contaminant biodegradation are governed by many environmental factors, including contaminant and cell biomass concentrations, temperature, pH, supply of nutrients, adequacy of carbon and energy sources, the presence of toxins such as heavy

ch16.qxd 10/9/03 7:01 PM Page 203

bioremediation metals, availability of contaminants to microorganisms (i.e., contact, contaminant solubility and hydrophobicity, and desorption from solids), time for acclimation, and availability of electron acceptors (Table 16.1). The supply and availability of electron acceptors is often cited as the controlling or limiting factor in the bioattenuation process. Aerobic biodegradation occurs in the presence of oxygen, whereas alternate electron acceptors, including nitrate, sulfate, trivalent iron (Fe3), and carbon dioxide, are utilized under anaerobic conditions. For relatively soluble petroleum hydrocarbons such as BTEX, the rate and extent of biological transformation of contaminants is typically much greater under aerobic conditions than under oxygen-limited conditions. For highly chlorinated solvents (e.g., PCE and TCE), the rate and extent of biological transformation, through reductive dechlorination, is greater under anaerobic conditions than under aerobic conditions, whereas less halogenated compounds (e.g., vinyl chloride) may be more amenable to aerobic biodegradation. The oxygen demand exerted by the microorganisms during petroleum hydrocarbon biodegradation generally exceeds the rate of oxygen replenishment, especially in areas of high contaminant concentration (i.e., near the source zone). In fact, the limiting dissolved oxygen concentration for aerobic biodegradation is approximately 2 mg/liter (Rao et al., 1997), although a study by J. Salanitro and coworkers published in Ground Water Monitoring and Review in 1997 suggests that this concentration may be as low as 0.2 mg/liter. The characteristic shape of a groundwater

203

contaminant plume may be partially explained by the presence or absence of oxygen as a terminal electron acceptor. Zones of aerobic biodegradation generally occur on the outermost and leading edges of the plume where contaminated groundwater meets with uncontaminated, well-oxygenated groundwater. Mixing of the two waters via dispersion provides an adequate supply of oxygen for the aerobic biodegradation process. However, contaminant biodegradation in the central region of the plume is generally governed by contaminant-specific anaerobic processes due to the rapid depletion of oxygen in these areas of high metabolic activity. The diffusion rate of oxygen in water is four orders of magnitude less than the diffusion rate in air; thus, the rate of oxygen consumption easily exceeds its rate of transport in water, which results in anaerobic conditions (Norris et al., 1994; Rao et al., 1997). Figure 16.2 presents a typical UST spill and indicates the areas or zones of aerobic and anaerobic biodegradation. The previous discussion illustrates the importance of oxygen as a driving force in the natural aerobic bioattenuation of subsurface contaminants. Oxygen may be naturally delivered to the contaminant plume either through mixing with uncontaminated groundwater or through reaeration from the overlying unsaturated zone. Reaeration may serve as a significant source of oxygen and is governed by many factors, including soil hydrogeologic properties, soil moisture, precipitation, and respiration rate of soil microorganisms. Macroporous sand materials can easily

FIGURE 16.2 The role of oxygen flux in the bioattenuation of hydrocarbon contaminants (adapted in part from Norris et al., 1994).

ch16.qxd 10/9/03 7:01 PM Page 204

204

the desk encyclopedia of microbiology

and rapidly contribute to the oxygen supply of a contaminant plume, whereas the diffusion of oxygen through clay materials is generally impeded, especially in the presence of moisture. Moisture may cause swelling of certain clay soils and reduces the number of air-filled pore spaces in the soil through which oxygen may easily diffuse. The respiration rate of soils may also serve as a significant oxygen sink and will restrict delivery of the gas to a contaminant plume.

B. Demonstration Several demonstrations have been completed to determine the efficacy of natural bioattenuation of contaminants in groundwater. A study by C. Y. Chiang and coworkers, published in Ground Water in 1989, investigated the aerobic bioattenuation of a petroleum hydrocarbon plume at a gas manufacturing plant in Michigan using a large network of monitoring wells. The contaminated aquifer was located 10–25 ft below the surface and was overlain by coarse sand. The initial mass of benzene at the site was approximately 10 kg; after a period of 21 months, the mass had been reduced to approximately 1.3 kg of benzene. A first-order bioattenuation rate of 0.0095/day was determined for the site. Analysis of groundwater samples throughout the study period revealed an inverse relationship between dissolved oxygen and benzene concentration in the plume. Samples from locations in which the BTX concentration was high were coupled with low dissolved oxygen concentrations as a result of increased microbial activity or biodegradation. Likewise, samples with low BTX concentrations contained higher dissolved oxygen concentrations due to the reduction in oxygen demand. Other natural attenuation processes were found to have very little effect on the reduction of benzene contaminant mass. Only 5% of benzene contaminant loss was attributed to volatilization from the groundwater, and sorption was thought to have little impact on the attenuation process based on the low organic carbon content of the aquifer material. Subsequent modeling and microcosm studies confirmed that natural aerobic bioattenuation was a dominant mechanism in the reduction of contaminant mass at this particular site. Bioattenuation of chlorinated solvents has also been demonstrated in field studies. In 1995, L. Semprini and coworkers investigated the biotransformation of chlorinated solvents in groundwater at a site in Michigan. A series of multilevel samplers bounding the width and length of the contaminant plume were used to determine the concentrations of TCE, isomers of DCE, vinyl chloride, and ethane with depth in the sandy aquifer. The results of the study

indicated that the reductive dechlorination of TCE had occurred over time in the plume based on the presence of less chlorinated compounds as well as the presence or absence of biochemical markers, including methane (methanogenesis) and sulfate (sulfate reduction). The transformation of TCE to isomers of DCE (predominantly cis-DCE) was associated with sulfate reducing conditions, whereas further transformation of cis-DCE to vinyl chloride and ethene was coupled with areas high in methanogenic activity. Although the mass of original TCE contaminant could not be established, Semprini and coworkers suggested that, on the basis of calculated flux rates for each of the contaminant compounds, TCE reduction to ethane was on the order of 20%.

VI. ENHANCED IN SITU BIOREMEDIATION/ BIOATTENUATION Many in situ bioremediation processes have been designed or engineered to maintain or accelerate the biodegradation of organic contaminants in soil and groundwater by supplying those materials which may limit the breakdown of the contaminant compound. For example, in petroleum hydrocarbon-contaminated environments which are nutrient limited, nitrogen, phosphorus, and other minerals may be added to the environmental system (either through soil tilling or through injection into groundwater) to stimulate growth of the contaminant-degrading microbial population. In many cases, the natural supply of oxygen is rapidly extinguished in areas of high metabolic activity, and therefore limits the rate and extent of biodegradation. Oxygen replenishment may also be achieved by soil tilling or by the introduction of air, pure oxygen, hydrogen peroxide (H2O2), or other oxygen-releasing materials into soil and groundwater systems. Microorganisms that have been previously acclimated to the contaminant of interest may also be mixed into soil or groundwater environments to stimulate the rate of contaminant decomposition. A recent approach to enhancing in situ treatment of chlorinated solvents is the introduction of electron donors such as hydrogen, lactic acid, etc. Installation of reactive barrier walls into groundwater aquifers may also improve the bioremediation of a variety of organic contaminants.

A. Biostimulation 1. Overview The earliest system of enhanced or engineered in situ bioremediation of groundwater was designed

ch16.qxd 10/9/03 7:01 PM Page 205

bioremediation by R. L. Raymond and co-workers with Sun Research and Development Company in Philadelphia and described in a 1978 American Petroleum Institute report titled “Field Application of Subsurface Biodegradation of Gasoline in a Sand Formation.” The “Raymond process” was a patented system [U.S. Patent No. 3,846,290 (1974)] in which biodegradation of petroleum hydrocarbons (namely, gasoline constituents) in groundwater by indigenous subsurface microorganisms was stimulated through the injection of nutrients and oxygen. Although subsurface microorganisms require many minerals, the two most common nutrient amendments are nitrogen (as ammonium or nitrate) and phosphorus (as phosphate). The necessary mass of each nutrient may be determined stoichiometrically, and the ratio between carbon, nitrogen, and phosphorus is typically 100:10:1 (C:N:P) (Norris et al., 1994). Many compounds have been used to increase the dissolved oxygen concentration of contaminated groundwater aquifers. Initial attempts to increase dissolved oxygen concentration in groundwater aquifers relied on air sparging of water before injection into the subsurface or sparging air into wells; however, the low solubility of oxygen in water (8–12 mg/liter) precluded maximum contaminant biodegradation. Subsequent methods for oxygen delivery resulted in greater solubility of the gas. Dissolved oxygen concentrations increase to approximately 40–50 mg/liter when sparging with pure oxygen, whereas injection of H2O2 can easily result in dissolved oxygen concentrations in the range of 250–500 mg/liter and higher, although H2O2 may be toxic to the microorganisms at higher concentrations (Rao et al., 1997). Oxygen-releasing compounds (e.g., ORC®, marketed by Regenesis, Inc.) are a more recent innovation for meeting the oxygen demand of microorganisms in subsurface environments. These formulations promote the slow release of oxygen which is produced when magnesium peroxide (the typical active ingredient in these compounds) reacts with water. Oxygen-releasing compounds may be added as a slurry injection or may be placed in a series of wells in the aquifer to form a barrier through which the contaminated groundwater flows. Oxygen is therefore introduced into the central regions of the plume (usually anaerobic) to promote aerobic biodegradation of the contaminant compound (DuPont et al., 1998). Bioaugmentation, or the introduction of contaminant-acclimated or genetically engineered microorganisms, is another method for enhancing biodegradation of contaminants. Although bioaugmentation has been successfully implemented in a variety of laboratoryscale and larger ex situ reactor systems, the success of bioaugmentation in soils and groundwater is limited

205

(Alexander, 1994). Some of the problems associated with adding an inoculum to soils or groundwater aquifers are the presence of environmental toxins and bacterial predators, adequate conditions for growth (i.e., nutrients, pH, temperature, etc.; see Table 16.1), contact with target contaminant, and cell movement or distribution (Alexander, 1994). The movement or distribution of the inoculum throughout the contaminated region of interest may be inhibited by sorption to solid surfaces, bacterial mobility, and the structure of the porous media. Microorganisms may be able to move freely through large macropores in the soil; however, their movement may be restricted through soil micropores in which larger pools of the contaminant may often reside (Alexander, 1994). A schematic of a typical biostimulation in situ remediation system is presented in Figure 16.3. Groundwater is initially drawn from a series of recovery wells downgradient from the source of contamination. The groundwater is then pumped to an aboveground treatment facility at which both nutrient amendments and oxygen (through either sparging or H2O2 injection) are added after the water is initially passed through a contaminant treatment scheme. After addition of chemicals, the groundwater is returned to the aquifer either through an injection well or through an infiltration gallery above the zone of contamination. This process continues until the contaminant concentration is reduced to the target level (DuPont et al., 1998). 2. Demonstration J. T. Wilson, J. M. Armstrong, and H. S. Rifai reported the bioremediation of an aviation gasoline spill at a Coast Guard air station in Traverse City, Michigan,

FIGURE 16.3 Simplified schematic of an in situ biostimulation remediation system (Raymond process).

ch16.qxd 10/9/03 7:01 PM Page 206

206

the desk encyclopedia of microbiology

using nutrient and H2O2 amendments (Flathman et al., 1994). The shallow sandy aquifer underlying the site was contaminated by approximately 25,000 gallons of aviation fuel emanating from an UST. Analysis of soil cores taken from the site identified a vertical zone of petroleum hydrocarbon contamination, and the remediation process was aimed at reducing the concentration of BTEX dissolved in the groundwater and bound in the aquifer material. A series of six monitoring wells were placed downgradient of the infiltration wells to quantify BTEX loss due to biodegradation. Nutrients introduced into the contaminated region included ammonium chloride, disodium phosphate, and potassium phosphate, whereas H2O2 was incrementally injected into the subsurface to allow for microbial acclimation (to a maximum of approximately 400 mg/liter oxygen). A high degree of BTEX removal was observed in each of the six monitoring wells. For example, BTEX was reduced from 1200 to 380 mg/liter at a monitoring well 83 ft from the infiltration wells. Benzene was most amenable to biodegradation in the groundwater, whereas the isomers of xylene were most resistant to biodegradation. Remediation of the aquifer material was also investigated by taking soil cores after completion of the experiment. The results indicated that the process generally led to a reduction in BTEX concentrations in the aquifer materials; however, concentrations of total petroleum hydrocarbons at each of the monitoring points remained fairly high. In areas in which oxygen concentration was low, nitrate was determined to be the terminal electron acceptor supporting most of the BTEX biodegradation.

