Barrier–environment interactions along the gut–brain axis and their influence on cognition and behaviour throughout the lifespan ==================================================================================================================================== * Sam E.J. Paton * José L. Solano * François Coulombe-Rozon * Manon Lebel * Caroline Menard ## Abstract Environment is known to substantially alter mental state and behaviour across the lifespan. Biological barriers such as the blood–brain barrier (BBB) and gut barrier (GB) are major hubs for communication of environmental information. Alterations in the structural, social and motor environment at different stages of life can influence function of the BBB and GB and their integrity to exert behavioural consequences. Importantly, each of these environmental components is associated with a distinct immune profile, glucocorticoid response and gut microbiome composition, creating unique effects on the BBB and GB. These barrier–environment interactions are sensitive to change throughout life, and positive or negative alterations at critical stages of development can exert long-lasting cognitive and behavioural consequences. Furthermore, because loss of barrier integrity is implicated in pathogenesis of mental disorders, the pathways of environmental influence represent important areas for understanding these diseases. Positive environments can be protective against stress- and age-related damage, raising the possibility of novel pharmacological targets. This review summarizes known mechanisms of environmental influence — such as social interactions, structural complexity and physical exercise — on barrier composition, morphology and development, and considers the outcomes and implications of these interactions in the context of psychiatric disorders. ## Environment, barriers and the brain Every organism is shaped by their surroundings through constant perception, consumption and physical engagement with environmental features such as chemical makeup, social targets, structural complexity and climate.1,2 All of these environmental components interact with the genome to influence phenotype and, thus, cognitive ability and behaviour, throughout development and aging. The idea that the environment could permanently affect the adult brain was popularized by Donald O. Hebb, who found that rats living in his home as pets performed much better on memory tasks than those raised in laboratory cages.3 Later work, especially from the laboratory of Mark Rosensweig, was crucial in deciphering the neurobiological underpinnings of this phenomenon; 4,5 today, we know that environmental conditions affect both neuronal plasticity and functional connectivity, as well as genesis of new neurons, glia and cerebral blood vessels.6–10 But the question remains, how exactly is external information translated into lasting effects on the brain? Although activity-dependent changes in neuronal plasticity offer an enticing and simplistic mechanism, evidence increasingly supports a role for changes to the periphery including the immune system, microbiome and glucocorticoid responses, which occur independently from the brain and yet are indispensable for some of the cognitive and behavioural consequences of environmental change.11–16 These systemic pathways for brain–environment communication, which remain poorly characterized, rely heavily on communication across specialized biological barriers, including the blood–brain barrier (BBB) and gut barrier (GB). The BBB and GB are conserved, highly specialized frontiers that protect the body from external disturbances while maintaining communication between anatomic compartments. The BBB, a dynamic endothelial membrane lining the lumen of blood vessels in the brain, is essential to cerebral homeostasis, supplying necessary nutrients and metabolites to neurons and glia while preventing entry of harmful substances from the bloodstream.17 The BBB is supported by an intricate cell network surrounding cerebral blood vessels termed the neurovascular unit, mainly comprising astrocytes, mural cells (including smooth muscle cells and pericytes), microglia and brain endothelial cells (Figure 1).18–20 The neurovascular unit performs many crucial functions including regulating BBB development and maintenance, and facilitating coupling between neuronal activity and cerebral blood flow.21 Specific BBB properties are largely conferred by brain endothelial cells that possess several unique features, including a tightly regulated transporter profile, restricted expression of adhesion molecules and a specialized tight junction architecture between neighbouring cells (Figure 1).17,22 Importantly, this composition is highly sensitive to circulating signals from the periphery, including cytokines and immune cells, glucocorticoid levels and gut-derived bacterial metabolites.23 ![Fig. 1](http://jpn.ca/https://www.jpn.ca/content/jpn/48/3/E190/F1.medium.gif) [Fig. 1](http://jpn.ca/content/48/3/E190/F1) Fig. 1 Biological barriers are key interfaces between environment and the body. The blood–brain barrier (BBB, inner left) is a dynamic and highly selective membrane between the endothelial cells that line the blood vessels of the brain. Specific barrier properties are conferred by specialized tight junction proteins (e.g., claudin-5) and by occludins anchored to the cytoskeleton by scaffold proteins, including zonula occludens (ZO). The BBB is encircled by an intricate network of cells termed the neurovascular unit, mainly comprising the following: astrocytes, which facilitate neuronal communication with blood vessels; mural cells including smooth muscle cells (on arteries and arterioles) and pericytes, which contribute to contractile properties and morphology of vessels; and microglia, the resident immune cells of the brain. Meanwhile, the gut barrier (GB, inner right) is a layer of epithelial cells termed enterocytes lining the lumen of the intestine. These cells are connected by a unique network of tight junctions — particularly claudins, as well as desmosomes and gap junctions — that regulate passage of substances from the gut lumen to the body. The GB is in close contact with the intestinal microbiome and is sensitive to changes in bacterial function. In addition, the GB communicates with enteric neurons, blood vessels, mast cells and immune cells, making it a key signalling hub. Both of these barriers mediate communication between the environment and the body in humans and in rodent models. Distinct environmental components — such as social interactions, structural complexity and locomotor activity — which can be translated from rodents (outer right) to humans (outer left), influence the barriers in specific ways to produce unique physiologic effects. VE = vascular endothelial. The GB — an epithelial membrane embedded in the mucosal layer of the gut lumen — plays a crucial role in homeostasis of the gastrointestinal tract. Given its close contact with the external environment, the GB is a converging centre for interactions between diet, microbiome, immune system and enteric efferent neurons (Figure 1).24,25 A brick-like tight junction pattern along the extended site of contact between gut epithelial cells closely regulates passage of fluids and metabolites from the gut lumen to the bloodstream (Figure 1).26 The GB interacts physically and chemically with intestinal microflora, which are sensitive to environmental changes and communicate with the body through metabolite levels, such as short-chain fatty acids (SCFAs), as well as microbial surface proteins, including peptidoglycans and lipopolysaccharide.27,28 Produced in varying amounts by specific bacterial strains, SCFAs and lipopolysaccharide are key messengers that influence cognition and behaviour through a communication network known as the microbiota–gut–brain axis.28,29 Despite their differences, the BBB and GB are both integral to survival throughout the lifespan. Indeed, each is required for normal embryonic development, beginning at early stages; loss of barrier integrity in later life is associated with cognitive and physiologic deficits of aging.30–34 Cooperation and communication between the BBB and the GB are therefore important aspects of normal physiology, mediated by a constant exchange of peripheral signals in response to environmental demands. Much of the environmental influence on cognition and behaviour throughout life is dependent on translation of external signals to the brain via the periphery. Importantly, the BBB and GB are not simple mediators of this communication, but rather active signalling hubs that sense and respond to relevant information through changes in receptor profile, gene expression and barrier permeability (Figure 2). The peripheral immune system is a major pathway for these interactions, as levels of circulating cytokines and leukocytes are highly sensitive to surrounding environment and fluctuate with conditions such as temperature and air pollution. 11,35 These produce a variety of barrier effects; for example, elevated levels of proinflammatory cytokines like tumour necrosis factor-α (TNF-α) and interleukin-6 (IL-6) suppress transcription of tight junction proteins and diminish BBB and GB integrity to allow infiltration of inflammatory mediators into the bloodstream and the brain.36–38 The microbiota–gut–brain axis provides an alternative avenue for interaction, where microbial metabolites such as SCFAs, which vary with environmental conditions, can influence intestinal barrier permeability, as well as diffuse across the GB into the circulation to influence leukocyte count, BBB integrity and neuronal activity.28,39 Meanwhile, lipopolysaccharide leaks into the bloodstream via a paracellular route between epithelial cells, where it forms a complex with circulating soluble binding proteins (e.g., lipopolysaccharide binding protein), which can alter the BBB and cognitive function.27,36 Finally, the physiologic stress response via the hypothalamic–pituitary–adrenal (HPA) axis communicates with and across barriers through levels of circulating glucocorticoids. When perceived stress initiates HPA activation, steroids — such as cortisol (in humans) or corticosterone (in rodents) — are released into the blood, where they interact with barriers through their binding with 2 kinds of receptors, the mineralocorticoid and glucocorticoid receptors.40,41 The binding of these steroids in the barrier modifies their permeability and responsiveness to subsequent stimuli.42,43 ![Fig. 2](http://jpn.ca/https://www.jpn.ca/content/jpn/48/3/E190/F2.medium.gif) [Fig. 2](http://jpn.ca/content/48/3/E190/F2) Fig. 2 Pathways of environment–barrier communication. The blood–brain barrier (BBB) and gut barrier (GB) communicate with each other and with the environment through a complex network of interactions. The main avenues for signalling are the hypothalamic–pituitary–adrenal (HPA) axis (red), the immune system (blue) and the microbiota–gut–brain axis (yellow). Information received from the environment produces changes in neuronal activity or microbiome function that are sensed by the GB and BBB and are subsequently transduced into circulating signals, enabling the brain to be affected by shifts in the gut and vice versa. LPS = lipopolysaccharide, SCFA = short-chain fatty acid. These interactions take on a special importance in the context of disease, as growing evidence suggests that disruption of environment–barrier communication is an early step in pathogenesis of mood disorders. Several lines of evidence have recently come together to support this notion. Disruption of the BBB — measured with magnetic