Exploring the Role of Neuroplasticity - Neurociência (2025)

Exploring the Role of Neuroplasticity - Neurociência (4)

Exploring the Role of Neuroplasticity - Neurociência (5)

Exploring the Role of Neuroplasticity - Neurociência (6)

Exploring the Role of Neuroplasticity - Neurociência (7)

Exploring the Role of Neuroplasticity - Neurociência (8)

Exploring the Role of Neuroplasticity - Neurociência (9)

Exploring the Role of Neuroplasticity - Neurociência (10)

Exploring the Role of Neuroplasticity - Neurociência (11)

Exploring the Role of Neuroplasticity - Neurociência (12)

Exploring the Role of Neuroplasticity - Neurociência (13)

Prévia do material em texto

<p>Citation: Marzola, P.; Melzer, T.;</p><p>Pavesi, E.; Gil-Mohapel, J.; Brocardo,</p><p>P.S. Exploring the Role of</p><p>Neuroplasticity in Development,</p><p>Aging, and Neurodegeneration. Brain</p><p>Sci. 2023, 13, 1610. https://doi.org/</p><p>10.3390/brainsci13121610</p><p>Academic Editor: Mahesh</p><p>Kandasamy</p><p>Received: 23 October 2023</p><p>Revised: 16 November 2023</p><p>Accepted: 18 November 2023</p><p>Published: 21 November 2023</p><p>Copyright: © 2023 by the authors.</p><p>Licensee MDPI, Basel, Switzerland.</p><p>This article is an open access article</p><p>distributed under the terms and</p><p>conditions of the Creative Commons</p><p>Attribution (CC BY) license (https://</p><p>creativecommons.org/licenses/by/</p><p>4.0/).</p><p>brain</p><p>sciences</p><p>Review</p><p>Exploring the Role of Neuroplasticity in Development, Aging,</p><p>and Neurodegeneration</p><p>Patrícia Marzola 1 , Thayza Melzer 1, Eloisa Pavesi 1 , Joana Gil-Mohapel 2,3,* and Patricia S. Brocardo 1,*</p><p>1 Department of Morphological Sciences and Graduate Neuroscience Program, Center of Biological Sciences,</p><p>Federal University of Santa Catarina, Florianopolis 88040-900, SC, Brazil; patriciarmarzola@gmail.com (P.M.);</p><p>melzer.th@gmail.com (T.M.); eloisapavesi@gmail.com (E.P.)</p><p>2 Division of Medical Sciences, University of Victoria, Victoria, BC V8P 5C2, Canada</p><p>3 Island Medical Program, Faculty of Medicine, University of British Columbia, Victoria, BC V8P 5C2, Canada</p><p>* Correspondence: jgil@uvic.ca (J.G.-M.); patricia.brocardo@ufsc.br (P.S.B.)</p><p>Abstract: Neuroplasticity refers to the ability of the brain to reorganize and modify its neural con-</p><p>nections in response to environmental stimuli, experience, learning, injury, and disease processes. It</p><p>encompasses a range of mechanisms, including changes in synaptic strength and connectivity, the</p><p>formation of new synapses, alterations in the structure and function of neurons, and the generation</p><p>of new neurons. Neuroplasticity plays a crucial role in developing and maintaining brain function,</p><p>including learning and memory, as well as in recovery from brain injury and adaptation to environ-</p><p>mental changes. In this review, we explore the vast potential of neuroplasticity in various aspects of</p><p>brain function across the lifespan and in the context of disease. Changes in the aging brain and the</p><p>significance of neuroplasticity in maintaining cognitive function later in life will also be reviewed. Fi-</p><p>nally, we will discuss common mechanisms associated with age-related neurodegenerative processes</p><p>(including protein aggregation and accumulation, mitochondrial dysfunction, oxidative stress, and</p><p>neuroinflammation) and how these processes can be mitigated, at least partially, by non-invasive and</p><p>non-pharmacologic lifestyle interventions aimed at promoting and harnessing neuroplasticity.</p><p>Keywords: aging; cognitive function; lifestyle interventions; neurodegeneration; neurodevelopment;</p><p>neuroplasticity</p><p>1. Introduction</p><p>The concept of neuroplasticity was first introduced by William James in 1890, and a few</p><p>decades later, Jerzy Konorski coined the term “neural plasticity” [1,2]. Neuroplasticity refers</p><p>to changes in brain structure and function throughout the lifespan. Neuroplasticity enables</p><p>the brain to change and adapt to intrinsic or extrinsic stimuli by reorganizing its structure,</p><p>function, or connections, resulting in physiological and morphological modifications. This</p><p>dynamic process allows us to adjust to different experiences and circumstances and plays a</p><p>significant role in learning, memory, and recovery from brain injuries [3].</p><p>Due to the multifaceted nature of neuroplasticity, different types of plasticity can</p><p>impact brain structure and function [4,5]. Structural neuroplasticity refers to changes in the</p><p>physical structures of neurons and neural networks, including the number, shape, strength,</p><p>and connectivity of synapses [6], thus enabling the brain to adapt to changing environments</p><p>and experiences. Numerous studies have indicated that structural plasticity occurs during</p><p>development and continues into adulthood [7–9]. On the other hand, functional neuro-</p><p>plasticity refers to changes in neural network properties that involve efficiency, strength,</p><p>and synchrony changes of synapses. Functional plasticity occurs rapidly, affecting various</p><p>cognitive and behavioral processes relating to attention, memory, and perception [5,10].</p><p>One well-studied example of structural neuroplasticity is adult neurogenesis, the pro-</p><p>cess by which new neurons are generated in the adult brain. This process occurs primarily</p><p>in the subventricular zone (SVZ) that lines the lateral ventricles and in the dentate gyrus</p><p>Brain Sci. 2023, 13, 1610. https://doi.org/10.3390/brainsci13121610 https://www.mdpi.com/journal/brainsci</p><p>https://doi.org/10.3390/brainsci13121610</p><p>https://doi.org/10.3390/brainsci13121610</p><p>https://creativecommons.org/</p><p>https://creativecommons.org/licenses/by/4.0/</p><p>https://creativecommons.org/licenses/by/4.0/</p><p>https://www.mdpi.com/journal/brainsci</p><p>https://www.mdpi.com</p><p>https://orcid.org/0000-0001-8152-5674</p><p>https://orcid.org/0000-0003-0428-6694</p><p>https://orcid.org/0000-0003-4982-1662</p><p>https://doi.org/10.3390/brainsci13121610</p><p>https://www.mdpi.com/journal/brainsci</p><p>https://www.mdpi.com/article/10.3390/brainsci13121610?type=check_update&version=1</p><p>Brain Sci. 2023, 13, 1610 2 of 32</p><p>of the hippocampus, a brain region essential for learning and memory [9,11–13]. Several</p><p>studies have shown that increased physical activity, exposure to enriched environments,</p><p>and certain drugs can enhance neurogenesis and improve learning and memory [8,14–16].</p><p>Another example of structural neuroplasticity is dendritic spine remodeling, the process</p><p>by which dendritic spines change in size, shape, and number in response to experience.</p><p>Animal studies have shown that dendritic spine remodeling also plays a crucial role in</p><p>learning and memory [17,18].</p><p>Functional neuroplasticity is thought to underlie memory formation, skill acquisition,</p><p>and recovery from injury. An example of functional neuroplasticity is long-term potentia-</p><p>tion (LTP), the persistent strengthening of synapses in response to repeated stimulation.</p><p>LTP is thought to be a key mechanism underlying learning and memory [19]. Conversely,</p><p>long-term depression (LTD) is the persistent weakening of synapses and also plays a role</p><p>in learning and memory [20,21]. Another example of functional neuroplasticity is cortical</p><p>reorganization, the process by which the brain’s sensory maps can change in response to</p><p>experience or injury. Learning new abilities results in changes to functional connectivity</p><p>among brain areas involved with motor control, sensory processing, and attention. For</p><p>example, blind individuals can have enhanced sensory processing in other modalities, such</p><p>as touch and hearing, due to cortical reorganization [22].</p><p>In the perinatal and early childhood periods, the brain undergoes rapid and extensive</p><p>growth and development, during which plasticity is particularly high. Studies have</p><p>shown that this period is characterized by a heightened sensitivity to environmental input,</p><p>which facilitates the formation of new neural connections [23]. In contrast, plasticity in</p><p>later stages of the lifespan is more tightly regulated and context-dependent. Changes in</p><p>neural activity, environmental factors, and behavioral outcomes can trigger the release of</p><p>specific neurotransmitters, enabling changes in neural connections only under contextual</p><p>conditions that facilitate plasticity [24]. Moreover, recent studies have suggested that the</p><p>regulation of plasticity in the mature brain occurs as a continuum, with different levels of</p><p>plasticity occurring under different conditions [25]. These findings suggest that plasticity</p><p>is a dynamic process that can be modulated and affected by various factors, including</p><p>age, experience, and environmental conditions. Understanding these factors can aid in</p><p>developing effective strategies to harness the power of neuroplasticity and minimize its</p><p>negative effects, leading to better treatments and outcomes for various neurological and</p><p>neurodegenerative conditions.</p><p>2. Neuroplasticity</p><p>Neuroplasticity,</p><p>adults [321], and promoted cognitive function</p><p>in individuals with mild cognitive impairment and dementia [322–324]. A systematic re-</p><p>view and meta-analysis of 17 randomized controlled trials found that exercise interventions</p><p>improved cognitive function, including memory, attention, processing speed, and executive</p><p>function, in older adults [325]. A separate meta-analysis of 29 randomized controlled trials</p><p>also found that exercise interventions were associated with significant improvements in</p><p>cognitive function in healthy older adults [326]. A recent study using transcranial mag-</p><p>netic stimulation (TMS) found that older adults who engaged in regular physical exercise</p><p>had more remarkable cortical plasticity than those who did not exercise regularly [327].</p><p>This suggests that exercise directly impacts the ability of the brain to adapt and change</p><p>in response to environmental stimuli during aging. In addition, a study using magnetic</p><p>resonance imaging (MRI) found that older adults who engaged in regular physical activity</p><p>had greater gray matter volume in the prefrontal cortex, a brain region important for</p><p>higher-order cognitive functions and decision making [328].</p><p>Together, these studies suggest that exercise can be a powerful tool for promoting</p><p>neuroplasticity and cognitive health during aging. By increasing trophic support through</p><p>the production of BDNF and promoting changes in brain structure and function, exercise</p><p>may help protect the brain against age-related cognitive decline and improve the quality of</p><p>life in later years.</p><p>4.4.2. Cognitive Stimulation and Socialization</p><p>Several lines of evidence have suggested that cognitive stimulation can effectively</p><p>promote neuroplasticity and brain health during aging. Cognitive stimulation encompasses</p><p>activities that challenge the brain, such as reading, writing, playing cognitive games, or</p><p>learning new skills. These activities promote the formation of new neuronal connections</p><p>and can help maintain cognitive function during aging [329]. A systematic review and meta-</p><p>analysis found that engaging in mentally stimulating activities was associated with a lower</p><p>risk of cognitive decline and dementia [330]. Older adults engaged in mentally stimulating</p><p>activities had greater gray matter volume in brain regions important for memory and</p><p>cognitive function [331], and an active cognitive lifestyle is associated with a more favorable</p><p>cognitive trajectory in older persons [332]. Leisure activities such as reading, playing board</p><p>games, playing musical instruments, and dancing were associated with a reduced risk of</p><p>dementia in individuals older than 75 [333]. Learning new skills is one way to engage in</p><p>mentally stimulating activities. In fact, learning a new skill, such as juggling, was associated</p><p>with changes in brain structure and function, including increased gray matter volume in</p><p>the visual and motor areas of the brain [81,82].</p><p>Socially interacting with others, such as friends and family, and engaging in social</p><p>activities can also promote cognitive and emotional stimulation. Older adults who engaged</p><p>in social activities, such as volunteering or participating in community events, had a lower</p><p>risk of cognitive decline than those who did not engage in such activities [334]. On the other</p><p>hand, a study found that social isolation was associated with a higher risk of dementia in</p><p>older adults. This study followed over 2000 older adults for up to 7 years and found that</p><p>those who were socially isolated had a 60% higher risk of developing dementia than those</p><p>with social support [335]. Of note, socialization has been shown to promote the formation</p><p>of new neuronal connections and enhance cognitive function. Indeed, socializing with</p><p>Brain Sci. 2023, 13, 1610 18 of 32</p><p>others was associated with increased gray matter volume in brain regions important for</p><p>memory and social cognition [336].</p><p>Furthermore, several studies showed that genetic and lifestyle factors play a role in</p><p>determining the individual risk of dementia and cognitive impairment. The ε4 allele of the</p><p>apolipoprotein E (APOE ε4) gene is the strongest known genetic risk factor for dementia</p><p>and cognitive impairment. The association between APOE ε4 and faster cognitive decline</p><p>was reduced in participants who were regularly engaged in productive activities [337].</p><p>4.4.3. Diet and Caloric Restriction</p><p>Diet plays a significant role in promoting neuroplasticity, especially during the aging</p><p>process. The brain is a complex organ that requires numerous nutrients, including vitamins,</p><p>minerals, antioxidants, and essential fatty acids [338]. Several studies have found that a</p><p>healthy diet rich in fruits, vegetables, whole grains, and lean protein provides the nutrients</p><p>necessary for optimal brain function [339,340].</p><p>In particular, the role of omega-3 fatty acids in promoting neuroplasticity is well</p><p>documented. These fatty acids are essential for brain health and are found in high con-</p><p>centrations in fatty fish such as salmon, sardines, and mackerel [341]. For example, a</p><p>randomized controlled trial involving older adults with mild cognitive impairment found</p><p>that supplementation with omega-3 fatty acids for 12 weeks significantly improved cogni-</p><p>tive function compared to the placebo group [342]. In a different study, supplementation</p><p>with omega-3 fatty acids was also shown to improve cognitive function in healthy older</p><p>adults [343]. Furthermore, Dullemeijer et al. (2007) found that higher plasma levels of n-3</p><p>polyunsaturated fatty acids were associated with a reduced decline in sensorimotor speed</p><p>and complex cognitive processing in older adults [344]. In addition to omega-3 fatty acids,</p><p>macro and micronutrients present in balanced diets, such as B vitamins and flavonoids,</p><p>can prevent or mitigate age-related degenerative processes [345].</p><p>Conversely, diets high in saturated and trans fats have been linked to cognitive</p><p>decline and dementia. A systematic review and meta-analysis of prospective studies</p><p>found that a higher intake of saturated fats was associated with a higher risk of cognitive</p><p>impairment and dementia [346]. Similarly, a study by Morris et al. found that a diet high</p><p>in saturated fat was associated with a greater risk of developing AD [347]. In agreement,</p><p>Solfrizzi et al. (2006) found that a diet high in trans fats was associated with cognitive</p><p>decline in older adults [348]. Of note, trans fats have been shown to have adverse effects on</p><p>brain function, as they can impair synaptic plasticity, alter membrane composition, and</p><p>increase neuroinflammation [346]. These findings suggest that diets high in saturated and</p><p>trans fats may negatively impact brain health and should be avoided.</p><p>Caloric restriction is another dietary strategy that promotes neuroplasticity and im-</p><p>proves cognitive function. Caloric restriction involves reducing calorie intake by a certain</p><p>percentage while maintaining adequate nutrition [349]. Animal studies have demonstrated</p><p>that caloric restriction can enhance neuroplasticity by increasing the production of neu-</p><p>rotrophic factors, such as BDNF [350,351]. Recent human studies have also suggested that</p><p>caloric restriction may benefit brain function. A randomized controlled trial of overweight</p><p>adults found that a 25% reduction in calorie intake for two years resulted in significant im-</p><p>provements in verbal memory and executive function compared to the control group [352].</p><p>4.4.4. Sleep Hygiene and Quality of Sleep</p><p>Sleep is critical in maintaining brain health and cognitive function, particularly in older</p><p>adults. Numerous studies have demonstrated the importance of sleep in promoting neuro-</p><p>plasticity, particularly during learning and memory consolidation [353,354]. Sleep is also</p><p>essential for clearing out toxic waste products and allowing the brain to regenerate [355].</p><p>Sleep deprivation has been associated with cognitive decline, particularly in older</p><p>adults. Indeed, chronic sleep deprivation has been linked to a faster rate of cognitive</p><p>decline in this population [356], while poor sleep quality was associated</p><p>with a greater</p><p>risk of developing dementia [357]. Recently, a study by Lucey and colleagues (2021) found</p><p>Brain Sci. 2023, 13, 1610 19 of 32</p><p>that disrupted sleep was associated with increased levels of AD-related proteins in the</p><p>cerebrospinal fluid [358].</p><p>Given this, and to promote optimal brain function, it is recommended that older</p><p>adults sleep 7–8 h per night [359]. Also, maintaining good sleep hygiene practices, such</p><p>as avoiding caffeine and electronic devices before bedtime and creating a relaxing sleep</p><p>environment, can help improve the quality and quantity of sleep. Several lines of evidence</p><p>have also highlighted the importance of regular exercise, healthy eating, and managing</p><p>stress for promoting good sleep and overall brain health [360,361].</p><p>5. Conclusions</p><p>Neurodevelopmental exposures, numerous lifestyle factors, acute neurological pro-</p><p>cesses (such as stroke and TBI), and neurodegenerative processes (such as AD and PD)</p><p>can all disrupt neuroplasticity, leading to impairments in motor skills, affective behaviors,</p><p>and cognitive function. Nevertheless, recent studies have highlighted the brain’s ability</p><p>to compensate for these impairments through processes involving neural reorganization,</p><p>which consists of recruiting other brain regions and neuronal circuits to compensate for the</p><p>damaged ones [362]. Psychological traits, such as personality, motivation, and attention,</p><p>also play a significant role in neuroplasticity mechanisms. For instance, individuals with</p><p>high levels of motivation have been shown to exhibit greater neuroplasticity than those</p><p>with low levels of motivation [363].</p><p>Understanding individual differences in experience-dependent neuroplasticity mecha-</p><p>nisms is critical for developing personalized approaches to improving cognitive function</p><p>and promoting recovery from brain injury or disease. In addition, promoting healthy</p><p>lifestyles, including physical exercise, cognitive and social stimulation, a healthy and bal-</p><p>anced diet, and good sleep hygiene, can go a long way in preventing or halting many</p><p>age-related conditions and promoting overall brain health. By promoting healthy lifestyles</p><p>and optimizing personalized treatment options that regulate and promote neuroplasticity,</p><p>we can effectively harness the power of brain plasticity.</p><p>Author Contributions: P.M., T.M. and E.P.: writing of original manuscript draft. J.G.-M.: funding</p><p>acquisition, conceptualization, review and editing of the final manuscript draft. P.S.B.: funding</p><p>acquisition, conceptualization, supervision, review and editing of the manuscript. All authors have</p><p>read and agreed to the published version of the manuscript.</p><p>Funding: This work was supported by the Fundação de Amparo à Pesquisa e Inovação do Estado de</p><p>Santa Catarina (FAPESC, Florianópolis, SC, Brazil, 2021TR1523). P.S.B. received a Conselho Nacional</p><p>de Desenvolvimento Científico e Tecnológico (CNPq; Brazil) Research Productivity Fellowship. J.G.M.</p><p>acknowledges funding from the University of Victoria (UVic, Victoria, BC, Canada) – São Paulo</p><p>Research Foundation (FAPESP, São Paulo, SP, Brazil) SPRINT partnership (UVic-FAPESP SPRINT</p><p>1/2018).</p><p>Institutional Review Board Statement: Not applicable.</p><p>Informed Consent Statement: Not applicable.</p><p>Data Availability Statement: Not applicable.</p><p>Conflicts of Interest: The authors declare no conflict of interest.</p><p>References</p><p>1. James, W. Habits. In The Principles of Psychology; Henry Holt and Company: New York, NY, USA, 1890; pp. 104–127.</p><p>2. Konorski, J. Conditioned Reflexes and Neuron Organization, Facsimile Reprint of the 1948; Cambridge University Press:</p><p>Cambridge, UK, 1968.</p><p>3. Voss, P.; Thomas, M.E.; Cisneros-Franco, J.M.; de Villers-Sidani, É. Dynamic Brains and the Changing Rules of Neuroplasticity:</p><p>Implications for Learning and Recovery. Front. Psychol. 2017, 8, 1657. [CrossRef]</p><p>4. Draganski, B.; May, A. Training-Induced Structural Changes in the Adult Human Brain. Behav. Brain Res. 2008, 192, 137–142.</p><p>[CrossRef]</p><p>5. Frizzell, T.O.; Phull, E.; Khan, M.; Song, X.; Grajauskas, L.A.; Gawryluk, J.; D’Arcy, R.C.N. Imaging Functional Neuroplasticity in</p><p>Human White Matter Tracts. Brain Struct. Funct. 2022, 227, 381–392. [CrossRef]</p><p>https://doi.org/10.3389/fpsyg.2017.01657</p><p>https://doi.org/10.1016/j.bbr.2008.02.015</p><p>https://doi.org/10.1007/s00429-021-02407-4</p><p>Brain Sci. 2023, 13, 1610 20 of 32</p><p>6. Leuner, B.; Gould, E. Structural Plasticity and Hippocampal Function. Annu. Rev. Psychol. 2010, 61, 111–140. [CrossRef]</p><p>7. Chen, X.; Zhou, X.; Yang, L.; Miao, X.; Lu, D.-H.; Yang, X.-Y.; Zhou, Z.-B.; Kang, W.-B.; Chen, K.-Y.; Zhou, L.-H.; et al. Neonatal</p><p>Exposure to Low-Dose (1.2%) Sevoflurane Increases Rats’ Hippocampal Neurogenesis and Synaptic Plasticity in Later Life.</p><p>Neurotox. Res. 2018, 34, 188–197. [CrossRef]</p><p>8. Garthe, A.; Roeder, I.; Kempermann, G. Mice in an Enriched Environment Learn More Flexibly Because of Adult Hippocampal</p><p>Neurogenesis. Hippocampus 2016, 26, 261–271. [CrossRef]</p><p>9. Kempermann, G.; Song, H.; Gage, F.H. Neurogenesis in the Adult Hippocampus. Cold Spring Harb. Perspect. Biol. 2015, 7, a018812.</p><p>[CrossRef]</p><p>10. Takeuchi, N.; Izumi, S.-I. Rehabilitation with Poststroke Motor Recovery: A Review with a Focus on Neural Plasticity. Stroke Res.</p><p>Treat. 2013, 2013, 128641. [CrossRef]</p><p>11. Aimone, J.B.; Li, Y.; Lee, S.W.; Clemenson, G.D.; Deng, W.; Gage, F.H. Regulation and Function of Adult Neurogenesis: From</p><p>Genes to Cognition. Physiol. Rev. 2014, 94, 991–1026. [CrossRef]</p><p>12. Jurkowski, M.P.; Bettio, L.; Woo, E.K.; Patten, A.; Yau, S.-Y.; Gil-Mohapel, J. Beyond the Hippocampus and the SVZ: Adult</p><p>Neurogenesis Throughout the Brain. Front. Cell. Neurosci. 2020, 14, 576444. [CrossRef]</p><p>13. Zhao, C.; Deng, W.; Gage, F.H. Mechanisms and Functional Implications of Adult Neurogenesis. Cell 2008, 132, 645–660.</p><p>[CrossRef]</p><p>14. Kempermann, G.; Kuhn, H.G.; Gage, F.H. More Hippocampal Neurons in Adult Mice Living in an Enriched Environment. Nature</p><p>1997, 386, 493–495. [CrossRef]</p><p>15. Lazarov, O.; Mattson, M.P.; Peterson, D.A.; Pimplikar, S.W.; van Praag, H. When Neurogenesis Encounters Aging and Disease.</p><p>Trends Neurosci. 