B. Electron donor delivery 1. Overview The delivery of electron donors to groundwater systems is another in situ biostimulation approach to promote or accelerate the biotransformation of chlorinated solvent compounds. Unlike many of the petroleum hydrocarbon contaminants that may be directly utilized by microorganisms to obtain cell energy and promote cell growth, chlorinated solvents are not often used as primary substrates. However, chlorinated solvents may be biodegraded or biotransformed under either aerobic or anaerobic conditions. Under aerobic conditions, biodegradation can be achieved through cometabolic processes in which enzymes capable of breaking down the target contaminant are fortuitously expressed during the biodegradation of other primary substrates. More frequently, chlorinated compounds are biotransformed to less chlorinated products under anaerobic conditions through the process of reductive

dechlorination. Microorganisms require a sufficient supply of electron donors in order to carry out the reductive dechlorination process. A variety of potential electron donors have been evaluated both in field and in laboratory studies and include acetate, methane, hydrogen, ammonia, benzoate, lactate, and methanol. Cocontaminants such as petroleum hydrocarbons may also serve as electron donors in subsurface environments. Many of the same difficulties experienced with the introduction of nutrients, microorganisms, and other materials into subsurface environments also occur with electron donor delivery (Norris et al., 1994). Excessive microbial growth may result near the point of injection due to the high electron donor concentration and availability. This problem may be further exacerbated by the simultaneous introduction of other materials that are essential for bacterial growth (i.e., oxygen and nutrients). 2. Demonstration In 1997, in a paper titled “Scale-up Issues for in situ Anaerobic Tetrachloroethene Bioremediation” M. D. Lee and co-workers reported laboratory and field experiments relating to electron donor selection and delivery to enhance anaerobic PCE biotransformation. Microcosm studies were used to determine the most efficient substrates to effect PCE dechlorination. The substrates included yeast extract [38% carbon (C)], wastewater (4.5% C), cheese whey permeate (26% C), molasses (29% C), corn steep liquor (17% C), manure tea (3% C), sodium benzoate (58% C), and acetate (29% C). The field site under investigation was a landfill associated with a chemical plant in Texas. The saturated zone of the aquifer underlying this site consisted primarily of sand. A series of injection, withdrawal, and monitoring wells were used in the field experiments which were designed to determine an optimal electron donor delivery system. The initial microcosm study investigated dechlorination of chlorinated ethenes (23 M PCE, 0.6 M TCE, and 3.3 M 1,2-DCE) using yeast extract, wastewater, molasses, corn steep liquor, and manure tea at varying initial carbon loadings. A high degree of carbon utilization was achieved in each microcosm (60% TOC removal), and PCE was transformed to vinyl chloride at carbon concentrations 60 mg/liter. Cell counting experiments indicated a positive relationship between the amount of carbon in the microcosm and the number of microorganisms present. A second microcosm experiment, using sodium benzoate, sodium acetate, corn steep liquor, and molasses, studied the effect of very high organic loadings on both PCE

ch16.qxd 10/9/03 7:01 PM Page 207

bioremediation dechlorination and microorganism concentration. In contrast to the results from the first study, higher organic loadings inhibited TOC removal and resulted in either no transformation or only partial transformation of PCE. The field tracer experiments compared the injection and distribution of the substrate and nutrient amendments under three conditions: tracer (bromide) injection at the rate of groundwater flux, tracer (iodide) injection at a rate of 60 times groundwater flux, and cross-gradient injection [bromide, substrate, and nutrients (N, P)] at a rate of 3.8 liters/min for the withdrawal wells (two) and the injection well. Distribution or dispersion was enhanced when the injection rate was increased to 60 times the natural groundwater gradient. The third condition, employing the cross-gradient injection scheme, further increased dispersion of the bromide tracer. Measurements of TOC also indicated that sodium benzoate was successfully transported downgradient from the injection well. However, transport of both the injected nutrients and electron acceptor (sulfate) was retarded either by sorption to the aquifer solids or by biodegradation processes.

C. Bioventing 1. Overview Bioventing is another in situ technique designed to stimulate aerobic biodegradation of contaminants by replenishing oxygen levels in oxygen-depleted, unsaturated zone environments. The process was first described by J. T. Wilson and C. H. Ward in an article published in 1986 in the Journal of Industrial Microbiology. R. E. Hinchee, R. R. DuPont, R. N. Miller, and others were responsible for developing and testing the process (Leeson and Hinchee, 1997). Bioventing has been successfully applied at several petroleum hydrocarbon-contaminated sites and may also be implemented to remediate sites contaminated with chlorinated solvents, although it typically requires the addition of substrates to promote cometabolism of the chlorinated hydrocarbons (Norris et al., 1994). A recent report suggests that bioventing is being used at less than 1% of UST contaminated soil sites. However, this percentage translates into the application of bioventing at more than 800 UST sites (USEPA, 1997). Bioventing is designed to emphasize biodegradation over contaminant volatilization; however, the relationship between the two mechanisms may be determined by both the properties of the contaminant of interest (e.g., molecular weight and vapor pressure) and site conditions (Norris et al., 1994;

207

Leeson and Hinchee, 1997). Hydrocarbon contaminants with higher vapor pressures will tend to volatilize with injection of air and may not be suitable for bioventing, whereas biodegradation of heavy hydrocarbons with lower vapor pressures may be achieved using this treatment technology (Cookson, 1995). In addition to contaminant characteristics, soil properties play a major role in the success of bioventing applications. These properties include physical characteristics, such as hydraulic conductivity, gas permeability, and moisture content, as well as properties that will affect the growth of microorganisms (in addition to oxygen supply), such as temperature, pH, and nutrient supply. Bioventing applications are best suited for soils with high gas permeabilities so that the injected air readily moves through the contaminated soil matrix (Leeson and Hinchee, 1997; Ward et al., 1997). The flow of air may be severely restricted in low-permeability soils and will subsequently impact the extent of contaminant biodegradation. Gravel and sand may be considered highly gaspermeable materials, whereas silts and clays represent soils with lower gas permeabilities. Although moisture is required by soil microbes for contaminant metabolism, high percentages of water in the soil will negatively impact the flow of oxygen in the soil pores. The majority of pore spaces in high moisture content soils are filled with water, and the diffusion of oxygen through water is much slower than through air. As with other treatment technologies, microbial growth factors (temperature, pH, and nutrients) may also impact the rate and extent of biodegradation in bioventing applications. Higher temperatures are generally associated with higher rates of contaminant biodegradation. However, Leeson and Hinchee (1997) pointed out that psychrophilic microorganisms at a field test site in Alaska were able to signifycantly biodegrade petroleum hydrocarbons in the subsurface. Nutrient amendment also may need to be considered in deficient subsurface soils. Delivery or transport of nutrients to the target zone may be difficult and will also depend on the soil physical properties. Soils generally contain sufficient nutrients to support the biodegradation of hydrocarbon contaminants, and the advantage of adding nutrients to subsurface soils has not been firmly established (Leeson and Hinchee, 1997). Figure 16.4 presents a schematic of a bioventing system (Leeson and Hinchee, 1997). Bioventing is accomplished by advective flow of air through a series of vent wells into areas that have exhausted their supply of oxygen in the soil gas. Monitoring points are distributed throughout the zone of contamination to determine both the decrease in oxygen concentration

ch16.qxd 10/9/03 7:01 PM Page 208

208

the desk encyclopedia of microbiology

FIGURE 16.4 Schematic of a bioventing system (adapted from Leeson and Hinchee, 1997).

and the increase in the carbon dioxide concentration (primary product of biodegradation). Low airflow rates are usually incorporated into the system design to favor contaminant biodegradation over volatilization. In theory, contaminant biodegradation should be much greater than volatilization; therefore, management of gases that are transported to the soil surface is usually not required. If volatilization is high, a separate extraction and treatment system will have to be installed to handle the vapors being generated by the bioventing system. Bioventing is frequently applied in cooperation with and usually following soil vapor extraction (SVE) for vadose zone remediation.

2. Demonstration One of the earliest and best known bioventing remediation systems was implemented at a site at Hill Air Force Base in Utah where 27,000 gallons of jet fuel were spilled (Leeson and Hinchee, 1997). Fuel contamination migrated to a depth of approximately 65 ft in the unsaturated zone, which consisted primarily of sand and gravel constituents. The average contaminant concentration in the soils was approximately 400 mg/kg. A series of vent wells were installed over the length of the contamination to evaluate the potential of bioventing. Enhanced bioventing using both moisture and nutrient addition was also investigated. The system was initially constructed to investigate contaminant volatilization and capture at higher air flow rates (SVE); however, after several months of testing,

lower airflow rates were employed to favor biodegradation over volatilization. A series of soil samples were taken with depth before and after implementation of the SVE and bioventing remediation system at the site. After application of SVE and bioventing, nearly all of the posttreatment hydrocarbon concentrations in the soils were 5 mg/kg, indicating successful cleanup of the site. It was estimated that approximately 1500 lbs of hydrocarbon fuel was removed through volatilization, whereas 93,000 lbs of fuel was removed through biodegradation. Much of the biodegradation of the fuel was attributed to the bioventing phase of cleanup. Enhanced bioventing by adding moisture and nutrients yielded mixed results. The addition of moisture resulted in a significant increase in contaminant biodegradation, whereas addition of nutrients (N and P) did not enhance removal of the petroleum hydrocarbons.

D. Permeable reactive barriers 1. Overview The use of in situ permeable reactive barriers is another method that introduces reactants which may stimulate the biotic or abiotic transformation of environmental contaminants. As opposed to other in situ remediation methods that rely on injection of reactants (i.e., oxygen and nutrients) and their transport with groundwater flow through the contaminated aquifer, stationary barrier walls containing the reactive porous media are

ch16.qxd 10/9/03 7:01 PM Page 209

209

bioremediation placed into the subsurface using a variety of methods. The contaminated groundwater is directed to and passes through the reactive barrier, thus enabling reaction between the porous media and the contaminant of interest. Permeable reactive barriers are typically used to treat target contaminants, including petroleum hydrocarbons and chlorinated solvents (Table 16.3). A schematic of an in situ permeable reactive barrier is presented in Figure 16.5. The barrier may span the width of the zone of contamination or a series of sheet pilings may be inserted into the subsurface which direct the contaminated water through the reactive barrier. This “funnel and gate” system was devised R. C. Starr, J. A. Cherry, and other researchers at the University of Waterloo and published in 1994 in Ground Water. In cases in which the reactants in the barrier may be rapidly extinguished due to biochemical reactions, these materials may be replenished by designing replacement cassettes that may be easily exchanged in the barrier. Cassettes can also be placed in series to target the biodegradation or biotransformation of a particular contaminant or to remediate groundwater with many contaminants, each of which has specific requirements for degradation. Permeable reactive barrier technologies may also incorporate a series of smaller reactive walls if the size of the plume is large, thus facilitating easier removal and replacement when compared to a single large wall. Starr, Cherry, and others pointed out that the most important considerations for the design and application

of this type of system include (i) time required to effect desired biochemical reaction (combination of groundwater flow rate, reaction rate, influent contaminant concentration, and target effluent contaminant concentration), (ii) the potential formation of hazardous products resulting from reactions within the reactive barrier, and (iii) costs associated with installation and media regeneration. Examples of reactants or amendments used in permeable reactive barrier technologies to enhance contaminant biodegradation and transformation include nutrient amendments (N and P), oxygen addition using oxygen-releasing compounds or biosparging (introduction of air), and introduction of zero valent iron to achieve abiotic reduction of chlorinated compounds. 2. Demonstration J. F. Barker, J. F. Devlin, and co-workers demonstrated the use of permeable reactive barrier remediation technologies at the Canadian Forces Base Borden site in Ontario, Canada. The work was documented in a 1998 report sponsored by the Department of Defence Advanced Applied Technology Demonstration Facility (AATDF) at Rice University. Contaminants within the groundwater plume targeted for remediation using this in situ technology included carbon tetrachloride, PCE, and toluene. Remediation of the highly chlorinated aliphatics (i.e., carbon tetrachloride and PCE) was accomplished through both biotic

FIGURE 16.5 Funnel and gate system to remediate contaminated groundwater using permeable reactive barriers (adapted in part from Starr and Cherry, 1994).

ch16.qxd 10/9/03 7:01 PM Page 210

210

the desk encyclopedia of microbiology

and abiotic reduction processes, whereas biodegradation of the less chlorinated aliphatics and toluene was achieved in an aerobic treatment zone. Three test gates containing different sequences of reactive cassette barriers were installed at the site. Gate 1 consisted of two granular iron cassettes (to effect abiotic reductive dechlorination of chlorinated hydrocarbons) followed by a cassette containing oxygen-releasing compounds. Gate 2 did not contain reactive media and served as a control and/or natural attenuation gate. Gate 3 contained a benzoate injection well (to serve as substrate for microorganisms and to achieve anaerobic conditions for microbially mediated reductive dechlorination) followed by a biosparging wall. Initial spiked contaminant concentrations in the groundwater were approximately 1 or 2 mg/liter for carbon tetrachloride and PCE and 10 mg/liter for toluene. Results from Gate 1 indicated that PCE was completely transformed to ethane and ethene (halflife 0.5 days). Carbon tetrachloride was transformed to chloroform prior to reaching the gate and was rapidly dechlorinated in the iron barrier to dichloromethane. High pH next to the granular iron cassette required emplacement of the ORC downgradient at lower pH to stimulate oxygen release and toluene biodegradation. Gate 2 (natural biodegradation and biotransformation) results also indicated biotransformation of carbon tetrachloride and chloroform (each having a half-life of approximately 11 days). In Gate 3, PCE was successfully transformed to cis-DCE. Subsequent passage of the contaminated groundwater in Gate 3 through the biosparging wall (aerobic zone) resulted in biodegradation of toluene as well as the less chlorinated end products of reductive dechlorination produced in the anaerobic zone (e.g., cis-DCE).

VII. ENHANCED EX SITU BIOREMEDIATION Like in situ processes, enhanced ex situ bioremediation processes are engineered to accelerate the biodegradation or biotransformation of organic contaminants in soils and solids. In many cases, ex situ technologies generally offer better control of the parameters that govern biodegradation than do in situ techniques. Ex situ bioremediation technologies include land farming, composting, and slurry reactors. Land treatment employs nutrient addition and aeration (by tilling) to stimulate biodegradation of land-applied wastes. Composting of contaminated soils and sludges is a high-temperature, exothermic process in which bulking agents (e.g., wood chips and mulch) and, in some cases, nutrients are added to encourage biodegrada-

tion. Finally, ex situ bioslurry reactors may be used to treat either contaminated liquids or solids through nutrient addition, aeration, and bioaugmentation.