resonance imaging by injection of contrasting agents or in postmortem tissue with markers of BBB integrity — and the GB — inferred from blood serum markers of GB permeability — have been observed in both males and females with diverse psychiatric conditions including major depressive disorder (MDD), bipolar disorder and schizophrenia.36,38,44–47 In mice, chronic social or psychological stress is sufficient to induce depressive and anxiety-like symptoms via damage to the BBB and GB (Doney and colleagues25 provide a review of BBB and GB disruption in various rodent models of stress).36,38,48–50 Environmental stressors such as social isolation, low socioeconomic status or sedentary lifestyle have been associated with systemic inflammation and risk for developing psychiatric disorders in both rodents and humans.51–55 Meanwhile, positive environments can protect against disease; in humans, physical exercise, high socioeconomic status and social support decrease risk for psychiatric disorders,54,56,57 while enriched housing or access to a running wheel have strong antidepressant effects in mouse models of stress.58,59 Finally, several reports have suggested that the positive behavioural outcomes of enrichment could involve changes to the neurovasculature and gut epithelium.8,60–64 As current monoamine-focused antidepressant and anxiolytic medications are, unfortunately, not always effective,65 positive environmental modifications to the BBB and GB could offer new and attractive pharmacological targets for mood-related disorders. Although the BBB and GB are increasingly recognized for their major role in environmental influence on cognition, behaviour and emotion in health and disease, the literature on this subject is broad, confusing and, at times, contradictory. Therefore, we aim to summarize known mechanisms of environmental influence on barrier activity, morphology and development, as well as to consider the outcomes and implications of these interactions in the context of psychiatric disorders to identify directions for future research. ## Distinct environmental features differentially modulate biological barriers An organism’s environment is extraordinarily complex, with many diverse aspects that can combine or cancel out as they interact with the genome to shape behaviour and cognitive function. Investigation of these relationships in a laboratory setting must strike a balance between feasibility and ethological validity. The most common means of studying neural consequences of various external conditions in the laboratory is the addition of extra factors to the home cage, a paradigm generally referred to as environmental enrichment.2,5 However, what enrichment actually means has been subject to various interpretations over the years. Cage layouts in these experiments tend to vary greatly among laboratories, and the literature is replete with a heterogeneous mix of enrichment paradigms including larger cages, multiple levels, cage mates, running wheels, shelters, toys, enhanced diet and added bedding, often combined.66 The consensus definition of enrichment as “a combination of complex inanimate and social stimulation”2 suggests an interweaving of factors that are not easily separated. However, it is increasingly apparent that distinct environmental components have unique effects on the brain and, therefore, their effects should be studied independently. 62,67–72 Here, we adopt a conventional division of 3 main facets of environmental enrichment, namely social interactions, structural complexity and physical exercise.1,2 This division is attractive because it allows extrapolation of findings between human and rodent studies, thereby enhancing translation between clinical and preclinical reports73 (Figure 1). We consider the interactions of each environmental component with the BBB and GB. ### Social interactions Humans are inherently social animals, and interaction with other individuals is a fundamental necessity, as much for emotional well-being as for normal development and physiology of the brain and neural circuits.56,74,75 Similarly, rodents participate in a complex array of social relationships that range from sexual interactions and family bonding to aggression and dominance hierarchies.76 Given that social encounters play such an important role in our lives, isolation is consequently linked to a host of unfavourable outcomes including risk of depression, cognitive decline, cardiovascular dysfunction, systemic inflammation and immune dysregulation. 56,74,77 Indeed, both social isolation and chronic social defeat stress disrupt neuronal reward circuits in the striatum of male and female mice.78,79 Evaluation of social interactions in humans is generally based on self-reported questionnaires, and tends to focus on subjective rather than objective measures of socialization. Far from being a shortfall of these studies, reports have suggested that physiologic effects are driven largely by perceived rather than actual social support or strain.77,80 In rodents, common manipulations include social stress through isolation or social defeat, while dominance in hierarchies is often assessed in pairs (for instance, with the tube test).38,81–83 Recently, advances in machine learning have enabled detailed tracking and analysis of maternal care and complex social interactions in animals.84,85 The physiologic outcomes of socialization extend far beyond the brain. In humans, for example, social support can buffer a stress-induced increase in concentrations of circulating glucocorticoids.86 Further, social interactions influence markers of systemic inflammation, with reported support from family being associated with a decrease in IL-6, while family strain increased serum levels of this cytokine. Support and strain from spousal relationships were associated with reduced or elevated levels of E-selectin, respectively.87 Interleukin-6 is a marker of systemic inflammation,38 and circulating E-selectin has been proposed as a marker of BBB permeability;49 thus, these findings suggest that positive social interactions can reduce immune response and promote barrier integrity. These fluctuations have also been observed in rodents; social instability in adolescent rats alters hippocampal gene expression and the composition of the gut microbiome, and social isolation in adult male CD-1 mice modifies the cytokine profile and behavioural response to immune challenge.88,89 Moreover, in social hierarchies among males, dominant CD-1 mice possess an immune profile and leukocyte transcriptional patterns that are distinct from submissive mice.90 These links between environmental, systemic and behavioural changes during social encounters imply communication of relevant information across the GB and BBB (Figure 3). ![Fig. 3](http://jpn.ca/https://www.jpn.ca/content/jpn/48/3/E190/F3.medium.gif) [Fig. 3](http://jpn.ca/content/48/3/E190/F3) Fig. 3 Mechanisms of environmental influence on the blood–brain and gut barriers. Social interaction, structural complexity and locomotor activity are distinct components of both the human and rodent environment that produce unique effects on the gut barrier, periphery and blood–brain barrier. AQP = aquaporin; Bdnf = brain-derived neurotrophic factor; bFgf = basic fibroblast growth factor; Cldn = claudin; CRH = corticotropin-releasing hormone; IL = interleukin; LPS = lipopolysaccharide; NF = nuclear factor; GSK = glycogen synthase kinase; HCAR = hydroxycarboxylic acid receptor; Ocln = occludin; SCFA = short-chain fatty acid; TNF = tumour necrosis factor; Vegf = vascular endothelial growth factor; ZO = zonula occludens. Correspondingly, social experiences affect, and are affected by, sex-specific properties of cells in the neurovascular unit. For example, microglia, which are highly sensitive to environmental factors, monitor the generation of new cells during development and contribute to sex-specific social tendencies via pruning of newborn astrocytes in mice.91,92 Social isolation, meanwhile, appears to induce astrocyte reactivity, as evidenced by a report of elevated expression of glial fibrillary astrocytic protein in the hippocampus of isolated mice.93 A further study suggests that isolation impairs astrocytic connectivity, reducing cell body volume and process complexity.94 Although it has not been directly studied, socialization-related adaptations in these cells likely modify function of the neurovascular unit and BBB integrity to influence behavioural changes.95 In brain endothelial cells, models of social stress, including social isolation and social defeat stress, lead to dysregulation of tight junction proteins and subsequent loss of BBB integrity in both male and female mice.38,49,81,83,96 Interestingly, this effect is dependent on timescale; in rats, short duration of isolation stress (7 d) produces deficits in the tight junction protein claudin-5, while longer durations (6 wk) actually increase mRNA expression of claudin-5 in the male hippocampus versus the female prefrontal cortex.81,83 Furthermore, targeted downregulation of claudin-5 at the BBB is sufficient to produce deficits in social interaction in male and female C57BL/6 mice.38,49 Although these findings hint at a connection between BBB integrity and social activity, the intricacies of this relationship remain unclear. Recent reports are helping to develop a mechanistic insight; for instance, dominant and submissive CD-1 mice display distinct immunological profiles, with dominance associated with a shift toward the adaptive immune system and, therefore, an increase in B-and T-cell activity.90 Active T cells can cross the BBB and infiltrate the brain to communicate information, and it is possible that the BBB mediates communication between the brain and immune system to influence social status. It is also likely that BBB phenotype is important for sex-specific behaviours. An elegant study involving *Drosophila* suggests that signalling through the moody receptor (a melatonin receptor homologue) in the BBB is required for normal male courtship behaviour.97 In humans, melatonin possesses antioxidant qualities that are important for maintaining BBB integrity, and is also involved in sex-specific sexual development, although a definitive link between the 2 functions remains to be proven.98 Finally, chronic social defeat stress increases expression of histone deacetylase-1, reducing histone acetylation and, thus, inactivating transcription of genes, including claudin-5, which has been proposed as a mechanism for development of stress-induced deficits in social behaviours.99 The gut is also strongly implicated in social interactions. In humans, HPA activation by an acute social stressor like public speech leads to a transient permeabilization of the GB, as measured by differential urinary excretion of ingested, nonmetabolized sugars.100 This effect can also be elicited by injection of corticotropin-releasing hormone and, in both cases, is attenuated by an ingestible inhibitor of enteric mast cell degranulation. Meanwhile, perceived social exclusion alters the composition of the gut microbiome, favouring species of the genus *Prevotella*, which has been previously linked with emotion perception and limbic system connectivity.29,101,102 Interestingly, in a group of rats with colon cancer, social interactions, but not environmentally complex housing, was found to modulate gastrointestinal morphology by increasing expression of occludin, a tight junction protein, and IL-10, an anti-inflammatory cytokine, in the intestinal mucosa and blood plasma.103 These changes can influence the brain as seen in prairie voles, for which socialization produces lasting enrichment in diversity, which correlates with changes in region-specific brain transcription and glial cell abundance.104 On the other hand, social stress from long-term social isolation alters blood pressure and volume in the small intestine among female rats, along with expression of BBB-related tight junctions.83 Furthermore, mice susceptible to social defeat stress are characterized by changes in gene expression of proinflammatory cytokines *IL1B* and *IL6* in the prefrontal cortex, in line with a higher abundance of *Bacteroidetes*, a genus related to gut dysbiosis. 