2010, 33, 569–579. [CrossRef]</p><p>16. van Praag, H.; Kempermann, G.; Gage, F.H. Running Increases Cell Proliferation and Neurogenesis in the Adult Mouse Dentate</p><p>Gyrus. Nat. Neurosci. 1999, 2, 266–270. [CrossRef]</p><p>17. Lai, C.S.W.; Franke, T.F.; Gan, W.-B. Opposite Effects of Fear Conditioning and Extinction on Dendritic Spine Remodelling. Nature</p><p>2012, 483, 87–91. [CrossRef]</p><p>18. Yang, G.; Pan, F.; Gan, W.-B. Stably Maintained Dendritic Spines Are Associated with Lifelong Memories. Nature</p><p>2009, 462, 920–924. [CrossRef]</p><p>19. Bliss, T.V.P.; Collingridge, G.L. A Synaptic Model of Memory: Long-Term Potentiation in the Hippocampus. Nature 1993, 361, 31–39.</p><p>[CrossRef]</p><p>20. Bear, M.F.; Malenka, R.C. Synaptic Plasticity: LTP and LTD. Curr. Opin. Neurobiol. 1994, 4, 389–399. [CrossRef]</p><p>21. Pinar, C.; Fontaine, C.J.; Triviño-Paredes, J.; Lottenberg, C.P.; Gil-Mohapel, J.; Christie, B.R. Revisiting the Flip Side: Long-Term</p><p>Depression of Synaptic Efficacy in the Hippocampus. Neurosci. Biobehav. Rev. 2017, 80, 394–413. [CrossRef]</p><p>22. Pascual-Leone, A.; Hamilton, R. Chapter 27 The Metamodal Organization of the Brain. Prog. Brain Res. 2001, 134, 427–445.</p><p>[CrossRef]</p><p>23. Knudsen, E.I. Sensitive Periods in the Development of the Brain and Behavior. J. Cogn. Neurosci. 2004, 16, 1412–1425. [CrossRef]</p><p>24. Monday, H.R.; Younts, T.J.; Castillo, P.E. Long-Term Plasticity of Neurotransmitter Release: Emerging Mechanisms and Contribu-</p><p>tions to Brain Function and Disease. Annu. Rev. Neurosci. 2018, 41, 299–322. [CrossRef]</p><p>25. Zuo, Y.; Lin, A.; Chang, P.; Gan, W.-B. Development of Long-Term Dendritic Spine Stability in Diverse Regions of Cerebral Cortex.</p><p>Neuron 2005, 46, 181–189. [CrossRef]</p><p>26. Kourosh-Arami, M.; Hosseini, N.; Komaki, A. Brain Is Modulated by Neuronal Plasticity during Postnatal Development.</p><p>J. Physiol. Sci. 2021, 71, 34. [CrossRef]</p><p>27. Johnston, M.V.; Ishida, A.; Ishida, W.N.; Matsushita, H.B.; Nishimura, A.;</p><p>Tsuji, M. Plasticity and Injury in the Developing Brain.</p><p>Brain Dev. 2009, 31, 1–10. [CrossRef]</p><p>28. Altman, J.; Das, G.D. Autoradiographic and Histological Evidence of Postnatal Hippocampal Neurogenesis in Rats. J. Comp.</p><p>Neurol. 1965, 124, 319–335. [CrossRef]</p><p>29. Ming, G.; Song, H. Adult Neurogenesis in the Mammalian Brain: Significant Answers and Significant Questions. Neuron</p><p>2011, 70, 687–702. [CrossRef]</p><p>30. Budday, S.; Steinmann, P.; Kuhl, E. Physical Biology of Human Brain Development. Front. Cell. Neurosci. 2015, 9, 257. [CrossRef]</p><p>31. Gage, F.H. Structural Plasticity of the Adult Brain. Dialogues Clin. Neurosci. 2004, 6, 135–141. [CrossRef]</p><p>32. Bhatt, S.; Diaz, R.; Trainor, P.A. Signals and Switches in Mammalian Neural Crest Cell Differentiation. Cold Spring Harb. Perspect.</p><p>Biol. 2013, 5, a008326. [CrossRef]</p><p>33. Kosodo, Y.; Röper, K.; Haubensak, W.; Marzesco, A.-M.; Corbeil, D.; Huttner, W.B. Asymmetric Distribution of the Apical Plasma</p><p>Membrane during Neurogenic Divisions of Mammalian Neuroepithelial Cells. EMBO J. 2004, 23, 2314–2324. [CrossRef]</p><p>34. Stiles, J.; Jernigan, T.L. The Basics of Brain Development. Neuropsychol. Rev. 2010, 20, 327–348. [CrossRef]</p><p>35. Gil-Sanz, C.; Franco, S.J.; Martinez-Garay, I.; Espinosa, A.; Harkins-Perry, S.; Müller, U. Cajal-Retzius Cells Instruct Neuronal</p><p>Migration by Coincidence Signaling between Secreted and Contact-Dependent Guidance Cues. Neuron 2013, 79, 461–477.</p><p>[CrossRef]</p><p>36. Patton, M.H.; Blundon, J.A.; Zakharenko, S.S. Rejuvenation of Plasticity in the Brain: Opening the Critical Period. Curr. Opin.</p><p>Neurobiol. 2019, 54, 83–89. [CrossRef]</p><p>https://doi.org/10.1146/annurev.psych.093008.100359</p><p>https://doi.org/10.1007/s12640-018-9877-3</p><p>https://doi.org/10.1002/hipo.22520</p><p>https://doi.org/10.1101/cshperspect.a018812</p><p>https://doi.org/10.1155/2013/128641</p><p>https://doi.org/10.1152/physrev.00004.2014</p><p>https://doi.org/10.3389/fncel.2020.576444</p><p>https://doi.org/10.1016/j.cell.2008.01.033</p><p>https://doi.org/10.1038/386493a0</p><p>https://doi.org/10.1016/j.tins.2010.09.003</p><p>https://doi.org/10.1038/6368</p><p>https://doi.org/10.1038/nature10792</p><p>https://doi.org/10.1038/nature08577</p><p>https://doi.org/10.1038/361031a0</p><p>https://doi.org/10.1016/0959-4388(94)90101-5</p><p>https://doi.org/10.1016/j.neubiorev.2017.06.001</p><p>https://doi.org/10.1016/S0079-6123(01)34028-1</p><p>https://doi.org/10.1162/0898929042304796</p><p>https://doi.org/10.1146/annurev-neuro-080317-062155</p><p>https://doi.org/10.1016/j.neuron.2005.04.001</p><p>https://doi.org/10.1186/s12576-021-00819-9</p><p>https://doi.org/10.1016/j.braindev.2008.03.014</p><p>https://doi.org/10.1002/cne.901240303</p><p>https://doi.org/10.1016/j.neuron.2011.05.001</p><p>https://doi.org/10.3389/fncel.2015.00257</p><p>https://doi.org/10.31887/DCNS.2004.6.2/fgage</p><p>https://doi.org/10.1101/cshperspect.a008326</p><p>https://doi.org/10.1038/sj.emboj.7600223</p><p>https://doi.org/10.1007/s11065-010-9148-4</p><p>https://doi.org/10.1016/j.neuron.2013.06.040</p><p>https://doi.org/10.1016/j.conb.2018.09.003</p><p>Brain Sci. 2023, 13, 1610 21 of 32</p><p>37. Huttenlocher, P.R.; Dabholkar, A.S. Regional Differences in Synaptogenesis in Human Cerebral Cortex. J. Comp. Neurol.</p><p>1997, 387, 167–178. [CrossRef]</p><p>38. Ismail, F.Y.; Fatemi, A.; Johnston, M.V. Cerebral Plasticity: Windows of Opportunity in the Developing Brain. Eur. J. Paediatr.</p><p>Neurol. 2017, 21, 23–48. [CrossRef]</p><p>39. Faust, T.E.; Gunner, G.; Schafer, D.P. Mechanisms Governing Activity-Dependent Synaptic Pruning in the Developing Mammalian</p><p>CNS. Nat. Rev. Neurosci. 2021, 22, 657–673. [CrossRef]</p><p>40. Fares, J.; Bou Diab, Z.; Nabha, S.; Fares, Y. Neurogenesis in the Adult Hippocampus: History, Regulation, and Prospective Roles.</p><p>Int. J. Neurosci. 2019, 129, 598–611. [CrossRef]</p><p>41. Eriksson, P.S.; Perfilieva, E.; Björk-Eriksson, T.; Alborn, A.-M.; Nordborg, C.; Peterson, D.A.; Gage, F.H. Neurogenesis in the Adult</p><p>Human Hippocampus. Nat. Med. 1998, 4, 1313–1317. [CrossRef]</p><p>42. Merkle, F.T.; Tramontin, A.D.; García-Verdugo, J.M.; Alvarez-Buylla, A. Radial Glia Give Rise to Adult Neural Stem Cells in the</p><p>Subventricular Zone. Proc. Natl. Acad. Sci. USA 2004, 101, 17528–17532. [CrossRef]</p><p>43. Kuhn, H.; Dickinson-Anson, H.; Gage, F. Neurogenesis in the Dentate Gyrus of the Adult Rat: Age-Related Decrease of Neuronal</p><p>Progenitor Proliferation. J. Neurosci. 1996, 16, 2027–2033. [CrossRef]</p><p>44. Lois, C.; Alvarez-Buylla, A. Long-Distance Neuronal Migration in the Adult Mammalian Brain. Science 1994, 264, 1145–1148.</p><p>[CrossRef]</p><p>45. Maggi, R.; Zasso, J.; Conti, L. Neurodevelopmental Origin and Adult Neurogenesis of the Neuroendocrine Hypothalamus. Front.</p><p>Cell. Neurosci. 2015, 8, 440. [CrossRef]</p><p>46. Kokoeva, M.V.; Yin, H.; Flier, J.S. Neurogenesis in the Hypothalamus of Adult Mice: Potential Role in Energy Balance. Science</p><p>2005, 310, 679–683. [CrossRef]</p><p>47. Villalba, A.; Götz, M.; Borrell, V. The Regulation of Cortical Neurogenesis. Curr. Top. Dev. Biol. 2021, 142, 1–66. [CrossRef]</p><p>48. Gould, E.; Reeves, A.J.; Graziano, M.S.A.; Gross, C.G. Neurogenesis in the Neocortex of Adult Primates. Science 1999, 286, 548–552.</p><p>[CrossRef]</p><p>49. Roeder, S.S.; Burkardt, P.; Rost, F.; Rode, J.; Brusch, L.; Coras, R.; Englund, E.; Håkansson, K.; Possnert, G.; Salehpour, M.; et al.</p><p>Evidence for Postnatal Neurogenesis in the Human Amygdala. Commun. Biol. 2022, 5, 366. [CrossRef]</p><p>50. Ponti, G.; Peretto, P.; Bonfanti, L. Genesis of Neuronal and Glial Progenitors in the Cerebellar Cortex of Peripuberal and Adult</p><p>Rabbits. PLoS ONE 2008, 3, e2366. [CrossRef]</p><p>51. Andreotti, J.P.; Prazeres, P.H.D.M.; Magno, L.A.V.; Romano-Silva, M.A.; Mintz, A.; Birbrair, A. Neurogenesis in the Postnatal</p><p>Cerebellum after Injury. Int. J. Dev. Neurosci. 2018, 67, 33–36. [CrossRef]</p><p>52. Ernst, A.; Alkass, K.; Bernard, S.; Salehpour, M.; Perl, S.; Tisdale, J.; Possnert, G.; Druid, H.; Frisén, J. Neurogenesis in the Striatum</p><p>of the Adult Human Brain. Cell 2014, 156, 1072–1083. [CrossRef]</p><p>53. Bruel-Jungerman, E.; Rampon, C.; Laroche, S. Adult Hippocampal Neurogenesis, Synaptic Plasticity and Memory: Facts and</p><p>Hypotheses. Rev. Neurosci. 2007, 18, 93–114. [CrossRef]</p><p>54. Koehl, M.; Abrous, D.N. A New Chapter in the Field of Memory: Adult Hippocampal Neurogenesis. Eur. J. Neurosci.</p><p>2011, 33, 1101–1114. [CrossRef]</p><p>55. Samuels, B.A.; Hen, R. Neurogenesis and Affective Disorders. Eur. J. Neurosci. 2011, 33, 1152–1159. [CrossRef]</p><p>56. Åberg, M.A.I.; Åberg, N.D.; Hedbäcker, H.; Oscarsson, J.; Eriksson, P.S. Peripheral Infusion of IGF-I Selectively Induces</p><p>Neurogenesis in the Adult Rat Hippocampus. J. Neurosci. 2000, 20, 2896–2903. [CrossRef]</p><p>57. Zigova, T.; Pencea, V.; Wiegand, S.J.; Luskin, M.B. Intraventricular Administration of BDNF Increases the Number of Newly</p><p>Generated Neurons in the Adult Olfactory Bulb. Mol. Cell. Neurosci. 1998, 11, 234–245. [CrossRef]</p><p>58. Yao, B.; Christian, K.M.; He, C.; Jin, P.; Ming, G.; Song, H. Epigenetic Mechanisms in Neurogenesis. Nat. Rev. Neurosci.</p><p>2016, 17, 537–549. [CrossRef]</p><p>59. Du Preez, A.; Onorato, D.; Eiben, I.; Musaelyan, K.; Egeland, M.; Zunszain, P.A.; Fernandes, C.; Thuret, S.; Pariante, C.M. Chronic</p><p>Stress Followed by Social Isolation Promotes Depressive-like Behaviour, Alters Microglial and Astrocyte Biology and Reduces</p><p>Hippocampal Neurogenesis in Male Mice. Brain. Behav. Immun. 2021, 91, 24–47. [CrossRef]</p><p>60. Malberg, J.E.; Eisch, A.J.; Nestler, E.J.; Duman, R.S. Chronic Antidepressant Treatment Increases Neurogenesis in Adult Rat</p><p>Hippocampus. J. Neurosci. 2000, 20, 9104–9110. [CrossRef]</p><p>61. Manev, H.; Uz, T.; Smalheiser, N.R.; Manev, R. Antidepressants Alter Cell Proliferation in the Adult Brain in Vivo and in Neural</p><p>Cultures in Vitro. Eur. J. Pharmacol. 2001, 411, 67–70. [CrossRef]</p><p>62. Micheli, L.; Ceccarelli, M.; D’Andrea, G.; Tirone, F. Depression and Adult Neurogenesis: Positive Effects of the Antidepressant</p><p>Fluoxetine and of Physical Exercise. Brain Res. Bull. 2018, 143, 181–193. [CrossRef]</p><p>63. Bettio, L.E.B.; Rajendran, L.; Gil-Mohapel, J. The Effects of Aging in the Hippocampus and Cognitive Decline. Neurosci. Biobehav.</p><p>Rev. 2017, 79, 66–86. [CrossRef]</p><p>64. Boldrini, M.; Fulmore, C.A.; Tartt,</p><p>A.N.; Simeon, L.R.; Pavlova, I.; Poposka, V.; Rosoklija, G.B.; Stankov, A.; Arango, V.;</p><p>Dwork, A.J.; et al. Human Hippocampal Neurogenesis Persists throughout Aging. Cell Stem Cell 2018, 22, 589–599.e5. [CrossRef]</p><p>65. Isaev, N.K.; Stelmashook, E.V.; Genrikhs, E.E. Neurogenesis and Brain Aging. Rev. Neurosci. 2019, 30, 573–580. [CrossRef]</p><p>66. Gipson, C.D.; Kupchik, Y.M.; Kalivas, P.W. Rapid, Transient Synaptic Plasticity in Addiction. Neuropharmacology 2014, 76, 276–286.</p><p>[CrossRef]</p><p>https://doi.org/10.1002/(SICI)1096-9861(19971020)387:2%3C167::AID-CNE1%3E3.0.CO;2-Z</p><p>https://doi.org/10.1016/j.ejpn.2016.07.007</p><p>https://doi.org/10.1038/s41583-021-00507-y</p><p>https://doi.org/10.1080/00207454.2018.1545771</p><p>https://doi.org/10.1038/3305</p><p>https://doi.org/10.1073/pnas.0407893101</p><p>https://doi.org/10.1523/JNEUROSCI.16-06-02027.1996</p><p>https://doi.org/10.1126/science.8178174</p><p>https://doi.org/10.3389/fncel.2014.00440</p><p>https://doi.org/10.1126/science.1115360</p><p>https://doi.org/10.1016/bs.ctdb.2020.10.003</p><p>https://doi.org/10.1126/science.286.5439.548</p><p>https://doi.org/10.1038/s42003-022-03299-8</p><p>https://doi.org/10.1371/journal.pone.0002366</p><p>https://doi.org/10.1016/j.ijdevneu.2018.03.002</p><p>https://doi.org/10.1016/j.cell.2014.01.044</p><p>https://doi.org/10.1515/REVNEURO.2007.18.2.93</p><p>https://doi.org/10.1111/j.1460-9568.2011.07609.x</p><p>https://doi.org/10.1111/j.1460-9568.2011.07614.x</p><p>https://doi.org/10.1523/JNEUROSCI.20-08-02896.2000</p><p>https://doi.org/10.1006/mcne.1998.0684</p><p>https://doi.org/10.1038/nrn.2016.70</p><p>https://doi.org/10.1016/j.bbi.2020.07.015</p><p>https://doi.org/10.1523/JNEUROSCI.20-24-09104.2000</p><p>https://doi.org/10.1016/S0014-2999(00)00904-3</p><p>https://doi.org/10.1016/j.brainresbull.2018.09.002</p><p>https://doi.org/10.1016/j.neubiorev.2017.04.030</p><p>https://doi.org/10.1016/j.stem.2018.03.015</p><p>https://doi.org/10.1515/revneuro-2018-0084</p><p>https://doi.org/10.1016/j.neuropharm.2013.04.032</p><p>Brain Sci. 2023, 13, 1610 22 of 32</p><p>67. Bicker, F.; Nardi, L.; Maier, J.; Vasic, V.; Schmeisser, M.J. Criss-crossing Autism Spectrum Disorder and Adult Neurogenesis.</p><p>J. Neurochem. 2021, 159, 452–478. [CrossRef]</p><p>68. Hansel, C. Deregulation of Synaptic Plasticity in Autism. Neurosci. Lett. 2019, 688, 58–61. [CrossRef]</p><p>69. Fries, G.R.; Saldana, V.A.; Finnstein, J.; Rein, T. Molecular Pathways of Major Depressive Disorder Converge on the Synapse. Mol.</p><p>Psychiatry 2023, 28, 284–297. [CrossRef]</p><p>70. Murphy, K.P.S.J.; Carter, R.J.; Lione, L.A.; Mangiarini, L.; Mahal, A.; Bates, G.P.; Dunnett, S.B.; Morton, A.J. Abnormal Synaptic</p><p>Plasticity and Impaired Spatial Cognition in Mice Transgenic for Exon 1 of the Human Huntington’s Disease Mutation. J. Neurosci.</p><p>2000, 20, 5115–5123. [CrossRef]</p><p>71. Babcock, K.R.; Page, J.S.; Fallon, J.R.; Webb, A.E. Adult Hippocampal Neurogenesis in Aging and Alzheimer’s Disease. Stem Cell</p><p>Rep. 2021, 16, 681–693. [CrossRef]</p><p>72. Magee, J.C.; Grienberger, C. Synaptic Plasticity Forms and Functions. Annu. Rev. Neurosci. 2020, 43, 95–117. [CrossRef]</p><p>73. Baltaci, S.B.; Mogulkoc, R.; Baltaci, A.K. Molecular Mechanisms of Early and Late LTP. Neurochem. Res. 2019, 44, 281–296.</p><p>[CrossRef]</p><p>74. Mainberger, F.; Langer, S.; Mall, V.; Jung, N.H. Impaired Synaptic Plasticity in RASopathies: A Mini-Review. J. Neural Transm.</p><p>2016, 123, 1133–1138. [CrossRef]</p><p>75. Bliss, T.V.P.; Lømo, T. Long-Lasting Potentiation of Synaptic Transmission in the Dentate Area of the Anaesthetized Rabbit</p><p>Following Stimulation of the Perforant Path. J. Physiol. 1973, 232, 331–356. [CrossRef]</p><p>76. Gall, C.M.; Le, A.A.; Lynch, G. Sex Differences in Synaptic Plasticity Underlying Learning. J. Neurosci. Res. 2023, 101, 764–782.</p><p>[CrossRef]</p><p>77. Lynch, M.A. Long-Term Potentiation and Memory. Physiol. Rev. 2004, 84, 87–136. [CrossRef]</p><p>78. Johnson, M.H. Functional Brain Development in Humans. Nat. Rev. Neurosci. 2001, 2, 475–483. [CrossRef]</p><p>79. Stiles, J. The Fundamentals of Brain Development: Integrating Nature and Nurture; Harvard University Press: Cambridge, MA, USA, 2008.</p><p>80. Pascual-Leone, A.; Amedi, A.; Fregni, F.; Merabet, L.B. THE PLASTIC HUMAN BRAIN CORTEX. Annu. Rev. Neurosci.</p><p>2005, 28, 377–401. [CrossRef]</p><p>81. Draganski, B.; Gaser, C.; Busch, V.; Schuierer, G.; Bogdahn, U.; May, A. Changes in Grey Matter Induced by Training. Nature</p><p>2004, 427, 311–312. [CrossRef]</p><p>82. Boyke, J.; Driemeyer, J.; Gaser, C.; Büchel, C.; May, A. Training-Induced Brain Structure Changes in the Elderly. J. Neurosci.</p><p>2008, 28, 7031–7035. [CrossRef]</p><p>83. Castrén, E.; Hen, R. Neuronal Plasticity and Antidepressant Actions. Trends Neurosci. 2013, 36, 259–267. [CrossRef]</p><p>84. Kleim, J.A.; Barbay, S.; Cooper, N.R.; Hogg, T.M.; Reidel, C.N.; Remple, M.S.; Nudo, R.J. Motor Learning-Dependent Synaptogen-</p><p>esis Is Localized to Functionally Reorganized Motor Cortex. Neurobiol. Learn. Mem. 2002, 77, 63–77. [CrossRef]</p><p>85. Gil-Mohapel, J.; Boehme, F.; Kainer, L.; Christie, B.R. Hippocampal Cell Loss and Neurogenesis after Fetal Alcohol Exposure:</p><p>Insights from Different Rodent Models. Brain Res. Rev. 2010, 64, 283–303. [CrossRef]</p><p>86. Miller, M.W. Effects of Prenatal Exposure to Alcohol on Postnatal Brain Development. Nat. Rev. Neurosci. 2018, 19, 639–651.</p><p>87. McEwen, B.S.; Bowles, N.P.; Gray, J.D.; Hill, M.N.; Hunter, R.G.; Karatsoreos, I.N.; Nasca, C. Mechanisms of Stress in the Brain.</p><p>Nat. Neurosci. 2015, 18, 1353–1363. [CrossRef]</p><p>88. Catala, M. Embryonic Development of the Central Nervous System. In Encyclopedia of Neuroscience; Springer: Berlin/Heidelberg,</p><p>Germany, 2021; pp. 196–202.</p><p>89. Copp, A.J.; Greene, N.D.E. Neural Tube Defects-Disorders of Neurulation and Related Embryonic Processes. Wiley Interdiscip.</p><p>Rev. Dev. Biol. 2013, 2, 213–227. [CrossRef]</p><p>90. Rakic, P. Specification of Cerebral Cortical Areas. Science 1988, 241, 170–176. [CrossRef]</p><p>91. Lemaire, V.; Koehl, M.; Le Moal, M.; Abrous, D.N. Prenatal Stress Produces Learning Deficits Associated with an Inhibition of</p><p>Neurogenesis in the Hippocampus. Proc. Natl. Acad. Sci. USA 2000, 97, 11032–11037. [CrossRef]</p><p>92. Martinez, S.; Alvarado-Mallart, R.M. Developmental Aspects of the Segmented Brain Hypothesis. Brain Res. Bull. 2002, 57, 301–305.</p><p>93. Perry, V.H.; Hume, D.A.; Gordon, S. Immunohistochemical Localization of Macrophages and Microglia in the Adult and</p><p>Developing Mouse Brain. Neuroscience 1985, 15, 313–326. [CrossRef]</p><p>94. Miller, F.D.; Gauthier-Fisher, A. Home at Last: Neural Stem Cell Niches Defined. Cell Stem Cell 2009, 4, 507–510. [CrossRef]</p><p>95. Paus, T. Growth of White Matter in the Adolescent Brain: Myelin or Axon? Brain Cogn. 2010, 72, 26–35. [CrossRef]</p><p>96. Kuan, C.-Y.; Roth, K.A.; Flavell, R.A.; Rakic, P. Mechanisms of Programmed Cell Death in the Developing Brain. Trends Neurosci.</p><p>2000, 23, 291–297. [CrossRef]</p><p>97. Bourgeois, J. Synaptogenesis, Heterochrony and Epigenesis in the Mammalian Neocortex. Acta Paediatr. 1997, 86, 27–33.</p><p>[CrossRef]</p><p>98. Scott, J.G.; McNaughton, A.J.; Polis, I.; Stein, D.J. Childhood Trauma and COMT Genotype Interact to Increase Hippocampal</p><p>Activation in Adulthood. Mol. Psychiatry 2006, 11, 1066–1067.</p><p>99. Hultman, C.M.; Sparen, P.; Takei, N.; Murray, R.M.; Cnattingius, S.; Geddes, J. Prenatal and Perinatal Risk Factors for Schizophre-</p><p>nia, Affective Psychosis, and Reactive Psychosis of Early Onset: Case-Control Study Prenatal and Perinatal Risk Factors for Early</p><p>Onset Schizophrenia, Affective Psychosis, and Reactive Psychosis. BMJ 1999, 318, 421–426. [CrossRef]</p><p>100. Lacagnina, S. The Developmental Origins of Health and Disease (DOHaD). Am. J. Lifestyle Med. 2020, 14, 47–50. [CrossRef]</p><p>https://doi.org/10.1111/jnc.15501</p><p>https://doi.org/10.1016/j.neulet.2018.02.003</p><p>https://doi.org/10.1038/s41380-022-01806-1</p><p>https://doi.org/10.1523/JNEUROSCI.20-13-05115.2000</p><p>https://doi.org/10.1016/j.stemcr.2021.01.019</p><p>https://doi.org/10.1146/annurev-neuro-090919-022842</p><p>https://doi.org/10.1007/s11064-018-2695-4</p><p>https://doi.org/10.1007/s00702-016-1609-3</p><p>https://doi.org/10.1113/jphysiol.1973.sp010273</p><p>https://doi.org/10.1002/jnr.24844</p><p>https://doi.org/10.1152/physrev.00014.2003</p><p>https://doi.org/10.1038/35081509</p><p>https://doi.org/10.1146/annurev.neuro.27.070203.144216</p><p>https://doi.org/10.1038/427311a</p><p>https://doi.org/10.1523/JNEUROSCI.0742-08.2008</p><p>https://doi.org/10.1016/j.tins.2012.12.010</p><p>https://doi.org/10.1006/nlme.2000.4004</p><p>https://doi.org/10.1016/j.brainresrev.2010.04.011</p><p>https://doi.org/10.1038/nn.4086</p><p>https://doi.org/10.1002/wdev.71</p><p>https://doi.org/10.1126/science.3291116</p><p>https://doi.org/10.1073/pnas.97.20.11032</p><p>https://doi.org/10.1016/0306-4522(85)90215-5</p><p>https://doi.org/10.1016/j.stem.2009.05.008</p><p>https://doi.org/10.1016/j.bandc.2009.06.002</p><p>https://doi.org/10.1016/S0166-2236(00)01581-2</p><p>https://doi.org/10.1111/j.1651-2227.1997.tb18340.x</p><p>https://doi.org/10.1136/bmj.318.7181.421</p><p>https://doi.org/10.1177/1559827619879694</p><p>Brain Sci. 2023, 13, 1610 23 of 32</p><p>101. Wadhwa, P.; Buss, C.; Entringer, S.; Swanson, J. Developmental Origins of Health and Disease: Brief History of the Approach and</p><p>Current Focus on Epigenetic Mechanisms. Semin. Reprod. Med. 2009, 27, 358–368. [CrossRef]</p><p>102. de Juan Romero, C.; Borrell, V. Genetic Maps and Patterns of Cerebral Cortex Folding. Curr. Opin. Cell Biol. 2017, 49, 31–37.</p><p>[CrossRef]</p><p>103. Toso, L.; Poggi, S.H.; Roberson, R.; Woodard, J.; Park, J.; Abebe, D.; Spong, C.Y. Prevention of Alcohol-Induced Learning Deficits</p><p>in Fetal Alcohol Syndrome Mediated through NMDA and GABA Receptors. Am. J. Obstet. Gynecol. 2006, 194, 681–686. [CrossRef]</p><p>104. Olney, J.W.; Wozniak, D.F.; Jevtovic-Todorovic, V.; Farber, N.B.; Bittigau, P.; Ikonomidou, C. Glutamate and GABA Receptor</p><p>Dysfunction in the Fetal Alcohol Syndrome. Neurotox. Res. 2002, 4, 315–325. [CrossRef]</p><p>105. Buss, C.; Entringer, S.; Wadhwa, P.D. Fetal Programming of Brain Development: Intrauterine Stress and Susceptibility to</p><p>Psychopathology. Sci. Signal. 2012, 5, pt7. [CrossRef]</p><p>106. Winkler, I.; Tervaniemi, M.; Schröger, E. Pre-Attentive Auditory Processing in the Human Brainstem and Cortex. Neurosci.</p><p>Biobehav. Rev. 2021, 122, 332–351.</p><p>107. Partanen, E.; Kujala, T.; Tervaniemi, M.; Huotilainen, M. Prenatal Music Exposure Induces Long-Term Neural Effects. PLoS ONE</p><p>2013, 8, e78946. [CrossRef]</p><p>108. Golub, M.D.; Sadtler, P.T.; Oby, E.R.; Quick, K.M.; Ryu, S.I.; Tyler-Kabara, E.C.; Batista, A.P.; Chase, S.M.; Yu, B.M. Learning by</p><p>Neural Reassociation. Nat. Neurosci. 2018, 21, 607–616. [CrossRef]</p><p>109. Giedd, J.N. Structural Magnetic Resonance Imaging of the Adolescent Brain. Ann. N. Y. Acad. Sci. 2004, 1021, 77–85. [CrossRef]</p><p>110. Bishop, D.V.M.; Snowling, M.J. Developmental Dyslexia and Specific Language Impairment: Same or Different? Psychol. Bull.</p><p>2004, 130, 858–886. [CrossRef]</p><p>111. Kuhl, P.K. Brain Mechanisms in Early Language Acquisition. Neuron 2010, 67, 713–727. [CrossRef]</p><p>112. Werker, J.F.; Hensch, T.K. Critical Periods in Speech Perception: New Directions. Annu. Rev. Psychol. 2015, 66, 173–196. [CrossRef]</p><p>113. Diamond, A. Executive Functions. Annu. Rev. Psychol. 2013, 64, 135–168. [CrossRef]</p><p>114. Hackman, D.A.; Farah, M.J. Socioeconomic Status and the Developing Brain. Trends Cogn. Sci. 2009, 13, 65–73. [CrossRef]</p><p>115. Bowlby, J. Attachment and Loss: Retrospect and Prospect. Am. J. Orthopsychiatry 1982, 52, 664–678. [CrossRef]</p><p>116. World Health Organization. Global Status Report on Violence Prevention 2014; World Health Organization: Geneva, Switzerland, 2014.</p><p>117. World Health Organization. Global Status Report on Preventing Violence against Children; World Health Organization: Geneva,</p><p>Switzerland, 2020.</p><p>118. Russell, G.; Lightman, S. The Human Stress Response. Nat. Rev. Endocrinol. 2019, 15, 525–534. [CrossRef]</p><p>119. McEwen, B.S. Neurobiological and Systemic Effects of Chronic Stress. Chronic Stress 2017, 1, 2470547017692328. [CrossRef]</p><p>120. Oomen, C.A.; Mayer, J.L.; De Kloet, E.R.; Joëls, M.; Lucassen, P.J. Brief Treatment with the Glucocorticoid Receptor Antagonist</p><p>Mifepristone Normalizes the Reduction in Neurogenesis after Chronic Stress. Eur. J. Neurosci. 2007, 26, 3395–3401. [CrossRef]</p><p>121. Liu, R.; Yang, X.D.; Liao, X.M.; Xie, X.M.; Su, Y.A.; Li, J.T.; Wang, X.D.; Si, T.M. Early Postnatal Stress Suppresses the Developmental</p><p>Trajectory of Hippocampal Pyramidal Neurons: The Role of CRHR1. Brain Struct. Funct. 2016, 221, 4525–4536. [CrossRef]</p><p>122. Fabricius, K.; Wörtwein, G.; Pakkenberg, B. The Impact of Maternal Separation on Adult Mouse Behaviour and on the Total</p><p>Neuron Number in the Mouse Hippocampus. Brain Struct. Funct. 2008, 212, 403–416. [CrossRef]</p><p>123. Chocyk, A.; Majcher-Maoelanka, I.; Dudys, D.; Przyborowska, A.; Wêdzony, K. Impact of Early-Life Stress on the Medial Prefrontal</p><p>Cortex Functions-a Search for the Pathomechanisms of Anxiety and Mood Disorders. Pharmacol. Rep. 2013, 65, 1462–1470.