A. Land treatment 1. Overview Land treatment or land farming refers to the accelerated aerobic biodegradation of organic wastes in either near-surface or excavated soils through the addition of nutrients, lime (pH control), and moisture and through increased aeration by tilling or other mechanical mixing (Loehr and Malina, 1986). Biodegradation is generally carried out by indigenous soil microorganisms, although some form of bioaugmentation may enhance the rate of degradation. Typically, land farming is an ex situ process whereby the contaminated soils are excavated from a site and sent to an engineered treatment unit (also termed a prepared bed system or reactor); however, in situ methods (nutrient addition and tilling) may be adequate to enhance biodegradation of the soil contaminants near the surface of excavated wastes that are mixed or tilled into the top soil layer (to a depth of approximately 1 ft). A wide variety of organic contaminants have been successfully treated in land farming applications, including petroleum hydrocarbons, pesticides, PCPs, PCBs, and PAHs (Cookson, 1995). Land farming typically requires the addition of nutrient amendments (namely, N and P) to the soils to enhance biodegradation of contaminants. The minerals may be introduced into the soil either as a solid or mixed with water and applied through a spraying system (Cookson, 1995). The spraying system may also provide needed moisture to soils and enhance contaminant biodegradation. Plowing, tilling, or other methods of mechanical mixing of the soils stimulates biodegradation through (i) mixing and distribution of soil amendments (nutrients, lime), (ii) distribution of contaminants (by breaking up soils) and increased contact between contaminants and microorganisms, and (iii) increased aeration of soils (increased oxygen supply) (DuPont et al., 1998; Ward et al., 1995; Cookson, 1995). Figure 16.6 presents an engineered land treatment system. Much like conventional municipal solid waste treatment units or landfills, controls or collection systems may be placed in the land treatment unit. To prevent groundwater contamination, the base of a typical land treatment unit is covered with a highly impermeable clay or geosynthetic (plastic) liner. A series of leachate recovery pipes are then placed near the base of the system to collect wastes that may percolate through the soil. A thick layer of sand covers the collection system, and the contaminated soil is then placed on the

ch16.qxd 10/9/03 7:01 PM Page 211

bioremediation

211

FIGURE 16.6 Typical land treatment unit (inset depicts liner configuration) [adapted in part from Cookson (1995) and Alexander (1994)].

sand layer. Collected leachate may be recycled or sent to another treatment system. A cover or enclosure may be added to the land treatment unit to eliminate rainfall percolation and off-site migration of contaminant vapors (Cookson, 1995; Alexander, 1994). 2. Demonstration In 1979, J. T. Dibble and R. Bartha published work (Soil Science) on the land treatment of hydrocarbon-contaminated soil resulting from a pipeline leak in New Jersey in which approximately 1.9 million liters of kerosene was spilled onto 1.5 hectares (ha) of an agricultural plot. After removal of approximately 200 m3 of heavily contaminated soil, both lime and nutrients (N, P, and potassium) were applied to the field soil (to a depth of 117 cm) to enhance biodegradation. Aerobic biodegradation of the kerosene contaminant was further stimulated by periodic tilling or mixing of the soil to promote distribution of oxygen. The applied nutrient concentrations were 200 kg nitrogen/ha, 20 kg phosphorus/ha, and 17 kg potassium/ha and were added at two different times after an initial loading of 6350 kg/ha of lime. The contaminated soil was characterized as a welldrained, sandy loam soil. Kerosene biodegradation was monitored during a 24-month period to determine the efficiency of the treatment system. Over the course of the experiment, kerosene concentrations in the soil decreased from 8700 mg/kg to very

low levels in the upper 30 cm of the soil. Likewise, kerosene concentration in the lower portion of the soil (30- to 45-cm depth) also decreased over the 24-month period to 3000 mg/kg. Biodegradation was determined to be the primary mechanism for removal (compared to volatilization) based on the disappearance patterns of compounds of varying molecular weight. The rate of biodegradation in the upper portion of the soil was initially greater than the rate of biodegradation in the lower portion. However, more rapid kerosene biodegradation was observed in the lower portion of the soil after 6 months of system operation and evaluation. Temperature also played a major role in the biodegradation of the kerosene contaminants. The greatest decreases in kerosene concentration due to biodegradation occurred during time periods when temperatures were at or higher than 20 C; contaminant removal was diminished at lower temperatures.

B. Composting 1. Overview Composting is another ex situ process in which organic compounds are biodegraded, biotransformed, or otherwise stabilized by mesophilic and thermophilic bacteria. Addition of other readily biodegradable materials, bulking agents, moisture, and possibly nutrients to contaminated soils enhances or stimulates the composting

ch16.qxd 10/9/03 7:01 PM Page 212

212

the desk encyclopedia of microbiology

process (Ro et al., 1998) Nitroaromatics, petroleum hydrocarbons, PAHs, chlorinated phenols, and pesticides are classes of contaminants that are amendable to biodegradation using a composting system (Table 16.3). Composting at high temperatures typically accelerates the biodegradation or biotransformation process and has historically been required to kill pathogenic organisms remaining in wastewater treatment sludges; however, high-temperature composting of contaminated soils and other solids may not be required due to the absence of pathogens in these materials (Cookson, 1995). As a result, composting of contaminated soils may occur either under mesophilic (15–45 C) conditions or under thermophilic (50–70 C) conditions. The most suitable temperatures for composting range from 55 to 60 C, whereas temperatures higher than 70 C severely inhibit the ability of the microorganisms to metabolize or transform the contaminants (Ro et al., 1998). Bulking agents are used to increase the pore space or porosity of contaminated soil which results in greater air movement through the compost pile and higher rates of aerobic biodegradation. Typical bulking agents used in composting include wood chips, straw, tree bark, and plant matter (Alexander, 1994). Moisture content may also be a controlling parameter in the composting of contaminated soils. Excess moisture will impede the diffusion of oxygen through the compost pile and therefore restrict aerobic biodegradation. Optimal percentage moisture contents in compost piles range from 50 to 65% (Ro et al., 1998). Cookson (1995) reports that hydrocarbon biodegradation in a composting system is optimized at a moisture content of 60%, whereas slightly lower moisture contents are suitable for other hazardous constituents. The three most common types of composting systems are the windrow, static pile, and in-vessel systems. In windrow composting systems, the composting materials (mixture of organic material, contaminated soil, and bulking agents) are placed in long rows and are turned or mixed using a mechanical device to provide aeration or oxygen replenishment. Static pile systems are similarly arranged in long rows of composting material, but oxygen is supplied through a series of pipes that are placed at the base of the piles. Finally, invessel systems are typically enclosed units in which composting materials are transported on a conveyor system through a reaction vessel that is responsible for mixing, forced aeration, and temperature control (Cookson, 1995). In-vessel systems allow for greater engineering control (i.e., capture of volatile compounds and temperature and aeration regulation) and may greatly accelerate the biodegradation process when compared to the other two types of composting systems.

2. Demonstration In 1992, R. T. Williams, P. S. Ziegenfuss, and W. E. Sisk published work in the Journal of Industrial Microbiology on the static pile composting of explosives contaminated sediments from two U.S. Army facilities— Louisiana Army Ammunition Plant (LAAP) and Badger Army Ammunition Plant (BAAP). Target contaminants to be biodegraded using this remediation scheme included TNT, RDX, HMX, tetryl, and nitrocellulose (NC). Additives to the contaminated soils included alfalfa, straw, manure, wood chips, and horse feed in varying combinations along with nitrogen, phosphorus, potassium, and water. The initial concentration of contaminants in the LAAP soil included 56,800 mg/kg TNT, 17,900 mg/kg RDX, 2390 mg/kg HMX, and 650 mg/kg tetryl, whereas the initial concentration of NC in the BAAP soil was 18,800 mg/kg. Compost pile temperatures of the various configurations (two LAAP piles and four BAAP piles) were set at 35 and 55 C to investigate composting of explosives under both mesophilic and thermophilic conditions. The results indicated successful remediation of the contaminants using various combinations of bulking agents and temperatures. For the LAAP soil static piles, the combined concentration of explosives decreased from 17,870 to 74 mg/kg in the thermophilic pile, whereas mesophilic conditions effected a concentration decrease from 16,560 to 326 mg/kg during the 153-day test cycle. Half-lives for TNT, RDX, and HMX under thermophilic conditions were 12, 17, and 23 days, respectively. Half-lives of the contaminants under mesophilic conditions were nearly double those calculated under thermophilic conditions. Very low levels of amino transformation products (e.g., 2-amino-4,6-dinitrotoluene, 4-amino-2,6dinitrotoluene) were detected at the end of the test cycle. Nitrocellulose concentrations were dramatically reduced in the BAAP soil static piles. Under thermophilic conditions, NC concentration in two of the test piles was reduced from an average of approximately 13,090 mg/kg to approximately 20 mg/kg. R. Valo and M. Slkinoja-Salonen (1986) investigated windrow composting of chlorophenol-contaminated soils from a sawmill in Finland. Two 50-m3 compost piles were constructed and contained contaminated soil along with bark and ash. Nitrogen, phosphorus, and potassium were also added to the compost pile in the aqueous phase to ensure adequate nutrient levels throughout the system. The initial concentration of chlorophenols in the soil ranged from 400 to 500 mg/kg soil, whereas the initial concentration in the compost ranged from 200 to 300 mg/kg. The composting unit was monitored over the course of 17 months with the

ch16.qxd 10/9/03 7:01 PM Page 213

bioremediation majority of sampling and analysis being conducted during the first 5 months of operation (summer months, presumably when microbial activity was greatest). The results of the study indicated a significant decrease in the concentration of chlorophenols in the composting unit. Most of the contaminant loss or destruction occurred during the first few months of operation. The concentration of chlorophenol decreased rapidly to approximately 30 mg/kg after a few months of operation, and the concentration decreased to approximately 15 mg/kg by the end of the test period. Bacterial identification and counts also indicated that the compost material contained a greater number of pentachlorophenol-degrading microorganisms when compared to an agricultural clay soil that was not previously contaminated by chlorophenols. Subsequent jar tests on the compost material with radiolabeled pentachlorophenol confirmed that biodegradation was a major removal mechanism because approximately 30% of the radiolabeled carbon evolved as 14CO2 during a 40-day test period.

C. Bioslurry processes 1. Overview Slurry reaction systems are another type of ex situ bioremediation process in which biodegradation of contaminants is effected either in a highly controlled bioslurry reactor or in a waste lagoon. Typically, contaminated soils, sediments, or other solids are added to the system and mixed with a host of amendments, including water, nutrients, oxygen (if an aerobic environment is desired), surfactants to enhance contaminant mobilization, and/or microorganisms that may specifically biodegrade or biotransform the contaminant of interest. Bioslurry systems, especially enclosed bioreactors, offer a high degree of engineering control. Most of the major parameters that impact contaminant biodegradation (e.g., temperature, pH, and nutrient addition) may be monitored and adjusted using this remediation approach. Many contaminant compound classes are amenable to biodegradation using bioslurry processes and include phenols, chlorinated phenols, PAHs, pesticides, chlorinated hydrocarbons, and petroleum hydrocarbons (Cookson, 1995; Alexander, 1994). Highly viscous contaminants such as tars and certain oils may not be appropriate for bioslurry treatment. Treatment of the contaminated material may occur either in a bioslurry reactor or in a lagoon. The bioreactor system may consist of a single reactor or a series of reactors, and these reactors are typically closed to the atmosphere to prevent escape of volatile compounds and to maintain system control. Both the size and the

213

volume of the bioslurry reactors can be highly variable. The percentage solids concentration in these reactors ranges from 5 to 50% and depends on both the physical properties of the soil and the characteristics of the treatment system. Reactors in series offer a potential method of creating both aerobic and anaerobic environments in sequence, and this type of system may be very efficient in coupling the reductive dechlorination of highly chlorinated solvents (anaerobic conditions) with the subsequent metabolism of less chlorinated compounds (aerobic conditions). Typically, engineered waste lagoons vary in size and may be equipped with a variety of mixers and aerators to provide oxygen to the system. As in land treatment applications, a highly impermeable layer of clay soil or synthetic liner should be situated at the base of waste lagoons to prevent contaminant percolation into groundwater. The continuously mixed bioreactors or waste lagoons have significant engineering advantages compared to other ex situ technologies such as composting and land farming, although bioreactor systems may be costly to operate (Cookson, 1995). The mixing of the solids within the reactor enhances the distribution of both the solid materials containing the contaminants and the amendments that are added to the reactor system. Increased aeration and mass transfer of oxygen throughout the bioreactor as a result of mixing or sparging encourages aerobic biodegradation of the contaminants. If the bioslurry system contains volatile compounds, the rate of volatilization may be increased due to agitation and mixing. Both the accessibility of the contaminant to the microorganisms (contact) and the fraction of bioavailable contaminant are increased in this type of reactor system. Bioslurry processes promote the breakup of aggregated soil particles that may sequester or contain high concentrations of the target contaminant. This breakup of larger particles into smaller ones leads to a greater soil surface area to volume ratio and thus may increase contaminant desorption by maximizing surface contact with the aqueous phase. Contaminated clay soils may be especially amenable to bioslurry treatment processes due to their decreased permeability and difficulty in treating in situ. Organic contaminants adsorb strongly to adhesive clay materials and may be easily entrapped in the intra- and interparticle pore spaces, and subsequent transport of materials (i.e., nutrients, oxygen or other electron acceptor, and microorganisms) through clay soils is very difficult. 2. Demonstration In 1991, G. C. Compeau and coworkers reported results from laboratory-scale experiments used in the design

ch16.qxd 10/9/03 7:01 PM Page 214

214

the desk encyclopedia of microbiology

and implementation of a full-scale bioslurry reactor treatment system. The full-scale system was designed to treat 3400 cubic yards of PCP-contaminated soil from a spill. PCP concentrations in the soil were variable, with a maximum of 9000 mg/kg. Initial testing of the contaminated soil indicated that the indigenous microflora were not capable of breaking down the PCP contaminant; therefore, subsequent bench and full-scale treatment systems incorporated specialized PCP-degrading microorganisms to augment the existing soil organisms and stimulate biodegradation of the contaminant. The laboratory-scale bioslurry treatability studies indicated that for a variety of solids with concentrations ranging from 5 to 40%, PCP could be successfully biodegraded when the reactor was inoculated with the PCP-degrading culture. For example, in the 40% solids concentration bioslurry test with an initial concentration of approximately 275 mg/liter, very little of the PCP was removed from the system during the initial 13-day test period. After inoculation, nearly all the PCP in the reactor was biodegraded during a 10-day period. The laboratory studies also revealed that the majority of the PCP contaminant resided within the more coarse particles, which may not be easily suspended in a bioslurry reactor system. As a result, design of the fullscale treatment system was amended to include a washing step to desorb the PCP from the coarse particles. The wash solution containing PCP along with the finer unwashed soil particles represented the major influents to the bioslurry reactors. A pair of 25,000-gallon bioslurry reactors were used in the full-scale cleanup operation. Nitrogen and phosphorus were added to the reactors to meet microbial growth requirements. Results from the full-scale experiment supported the laboratory findings that inoculation was necessary to stimulate PCP biodegradation. A testing period of 2 weeks was required to decrease PCP soil concentrations from 370 to 0.5 mg/kg in one of the inoculated reactors. This degree of treatment was also reached in the second reactor, but only after addition of the inoculum following a 7-day test period in which biodegradation did not occur. It is generally believed that bioaugmentation decreases the lag time in biodegradation studies but that, given time, selection processes will result in microbial populations capable of degrading target contaminants.