105 A recent study suggests a mechanism for microbiome influence on social behaviour in which gut dysbiosis leads to diminished bacterial production of butyric acid — a prominent SCFA that normally diffuses across the GB and BBB to promote synaptic plasticity by inhibition of histone deacetylase 2 activity — thereby increasing transcriptional suppression and a subordinate state.106 These findings highlight that both the GB and BBB are important components in mediating communication between the brain, periphery and environment during social interactions. ### Structural complexity Structural enrichment is a paradigm stretching back to Donald O. Hebb and his pet rats.3 Novel and diverse environmental features offer a continual opportunity for informal learning and accelerate development of neural circuits and cerebral vascular networks.107,108 In humans, for example, high socioeconomic status is associated with better housing quality, leisure time and more opportunities for travel, leading to a greater frequency of novel experiences, compared with low socioeconomic status. Reproduction of this concept for preclinical studies with rodents varies widely between research groups, but generally involves larger caging, multiple levels in the environment, space for shelter, extra nesting material and manipulable objects or toys, which collectively provide an enhanced visual, olfactory and somatosensory landscape.2,66 Despite poor standardization and diversity of experimental setups, the behavioural and neural outcomes of environmental complexity in rodents have been remarkably consistent. Indeed, mice housed with some kind of structural enrichment generally show enhanced learning and memory performance,5 and diminished anxiety-like behaviour.109,110 One of the most common biological observations in rats and mice with complex housing is the genesis of new neurons in the hippocampus, 9 although it is unclear if this is sufficient to explain behavioural outcomes.2,111 Further neural consequences of this enrichment include increased dendritic branching, neuronal firing rate and synaptic plasticity in several cortical and striatal brain regions.6,7,9,112,113 However, it appears likely that systemic adaptations play an indispensable role in driving cognitive and behavioural effects of environmental complexity. Modifications to CD8+ T-cell transcription and inflammatory response, for example, appear to be essential components of environmental complexity–driven effects on hippocampal neurogenesis.16 Moreover, elevated levels of circulating SCFAs from shifts in microbiome composition are indispensable for positive effects of structural enrichment on neuronal plasticity and microglial function in the visual cortex.13 Finally, both novel and complex environmental features influence the responsiveness of the HPA axis. Enriched housing in CD-1 mice appears to slightly increase basal levels of circulating corticosterone; 114 furthermore, exposure to novel objects and larger cages elevates corticosterone in the blood of male C57BL/6 mice.115 The importance of these peripheral responses to the cognitive outcomes of structural enrichment suggests that some of this external information must cross the BBB and GB to reach the brain (Figure 3). Blood vessels in the brain and the BBB are indeed shown to be actively modified by learning and memory tasks,115–118 indicating a role for the neurovasculature in mediating cognitive and behavioural outcomes of structural enrichment. Correspondingly, several reports have indicated that enhanced blood vessel size and density compensate for neuronal changes in rodents with complex housing.4,8,63,119 These cerebrovascular adaptations arise through an accumulation of minute effects of environmental complexity on various cells of the neurovascular unit. For example, complex housing increases astrocytic arborization and plasticity in the rat dentate gyrus,120 and promotes tighter astrocyte–synapse interactions, which could improve coupling between neurons and the neurovascular unit.121 Meanwhile, microglia from mice housed with structural enrichment exhibit diminished reactivity and potentially enhanced polarization toward a protective, anti-inflammatory phenotype.12,122 Accumulating evidence also highlights the role of growth factors in the effects of complex housing, such as brain-derived neurotrophic factor and fibroblast growth factor 2, both of which are known to influence endothelial proliferation and genesis of blood vessels.60,110,123–125 Further, preclinical studies that employ environmental complexity as a protective intervention in disease models implicate the BBB as an important site for preventive adaptations. Angiogenesis and blood vessel density have been linked to the protective effects of environmental complexity against several disease models in rodents, including stroke,126 diabetes127 and aging.128 In a set of experiments using the chronic cerebral hypoperfusion model of vascular dementia in rats, environmental complexity, combined with access to a running wheel, improved memory deficits and reliably prevented disease-related loss of tight junction proteins, namely claudin-5 and zonula occluden-1, suggesting a neuroprotective adaptation at the neurovascular unit.129–131 This protective effect appears to be mediated by inhibition of β-catenin phosphorylation132 and rescue of autophagy dysfunction.131 The GB also plays an important role in mediating the effects of complex environments. Indeed, alterations to the gut microbiome that are induced by complex housing combined with a running wheel facilitate environment-related synaptic plasticity and microglia arborization in the visual cortex via increased circulating levels of butyrate, an SCFA.13 Access to physical exercise, combined with a complex environment, increases microbial production of SCFAs, which appears to be a key factor in driving neurogenesis in the hippocampus, a feature observed with structural enrichment in mice.9 It also accounts for a portion of enrichment-related increases in expression of hippocampal growth factors.133 In disease conditions, mice in structurally enriched cages possess a distinct microbiome composition when compared with standard housing,134 which could represent a confounding factor in studies seeking mechanistic insights for brain disorders. A similar impact was observed for the immune system, with complex housing promoting an anti-inflammatory response in both the blood and gut mucosa.103 In fact, combining complex housing with a running wheel can enhance the intestinal epithelium’s response for wound repair in the context of colon cancer.135 Finally, environmental complexity can modulate gut microbiome composition by increasing levels of *Lactobacillus*, a bacterium associated with anti-inflammatory effects.136 These findings shed light on a possible role for the GB in mediating gut–brain communication in complex environments. ### Physical exercise Physical activity is one of the single most important modifiable factors for individual health and has been proposed as the critical environmental variable in regulating health outcomes. 71,72,137 Regular voluntary exercise is linked to several beneficial effects, including improvements to memory and learning, as well as decreased risk of cardiovascular and psychiatric diseases.57,138 Moreover, exercise is a strong natural reward that enhances neuronal activity in key brain areas for reward processing.139 However, despite the inherent motivation, environmental constraints profoundly influence the possibility for an organism to be active. In humans, for example, community levels of physical activity correlate positively with availability of parks and public spaces, and negatively with socioeconomic barriers to space for exercise. 140,141 Likewise, mice with free access to a running wheel will habitually run up to 15 km per day, a behaviour not observed in standard laboratory cages.142 Therefore, locomotion can be seen not just objectively as quantifiable movement, but more broadly as a measure of external influence on expressed behaviours, a shift that emphasizes the environmental aspects of physical exercise. Activity is commonly evaluated among human participants through questionnaires about lifestyle and exercise, or, in clinical studies, as an intervention, whereby a subset of patients performs a predetermined exercise regimen. Meanwhile, locomotion studies in rodents are limited to monitoring of regular movement within the home cage, or with access to a treadmill or running wheel.143 Although exercise is linked to many changes in the brain — including elevated striatal dopamine and enhanced synaptic plasticity and hippocampal neurogenesis — it also produces broad systemic effects.69,137,144 Among both humans and rodents, physical activity produces changes to the immune system, including a distinct cytokine profile that differs slightly but significantly from that elicited by infection or damage. In contrast to the pathogen response, strictly proinflammatory cytokines such as TNF-α and IL-1β are not significantly increased after physical exercise.138 Instead, IL-6 alone is heavily secreted from contracting skeletal muscles, promoting a downstream anti-inflammatory effect by stimulating release of IL-10.145,146 Further, exercise is linked to slight shifts in microbiome composition, generating a pronounced increase in circulating levels of butyrate.147,148 Glucocorticoid release is elicited by acute exercise in an intensity-dependent manner, and chronic exercise can elevate baseline levels of cortisol.149 Furthermore, a brief period of treadmill running dampens leukocyte sensitivity to elevated glucocorticoid levels and reduces risk of HPA over-reaction.150 However, forced physical exercise is detrimental, with similar outcomes as exposure to chronic stressors. It promotes higher expression of inflammatory markers and HPA reactivity, which in turn are related to anxiety-like behaviours.151 Paradoxically, voluntary exercise–induced elevation of IL-6 and corticosterone is also similar to the effects of chronic stress, but produces completely opposite effects on the brain.152 Although stress can be detrimental, voluntary exercise is often associated with beneficial outcomes including improved memory and reduced anxiety-like behaviours.64,69 An important difference that could explain the behavioural outcomes of physical exercise versus stress exposure, despite common biology, is controllability. In fact, controllable events might even promote control over unpredictable events153 and produce differential HPA activation or immune profiles.154,155 Interestingly, voluntary exercise can be seen as a controllable event that produces several changes to the BBB and GB (Figure 3). A better understanding of the impact of physical exercise on these barriers could help its promotion in the context of mental health or other conditions. Among humans, physical activity produces an increase in both blood supply to the brain and blood oxygenation within minutes, especially in the prefrontal cortex