</p><p>[CrossRef]</p><p>124. Herzberg, M.P.; Gunnar, M.R. Early Life Stress and Brain Function: Activity and Connectivity Associated with Processing</p><p>Emotion and Reward. Neuroimage 2020, 209, 116493. [CrossRef]</p><p>125. McEwen, B.S.; Gianaros, P.J. Central Role of the Brain in Stress and Adaptation: Links to Socioeconomic Status, Health, and</p><p>Disease. Ann. N. Y. Acad. Sci. 2010, 1186, 190–222. [CrossRef]</p><p>126. Hair, N.L.; Hanson, J.L.; Wolfe, B.L.; Pollak, S.D. Association of Child Poverty, Brain Development, and Academic Achievement.</p><p>JAMA Pediatr. 2015, 169, 822–829. [CrossRef]</p><p>127. Enoch, M.-A. The Role of Early Life Stress as a Predictor for Alcohol and Drug Dependence. Psychopharmacology 2011, 214, 17–31.</p><p>[CrossRef]</p><p>128. Rothman, E.F.; Edwards, E.M.; Heeren, T.; Hingson, R.W. Adverse Childhood Experiences Predict Earlier Age of Drinking Onset:</p><p>Results from a Representative US Sample of Current or Former Drinkers. Pediatrics 2008, 122, 298–304. [CrossRef]</p><p>129. Pilowsky, D.J.; Keyes, K.M.; Hasin, D.S. Adverse Childhood Events and Lifetime Alcohol Dependence. Am. J. Public Health</p><p>2009, 99, 258–263. [CrossRef]</p><p>130. Hyman, S.M.; Paliwal, P.; Chaplin, T.M.; Mazure, C.M.; Rounsaville, B.J.; Sinha, R. Severity of Childhood Trauma Is Predictive of</p><p>Cocaine Relapse Outcomes in Women but Not Men. Drug Alcohol Depend. 2008, 92, 208–216. [CrossRef]</p><p>131. Blakemore, S.-J.; Choudhury, S. Development of the Adolescent Brain: Implications for Executive Function and Social Cognition.</p><p>J. Child Psychol. Psychiatry 2006, 47, 296–312. [CrossRef]</p><p>132. Fareri, D.S.; Gabard-Durnam, L.J.; Goff, B.; Tottenham, N. Developmental Trajectories of Amygdala-Prefrontal Morphology and</p><p>Functioning in Adolescents: A Longitudinal Study. Neuroimage 2015, 118, 239–247.</p><p>133. Gee, D.G.; Humphreys, K.L.; Flannery, J.; Goff, B.; Telzer, E.H.; Shapiro, M.; Hare, T.A.; Bookheimer, S.Y.; Tottenham, N. A Devel-</p><p>opmental Shift from Positive to Negative Connectivity in Human Amygdala–Prefrontal Circuitry. J. Neurosci. 2013, 33, 4584–4593.</p><p>[CrossRef]</p><p>https://doi.org/10.1055/s-0029-1237424</p><p>https://doi.org/10.1016/j.ceb.2017.11.009</p><p>https://doi.org/10.1016/j.ajog.2006.01.003</p><p>https://doi.org/10.1080/1029842021000010875</p><p>https://doi.org/10.1126/scisignal.2003406</p><p>https://doi.org/10.1371/journal.pone.0078946</p><p>https://doi.org/10.1038/s41593-018-0095-3</p><p>https://doi.org/10.1196/annals.1308.009</p><p>https://doi.org/10.1037/0033-2909.130.6.858</p><p>https://doi.org/10.1016/j.neuron.2010.08.038</p><p>https://doi.org/10.1146/annurev-psych-010814-015104</p><p>https://doi.org/10.1146/annurev-psych-113011-143750</p><p>https://doi.org/10.1016/j.tics.2008.11.003</p><p>https://doi.org/10.1111/j.1939-0025.1982.tb01456.x</p><p>https://doi.org/10.1038/s41574-019-0228-0</p><p>https://doi.org/10.1177/2470547017692328</p><p>https://doi.org/10.1111/j.1460-9568.2007.05972.x</p><p>https://doi.org/10.1007/s00429-016-1182-4</p><p>https://doi.org/10.1007/s00429-007-0169-6</p><p>https://doi.org/10.1016/S1734-1140(13)71506-8</p><p>https://doi.org/10.1016/j.neuroimage.2019.116493</p><p>https://doi.org/10.1111/j.1749-6632.2009.05331.x</p><p>https://doi.org/10.1001/jamapediatrics.2015.1475</p><p>https://doi.org/10.1007/s00213-010-1916-6</p><p>https://doi.org/10.1542/peds.2007-3412</p><p>https://doi.org/10.2105/AJPH.2008.139006</p><p>https://doi.org/10.1016/j.drugalcdep.2007.08.006</p><p>https://doi.org/10.1111/j.1469-7610.2006.01611.x</p><p>https://doi.org/10.1523/JNEUROSCI.3446-12.2013</p><p>Brain Sci. 2023, 13, 1610 24 of 32</p><p>134. Blakemore, S.-J. Development of the Social Brain</p><p>in Adolescence. J. R. Soc. Med. 2012, 105, 111–116. [CrossRef]</p><p>135. Tottenham, N.; Galván, A. Stress and the Adolescent Brain. Neurosci. Biobehav. Rev. 2016, 70, 217–227. [CrossRef]</p><p>136. Sowell, E.R.; Peterson, B.S.; Thompson, P.M.; Welcome, S.E.; Henkenius, A.L.; Toga, A.W. Mapping Cortical Change across the</p><p>Human Life Span. Nat. Neurosci. 2003, 6, 309–315. [CrossRef]</p><p>137. Evans, J.; Sumners, C.; Moore, J.; Huentelman, M.J.; Deng, J.; Gelband, C.H.; Shaw, G. Characterization of Mitotic Neurons</p><p>Derived From Adult Rat Hypothalamus and Brain Stem. J. Neurophysiol. 2002, 87, 1076–1085. [CrossRef]</p><p>138. Parent, A.; Côté, P.Y.; Lavoie, B. Chemical Anatomy of Primate Basal Ganglia. Prog. Neurobiol. 1995, 46, 131–197. [CrossRef]</p><p>139. Suzuki, S.O.; Goldman, J.E. Multiple Cell Populations in the Early Postnatal Subventricular Zone Take Distinct Migratory</p><p>Pathways: A Dynamic Study of Glial and Neuronal Progenitor Migration. J. Neurosci. 2003, 23, 4240–4250. [CrossRef]</p><p>140. Shapiro, L.A.; Ng, K.; Zhou, Q.-Y.; Ribak, C.E. Subventricular Zone-Derived, Newly Generated Neurons Populate Several</p><p>Olfactory and Limbic Forebrain Regions. Epilepsy Behav. 2009, 14, 74–80. [CrossRef]</p><p>141. Zhao, M.; Momma, S.; Delfani, K.; Carlén, M.; Cassidy, R.M.; Johansson, C.B.; Brismar, H.; Shupliakov, O.; Frisén, J.; Janson, A.M.</p><p>Evidence for Neurogenesis in the Adult Mammalian Substantia Nigra. Proc. Natl. Acad. Sci. USA 2003, 100, 7925–7930. [CrossRef]</p><p>142. Magavi, S.S.; Leavitt, B.R.; Macklis, J.D. Induction of Neurogenesis in the Neocortex of Adult Mice. Nature 2000, 405, 951–955.</p><p>[CrossRef]</p><p>143. Bernier, P.J.; Bédard, A.; Vinet, J.; Lévesque, M.; Parent, A. Newly Generated Neurons in the Amygdala and Adjoining Cortex of</p><p>Adult Primates. Proc. Natl. Acad. Sci. USA 2002, 99, 11464–11469. [CrossRef]</p><p>144. Cao, Q.-L.; Howard, R.M.; Dennison, J.B.; Whittemore, S.R. Differentiation of Engrafted Neuronal-Restricted Precursor Cells Is</p><p>Inhibited in the Traumatically Injured Spinal Cord. Exp. Neurol. 2002, 177, 349–359. [CrossRef]</p><p>145. Dayer, A.G.; Cleaver, K.M.; Abouantoun, T.; Cameron, H.A. New GABAergic Interneurons in the Adult Neocortex and Striatum</p><p>Are Generated from Different Precursors. J. Cell Biol. 2005, 168, 415–427. [CrossRef]</p><p>146. Inta, D.; Alfonso, J.; von Engelhardt, J.; Kreuzberg, M.M.; Meyer, A.H.; van Hooft, J.A.; Monyer, H. Neurogenesis and Widespread</p><p>Forebrain Migration of Distinct GABAergic Neurons from the Postnatal Subventricular Zone. Proc. Natl. Acad. Sci. USA</p><p>2008, 105, 20994–20999. [CrossRef]</p><p>147. Florio, M.; Huttner, W.B. Neural Progenitors, Neurogenesis and the Evolution of the Neocortex. Development 2014, 141, 2182–2194.</p><p>[CrossRef]</p><p>148. Zecevic, N.; Rakic, P. Development of Layer I Neurons in the Primate Cerebral Cortex. J. Neurosci. 2001, 21, 5607–5619. [CrossRef]</p><p>149. Jhaveri, D.J.; Tedoldi, A.; Hunt, S.; Sullivan, R.; Watts, N.R.; Power, J.M.; Bartlett, P.F.; Sah, P. Evidence for Newly Generated</p><p>Interneurons in the Basolateral Amygdala of Adult Mice. Mol. Psychiatry 2018, 23, 521–532. [CrossRef]</p><p>150. Lee, D.A.; Bedont, J.L.; Pak, T.; Wang, H.; Song, J.; Miranda-Angulo, A.; Takiar, V.; Charubhumi, V.; Balordi, F.;</p><p>Takebayashi, H.; et al. Tanycytes of the Hypothalamic Median Eminence Form a Diet-Responsive Neurogenic Niche. Nat.</p><p>Neurosci. 2012, 15, 700–702. [CrossRef]</p><p>151. Li, J.; Tang, Y.; Cai, D. IKKβ/NF-KB Disrupts Adult Hypothalamic Neural Stem Cells to Mediate a Neurodegenerative Mechanism</p><p>of Dietary Obesity and Pre-Diabetes. Nat. Cell Biol. 2012, 14, 999–1012. [CrossRef]</p><p>152. Bernstein, P.L.; Zuo, M.; Cheng, M.-F. Social Condition Affects the Courtship Behavior of Male Ring Doves with Posterior Medial</p><p>Hypothalamic Lesions. Behav. Neural Biol. 1993, 59, 120–125. [CrossRef]</p><p>153. Fowler, C.D.; Liu, Y.; Ouimet, C.; Wang, Z. The Effects of Social Environment on Adult Neurogenesis in the Female Prairie Vole.</p><p>J. Neurobiol. 2002, 51, 115–128. [CrossRef]</p><p>154. Cheng, M.-F.; Peng, J.-P.; Chen, G.; Gardner, J.P.; Bonder, E.M. Functional Restoration of Acoustic Units and Adult-Generated</p><p>Neurons after Hypothalamic Lesion. J. Neurobiol. 2004, 60, 197–213. [CrossRef]</p><p>155. Zhang, Y.; Kim, M.S.; Jia, B.; Yan, J.; Zuniga-Hertz, J.P.; Han, C.; Cai, D. Hypothalamic Stem Cells Control Ageing Speed Partly</p><p>through Exosomal MiRNAs. Nature 2017, 548, 52–57. [CrossRef]</p><p>156. Plakkot, B.; Di Agostino, A.; Subramanian, M. Implications of Hypothalamic Neural Stem Cells on Aging and Obesity-Associated</p><p>Cardiovascular Diseases. Cells 2023, 12, 769. [CrossRef]</p><p>157. Duman, R.S.; Nakagawa, S.; Malberg, J.E. Regulation of Adult Neurogenesis by Antidepressant Treatment. Neuropsychopharmacology</p><p>2001, 25, 836–844. [CrossRef]</p><p>158. Santarelli, L.; Saxe, M.; Gross, C.; Surget, A.; Battaglia, F.; Dulawa, S.; Weisstaub, N.; Lee, J.; Duman, R.; Arancio, O.; et al.</p><p>Requirement of Hippocampal Neurogenesis for the Behavioral Effects of Antidepressants. Science 2003, 301, 805–809. [CrossRef]</p><p>159. Castello, N.A.; Nguyen, M.H.; Tran, J.D.; Cheng, D.; Green, K.N.; LaFerla, F.M. Ketamine-Induced Neurogenesis in Prefrontal</p><p>Cortex Contributes to Antidepressant-like Effects. Proc. Natl. Acad. Sci. USA 2020, 117, 28794–28804.</p><p>160. Gil-Mohapel, J.; Simpson, J.M.; Christie, B.R. Modulation of Adult Neurogenesis by Physical Exercise and Environmental</p><p>Enrichment: Insights for the Treatment of Neurological Disorders. In Adult Neurogenesis and CNS Diseases; Transworld Research</p><p>Network: Kerala, India, 2010; pp. 125–150.</p><p>161. van Praag, H.; Christie, B.R.; Sejnowski, T.J.; Gage, F.H. Running Enhances Neurogenesis, Learning, and Long-Term Potentiation</p><p>in Mice. Proc. Natl. Acad. Sci. USA 1999, 96, 13427–13431. [CrossRef]</p><p>162. Yau, S.; Gil-Mohapel, J.; Christie, B.R.; So, K. Physical Exercise-Induced Adult Neurogenesis: A Good Strategy to Prevent</p><p>Cognitive Decline in Neurodegenerative Diseases? Biomed Res. Int. 2014, 2014, 403120. [CrossRef]</p><p>163. Christian, P.; Stewart, C.P.; Omumbo, J.A. Impact of Maternal and Child Nutrition on Health. Annu. Rev. Nutr. 2019, 39, 213–231.</p><p>https://doi.org/10.1258/jrsm.2011.110221</p><p>https://doi.org/10.1016/j.neubiorev.2016.07.030</p><p>https://doi.org/10.1038/nn1008</p><p>https://doi.org/10.1152/jn.00088.2001</p><p>https://doi.org/10.1016/0301-0082(95)80010-6</p><p>https://doi.org/10.1523/JNEUROSCI.23-10-04240.2003</p><p>https://doi.org/10.1016/j.yebeh.2008.09.011</p><p>https://doi.org/10.1073/pnas.1131955100</p><p>https://doi.org/10.1038/35016083</p><p>https://doi.org/10.1073/pnas.172403999</p><p>https://doi.org/10.1006/exnr.2002.7981</p><p>https://doi.org/10.1083/jcb.200407053</p><p>https://doi.org/10.1073/pnas.0807059105</p><p>https://doi.org/10.1242/dev.090571</p><p>https://doi.org/10.1523/JNEUROSCI.21-15-05607.2001</p><p>https://doi.org/10.1038/mp.2017.134</p><p>https://doi.org/10.1038/nn.3079</p><p>https://doi.org/10.1038/ncb2562</p><p>https://doi.org/10.1016/0163-1047(93)90834-5</p><p>https://doi.org/10.1002/neu.10042</p><p>https://doi.org/10.1002/neu.20014</p><p>https://doi.org/10.1038/nature23282</p><p>https://doi.org/10.3390/cells12050769</p><p>https://doi.org/10.1016/S0893-133X(01)00358-X</p><p>https://doi.org/10.1126/science.1083328</p><p>https://doi.org/10.1073/pnas.96.23.13427</p><p>https://doi.org/10.1155/2014/403120</p><p>Brain Sci. 2023, 13, 1610 25 of 32</p><p>164. Edlow, A.G. Maternal Obesity and Neurodevelopmental and Psychiatric Disorders in Offspring. Prenat. Diagn. 2017, 37, 95–110.</p><p>[CrossRef]</p><p>165. Rivera, H.M.; Christiansen, K.J.; Sullivan, E.L. The Role of Maternal Obesity in the Risk of Neuropsychiatric Disorders. Front.</p><p>Neurosci. 2015, 9, 194. [CrossRef]</p><p>166. Dearden, L.; Ozanne, S.E. Early Life Origins of Metabolic Disease: Developmental Programming of Hypothalamic Pathways</p><p>Controlling Energy Homeostasis. Front. Neuroendocrinol. 2015, 39, 3–16. [CrossRef]</p><p>167. Bae-Gartz, I.; Janoschek, R.; Breuer, S.; Schmitz, L.; Hoffmann, T.; Ferrari, N.; Branik, L.; Oberthuer, A.; Kloppe, C.-S.;</p><p>Appel, S.; et al. Maternal Obesity Alters Neurotrophin-Associated MAPK Signaling in the Hypothalamus of Male Mouse Off-</p><p>spring. Front. Neurosci. 2019, 13, 962. [CrossRef]</p><p>168. Schmitz, L.; Kuglin, R.; Bae-Gartz, I.; Janoschek, R.; Appel, S.; Mesaros, A.; Jakovcevski,</p><p>I.; Vohlen, C.; Handwerk, M.;</p><p>Ensenauer, R.; et al. Hippocampal Insulin Resistance Links Maternal Obesity with Impaired Neuronal Plasticity in Adult Off-</p><p>spring. Psychoneuroendocrinology 2018, 89, 46–52. [CrossRef]</p><p>169. Van Dam, J.M.; Garrett, A.J.; Schneider, L.A.; Hodyl, N.A.; Goldsworthy, M.R.; Coat, S.; Rowan, J.A.; Hague, W.M.; Pitcher, J.B.</p><p>Reduced Cortical Excitability, Neuroplasticity, and Salivary Cortisol in 11–13-Year-Old Children Born to Women with Gestational</p><p>Diabetes Mellitus. EBioMedicine 2018, 31, 143–149. [CrossRef]</p><p>170. Yu, L.; Zhong, X.; He, Y.; Shi, Y. Butyrate, but Not Propionate, Reverses Maternal Diet-Induced Neurocognitive Deficits in</p><p>Offspring. Pharmacol. Res. 2020, 160, 105082. [CrossRef]</p><p>171. Mendes-da-Silva, C.; Lemes, S.F.; Baliani, T.d.S.; Versutti, M.D.; Torsoni, M.A. Increased Expression of Hes5 Protein in Notch</p><p>Signaling Pathway in the Hippocampus of Mice Offspring of Dams Fed a High-fat Diet during Pregnancy and Suckling. Int. J.</p><p>Dev. Neurosci. 2015, 40, 35–42. [CrossRef]</p><p>172. Ge, Q.; Hu, X.; Ma, N.; Sun, M.; Zhang, L.; Cai, Z.; Tan, R.; Lu, H. Maternal High-salt Diet during Pregnancy Impairs Synaptic</p><p>Plasticity and Memory in Offspring. FASEB J. 2021, 35, e21244. [CrossRef]</p><p>173. Wasserman, G.A.; Liu, X.; Parvez, F.; Ahsan, H.; Factor-Litvak, P.; van Geen, A.; Slavkovich, V.; Lolacono, N.J.; Cheng, Z.;</p><p>Hussain, I.; et al. Water Arsenic Exposure and Children’s Intellectual Function in Araihazar, Bangladesh. Environ. Health Perspect.</p><p>2004, 112, 1329–1333. [CrossRef]</p><p>174. May, P.A.; Chambers, C.D.; Kalberg, W.O.; Zellner, J.; Feldman, H.; Buckley, D.; Kopald, D.; Hasken, J.M.; Xu, R.; Honerkamp-</p><p>Smith, G.; et al. Prevalence of Fetal Alcohol Spectrum Disorders in 4 US Communities. JAMA 2018, 319, 474. [CrossRef]</p><p>175. Brancato, A.; Castelli, V.; Lavanco, G.; Marino, R.A.M.; Cannizzaro, C. In Utero ∆9-Tetrahydrocannabinol Exposure Confers</p><p>Vulnerability towards Cognitive Impairments and Alcohol Drinking in the Adolescent Offspring: Is There a Role for Neuropeptide</p><p>Y? J. Psychopharmacol. 2020, 34, 663–679. [CrossRef]</p><p>176. Volkow, N.D.; Jones, E.B. Have We Entered a New Era of Prenatal Cocaine Exposure? Neuropharmacology 2019, 151, 117–124.</p><p>177. Grandjean, P.; Landrigan, P.J. Neurobehavioural Effects of Developmental Toxicity. Lancet Neurol. 2014, 13, 330–338. [CrossRef]</p><p>178. Dolk, H.; Lopes dos Santos, M.; Soares, P. Congenital Rubella Syndrome in the Era of Global Elimination of Measles and Rubella.</p><p>Lancet 2017, 5, e287–e288.</p><p>179. Mullins, E.; Evans, D.; Viner, R.M.; O’Brien, P. The Impact of the COVID-19 Pandemic on Perinatal Mental Health. J. Perinat. Med.</p><p>2021, 49, 327–329.</p><p>180. Kancherla, V.; Razzaghi, H.; Okoniewski, K.C.; Wong, K.K.; Salihu, H.M.; Kirby, R.S.; Anjohrin, S.B. Increased Proportion of</p><p>Reported Congenital Anomalies during the COVID-19 Pandemic. Birth Defects Res. 2021, 113, 1079–1087.</p><p>181. Heidarzadeh, M.; Taheri, M.; Mazaheripour, Z.; Abbasi-Khameneh, F. The Incidence of Congenital Anomalies in Newborns</p><p>before and during the Covid-19 Pandemic. Ital. J. Pediatr. 2022, 48, 174. [CrossRef]</p><p>182. Black, M.M.; Walker, S.P.; Fernald, L.C.H.; Andersen, C.T.; DiGirolamo, A.M.; Lu, C.; McCoy, D.C.; Fink, G.; Shawar, Y.R.;</p><p>Shiffman, J.; et al. Early Childhood Development Coming of Age: Science through the Life Course. Lancet 2017, 389, 77–90.</p><p>[CrossRef]</p><p>183. Vinuesa, A.; Bentivegna, M.; Calfa, G.; Filipello, F.; Pomilio, C.; Bonaventura, M.M.; Lux-Lantos, V.; Matzkin, M.E.; Gregosa, A.;</p><p>Presa, J.; et al. Early Exposure to a High-Fat Diet Impacts on Hippocampal Plasticity: Implication of Microglia-Derived</p><p>Exosome-like Extracellular Vesicles. Mol. Neurobiol. 2019, 56, 5075–5094. [CrossRef]</p><p>184. Arcego, D.M.; Olivo, L.B.; Moraes, R.O.; Garcia, E.d.S.; Silveira, A.C.; Krolow, R.; Couto-Pereira, N.d.S.; Lampert, C.;</p><p>Toniazzo, A.P.; Nicola, F.d.C.; et al. Neurometabolic Effects of Sweetened Solution Intake during Adolescence Related to</p><p>Depressive-like Phenotype in Rats. Nutrition 2020, 75–76, 110770. [CrossRef]</p><p>185. Lee, H.; Park, H.; Ha, E.; Hong, Y.-C.; Ha, M.; Park, H.; Kim, B.-N.; Lee, B.; Lee, S.-J.; Lee, K.Y.; et al. Effect of Breastfeeding</p><p>Duration on Cognitive Development in Infants: 3-Year Follow-up Study. J. Korean Med. Sci. 2016, 31, 579. [CrossRef]</p><p>186. Abbink, M.R.; Schipper, L.; Naninck, E.F.G.; de Vos, C.M.H.; Meier, R.; van der Beek, E.M.; Lucassen, P.J.; Korosi, A. The Effects of</p><p>Early Life Stress, Postnatal Diet Modulation, and Long-Term Western-Style Diet on Later-Life Metabolic and Cognitive Outcomes.</p><p>Nutrients 2020, 12, 570. [CrossRef]</p><p>187. Ruel, M.T.; Alderman, H. Nutrition-Sensitive Interventions and Programmes: How Can They Help to Accelerate Progress in</p><p>Improving Maternal and Child Nutrition? Lancet 2013, 382, 536–551. [CrossRef]</p><p>188. Victora, C.G.; Adair, L.; Fall, C.; Hallal, P.C.; Martorell, R.; Richter, L.; Sachdev, H.S. Maternal and Child Undernutrition:</p><p>Consequences for Adult Health and Human Capital. Lancet 2008, 371, 340–357. [CrossRef]</p><p>https://doi.org/10.1002/pd.4932</p><p>https://doi.org/10.3389/fnins.2015.00194</p><p>https://doi.org/10.1016/j.yfrne.2015.08.001</p><p>https://doi.org/10.3389/fnins.2019.00962</p><p>https://doi.org/10.1016/j.psyneuen.2017.12.023</p><p>https://doi.org/10.1016/j.ebiom.2018.04.011</p><p>https://doi.org/10.1016/j.phrs.2020.105082</p><p>https://doi.org/10.1016/j.ijdevneu.2014.11.005</p><p>https://doi.org/10.1096/fj.202001890R</p><p>https://doi.org/10.1289/ehp.6964</p><p>https://doi.org/10.1001/jama.2017.21896</p><p>https://doi.org/10.1177/0269881120916135</p><p>https://doi.org/10.1016/S1474-4422(13)70278-3</p><p>https://doi.org/10.1186/s13052-022-01368-6</p><p>https://doi.org/10.1016/S0140-6736(16)31389-7</p><p>https://doi.org/10.1007/s12035-018-1435-8</p><p>https://doi.org/10.1016/j.nut.2020.110770</p><p>https://doi.org/10.3346/jkms.2016.31.4.579</p><p>https://doi.org/10.3390/nu12020570</p><p>https://doi.org/10.1016/S0140-6736(13)60843-0</p><p>https://doi.org/10.1016/S0140-6736(07)61692-4</p><p>Brain Sci. 2023, 13, 1610 26 of 32</p><p>189. Prado, E.L.; Abbeddou, S.; Adu-Afarwuah, S.; Arimond, M.; Ashorn, P.; Ashorn, U.; Brown, K.H.; Hess, S.Y.; Lartey, A.;</p><p>Maleta, K.; et al. Linear Growth and Child Development in Burkina Faso, Ghana, and Malawi. Pediatrics 2016, 138, e20154698.</p><p>[CrossRef]</p><p>190. Granat-McGregor, S.M.; Powell, C.A.; Walker, S.P. Nutritional Supplementation, Cognitive and Scholastic Outcomes of Kenyan</p><p>Children. J. Nutr. 2007, 137, 2694–2699.</p><p>191. Shonkoff, J.P.; Garner, A.S.; Siegel, B.S.; Dobbins, M.I.; Earls, M.F.; Garner, A.S.; McGuinn, L.; Pascoe, J.; Wood, D.L. The Lifelong</p><p>Effects of Early Childhood Adversity and Toxic Stress. Pediatrics 2012, 129, e232–e246. [CrossRef]</p><p>192. Campbell, F.; Conti, G.; Heckman, J.J.; Moon, S.H.; Pinto, R.; Pungello, E.; Pan, Y. Early Childhood Investments Substantially</p><p>Boost Adult Health. Science 2014, 343, 1478–1485. [CrossRef]</p><p>193. Yoshikawa, H.; Weiland, C.; Brooks-Gunn, J.; Burchinal, M.R.; Espinosa, L.M.; Gormley, W.T.; Ludwig, J.; Magnuson, K.A.;</p><p>Phillips, D.; Zaslow, M.J. Investing in Our Future: The Evidence Base on Preschool Education; Society for Research in Child Develop-</p><p>ment, Foundation for Child Development: Washington, DC, USA, 2013.</p><p>194. Sheridan, M.A.; Fox, N.A.; Zeanah, C.H.; McLaughlin, K.A.; Nelson, C.A. Variation in Neural Development as a Result of</p><p>Exposure to Institutionalization Early in Childhood. Proc. Natl. Acad. Sci. USA 2012, 109, 12927–12932. [CrossRef]</p><p>195. Nelson, C.A.; Thomas, K.M.; de Haan, M. Neural Bases of Cognitive Development. In Handbook of Child Psychology; John</p><p>Wiley & Sons, Inc.: Hoboken, NJ, USA, 2007; pp. 1–56.</p><p>196. Kempermann, G. Environmental Enrichment, New Neurons and the Neurobiology of Individuality. Nat. Rev. Neurosci. 2019, 20, 235–245.</p><p>[CrossRef]</p><p>197. Bergami, M.; Masserdotti, G.; Temprana, S.G.; Motori, E.; Eriksson, T.M.; Göbel, J.; Yang, S.M.; Conzelmann, K.-K.; Schinder, A.F.;</p><p>Götz, M.; et al. A Critical Period for Experience-Dependent Remodeling of Adult-Born Neuron Connectivity. Neuron</p><p>2015, 85, 710–717. [CrossRef]</p><p>198. Barichello, T.; Fagundes, G.D.; Generoso, J.S.; Dagostin, C.S.; Simões, L.R.;</p><p>Vilela, M.C.; Comim, C.M.; Petronilho, F.; Quevedo, J.;</p><p>Teixeira, A.L. Environmental Enrichment Restores Cognitive Deficits Induced by Experimental Childhood Meningitis. Rev. Bras.</p><p>Psiquiatr. 2014, 36, 322–329. [CrossRef]</p><p>199. Troller-Renfree, S.V.; Costanzo, M.A.; Duncan, G.J.; Magnuson, K.; Gennetian, L.A.; Yoshikawa, H.; Halpern-Meekin, S.;</p><p>Fox, N.A.; Noble, K.G. The Impact of a Poverty Reduction Intervention on Infant Brain Activity. Proc. Natl. Acad. Sci. USA</p><p>2022, 119, e2115649119. [CrossRef]</p><p>200. Vogel Ciernia, A.; Laufer, B.I.; Dunaway, K.W.; Mordaunt, C.E.; Coulson, R.L.; Totah, T.S.; Stolzenberg, D.S.; Frahm, J.C.;</p><p>Singh-Taylor, A.; Baram, T.Z.; et al. Experience-Dependent Neuroplasticity of the Developing Hypothalamus: Integrative</p><p>Epigenomic Approaches. Epigenetics 2018, 13, 318–330. [CrossRef]</p><p>201. Marrocco, J.; McEwen, B.S. Sex in the Brain: Hormones and Sex Differences. Dialogues Clin. Neurosci. 2016, 18, 373–383. [CrossRef]</p><p>202. Duarte-Guterman, P.; Yagi, S.; Chow, C.; Galea, L.A.M. Hippocampal Learning, Memory, and Neurogenesis: Effects of Sex and</p><p>Estrogens across the Lifespan in Adults. Horm. Behav. 2015, 74, 37–52. [CrossRef]</p><p>203. Triviño-Paredes, J.; Patten, A.R.; Gil-Mohapel, J.; Christie, B.R. The Effects of Hormones and Physical Exercise on Hippocampal</p><p>Structural Plasticity. Front. Neuroendocrinol. 2016, 41, 23–43. [CrossRef]</p><p>204. Hojo, Y.; Murakami, G.; Mukai, H.; Higo, S.; Hatanaka, Y.; Ogiue-Ikeda, M.; Ishii, H.; Kimoto, T.; Kawato, S. Estrogen Synthesis in</p><p>the Brain—Role in Synaptic Plasticity and Memory. Mol. Cell. Endocrinol. 2008, 290, 31–43. [CrossRef]</p><p>205. Hojo, Y.; Hattori, T.; Enami, T.; Furukawa, A.; Suzuki, K.; Ishii, H.; Mukai, H.; Morrison, J.H.; Janssen, W.G.M.; Kominami, S.; et al.</p><p>Adult Male Rat Hippocampus Synthesizes Estradiol from Pregnenolone by Cytochromes P45017α and P450 Aromatase Localized</p><p>in Neurons. Proc. Natl. Acad. Sci. USA 2004, 101, 865–870. [CrossRef]</p><p>206. Kato, A.; Hojo, Y.; Higo, S.; Komatsuzaki, Y.; Murakami, G.; Yoshino, H.; Uebayashi, M.; Kawato, S. Female Hippocampal</p><p>Estrogens Have a Significant Correlation with Cyclic Fluctuation of Hippocampal Spines. Front. Neural Circuits 2013, 7, 149.</p><p>[CrossRef]</p><p>207. Okamoto, M.; Hojo, Y.; Inoue, K.; Matsui, T.; Kawato, S.; McEwen, B.S.; Soya, H. Mild Exercise Increases Dihydrotestosterone in</p><p>Hippocampus Providing Evidence for Androgenic Mediation of Neurogenesis. Proc. Natl. Acad. Sci. USA 2012, 109, 13100–13105.</p><p>[CrossRef]</p><p>208. Prange-Kiel, J.; Wehrenberg, U.; Jarry, H.; Rune, G.M. Para/Autocrine Regulation of Estrogen Receptors in Hippocampal Neurons.</p><p>Hippocampus 2003, 13, 226–234. [CrossRef]</p><p>209. Camacho-Arroyo, I.; Piña-Medina, A.G.; Bello-Alvarez, C.; Zamora-Sánchez, C.J. Sex Hormones and Proteins Involved in Brain</p><p>Plasticity. Vitam. Horm. 2020, 114, 145–165. [CrossRef]</p><p>210. Fester, L.; Rune, G.M. Sex Neurosteroids: Hormones Made by the Brain for the Brain. Neurosci. Lett. 2021, 753, 135849. [CrossRef]</p><p>211. Brunne, B.; Rune, G.M. Sex Neurosteroidogenesis and Hippocampal Network Maintenance. Curr. Opin. Endocr. Metab. Res.</p><p>2022, 23, 100316. [CrossRef]</p><p>212. McEwen, B.S.; Akama, K.T.; Spencer-Segal, J.L.; Milner, T.A.; Waters, E.M. Estrogen Effects on the Brain: Actions beyond the</p><p>Hypothalamus via Novel Mechanisms. Behav. Neurosci. 2012, 126, 4–16. [CrossRef]</p><p>213. Brann, D.W.; Lu, Y.; Wang, J.; Zhang, Q.; Thakkar, R.; Sareddy, G.R.; Pratap, U.P.; Tekmal, R.R.; Vadlamudi, R.K. Brain-Derived</p><p>Estrogen and Neural Function. Neurosci. Biobehav. Rev. 2022, 132, 793–817. [CrossRef]</p><p>214. Lu, Y.; Sareddy, G.R.; Wang, J.; Wang, R.; Li, Y.; Dong, Y.; Zhang, Q.; Liu, J.; O’Connor, J.C.; Xu, J.; et al. Neuron-Derived Estrogen</p><p>Regulates Synaptic Plasticity and Memory. J. Neurosci. 