VIII. SUMMARY Bioremediation refers to the transformation of organic wastes by microorganisms into biomass, carbon dioxide, water, and inorganic salts, depending on the structure of the compounds in the waste. These organic waste

materials may also be biotransformed to less toxic compounds or compounds that may be more amenable to complete mineralization. Many in situ and ex situ technologies are currently available to address organic waste contamination in both soils and groundwater. Natural bioattenuation is an in situ method for remediating contaminated subsurface soils and groundwater. It is a nonintrusive method that takes advantage of the abilities of natural microflora in subsurface environments to biodegrade organic contaminants. Although natural bioattenuation does not require complex engineered systems, monitoring for contaminant concentration and biochemical markers is necessary to validate the efficiency of this type of treatment option. Enhanced in situ bioremediation technologies include the Raymond process (biostimulation), electron donor delivery, bioventing, and permeable reactive barriers. Biostimulation technology involves the addition of nutrients (namely, N and P), oxygen, and perhaps microorganisms to groundwater aquifers to enhance biodegradation of organic contaminants. Oxygen may be added to the groundwater by injection of air, pure oxygen, hydrogen peroxide, or oxygen releasing compounds. Injection of microorganisms into the subsurface, or bioaugmentation, also may be an option for biodegradation enhancement; however, to date there has been little success in field-scale experiments. In situ biodegradation and biotransformation of chlorinated organic compounds may be accelerated by the injection of electron donors, such as lactate, methanol, and hydrogen, to the contaminant region of interest in groundwater. In a cometabolic process, breakdown of the primary substrate (e.g., methane) leads to the fortuitous expression of enzymes that are capable of oxidizing highly chlorinated organics such as TCE. Biotransformation through reductive dechlorination also requires an adequate supply of electron donors to serve as a primary substrate for the microorganisms that mediate this process. Bioventing of contaminated subsurface unsaturated zone soils focuses on delivering oxygen to areas in which oxygen has been previously depleted due to microbiological reactions in the soil. This treatment technology is typically applied to soils that have high permeabilities and thus a greater ability to transfer oxygen to the contaminant region. The treatment technology is designed to favor contaminant biodegradation over volatilization, but site hydrogeology and contaminant physical properties may also have an impact on the effectiveness of the two mechanisms. Permeable reactive barriers provide another in situ method in which groundwater is routed through a wall or barrier containing materials that will enhance either the biodegradation or biotransformation of

ch16.qxd 10/9/03 7:01 PM Page 215

215

bioremediation organic contaminants. Materials contained in the reaction zone may include both nutrients and oxygen releasing compounds to enhance aerobic biodegradation of contaminants or other reactive agents such as zero-valent iron that lead to the abiotic reductive dechlorination of chlorinated solvents. Land treatment or land farming is an ex situ method to treat contaminated soils and sludges. Excavated materials are placed either in an engineered treatment unit or on top of the natural soil surface. Biodegradation in these systems is enhanced through tilling and mixing to promote aeration, nutrient amendment, and possibly the addition of contaminant-degrading microorganisms. Ex situ composting of contaminated soils is achieved by addition of bulking agents such as wood chips to promote aeration, nutrients, and readily biodegradable materials to the contaminated soils. These mixtures are then placed in windrow, static pile, or in-vessel composting systems in which higher temperature, thermophilic conditions are initiated to accelerate the biodegradation process. Finally, bioslurry processes promote treatment of contaminated soils, solids, and sludges by placement in bioslurry reactors and mixing with water, nutrients, oxygen, microorganisms, or other materials that might enhance the biodegradation or biotransformation of the contaminant. Bioslurry processes offer a high degree of control over the parameters that influence biodegradation, including temperature, pH, and nutrient concentration, but they can be expensive when compared to land treatment and composting. Bioremediation processes for the treatment of contaminated environmental media such as soils and the saturated (aquifer) and unsaturated (vadose) zones of the subsurface may prove to be the most cost-effective treatment options depending on site-specific remedial objectives. In common with most other remediation technologies, bioremediation processes are usually applied as part of a system or treatment in conjunction with other complementing processes to obtain optimal results.

Remediation Technology Design & Application: Bioremediation” (W. C. Anderson, Ed.). American Academy of Environmental Engineers, Annapolis, MD. Flathman, P. E., Jerger, D. E., and Exner, J. H. (Eds.) (1994). “Bioremediation: Field Experience.” Lewis, Boca Raton, FL. Leeson, A., and Hinchee, R. E. (1997). “Soil Bioventing: Principles and Practice.” Lewis, Boca Raton, FL. Loehr, R. C., and Malina, J. F. (Eds.) (1986). “Land Treatment—A Hazardous Waste Management Alternative.” Center for Research in Water Resources, University of Texas at Austin/Van Nostrand Reinhold, Austin/New York. Madigan, M. T., Martinko, J. M., and Parker, J. (1997). “Biology of Microorganisms,” 8th ed. Prentice-Hall, Upper Saddle River, NJ. Norris, R. D., Hinchee, R. E., Brown, R., McCarty, P. L., Semprini, L., Wilson, J. T., Kampbell, D. H., Reinhard, M., Bouwer, E. J., Borden, R. C., Vogel, T. M., Thomas, J. M., and Ward, C. H. (1994). “Handbook of Bioremediation.” Lewis, Boca Raton, FL. Pieper, D. H., and Reineke, W. (2000). Engineering bacteria for bioremediation. Curr. Opin. Biotechnol. 11, 262–70. Rao, P. S., Brown, R. A., Allen-King, R. M., Cooper, W. J., Gardner, W. R., Gollin, M. A., Hellman, T. M., Heminway, D. F., Luthy, R. G., Olsen, R. L., Palmer, P. A., Pohland, F. G., Rappaport, A. B., Sara, M. N., Syrrist, D. M., and Wagner, B. J. (1997). “Innovations in Ground Water and Soil Cleanup: From Concept to Commercialization.” National Academy Press, Washington, DC. Rittman, B. E., Alvarez-Cohen, L., Bedient, P. B., Brown, R. A., Chappelle, F. H., Kitanidis, P. K., Mahaffey, W. R., Norris, R. D., Salanitro, J. P., Shauver, J. M., Tiedje, J. M., Wilson, J. T., and Wolfe, R. S. (1993). “In Situ Bioremediation: When Does It Work?” National Academy Press, Washington, DC. Ro, K. S., Preston, K. T., Seiden, S., and Bergs, M. A. (1998). Remediation composting process principles: Focus on soils contaminated with explosive compounds. Crit. Rev. Environ. Sci. Technol. 28(3), 253–282. Spain, J. C. (Ed.) (1995). “Biodegradation of Nitroaromatic Compounds.” Plenum, New York. U.S. Environmental Protection Agency (USEPA) (1997). Cleaning up the nation’s waste sites: Markets and technology trends, EPA 542-R-96-005. USEPA, Washington, DC. Ward, C. H., Loehr, R. L., Norris, R., Nyer, E., Piotrowski, M., Spain, J., and Wilson, J. (1995). “Innovative Site Remediation Technology: Bioremediation” (W. C. Anderson, Ed.). American Academy of Environmental Engineers, Annapolis, MD. Ward, C. H., Cherry, J. A., and Scalf, M. R. (Eds.) (1997). “Subsurface Restoration.” Ann Arbor Press, Chelsea, MI. Young, L. Y., and Cerniglia, C. E. (Eds.) (1994). “Microbial Transformation and Degradation of Toxic Organic Chemicals.” Wiley–Liss, New York.

BIBLIOGRAPHY

WEBSITES

Alexander, M. (1994). “Biodegradation and Bioremediation.” Academic Press, San Diego. Alleman, C. B., Leeson, A. (Eds.) (1999). “Bioreactor and Ex Situ Biological Treatment Technologies: The Fifth International In Situ and On-Site Bioremediation Symposium: San Diego, California.” Battelle. Chaudry, G. R. (Ed.) (1994). “Biological Degradation and Bioremediation of Toxic Chemicals.” Dioscorides Press, Portland, OR. Cookson, J. T. (1995). “Bioremediation Engineering: Design and Application.” McGraw-Hill, New York. DuPont, R. R., Bruell, C. J., Downey, D. C., Huling, S. G., Marley, M. C., Norris, R. D., and Pivetz, B. (1998). “Innovative Site

The University of Minnesota Biocatalysis/Biodegradation Database http://umbbd.ahc.umn.edu/ The Bangor (Wales) Biodegradation Group http://biology.bangor.ac.uk/research/biodegradation/ The Electronic Journal of Biotechnology http://www.ejb.org/ List of biotechnology organizations and research institutes http://www.ejb.org/feedback/borganizations.html The Biodegradative Strain Database http://bsd.cme.msu.edu/bsd/index.html Bioremediation Discussion Group http://bioremediationgroup.org

ch17.qxd 10/9/03 7:02 PM Page 216

17 Biosensors Yoko Nomura and Isao Karube Tokyo University of Technology

GLOSSARY

measurements, such as in medical diagnosis, food quality control, and environmental monitoring. glucose oxidase An enzyme which catalyzes glucose oxidation. It is usually suitable for industrial use because of its high stability. It is one of the typical biological sensing elements and was used in the first biosensor. transducer An electronic signal-transducing element that can convert a change in the concentration of a product of a biological reaction into an electronic signal. Examples are electrodes and optical apparatus.

biochemical oxygen demand (BOD) The amount of dissolved oxygen (DO) needed to biologically degrade the organic compounds in an aquatic environment. BOD measurements are conventionally carried out according to the BOD 5-day method which measures the DO by titration (modified Winkler method) before and after a 5-day sample incubation period during which biodegradation occurs. biological sensing element A biomolecule or a biomaterial used in a biosensor for analyte recognition; sometimes referred to as a biological recognition element. It undergoes a specific biological reaction with the analyte so that the analyte can be selectively detected by the biosensor. Examples include enzymes, antibodies, DNA oligomers, and microorganisms. flow injection analysis (FIA) A technique sometimes used in flow-type biosensors; developed in the 1970s. The sample is injected directly into carrier solution running through fine tubes of manifolds which include an electrical detection apparatus. Reactions such as those resulting in fluorescence or chemiluminescence occur in situ and are detected as electronic signals. FIA systems afford precise control of the mixing ratio of injected samples and carrier solutions, which results in highly reproducible measurements. Because the detection by FIA is simple, rapid, precise, and continuous, it is used for various The Desk Encyclopedia of Microbiology ISBN: 0-12-621361-5

A Biosensor is broadly defined as a sensing system which uses biological reactions such as enzymatic or immunological reactions. Most biosensors are composed of a biological sensing element and a transducer. Many biosensors (Cass, 1990; Suzuki, 1990; Buerk, 1993) have been constructed since the first report describing the development of the first enzyme sensor for glucose measurement by Clark in 1962 (Buerk, 1993). This biosensor measured the product of an enzyme reaction using an electrode, which was a remarkable achievement even though the enzyme was not immobilized on the electrode. Today’s basic biosensors originated from the investigation by Updike and Hicks in 1967 (Buerk, 1993). Their sensor combined membrane-immobilized glucose oxidase with an oxygen electrode, and oxygen measurements were carried out before and after the enzyme reaction. Many other biological elements and

216

Copyright © 2003 Elsevier Ltd All rights of reproduction in any form reserved

ch17.qxd 10/9/03 7:02 PM Page 217

217

biosensors transducers have been examined since this first glucose biosensor was developed. For example, in 1977 Karube reported the first microbial sensor which used the whole cell as a biological sensing element. Many immuno-sensors using antibodies were fabricated in 1980s and recently DNA, RNA, and even artificial recognition elements have been employed. Transducers have also been improved, and novel transducing elements such as optical detectors, including surface plasmon resonance (SPR) detectors, are widely used in modern biosensors.