and motor areas.156–158 In rodent models, long-lasting increases of blood flow caused by several weeks of physical exercise is associated with increased blood vessel density in the cortex, striatum and cerebellum.8,67,159,160 Exercise also strengthens interactions between cells of the neurovascular unit to produce a healthier BBB. For instance, access to treadmill exercise boosts astrocyte density in the hippocampus of male rats161 and in the prefrontal cortex of male C57BL/6 mice.162 Wheel running increases astrocyte reactivity, as measured by cell body area, and staining of aquaporin-4 — a water channel localized on astrocyte end-feet that is heavily involved in communication between astrocytes and endothelial cells of the BBB — in both the hippocampus and prefrontal cortex of male Wistar rats.163 Aquaporin-4 is involved in various functions such as regulation of extracellular space volume, cerebrospinal fluid circulation, waste clearance, neuroinflammation, calcium signalling, cell migration and more; thus, it is directly involved in BBB homeostasis and permeability.164 Physical exercise changes the cellular distribution of aquaporin-4, with higher concentration at the end-feet. 165 Increased astrocytic coverage of blood vessels was also reported in mice with access to physical exercise (30-min sessions, 3 times/wk for 12 wk).61 Furthermore, smooth muscle cells along cerebral capillaries, including pericytes, express high levels of a lactate receptor, hydrocarboxylic acid receptor-1, after long-term treadmill running among C57BL/6 mice. Subsequent downstream signalling of hydrocarboxylic acid receptor 1 leads to release of vascular endothelial growth factor (VEGF) and increased angiogenesis,160 a finding that coincides with earlier studies linking elevated VEGF expression to neurogenic and behavioural changes after exercise.166,167 This link provides evidence that the BBB acts as a mediator for cerebral and systemic effects of environmental change. Physical exercise also promotes strength and maintenance of tight junctions between endothelial cells of the BBB and protects against damage-induced loss of barrier integrity. Among male C57BL/6 mice, access to a running wheel enhanced expression of occludin in the brain and prevented extravasation of injected tumour cells;168 among male Wistar rats, a graded treadmill exercise rescued expression of striatal claudin-5 after induction of type I diabetes.169 Souza and colleagues170 linked exercise-related protection of tight junction proteins to reductions in inflammatory signalling and oxidative stress of the central nervous system. The protective antioxidant effects of physical exercise in the brain appear to be mediated by inhibition of glycogen synthase kinase-3 (GSK-3), a redox-sensitive enzyme that destabilizes tight junction architecture via β-catenin phosphorylation.171 In vitro studies have found that inhibiting GSK-3 in brain endothelial cells reduces inflammatory signalling and improves the half-life of tight junction proteins, namely claudin-5 and occludin.172,173 In mice, voluntary wheel running could inhibit GSK-3 by increasing serum concentrations of the clusterin protein, which binds strongly to the low-density lipoprotein receptor-related protein 8 on microvascular endothelial cells in the brain.174 Activation of this receptor suppresses GSK-3 activity through a pathway known to promote BBB integrity in vitro.175,176 Excessive activity (> 60 min at > 70% of work capacity) can promote a leaky gut by activation of the HPA axis, along with elevated body temperature, leading to hypoperfusion of abdominal organs and subsequent changes in level of nutrients and immune signals reaching the GB.177,178 This effect produces a transient increase in permeability of the epithelial barrier, as measured with a sugar absorption test, as well as increased circulating markers of intestinal stress.178,179 In rodents, voluntary physical exercise influences inflammatory gene expression in intestinal leukocytes, downregulating proinflammatory cytokines while increasing levels of anti-inflammatory factors and antioxidant enzymes.180,181 Similarly, in humans, short periods of exercise increase lymphocyte levels in the blood, but diminish expression of toll-like receptors, suggesting that physical activity can modulate the immune response to pathogens and stressors.182,183 Physical exercise increases diversity of the gut microbiome, according to a direct comparison of samples from athletes and people with a sedentary lifestyle.184 This effect may be mediated by innate immune cells that condition the gut microbiome toward optimal composition of serotonin and SCFA-producing bacteria.147,185 The microbiome of people with low body weight despite a sedentary lifestyle is similar to that elicited by exercise, suggesting that a specific bacterial profile could be associated with physical health.186 Indeed, randomized controlled trials involving children with obesity showed that deleterious gut microbiome and proinflammatory signalling could be reversed to the levels of healthy children via a physical exercise intervention.187 As for adults with obesity, 6 weeks of physical exercise was sufficient to modify the diversity and composition of the gut microbiome by increasing SCFAs and butyrate-producing bacteria in relation to body mass and loss of weight.188 At the mechanistic level, butyrate was found to be responsible for assembly and maintenance of tight junction proteins in cultured human intestinal epithelial cells,189 suggesting that exercise-induced microbial production of butyrate could promote a healthy GB. ## Barrier–environment interactions in development and aging Most of this evidence describes how environmental components influence the normal functioning of the BBB and GB during adult life; however, they do not explicitly consider the impact of early challenges or the long-term effects of environmental conditions on barrier function later in life. Across the lifespan, from the maternal environment to life as an older adult, environmental conditions have the potential to modify barrier functioning; indeed, enriched environments may improve function and the expression of anti-inflammatory factors, whereas detrimental environmental conditions could lead to improper functioning and exacerbated inflammation. 73,190,191 Brain maturation and development is a continuous process; different developmental stages have characteristic behavioural, neurologic and immune profiles. This progression between life stages is similar across humans and rodents and is mainly mediated by preprogrammed physiologic changes in cell and hormonal activity.192 Throughout development, there are periods (e.g., in embryo, childhood, adolescence) of increased sensitivity to the surrounding environment’s threats, like stress,40 as well as to beneficial, stimulating conditions. Traditionally, these windows of sensitivity are thought to be mainly determined by synaptic changes or neuronal growth, but the BBB and the GB also have specific developmental milestones during those sensitive periods. For instance, during the first postnatal days there is an increase in both astrocyte expression in the brain and microbial colonization of the gut that will help sustain normal neuronal wiring and cognitive function.30,33 Here, we discuss how the function of the barriers can be improved or harmed by environmental features from a lifespan and life stage perspective (further details about BBB development can be found elsewhere)30,33,193,194. In addition, we highlight research opportunities for which a developmental perspective may contribute to understanding of the relationship between barrier function and mental health. ### Embryogenesis and development During the first embryonic or fetal stages (embryonic day 10 in rodents and gestational week 8 in humans) the permeability of the BBB is set to protect the growing brain against potential threats,195 and the GB prepares for complex diets by triggering specific cell differentiation of the intestine.194 However, the maternal environment, in combination with embryonal or fetal age, can modify the development of these barriers and their susceptibility to factors such as toxins,196,197 viruses198,199 or unhealthy diet.200 Environmental modification in the sociability, complexity (e.g., socioeconomic status in humans) and physical activity of the mother during pregnancy can produce positive or negative transgenerational effects by setting a specific proinflammatory footprint in the offspring.201–204 These modifications may even contribute to future stress reactivity by changing release of stress hormones and expression of brain glucocorticoid receptors.205,206 For instance, environmental enrichment by structural complexity during pregnancy alters inflammatory responses mediated by circulating cytokines (IL-6, IL-10 and TNF-α) and lipopolysaccharide-induced expression of IL-22 and prostaglandin E2.207 This immune response profile promotes healthy offspring and may help with the normal development of motor skills. Likewise, in a transgenic mouse model of Alzheimer disease, physical exercise during pregnancy showed long-term effects on the offspring’s neurovascular function by improving cerebral oxygen and nutrient supply, increasing angiogenesis and BBB transporter activity and reducing microglia reactivity.208 Furthermore, positive environmental conditions for the mother can reduce behavioural outcomes associated with anxiety during the offspring’s adolescence or adult life.209 On the other hand, a detrimental environment such as social isolation and reduced complexity during pregnancy may negate the beneficial effects on synaptic transmission driven by access to an enrichment environment. 210 Housing mothers with a whole ensemble of enrichment during pregnancy — combining social interaction, physical exercise and structural complexity — can prevent the metabolic disturbances generated by hypoxic–ischemic events in the offspring’s early life and their impact in adult memory impairment.211 Positive cognitive outcomes are associated with long-term increases in the expression of growth-related proteins, downregulation of amyloid precursors and proper glucose metabolism. Considering the crucial role of BBB glucose transporters to meet the brain’s high energy demands, environmental enrichment may promote BBB-related transgenerational features that support brain metabolism and, ultimately, plasticity. After birth, the growing BBB and GB undergo additional critical periods before they reach full maturation and functioning. Environmental modifications during postnatal and early-life periods — childhood and adolescence — play key roles in improving or altering barriers’ properties in the short and long term. They have been mostly investigated using paradigms of early-life adversity, such as maternal separation, limited besting and nesting, social isolation or sensorial deprivation, and as short- and long-term effects centred on brain connectivity and neuronal function.51,212,213 However, these postnatal periods also see expansion and refinement of the brain vascular architecture, with changes in endothelial transport and astrocyte expression.30,214 These features of BBB development suggest that neurovascular function may be sensitive to positive and negative environmental interventions; thus, access to an enriched environment early in life could potentially exert lasting changes in brain and behaviour. Indeed, structural complexity during postnatal development is associated with increased vascular branching and expression of the angiogenic VEGF and glucose transporters, at least in