2019, 39, 2792–2809. [CrossRef]</p><p>https://doi.org/10.1542/peds.2015-4698</p><p>https://doi.org/10.1542/peds.2011-2663</p><p>https://doi.org/10.1126/science.1248429</p><p>https://doi.org/10.1073/pnas.1200041109</p><p>https://doi.org/10.1038/s41583-019-0120-x</p><p>https://doi.org/10.1016/j.neuron.2015.01.001</p><p>https://doi.org/10.1590/1516-4446-2014-1443</p><p>https://doi.org/10.1073/pnas.2115649119</p><p>https://doi.org/10.1080/15592294.2018.1451720</p><p>https://doi.org/10.31887/DCNS.2016.18.4/jmarrocco</p><p>https://doi.org/10.1016/j.yhbeh.2015.05.024</p><p>https://doi.org/10.1016/j.yfrne.2016.03.001</p><p>https://doi.org/10.1016/j.mce.2008.04.017</p><p>https://doi.org/10.1073/pnas.2630225100</p><p>https://doi.org/10.3389/fncir.2013.00149</p><p>https://doi.org/10.1073/pnas.1210023109</p><p>https://doi.org/10.1002/hipo.10075</p><p>https://doi.org/10.1016/bs.vh.2020.04.002</p><p>https://doi.org/10.1016/j.neulet.2021.135849</p><p>https://doi.org/10.1016/j.coemr.2022.100316</p><p>https://doi.org/10.1037/a0026708</p><p>https://doi.org/10.1016/j.neubiorev.2021.11.014</p><p>https://doi.org/10.1523/JNEUROSCI.1970-18.2019</p><p>Brain Sci. 2023, 13, 1610 27 of 32</p><p>215. Bettio, L.E.B.; Thacker, J.S.; Rodgers, S.P.; Brocardo, P.S.; Christie, B.R.; Gil-Mohapel, J. Interplay between Hormones and Exercise</p><p>on Hippocampal Plasticity across the Lifespan. Biochim. Biophys. Acta-Mol. Basis Dis. 2020, 1866, 165821. [CrossRef]</p><p>216. López-Otín, C.; Blasco, M.A.; Partridge, L.; Serrano, M.; Kroemer, G. The Hallmarks of Aging. Cell 2013, 153, 1194–1217. [CrossRef]</p><p>217. United Nations. World Population Ageing 2019; United Nations: New York, NY, USA, 2020.</p><p>218. United Nations. World Population Ageing 2019: Highlights; United Nations: New York, NY, USA, 2019.</p><p>219. World Health Organization. World Report on Ageing and Health; World Health Organization: Geneva, Switzerland, 2015.</p><p>220. Centers for Disease Control and Prevention. Diabetes and Aging; Centers for Disease Control and Prevention: Atlanta, GA, USA, 2021.</p><p>221. American Cancer Society. Cancer Facts and Figures 2021; American Cancer Society: Atlanta, GA, USA, 2021.</p><p>222. Beard, J.R.; Officer, A.; de Carvalho, I.A.; Sadana, R.; Pot, A.M.; Michel, J.-P.; Lloyd-Sherlock, P.; Epping-Jordan, J.E.;</p><p>Peeters, G.M.E.E.; Mahanani, W.R.; et al. The World Report on Ageing and Health: A Policy Framework for Healthy Ageing.</p><p>Lancet 2016, 387, 2145–2154. [CrossRef]</p><p>223. Childs, B.G.; Durik, M.; Baker, D.J.; van Deursen, J.M. Cellular Senescence in Aging and Age-Related Disease: From Mechanisms</p><p>to Therapy. Nat. Med. 2015, 21, 1424–1435. [CrossRef]</p><p>224. Navarro, A.; Boveris, A. The Mitochondrial Energy Transduction System and the Aging Process. Am. J. Physiol. Physiol.</p><p>2007, 292, C670–C686. [CrossRef]</p><p>225. Muñoz-Espín, D.; Serrano, M. Cellular Senescence: From Physiology to Pathology. Nat. Rev. Mol. Cell Biol. 2014, 15, 482–496.</p><p>[CrossRef]</p><p>226. Stern, Y. Cognitive Reserve</p><p>Brain Sci. 2023, 12, x FOR PEER REVIEW 31 of 36</p><p>211. Brunne, B.; Rune, G.M. Sex Neurosteroidogenesis and Hippocampal Network Maintenance. Curr. Opin. Endocr. Metab. Res. 2022,</p><p>23, 100316. https://doi.org/10.1016/j.coemr.2022.100316.</p><p>212. McEwen, B.S.; Akama, K.T.; Spencer-Segal, J.L.; Milner, T.A.; Waters, E.M. Estrogen Effects on the Brain: Actions beyond the</p><p>Hypothalamus via Novel Mechanisms. Behav. Neurosci. 2012, 126, 4–16. https://doi.org/10.1037/a0026708.</p><p>213. Brann, D.W.; Lu, Y.; Wang, J.; Zhang, Q.; Thakkar, R.; Sareddy, G.R.; Pratap, U.P.; Tekmal, R.R.; Vadlamudi, R.K. Brain-Derived</p><p>Estrogen and Neural Function. Neurosci. Biobehav. Rev. 2022, 132, 793–817. https://doi.org/10.1016/j.neubiorev.2021.11.014.</p><p>214. Lu, Y.; Sareddy, G.R.; Wang, J.; Wang, R.; Li, Y.; Dong, Y.; Zhang, Q.; Liu, J.; O�Connor, J.C.; Xu, J.; et al. Neuron-Derived Estrogen</p><p>Regulates Synaptic Plasticity and Memory. J. Neurosci. 2019, 39, 2792–2809. https://doi.org/10.1523/JNEUROSCI.1970-18.2019.</p><p>215. Bettio, L.E.B.; Thacker, J.S.; Rodgers, S.P.; Brocardo, P.S.; Christie, B.R.; Gil-Mohapel, J. Interplay between Hormones and Exer-</p><p>cise on Hippocampal Plasticity across the Lifespan. Biochim. Biophys. Acta-Mol. Basis Dis. 2020, 1866, 165821.</p><p>https://doi.org/10.1016/j.bbadis.2020.165821.</p><p>216. López-Otín, C.; Blasco, M.A.; Partridge, L.; Serrano, M.; Kroemer, G. The Hallmarks of Aging. Cell 2013, 153, 1194–1217.</p><p>https://doi.org/10.1016/j.cell.2013.05.039.</p><p>217. United Nations.</p><p>World Population Ageing 2019; United Nations: New York, NY, USA, 2020.</p><p>218. United Nations. World Population Ageing 2019: Highlights; United Nations: New York, NY, USA, 2019.</p><p>219. World Health Organization. World Report on Ageing and Health; World Health Organization: Geneva, Switzerland, 2015.</p><p>220. Centers for Disease Control and Prevention. Diabetes and Aging; Centers for Disease Control and Prevention: Atlanta, GA, USA, 2021.</p><p>221. American Cancer Society. Cancer Facts and Figures 2021; American Cancer Society: Atlanta, GA, USA, 2021.</p><p>222. Beard, J.R.; Officer, A.; de Carvalho, I.A.; Sadana, R.; Pot, A.M.; Michel, J.-P.; Lloyd-Sherlock, P.; Epping-Jordan, J.E.; Peeters,</p><p>G.M.E.E.; Mahanani, W.R.; et al. The World Report on Ageing and Health: A Policy Framework for Healthy Ageing. Lancet</p><p>2016, 387, 2145–2154. https://doi.org/10.1016/S0140-6736(15)00516-4.</p><p>223. Childs, B.G.; Durik, M.; Baker, D.J.; van Deursen, J.M. Cellular Senescence in Aging and Age-Related Disease: From Mechanisms</p><p>to Therapy. Nat. Med. 2015, 21, 1424–1435. https://doi.org/10.1038/nm.4000.</p><p>224. Navarro, A.; Boveris, A. The Mitochondrial Energy Transduction System and the Aging Process. Am. J. Physiol. Physiol. 2007,</p><p>292, C670–C686. https://doi.org/10.1152/ajpcell.00213.2006.</p><p>225. Muñoz-Espín, D.; Serrano, M. Cellular Senescence: From Physiology to Pathology. Nat. Rev. Mol. Cell Biol. 2014, 15, 482–496.</p><p>https://doi.org/10.1038/nrm3823.</p><p>226. Stern, Y. Cognitive Reserve☆. Neuropsychologia 2009, 47, 2015–2028. https://doi.org/10.1016/j.neuropsychologia.2009.03.004.</p><p>227. Stern, Y.; Arenaza-Urquijo, E.M.; Bartrés-Faz, D.; Belleville, S.; Cantilon, M.; Chetelat, G.; Ewers, M.; Franzmeier, N.; Kemper-</p><p>mann, G.; Kremen, W.S.; et al. Whitepaper: Defining and Investigating Cognitive Reserve, Brain Reserve, and Brain Mainte-</p><p>nance. Alzheimers Dement. 2020, 16, 1305–1311. https://doi.org/10.1016/j.jalz.2018.07.219.</p><p>228. McQuail, J.A.; Dunn, A.R.; Stern, Y.; Barnes, C.A.; Kempermann, G.; Rapp, P.R.; Kaczorowski, C.C.; Foster, T.C. Cognitive Re-</p><p>serve in Model Systems for Mechanistic Discovery: The Importance of Longitudinal Studies. Front. Aging Neurosci. 2021, 12,</p><p>607685. https://doi.org/10.3389/fnagi.2020.607685.</p><p>229. Ionescu-Tucker, A.; Cotman, C.W. Emerging Roles of Oxidative Stress in Brain Aging and Alzheimer�s Disease. Neurobiol. Aging</p><p>2021, 107, 86–95. https://doi.org/10.1016/j.neurobiolaging.2021.07.014.</p><p>230. Baracaldo-Santamaría, D.; Avendaño-Lopez, S.S.; Ariza-Salamanca, D.F.; Rodriguez-Giraldo, M.; Calderon-Ospina, C.A.; Gon-</p><p>zález-Reyes, R.E.; Nava-Mesa, M.O. Role of Calcium Modulation in the Pathophysiology and Treatment of Alzheimer�s Disease.</p><p>Int. J. Mol. Sci. 2023, 24, 9067. https://doi.org/10.3390/ijms24109067.</p><p>231. Lesuis, S.L.; Hoeijmakers, L.; Korosi, A.; de Rooij, S.R.; Swaab, D.F.; Kessels, H.W.; Lucassen, P.J.; Krugers, H.J. Vulnerability</p><p>and Resilience to Alzheimer�s Disease: Early Life Conditions Modulate Neuropathology and Determine Cognitive Reserve.</p><p>Alzheimers. Res. Ther. 2018, 10, 95. https://doi.org/10.1186/s13195-018-0422-7.</p><p>232. Jellinger, K.A.; Attems, J. Neuropathological Approaches to Cerebral Aging and Neuroplasticity. Dialogues Clin. Neurosci. 2013,</p><p>15, 29–43. https://doi.org/10.31887/DCNS.2013.15.1/kjellinger.</p><p>233. Hy, L.X.; Keller, D.M. Prevalence of AD among Whites. Neurology 2000, 55, 198–204. https://doi.org/10.1212/WNL.55.2.198.</p><p>234. Niu, H.; Álvarez-Álvarez, I.; Guillén-Grima, F.; Aguinaga-Ontoso, I. Prevalence and Incidence of Alzheimer�s Disease in Eu-</p><p>rope: A Meta-Analysis. Neurologia 2017, 32, 523–532. https://doi.org/10.1016/j.nrleng.2016.02.009.</p><p>235. Knopman, D.S.; Amieva, H.; Petersen, R.C.; Chételat, G.; Holtzman, D.M.; Hyman, B.T.; Nixon, R.A.; Jones, D.T. Alzheimer</p><p>Disease. Nat. Rev. Dis. Prim. 2021, 7, 33. https://doi.org/10.1038/s41572-021-00269-y.</p><p>236. Guo, J.; Huang, X.; Dou, L.; Yan, M.; Shen, T.; Tang, W.; Li, J. Aging and Aging-Related Diseases: From Molecular Mechanisms</p><p>to Interventions and Treatments. Signal Transduct. Target. Ther. 2022, 7, 391. https://doi.org/10.1038/s41392-022-01251-0.</p><p>237. Verkhratsky, A.; Lazareva, N.; Semyanov, A. Glial Decline and Loss of Homeostatic Support Rather than Inflammation Defines</p><p>Cognitive Aging. Neural Regen. Res. 2022, 17, 565. https://doi.org/10.4103/1673-5374.320979.</p><p>238. Barrientos, R.M.; Kitt, M.M.; Watkins, L.R.; Maier, S.F. Neuroinflammation in the Normal Aging Hippocampus. Neuroscience</p><p>2015, 309, 84–99. https://doi.org/10.1016/j.neuroscience.2015.03.007.</p><p>239. Wang, X.; Michaelis, E.K. Selective Neuronal Vulnerability to Oxidative Stress in the Brain. Front. Aging Neurosci. 2010, 2, 12.</p><p>https://doi.org/10.3389/fnagi.2010.00012.</p><p>240. Grimm, A.; Eckert, A. Brain Aging and Neurodegeneration: From a Mitochondrial Point of View. J. Neurochem. 2017, 143, 418–</p><p>431. https://doi.org/10.1111/jnc.14037.</p><p>. Neuropsychologia 2009, 47, 2015–2028. [CrossRef]</p><p>227. Stern, Y.; Arenaza-Urquijo, E.M.; Bartrés-Faz, D.; Belleville, S.; Cantilon, M.; Chetelat, G.; Ewers, M.; Franzmeier, N.;</p><p>Kempermann, G.; Kremen, W.S.; et al. Whitepaper: Defining and Investigating Cognitive Reserve, Brain Reserve, and Brain</p><p>Maintenance. Alzheimers Dement. 2020, 16, 1305–1311. [CrossRef]</p><p>228. McQuail, J.A.; Dunn, A.R.; Stern, Y.; Barnes, C.A.; Kempermann, G.; Rapp, P.R.; Kaczorowski, C.C.; Foster, T.C. Cognitive Reserve</p><p>in Model Systems for Mechanistic Discovery: The Importance of Longitudinal Studies. Front. Aging Neurosci. 2021, 12, 607685.</p><p>[CrossRef]</p><p>229. Ionescu-Tucker, A.; Cotman, C.W. Emerging Roles of Oxidative Stress in Brain Aging and Alzheimer’s Disease. Neurobiol. Aging</p><p>2021, 107, 86–95. [CrossRef]</p><p>230. Baracaldo-Santamaría, D.; Avendaño-Lopez, S.S.; Ariza-Salamanca, D.F.; Rodriguez-Giraldo, M.; Calderon-Ospina, C.A.;</p><p>González-Reyes, R.E.; Nava-Mesa, M.O. Role of Calcium Modulation in the Pathophysiology and Treatment of Alzheimer’s</p><p>Disease. Int. J. Mol. Sci. 2023, 24, 9067. [CrossRef]</p><p>231. Lesuis, S.L.; Hoeijmakers, L.; Korosi, A.; de Rooij, S.R.; Swaab, D.F.; Kessels, H.W.; Lucassen, P.J.; Krugers, H.J. Vulnerability</p><p>and Resilience to Alzheimer’s Disease: Early Life Conditions Modulate Neuropathology and Determine Cognitive Reserve.</p><p>Alzheimers. Res. Ther. 2018, 10, 95. [CrossRef]</p><p>232. Jellinger, K.A.; Attems, J. Neuropathological Approaches to Cerebral Aging and Neuroplasticity. Dialogues Clin. Neurosci.</p><p>2013, 15, 29–43. [CrossRef]</p><p>233. Hy, L.X.; Keller, D.M. Prevalence of AD among Whites. Neurology 2000, 55, 198–204. [CrossRef]</p><p>234. Niu, H.; Álvarez-Álvarez, I.; Guillén-Grima, F.; Aguinaga-Ontoso, I. Prevalence and Incidence of Alzheimer’s Disease in Europe:</p><p>A Meta-Analysis. Neurologia 2017, 32, 523–532. [CrossRef]</p><p>235. Knopman, D.S.; Amieva, H.; Petersen, R.C.; Chételat, G.; Holtzman, D.M.; Hyman, B.T.; Nixon, R.A.; Jones, D.T. Alzheimer</p><p>Disease. Nat. Rev. Dis. Prim. 2021, 7, 33. [CrossRef]</p><p>236. Guo, J.; Huang, X.; Dou, L.; Yan, M.; Shen, T.; Tang, W.; Li, J. Aging and Aging-Related Diseases: From Molecular Mechanisms to</p><p>Interventions and Treatments. Signal Transduct. Target. Ther. 2022, 7, 391. [CrossRef]</p><p>237. Verkhratsky, A.; Lazareva, N.; Semyanov, A. Glial Decline and Loss of Homeostatic Support Rather than Inflammation Defines</p><p>Cognitive Aging. Neural Regen. Res. 2022, 17, 565. [CrossRef] [PubMed]</p><p>238. Barrientos, R.M.; Kitt, M.M.; Watkins, L.R.; Maier, S.F. Neuroinflammation in the Normal Aging Hippocampus. Neuroscience</p><p>2015, 309, 84–99. [CrossRef]</p><p>239. Wang, X.; Michaelis, E.K. Selective Neuronal Vulnerability to Oxidative Stress in the Brain. Front. Aging Neurosci. 2010, 2, 12.</p><p>[CrossRef] [PubMed]</p><p>240. Grimm, A.; Eckert, A. Brain Aging and Neurodegeneration: From a Mitochondrial Point of View. J. Neurochem. 2017, 143, 418–431.</p><p>[CrossRef]</p><p>241. Johnson, F.B.; Sinclair, D.A.; Guarente, L. Molecular Biology of Aging. Cell 1999, 96, 291–302. [CrossRef] [PubMed]</p><p>242. Khachaturian, Z.S. The Role of Calcium Regulation in Brain Aging: Reexamination of a Hypothesis. Aging Clin. Exp. Res.</p><p>1989, 1, 17–34. [CrossRef]</p><p>243. Foster, T.C. Calcium Homeostasis and Modulation of Synaptic Plasticity in the Aged Brain. Aging Cell 2007, 6, 319–325. [CrossRef]</p><p>244. Gant, J.C.; Chen, K.-C.; Kadish, I.; Blalock, E.M.; Thibault, O.; Porter, N.M.; Landfield, P.W. Reversal of Aging-Related Neuronal</p><p>Ca2+ Dysregulation and Cognitive Impairment by Delivery of a Transgene Encoding FK506-Binding Protein 12.6/1b to the</p><p>Hippocampus. J. Neurosci. 2015, 35, 10878–10887. [CrossRef]</p><p>245. Kumar, A.; Foster, T.C. Alteration in NMDA Receptor Mediated Glutamatergic Neurotransmission in the Hippocampus During</p><p>Senescence. Neurochem. Res. 2019, 44, 38–48. [CrossRef]</p><p>https://doi.org/10.1016/j.bbadis.2020.165821</p><p>https://doi.org/10.1016/j.cell.2013.05.039</p><p>https://doi.org/10.1016/S0140-6736(15)00516-4</p><p>https://doi.org/10.1038/nm.4000</p><p>https://doi.org/10.1152/ajpcell.00213.2006</p><p>https://doi.org/10.1038/nrm3823</p><p>https://doi.org/10.1016/j.neuropsychologia.2009.03.004</p><p>https://doi.org/10.1016/j.jalz.2018.07.219</p><p>https://doi.org/10.3389/fnagi.2020.607685</p><p>https://doi.org/10.1016/j.neurobiolaging.2021.07.014</p><p>https://doi.org/10.3390/ijms24109067</p><p>https://doi.org/10.1186/s13195-018-0422-7</p><p>https://doi.org/10.31887/DCNS.2013.15.1/kjellinger</p><p>https://doi.org/10.1212/WNL.55.2.198</p><p>https://doi.org/10.1016/j.nrl.2016.02.016</p><p>https://doi.org/10.1038/s41572-021-00269-y</p><p>https://doi.org/10.1038/s41392-022-01251-0</p><p>https://doi.org/10.4103/1673-5374.320979</p><p>https://www.ncbi.nlm.nih.gov/pubmed/34380892</p><p>https://doi.org/10.1016/j.neuroscience.2015.03.007</p><p>https://doi.org/10.3389/fnagi.2010.00012</p><p>https://www.ncbi.nlm.nih.gov/pubmed/20552050</p><p>https://doi.org/10.1111/jnc.14037</p><p>https://doi.org/10.1016/S0092-8674(00)80567-X</p><p>https://www.ncbi.nlm.nih.gov/pubmed/9988222</p><p>https://doi.org/10.1007/BF03323872</p><p>https://doi.org/10.1111/j.1474-9726.2007.00283.x</p><p>https://doi.org/10.1523/JNEUROSCI.1248-15.2015</p><p>https://doi.org/10.1007/s11064-018-2634-4</p><p>Brain Sci. 2023, 13, 1610 28 of 32</p><p>246. Santos-Parker, J.R.; LaRocca, T.J.; Seals, D.R. Aerobic Exercise and Other Healthy Lifestyle Factors That Influence Vascular Aging.</p><p>Adv. Physiol. Educ. 2014, 38, 296–307. [CrossRef]</p><p>247. Simpson, R.J.; Kunz, H.; Agha, N.; Graff, R. Exercise and the Regulation of Immune Functions. Prog. Mol. Biol. Transl. Sci.</p><p>2015, 135, 355–380. [CrossRef]</p><p>248. Fjell, A.M.; Walhovd, K.B. Structural Brain Changes in Aging: Courses, Causes and Cognitive Consequences. Rev. Neurosci.</p><p>2010, 21, 187–222. [CrossRef]</p><p>249. Beyer, K.; Domingo-Sàbat, M.; Ariza, A.; Guillazo-Blanch, G. Molecular Anatomy of the Nigrostriatal Pathway in the Rat: A</p><p>Review. Neuroscience 2012, 227, 70–83.</p><p>250. Gil, J.M.; Rego, A.C. Mechanisms of Neurodegeneration in Huntington’s Disease. Eur. J. Neurosci. 2008, 27, 2803–2820. [CrossRef]</p><p>251. Li, Y.; Liu, W.; Oo, T.F.; Wang, L.; Tang, Y.; Jackson-Lewis, V.; Zhou, C.; Geghman, K.; Bogdanov, M.; Przedborski, S.; et al. Mutant</p><p>LRRK2R1441G BAC Transgenic Mice Recapitulate Cardinal Features of Parkinson’s Disease. Nat. Neurosci. 2009, 12, 826–828.</p><p>[CrossRef]</p><p>252. Alzheimer’s Association. 2022 Alzheimer’s Disease Facts and Figures, More Than Normal Aging: Understanding Mild Cognitive</p><p>Impairment; Alzheimer’s Association: Chicago, IL, USA, 2022.</p><p>253. Parkinson’s Foundation. Statistics on Parkinson’s; Parkinson’s Foundation: New York, NY, USA, 2021.</p><p>254. World Health Organization. Dementia; World Health Organization: Geneva, Switzerland, 2019.</p><p>255. Bhat, A.H.; Dar, K.B.; Anees, S.; Zargar, M.A.; Masood, A.; Sofi, M.A.; Ganie, S.A. Oxidative Stress, Mitochondrial Dysfunction</p><p>and Neurodegenerative Diseases; a Mechanistic Insight. Biomed. Pharmacother. 2015, 74, 101–110. [CrossRef]</p><p>256. Sweeney, P.; Park, H.; Baumann, M.; Dunlop, J.; Frydman, J.; Kopito, R.; McCampbell, A.; Leblanc, G.; Venkateswaran, A.;</p><p>Nurmi, A.; et al. Protein misfolding in neurodegenerative diseases: Implications and strategies. Transl Neurodegener. 2017, 6, 1–13.</p><p>[CrossRef]</p><p>257. DiFiglia, M.; Sapp, E.; Chase, K.O.; Davies, S.W.; Bates, G.P.; Vonsattel, J.P.; Aronin, N. Aggregation of Huntingtin in Neuronal</p><p>Intranuclear Inclusions and Dystrophic Neurites in Brain. Science 1997, 277, 1990–1993. [CrossRef]</p><p>258. Moreno-Gonzalez, I.; Edwards III, G.; Salvadores, N. Shining a Light on Protein Misfolding and Aggregation in Neurological</p><p>Diseases. Protein Sci. 2018, 27, 1820–1831.</p><p>259. Sun, N.; Youle, R.J.; Finkel, T. The Mitochondrial Basis of Aging. Mol. Cell 2016, 61, 654–666. [CrossRef]</p><p>260. Hroudová, J.; Singh, N.; Fišar, Z. Mitochondrial Dysfunctions in Neurodegenerative Diseases: Relevance to Alzheimer’s Disease.</p><p>Biomed Res. Int. 2014, 2014, 175062. [CrossRef]</p><p>261. Li, M.; Fu, X.; Xie, W.; Guo, W.; Li, B.; Cui, R.; Yang, W. Effect of Early Life Stress on the Epigenetic Profiles in Depression. Front.</p><p>Cell Dev. Biol. 2020, 8, 867. [CrossRef]</p><p>262. Niranjan, R. The Role of Inflammatory and Oxidative Stress Mechanisms in the Pathogenesis of Parkinson’s Disease: Focus on</p><p>Astrocytes. Mol. Neurobiol. 2014, 49, 28–38. [CrossRef]</p><p>263. das Neves, S.P.; Sousa, J.C.; Sousa, N.; Cerqueira, J.J.; Marques, F. Altered astrocytic function in experimental neuroinflammation</p><p>and multiple sclerosis. Glia 2021, 69, 1341–1368. [CrossRef]</p><p>264. Xie, Z.; Morgan, T.E.; Rozovsky, I.; Finch, C.E.; Longo, D.L. The Inflammation-Coagulation Connection in Brain Aging and</p><p>Alzheimer’s Disease. Neurosci. Biobehav. Rev. 2013, 37, 1572–1587.</p><p>265. Prusiner, S.B. A Unifying Role for Prions in Neurodegenerative Diseases. Science 2012, 336, 1511–1513. [CrossRef]</p><p>266. Powers, E.T.; Balch, W.E. Diversity in the Origins of Proteostasis Networks—A Driver for Protein Function in Evolution. Nat. Rev.</p><p>Mol. Cell Biol. 2013, 14, 237–248. [CrossRef]</p><p>267. Hoozemans, J.J.M.; van Haastert, E.S.; Nijholt, D.A.T.; Rozemuller, A.J.M.; Eikelenboom, P.; Scheper, W. The Unfolded Protein</p><p>Response Is Activated in Pretangle Neurons in Alzheimer’s Disease Hippocampus. Am. J. Pathol. 2009, 174, 1241–1251. [CrossRef]</p><p>268. Hetz, C.; Saxena, S. ER Stress and the Unfolded Protein Response in Neurodegeneration. Nat. Rev. Neurol. 2017, 13, 477–491.</p><p>[CrossRef]</p><p>269. Smith, H.L.; Mallucci, G.R. The Unfolded Protein Response: Mechanisms and Therapy of Neurodegeneration. Brain</p><p>2016, 139, 2113–2121. [CrossRef]</p><p>270. Hoozemans, J.J.M.; Veerhuis, R.; Van Haastert, E.S.; Rozemuller, J.M.; Baas, F.; Eikelenboom, P.; Scheper, W. The Unfolded Protein</p><p>Response Is Activated in Alzheimer’s Disease. Acta Neuropathol. 2005, 110, 165–172. [CrossRef]</p><p>271. Bellucci, A.; Navarria, L.; Zaltieri, M.; Falarti, E.; Bodei, S.; Sigala, S.; Battistin, L.; Spillantini, M.; Missale, C.; Spano, P. Induction of</p><p>the Unfolded Protein Response by α-Synuclein in Experimental Models of Parkinson’s Disease. J. Neurochem. 2011, 116, 588–605.</p><p>[CrossRef]</p><p>272. Briston, T.; Hicks, A.R. Mitochondrial Dysfunction and Neurodegenerative Proteinopathies: Mechanisms and Prospects for</p><p>Therapeutic Intervention. Biochem. Soc. Trans. 2018, 46, 829–842. [CrossRef]</p><p>273. Kraytsberg, Y.; Kudryavtseva, E.; McKee, A.C.; Geula, C.; Kowall, N.W.; Khrapko, K. Mitochondrial DNA Deletions Are Abundant</p><p>and Cause Functional Impairment in Aged Human Substantia Nigra Neurons. Nat. Genet. 2006, 38, 518–520. [CrossRef]</p><p>274. Rhein, V.; Baysang, G.; Rao, S.; Meier, F.; Bonert, A.; Müller-Spahn, F.; Eckert, A. Amyloid-Beta Leads to Impaired Cellular</p><p>Respiration, Energy Production and Mitochondrial Electron Chain Complex Activities in Human Neuroblastoma Cells. Cell. Mol.</p><p>Neurobiol. 2009, 29, 1063–1071. [CrossRef]</p><p>275. Du, J.; Wang, Y.; Hunter, R.; Wei, Y.; Blumenthal, R.; Falke, C.; Khairova, R.; Zhou, R.; Yuan, P.; Machado-Vieira, R.; et al. Dynamic</p><p>Regulation of Mitochondrial Function by Glucocorticoids. Proc. Natl. Acad. Sci. USA 2009, 106, 3543–3548. [CrossRef]</p><p>https://doi.org/10.1152/advan.00088.2014</p><p>https://doi.org/10.1016/bs.pmbts.2015.08.001</p><p>https://doi.org/10.1515/REVNEURO.2010.21.3.187</p><p>https://doi.org/10.1111/j.1460-9568.2008.06310.x</p><p>https://doi.org/10.1038/nn.2349</p><p>https://doi.org/10.1016/j.biopha.2015.07.025</p><p>https://doi.org/10.1186/s40035-017-0077-5</p><p>also known as brain plasticity or neural plasticity, is the biological</p><p>capacity of the brain to adapt physiologically or even alter its anatomical structure in</p><p>response to stimuli or damage [26]. This ability is central to learning, memory, injury</p><p>recovery, and adaptation to environmental changes [27].</p><p>2.1. Structural Neuroplasticity</p><p>Structural neuroplasticity refers to physical changes to neural circuits, including the</p><p>growth of new dendritic spines, axonal sprouting, and even neurogenesis. In particular, neu-</p><p>rogenesis refers to generating new functional neurons, a multifaceted and tightly regulated</p><p>process involving the proliferation, differentiation, and integration of new neurons from</p><p>neural precursor cells. Each stage is characterized by the activation and presence of distinct</p><p>transcriptional factors and markers [12,28–30]. Structural neuroplasticity is essential to</p><p>rewiring the brain and has implications for recovery after brain injury, neurodevelopment,</p><p>and adaptations to sensory input alterations throughout life [31].</p><p>2.1.1. Developmental Neurogenesis and Synaptogenesis</p><p>The development of the central nervous system (CNS) begins in the early weeks</p><p>following fertilization, shortly after formation of the three embryonic germ layers, ectoderm,</p><p>mesoderm, and endoderm, a crucial phase during embryonic development. The CNS</p><p>Brain Sci. 2023, 13, 1610 3 of 32</p><p>derives from the differentiation of multipotent cells present in the ectoderm through the</p><p>formation of the neural plate in the dorsal region of the embryo. This neural plate will</p><p>fold its crest in the craniocaudal and rostrocaudal directions, forming the neural tube at</p><p>embryonic day 30 in the human [32]. The closure of the neural tube marks the beginning</p><p>of rapid brain enlargement from two groups of cells: neural stem cells (NSCs), which are</p><p>multipotent cells that can give rise to various types of neural cells, including neurons,</p><p>astrocytes, and oligodendrocytes, and express markers such as Sox 2 and Nestin; and</p><p>neural precursor cells (NPCs), which are immediate descendants of neural stem cells, are</p><p>committed to a neuronal fate and express markers such as Pax6, Dlx2, and Tbr2 [30,32].