I. PRINCIPLE OF BIOSENSORS A biosensor is a detection system composed of biological sensing elements, such as enzymes, antibodies, microorganisms, and DNA, and an electronic signaltransducing element (Fig. 17. 1) (Buerk, 1993). A transducer can convert a change in the concentration of a product of a biological reaction into an electronic signal. For example, an oxygen electrode converts a change in the oxygen concentration of a sample caused by enzymes or biodegradation into a change in electric current. The transducers used in biosensors include electrodes, piezo-electric quartz crystals, and optics. Many biosensors can be applied to important fields such as diagnosis and environmental monitoring. The principle of a simple glucose sensor using glucose oxidase (GOD) is shown in Fig. 17.2. The target analyte, glucose, is a substrate of GOD. Glucose diffuses into the membrane and a biological reaction occurs within the membrane. When GOD catalyzes the glucose oxidation, dissolved oxygen in the sample is consumed and gluconic lactone and hydrogen peroxide are produced. The oxygen electrode detects a change in dissolved oxygen concentration of the

sample as a change in electric current. Since the change in dissolved oxygen concentration is a result of the glucose oxidation, the glucose concentration of the sample can be measured. The biosensor does not directly detect the target analyte (glucose). Instead, it measures the change in the concentration of a co-reactant (oxygen) or a co-product (hydrogen peroxide) of the reaction catalyzed by the immobilized biologicalsensing material (i.e., GOD). Biosensor configurations are categorized into two types; batch type (a) and flow type (b) as depicted in Fig. 17.3 (Buerk, 1993). In flow-type sensors, a bioreactor, a column stuffed with biomaterial-immobilized beads, can be incorporated separately from a transducer. Flow-type biosensors are very useful for continuous monitoring of target analytes. Immobilization of biomaterials is usually required for biosensor fabrication. Typical immobilization carriers are glass, alginate and artificial resin beads, and membranes. Disposable biosensors such as amperometric glucose sensors for medical diagnosis are examples of a batch-type sensor. Flow injection analysis (FIA) is a technique sometimes used in flow-type biosensors. Because biosensors using FIA yield very rapid and accurate measurements, these have been applied to various fields, such as environmental monitoring and food quality control. In a typical biosensor study, parameters such as the amount of immobilized biological-sensing elements (e.g., enzymes), pH, and temperature of the buffer are optimized, and then a calibration curve is generated using standard solutions containing known concentrations of the analyte. Figure 17.4 shows a typical calibration curve of a biosensor. Calibration curves are normally obtained by one of two methods: from reaction curves of the biological co-reaction measurement (Fig. 17.4a) or the biological co-product measurement

)

Electronic signal

FIGURE 17.1 Biological elements are combined with transducers when biosensors are fabricated. Electrodes and optical devices are widely used as transducers in biosensors. A piezoelectric device such as a quartz crystal detects a weight change before and after the biological reaction as applied to immuno- and DNA sensors. Transducers convert a change in the concentration of the compound into an electronic signal.

ch17.qxd 10/9/03 7:02 PM Page 218

218

the desk encyclopedia of microbiology

FIGURE 17.2 Principle of a glucose sensor. The analyte is glucose, and the biological-sensing element of the biosensors is glucose oxidase (GOD). The membrane-immobilized GOD is combined with an oxygen electrode as a transducer. An oxygen electrode generates electric current depending on the dissolved oxygen concentration. Glucose and oxygen permeate into the membrane-immobilized GOD. (a) Dissolved oxygen is not consumed when a sample which does not contain glucose such as pure buffer solution is measured by the glucose sensor. The sample is air-saturated and the oxygen electrode produces stable high current. This value is defined as the baseline current of the sensor. (b) A sample containing glucose is measured by the glucose sensor. GOD catalyzes the glucose oxidation using oxygen and dissolved oxygen is consumed. The oxygen concentration is lower than that in (a) and the electric current decreases from the baseline current. (c) Typical reaction curve of the sensor is illustrated. A, the baseline current: B1–B3, the electric currents obtained from samples containing glucose. The differences between A and B1–B3 are the sensor responses (C1–C3). As glucose concentration increases, the sensor response will also increase.

(Fig. 17.4b). In both cases the sensor responses are calculated from the difference between the values of pure buffer and the sample containing the analyte. The sensitivity of a biosensor is defined as the slope of the linear range of the calibration curve. The linear range is between the lower detection limit (C1) and the nonlinear profile at higher concentration (C6, 7,

FIGURE 17.3 Two types of biosensor measurements. The illustrated biosensor is a combination of an electrode and a membraneimmobilized biomaterial. For example, adsorption or entrapment methods may be used to immobilize biomaterials. The membrane with immobilized biomaterial such as cellulose acetate membrane (0.45 m) is attached to the electrode by an O-ring. (a) Batch-type measurement. The biosensor is immersed in the buffer solution and the analyte solution is directly injected into the buffer solution. (b) Flow-type measurement. The biosensor is attached to a flow cell and a flow-type biosensor is fabricated. The buffer solution is propelled by a peristaltic pump and the analyte solution is injected into the flow line. The buffer solution mixed with the analyte is introduced to the flow cell and is measured by the biosensor.

and 8). The biosensor response (biosensor output signal Rs) obtained from the sample measurement is substituted into the calibration curve in Fig. 17.4 and the analyte concentration of the sample can be calculated (Cx). Reproducibility and the life-time of a biosensor are usually examined, and selectivity must also be evaluated in some cases. Biosensors have replaced conventional methods, which are often complicated, time-consuming, expensive, and require pretreatment or clean-up of real samples prior to analysis. Biosensors generally have the following advantages compared to other analytical methods: 1. Rapid and convenient detection. 2. Direct measurement of real samples. 3. Very specific detection. On the other hand, stability and reproducibility have been problematic for biosensors due to the inherent instability of biomaterials used as sensing

ch17.qxd 10/9/03 7:02 PM Page 219

219

biosensors

II. APPLICATIONS OF BIOSENSORS Biosensors have been applied in many important fields, such as food quality control, environmental monitoring, and medical diagnosis. Recently, some groups reported biosensors for detecting chemical or toxic agents for use in military settings (Buerk, 1993). A few examples of biosensors in practical use in major fields are described in the following sections.

A. Food analysis

FIGURE 17.4 Typical calibration curve of a biosensor. The sample injections were carried out eight times (Sn). (a) A reaction curve of a biosensor based on the measurement of co-reactants. Co-reactant is consumed during the measurement and the sensor output such as the electric current in the figure decreases as the analyte concentration increases. The baseline current is higher than that of the sample containing the analyte. The sensor response (Rn) is calculated by subtracting the measurement value (e.g. the electric current) of the sample containing the analyte from the baseline value. (b) A reaction curve of a biosensor based on the measurement of co-products. Coproduct or sensor output increases as the analyte concentration increases. The baseline value is less than that of the sample containing the analyte. The sensor response (Rn) is obtained by subtracting the baseline value from the measurement value of the sample containing the analyte. (c) A calibration curve. The dynamic range of a biosensor is the linear correlation from the detection limit C1–C5. The sensitivity of the biosensor, for example, is calculated as (R4 R2)/(C4 C2). Rs is obtained when an unknown concentration sample is measured by the biosensor and it is substituted into the linear correlation, and the sample concentration Cs is obtained.

elements. Although biosensors have disadvantages, numerous investigations on biosensors to date have helped to overcome at least some of these difficulties, allowing practical application of many biosensors in the real world.

Many biosensors such as enzyme sensors have been developed for food analysis and food quality control. Enzyme sensors are used for the measurement of sugars, such as glucose, sucrose, and fructose. Vitamin C (ascorbic acid) and glutamate in food are also measured by biosensors. Some of these sensors are also being used in the medical field and for monitoring waste-water. Many of these sensors operate on the same principle as described previously for the glucose sensor shown in Fig.17. 2. The freshness of fish meat can also be measured by enzyme sensors. The freshness sensor that detects the degradation products of adenosine triphosphate (ATP) in fish meat, as well as many other enzyme sensors, achieves very high sensitivity by combining an FIA system coupled with chemiluminescence detection. Microbial sensors are also applied to food quality analysis for measuring free fatty acids in milk (Schmidt et al., 1996), alcohol, or acetic acid (Suzuki, 1990). The free fatty acid sensor uses Arthrobacter nicotianase, and both the alcohol and the acetic acid sensors use same bacterium, Tricosposporon blassicae.

B. Medical diagnosis Enzyme sensors and microbial sensors for antibiotics and vitamins have been fabricated since the 1970s. Diabetes has been a particularly important target for many biosensors intended for medical use. Glucose sensors have been applied to diagnosis and monitoring of diabetes patients and to food quality analysis. Insulin biosensors have also been developed for use in diabetes treatment. Since the first glucose sensor using an oxygen electrode was reported, numerous glucose sensors have been developed using techniques aimed at practical use. For example, screen-printing (Nagata et al., 1995) and micromachining techniques (Hiratsuka et al., 1998) have been applied to glucose sensors. Disposable biosensors have very promising applications in medical diagnosis, such as monitoring blood glucose concentrations in diabetes patients.

ch17.qxd 10/9/03 7:02 PM Page 220

220

the desk encyclopedia of microbiology

Matsushita Company (Osaka, Japan) has commercialized a circular-type glucose sensor which utilizes semiconductor technology.

C. Environmental monitoring Biosensors can rapidly detect and measure eutoriphicants, toxicants, and biochemical oxygen demand (BOD) in the environment. Enzyme immuno and microbial sensors have been mainly developed to detect pollutants in the environment. Biosensors for environmental monitoring have been reviewed (Karube et al., 1995). The phosphate sensor is a typical example of an enzyme sensor for environmental monitoring. In the past few years, dramatic improvements have been made in phosphate detection systems using enzyme sensors (Nakamura et. al., 1997), and it is expected that some of these sensors will be used in the field in the near future. Many biosensors have been developed to detect toxicants in the environment such as pesticides and cyanide. Jeanty and Marty (1998) reported organophosphates detection systems based on acetylcholine esterase inhibition. Immuno-sensors, in place of conventional enzyme-linked immuno sorbent assays are also frequently used to detect pesticides. Carl et al. (1997) fabricated an immuno-sensor which detects chemical endocrine disrupters (PCB) using a screenprinting technique, and recently Seifert and Hock (1998) investigated a novel sensor which uses a human estrogen receptor and an SPR-detection system. Microbial sensors have been constructed which detect cyanide and detergents. Karube et al. (1995) reviewed developments in microbial sensors for environmental monitoring. The BOD sensor, which uses an omnivorous yeast Trichosporon cutaneum, is a well-known example of a microbial biosensor applied to environmental

monitoring (Cass, 1990). The microbial BOD sensor measures the oxygen uptake by the respiratory system of the microorganism which changes as a result of the biodegradation of organic compounds in the sample. (Fig. 17.5). The microbial BOD sensor systems have been commercialized and marketed since 1983 by several companies (DKK, Nisshin Denki, and Central Kagaku Co., Tokyo).

III. CURRENT TOPICS IN BIOSENSORS Enzyme-, immuno-, and microbial sensors have become very popular. Sensors that take advantage of new transducing techniques such as SPR and a charge-coupled device have increasingly been reported (Buerk, 1993). Novel molecular recognition elements, such as DNA oligomers having specific affinity to various target molecules, have also been tested for use in biosensors. Stability of a biosensor is very important, especially when the sensors must be used continuously in the field. Microorganisms and few stable enzymes are suitable for this purpose. However, many enzymes and antibodies used in biosensors are not stable enough for long-term practical use. Molecularly imprinted polymers (MIPs) have been employed in place of antibodies to construct biosensors with enhanced stability (Kriz et al., 1995). These polymers are prepared by polymerization of various monomers in the presence of the target compound which acts as a template. After the polymer is removed from the template compound, the polymer retains the memory of the template and can selectively rebind the template compound. The use of MIPs to replace biomolecules in biosensors has advantages such as enhanced stability and inexpensive cost, which may expand the scope and applicability of the biosensors of tomorrow.

ACKNOWLEDGMENT We thank Yohei Yokobayashi at The Scripps Research Institute for his help and advice in preparing the manuscript. FIGURE 17.5 A schematic of a basic microbial BOD sensor. A microbial electrode consists of a membrane-immobilized T. cutaneum and an oxygen electrode. BOD measurement is performed aerobically using phosphate buffer at 30 C. Glucose–glutamic acid solution is used as the standard solution for preparation of a calibration curve. After the standard solution measurement is obtained, real samples are examined and the BOD value is estimated. This system has been commercialized and the measurements are carried out automatically and continuously.

BIBLIOGRAPHY Buerk, D. G. (1993). “Biosensors.” Technomic, Lancaster, Pennsylvania. Carl, M. D., Iionti, I., Taccini, M., Cagnini, A., and Mascini, M. (1997). Disposable screen-printed electrode for the immunochemical detection of polychlorinated biphenyls. Anal. Chim. Acta 324, 189–197.

ch17.qxd 10/9/03 7:02 PM Page 221

221

biosensors Cass, A. E. G. (1990). “Biosensor.” IRL/Oxford University Press, New York. Cosnier, S. (2000). Biosensors based on immobilization of biomolecules by electrogenerated polymer films. New perspectives. Appl. Biochem. Biotechnol. 89, 127–138. Hiratsuka, A., Sasaki, S., and Karube, I. (1998). A self-contained glucose sensor chip with arrayed capillaries. Electro-analysis 10, 231–235. Jeanty, G., and Marty, J. L. (1998). Detection of paraoxon by continuous flow system based enzyme sensor. Biosensor Bioelectronics 13, 213–218. Karube, I., Nomura, Y., and Arikawa, Y. (1995). Biosensors for environmental control. Trends Anal. Chem. 14, 295–299. Kriz, D., Ramstrom, O., Svensson, A., and Mosbach, K. (1995). Introducing biomimetic sensors based on molecularly imprinted polymers as recognition elements. Anal. Chem. 67, 2142–2144. Nagata, R., Yokoyama, K., Clark, S. A., and Karube, I. (1995). A glucose sensor fabricated by the screen printing technique. Biosensor Bioelectronics 10, 261–267. Nakamura, H., Ikebukuro, I., McNiven, S., Karube, I., Yamamoto, H., Hayashi, K., Suzuki, M., and Kubo, I. (1997). A chemiluminescent FIA biosensor for phosphate ion monitoring using pyruvate oxidase. Biosensor Bioelectronics 12, 959–966. Schmidt, A., Gabisch, C. S., and Bilitewski, U. (1996). Microbial biosensor for free fatty acids using an oxygen electrode based on thick film technology. Biosensor Bioelectronics 11, 1139–1145.