the rodent visual cortex.107,215 Structural complexity with VEGF administration, either in the visual cortex or in the hippocampus dentate gyrus, improves execution time and performance in a spatial memory task.117 Intriguingly, opportunities for social interactions after stressful events during adolescence counteracts deleterious stress effects by reducing aggressivity and anxiety- and depression-like behaviours in adulthood. These behavioural adaptations appear to be related to changes in corticosterone levels and increased expression of cell adhesion molecules associated with cell migration and outgrowth during development.216 Maternal separation can induce visceral hypersensitivity in rats, which can be improved via nutritional approaches early in life;217 however, how access to an enriched environment, physical exercise or social interactions could affect GB development remain understudied. ### Later life and aging On the other end of the lifespan, aging is generally associated with progressive decline of cognitive and physical health capacities, with cellular and molecular changes driving metabolic impairment, synaptic loss or alterations, neuroendocrine senescence and cerebrovascular decline.31,218,219 Onset of aging-related pathologies is linked with complex interactions between genetic and environmental factors, which can alleviate or exacerbate programmed senescence process for various systems or cell types that might increase susceptibility for conditions like Alzheimer or Parkinson diseases.31,219 Core features of age-related cognitive impairments, including dementias, are vascular dysfunction and BBB hyperpermeability through loss of occludin, zonula occluden-1, astrocytes and pericytes.220,221 Despite the progressive decline of cognitive processes, access to environmental enrichment during aging has positive effects in delaying onset of several pathologies, sustaining neurovascular integrity and activity, and preserving function of other brain systems during normal aging.62,73,222 Complementary environmental features can counteract age-related impairments. For instance, the whole ensemble of social, structural and locomotor enrichment increases lifespan and reduces anxiety-like behaviour, which appear to be associated with proper neurovascular function as measured by high glucose metabolism, expression of angiogenic markers and reduced expression of inflammatory genes in the brain.119,223–225 Interestingly, voluntary physical exercise during middle or older age promotes expression of the proangiogenic growth factor, VEGF; prevents age-related astrocyte, pericyte and myelin loss in the hippocampus; and reduces vascular leakage, possibly through higher vascular coverage by pericytes. 223,225 These beneficial effects of voluntary physical exercise on neurovascular health are associated with conserved motivation to engage in spontaneous behaviours, an indirect measure of cognitive function.225 Furthermore, long periods of social interaction during the transition from middle age to older age improves rodent performance in the novel object recognition task, which appears to be driven by alterations in corticosterone levels and reduced accumulation of vascular amyloid depositions.62 These findings support an extended window of opportunity during aging to intervene with the goal of sustaining BBB and GB integrity and promoting optimal function, leading to proper mental and physical health during this life stage. Environmental enrichment can act as a preventive strategy to overcome the negative short- and long-term effects of stressful early-life events and age-programmed senescence on stress response, cell loss and decline of neurovascular function.53,73,226 Its positive outcomes seem to be mediated by changes in glucose metabolism, angiogenic factors that control vascular integrity and glial cell reactivity, leading to reduce maladaptive behaviours and preservation of cognitive function throughout the lifespan.117,191,227,228 It seems that vascularization and BBB integrity are complementary factors relevant to the promotion of learning and memory processes or emotional regulation.117 Most current research in this area has focused on neurodevelopmental disorders such as neonatal hypoxia or neurodegenerative conditions such as Alzheimer disease or dementia. Moreover, these studies have involved only a small subset of brain areas, highlighting the need to expand knowledge about how positive environment influences development and describe its role for other behavioural domains, such as mood disorders. Another research avenue will be to determine the role of each component of environmental enrichment in development, as most of the research to date has focused on the general effect of the enrichment environment without considering the effects of each feature independently or comparing them with each other. Few studies have directly compared beneficial effects of one environmental feature with other features or the summative effect of all components; thus, it is impossible to rule out that single strategies may be more effective in specific contexts.62 Identification of the main element leading to behavioural improvement will help to establish translational low-cost, highly effective strategies in the promotion of healthy lifestyle and improved mental health. ## Barrier–environment interactions in mood disorder pathogenesis The importance of studying environment–barrier interactions is heightened in the context of brain disorders. Psychiatric conditions like MDD, bipolar disorder and schizophrenia rank among the leading causes of disability worldwide, affecting nearly 1 in 4 people.229 Despite high prevalences — 7% for MDD, 3% for bipolar disorder and 1% for schizophrenia — and severe effects on quality of life, commercially available therapeutics have advanced only slightly in the past 30 years, while between 30%–50% of all affected individuals are slightly responsive or nonresponsive to current treatments. 65,230 Environment constitutes an important proportion of risk for these diseases, with genetic contributions estimated at 60% of risk for schizophrenia to as low as 30% for MDD.231 This suggests disruption of pathways for environment–brain communication, like the BBB and GB, in the pathogenesis of mood disorders. Correspondingly, loss of barrier integrity is increasingly recognized as a hallmark feature of neurodegenerative disorders, with dysfunction of BBB and GB reported among people with MDD,36,38,49,232,233 bipolar disorder45,46 and schizophrenia.44,47,234 Despite the well-known involvement of the environment, BBB and GB in the pathogenesis of mood disorders, the role of communication between these systems remains unclear. This section discusses a possible role for environment–barrier interactions in progression of psychiatric conditions and identifies areas for future research. ### Environment as a source of stress Past and present environments can promote or prevent the onset of mental health disorders through modification of the stress response. Negative or impoverished conditions — such as low socioeconomic status, social isolation and sedentary lifestyle — are sources of chronic stress linked to peripheral inflammation, 55,235 gut dysbiosis236 and risk for development of mood disorders.54,56 However, positive and stimulating environmental conditions also confer a degree of stress; social interactions, structural complexity and physical exercise all increase HPA activation (Figure 3), and yet are associated with beneficial effects on learning and memory via adaptations in the BBB and GB. Moreover, preclinical studies have found anti-depressant effects of physical exercise and complex housing in the context of social defeat stress.58,59 What determines whether the influence of a given environment is positive or negative? Part of the answer comes from the inherent properties of the stress response, which is described as an allostatic system, meaning it can adapt to environmental demands, changing its equilibrium point through sustained alterations in allostatic mediators like cytokines and glucocorticoids.237 The dynamic nature of allostatic systems is crucial to maintaining homeostatic mechanisms during periods of external change.238,239 However, in the presence of a severe or prolonged stressor, these systems are susceptible to allostatic overload, at which point the capacity to maintain stability is exceeded, resulting in illness or death.237,240 Importantly, in the face of a given stress, some people will experience allostatic overload and pathology, while others will be able to cope and return to baseline. This capacity for returning to baseline after stress is termed resilience, a concept that has come to shape the way health and disease are conceptualized.241,242 In the case of psychiatric illness, the stress response is elevated to the point of damaging the BBB to induce neuroinflammation and cognitive deficits,38 while a subthreshold, or “good,” stress, can increase the functional range of the system and enable an organism to weather greater fluctuations in the future.1 This framework clearly explains how a stimulating environment could act as a low-grade stressor to actually enhance an organism’s capacity to deal with future challenges — a phenomenon that has been referred to both as buffering56 and inoculating against1 the effects of chronic stress. ### Barrier–environment interactions mediate stress resilience As critical frontiers for communication between the environment, periphery and brain, the BBB and GB are key loci for adaptations that determine the influence of the environment on cognition and behaviour. These barriers are known to play a crucial role in mediating resilience versus susceptibility to chronic stress. For instance, inflammation-related epigenetic suppression of the claudin-5 promoter in endothelial cells of the BBB is a key predictor of susceptibility to social defeat stress in male mice.99 Among female mice, 6 days of chronic variable stress alters gut microbiome composition and disrupts GB integrity, resulting in elevated levels of circulating llipopolysaccharide-binding protein and behavioural deficits.25,36,49 However, the involvement of the BBB and GB in effects of positive environments on stress response and mental health is less clear. One preclinical study reported that peripheral injection of fibroblast growth factor-2, a growth factor associated with BBB integrity and positive cognitive outcomes of structural enrichment, is sufficient to diminish anxious behaviour in a study of selectively bred, high-anxiety rats.110 In another report, physical exercise prevented depression-like behaviour that would normally have been induced by chronic unpredictable stress exposure by increasing astrocyte density and proliferation in the hippocampus. 