</p><p>Between weeks 4 and 5 of human embryonic development begins a phase known as</p><p>interkinetic nuclear migration of NSCs and NPCs, which results in the symmetric division</p><p>along the ventricular edge. This early proliferation leads to an exponential increase in the</p><p>pool of progenitor cells that contribute to the expansion of surface area and thickness of</p><p>the ventricular zone [30,33,34]. Around gestational week 5 (human embryonic day 42), the</p><p>NPCs located in the ventricular zone, referred to as radial glial cells, begin to switch from</p><p>symmetric to asymmetric cell division, generating one daughter cell that remains in the</p><p>ventricular zone as a radial glial cell and a postmitotic neuron, marking the beginning of</p><p>neurogenesis itself [33]. The process of differentiation of NPCs into neurons involves the</p><p>successive expression of specific transcription factors and proteins, including Neurogenin</p><p>(Ngn) and Mash 1 (Ascl1), doublecortin (DCX), βIII-tubulin (Tuj1), and finally NeuN,</p><p>a marker of mature neurons [34]. Of note, new neurons also need to migrate to their</p><p>appropriate locations within the brain. This process involves the support of radial glial cells</p><p>and Cajal–Retzius cells, which create pathways such as the reelin pathway to aid migrating</p><p>neurons in reaching their final destination [35].</p><p>A final step in the neurogenic process involves establishing functional connections</p><p>between the newly generated neurons to form neural circuits. This process relies on</p><p>synaptic plasticity or synaptogenesis. Synaptogenesis begins approximately in human</p><p>gestational week 27 and continues to occur after birth. During the postnatal period,</p><p>synapses are produced rapidly; by age two, the number of synapses is estimated to be</p><p>twice the number in the adult brain. Indeed, synaptogenesis is an incredibly dynamic</p><p>process in the human cerebral cortex in infancy and childhood [27], with the postnatal</p><p>period being marked by enhanced experience-dependent sensitivity to sensory information.</p><p>In addition, synaptic strength and efficacy alterations occur during the development [27],</p><p>allowing the developing brain to adapt to environmental stimuli. This “critical period” is</p><p>not sustained into adulthood, though, thus restricting the ability to indiscriminately store</p><p>new sensory information [36]. Indeed, the number of synapses falls over the subsequent</p><p>years and into adolescence through a process referred to as synaptic pruning, through</p><p>which necessary synapses are preserved and redundant ones are eliminated [37]. These</p><p>findings support the idea of “windows of opportunity” that enable the construction and</p><p>consolidation of experience-dependent structural and functional brain connections during</p><p>the neurodevelopment period [38] and explain why children can readily acquire new</p><p>languages (and other skills). At the same time, this ability requires much more effort and</p><p>attention later in life [39].</p><p>Of note, in addition to being influenced by environmental stimuli, the various stages</p><p>of developmental neurogenesis and synaptogenesis are tightly regulated by numerous</p><p>intrinsic factors [32,34], including transcription factors, growth factors (such as brain-</p><p>derived neurotrophic factor (BDNF) and insulin-like growth factor (IGF)), cell adhesion</p><p>molecules (such as N-cadherin, which aids in cell migration and synaptic connection</p><p>formation), and signaling molecules (such as Notch, Sonic Hedgehog (SHH), Wnt, and</p><p>fibroblast growth factor (FGF)).</p><p>2.1.2. Adult Neurogenesis</p><p>Although the original evidence in support of adult neurogenesis dates back to the</p><p>1960s with the pioneering work of the American neuroscientist Joseph Altman [28], it was</p><p>Brain Sci. 2023, 13, 1610 4 of 32</p><p>only in the mid-1990s that this phenomenon became generally accepted [40] following the</p><p>seminal work by Eriksson et al. (1998), who demonstrated the incorporation of bromod-</p><p>eoxyuridine (BrdU; a nucleotide analog) into the DNA of newly generated neurons in the</p><p>human hippocampal dentate gyrus [41]. Since then, numerous studies have confirmed</p><p>that neural stem cells are indeed present in juvenile and adult brains [42] and that neu-</p><p>rogenesis continues to occur in select regions of the adult mammalian brain [14,43], the</p><p>most important and widely studied being the subgranular zone (SGZ) of the hippocampal</p><p>dentate gyrus [28] and the subventricular zone (SVZ) of the lateral ventricles [44]. Newborn</p><p>neurons have also been described in other brain regions, referred to as “noncanonical” neu-</p><p>rogenic areas [12], including the hypothalamus [45,46], neocortex [47,48], amygdala [49],</p><p>cerebellum [50,51], and striatum [42,52]. Of note, adult neurogenesis is thought to have</p><p>functional significance. For example, adult hippocampal neurogenesis is involved in several</p><p>emotional and cognitive functions, including spatial learning, memory, pattern separation,</p><p>and mood regulation [11,53–55].</p><p>Similarly to developmental neurogenesis, adult neurogenesis is also modulated by sev-</p><p>eral intrinsic and extrinsic factors, including trophic support [56,57], epigenetic factors [58],</p><p>physical activity [16], stress [59], environmental enrichment [14], and pharmacological inter-</p><p>ventions, such as antidepressants [60–62]. On the other hand, a reduction or dysregulation</p><p>of the neurogenic processes may contribute, at least in part, to alterations in cognition and</p><p>emotional resilience seen with normal aging [63–65], as well as with various psychiatric and</p><p>neurodegenerative conditions, including addiction [66], autism spectrum disorder [67,68],</p><p>major depressive disorder [69], Huntington’s disease (HD) [70], and Alzheimer’s disease</p><p>(AD) [71].</p><p>2.2. Functional Neuroplasticity</p><p>In contrast to structural neuroplasticity, functional neuroplasticity refers to changes in</p><p>the functional organization of neural circuits. As mentioned in this review, synaptogenesis</p><p>represents a structural aspect</p><p>https://doi.org/10.1126/science.277.5334.1990</p><p>https://doi.org/10.1016/j.molcel.2016.01.028</p><p>https://doi.org/10.1155/2014/175062</p><p>https://doi.org/10.3389/fcell.2020.00867</p><p>https://doi.org/10.1007/s12035-013-8483-x</p><p>https://doi.org/10.1002/glia.23940</p><p>https://doi.org/10.1126/science.1222951</p><p>https://doi.org/10.1038/nrm3542</p><p>https://doi.org/10.2353/ajpath.2009.080814</p><p>https://doi.org/10.1038/nrneurol.2017.99</p><p>https://doi.org/10.1093/brain/aww101</p><p>https://doi.org/10.1007/s00401-005-1038-0</p><p>https://doi.org/10.1111/j.1471-4159.2010.07143.x</p><p>https://doi.org/10.1042/BST20180025</p><p>https://doi.org/10.1038/ng1778</p><p>https://doi.org/10.1007/s10571-009-9398-y</p><p>https://doi.org/10.1073/pnas.0812671106</p><p>Brain Sci. 2023, 13, 1610 29 of 32</p><p>276. Schon, E.A.; Przedborski, S. Mitochondria: The Next (Neurode)Generation. Neuron 2011, 70, 1033–1053. [CrossRef]</p><p>277. Nunomura, A.; Moreira, P.I.; Castellani, R.J.; Lee, H.; Zhu, X.; Smith, M.A.; Perry, G. Oxidative Damage to RNA in Aging and</p><p>Neurodegenerative Disorders. Neurotox. Res. 2012, 22, 231–248. [CrossRef]</p><p>278. Mishra, S.R.; Mahapatra, K.K.; Behera, B.P.; Patra, S.; Bhol, C.S.; Panigrahi, D.P.; Praharaj, P.P.; Singh, A.; Patil, S.; Dhiman, R.; et al.</p><p>Mitochondrial dysfunction as a driver of NLRP3 inflammasome activation and its modulation through mitophagy for potential</p><p>therapeutics. Int. J. Biochem Cell Biol. 2021, 136, 106013. [CrossRef]</p><p>279. Choo, Y.S. Mutant Huntingtin Directly Increases Susceptibility of Mitochondria to the Calcium-Induced Permeability Transition</p><p>and Cytochrome c Release. Hum. Mol. Genet. 2004, 13, 1407–1420. [CrossRef]</p><p>280. Gallart-Palau, X.; Lee, B.S.; Adav, S.S.; Qian, J.; Serra, A.; Park, J.E.; Lai, M.K.; Chen, C.P.; Kalaria, R.N.; Sze, S.K. Gender</p><p>differences in white matter pathology and mitochondrial dysfunction in Alzheimer’s disease with cerebrovascular disease. Mol.</p><p>Brain. 2016, 17, 27. [CrossRef]</p><p>281. Hansson Petersen, C.A.; Alikhani, N.; Behbahani, H.; Wiehager, B.; Pavlov, P.F.; Alafuzoff, I.; Leinonen, V.; Ito, A.; Winblad, B.;</p><p>Glaser, E.; et al. The Amyloid β-Peptide Is Imported into Mitochondria via the TOM Import Machinery and Localized to</p><p>Mitochondrial Cristae. Proc. Natl. Acad. Sci. USA 2008, 105, 13145–13150. [CrossRef]</p><p>282. Schapira, A.H.V. Mitochondrial Dysfunction in Parkinson’s Disease. Cell Death Differ. 2007, 14, 1261–1266. [CrossRef]</p><p>283. Surmeier, D.J.; Obeso, J.A.; Halliday, G.M. Parkinson’s Disease Is Not Simply a Prion Disorder. J. Neurosci. 2017, 37, 9799–9807.</p><p>[CrossRef]</p><p>284. McGeer, P.L.; McGeer, E.G. Inflammation and Neurodegeneration in Parkinson’s Disease. Parkinsonism Relat. Disord.</p><p>2004, 10, S3–S7. [CrossRef]</p><p>285. Mandrekar-Colucci, S.; Karlo, J.C.; Landreth, G.E. Mechanisms Underlying the Rapid Peroxisome Proliferator-Activated Receptor-</p><p>γ-Mediated Amyloid Clearance and Reversal of Cognitive Deficits in a Murine Model of Alzheimer’s Disease. J. Neurosci.</p><p>2012, 32, 10117–10128. [CrossRef]</p><p>286. Heneka, M.T.; Carson, M.J.; El Khoury, J.; Landreth, G.E.; Brosseron, F.; Feinstein, D.L.; Jacobs, A.H.; Wyss-Coray, T.; Vitorica, J.;</p><p>Ransohoff, R.M.; et al. Neuroinflammation in Alzheimer’s Disease. Lancet Neurol. 2015, 14, 388–405. [CrossRef]</p><p>287. Liddelow, S.A.; Guttenplan, K.A.; Clarke, L.E.; Bennett, F.C.; Bohlen, C.J.; Schirmer, L.; Bennett, M.L.; Münch, A.E.; Chung, W.-S.;</p><p>Peterson, T.C.; et al. Neurotoxic Reactive Astrocytes Are Induced by Activated Microglia. Nature 2017, 541, 481–487. [CrossRef]</p><p>288. Ising, C.; Heneka, M.T. Functional and Structural Damage of Neurons by Innate Immune Mechanisms during Neurodegeneration.</p><p>Cell Death Dis. 2018, 9, 120. [CrossRef]</p><p>289. Lučiūnaitė, A.; McManus, R.M.; Jankunec, M.; Rácz, I.; Dansokho, C.; Dalgėdienė, I.; Schwartz, S.; Brosseron, F.; Heneka, M.T.</p><p>Soluble Aβ Oligomers and Protofibrils Induce NLRP3 Inflammasome Activation in Microglia. J. Neurochem. 2020, 155, 650–661.</p><p>[CrossRef]</p><p>290. La Vitola, P.; Balducci, C.; Baroni, M.; Artioli, L.; Santamaria, G.; Castiglioni, M.; Cerovic, M.; Colombo, L.; Caldinelli, L.;</p><p>Pollegioni, L.; et al. Peripheral Inflammation Exacerbates A-synuclein Toxicity and Neuropathology in Parkinson’s Models.</p><p>Neuropathol. Appl. Neurobiol. 2021, 47, 43–60. [CrossRef]</p><p>291. Colombo, E.; Farina, C. Astrocytes: Key Regulators of Neuroinflammation. Trends Immunol. 2016, 37, 608–620. [CrossRef]</p><p>292. Rodríguez-Arellano, J.J.; Parpura, V.; Zorec, R.; Verkhratsky, A. Astrocytes in Physiological Aging and Alzheimer’s Disease.</p><p>Neuroscience 2016, 323, 170–182. [CrossRef]</p><p>293. Coppedè, F.; Mancuso, M.; Siciliano, G.; Migliore, L.; Murri, L. Genes and the environment in neurodegeneration. Biosci. Rep.</p><p>2006, 26, 341–367. [CrossRef]</p><p>294. Ulrich, J.D.; Holtzman, D.M. TREM2 Function in Alzheimer’s Disease and Neurodegeneration. ACS Chem. Neurosci. 2016, 7, 420–427.</p><p>[CrossRef]</p><p>295. Gialluisi, A.; Reccia, M.G.; Modugno, N.; Nutile, T.; Lombardi, A.; Di Giovannantonio, L.G.; Pietracupa, S.; Ruggiero, D.; Scala, S.;</p><p>Gambardella, S.; et al. Identification of Sixteen Novel Candidate Genes for Late Onset Parkinson’s Disease. Mol. Neurodegener.</p><p>2021, 16, 35. [CrossRef]</p><p>296. Costa, L.G. Neurotoxicity of Pesticides: A Brief Review. Front. Biosci. 2008, 13, 1240. [CrossRef]</p><p>297. Itzhaki, R.F.; Lathe, R.; Balin, B.J.; Ball, M.J.; Bearer, E.L.; Braak, H.; Bullido, M.J.; Carter, C.; Clerici, M.; Cosby, S.L.; et al. Microbes</p><p>and Alzheimer’s Disease. J. Alzheimers Dis. 2016, 51, 979–984. [CrossRef]</p><p>298. McKee, A.C.; Stein, T.D.; Nowinski, C.J.; Stern, R.A.; Daneshvar, D.H.; Alvarez, V.E.; Lee, H.-S.; Hall, G.; Wojtowicz, S.M.;</p><p>Baugh, C.M.; et al. The Spectrum of Disease in Chronic Traumatic Encephalopathy. Brain 2013, 136, 43–64. [CrossRef]</p><p>299. Morrison, J.H.; Baxter, M.G. The Ageing Cortical Synapse: Hallmarks and Implications for Cognitive Decline. Nat. Rev. Neurosci.</p><p>2012, 13, 240–250. [CrossRef]</p><p>300. Lövdén, M.; Bäckman, L.; Lindenberger, U.; Schaefer, S.; Schmiedek, F. A Theoretical Framework for the Study of Adult Cognitive</p><p>Plasticity. Psychol. Bull. 2010, 136, 659–676. [CrossRef]</p><p>301. Burke, S.N.; Barnes, C.A.; Seneczko, K. Decreased Hippocampal LTP in Aged Rats Associated with Deficits in Explicit Memory</p><p>Retrieval. Hippocampus 2010, 20, 817–827.</p><p>302. Spencer, J.L.; Waters, E.M.; Milner, T.A.; Lee, F.S.; McEwen, B.S. BDNF Variant Val66Met Interacts with Estrous Cycle in the</p><p>Control of Hippocampal Function. Proc. Natl. Acad. Sci. USA 2010, 107, 4395–4400. [CrossRef]</p><p>303. Amani, M.; Lauterborn, J.C.; Le, A.A.; Cox, B.M.; Wang, W.; Quintanilla, J.; Cox, C.D.; Gall, C.M.; Lynch, G. Rapid Aging in the</p><p>Perforant Path Projections to the Rodent Dentate Gyrus. J. Neurosci. 2021, 41, 2301–2312. [CrossRef]</p><p>https://doi.org/10.1016/j.neuron.2011.06.003</p><p>https://doi.org/10.1007/s12640-012-9331-x</p><p>https://doi.org/10.1016/j.biocel.2021.106013</p><p>https://doi.org/10.1093/hmg/ddh162</p><p>https://doi.org/10.1186/s13041-016-0205-7</p><p>https://doi.org/10.1073/pnas.0806192105</p><p>https://doi.org/10.1038/sj.cdd.4402160</p><p>https://doi.org/10.1523/JNEUROSCI.1787-16.2017</p><p>https://doi.org/10.1016/j.parkreldis.2004.01.005</p><p>https://doi.org/10.1523/JNEUROSCI.5268-11.2012</p><p>https://doi.org/10.1016/S1474-4422(15)70016-5</p><p>https://doi.org/10.1038/nature21029</p><p>https://doi.org/10.1038/s41419-017-0153-x</p><p>https://doi.org/10.1111/jnc.14945</p><p>https://doi.org/10.1111/nan.12644</p><p>https://doi.org/10.1016/j.it.2016.06.006</p><p>https://doi.org/10.1016/j.neuroscience.2015.01.007</p><p>https://doi.org/10.1007/s10540-006-9028-6</p><p>https://doi.org/10.1021/acschemneuro.5b00313</p><p>https://doi.org/10.1186/s13024-021-00455-2</p><p>https://doi.org/10.2741/2758</p><p>https://doi.org/10.3233/JAD-160152</p><p>https://doi.org/10.1093/brain/aws307</p><p>https://doi.org/10.1038/nrn3200</p><p>https://doi.org/10.1037/a0020080</p><p>https://doi.org/10.1073/pnas.0915105107</p><p>https://doi.org/10.1523/JNEUROSCI.2376-20.2021</p><p>Brain Sci. 2023, 13, 1610 30 of 32</p><p>304. Buss, E.W.; Corbett, N.J.; Roberts, J.G.; Ybarra, N.; Musial, T.F.; Simkin, D.; Molina-Campos, E.; Oh, K.-J.; Nielsen, L.L.;</p><p>Ayala, G.D.; et al. Cognitive Aging Is Associated with Redistribution of Synaptic Weights in the Hippocampus. Proc.</p><p>Natl. Acad.</p><p>Sci. USA 2021, 118, e1921481118. [CrossRef]</p><p>305. Yu, X.-W.; Oh, M.M.; Disterhoft, J.F. CREB, Cellular Excitability, and Cognition: Implications for Aging. Behav. Brain Res.</p><p>2017, 322, 206–211. [CrossRef]</p><p>306. Gharsemian-Shirvan, M.; Khodagholi, F.; Shaerzadeh, F. Age-Related Changes in Dendritic Arborization, Spine Morphology, and</p><p>Synaptic Density: An Ultrastructural Study on CA1 Pyramidal Neurons of Rat Hippocampus. Synapse 2020, 74, e22101.</p><p>307. Lilja, A.M.; Röjdner, J.; Mustafiz, T.; Thomé, C.M.; Storelli, E.; Gonzalez, D.; Unger-Lithner, C.; Greig, N.H.; Nordberg, A.;</p><p>Marutle, A. Age-Dependent Neuroplasticity Mechanisms in Alzheimer Tg2576 Mice Following Modulation of Brain Amyloid-β</p><p>Levels. PLoS ONE 2013, 8, e58752. [CrossRef]</p><p>308. Torres, L.L.; Quaglio, N.B.; de Souza, G.T.; Garcia, R.C.T. Synaptic Plasticity, Dementia and Alzheimer Disease. In Synaptic</p><p>Plasticity in Neurodegenerative Diseases; Academic Press: Cambridge, MA, USA, 2020; pp. 67–90.</p><p>309. Demars, M.; Hu, Y.-S.; Gadadhar, A.; Lazarov, O. Impaired Neurogenesis Is an Early Event in the Etiology of Familial Alzheimer’s</p><p>Disease in Transgenic Mice. J. Neurosci. Res. 2010, 88, 2103–2117. [CrossRef]</p><p>310. Gil-Mohapel, J.; Brocardo, P.S.; Choquette, W.; Gothard, R.; Simpson, J.M.; Christie, B.R. Hippocampal Neurogenesis Levels</p><p>Predict WATERMAZE Search Strategies in the Aging Brain. PLoS ONE 2013, 8, e75125. [CrossRef]</p><p>311. Fjell, A.M.; Walhovd, K.B.; Fennema-Notestine, C.; McEvoy, L.K.; Hagler, D.J.; Holland, D.; Brewer, J.B.; Dale, A.M. One-Year</p><p>Brain Atrophy Evident in Healthy Aging. J. Neurosci. 2009, 29, 15223–15231. [CrossRef]</p><p>312. Hötting, K.; Röder, B. Beneficial Effects of Physical Exercise on Neuroplasticity and Cognition. Neurosci. Biobehav. Rev.</p><p>2013, 37, 2243–2257. [CrossRef]</p><p>313. Franzén, E.; Johansson, H.; Freidle, M.; Ekman, U.; Wallén, M.B.; Schalling, E.; Lebedev, A.; Lövdén, M.; Holmin, S.;</p><p>Svenningsson, P.; et al. The EXPANd Trial: Effects of Exercise and Exploring Neuroplastic Changes in People with Parkinson’s</p><p>Disease: A Study Protocol for a Double-Blinded Randomized Controlled Trial. BMC Neurol. 2019, 19, 280. [CrossRef]</p><p>314. Garzón, B.; Lövdén, M.; de Boer, L.; Axelsson, J.; Riklund, K.; Bäckman, L.; Nyberg, L.; Guitart-Masip, M. Role of Dopamine</p><p>and Gray Matter Density in Aging Effects and Individual Differences of Functional Connectomes. Brain Struct. Funct.</p><p>2021, 226, 743–758. [CrossRef]</p><p>315. Katz, M.J.; Fratiglioni, L.; Wang, H.X.; Frisoni, G.B.; Bäckman, L. Maintaining Cognitive Health in Aging: The Role of Cognitive</p><p>Reserve. Alzheimers. Res. Ther. 2021, 13, 1–11.</p><p>316. Gavelin, H.M.; Dong, C.; Minkov, R.; Bahar-Fuchs, A.; Ellis, K.A.; Lautenschlager, N.T.; Mellow, M.L.; Wade, A.T.; Smith, A.E.;</p><p>Finke, C.; et al. Combined physical and cognitive training for older adults with and without cognitive impairment: A systematic</p><p>review and network meta-analysis of randomized controlled trials. Ageing Res Rev. 2021, 66, 101232. [CrossRef]</p><p>317. Duzel, E.; van Praag, H.; Sendtner, M. Can Physical Exercise in Old Age Improve Memory and Hippocampal Function? Brain</p><p>2016, 139, 662–673. [CrossRef]</p><p>318. Erickson, K.I.; Kramer, A.F. Aerobic Exercise Effects on Cognitive and Neural Plasticity in Older Adults. Br. J. Sports Med.</p><p>2008, 43, 22–24. [CrossRef]</p><p>319. Vaynman, S.; Ying, Z.; Gomez-Pinilla, F. Hippocampal BDNF Mediates the Efficacy of Exercise on Synaptic Plasticity and</p><p>Cognition. Eur. J. Neurosci. 2004, 20, 2580–2590. [CrossRef]</p><p>320. Gutierrez, R.M.; Hidalgo-Lanussa, O.; Lattig, M.C.; Velez-Pardo, C. Exercise Intervention in Transgenic Alzheimer’s Rats Improves</p><p>Hippocampal GABAergic Transmission and Cognition. Behav. Brain Res. 2018, 346, 136–143.</p><p>321. Erickson, K.I.; Voss, M.W.; Prakash, R.S.; Basak, C.; Szabo, A.; Chaddock, L.; Kim, J.S.; Heo, S.; Alves, H.; White, S.M.; et al.</p><p>Exercise Training Increases Size of Hippocampus and Improves Memory. Proc. Natl. Acad. Sci. USA 2011, 108, 3017–3022.</p><p>[CrossRef]</p><p>322. van de Rest, O.; Berendsen, A.A.M.; Haveman-Nies, A.; de Groot, L.C.P.G.M. Dietary Patterns, Cognitive Decline, and Dementia:</p><p>A Systematic Review. Adv. Nutr. 2015, 6, 154–168. [CrossRef]</p><p>323. Heyn, P.; Abreu, B.C.; Ottenbacher, K.J. The Effects of Exercise Training on Elderly Persons with Cognitive Impairment and</p><p>Dementia: A Meta-Analysis. Arch. Phys. Med. Rehabil. 2004, 85, 1694–1704. [CrossRef]</p><p>324. Lautenschlager, N.T.; Cox, K.L.; Flicker, L.; Foster, J.K.; van Bockxmeer, F.M.; Xiao, J.; Greenop, K.R.; Almeida, O.P. Effect of</p><p>Physical Activity on Cognitive Function in Older Adults at Risk for Alzheimer Disease. JAMA 2008, 300, 1027. [CrossRef]</p><p>325. Smith, P.J.; Blumenthal, J.A.; Hoffman, B.M.; Cooper, H.; Strauman, T.A.; Welsh-Bohmer, K.; Browndyke, J.N.; Sherwood, A.</p><p>Aerobic Exercise and Neurocognitive Performance: A Meta-Analytic Review of Randomized Controlled Trials. Psychosom. Med.</p><p>2010, 72, 239–252. [CrossRef]</p><p>326. Northey, J.M.; Cherbuin, N.; Pumpa, K.L.; Smee, D.J.; Rattray, B. Exercise Interventions for Cognitive Function in Adults Older</p><p>than 50: A Systematic Review with Meta-Analysis. Br. J. Sports Med. 2018, 52, 154–160. [CrossRef]</p><p>327. McGregor, K.M.; Crosson, B.; Mammino, K.; Omar, J.; García, P.S.; Nocera, J.R. Influences of 12-Week Physical Activity Interven-</p><p>tions on TMS Measures of Cortical Network Inhibition and Upper Extremity Motor Performance in Older Adults—A Feasibility</p><p>Study. Front. Aging Neurosci. 2018, 9, 422. [CrossRef]</p><p>328. Colcombe, S.J.; Erickson, K.I.; Scalf, P.E.; Kim, J.S.; Prakash, R.; McAuley, E.; Elavsky, S.; Marquez, D.X.; Hu, L.; Kramer, A.F.</p><p>Aerobic Exercise Training Increases Brain Volume in Aging Humans. J. Gerontol. Ser. A Biol. Sci. Med. Sci. 2006, 61, 1166–1170.</p><p>[CrossRef]</p><p>https://doi.org/10.1073/pnas.1921481118</p><p>https://doi.org/10.1016/j.bbr.2016.07.042</p><p>https://doi.org/10.1371/annotation/79c850e6-4494-4533-b68d-ef4eb72cd65a</p><p>https://doi.org/10.1002/jnr.22387</p><p>https://doi.org/10.1371/journal.pone.0075125</p><p>https://doi.org/10.1523/JNEUROSCI.3252-09.2009</p><p>https://doi.org/10.1016/j.neubiorev.2013.04.005</p><p>https://doi.org/10.1186/s12883-019-1520-2</p><p>https://doi.org/10.1007/s00429-020-02205-4</p><p>https://doi.org/10.1016/j.arr.2020.101232</p><p>https://doi.org/10.1093/brain/awv407</p><p>https://doi.org/10.1136/bjsm.2008.052498</p><p>https://doi.org/10.1111/j.1460-9568.2004.03720.x</p><p>https://doi.org/10.1073/pnas.1015950108</p><p>https://doi.org/10.3945/an.114.007617</p><p>https://doi.org/10.1016/j.apmr.2004.03.019</p><p>https://doi.org/10.1001/jama.300.9.1027</p><p>https://doi.org/10.1097/PSY.0b013e3181d14633</p><p>https://doi.org/10.1136/bjsports-2016-096587</p><p>https://doi.org/10.3389/fnagi.2017.00422</p><p>https://doi.org/10.1093/gerona/61.11.1166</p><p>Brain Sci. 2023, 13, 1610 31 of 32</p><p>329. Maillot, P.; Perrot, A.; Hartley, A. Effects of Interactive Physical-Activity Video-Game Training on Physical and Cognitive Function</p><p>in Older Adults. Psychol. Aging 2012, 27, 589–600. [CrossRef]</p><p>330. Yates, L.A.; Ziser, S.; Spector, A.; Orrell, M. Cognitive Leisure Activities and Future Risk of Cognitive Impairment and Dementia:</p><p>Systematic Review and Meta-Analysis. Int. Psychogeriatr. 2016, 28, 1791–1806. [CrossRef]</p><p>331. Park, D.C.; Bischof, G.N. The Aging Mind: Neuroplasticity in Response to Cognitive Training. Dialogues Clin. Neurosci. 2013, 15, 109–119.</p><p>[CrossRef]</p><p>332. Marioni, R.E.; Valenzuela, M.J.; van den Hout, A.; Brayne, C.; Matthews, F.E. Active Cognitive Lifestyle Is Associated with</p><p>Positive Cognitive Health Transitions and Compression of Morbidity from Age Sixty-Five. PLoS ONE 2012, 7, e50940. [CrossRef]</p><p>333. Verghese, J.; Lipton, R.B.; Katz, M.J.; Hall, C.B.; Derby, C.A.; Kuslansky, G.; Ambrose, A.F.; Sliwinski, M.; Buschke, H. Leisure</p><p>Activities and the Risk of Dementia in the Elderly. N. Engl. J. Med. 2003, 348, 2508–2516. [CrossRef]</p><p>334. Fabrigoule, C.; Letenneur, L.; Dartigues, J.F.; Zarrouk, M.; Commenges, D.; Barberger-Gateau, P. Social and Leisure Activities and</p><p>Risk of Dementia: A Prospective Longitudinal Study. J. Am. Geriatr. Soc. 1995, 43, 485–490. [CrossRef]</p><p>335. Holwerda, T.J.; Deeg, D.J.H.;</p><p>Beekman, A.T.F.; van Tilburg, T.G.; Stek, M.L.; Jonker, C.; Schoevers, R.A. Feelings of Loneliness, but</p><p>Not Social Isolation, Predict Dementia Onset: Results from the Amsterdam Study of the Elderly (AMSTEL). J. Neurol. Neurosurg.</p><p>Psychiatry 2014, 85, 135–142. [CrossRef]</p><p>336. Spreng, R.N.; Turner, G.R. Structural Covariance of the Default Network in Healthy and Pathological Aging. J. Neurosci.</p><p>2013, 33, 15226–15234. [CrossRef]</p><p>337. Zhang, Y.; Fu, S.; Ding, D.; Lutz, M.W.; Zeng, Y.; Yao, Y. Leisure Activities, APOE E4, and Cognitive Decline: A Longitudinal</p><p>Cohort Study. Front. Aging Neurosci. 2021, 13, 736201. [CrossRef]</p><p>338. Gomez-Pinilla, F. The Combined Effects of Exercise and Foods in Preventing Neurological and Cognitive Disorders. Prev. Med.</p><p>(Baltim.) 2011, 52, S75–S80. [CrossRef]</p><p>339. Morris, M.C.; Tangney, C.C.; Wang, Y.; Sacks, F.M.; Bennett, D.A.; Aggarwal, N.T. MIND Diet Associated with Reduced Incidence</p><p>of Alzheimer’s Disease. Alzheimers Dement. 2015, 11, 1007–1014. [CrossRef]</p><p>340. Tangney, C.C.; Kwasny, M.J.; Li, H.; Wilson, R.S.; Evans, D.A.; Morris, M.C. Adherence to a Mediterranean-Type Dietary Pattern</p><p>and Cognitive Decline in a Community Population. Am. J. Clin. Nutr. 2011, 93, 601–607. [CrossRef]</p><p>341. Innis, S.M. Dietary Omega 3 Fatty Acids and the Developing Brain. Brain Res. 2008, 1237, 35–43. [CrossRef]</p><p>342. Konagai, C.; Yanagimoto, K.; Hayamizu, K.; Han, L.; Tsuji, T.; Koga, Y. Effects of Krill Oil Containing N-3 Polyunsaturated Fatty</p><p>Acids in Phospholipid Form on Human Brain Function: A Randomized Controlled Trial in Healthy Elderly Volunteers. Clin.