Seifert, M., and Hock, B. (1998). Analytics of estrogens and xenoestrogens in the environmental using a SPR-Biosensor, Proc. Biosensor 98, 47. Stefan, R. I., van Staden, J. F., Aboul-Enein, H. Y. (2000). Immunosensors in clinical analysis. Fresenius J. Anal. Chem. 366, 659–668. Stephens, D. (2001). New genetically encoded ‘biosensors’. Trends Cell Biol. 11, 241. Suzuki, S. (1990). “Biosensor,” 5th ed. Kohdan-sha, Tokyo. (In Japanese) Van Regenmortel, M. H. (2000). Analysing structure–function relationships with biosensors. Cell Mol. Life Sci. 58, 794–800.

WEBSITES Website by K. Bruce Jacobson of the Oak Ridge Ridge National Laboratory http://www.ornl.gov/ORNLReview/rev29_3/text/biosens.htm The Electronic Journal of Biotechnology http://www.ejb.org/ List of biotechnology organizations and research institutes http://www.ejb.org/feedback/borganizations.html

ch18.qxd 10/9/03 7:03 PM Page 222

18 Cell membrane: structure and function Robert J. Kadner University of Virginia

and other eukaryotic cells possess, in addition to the plasma membrane, numerous intracellular membranes which form the organelles that perform specialized metabolic functions. Bacterial and archaeal cells typically lack intracellular membrane organelles and contain only the single cytoplasmic membrane, perhaps surrounded by an outer membrane. The cytoplasmic membrane of bacteria is typically composed of simple phospholipids that form a membrane bilayer, into which are inserted a large number of different proteins. The phospholipid bilayer forms the osmotic barrier that prevents movement of most materials into or out of the cell. The various membrane proteins carry out numerous important functions, including the generation and storage of metabolic energy and the regulation of uptake and release of all nutrients and metabolic products. Membrane proteins recognize and transmit many signals that reflect changes in environmental conditions and trigger an appropriate cellular response. They also play key roles in the control of cell growth and division, bacterial movement, and the export of surface proteins and carbohydrates.

GLOSSARY ABC proteins Proteins that contain the widely conserved ATP-binding cassette, a motif that couples energy from ATP binding and hydrolysis to various transport processes. detergent A molecule with polar and nonpolar portions that can disrupt membranes by stabilizing the dispersion of hydrophobic lipids and proteins in water. hydrophobic Molecules or portions of molecules that cannot form hydrogen bonds or other polar interactions with water. hydrophobic effect The tendency of hydrophobic regions of molecules to avoid contact with water. osmolarity The tendency of water to flow across a membrane in the direction of the more concentrated solution. proton motive force The electrochemical measure of the transmembrane gradient of protons, consisting of an electrical potential due to separation of charge and the pH gradient due to different concentration of protons. symporter A transport system in which movement of the coupling ion moves in the same direction as the substrate molecule, in contrast to an antiporter or uniporter.

A. Ultrastructure of cell membranes

Every Cell Possesses a Surface Membrane that separates it from the environment or from other cells. Animal cells

Cell membranes are readily visible when thin sections of cells are stained with heavy metals and viewed in

The Desk Encyclopedia of Microbiology ISBN: 0-12-621361-5

I. ULTRASTRUCTURE AND THE ROLE OF CYTOPLASMIC MEMBRANE

222

Copyright © 2003 Elsevier Ltd All rights of reproduction in any form reserved

ch18.qxd 10/9/03 7:03 PM Page 223

cell membrane: structure and function

223

the electron microscope. They appear as a characteristic triple-layered structure with two dark electrondense layers, representing the region of the lipid head groups, surrounding a light layer which reflects the hydrophobic central portion of the membrane bilayer. All cellular membranes appear quite similar by this technique, regardless of the source of the membrane or their protein content (Fig. 18.1a). Most biological membranes are 4 or 5 nm in width. In the technique of freeze-fracture electron microscopy a knife blow is used to split a frozen sample of cells. The fracture plane often extends along or through the weakly connected central section of a membrane bilayer. This technique can reveal the presence and density of proteins embedded in and spanning through the membrane. Figure 18.1 supports the fluid mosaic model of membrane structure in which the polar membrane lipids form a lamellar, or leaf-like, bilayer in which their nonpolar portions face each other in the central region and their polar regions are on the outside. Integral membrane proteins span across the bilayer, but can diffuse within the plane of the bilayer and even associate into large complexes. Many peripheral membrane proteins do not span across the membrane and can be transiently bound through hydrophobic anchors or by association with other membrane proteins or the lipid head groups.

B. Role as osmotic barrier The cytoplasmic membrane is the osmotic barrier of the cell, owing to its ability to restrict the passage of salts and polar organic compounds. If a cell is placed in a medium in which the osmolarity is higher or lower than the osmolarity of the cytoplasm, water will flow across the cytoplasmic membrane out or into the cell, respectively. This osmotic flow of water occurs in response to the natural forces that seek to eliminate gradients or differences in the concentration of water on the two sides of the membrane. Hence, the cytoplasm either shrinks or swells under these two conditions as a result of the loss or gain of water. In most bacteria, the cell does not change size owing to the presence of its rigid cell wall.

C. Role as cell boundary The cytoplasmic membrane is the boundary between the cell and its surroundings and thus must regulate the passage of nutrients and metabolic products. The presence of the hydrophobic layer formed by the membrane lipids greatly restricts the passage of any polar molecules and of macromolecules. It prevents

FIGURE 18.1 Structure of bacterial membranes. (a) Electron micrograph of a thin section of the fish pathogen, Aeromonas salmonicida. Cells were embedded in plastic, cut to a thin slice, stained with heavy metals (uranyl acetate and lead citrate), and visualized by transmission electron microscopy. The cytoplasmic membrane is seen as the triple-layered structure bounding the cytoplasm. Outside the 7.5-nm thick cytoplasmic membrane is the periplasmic space, the triple-layered outer membrane, and the thick surface S layer. (b) Electron micrograph of a freeze-fractured sample. A suspension of cells of Bacillus licheniformis was frozen, and the block was shattered by a sharp blow with a knife edge. The fracture plane ran occasionally though membrane bilayers. A carbon replica of the two faces thus revealed was made, the organic material was etched away with acid, and surface structures were enhanced by shadowing with a beam of platinum atoms. The image is viewed in transmission electron microscope. The two faces show the two halves of the cytoplasmic membrane layer, indicating the presence of particles that are embedded in and span the membranes (courtesy of Terrance Beveridge, University of Guelph, Canada).

ch18.qxd 10/9/03 7:03 PM Page 224

224

the desk encyclopedia of microbiology

the loss of cellular macromolecules and metabolic intermediates.

D. Regulation of transport Transport systems allow the passage of specific molecules, such as ion, nutrients, and metabolic products, either into or out of the cell. Bacteria in general have a large number of very specific transport systems that carry out active transport. They consume some form of metabolic energy to be able to pump their substrate from a low concentration on one side of the membrane to a much higher concentration on the other side. Transport is an integral part of the universal process of bioenergetics, which refers to the formation and consumption of sources of cellular energy, most of which involve transmembrane ion gradients.

E. Role in cell growth and division Expansion of the membrane surface is intimately related to growth rate of any cell, and all components must be inserted in a timely manner to allow cells to expand in size. Cell division requires a carefully controlled process whereby the membranes of the parental cell pinch together, fuse, and separate to create two progeny cells, without loss of internal material. The membrane contains export systems of control the release of structural components to the cell surface and other secreted factors beyond the cell. Other membranelocalized protein complexes regulate the process of initiation of DNA replication, separation of chromosomes into the dividing bacterial cells, and in-growth of the cell surface that occurs during cell division.

groups of glycerol, usually with a saturated fatty acid at the 1-position and an unsaturated fatty acid at the 2-position. To the third hydroxyl group of glycerol is attached a phosphate moiety and to it the head group. In bacteria, the range of head groups is narrow, and the phospholipids in Escherichia coli are approximately 75% phosphatidyl ethanolamine (PE) and 20% phosphatidyl glycerol (PG), and the remainder is cardiolipin (diphosphatidyl glycerol), phosphatidyl serine, and trace amounts of other phospholipids (Fig. 18.2). Other bacteria possess more complex types of membrane lipids, although these lipids are usually much less complex than those in the plasma membrane of animal cells. Some bacteria possess phosphatidyl choline, or lecithin, which is characteristic of higher organisms. Other bacteria produce glycolipids, such as monogalactosyl diglyceride. The membrane lipids from archaea are quite different from those in bacteria and eukarya. Their hydrocarbon chains are based on isoprenoid units and these are linked to the glycerol backbone in an ether, rather than ester, linkage. In some archaea, a glycerol backbone and head group are attached to both ends of a pair of isoprenoid units. Sterols, such as cholesterol in mammalian cells or ergosterol in fungi, are invariant features of membranes in eukaryal cells, in which they appear to stiffen the membrane by increasing the degree of order of the hydrocarbon chains. Sterols are not commonly found in bacteria and archaea, except for the cell wall-less Mycoplasma. Very complex lipids, including the very long, branched mycolic acids, are common in Mycobacterium but occur in a very thick and rigid outer layer rather than in the cytoplasmic membrane.

B. Hydrophobic effect II. STRUCTURE AND PROPERTIES OF MEMBRANE LIPIDS A. Lipid composition The key ingredients of biological membranes are polar lipids, primarily phospholipids. In most bacteria, phospholipids consist of two fatty acids, usually with 16–18 carbon atoms in the hydrocarbon chain with zero or one cis-double bond. The fatty acid content changes in response to environmental conditions, particularly temperature. As described later, lower growth temperatures result in a higher degree of fatty acid unsaturation, which has dramatic effects on the membrane’s fluidity and function. Some fatty acids are branched or contain cyclopropane rings. Fatty acids are joined in ester linkage to two of the hydroxyl

Phospholipids spontaneously form membranous structures when suspended in aqueous solution. The forces that drive their assembly into a bilayer or more complex structure are called the hydrophobic effect. This organizing force depends on the ability of water molecules to donate and accept hydrogen bonds from one another to form an extensive network of water molecules transiently linked through hydrogen bonds and polar interactions. Polar molecules can participate in this interactive network of water molecules and are thereby able to dissolve in aqueous solution. In contrast, hydrophobic or nonpolar molecules, such as a long hydrocarbon or aromatic chain, are unable to participate in the hydrogen-bonded network of water molecules. For them to dissolve in water would result in loss of the energy resulting from the intrusion in the mobile water bonding and from the organization of

ch18.qxd 10/9/03 7:03 PM Page 225

cell membrane: structure and function

225

FIGURE 18.2 Chemical structures of the fatty acids and phospholipids that comprise the bulk of the membrane lipids in E. coli. For the phospholipids, the R groups represent a fatty acid.

water molecules into a cage-like structure around the intruding nonpolar molecule. It is the loss of the energy of interaction between the water molecules and the entropic cost of organizing them that drive the nonpolar molecules to associate with one another, out of contact with the water. Hence, non-polar lipids tend to form oil droplets in water, so as to present the smallest possible hydrophobic surface to the water.

C. Membrane bilayer Polar lipids, such as phospholipids, have chemical structures of two different natures. The hydrophobic acyl chains strive to be sequestered from contact with water, whereas the charged and polar head groups seek contact with aqueous ions to help dissipate their electrical charge and to form a hydrogen-bonded network with water. Thus, polar lipids are driven by basic physical characteristics to form aggregate structures in which the hydrophobic portions are segregated out of contact with water while the head groups face

the water. There are numerous ways in which these requirements can be accommodated. Of greatest biological relevance is the lamellar bilayer, in which large flat surfaces of bilayer form with the acyl chains facing each other on the inside and the head groups facing the solution on the outside (Figs. 18.3 and 18.4). Other nonlamellar structures, such as hexagonal phases, can form under certain conditions of head group, temperature, and salt concentration. The propensity of different lipids to form nonlamellar bilayers is a function, in part, of the relative cross-sectional areas of the hydrophobic acyl chains in reference to the area of the head group. When the areas of the polar and nonpolar parts are similar, a lamellar bilayer is favored. If the nonpolar part is substantially smaller or larger than the head group, formation of a spherical micelle or an inverted structure, such as the Hexll phase (hexagonal phase II), respectively, is favored. When most biological phospholipids are dried into a film from a solution in an organic solvent and then water is added, the lipids spontaneously form a

ch18.qxd 10/9/03 7:03 PM Page 226

226

the desk encyclopedia of microbiology

FIGURE 18.3 Schematic representation of the lipid bilayer or lamellar arrangement. The polar head groups of the lipids face the water, and the hydrocarbon acyl chains are segregated away from the water in the interior of the bilayer.

multilamellar liposome in which concentric bilayers assemble like an onion. When these liposomes are sonicated (disrupted by intense ultrasonic irradiation), they break down into small unilamellar vesicles. These are small spherical particles with a single membrane bilayer surrounding an aqueous cavity.