</p><p>Interv. Aging 2013, 8, 1247–1257. [CrossRef]</p><p>343. Witte, A.V.; Kerti, L.; Hermannstädter, H.M.; Fiebach, J.B.; Schreiber, S.J.; Schuchardt, J.P.; Hahn, A.; Flöel, A. Long-Chain Omega-3</p><p>Fatty Acids Improve Brain Function and Structure in Older Adults. Cereb. Cortex 2014, 24, 3059–3068. [CrossRef]</p><p>344. Dullemeijer, C.; Durga, J.; Brouwer, I.A.; van de Rest, O.; Kok, F.J.; Brummer, R.-J.M.; van Boxtel, M.P.; Verhoef, P. N−3 Fatty Acid</p><p>Proportions in Plasma and Cognitive Performance in Older Adults. Am. J. Clin. Nutr. 2007, 86, 1479–1485. [CrossRef]</p><p>345. Melzer, T.M.; Manosso, L.M.; Yau, S.; Gil-Mohapel, J.; Brocardo, P.S. In Pursuit of Healthy Aging: Effects of Nutrition on Brain</p><p>Function. Int. J. Mol. Sci. 2021, 22, 5026. [CrossRef]</p><p>346. Wu, L.; Sun, D.; Qu, X.; Zhang, R.; Wang, Y. Dietary Fat Intake and Risk of Cognitive Impairment or Dementia: A Systematic</p><p>Review and Meta-Analysis. Ann. Med. 2018, 50, 698–712.</p><p>347. Morris, M.C.; Evans, D.A.; Bienias, J.L.; Tangney, C.C.; Bennett, D.A.; Aggarwal, N.; Schneider, J.; Wilson, R.S. Dietary Fats and</p><p>the Risk of Incident Alzheimer Disease. Arch. Neurol. 2003, 60, 194. [CrossRef]</p><p>348. Solfrizzi, V.; Colacicco, A.M.; D’Introno, A.; Capurso, C.; Torres, F.; Rizzo, C.; Capurso, A.; Panza, F. Dietary Intake of Unsaturated</p><p>Fatty Acids and Age-Related Cognitive Decline: A 8.5-Year Follow-up of the Italian Longitudinal Study on Aging. Neurobiol.</p><p>Aging 2006, 27, 1694–1704. [CrossRef]</p><p>349. Mattson, M.P.; Longo, V.D.; Harvie, M. Impact of Intermittent Fasting on Health and Disease Processes. Ageing Res. Rev.</p><p>2017, 39, 46–58. [CrossRef]</p><p>350. Lee, J.; Duan, W.; Long, J.M.; Ingram, D.K.; Mattson, M.P. Dietary Restriction Increases the Number of Newly Generated Neural</p><p>Cells, and Induces BDNF Expression, in the Dentate Gyrus of Rats. J. Mol. Neurosci. 2000, 15, 99–108. [CrossRef]</p><p>351. Stranahan, A.M.; Lee, K.; Martin, B.; Maudsley, S.; Golden, E.; Cutler, R.G.; Mattson, M.P. Voluntary Exercise and Caloric</p><p>Restriction Enhance Hippocampal Dendritic Spine Density and BDNF Levels in Diabetic Mice. Hippocampus 2009, 19, 951–961.</p><p>[CrossRef]</p><p>352. Witte, A.V.; Fobker, M.; Gellner, R.; Knecht, S.; Flöel, A. Caloric Restriction Improves Memory in Elderly Humans. Proc. Natl.</p><p>Acad. Sci. USA 2009, 106, 1255–1260. [CrossRef]</p><p>353. Marshall, L.; Helgadóttir, H.; Mölle, M.; Born, J. Boosting Slow Oscillations during Sleep Potentiates Memory. Nature</p><p>2006, 444, 610–613. [CrossRef]</p><p>354. Stickgold, R. Sleep-Dependent Memory Consolidation. Nature 2005, 437, 1272–1278. [CrossRef]</p><p>355. Xie, L.; Kang, H.; Xu, Q.; Chen, M.J.; Liao, Y.; Thiyagarajan, M.; O’Donnell, J.; Christensen, D.J.; Nicholson, C.; Iliff, J.J.; et al. Sleep</p><p>Drives Metabolite Clearance from the Adult Brain. Science 2013, 342, 373–377. [CrossRef]</p><p>356. Cross, N.E.; Carrier, J.; Postuma, R.B.; Gosselin, N.; Kakinami, L.; Thompson, C.; Chouchou, F.; Dang-Vu, T.T. Association</p><p>between insomnia disorder and cognitive function in middle-aged and older adults: A cross-sectional analysis of the Canadian</p><p>Longitudinal Study on Aging. Sleep 2019, 42, zsz114. [CrossRef]</p><p>https://doi.org/10.1037/a0026268</p><p>https://doi.org/10.1017/S1041610216001137</p><p>https://doi.org/10.31887/DCNS.2013.15.1/dpark</p><p>https://doi.org/10.1371/journal.pone.0050940</p><p>https://doi.org/10.1056/NEJMoa022252</p><p>https://doi.org/10.1111/j.1532-5415.1995.tb06093.x</p><p>https://doi.org/10.1136/jnnp-2012-302755</p><p>https://doi.org/10.1523/JNEUROSCI.2261-13.2013</p><p>https://doi.org/10.3389/fnagi.2021.736201</p><p>https://doi.org/10.1016/j.ypmed.2011.01.023</p><p>https://doi.org/10.1016/j.jalz.2014.11.009</p><p>https://doi.org/10.3945/ajcn.110.007369</p><p>https://doi.org/10.1016/j.brainres.2008.08.078</p><p>https://doi.org/10.2147/CIA.S50349</p><p>https://doi.org/10.1093/cercor/bht163</p><p>https://doi.org/10.1093/ajcn/86.5.1479</p><p>https://doi.org/10.3390/ijms22095026</p><p>https://doi.org/10.1001/archneur.60.2.194</p><p>https://doi.org/10.1016/j.neurobiolaging.2005.09.026</p><p>https://doi.org/10.1016/j.arr.2016.10.005</p><p>https://doi.org/10.1385/JMN:15:2:99</p><p>https://doi.org/10.1002/hipo.20577</p><p>https://doi.org/10.1073/pnas.0808587106</p><p>https://doi.org/10.1038/nature05278</p><p>https://doi.org/10.1038/nature04286</p><p>https://doi.org/10.1126/science.1241224</p><p>https://doi.org/10.1093/sleep/zsz114</p><p>Brain Sci. 2023, 13, 1610 32 of 32</p><p>357. Lim, A.S.P.; Kowgier, M.; Yu, L.; Buchman, A.S.; Bennett, D.A. Sleep Fragmentation and the Risk of Incident Alzheimer’s Disease</p><p>and Cognitive Decline in Older Persons. Sleep 2013, 36, 1027–1032. [CrossRef]</p><p>358. Lucey, B.P.; Wisch, J.; Boerwinkle, A.H.; Landsness, E.C.; Toedebusch, C.D.; McLeland, J.S.; Butt, O.H.; Hassenstab, J.; Morris, J.C.;</p><p>Ances, B.M.; et al. Sleep and Longitudinal Cognitive Performance in Preclinical and Early Symptomatic Alzheimer’s Disease.</p><p>Brain 2021, 144, 2852–2862. [CrossRef]</p><p>359. National Sleep Foundation. National Sleep Foundation Recommends New Sleep Times; National Sleep Foundation: Washington,</p><p>DC, USA, 2022.</p><p>360. Luyster, F.S.; Strollo, P.J.; Zee, P.C.; Walsh, J.K. Sleep: A Health Imperative. Sleep 2012, 35, 727–734. [CrossRef]</p><p>361. Saper, C.B.; Chou, T.C.; Scammell, T.E. The Sleep Switch: Hypothalamic Control of Sleep and Wakefulness. Trends Neurosci.</p><p>2001, 24, 726–731. [CrossRef]</p><p>362. Wu, C.W.; van Gelderen, P.; Hanakawa, T.; Yaseen, M.A.; Duyn, J.H. Compensation after Stroke: Plasticity of Intrinsic Connectivity</p><p>Networks. Neuroimage 2021, 244, 118532.</p><p>363. Lövdén, M.; Schaefer, S.; Noack, H.; Kanowski, M.; Kaufmann, J.; Tempelmann, C.; Bodammer, N.C.; Kühn, S.; Heinze, H.-J.;</p><p>Lindenberger, U.; et al. Performance-Related Increases in Hippocampal N-Acetylaspartate (NAA) Induced by Spatial Navigation</p><p>Training Are Restricted to BDNF Val Homozygotes. Cereb. Cortex 2011, 21, 1435–1442. [CrossRef]</p><p>Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual</p><p>author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to</p><p>people or property resulting from any ideas, methods, instructions or products referred to in the content.</p><p>https://doi.org/10.5665/sleep.2802</p><p>https://doi.org/10.1093/brain/awab272</p><p>https://doi.org/10.5665/sleep.1846</p><p>https://doi.org/10.1016/S0166-2236(00)02002-6</p><p>https://doi.org/10.1093/cercor/bhq230</p><p>Introduction</p><p>Neuroplasticity</p><p>Structural Neuroplasticity</p><p>Developmental Neurogenesis and Synaptogenesis</p><p>Adult Neurogenesis</p><p>Functional Neuroplasticity</p><p>Neurodevelopment</p><p>and Neuroplasticity</p><p>Prenatal Stage (from Conception until Birth)</p><p>Infancy and Childhood</p><p>Adolescence</p><p>Adulthood</p><p>Prenatal Factors That Impact Neurodevelopment and Neuroplasticity</p><p>Postnatal Factors That Impact Neurodevelopment and Neuroplasticity</p><p>Sex Hormones and Neuroplasticity</p><p>Aging, Neurodegeneration, and Neuroplasticity</p><p>Physiological Aging</p><p>Neurodegeneration</p><p>Protein Aggregation in Neurodegeneration</p><p>Mitochondrial Dysfunction and Oxidative Stress in Neurodegeneration</p><p>Neuroinflammation in Neurodegeneration</p><p>Genetic and Environmental Factors in Neurodegeneration</p><p>Correlation between Aging, Neurodegeneration, and Neuroplasticity</p><p>Non-Pharmacologic and Non-Invasive Strategies to Promote Neuroplasticity during Aging</p><p>Physical Exercise</p><p>Cognitive Stimulation and Socialization</p><p>Diet and Caloric Restriction</p><p>Sleep Hygiene and Quality of Sleep</p><p>Conclusions</p><p>References</p><p>of plasticity but also has functional implications. The forma-</p><p>tion of new synapses or the strengthening of existing ones can enhance the communication</p><p>and transmission of signals among neurons, leading to functional changes in neuronal</p><p>circuits. These adaptations include two forms of synaptic plasticity, long-term potentiation</p><p>(LTP) and long-term depression (LTD), through which the strength of synaptic connections</p><p>between neurons can change in response to different patterns of neuronal activity, thus</p><p>contributing to memory formation, skill acquisition, and habituation [72]. According to this</p><p>hypothesis, learning or experiencing something new can strengthen synaptic connections.</p><p>This, in turn, increases the efficiency of synaptic neurotransmission, ultimately aiding in</p><p>memory consolidation and information recall [73,74].</p><p>The term LTP was first introduced in 1973 by Bliss and Lomo [75]. The authors</p><p>demonstrated the long-lasting increase of synaptic strength in the dentate gyrus of the</p><p>hippocampus following high-frequency stimulation. Later, in 1993, Bliss and Collingridge</p><p>expanded on these findings and discussed the potential implications of LTP in the context of</p><p>a cellular memory model [19]. LTP is one of the most well-studied mechanisms underlying</p><p>neuroplasticity and is a specific cellular and synaptic process in which the strength of</p><p>a synapse is increased, resulting in more efficient transmission of signals between the</p><p>presynaptic and the postsynaptic neurons. Mechanistically, LTP involves changes that</p><p>can last for an extended period (days, weeks, or even years) in the synaptic structure,</p><p>such as an increase in the size and shape of postsynaptic dendritic spines as well as the</p><p>increase in the area of postsynaptic density (PSD) and the number of neurotransmitter</p><p>receptors of the postsynaptic membrane in response to the calcium-dependent activation of</p><p>N-methyl-D-aspartate (NMDA) receptors [76]. LTP is often described using the principle</p><p>of Hebbian plasticity, which states that synapses that are repeatedly active at the same</p><p>time tend to strengthen their connections. This principle aligns with the idea that the</p><p>brain adapts to experiences and reinforces the neural pathways associated with those</p><p>experiences [77].</p><p>Brain Sci. 2023, 13, 1610 5 of 32</p><p>On the other hand, LTD is the process of decreasing synaptic strength, resulting in a</p><p>less efficient transmission of signals in response to the depolarization of the postsynaptic</p><p>neuron for an extended period. LTD helps maintain synaptic connections’ overall balance</p><p>and efficiency, which is essential for synaptic homeostasis [21]. Both LTP and LTD are</p><p>critical for the adaptative capabilities of the CNS and allow neural circuits to adjust their</p><p>connections and synaptic strength in response to experiences, cognitive function, memory</p><p>consolidation, and habituation [72,73].</p><p>In summary, functional and structural plasticity play an important role in brain func-</p><p>tion, and changes in neuroplasticity may be associated with diseases and disorders of the</p><p>CNS. It is worth noting that neuroplasticity is most robust during development, but it</p><p>persists throughout life. This fact has significant implications for understanding brain</p><p>function, recovery from brain injury, and potentially treating neurological and psychiatric</p><p>disorders.</p><p>3. Neurodevelopment and Neuroplasticity</p><p>The brain is arguably the most complex organ in the human body, and its devel-</p><p>opment is a continuous process that starts early in gestation and continues throughout</p><p>adulthood [78]. Neurodevelopment is a highly orchestrated process involving the pro-</p><p>liferation, migration, differentiation, and maturation of neurons and the formation and</p><p>refinement of synaptic connections between them [79]. Normal neurodevelopment is essen-</p><p>tial for the proper functioning of the nervous system, and any disruption in this process can</p><p>lead to a wide range of neurodevelopmental and neurological disorders, such as autism</p><p>spectrum disorder (ASD), attention-deficit/hyperactivity disorder (ADHD), schizophrenia,</p><p>and epilepsy [80]. The causes of these disorders are complex and multifactorial, involving</p><p>genetic and environmental factors that can affect brain development and function [7]. Stud-</p><p>ies have shown that neuroplasticity involves many aspects of brain development, including</p><p>forming and refining neural connections during early development and acquiring new</p><p>skills and abilities throughout life [81,82]. In addition, neuroplasticity plays a crucial role in</p><p>recovery from brain injury and stroke and in treating neurological and psychiatric disorders,</p><p>such as depression and anxiety [83,84].</p><p>On the other hand, neuroplasticity disruptions can adversely affect neurodevelopment.</p><p>Exposure to toxic substances during development, such as alcohol or drugs, can impair</p><p>neuroplasticity and disrupt normal brain development [85,86]. Similarly, experiences with</p><p>chronic stress or trauma can impair neuroplasticity and lead to permanent changes in brain</p><p>structure and function [87]. Understanding the role of neuroplasticity in normal brain</p><p>development is critical for identifying and addressing factors that can disrupt this process</p><p>and lead to neurodevelopmental and neurological disorders. This section discusses the</p><p>stages of normal neurodevelopment and the factors influencing this process.</p><p>3.1. Prenatal Stage (from Conception until Birth)</p><p>Neurodevelopment is a critical process that begins approximately two to three weeks</p><p>after conception with the formation of the neural tube [88]. Neurulation involves the folding</p><p>and fusion of the lateral ends of the neural plate and is a crucial stage in the formation</p><p>of the brain and spinal cord. Abnormalities during neurulation can result in neural tube</p><p>defects, such as spina bifida [89]. Following neurulation, neurogenesis and neuronal</p><p>migration occur, which are also critical processes in neurodevelopment. Neurogenesis</p><p>occurs primarily during the first trimester of pregnancy and in specific brain regions, such</p><p>as the ventricular zone [29]. Neuronal migration occurs during the second trimester of</p><p>pregnancy. It is essential for proper brain development, allowing neurons to form correct</p><p>connections with other neurons and establish functional neural circuits [90].</p><p>During neurogenesis and neural migration, environmental factors such as stress,</p><p>nutrition, alcohol exposure, and sensory input can influence the rate and direction of these</p><p>processes. For example, exposure to stress hormones during gestation has been shown to</p><p>Brain Sci. 2023, 13, 1610 6 of 32</p><p>alter the timing of neurogenesis and the migration of new neurons, leading to changes in</p><p>brain structure and function [91].</p><p>Another significant event during embryonic development is the division of the neural</p><p>tube into three primary brain vesicles, which give rise to the different structures of the</p><p>brain [92]. Microglia migrate into the developing brain during the early stages of ges-</p><p>tation, around 4–5 weeks after fertilization, and establish the pool of resident immune</p><p>cells [93]. Gliogenesis, which produces region- and subtype-specific glia, begins at 22 weeks</p><p>of gestation and continues throughout adulthood [94]. Glial cells also aid in the myelina-</p><p>tion of neurons at approximately 32 weeks of pregnancy, a process that continues after</p><p>birth and into adulthood [95]. At 18 weeks of gestation, the excess of cells is eliminated</p><p>through apoptosis, a form of programmed cell death, resulting in the refinement of cell</p><p>populations and ensuring proper development and synaptic connectivity in the mature</p><p>brain [96]. Synaptogenesis begins in utero at approximately 27 weeks of gestation but</p><p>predominantly occurs after birth, coinciding with the growth of dendrites and axons and</p><p>axonal myelination [38]. The pruning of excess synapses and dendritic processes continues</p><p>after birth, a necessary process for proper neural network formation, with disruptions of</p><p>this step being linked to various neurological disorders [97]. By the end of the prenatal pe-</p><p>riod, major</p><p>fiber pathways, such as the thalamocortical pathway, have been established [34].</p><p>These critical stages of brain development are essential for a healthy and functional brain.</p><p>Conversely, disruptions in fetal brain development have been linked to an increased risk</p><p>of psychiatric disorders, such as autism and schizophrenia [98]. The complexity of these</p><p>processes highlights the importance of proper prenatal care, including good nutrition and</p><p>avoiding harmful substances such as alcohol and drugs, which can adversely affect brain</p><p>development [85,99].</p><p>The developmental origins of the health and disease hypothesis (DOHaD) propose</p><p>that environmental exposure during early life (particularly during the prenatal period)</p><p>can permanently influence the long-term development of disease. The initial studies</p><p>addressing this relationship observed the association between gestational malnutrition</p><p>and the phenotypes of the offspring, as well as the risk of developing metabolic diseases</p><p>such as obesity, diabetes, and cardiovascular diseases later in life. In accordance with</p><p>this, later studies have identified epigenetic modifications in fetal DNA as a response to</p><p>environmental stimuli, which can permanently alter protein expression and phenotypes in</p><p>the offspring. Of note, some of the environmental factors causing epigenetic modifications</p><p>besides maternal nutrition include smoking, maternal stress, and infection [100,101].</p><p>During prenatal development, genetic information plays a crucial role in orchestrating</p><p>events that determine the formation and refinement of neural connections. A complex</p><p>sequence of guidance molecules instructs newly born cells on what type of neurons to</p><p>become and where to go, while genetically determined intrinsic neural activity instructs the</p><p>refinement of axonal projections, which are guided by molecular cues to their approximate</p><p>target area [102]. While these events are critical for proper brain development, prenatal dis-</p><p>ruptions can negatively affect neuroplasticity. For example, prenatal exposure to teratogenic</p><p>factors such as alcohol can interfere with glutamatergic and GABAergic neurotransmitter</p><p>function, destabilizing previously tentative synapses [103,104]. This interference can lead</p><p>to structurally different yet functionally viable circuits. Studies have also shown that</p><p>neuroplastic changes during the prenatal period can have long-lasting effects on brain</p><p>development and behavior. For example, maternal stress during pregnancy has been linked</p><p>to altered brain development and increased risk for behavioral disorders such as ADHD</p><p>and ASD in the offspring, negatively impacting cognitive and emotional outcomes later in</p><p>life and increasing the risk for mental health conditions in adulthood [105].</p><p>Interestingly, through non-invasive techniques such as fetal magnetoencephalography</p><p>(fMEG) that record neural responses to external stimuli like music, speech, and touch [106],</p><p>it has been shown that exposure to music during the prenatal period can enhance the</p><p>development of the auditory system and improve cognitive function later in life [107]. In</p><p>addition, studies have shown that the fetus can recognize and respond to familiar voices,</p><p>Brain Sci. 2023, 13, 1610 7 of 32</p><p>including the mother’s voice. This recognition can be attributed to the development of the</p><p>auditory system, which is functional by the 16th week of gestation [108].</p><p>3.2. Infancy and Childhood</p><p>After birth, the human brain undergoes refinement and reorganization, particularly</p><p>during sensitive and critical periods known as “windows of brain plasticity,” which are</p><p>most pronounced in early childhood but continue into adolescence and adulthood [38].</p><p>During this stage, the brain undergoes an extraordinary growth spurt, with neurons form-</p><p>ing connections at an astonishing rate. As the brain triples in size during the first two years</p><p>of life, it builds an immense network of neural circuits that enables the processing of sen-</p><p>sory information and the development of higher-order cognitive functions [109]. Studies</p><p>have shown that early experiences and environmental factors are essential in shaping the</p><p>developing brain. For example, language development begins early in life. It involves a</p><p>complex interplay of genetic and environmental factors [110] and is strongly influenced by</p><p>early exposure to language and sounds [111,112].</p><p>Another critical aspect of neurodevelopment in childhood (as well as adolescence;</p><p>see below) is the development of executive skills such as attention, working memory, and</p><p>self-control. These skills are important for academic achievement, social functioning, and</p><p>well-being [113]. The development of executive functioning skills is influenced by several</p><p>factors, including genetics, environmental factors, and experiences [114]. Brain neuroplas-</p><p>ticity during infancy is also essential for developing social and emotional skills. Infants</p><p>develop the ability to recognize and respond to emotional cues and to form secure bonds</p><p>with caregivers. These skills, critical for social and emotional development, are shaped by</p><p>early experiences and essential for later well-being [115]. The growth and development</p><p>of the brain during this period lays the foundation for later cognitive, social, and emo-</p><p>tional abilities. Therefore, providing infants and children with a nurturing and stimulating</p><p>environment that promotes and supports optimal brain development is essential.</p><p>Windows of opportunity are necessary for developing new skills; however, they</p><p>leave the brain vulnerable to the harmful effects of the environment. In the same way</p><p>that environmental stimuli lead to necessary and expected neuronal remodeling while</p><p>learning a new skill that will be important throughout life, adverse situations can potentially</p><p>result in maladaptive changes. The World Health Organization (WHO) has coined the</p><p>term early stress to refer to adverse situations that occur during childhood, from birth to</p><p>18 years of age. Early stress can include coping with conditions such as living in extreme</p><p>poverty, violence, abuse (physical, sexual, or psychological), neglect, and grief, among</p><p>other examples. According to the WHO, around one billion children and adolescents are</p><p>exposed to some type of early stress every year [116,117]. When a child experiences a form</p><p>of early stress, the hypothalamic–pituitary–adrenal (HPA) axis is activated, resulting in the</p><p>release of cortisol, which in turn activates the sympathetic nervous system, thus triggering</p><p>the fight-or-flight response as a defense mechanism [118].</p><p>However, when the stress response is chronic and exacerbated, it leads to adverse</p><p>effects, including deregulating the HPA axis. Indeed, the sustained release of high levels of</p><p>cortisol into the blood plasma can result in the disruption of the feedback loop that regulates</p><p>the HPA axis, causing resistance to cortisol and ultimately resulting in the damage of various</p><p>brain regions, including the hippocampus [119,120]. Indeed, when in excess, cortisol (or</p><p>corticosterone in rodents) causes damage to dendritic arborization, the morphology of</p><p>dendritic spines, and the synaptic integrity of hippocampal neurons. Male C57BL/6N mice</p><p>that were subjected to an environmental stress protocol (insufficient sawdust in the housing</p><p>box) between postnatal days 2 and 9 showed decreased arborization and lower density</p><p>of dendritic spines in pyramidal neurons in the CA3 region of the hippocampus [121].</p><p>Furthermore, exposure to early stress induces decreased neurogenesis in the dentate gyrus</p><p>of the hippocampus in C57 adult mice [122].</p><p>Notably, the impact of hyperactivation of the HPA axis on brain morphology and</p><p>behavior (including the development of self-destructive behaviors, less tolerance to adverse</p><p>Brain Sci. 2023, 13, 1610 8 of 32</p><p>everyday situations, greater vulnerability to substance abuse, and difficulties in interper-</p><p>sonal relationships) is well documented in the literature [123,124]. Indeed, reductions in</p><p>adult neurogenesis, dendritic arborization, and glucocorticoid receptor</p><p>density changes</p><p>have been observed in the hippocampus of adults who experienced early stress [124,125].</p><p>Furthermore, exposure to early stress has also been associated with decreased gray matter</p><p>and increased psychiatric disorders, including anxiety and depression [126]. Further-</p><p>more, studies have demonstrated the relationship between the experience of early stress</p><p>in childhood and the development of alcohol dependence later in life [127]. A survey of</p><p>3592 adults on their drinking habits and history of early stress showed that the average</p><p>age at which alcohol intake began was lower in people with traumatic experiences. While</p><p>most participants said they drink to socialize and feel good, around 10% said they use</p><p>alcohol to “deal with problems and stress” [128]. In addition, Pilowsky and collaborators</p><p>(2009) demonstrated an association between traumatic events occurring in childhood and</p><p>adolescence with a greater frequency of heavy episodic drinking and an early onset of</p><p>alcohol consumption, with data highlighting that experiencing two or more traumatic</p><p>events early in life increased the propensity for alcohol dependence in adulthood. The</p><p>chronicity and severity of these episodes were also associated with the risk of relapse in</p><p>female patients who were undergoing treatment for cocaine addiction [129,130].</p><p>3.3. Adolescence</p><p>As children grow into adolescence, their brains mature, accompanied by significant</p><p>cognitive, social, and emotional development. Indeed, neuroimaging studies have shown</p><p>that the adolescent brain undergoes considerable changes in the prefrontal cortex, which is</p><p>responsible for higher-order cognitive functions, such as decision making, impulse control,</p><p>attention, and working memory [131]. The prefrontal cortex is also involved in social and</p><p>emotional processing, and its development during adolescence is crucial for acquiring</p><p>social and emotional skills, including navigating complex social relationships, empathizing</p><p>with others, and regulating emotions. Indeed, the prefrontal cortex undergoes significant</p><p>changes in its structural and functional connectivity during adolescence, and these changes</p><p>have been related to improvements in social cognition and emotion regulation [132,133].</p><p>Moreover, recent studies have highlighted the role of the social brain network, which</p><p>includes regions such as the medial prefrontal cortex, the temporoparietal junction, and</p><p>the amygdala, in social and emotional processing during adolescence [134,135]. These</p><p>brain regions are involved in social cognition, empathy, and emotional regulation, and their</p><p>development during adolescence is crucial for social and emotional competence.</p><p>Understanding the neurobiological changes during adolescence is crucial for promot-</p><p>ing healthy brain development and preventing mental health conditions during this critical</p><p>period of life. One of the most significant changes during adolescence is synaptic pruning,</p><p>which involves the elimination of unnecessary synapses and neural connections. This</p><p>process makes the brain more efficient by reducing neural noise and enhancing information</p><p>processing [136]. Synaptic pruning occurs mainly in the prefrontal cortex but also affects</p><p>other brain regions, such as the hippocampus and amygdala [37].</p><p>3.4. Adulthood</p><p>In adulthood, the rate of neurodevelopment slows down significantly. However,</p><p>the brain retains the capacity to form new neurons and connections and adapt to new</p><p>experiences throughout life. One mechanism through which the brain can continue to</p><p>adapt is adult neurogenesis, which is thought to play an important role in learning and</p><p>memory, as well as in mood regulation and the stress response [11,53–55].</p><p>In addition to the hippocampus and the SVZ, additional brain regions have also</p><p>emerged as sites where adult neurogenesis can take place [12]. Indeed, animal studies</p><p>have delineated neurogenic loci encompassing the hypothalamus [137], striatum [138–140],</p><p>substantia nigra (SN) [141], cerebral cortex [142], and amygdala [143]. Evidence indicates</p><p>that the genesis of neurons in these newly identified neurogenic areas is attributable to</p><p>Brain Sci. 2023, 13, 1610 9 of 32</p><p>the migration of NSPCs, typically originating from the SVZ [140,143–147]. Concurrently,</p><p>some studies have challenged this notion and proposed the existence of endogenous pools</p><p>of NSPCs within these regions, capable of local replication and integration into neuronal</p><p>circuits [137,138,148,149].</p><p>The hypothalamus, one of the major regulatory centers in the brain, controls various</p><p>homeostatic processes, and hypothalamic neural stem cells (htNSCs) have been shown to in-</p><p>terfere with these processes. Indeed, the hypothalamic neurogenesis is thought to influence</p><p>metabolism and fat storage, as evidenced by multiple studies on the impacts of a high-fat</p><p>diet (HFD) in mice [46,150,151]. Furthermore, neurogenesis within the hypothalamus is</p><p>also thought to contribute to behavioral and sexual functions, as elucidated in studies</p><p>by Bernstein et al. (1993) [152], Fowler et al. (2002) [153], and Cheng et al. (2004) [154].</p><p>Additionally, emerging research suggests that neurogenesis in the hypothalamus undergoes</p><p>alterations during aging [155], prompting investigations into the potential implications of</p><p>age-related changes in hypothalamic neurogenesis on overall physiological homeostasis</p><p>and cognitive functions [156].</p><p>Of note, deficits in adult hippocampal neurogenesis (as well as other forms of struc-</p><p>tural and functional plasticity) have been implicated not only in normal aging [63], but</p><p>also in various psychiatric [69] and neurodegenerative [70,71] conditions. Conversely,</p><p>classic antidepressants such as monoamine oxidase inhibitors (MAOIs), tricyclic antide-</p><p>pressants, and selective serotonin reuptake inhibitors (SSRIs) have been shown to possess</p><p>pro-neurogenic properties, and these are thought to mediate, at least in part, their antide-</p><p>pressant effects [60,61,157,158]. More recently, ketamine, an anesthetic with antidepressant</p><p>properties, was also shown to increase adult hippocampal neurogenesis in rodents [159].</p><p>Other studies have explored the potential of environmental enrichment (such as exposing</p><p>animals to a stimulating environment with toys and social interactions) [8,14,160] and</p><p>physical exercise [160–162] in promoting adult hippocampal neurogenesis. These findings</p><p>highlight the exciting potential of pharmacologic and non-pharmacologic interventions in</p><p>promoting adult hippocampal neurogenesis and potentially improving human brain health</p><p>and cognitive function.</p><p>3.5. Prenatal Factors That Impact Neurodevelopment and Neuroplasticity</p><p>Several prenatal factors can significantly impact neurodevelopment, and multiple</p><p>lines of research have identified maternal nutrition, exposure to toxins (e.g., alcohol and</p><p>illicit drugs), and infection as crucial determinants. Insufficient nutrition during preg-</p><p>nancy can lead to low birth weight and impaired cognitive development [163]. Moreover,</p><p>maternal nutrition has been associated in preclinical and clinical research with altered</p><p>neuropsychiatric outcomes in the offspring [164,165]. Of note, insulin has been identified as</p><p>a critical modulator of neuronal network development during the early phases of life [166].</p><p>Studies have shown an association between impaired insulin signaling in the hippocam-</p><p>pus of adolescent and adult offspring of obese mice with impairments in hippocampal</p><p>gene expression, neurogenesis, and synaptic plasticity [167–170]. Furthermore, a recent</p><p>study in rodents also suggested that an appropriate maternal diet, especially fiber-rich,</p><p>could regulate and reverse these neurocognitive alterations [170]. One possible mechanism</p><p>underlying the effects of a maternal high-fat diet on these neurodevelopment outcomes</p><p>is related to altered Notch 1 signaling activation, which in turn is thought to inhibit the</p><p>proliferation and differentiation of neural progenitors [171]. In addition, maternal excess</p><p>salt intake has also been associated with changes in brain development and neural plas-</p><p>ticity in rodents, particularly concerning synaptic transmission and neuroplasticity in the</p><p>hippocampus [172].</p><p>Exposure to toxins, including alcohol, illicit drugs, and heavy metals such as lead,</p><p>significantly impacts the developing brain, potentially resulting in brain damage and devel-</p><p>opmental delays [173]. These toxic substances can enter the body through various sources,</p><p>including environmental pollution, contaminated food and water, and maternal substance</p><p>use. Alcohol exposure during pregnancy has been linked to fetal alcohol spectrum disor-</p><p>Brain Sci. 2023, 13, 1610 10 of 32</p><p>ders (FASDs), a range of neurodevelopmental and behavioral problems that can result in</p><p>lifelong disabilities and neurocognitive abnormalities [85,174].</p><p>Consumption of cannabis (marijuana) during the prenatal period has also been shown</p><p>to affect neurodevelopmental processes. Indeed, in utero exposure to ∆9-tetrahydrocannabinol</p><p>was associated with behavioral alterations in adolescent rats, including impairment in aver-</p><p>sive limbic memory, decreased instrumental learning, and increased alcohol</p><p>consumption [175]. Similarly, exposure to illicit drugs, including stimulants (such as</p><p>cocaine and methamphetamine) and opioids (such as heroin), can cause a range of adverse</p><p>effects, including congenital disabilities, low birth weight, and developmental delays [176].</p><p>Exposure to heavy metals can also cause irreversible damage to the developing brain,</p><p>leading to a range of cognitive, behavioral, and developmental problems [177]. Maternal</p><p>infections during pregnancy can also have severe consequences for fetal development.</p><p>For example, rubella infection during pregnancy can lead to congenital malformations,</p><p>including hearing loss and intellectual disabilities [178]. Although the association between</p><p>COVID-19 and congenital anomalies in babies conceived and born during the pandemic is</p><p>still unclear due to the lack of knowledge on fetal and perinatal complications following</p><p>COVID-19 infection, some studies have shown an increase in the rate of CNS congenital</p><p>anomalies during the pandemic [179–181]. The long-term neuroplastic changes caused by</p><p>COVID-19 infections are still unclear and require further investigation.</p><p>3.6. Postnatal Factors That Impact Neurodevelopment and Neuroplasticity</p><p>After birth, several postnatal factors can significantly influence neurodevelopment</p><p>and neuroplasticity. Nutrition, social interaction, and environmental factors can all affect</p><p>brain development during the postnatal period. Nutritional status is a critical determinant</p><p>of brain growth and development, particularly during the first 1000 days of life [182]. In</p><p>rodents, a high-fat diet during the postnatal period can negatively impact cognition and</p><p>synaptic plasticity and promote neuroinflammation and microglial activation [183]. For</p><p>example, ingestion of sweeteners, such as aspartame and sucralose, can induce changes</p><p>in behavior and neuroplasticity, including decreases in hippocampal neurogenesis and</p><p>BDNF levels in rats in a sex-dependent manner [184]. In humans, breastfeeding has been</p><p>associated with improved metabolic and neurocognitive health outcomes in infants [185],</p><p>effects that are thought to be mediated, at least in part, by the unique properties and</p><p>lipid composition of maternal milk [186]. In addition, several studies have shown that</p><p>inadequate nutrition during infancy and early childhood can lead to long-term cognitive</p><p>deficits, reduced academic achievement, and behavioral problems [187,188]. In contrast,</p><p>adequate nutrition during this period can promote optimal brain development, including</p><p>improved cognitive and behavioral outcomes [189,190].</p><p>As explained above (see Section 3.2), the early years of life are a period of significant</p><p>neuroplasticity, and experiences during this time can shape the structure and function</p><p>of the brain [191]. Social interaction and early childhood experiences, such as language</p><p>exposure, are critical for optimal brain development. Studies have shown that children</p><p>who experience high-quality early care and education, including language-rich environ-</p><p>ments, have better cognitive, social–emotional, and academic outcomes than those who</p><p>do not [192,193]. Conversely, lack of stimulation, social deprivation, and neglect can have</p><p>long-lasting adverse effects on brain development, including reduced cortical thickness</p><p>and gray matter volume [194] and increased risk of cognitive deficits and developmental</p><p>delays [195].</p><p>Life experience and environmental enrichment positively modulate behavior and neu-</p><p>roplasticity during critical periods and throughout life [36,196]. Environmental enrichment</p><p>during childhood, adolescence, and adulthood has been shown to promote neurogenesis</p><p>and affect the pattern of monosynaptic inputs in animal models [14,160,197]. Additionally,</p><p>environmental enrichment has been shown to improve spatial learning performance and</p><p>neuroplasticity while increasing hippocampal volume and BDNF levels in various animal</p><p>models [160,198]. In humans, evaluating the impact of environmental enrichment on brain</p><p>Brain Sci. 2023, 13, 1610 11 of 32</p><p>neuroplasticity and function is much more complex due to the number of potential con-</p><p>founding variables. However, a recent randomized control trial has assessed the effects</p><p>of environment and neurodevelopment and concluded that interventions that can reduce</p><p>poverty could promote changes in children’s brain function and the development of higher-</p><p>order cognitive skills. This study found that infant neuroplasticity was positively correlated</p><p>with economic status, which in turn is known to impact numerous socio-economic factors</p><p>that can influence a child’s well-being, including household income and expenses, type</p><p>and amount of work of the mother, parenting behavior, and overall family wellbeing and</p><p>stress [199]. Social interactions can also be considered a form of environmental enrichment</p><p>known to modulate neuroplasticity. In particular, maternal contact has been shown to im-</p><p>pact neurodevelopment and to have long-lasting effects on behavior. Appropriate maternal</p><p>care during the first postnatal week can promote life-long stress resilience in rodents [200].</p><p>In conclusion, neuroplasticity is a critical determinant of brain growth and develop-</p><p>ment not only prenatally but also during the postnatal period. Several prenatal factors,</p><p>such as maternal nutrition, exposure to toxins, and infection, can potentially result in or</p><p>contribute to the offspring’s cognitive, behavioral, and developmental problems. On the</p><p>other hand, an appropriate maternal diet, especially one rich in fiber, can help prevent neu-</p><p>rocognitive alterations caused by a maternal high-fat diet. The early years of postnatal life</p><p>are also particularly significant for brain development, as experiences during this time can</p><p>shape the structure and function of the brain. Adequate nutrition, language-rich environ-</p><p>ments, high-quality early care, education, and rewarding social interactions and stimulation</p><p>are critical for optimal brain development during this period. Environmental enrichment</p><p>during childhood and adolescence can increase neuroplasticity and improve cognitive</p><p>outcomes. In contrast, exposure to toxins, social deprivation, neglect, and harmful environ-</p><p>mental factors can have a long-lasting negative impact on brain development, potentially</p><p>leading to long-term cognitive deficits and behavioral and psychological disturbances.</p><p>3.7. Sex Hormones and Neuroplasticity</p><p>Sex hormones are now known to have widespread actions in both the male and female</p><p>brains, through mechanisms thought to involve both genomic and nongenomic receptors.</p><p>Indeed, many neural and behavioral functions are affected by sex hormones such as estro-</p><p>gens, including mood, cognitive function, blood pressure regulation, motor coordination,</p><p>pain, and opioid sensitivity [201]. Moreover, sex-specific differences have been reported</p><p>with regards to hippocampal-dependent cognition and neurogenesis, suggesting that sex</p><p>hormones are involved in these processes.</p><p>Indeed, estrogens have been shown to modulate</p><p>certain forms of spatial and contextual memory, as well as different forms of neuroplasticity</p><p>including neurogenesis, primarily in the adult female hippocampus [202,203].</p><p>Peripheral sex steroid hormones, including estrogens, progesterone, testosterone, and</p><p>other androgens, are able to cross the blood–brain barrier and reach the brain. Furthermore,</p><p>hippocampal neurons are capable of synthesizing sex steroids de novo from cholesterol,</p><p>since neural cells express all the enzymes required for the synthesis of estradiol and testos-</p><p>terone, the end products of sex steroidogenesis [204–209]. Regarding 17β-estradiol (E2)</p><p>in particular, its synthesis in hippocampal neurons is homeostatically controlled by Ca2+</p><p>transients and is regulated by the release of gonadotropin-releasing hormone (GnRH) [210].</p><p>Indeed, release of GnRH from GnRH-positive neurons in the hippocampus is thought to</p><p>regulate the local synthesis of sex steroids in a sex-dependent manner and thus contribute</p><p>to the sexual differentiation of hippocampal neurons during the perinatal period [210,211].</p><p>Of note, a GnRH-induced increase in estradiol synthesis appears to provide a link between</p><p>the hypothalamus and the hippocampus, and this may underlie, at least in part, estrous</p><p>cyclicity of spine density in the female hippocampus [206,210]. Furthermore, sex hormones</p><p>can initiate gene transcription and activate signaling cascades by utilizing genomic and</p><p>non-genomic [201,212] mechanisms that play a key role in coordinating various physio-</p><p>logical and pathological neuroplasticity-related events, such as formation or remodeling</p><p>of dendritic spines, neurogenesis, synaptogenesis, and myelination modulation [213]. For</p><p>Brain Sci. 2023, 13, 1610 12 of 32</p><p>example, the study by Lu et al. in 2019, employing a forebrain-neuron-specific aromatase</p><p>knock-out mouse model, provided compelling genetic evidence of the involvement of</p><p>neuron-derived E2 in modulating AKT-ERK and CREB-BDNF signaling cascades. This</p><p>study established that neuron-derived E2 is essential for normal expression of LTP and</p><p>other forms of synaptic plasticity, as well as cognitive function in both male and female</p><p>brains [214]. However, it has also been proposed that whereas E2 appears to be essential</p><p>to maintaining synaptic transmission and synaptic connectivity in the female hippocam-</p><p>pus, dihydrotestosterone appears to be crucial for synaptic transmission and synaptic</p><p>connectivity in the male hippocampus [207,210,211]. As the expression of sex hormones</p><p>varies throughout the lifespan, its effects on neuroplasticity during distinct periods of</p><p>development, adulthood, and aging must also be considered. Along these lines, strategies</p><p>aimed at restoring and/or maintaining normal hormone levels in the brain throughout the</p><p>lifespan such as physical exercise are thought to be beneficial in promoting brain health in</p><p>general and neuroplasticity in particular [215].</p><p>4. Aging, Neurodegeneration, and Neuroplasticity</p><p>4.1. Physiological Aging</p><p>Aging is a natural process affecting various physiological systems, including the</p><p>immune, cardiovascular, musculoskeletal, and nervous systems [216]. According to the</p><p>United Nations, the proportion of people over 60 is projected to reach 25% by 2050 [217,218],</p><p>reflecting a global trend towards population aging. As the population ages worldwide, the</p><p>incidence of age-related diseases and the associated healthcare costs also increase.</p><p>The World Health Organization (WHO) defines healthy aging as the process of opti-</p><p>mizing opportunities for physical, mental, and social well-being to enable older individuals</p><p>to maintain their functional abilities that allow them to participate actively in society [219].</p><p>This definition encompasses not only the absence of disease but also emphasizes the im-</p><p>portance of maintaining functional capacity and engagement in activities that contribute</p><p>to a fulfilling life. For older adults, healthy aging involves healthy lifestyle habits and</p><p>behaviors, including healthy nutrition, regular physical activity, and avoiding smoking,</p><p>excessive drinking, and illicit drugs.</p><p>Aging increases the risk of developing chronic diseases such as cardiovascular disease,</p><p>diabetes, and cancer. For example, type 2 diabetes is more prevalent in older adults,</p><p>with approximately one in four adults over the age of 65 being affected by this metabolic</p><p>disorder [220]. Cancer incidence also increases with age, with approximately 60% of</p><p>all cancer cases occurring in adults over the age of 65 [221]. Moreover, ischemic heart</p><p>disease, stroke, and chronic obstructive pulmonary disease (COPD) are common causes</p><p>of mortality in the elderly population [222]. Additionally, aging is characterized by a</p><p>decline in immune function linked to the accumulation of senescent cells, which secrete</p><p>pro-inflammatory cytokines and contribute to chronic inflammation, ultimately resulting</p><p>in tissue damage [223]. Indeed, aging is associated with cellular senescence, a state of</p><p>irreversible growth arrest, which can be induced by various stressors, such as oxidative</p><p>stress, resulting in the damage of cellular proteins, lipids, and DNA [224], as well as</p><p>telomere shortening [225].</p><p>One of the critical differences between physiological and pathological brain aging</p><p>relates to cognitive reserve and the maintenance of cognition in physiological aging versus</p><p>the presence of notable cognitive decline in pathological aging. Indeed, the intrinsic</p><p>capacity of the nervous system to regenerate and compensate for neuronal loss (i.e., the</p><p>“brain reserve”) may counteract neurodegeneration and its consequences [226]. Within this</p><p>scenario, environmental (e.g., lifestyle) and genetic factors influence the degree of resiliency</p><p>to aging, thus influencing how much the intrinsic “brain reserve” can help prevent age-</p><p>related neuronal damage and loss. Cognitive reserve depends on how efficiently the system</p><p>can tap into the brain reserve [227]. In animal studies, several intrinsic and extrinsic factors,</p><p>including sex, body weight, physical activity, sleep duration, and anxiety and stress, can all</p><p>affect the cognitive reserve and the onset of cognitive impairments with aging [228].