D. Lipid phase behavior Above a certain temperature, the lipid molecules in a bilayer can move rapidly within the plane of the membrane but do not move very far in or out of the membrane owing to the hydrophobic effect. In this liquid crystalline state, the acyl chains are parallel to each other but undergo frequent rotations around the carbon bonds to produce kinks in the chain. These kinks provide transient discontinuities in the hydrophobic barrier that allow movement of other lipid molecules within the membrane and of water molecules across the membrane. As the suspension of membranes is cooled below a critical temperature (Tm), there is a transition of the lipids from the liquid crystalline phase to the rigid gel phase. In this phase, all the hydrocarbon chains form the all-trans configuration, which increases the bilayer thickness and greatly decreases diffusion of the lipids within the membrane and of permeants across the membrane. This transition reflects the motion and packing of the acyl chains. The critical temperature at which the gel-to-liquid crystalline phase transition occurs is dependent on the

FIGURE 18.4 Representation of the orientation of the acyl chains and head groups of a phospholipid in a bilayer. This orientation of phosphatidylethanolamine is seen in crystal structures. The solid circles represent oxygen atoms; the heavy lines represent the glycerol; the thin lines are ethanolamine; and the gray lines are the hydrocarbon chains of the two fatty acids, all in their most extended configuration.

lipid composition, including the nature of the head group and the lipid chains. The longer the hydrocarbon chains, the higher the Tm for transition to the liquid crystalline state. The presence of unsaturated fatty acids with one or more double bonds has a very dramatic effect reducing the Tm by as much as 60 C. The effect of the double bond is greatest when it is in the middle of the acyl chain. The double bond introduces a permanent kink or bend in the chain that interferes with packing of the chains in the gel state. Most cells adjust their lipid composition to the growth temperature to ensure that their membrane remains in the liquid crystalline state. Most membrane proteins are excluded from or are inactive in the rigid gel phase membranes. Another lipid phase transition can occur at temperature higher than Tm. This corresponds to the change of certain lipids from a lamellar to a nonlamellar configuration.

ch18.qxd 10/9/03 7:03 PM Page 227

cell membrane: structure and function

III. STRUCTURE AND PROPERTIES OF MEMBRANE PROTEINS Although the lipids are very important for determining the barrier functions of the cell membrane, the membrane proteins confer most of the important functions of biological membranes. There are many different types of membrane proteins, reflecting their very different functions. Integral membrane proteins are those that cross the membrane with one or more transmembrane segments and are usually exposed to both sides of the membrane. They cannot be easily removed from the membrane unless detergents are added to disrupt the bilayer structure. Some integral membrane proteins are stably anchored to the membrane, usually by covalent attachment to a lipid such as a fatty acid, isoprenoid, phosphatidyl inositol, or a lipoprotein derivative. The membrane-spanning proteins include most of the functionally important transporters and signal receptors. Peripheral membrane proteins are defined operationally as those that are readily removed from association with the membrane by procedures that do not disrupt the bilayer, such as washing with high salt, urea, or sodium carbonate at pH 11. These proteins generally lack transmembrane segments, although it is recognized that some peripheral proteins can transiently insert a segment across the membrane as part of their normal function. Some integral membrane proteins possess one or two transmembrane segments only, and the bulk of these proteins is located in the aqueous solutions on one or both sides of the membrane. The nonmembranous segments of these proteins are similar in character to that of a normal soluble globular protein. Charged or polar amino acid residues line the surfaces exposed to the water, and nonpolar amino acid residues form the interior of the domain, driven into this sequestered state by the same hydrophobic effect that stabilizes the membrane lipid structure. The membrane-spanning portion of these proteins has a very limited range of composition and structures. Owing to the very hydrophobic environment of the interior of the membrane bilayer, the presence of charged or polar residues or of unpaired hydrogen bonds is energetically unfavorable. Thus, the amino acid residues that comprise single transmembrane segments are generally highly hydrophobic. In addition, an helix is the only peptide conformation in which all the hydrogen bonding possibilities of the polypeptide backbone are satisfied. Thus, single or double transmembrane segments are most likely to be non-polar -helical segments, with lengths of approximately 20 amino acids

227

(approximately six turns of the helix), which is sufficient to span the width of the membrane. Many membrane proteins cross the membrane multiple times and insert the bulk of their mass within the membrane. Transport proteins typically have 10–14 transmembrane segments, and a major class of signal receptors have seven transmembrane segments. These proteins have a very different character than that of the multidomain bitopic proteins described previously. The large surface of the protein that is imbedded in the membrane and exposed to the hydrophobic environment of the hydrocarbon lipid chains is highly hydrophobic and cannot possess charged amino acid residues. Other surfaces of these proteins are exposed to the solution on either side of the membrane. These surfaces, which are composed of the loops joining transmembrane segments, must possess mainly polar residues capable of remaining soluble in water. The presence of the very different surfaces of membrane proteins, two polar belts and one very nonpolar belt, holds the protein tightly within the membrane bilayer and restricts its movement out of the membrane. The transmembrane segments of such a polytopic protein are -helical in the few proteins for which detailed structural information is available (the photosynthetic reaction center, some electron transport complexes, bacteriorhodopsin, the vitamin B12 transporter, and lactose permease). The amino acid residues that comprise transmembrane segments need not be all nonpolar. These residues can be exposed to different environments, namely the lipid hydrocarbon chains, the neighboring -helical transmembrane segments, and a potential water-filled channel. Thus, the residues of these transmembrane segments exhibit periodic variability, with very non-polar residues along one face and generally polar residues along the opposite face. In contrast, the transmembrane segments of most bacterial outer membrane proteins are composed of antiparallel sheets of approximately eight amino acid residues in length. Proteins are inserted in cellular membranes in a defined orientation. The orientation of the entire protein appears to be determined by the orientation of its individual transmembrane segments. The orientation of each transmembrane segment is determined mainly by the nature of the charged residues flanking the transmembrane segment rather than by the residues within the transmembrane segment. The “inside-positive” rule seems to govern the orientation of transmembrane segments in bacteria, and it states that relatively short, positively charged loops are retained on the cytoplasmic side of the membrane. The orientation of a protein can be affected by its association with other cellular or

ch18.qxd 10/9/03 7:03 PM Page 228

228

the desk encyclopedia of microbiology

membrane proteins or by the presence of a signal sequence that directs the protein into the secretory pathway. The orientation of a membrane protein can be determined experimentally by detecting the sites of action of proteases or other enzymes added to one side of the membrane. A simpler and more generally informative approach makes use of fusions of a part of the tested protein to a topological reporter, which is an enzyme whose activity depends on which side of the membrane it resides. It has thus been possible to gain a considerable degree of understanding of the structure of many membrane proteins, even without high-resolution crystallographic information.

IV. FUNCTIONS OF CYTOPLASMIC MEMBRANE The cytoplasmic membrane of bacteria is a very busy site at which an almost bewildering number of important processes occur. It carries out most of the reactions that are handled by the many organelles of eukaryotic cells. Since bacteria are generally far more metabolically capable and diverse than eukaryotic cells, it is not surprising that an estimated one-fourth of the cell’s proteins are membrane associated. One of the key principles in biology is the basic universality of bioenergetics, which refers to the processes of energy generation, storage, and utilization in cells. The major feature of these processes is the use of ion gradients that are formed across cellular membranes, such as the membranes of mitochondria and chloroplasts in higher organisms or the cytoplasmic membrane of bacteria. In most systems, the gradient of protons is the central factor in bioenergetics, although gradients of sodium ions are used by some bacteria for energy generation and by most eukaryotic cells for cellular signaling and nutrient transport. The chemiosmotic proposal, initially made by Peter Mitchell, that ion gradients are the intermediate between the processes of electron transport and the formation of ATP has been overwhelmingly accepted. In bacteria, ion gradients are also used to drive several types of active transport systems, bacterial motility, and protein secretion.

A. Energy generation Energy generation refers to the trapping in a metabolically useful form of the energy absorbed from sunlight or released by reduction of some inorganic molecule or the oxidation or breakdown of an organic molecule serving as energy source. Bacteria can generate

energy by many different processes. In simple fermentative pathways, such as the glycolytic breakdown of glucose to lactate, ATP can be formed during several enzymatic steps by the process of substrate level phosphorylation. Important energy-producing processes use electron transport chains to pass electrons from a carrier of high negative redox potential to carriers of successively lower energy states. During respiration, electrons enter these chains following transfer from an organic molecule and are ultimately transferred to an inorganic electron acceptor. During photosynthesis electrons are excited to a higher energy state following absorption of light by a chlorophyll-related molecule. Figure 18.5 summarizes several key steps of microbial energy generation. During the processes of electron transport to carriers of successively higher redox potential, there is a separation of charge across the membrane. This is ultimately coupled to the movement of protons from one side of the membrane to the other, resulting in the formation of a proton motive force (pmf) which has two aspects. Movement of the positively charged proton creates an electrical charge across the membrane, which is termed . In bacteria and mitochondria, in which electron transport results in the release of protons, the electrical charge is negative inside and can be 100–200 mV. Proton pumping also results in a difference in proton concentration, or pH, across the membrane, such that the exterior is usually more acidic than the interior. Photosynthetic or respiratory electron transport thus results in proton pumping and creation of the pmf. The proton gradient can be tapped to bring about the formation of ATP, which is the ultimate energy source for most energy-requiring processes in the cytoplasm. Synthesis of ATP is carried out by a family of protein complexes, which include the F1F0 protontranslocating ATPases of mitochondria and bacteria. Related complexes are found in chloroplasts and archaea. These protein complexes contain a membraneembedded sector, the F0 portion, which includes the pathway to allow the protons to flow back into the cell in response to the chemical and electrical forces acting on them. The F1 portion of the complex contains the sites for conversion of ADP Pi to ATP, and the energy for this process is coupled to the movement of protons. The stoichiometry of the process is such that entry of three or four protons results in the formation of one molecule of ATP. The individual steps of proton movement through the F1F0-ATPase and ATP synthesis or hydrolysis are tightly coupled under most conditions to prevent wasteful loss of the pmf or of the ATP pool in the cell and consequent heat generation. In bacteria, this ATP synthase can

ch18.qxd 10/9/03 7:03 PM Page 229

cell membrane: structure and function

229

FIGURE 18.5 Summary of some processes of generation of metabolic energy in bacteria. (Left) Components of respiratory electron transport chains, in which specific substrate dehydrogenases oxidize their substrate, transfer the released electrons to membrane quinones, and in some cases extrude protons to the exterior. The electrons of the reduced quinones are transferred by the terminal oxidase or reductase to the terminal electron acceptor, such as oxygen, with the extrusion of additional protons. (Bottom) Photosynthetic systems whereby absorption of light is converted to a transmembrane gradient of protons. (Right) Two examples in which substrate/product exchange and metabolism result in generation of pmf. (Top) The electrical and chemical components of the proton motive force and a representation of the action of the F1F0ATP synthase which interconverts the proton gradient and ATP synthesis or hydrolysis.

function in a reversible manner to allow ATP that was generated by substrate-level phosphorylation to drive formation of a proton gradient which can then be used to drive transport systems or motility. Movement of protons causes the F0 sector to rotate within the membrane, like the action of a turbine. The rotation of the F0 sector is coupled to changes in the conformation of the nucleotide-binding sites in the stationary F1 sector, which is linked to interconversion of ADP Pi to ATP. 1. Photosynthesis Photosynthesis traps the energy of sunlight and converts it into metabolically useful forms such as ATP during cyclic electron transport and NADPH during noncyclic electron transport processes. In some organisms, photosynthetic electron transport is coupled to the formation of oxygen from water—the process that is essential for aerobic life. There are numerous

pigments in cells that absorb light energy for use in photosynthesis, but the most important of these are the chlorophylls. Chlorophylls are porphyrin molecules, similar to heme, but containing a magnesium atom instead of iron. Some pigments are carried in protein molecules that serve as antennae or light-harvesting complexes, but the key processes occur in a protein complex called the photosynthetic reaction center, which typically consists of three proteins that spread across the membrane and contain bacteriochlorophyll, bacteriopheophytin, menaquinone, and nonheme iron as electron carriers. The light energy is ultimately absorbed by a chlorophyll molecule and this energy excites an electron to a higher energy level. This excited electron passes through a series of electron-carrying prosthetic groups within the reaction center and then out through the pool of membrane-bound quinones, which transfer the electron to a cytochrome-containing electron transport chain. Passage of the electron through the electron transport

ch18.qxd 10/9/03 7:03 PM Page 230

230

the desk encyclopedia of microbiology

chain is coupled to pumping of protons across the specialized membranes containing the photosynthetic apparatus. In cyclic phosphorylation, as carried out in the photosynthetic bacteria, the electron ultimately returns to the chlorophyll molecules after transfer to the periplasmic heme protein, cytochrome c. In the process of noncyclic phosphorylation in plants and cyanobacteria, the electron can be transferred ultimately to pyridine nucleotides for use as a reductant in biosynthetic processes. In this process, the electron can be replaced on chlorophyll by another lightabsorption process that removes electrons from water to create oxygen. A completely different system for conversion of light into metabolic energy is present in Halobacterium salinarum, an extremely halophilic archaeon that thrives in very saline environments such as the Dead Sea or brine evaporation ponds. These bacteria produce patches of membrane that are densely packed with the membrane protein, bacteriorhodopsin, having seven transmembrane helices and covalently bound retinal as in the visual pigment in mammalian retina. Light absorption by the retinal causes the isomerization of one of the double bonds in the molecule, causing a change in the conformation of the protein which results in the change of the pK of several acidic groups on either side of the membrane. The consequence of these changes is that a proton is released from the bacteriorhodopsin on the outside of the membrane and replaced by one from the cytoplasm. In this way, light is directly converted into a transmembrane proton gradient without the requirement of an electron transport chain.

oxygen or other acceptor is coupled to proton movement across the membrane. In E. coli, electron donors for respiration include NADH, succinate, glycerol 3-P, formate, lactate, pyruvate, hydrogen, and glucose. Electron acceptors include oxygen, nitrate, nitrite, fumarate, dimethylsulfoxide, and trimethylamineN-oxide. Typical respiratory systems contain two to four transmembrane protein complexes. These include substrate-specific dehydrogenases, which transfer electrons from the donor to quinones in the membrane. The reduced quinones migrate to another protein complex which accepts electrons from them and transfers the electrons to cytochromes and ultimately to the terminal electron acceptor. The transmembrane protein complexes often contain flavin and/or nonheme iron and are arranged in the membrane in such a way that the passage of electron results in the release of proton to the outside or its consumption from the cytoplasm, i.e. the formation of the pmf.