</p><p>Brain Sci. 2023, 13, 1610 13 of 32</p><p>On the other hand, regulation of neuroinflammation and oxidative stress, as well as</p><p>maintenance of calcium homeostasis, can promote cellular resilience and neuronal circuit</p><p>adaptation and ultimately increase cognitive reserve [229–231]. Nevertheless, the aging</p><p>brain does experience significant structural and functional changes, particularly affecting</p><p>the hippocampus and cerebral cortex [63,232]. Depending on the cognitive reserve of the</p><p>brain, alterations in neurons and their connectivity may decrease cognitive function [63]</p><p>and increase the risk of developing age-related neurological disorders such as AD and</p><p>Parkinson’s disease (PD) [233–236].</p><p>In addition to affecting neuronal structure and function, aging has also been associated</p><p>with astrocytic dysfunction and subsequent synaptic disturbances. Indeed, in old animals,</p><p>deficiencies in astrocytic glutamate uptake have been related to synaptic plasticity im-</p><p>pairments and age-related cognitive decline. Furthermore, astrocytes’ role in maintaining</p><p>antioxidant defenses may also be compromised by aging, which can further contribute to</p><p>neurodegeneration [237].</p><p>In addition, microglia (immune cells of the brain) and oligodendrocytes (myelinating</p><p>cells of the CNS) are also affected by aging. Indeed, an age-related reduction in both white</p><p>matter volume, myelinization, and microglia activation has been observed in the aged</p><p>brain, with an impairment in microglia activation being associated with decreased neuro-</p><p>protection and a compromised process of synapse elimination [237]. Neuroinflammation</p><p>is a well-established contributor to age-related cognitive impairments and hippocampal</p><p>neurogenesis deficits. The production of inflammatory mediators, including cytokines,</p><p>interleukins, and neurotrophins, as well as the activation</p><p>of glia and other immune cells,</p><p>can have a direct impact on synaptic plasticity and neurogenesis and with that exacerbate</p><p>the processes associated not only with normal aging but also with neurodegenerative</p><p>disorders [238,239].</p><p>One of the most notable changes associated with aging is mitochondrial dysfunction.</p><p>With age, mitochondria become less efficient at producing adenosine triphosphate (ATP)</p><p>and more prone to producing toxic reactive oxygen species (ROS) [216], which in turn can</p><p>cause neuronal dysfunction and, ultimately, cell death. Dysregulation of mitochondrial</p><p>function contributes to the pathogenesis of age-related diseases, including neurodegen-</p><p>erative disorders. Indeed, mitochondrial dysfunction has been implicated in AD and</p><p>PD [240]. The accumulation of mitochondrial DNA mutations has been observed in aged</p><p>mice, and this has been linked to increased oxidative damage [241]. Intracellular Ca2+</p><p>dysregulation has also been implicated in age-related cognitive decline [242,243]. Indeed,</p><p>an increase in intracellular Ca2+ levels in dorsal hippocampus CA1 pyramidal neurons</p><p>has been shown to contribute to memory impairment in aged animals [244]. Furthermore,</p><p>reduced NMDA receptor activation and LTP can impair Ca2+-dependent synaptic plasticity</p><p>in old animals [245].</p><p>Interventions targeting the various physiological systems affected by aging may in-</p><p>crease the lifespan and improve the quality of life of older individuals. For example,</p><p>physical exercise has been shown to improve cardiovascular function, increase neuroplas-</p><p>ticity, and promote the production of anti-inflammatory cytokines, all of which can help</p><p>mitigate the adverse effects of aging on the body and the brain [246,247]. Furthermore,</p><p>promoting healthy lifestyle habits such as maintaining a nutritious diet, engaging in regular</p><p>physical exercise, and avoiding smoking and excessive alcohol consumption can also help</p><p>minimize the impact of aging on individuals and society [63,162]. Indeed, maintaining</p><p>functional capacity and improving well-being can enhance the quality of life for older</p><p>individuals and drastically reduce healthcare costs.</p><p>4.2. Neurodegeneration</p><p>Neurodegeneration is characterized by the progressive loss of neuronal structure and</p><p>function, leading to irreversible neuronal damage and cell death [248]. Neurodegeneration</p><p>underlies the development of several neurodegenerative diseases, including AD, PD, as</p><p>well as HD, and amyotrophic lateral sclerosis (ALS) [249–251]. Neurodegenerative diseases</p><p>Brain Sci. 2023, 13, 1610 14 of 32</p><p>affect millions worldwide, with AD and PD being the most common neurodegenerative</p><p>disorders. In the United States, as many as 6.2 million people may have AD, as reported by</p><p>the Alzheimer’s Disease Association in 2022 [252]. Similarly, nearly a million Americans</p><p>are living with PD, according to the Parkinson’s Foundation [253]. The incidence of these</p><p>disorders is expected to triple by 2050, highlighting the need for effective prevention and</p><p>treatment strategies [254].</p><p>Multiple factors are known to contribute to the neurodegenerative processes that</p><p>culminate in neuronal damage and, ultimately, cell death [238,255]. For example, protein</p><p>misfolding, aggregation, and deposition have long been recognized as neuropathological</p><p>hallmarks common to many neurodegenerative disorders, including AD, PD, HD, and</p><p>ALS [250,256–258]. Moreover, mitochondrial dysfunction (which can result from genetic</p><p>mutations, exposure to environmental toxins, as well as physiological aging) [259], ROS</p><p>generation and accumulation, and a consequent increase in oxidative stress [250,260,261]</p><p>have all been shown to play a role in the pathogenesis of several neurodegenerative</p><p>disorders, including AD, PD, and HD. In addition, neuroinflammation has also been</p><p>implicated in the development of neurodegenerative diseases [262–264]. Other factors</p><p>contributing to aging and neurodegeneration (and consequent neuroplasticity impairment)</p><p>include genetic and environmental factors [236].</p><p>4.2.1. Protein Aggregation in Neurodegeneration</p><p>Neurodegenerative diseases are characterized by the accumulation and aggregation of</p><p>disease-specific proteins such as beta-amyloid and tau in AD, alpha-synuclein in PD, and</p><p>mutant huntingtin in HD [265]. These proteins misfold and form toxic oligomers and fibrils</p><p>that interfere with normal cellular functions, eventually leading to cell death. This process is</p><p>known as protein aggregation. The formation of abnormal protein aggregates is believed to</p><p>arise from disturbances in the proteostasis network. This tightly regulated system ensures</p><p>proper protein folding, trafficking, and degradation under normal conditions [266]. Disrup-</p><p>tions to the proteostasis network can result from genetic mutations, aging, environmental</p><p>stimuli, or a combination of several factors. Such disruptions can trigger the accumulation</p><p>of misfolded proteins, formation of protein aggregates, increased neuroinflammation and</p><p>oxidative stress, and activation of apoptotic pathways, ultimately culminating in neuronal</p><p>death.</p><p>The endoplasmic reticulum (ER) is a critical component of the proteostasis network</p><p>and is involved in the folding and processing of proteins [267]. Disruptions to ER func-</p><p>tion can accumulate misfolded proteins and trigger ER stress, activating the unfolded</p><p>protein response (UPR) [268]. The UPR signaling pathway activates adaptive pathways</p><p>to improve protein folding and promote quality control mechanisms and degradative</p><p>pathways [269]. Recent studies have shown that the UPR plays a role in the pathogenesis</p><p>of neurodegenerative diseases. For example, in AD, the UPR is activated in response to</p><p>the accumulation of beta-amyloid [270], while in PD, it is activated in response to the</p><p>accumulation of alpha-synuclein [271]. However, if the UPR fails to restore proteostasis or</p><p>if the accumulation of misfolded proteins exceeds the capacity of the cellular machinery to</p><p>degrade them, the UPR can also activate apoptotic pathways, thus resulting in neuronal</p><p>death [266].</p><p>4.2.2. Mitochondrial Dysfunction and Oxidative Stress in Neurodegeneration</p><p>Oxidative stress and mutations in mitochondrial DNA contribute to aging and neu-</p><p>rodegenerative diseases [272]. Several studies have shown that mitochondrial DNA muta-</p><p>tions accumulate in the aging brain and are associated with cognitive decline [261,273,274].</p><p>Moreover, increased levels of ROS have been shown to induce mitochondrial DNA mu-</p><p>tations and damage the mitochondrial respiratory chain, leading to mitochondrial dys-</p><p>function and neuronal death [275–277]. Furthermore, recent studies have shown that the</p><p>crosstalk between oxidative stress and other pathological mechanisms, such as protein</p><p>Brain Sci. 2023, 13, 1610 15 of 32</p><p>misfolding and inflammation, can further exacerbate mitochondrial damage and neuronal</p><p>death [278].</p><p>An increasing number of disease-specific proteins have been found to interact with</p><p>mitochondria. For example, mutant huntingtin has been shown to disrupt mitochondrial</p><p>function and dynamics in HD [279]. Moreover, dysfunctional mitochondria may precipitate</p><p>AD since beta-amyloid disrupts mitochondrial function and impairs energy production</p><p>in brain cells [280,281]. PD is also associated with mitochondrial dysfunction, including</p><p>impaired mitochondrial dynamics and oxidative stress [282], and oxidative stress has</p><p>been linked to alpha-synuclein accumulation in dopaminergic neurons, a hallmark of the</p><p>disease [283]. Together, these studies suggest that mitochondrial dysfunction is involved in</p><p>the pathogenesis of multiple neurodegenerative disorders.</p><p>4.2.3. Neuroinflammation in Neurodegeneration</p><p>Neuroinflammation is a complex process mediated by microglia and astrocytes and is</p><p>thought to play a role in several neurodegenerative diseases [284]. Microglia can assume</p><p>phagocytic phenotypes and release inflammatory cytokines in response to various stimuli,</p><p>including protein aggregates and pathogens [285]. For example, protein aggregates, such</p><p>as beta-amyloid in AD and alpha-synuclein in</p><p>PD, can activate microglia and induce</p><p>chronic neuroinflammation, leading to neuronal dysfunction and death [286,287]. Moreover,</p><p>oligomeric aggregates of beta-amyloid, tau, and alpha-synuclein can initiate both glial</p><p>and neuronal inflammation by activating several inflammatory pathways [286,288,289]. In</p><p>agreement, a recent study has shown that induced peripheral inflammation can potentiate</p><p>the adverse effects of alpha-synuclein oligomers by exacerbating neuroinflammation and</p><p>cognitive deficits in a synucleinopathy mouse model [290].</p><p>In response to injury or inflammation, activated astrocytes (i.e., astrogliosis) can also</p><p>contribute to neuroinflammation by releasing cytokines and chemokines, further activating</p><p>microglia and perpetuating neuroinflammation [291]. In agreement, a recent study showed</p><p>that activated microglia can promote the activation of neurotoxic reactive astrocytes (A1</p><p>type), resulting in both neuron and oligodendrocyte death. Of note, reactive A1 astrocytes</p><p>have been reported in various neurodegenerative diseases, including AD, PD, HD, ALS,</p><p>and multiple sclerosis [287]. In animal models of AD, reactive astrocytes were associated</p><p>with atrophy of glial fibrillary acidic protein (GFAP)-positive cells and the presence of</p><p>amyloid deposits in brain regions such as the hippocampus [292].</p><p>4.2.4. Genetic and Environmental Factors in Neurodegeneration</p><p>Genetic and environmental factors can also contribute to neuroinflammation and</p><p>neurodegeneration [293]. For example, mutations in the triggering receptor expressed on</p><p>the myeloid cells 2 (TREM2) gene, expressed by microglia, have been linked to an increased</p><p>risk of AD and other neurodegenerative diseases [294]. Moreover, genome-wide association</p><p>studies have identified new genetic risk factors for AD and PD [295].</p><p>In addition, environmental factors such as exposure to toxins, infection, and traumatic</p><p>brain injury (TBI) have also been shown to increase the risk of various neurodegenerative</p><p>diseases [296–298]. For example, exposure to pesticides, heavy metals, and solvents has</p><p>been linked to increased incidence of AD and PD [296]. Viral infections, such as herpes</p><p>simplex virus type 1 and human herpes virus 6, have also been associated with the de-</p><p>velopment of AD [297]. Furthermore, TBI has been linked to the development of chronic</p><p>traumatic encephalopathy, a neurodegenerative disease commonly found in athletes and</p><p>military veterans [298].</p><p>4.3. Correlation between Aging, Neurodegeneration, and Neuroplasticity</p><p>Several lines of evidence have supported a strong correlation between aging, neu-</p><p>rodegeneration, and neuroplasticity. With age, both neurogenesis (i.e., the generation of</p><p>new neurons) and synaptic plasticity (i.e., the ability of neurons to form new connections)</p><p>decline [299]. These changes are thought to contribute to the development and progression</p><p>Brain Sci. 2023, 13, 1610 16 of 32</p><p>of neurodegenerative processes by impairing the ability of the brain to compensate for the</p><p>effects of physiological aging and/or incurred damage while maintaining normal function.</p><p>On the other hand, engaging in activities that promote neuroplasticity, such as learning new</p><p>skills or engaging in regular physical exercise, has been shown to help maintain cognitive</p><p>function and slow cognitive decline in older adults [162,300]. These findings suggest that</p><p>interventions designed to enhance neuroplasticity may slow or potentially reverse the</p><p>effects of neurodegeneration in older adults.</p><p>The hippocampus and hippocampal neuroplasticity are particularly affected by the</p><p>aging process. Age-related changes in the hippocampus, such as increased oxidative stress,</p><p>neuroinflammation, altered gene expression, hormone imbalance, reduced neurogenesis,</p><p>and impaired synaptic plasticity, have all been associated with cognitive decline [63].</p><p>Indeed, several studies have shown that aging is associated with decreased synaptic</p><p>plasticity, including LTP and LTD, which are thought to underlie learning and memory. For</p><p>example, hippocampal LTP was reduced in older adults, and this decrease was associated</p><p>with poorer memory performance [301]. Of note, the decline in hippocampal LTP observed</p><p>in aging rats was also shown to be associated with a decrease in the expression of estrogen</p><p>receptors in the hippocampus [302]. Furthermore, aged animals were shown to have</p><p>decreased synaptic plasticity in the projections from the entorhinal cortex to the dentate</p><p>gyrus [303] and reduced neuronal excitability of CA1 pyramidal neurons [304]. Conversely,</p><p>improving cyclic AMP response element-binding protein (CREB) signalizing enhanced</p><p>cognitive performance in aged animals [305].</p><p>In addition, aging has also been associated with a decline in structural plasticity</p><p>throughout the brain, including the hippocampus. Indeed, aging is associated with re-</p><p>duced dendritic arborization and length, spine density [306–308], as well as decreased</p><p>synaptogenesis (i.e., formation of new synapses) [299]. Furthermore, adult neurogenesis</p><p>also dramatically declines with age, with the proportion of neuronal stem cells that survive</p><p>to become mature neurons being significantly reduced in the aged brain [309,310]. Of</p><p>note, such decreases have been shown to impact learning strategies in aged mice [310].</p><p>In humans, neuroimaging studies have shown that aging is associated with a decrease</p><p>in gray matter volume and cortical thickness, which may reflect a reduction in synaptic</p><p>density [311].</p><p>In summary, the decline in neuroplasticity accompanying aging has important im-</p><p>plications for cognitive function and the risk of developing neurodegenerative diseases.</p><p>Understanding the mechanisms underlying the age-related decline in neuroplasticity is</p><p>crucial for developing strategies to promote healthy aging and reduce the risk of age-</p><p>related neurological disorders. To this end, non-invasive strategies aimed at improving</p><p>neuroplasticity, including physical exercise, cognitive stimulation, social engagement, and</p><p>dietary interventions, hold promise in halting the course of neurodegeneration in the aging</p><p>brain [162,215,312,313].</p><p>4.4. Non-Pharmacologic and Non-Invasive Strategies to Promote Neuroplasticity during Aging</p><p>As described above, molecular and structural changes within the brain with aging</p><p>can contribute to a decline in brain function and neurodegeneration [314]. However, the</p><p>cognitive reserve (i.e., the ability of the brain to cope with damage and deterioration)</p><p>can significantly reduce the risk of dementia and other age-related neurodegenerative</p><p>conditions. Various non-invasive and non-pharmacological approaches have been shown</p><p>to increase the cognitive reserve and potentially counteract the deleterious effects of aging</p><p>by protecting the brain against age-associated neurodegenerative processes [315]. These</p><p>strategies, including physical exercise, environmental enrichment and social stimulation, a</p><p>healthy diet, and caloric restriction, as well as sleep hygiene, have been shown to enhance</p><p>brain plasticity and improve cognitive function in aging individuals [316] while also</p><p>counteracting several age-induced alterations in brain signaling, structure, and function [63].</p><p>The following sections outline some of the non-invasive and non-pharmacological strategies</p><p>proposed to increase neuroplasticity in the aging brain.</p><p>Brain Sci. 2023, 13, 1610 17 of 32</p><p>4.4.1. Physical Exercise</p><p>Physical exercise is a well-established, non-invasive strategy for promoting neuroplas-</p><p>ticity during aging [317,318]. Exercise is known to increase the production of growth factors,</p><p>such as BDNF, which promote the survival and growth of neurons and synapses and play</p><p>a key role in neuroplasticity [319]. In addition, exercise has also been shown to increase the</p><p>expression of synapsin-I, a presynaptic protein related to motor performance [320].</p><p>Various studies have now shown the beneficial effects of exercise in maintaining cog-</p><p>nitive function in aging [162,318,321,322]. Indeed, physical exercise increased hippocampal</p><p>volume, improved spatial memory in older</p>
  • Pós graduação Intervenção ABA para Autismo e Deficiência Intelectual A Disciplina 11 Módulo 1 Processos de Ensino I Disciplina 11 Módulo 1 Teste da Aula 3
  • Pós graduação Intervenção ABA para Autismo e Deficiência Intelectual A Disciplina 11 Módulo 1 Processos de Ensino Módulo 1 Teste da Aula 2
  • Considerando o enunciado acima e os estudos na disciplina de Desenvolvimento e aprendizagem motora, explique os conceitos das categorias de movimento de estabilidade, locomoção, manipulação e apresent
  • Considerando o enunciado acima e os estudos na disciplina de Desenvolvimento e aprendizagem motora, explique os conceitos das categorias de movimento de estabilidade, locomoção, manipulação e apresent
  • Considerando o enunciado acima e os estudos na disciplina de Desenvolvimento e aprendizagem motora, explique os conceitos das categorias de movimento de estabilidade, locomoção, manipulação e apresent
  • Considerando o enunciado acima e os estudos na disciplina de Desenvolvimento e aprendizagem motora, explique os conceitos das categorias de movimento de estabilidade, locomoção, manipulação e apresent
  • Considerando o enunciado acima e os estudos na disciplina de Desenvolvimento e aprendizagem motora, explique os conceitos das categorias de movimento de estabilidade, locomoção, manipulação e apresent
  • Considerando o enunciado acima e os estudos na disciplina de Desenvolvimento e aprendizagem motora, explique os conceitos das categorias de movimento de estabilidade, locomoção, manipulação e apresent
  • Considerando o enunciado acima e os estudos na disciplina de Desenvolvimento e aprendizagem motora, explique os conceitos das categorias de movimento de estabilidade, locomoção, manipulação e apresent
  • Considerando o enunciado acima e os estudos na disciplina de Desenvolvimento e aprendizagem motora, explique os conceitos das categorias de movimento de estabilidade, locomoção, manipulação e apresent
  • O estudo das mudanças no comportamento dos movimentos ao longo dos ciclos da vida, é uma das formas de observar o desenvolvimento motor. Neste sentido, o modelo heurístico da ampulheta de Gallahue, no
  • O estudo das mudanças no comportamento dos movimentos ao longo dos ciclos da vida, é uma das formas de observar o desenvolvimento motor. Neste sentido, o modelo heurístico da ampulheta de Gallahue, no
  • O estudo das mudanças no comportamento dos movimentos ao longo dos ciclos da vida, é uma das formas de observar o desenvolvimento motor. Neste sentido, o modelo heurístico da ampulheta de Gallahue, no
  • É possível destacar três funções executivas centrais: a inibição, que inclui o controle inibitório e o controle de interferências, a memória operac...
  • Como as funções executivas estão relacionadas ao desempenho em compreensão de leitura? A) As funções executivas não têm impacto no desempenho em c...
  • As funções executivas são um conjunto de habilidades necessárias para o controle e autorregulação do comportamento humano. Diamond (2013) divide as...
  • Leia as afirmativas abaixo, e assinale (V) para as verdadeiras e (F) para as falsas: ( ) Há níveis de consciência silábica; consciência intra-siláb...
  • Sobre os fatores complicadores para entendimento da relação simbólica, analise as sentenças apresentadas na e assinale a alternativa correta: I- Ap...
  • Invenção é o processo de criar algo novo que antes não existia, Pode ser um produto, um dispositivo, um método, uma técnica ou até mesmo uma ideia....
  • Enumere a segunda coluna de acordo com a primeira. I. Astrócitos. II. Oligodendrócitos. III. Micróglia. IV. Células ependimárias. A. Atua c...
  • A nalise as seguintes questões e marque a alternativa CORRETA: I. A memória é o processo em virtude do qual se associam coisas ou eventos no mu...
  • uais sao os dois principais plexos que estao envolvidos nos reflexos peristaltismo
  • quais sao os dois principais plexos que estao envolvidos nos reflexos peristaltismo
  • O que é fundamental que ocorra para que os indivíduos aprendam a ler? A) Reciclagem dos neurônios. B) Sinapse nervosa. C) Reconhecer os textos ...
  • Qual é a importância do estudo da Neuroanatomia Humana na sua formação acadêmica
  • Além da exploração dos sistemas e funcionamento do cérebro, ainda há sua abordagem do papel da cultura no desenvolvimento psicológico humano (HAZIN...
  • Assinale a alternativa correta sobre fatores de crescimento: a. PDGF-αα, αβ, ββ FATOR DE CRESCIMENTO DERIVADO DA PLAQUETA •      Quimiotático...
  • 10. (ANPEC 1996, Questão 5) Seja X uma variável aleatória com função de densidade  xf.(0) Se     bax abaxa b dx bax x     ln 1 222 , c...
Exploring the Role of Neuroplasticity - Neurociência (2025)
Top Articles
Latest Posts
Recommended Articles
Article information

Author: Arielle Torp

Last Updated:

Views: 6117

Rating: 4 / 5 (61 voted)

Reviews: 92% of readers found this page helpful

Author information

Name: Arielle Torp

Birthday: 1997-09-20

Address: 87313 Erdman Vista, North Dustinborough, WA 37563

Phone: +97216742823598

Job: Central Technology Officer

Hobby: Taekwondo, Macrame, Foreign language learning, Kite flying, Cooking, Skiing, Computer programming

Introduction: My name is Arielle Torp, I am a comfortable, kind, zealous, lovely, jolly, colorful, adventurous person who loves writing and wants to share my knowledge and understanding with you.