2. Respiration

B. Membrane transport

Respiration is the process whereby electrons from the metabolism of an energy source are transferred through a proton-pumping electron transport chain to some inorganic molecule. The most familiar form of respiration is aerobic respiration, in which oxygen serves as the ultimate electron acceptor. Owing to the ability of oxygen to accept electrons, aerobic respiration is the most energetically favorable, but some partially reduced forms of oxygen, hydrogen peroxide, superoxide anion, and hydroxyl radical, are extremely reactive and thus toxic to the organism. Many bacteria are capable of carrying out anaerobic respiration, in which the electrons are transferred to alternative acceptors, such as nitrate, nitrite, sulfate, or sulfite. These processes yield less energy but can occur in anoxic environments. All respiratory metabolism uses electron transport chains, whereby electron transfer from the donor to

Biological membranes form the permeability barrier separating the cell from its environment. The hydrophobic barrier of the membrane bilayer greatly restricts passage of polar molecules, although non-polar molecules can pass. Transport mechanisms exist to move nutrients and precursors into the cell and metabolic products, surface components, and toxic materials out of the cell. Several types of transport mechanisms and families of transporters have been identified.

3. Coupled processes Some bacteria couple the transport and metabolism of their energy source directly to the production of the pmf. An example of this very simple, but not very energy-rich, process is malo-lactate fermentation in Leuconostoc. The substrate malate is transported into the cell and converted to lactate, which leaves the cell in exchange for a new molecule of malate. The net result of this process is the movement of one negative change into the cell and the consumption of one proton inside the cell, which results in the creation of a pmf that is interior negative and alkaline.

1. Types of transport systems Transport can occur through energy-dependent and energy-independent processes. Several general classes of transport process have been identified. Passive diffusion occurs spontaneously without the involvement of metabolic energy or of transport proteins. It only allows the flow of material down a concentration

ch18.qxd 10/9/03 7:03 PM Page 231

cell membrane: structure and function gradient, and the rate of this process is a linear function of the concentration gradient. The rate of passive diffusion depends on the ability of the permeant to dissolve in the membrane bilayer and thus depends of the polarity of the permeant and its size. These factors are related to the ability of the permeant to fit into transient defects that form in the membrane bilayer. Only water and a few hydrophobic molecules use this mechanism for entry into bacteria. Facilitated diffusion requires the operation of a membrane protein for passage of the permeant across the membrane. These transporters merely provide a route for diffusion of their substrate down its concentration gradient, and thus the concentration of the substrate on both sides of the membrane will become equal. Transport is not dependent on the polarity of the substrate and usually exhibits stereospecificity, in which isomeric forms of the same compound are transported at very different rates. Because of the involvement of the transporter as a catalyst for movement, the rate of transport can be saturated in the same manner as an enzyme-catalyzed reaction. A possible example is the glycerol facilitator GlpF of E. coli, a transmembrane protein that allows glycerol and other small molecules to diffuse across the cytoplasmic

231

membrane at rates much faster than they cross lipid bilayers. However, this protein may act in the manner of a channel rather than a carrier. The overwhelming majority of transport systems in bacteria catalyze active transport and expend metabolic energy to allow the accumulation of even very low external concentrations of a nutrient to a much greater concentration inside the cell. Active transport is carried out by a transport protein or complex and thus exhibits substrate stereospecificity and rate saturation. The difference compared to facilitated diffusion is that the substrate can be accumulated at concentrations as much as 1 million times higher than that outside. This accumulation requires the expenditure of energy, and in the absence of energy many but not all transport mechanisms can carry out facilitated diffusion. These active transport systems differ in their molecular complexity and in the mechanism by which metabolic energy is coupled to substrate accumulation. It is important to distinguish active transport, in which the substrate is accumulated in unaltered form, from group translocation, in which the substrate is converted into a different molecule during the process of transport. Some types of transport processes in bacterial cells are summarized in Fig. 18.6.

FIGURE 18.6 Schematic representation of several types of active transport systems in bacteria. Top, transporters linked to the pmf; left, ATP-driven transports; bottom, examples of systems that carry out release of metabolic products of toxic chemicals. (Right) A presentation of all the transport systems known to mediate uptake or release of potassium in E. coli. Some of these transporters are ATP driven; others are coupled to the pmf; and MscL is a channel activated by mechanical stretch of the membrane.

ch18.qxd 10/9/03 7:03 PM Page 232

232

the desk encyclopedia of microbiology

2. ATP-driven active transport Several groups of transport mechanisms use the energy gained during ATP hydrolysis to drive active transport. One of these groups is called the P-type ion-translocating ATPases to indicate the fact that a phospho-enzyme is formed as an intermediate in their reaction cycle. Typically, these transport systems consist of a large polypeptide of approximately 100 kDa which spans the membrane and contains the site for ATP binding and the residue that is phosphorylated. A smaller subunit usually participates in the activity. The ATP is used to phosphorylate a specific acidic aspartate residue, and this phosphorylation causes a change in conformation of the transporter that is part of the ion pumping process. Several examples of this type of transport system have been extensively studied. The sodium/potassium ATPase in the plasma membrane of higher organisms is responsible for pumping sodium out and potassium into cells, thereby generating the ion gradients that are necessary for many steps of nutrient transport and for neural signal transmission. The electrical potential that exists across the plasma membrane of mammalian and some other cells is based mainly on the difference in sodium ion concentration that is maintained by the action of this transporter. Similarly, the calciumtranslocating ATPase located in the sarcoplasmic reticulum acts to lower the intracellular calcium concentrations that accumulate following the processes that initiate muscle contraction. Other P-type ATPases include the proton-translocating ATPase in the plasma membranes of fungi and plants, which establishes the gradient of protons that is used to drive many of these cells’ nutrient transport systems, and some of the transport systems for magnesium or potassium ions in bacteria. Although P-type ATPases function only in the transport of ions, another family of ATP-driven systems transports a wide range of substrates and is involved in the uptake of numerous types of nutrients and in the efflux of both surface and secreted macromolecules (proteins, carbohydrates, and lipids) and of toxic chemicals. This family of transporters is usually the largest family of any set of related genes in those organisms whose genomes have been completely sequenced. Although the subunit composition of these transporters differs, they are called the ABC family to indicate the presence in at least one subunit of a highly conserved ATP-binding cassette, a protein domain that couples ATP binding and hydrolysis to the transport process. A more descriptive name for these proteins is traffic ATPases. One subset of the family of ABC transporters includes a large group of nutrient uptake mechanisms

present only in bacteria and archaea and called periplasmic permeases. These transport systems, such as those for histidine, maltose, oligopeptides, etc., consist of a heterotetramer in which two highly hydrophobic transmembrane proteins with usually five or six membrane-spanning segments are associated with two subunits that contain the ATP-binding cassette and are mainly exposed to the cytoplasm. A fifth protein subunit is responsible for the substrate specificity of these transport systems. This substratebinding protein usually has very high affinity for the substrate and allows uptake of nutrients even in nanomolar-range concentrations. The structure of these substrate-binding proteins resembles a clam, with two large lobes hinged in the middle. The substrate binds to specific residues in both lobes, which close around the substrate molecule for carriage to the membrane-bound components and entry into the cell. In gram-negative bacteria, the substrate-binding protein floats freely in the periplasmic space between the cytoplasmic and outer membranes. Gram-positive bacteria lack the outer membrane and, to prevent its loss, the binding protein is tethered to the cytoplasmic membrane by a lipoprotein anchor. The mechanism by which ATP hydrolysis is coupled to the release of substrate from the binding protein and its movement across the membrane is currently being studied. A striking feature of these transporters is that they act in a unidirectional manner and only allow nutrient to enter the cell but not to be released. The basic features of the ABC transport process and homologous transport components also operate in the opposite direction in many processes of macromolecular export, including proteins and surface carbohydrates. 3. Transporters coupled to ion gradients In addition to ATP-driven active transport systems, bacteria possess many transporters in which the movement of their substrate is obligately coupled to the movement, in the same or opposite direction, of an ion. In this way, accumulation of a substrate is coupled to the expenditure of the gradient of the coupling ion, which is usually a proton or sodium ion. Transmembrane ion gradients are a very convenient source of energy for active transporters. Entry into the cell of at least three protons must occur for synthesis of one molecule of ATP. It is thus very economical for the cell if it can accumulate a molecule of substrate at the expenditure of one proton rather than having to expend one ATP molecule for the same purpose. Symport refers to the process in which the coupling ion moves in the same direction as the substrate, that is, when the downhill movement of a proton into the

ch18.qxd 10/9/03 7:03 PM Page 233

cell membrane: structure and function negative and alkaline interior of the cell is coupled to the uptake of substrate. Antiport is the reverse process, in which the two molecules move in opposite directions. If movement of substrate is coupled to movement of a proton in a 1 : 1 stoichiometry, then a pmf of 120 mV can achieve a 100-fold accumulation of substrate inside the cell. A pmf of 180 mV can allow a 1000-fold gradient of substrate. Uniporters allow coupling of the movement of a positively charged molecule to the pmf without movement of any other ion. The cationic molecule is drawn into the negatively charged cell interior simply by electrostatic attraction. These types of transporters are referred to as secondary active transport systems since they use the pmf that was generated by other means and do not use an immediate source of energy, such as ATP. Ion-coupled transport systems are inhibited by conditions that dissipate or prevent formation of the pmf, such as ionophores which allow ions to distribute across the membrane in response to the electrical and chemical gradients that act on it. An uncoupler or protonophore, such as 2,4-dinitrophenol or carbonylcyanide p-trifluoro-methoxy phenylhydrazone, is a hydrophobic molecule that can cross the membrane in either its ionized or its neutral form. Its presence allows protons to equilibrate across the membrane, thereby dissipating both the electrical and the chemical gradients of protons and thus the entire pmf. The ionophore valinomycin carries potassium ions and allows them to distribute across the membrane in response to electrical or chemical gradients. The addition of valinomycin to cells or membrane vesicles that have a pmf allows potassium ions to accumulate inside the negatively charged interior. This accumulation results in dissipation of the electrical potential . The ionophore nigericin carries protons and sodium or potassium ions across the membrane, but only in the process of exchange. The action of nigericin thus does not result in any net gain or loss of charge and thus does not dissipate . However, if there is a concentration gradient of protons, pH, nigericin allows the concentration gradient to dissipate, whereas the electrical potential is maintained or even increased. Ion-coupled transporters typically consist of a single polypeptide chain with 12 transmembrane segments, although examples with 10–14 transmembrane segments are known. It has been proposed that these proteins arose as the result of tandem duplication of a precursor protein with six transmembrane segments. In the well-studied E. coli lactose permease LacY, several major experiments demonstrated the coupling of lactose accumulation to the pmf. The magnitude of the pmf affects the magnitude of the accumulation ratio of lactose inside the cell in a direct manner indicative of a

233

1 : 1 stoichiometry of lactose and proton. When membrane vesicles are energized by provision of a substrate for the electron transport system, a pmf is generated and lactose is accumulated. Even when a final steadystate level of lactose accumulation is reached, the lactose is in continual movement in both directions across the membrane. At the steady state, the rates in and out of the vesicle are equal, although the internal concentration of lactose is much higher than the external concentration. This indicates that the energy has resulted in a decreased affinity of the carrier for lactose on the inside face of the membrane relative to its affinity on the outside. Instead of using the electron transport system, a transmembrane electrical potential, interior negative, can be generated experimentally by diluting vesicles loaded with a high concentration of potassium ions into a medium of low potassium concentration in the presence of valinomycin. The potassium ions flow out down their concentration gradient, carrying positive charge out of the vesicle and leaving behind an interior negative charge. This negative interior can attract protons into the vesicle through the lactose permease, thereby driving lactose accumulation. Finally, if unenergized vesicles are placed in an unbuffered solution that contains a high concentration of lactose, the lactose will flow into the vesicle, bringing along a proton and thereby causing a measurable decrease in the pH of the medium. All these results provide convincing evidence for the coupled movement of proton and lactose. Transporters are designed to prevent uncoupled movement of substrate without protons or of protons without substrate. If the latter case occurred, it would result in the operation of an uncoupler and allow the futile dissipation of the pmf. How the binding of a proton affects the affinity or binding of the substrate remains an intriguing and central question. It has been shown that the proton must bind before the lactose. If saturating concentrations of lactose are present on both sides of the membrane, the lactose transporter carries out their very rapid exchange independent of the release or re-binding of the proton. This result indicates that the reorientation of the loaded substrate-binding site from facing the interior to facing the exterior does not require changes in proton binding by LacY. If lactose is present on only on