243 Proliferation of astrocytes can be associated with more extensive astrocyte covering of blood vessels, a key feature of BBB integrity.20 This finding corresponds with results from a rat model of vascular dementia in which physical exercise prevented cognitive deficits by increasing astrocyte coverage of blood vessels.61 Further, in mice, oral administration of SCFAs — which are produced in higher levels by the gut after structural enrichment, social interactions and physical exercise — promoted anxiolytic-like behaviours and prevented social stress–inducing anhedonia, stress reactivity and increased intestinal permeability.244 Interestingly, it is known that SCFAs exert their effect mainly through free fatty acid receptor-2 and −3; however, studies of their role in mediating the gut–brain axis have focused on receptor modulation with chemogenetic tools or the development of allosteric modulators without behavioural evaluation;245 thus, it will be intriguing to evaluate the impact of these SCFA-related chemogenetic tools and allosteric modulators in the context of stress-related disorders. In humans, the finding that social support is associated with reductions in circulating levels of E-selectin is striking,87 as this molecule has been reported as a sex-specific biomarker for BBB disruption associated with MDD.49 Moreover, reports have linked socioeconomic status with gut and microbiome health,236 as well as better measurements of intestinal barrier integrity via a sugar permeability assay in children.246 Although evidence is scarce, it is increasingly apparent that environmental modifications of the neurovasculature and gut epithelium are important in development of stress resilience. This highlights a new opportunity for drug discovery, as the BBB and GB are increasingly seen as potential targets for novel antidepressant and antipsychotic therapies. Drug discovery programs that aim to reduce stress-induced damage by targeting pathological features of mood disorders are an obvious approach. For example, 2 recent preclinical studies — one employing pharmacological modulation of Wnt-7a signalling with the Gpr124/Reck agonist in stroke models, and another regulating the transforming growth factor-β pathway in experimental epilepsy using RepSox, an inhibitor of activin receptor-like kinase-5 — have shown that BBB integrity can be recovered by indirect upregulation of claudin-5 in brain blood vessels.247,248 However, these target-driven studies are limited in scope, focusing strictly on repair and thereby missing out on bigger-picture strategies associated with overall mental well-being.249 The proresilient effects of social interaction, environmental complexity and physical activity offer an alternative, phenotype-based method for drug development, making it possible to investigate pathways associated with prevention instead of treatment or to investigate stress-related changes underlying cognitive deficits and mood disorders. Several potential antidepressant targets can be gleaned from environment–barrier interactions (Figure 2), including levels of circulating factors like cytokines and SCFAs, expression of growth factors in the brain and activity of redox-sensitive enzymes like GSK-3 in brain endothelial cells. For decades, GSK-3 has been targeted in the context of psychiatric conditions, notably with lithium.250 The efficiency of antidepressant treatment has been linked to an inhibitory effect on GSK-3 activity; 251 nevertheless, its relevance in the treatment of MDD is unclear. Greater attention should be paid to these pathways in future studies to elucidate potential roles in promoting stress resilience at the BBB and GB. Ultimately, a better understanding of environment–barrier interactions in the context of psychiatric disorders will greatly improve understanding of these diseases, as well as capacity to prevent and treat them. ## Conclusion In the early 20th century, it was commonly held that the adult brain was a rigid organ, independent of external influence (Figure 4). The experiments of Donald O. Hebb3 helped to strike down this dogma and repopularize the idea that environment can change the brain, a concept that reaches back centuries.5,252 His findings of environmental influence on learning and memory in rats lent enormous support to the theory of synaptic plasticity, which ultimately changed the scope of future neuroscience; yet, this revolution was narrow-sighted, and the focus on neurons and the brain left systemic aspects of enrichment sidelined for years. Alhough it has been long known that environment can influence the immune system, circulating glucocorticoids and microbiome composition,14,52,146,148,253 only recently have the interactions between these systems and the brain been investigated. 13,16,88,114 As critical mediators of communication between anatomic compartments, biological barriers like the BBB and GB are major interfaces for translating environmental information between the brain and periphery. ![Fig. 4](http://jpn.ca/https://www.jpn.ca/content/jpn/48/3/E190/F4.medium.gif) [Fig. 4](http://jpn.ca/content/48/3/E190/F4) Fig. 4 Historical development of understanding of environmental influence on cognition, emotion and behaviour. (A) For the first half of the 20th century, it was widely believed that the adult brain was a rigid organ, insensitive to change from external factors. (B) Afterr Hebb’s demonstration of positive effects of a stimulating environment on learning and memory, it was established that environmental influence on neuronal plasticity and firing was responsible for long-term changes in mental state. (C) In recent years, systemic outcomes have been increasingly recognized as indispensable for positive outcomes of environmental enrichment. Thus, peripheral signals, mediated by the blood–brain and gut barriers, are important features acting in parallel to central plasticity changes to influence cognition, emotion and behaviour in response to shifting environments. HPA = hypothalamic–pituitary–adrenal. In this review, we have summarized a broad literature that describes multifaceted interactions between the environment, the BBB and the GB to argue that these barriers play a central, active role in external modulation of cognition, emotion and behaviour throughout the lifespan. Further, we emphasized an important early role for environment–barrier interactions in the pathogenesis of mood disorders and suggested potential targets for novel therapeutics. We focused our discussion on 3 major environmental components that have been identified as the key loci for environmental influence on cognition, behaviour and underlying neurobiology — social interactions, structural complexity and locomotion.1,2,62,68,70 This focus meant that several other factors such as environmental pollution, diet, temperature, auditory stimulation and drug and alcohol consumption — all of which are also known to affect behaviour and cognition — were beyond the scope of this review. However, these are also important variables that can modify barrier integrity and should not be neglected by future studies. In addition, we found that the literature lacked a standardized paradigm for studying environment in a laboratory setting, such as cage components used as structural enrichment, and duration of exposure to running wheels or treadmill exercise. This variability impedes comparison of results; moving forward, we suggest clear and transparent reporting of environmental conditions, as well as adoption of standardized paradigms, which will be crucial to future detailed investigations of environment–barrier interactions in health and disease. In a world where the number of diagnoses for psychiatric illnesses is growing in parallel with accumulation of environmental pollutants, feelings of loneliness and isolation, vast cultural and dietary shifts, sprawling suburbanization and more frequent extreme weather events that drive mass displacement, understanding the detailed interactions between environment and the brain should be of utmost importance, The BBB and GB are key loci for these investigations. Several questions remain unanswered, representing exciting opportunities for new research. We suggest 2 main areas of interest, namely characterizing relationships between socioeconomic status, social interactions and physical activity with BBB and GB health in human psychiatric disorders, and investigating molecular pathways involved in environment-associated protective adaptations at the BBB and gut epithelium for novel pharmaceutical targets. A fuller understanding of environment–barrier interactions will help in the development of effective therapies and preventive strategies for psychiatric conditions, as well as a better appreciation of how humans are affected by the worlds they inhabit. ## Footnotes * * Share first authorship. * **Competing interests:** None declared. * **Contributors:** All of the authors contributed to the conception and design of the work, drafted the manuscript, revised it critically for important intellectual content, gave final approval of the version to be published and agreed to be accountable for all aspects of the work. * **Funding:** This work was supported by the Canadian Institutes for Health Research (Project Grant No. 427011 to Caroline Menard), Fonds de recherche du Québec — Santé (Junior 2 salary award to Caroline Menard), Brain Canada 2019 Future Leaders in Canadian Brain Research (Caroline Menard) and Canada First Research Excellence Fund (Sentinel North Research Chair to Caroline Menard). Sam Paton and José Solano are funded by Canadian Institutes for Health Research MSc and NeuroQuebec PhD scholarships, respectively. * Received November 30, 2022. * Revision received March 1, 2023. * Accepted March 19, 2023. This is an Open Access article distributed in accordance with the terms of the Creative Commons Attribution (CC BY-NC-ND 4.0) licence, which permits use, distribution and reproduction in any medium, provided that the original publication is properly cited, the use is noncommercial (i.e., research or educational use), and no modifications or adaptations are made. See: [https://creativecommons.org/licenses/by-nc-nd/4.0/](https://creativecommons.org/licenses/by-nc-nd/4.0/) ## References 1. Crofton EJ, Zhang Y, Green TA. Inoculation stress hypothesis of environmental enrichment. Neurosci Biobehav Rev 2015;49:19–31. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1016/j.neubiorev.2014.11.017&link_type=DOI) 2. Kempermann G. Environmental enrichment, new neurons and the neurobiology of individuality. Nat Rev Neurosci 2019;20:235–45. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1038/s41583-019-0120-x&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=30723309&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) 3. Hebb DO. The organization of behavior; a neuropsychological theory. New York: Wiley; 1949:xix, 335. 4. Diamond MC, Krech D, Rosenzweig MR. The effects of an enriched environment on the histology of the rat cerebral cortex. J Comp Neurol 1964;123:111–20. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1002/cne.901230110&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=14199261&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) [Web of Science](http://jpn.ca/lookup/external-ref?access_num=A19644550B00004&link_type=ISI) 5. Rosenzweig MR, Bennett EL. Psychobiology of plasticity: effects of training and experience on brain and behavior. Behav Brain Res 1996;78:57–65. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1016/0166-4328(95)00216-2&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=8793038&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) [Web of Science](http://jpn.ca/lookup/external-ref?access_num=A1996UR89700009&link_type=ISI) 6. Baroncelli L, Braschi C, Spolidoro M, et al. Nurturing brain plasticity: impact of environmental enrichment. Cell Death Differ 2010;17:1092–103. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1038/cdd.2009.193&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=20019745&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) [Web of Science](http://jpn.ca/lookup/external-ref?access_num=000278726100004&link_type=ISI) 7. Eckert MJ, Bilkey DK, Abraham WC. Altered plasticity in hippocampal CA1, but not dentate gyrus, following long-term environmental enrichment. J Neurophysiol 2010;103:3320–9. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1152/jn.01037.2009&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=20393057&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) [Web of Science](http://jpn.ca/lookup/external-ref?access_num=000278493900032&link_type=ISI) 8. Ekstrand J, Hellsten J, Tingström A. Environmental enrichment, exercise and corticosterone affect endothelial cell proliferation in adult rat hippocampus and prefrontal cortex. Neurosci Lett 2008;442:203–7. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1016/j.neulet.2008.06.085&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=18625288&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) 9. Kempermann G, Kuhn HG, Gage FH. More hippocampal neurons in adult mice living in an enriched environment. Nature 1997;386:493–5. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1038/386493a0&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=9087407&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) [Web of Science](http://jpn.ca/lookup/external-ref?access_num=A1997WR25600051&link_type=ISI) 10. Manno FAM, An Z, Kumar R, et al. Environmental enrichment leads to behavioral circadian shifts enhancing brain-wide functional connectivity between sensory cortices and eliciting increased hippocampal spiking. Neuroimage 2022;252:119016. 11. Brod S, Gobbetti T, Gittens B, et al. The impact of environmental enrichment on the murine inflammatory immune response. JCI Insight 2017;2:e90723. 12. Chabry J, Nicolas S, Cazareth J, et al. Enriched environment decreases microglia and brain macrophages inflammatory phenotypes through adiponectin-dependent mechanisms: Relevance to depressive-like behavior. Brain Behav Immun 2015;50:275–87. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1016/j.bbi.2015.07.018&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=26209808&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) 13. Lupori L, Cornuti S, Mazziotti R, et al. The gut microbiota of environmentally enriched mice regulates visual cortical plasticity. Cell Rep 2022;38:110212. 14. Marashi V, Barnekow A, Ossendorf E, et al. Effects of different forms of environmental enrichment on behavioral, endocrinological, and immunological parameters in male mice. Horm Behav 2003;43:281–92. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1016/S0018-506X(03)00002-3&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=12694638&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) [Web of Science](http://jpn.ca/lookup/external-ref?access_num=000182658500002&link_type=ISI) 15. McQuaid RJ, Audet M-C, Jacobson-Pick S, et al. Environmental enrichment influences brain cytokine variations elicited by social defeat in mice. Psychoneuroendocrinology 2013;38:987–96. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1016/j.psyneuen.2012.10.003&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=23177512&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) [Web of Science](http://jpn.ca/lookup/external-ref?access_num=000320412400004&link_type=ISI) 16. Zarif H, Nicolas S, Guyot M, et al. CD8+ T cells are essential for the effects of enriched environment on hippocampus-dependent behavior, hippocampal neurogenesis and synaptic plasticity. Brain Behav Immun 2018;69:235–54. 17. Daneman R, Prat A. The blood-brain barrier. Cold Spring Harb Perspect Biol 2015;7:a020412. [Abstract/FREE Full Text](http://jpn.ca/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTE6ImNzaHBlcnNwZWN0IjtzOjU6InJlc2lkIjtzOjExOiI3LzEvYTAyMDQxMiI7czo0OiJhdG9tIjtzOjE5OiIvanBuLzQ4LzMvRTE5MC5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 18. Armulik A, Genové G, Mäe M, et al. Pericytes regulate the blood-brain barrier. Nature 2010;468:557–61. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1038/nature09522&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=20944627&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) [Web of Science](http://jpn.ca/lookup/external-ref?access_num=000284584200041&link_type=ISI) 19. Haruwaka K, Ikegami A, Tachibana Y, et al. Dual microglia effects on blood brain barrier permeability induced by systemic inflammation. Nat Commun 2019;10:5816. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1038/s41467-019-13812-z&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=31862977&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) 20. Hösli L, Zuend M, Bredell G, et al. Direct vascular contact is a hallmark of cerebral astrocytes. Cell Rep 2022;39:110599. 21. Andreone BJ, Lacoste B, Gu C. Neuronal and vascular interactions. Annu Rev Neurosci 2015;38:25–46. 22. Sweeney MD, Zhao Z, Montagne A, et al. Blood-brain barrier: from physiology to disease and back. Physiol Rev 2019;99:21–78. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1152/physrev.00050.2017&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=30280653&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) 23. Segarra M, Aburto MR, Acker-Palmer A. Blood-brain barrier dynamics to maintain brain homeostasis. Trends Neurosci 2021;44:393–405. 24. Chelakkot C, Ghim J, Ryu SH. Mechanisms regulating intestinal barrier integrity and its pathological implications. Exp Mol Med 2018;50:1–9. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1038/s12276-018-0116-z&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=30054468&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) 25. Doney E, Cadoret A, Dion-Albert L, et al. Inflammation-driven brain and gut barrier dysfunction in stress and mood disorders. Eur J Neurosci 2022;55:2851–94. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1111/ejn.15239&link_type=DOI) 26. Suzuki T. Regulation of intestinal epithelial permeability by tight junctions. Cell Mol Life Sci 2013;70:631–59. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1007/s00018-012-1070-x&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=22782113&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) 27. Ahn J, Hayes RB. Environmental influences on the human microbiome and implications for noncommunicable disease. Annu Rev Public Health 2021;42:277–92. 28. Cryan JF, O’Riordan KJ, Cowan CSM, et al. The microbiota-gutbrain axis. Physiol Rev 2019;99:1877–2013. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1152/physrev.00018.2018&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=31460832&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) 29. Lee S-H, Yoon S-H, Jung Y, et al. Emotional well-being and gut microbiome profiles by enterotype. Sci Rep 2020;10:20736. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1038/s41598-020-77673-z&link_type=DOI) 30. Coelho-Santos V, Shih AY. Postnatal development of cerebrovascular structure and the neurogliovascular unit. Wiley Interdiscip Rev Dev Biol 2020;9:e363. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1002/wdev.363&link_type=DOI) 31. Costea L, Mészáros Á, Bauer H, et al. The blood-brain barrier and its intercellular junctions in age-related brain disorders. Int J Mol Sci 2019;20:5472. 32. Dinan TG, Cryan JF. Gut instincts: microbiota as a key regulator of brain development, ageing and neurodegeneration: microbiota-gut-brain axis across the lifespan. J Physiol 2017;595:489–503. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1113/JP273106&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=27641441&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) 33. Jena A, Montoya CA, Mullaney JA, et al. Gut-brain axis in the early postnatal years of life: a developmental perspective. Front Integr Neurosci 2020;14:44. 34. Zhao Z, Nelson AR, Betsholtz C, et al. Establishment and dysfunction of the blood-brain barrier. Cell 2015;163:1064–78. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1016/j.cell.2015.10.067&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=26590417&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) 35. 1. Esser C , editor. Environmental influences on the immune system. Vienna (Austria): Springer Vienna; 2016. Available: [https://link.springer.com/book/10.1007/978-3-7091-1890-0](https://link.springer.com/book/10.1007/978-3-7091-1890-0) (accessed 2022 Oct. 31). 36. Doney E, Dion-Albert L, Coulombe-Rozon F, et al. Chronic stress exposure alters the gut barrier: sex-specific effects on microbiota and jejunum tight junctions. Biological Psychiatry Global Open Science 2023 May 8 . [Epub ahead of print] doi: 10.1016/j.bpsgos.2023.04.007 [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1016/j.bpsgos.2023.04.007&link_type=DOI) 37. Eto M, Kouroedov A, Cosentino F, et al. Glycogen synthase kinase-3 mediates endothelial cell activation by tumor necrosis factor-alpha. Circulation 2005;112:1316–22. [Abstract/FREE Full Text](http://jpn.ca/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTQ6ImNpcmN1bGF0aW9uYWhhIjtzOjU6InJlc2lkIjtzOjEwOiIxMTIvOS8xMzE2IjtzOjQ6ImF0b20iO3M6MTk6Ii9qcG4vNDgvMy9FMTkwLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 38. Menard C, Pfau ML, Hodes GE, et al. Social stress induces neurovascular pathology promoting depression. Nat Neurosci 2017;20:1752–60. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1038/s41593-017-0010-3&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=29184215&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) 39. Braniste V, Al-Asmakh M, Kowal C, et al. The gut microbiota influences blood-brain barrier permeability in mice. Sci Transl Med 2014;6:263ra158. [Abstract/FREE Full Text](http://jpn.ca/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTE6InNjaXRyYW5zbWVkIjtzOjU6InJlc2lkIjtzOjE0OiI2LzI2My8yNjNyYTE1OCI7czo0OiJhdG9tIjtzOjE5OiIvanBuLzQ4LzMvRTE5MC5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 40. Lupien SJ, McEwen BS, Gunnar MR, et al. Effects of stress throughout the lifespan on the brain, behaviour and cognition. Nat Rev Neurosci 2009;10:434–45. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1038/nrn2639&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=19401723&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) [Web of Science](http://jpn.ca/lookup/external-ref?access_num=000266199300011&link_type=ISI) 41. Salvador E, Shityakov S, Förster C. Glucocorticoids and endothelial cell barrier function. Cell Tissue Res 2014;355:597–605. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1007/s00441-013-1762-z&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=24352805&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) [Web of Science](http://jpn.ca/lookup/external-ref?access_num=000334173000010&link_type=ISI) 42. Mason BL, Pariante CM, Jamel S, et al. Central nervous system (CNS) delivery of glucocorticoids is fine-tuned by saturable transporters at the blood-CNS barriers and nonbarrier regions. Endocrinology 2010;151:5294–305. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1210/en.2010-0554&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=20881247&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) [Web of Science](http://jpn.ca/lookup/external-ref?access_num=000283231700023&link_type=ISI) 43. Tena-Garitaonaindia M, Arredondo-Amador M, Mascaraque C, et al. Modulation of intestinal barrier function by glucocorticoids: lessons from preclinical models. Pharmacol Res 2022;177:106056. 44. Greene C, Hanley N, Campbell M. Blood-brain barrier associated tight junction disruption is a hallmark feature of major psychiatric disorders. Transl Psychiatry 2020;10:373. 45. Kamintsky L, Cairns KA, Veksler R, et al. Blood-brain barrier imaging as a potential biomarker for bipolar disorder progression. Neuroimage Clin 2020;26:102049. 46. Kılıç F, Isık Ü, Demirdas A, et al. Serum zonulin and claudin-5 levels in patients with bipolar disorder. J Affect Disord 2020;266:37–42. 47. Severance EG, Gressitt KL, Stallings CR, et al. Discordant patterns of bacterial translocation markers and implications for innate immune imbalances in schizophrenia. Schizophr Res 2013;148:130–7. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1016/j.schres.2013.05.018&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=23746484&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) 48. Cheng Y, Desse S, Martinez A, et al. TNFα disrupts blood brain barrier integrity to maintain prolonged depressive-like behavior in mice. Brain Behav Immun 2018;69:556–67. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1016/j.bbi.2018.02.003&link_type=DOI) 49. Dion-Albert L, Cadoret A, Doney E, et al. Vascular and blood-brain barrier-related changes underlie stress responses and resilience in female mice and depression in human tissue. Nat Commun 2022;13:164. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1038/s41467-021-27604-x&link_type=DOI) 50. Matsuno H, Tsuchimine S, O’Hashi K, et al. Association between vascular endothelial growth factor-mediated blood-brain barrier dysfunction and stress-induced depression. Mol Psychiatry 2022;27:3822–32. 51. Cater M, Majdic G. How early maternal deprivation changes the brain and behavior? Eur J Neurosci 2022;55:2058–75. 52. DeVries AC, Glasper ER, Detillion CE. Social modulation of stress responses. Physiol Behav 2003;79:399–407. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1016/S0031-9384(03)00152-5&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=12954434&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) [Web of Science](http://jpn.ca/lookup/external-ref?access_num=000185291300007&link_type=ISI) 53. Kentner AC, Cryan JF, Brummelte S. Resilience priming: translational models for understanding resiliency and adaptation to early life adversity. Dev Psychobiol 2019;61:350–75. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1002/dev.21775&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=30311210&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) 54. Lorant V, Deliège D, Eaton W, et al. Socioeconomic inequalities in depression: a meta-analysis. Am J Epidemiol 2003;157:98–112. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1093/aje/kwf182&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=12522017&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) [Web of Science](http://jpn.ca/lookup/external-ref?access_num=000180359300003&link_type=ISI) 55. Teychenne M, Ball K, Salmon J. Sedentary behavior and depression among adults: a review. Int J Behav Med 2010;17:246–54. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1007/s12529-010-9075-z&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=20174982&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) [Web of Science](http://jpn.ca/lookup/external-ref?access_num=000290478100002&link_type=ISI) 56. Cohen S, Wills TA. Stress, social support, and the buffering hypothesis. Psychol Bull 1985;98:310–57. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1037/0033-2909.98.2.310&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=3901065&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) [Web of Science](http://jpn.ca/lookup/external-ref?access_num=A1985AQC4600006&link_type=ISI) 57. Spielman LJ, Little JP, Klegeris A. Physical activity and exercise attenuate neuroinflammation in neurological diseases. Brain Res Bull 2016;125:19–29. 58. Lehmann ML, Herkenham M. Environmental enrichment confers stress resiliency to social defeat through an infralimbic cortex-dependent neuroanatomical pathway. J Neurosci 2011;31:6159–73. [Abstract/FREE Full Text](http://jpn.ca/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6Njoiam5ldXJvIjtzOjU6InJlc2lkIjtzOjEwOiIzMS8xNi82MTU5IjtzOjQ6ImF0b20iO3M6MTk6Ii9qcG4vNDgvMy9FMTkwLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 59. Mul JD, Soto M, Cahill ME, et al. Voluntary wheel running promotes resilience to chronic social defeat stress in mice: a role for nucleus accumbens ΔFosB. Neuropsychopharmacology 2018;43:1934–42. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1038/s41386-018-0103-z&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=29875450&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) 60. Griñan-Ferré C, Pérez-Cáceres D, Gutiérrez-Zetina SM, et al. Environmental enrichment improves behavior, cognition, and brain functional markers in young senescence-accelerated prone mice (SAMP8). Mol Neurobiol 2016;53:2435–50. 61. Leardini-Tristão M, Andrade G, Garcia C, et al. Physical exercise promotes astrocyte coverage of microvessels in a model of chronic cerebral hypoperfusion. J Neuroinflammation 2020;17:117. 62. Robison LS, Francis N, Popescu DL, et al. Environmental enrichment: disentangling the influence of novelty, social, and physical activity on cerebral amyloid angiopathy in a transgenic mouse model. Int J Mol Sci 2020;21:843. 63. Sirevaag AM, Black JE, Shafron D, et al. Direct evidence that complex experience increases capillary branching and surface area in visual cortex of young rats. Brain Res 1988;471:299–304. [PubMed](http://jpn.ca/lookup/external-ref?access_num=3179754&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) 64. Williams ZAP, Szyszkowicz JK, Osborne N, et al. Sex-specific effects of voluntary wheel running on behavior and the gut microbiota- immune-brain axis in mice. Brain Behav Immun-Health 2023;30:100628. 65. Leichsenring F, Steinert C, Rabung S, et al. The efficacy of psychotherapies and pharmacotherapies for mental disorders in adults: an umbrella review and meta-analytic evaluation of recent meta-analyses. World Psychiatry 2022;21:133–45. 66. Ratuski AS, Weary DM. Environmental enrichment for rats and mice housed in laboratories: a metareview. Animals (Basel) 2022;12:414. 67. Isaacs KR, Anderson BJ, Alcantara AA, et al. Exercise and the brain: angiogenesis in the adult rat cerebellum after vigorous physical activity and motor skill learning. J Cereb Blood Flow Metab 1992;12:110–9. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1038/jcbfm.1992.14&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=1370068&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) [Web of Science](http://jpn.ca/lookup/external-ref?access_num=A1992GV64300013&link_type=ISI) 68. Olson AK, Eadie BD, Ernst C, et al. Environmental enrichment and voluntary exercise massively increase neurogenesis in the adult hippocampus via dissociable pathways. Hippocampus 2006;16:250–60. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1002/hipo.20157&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=16411242&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) [Web of Science](http://jpn.ca/lookup/external-ref?access_num=000235910900007&link_type=ISI) 69. van Praag H, Kempermann G, Gage FH. Running increases cell proliferation and neurogenesis in the adult mouse dentate gyrus. Nat Neurosci 1999;2:266–70. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1038/6368&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=10195220&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) [Web of Science](http://jpn.ca/lookup/external-ref?access_num=000081684500015&link_type=ISI) 70. Zhang X, Liu J-Y, Liao W-J, et al. Differential effects of physical and social enriched environment on angiogenesis in male rats after cerebral ischemia/reperfusion injury. Front Hum Neurosci 2021;15:622911. [PubMed](http://jpn.ca/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) 71. Mustroph ML, Chen S, Desai SC, et al. Aerobic exercise is the critical variable in an enriched environment that increases hippocampal neurogenesis and water maze learning in male C57BL/6J mice. Neuroscience 2012;219:62–71. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1016/j.neuroscience.2012.06.007&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=22698691&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) [Web of Science](http://jpn.ca/lookup/external-ref?access_num=000307417000006&link_type=ISI) 72. Rogers J, Vo U, Buret LS, et al. Dissociating the therapeutic effects of environmental enrichment and exercise in a mouse model of anxiety with cognitive impairment. Transl Psychiatry 2016;6:e794. 73. Queen NJ, Hassan QN, Cao L. Improvements to healthspan through environmental enrichment and lifestyle interventions: Where are we now? Front Neurosci 2020;14:605. 74. Cacioppo JT, Hawkley LC, Norman GJ, et al. Social isolation. Ann N Y Acad Sci 2011;1231:17–22. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1111/j.1749-6632.2011.06028.x&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=21651565&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) [Web of Science](http://jpn.ca/lookup/external-ref?access_num=000295542200003&link_type=ISI) 75. Moriguchi Y. The early development of executive function and its relation to social interaction: a brief review. Front Psychol 2014;5:388. 76. Shemesh Y, Sztainberg Y, Forkosh O, et al. High-order social interactions in groups of mice. eLife 2013;2:e00759. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.7554/eLife.00759&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=24015357&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) 77. Hawkley LC, Capitanio JP. Perceived social isolation, evolutionary fitness and health outcomes: a lifespan approach. Philos Trans R Soc Lond B Biol Sci 2015;370:20140114. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1098/rstb.2014.0114&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=25870400&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) 78. Krishnan V, Han M-H, Graham DL, et al. Molecular adaptations underlying susceptibility and resistance to social defeat in brain reward regions. Cell 2007;131:391–404. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1016/j.cell.2007.09.018&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=17956738&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) [Web of Science](http://jpn.ca/lookup/external-ref?access_num=000250507700027&link_type=ISI) 79. Park G, Ryu C, Kim S, et al. Social isolation impairs the prefrontal-nucleus accumbens circuit subserving social recognition in mice. Cell Rep 2021;35:109104. 80. Cacioppo JT, Hawkley LC, Thisted RA. Perceived social isolation makes me sad: 5-year cross-lagged analyses of loneliness and depressive symptomatology in the Chicago Health, Aging, and Social Relations Study. Psychol Aging 2010;25:453–63. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1037/a0017216&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=20545429&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) [Web of Science](http://jpn.ca/lookup/external-ref?access_num=000278702500019&link_type=ISI) 81. Alshammari TK, Alghamdi HM, Alduhailan HE, et al. Examining the central effects of chronic stressful social isolation on rats. Biomed Rep 2020;13:56. 82. Fulenwider HD, Caruso MA, Ryabinin AE. Manifestations of domination: assessments of social dominance in rodents. Genes Brain Behav 2022;21:e12731. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1111/gbb.12731&link_type=DOI) 83. Karailiev P, Hlavacova N, Chmelova M, et al. Tight junction proteins in the small intestine and prefrontal cortex of female rats exposed to stress of chronic isolation starting early in life. Neurogastroenterol Motil 2021;33:e14084. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1111/nmo.14084&link_type=DOI) 84. Lauer J, Zhou M, Ye S, et al. Multi-animal pose estimation, identification and tracking with DeepLabCut. Nat Methods 2022;19:496–504. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1038/s41592-022-01443-0&link_type=DOI) 85. Winters C, Gorssen W, Ossorio-Salazar VA, et al. Automated procedure to assess pup retrieval in laboratory mice. Sci Rep 2022;12:1663. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1038/s41598-022-05641-w&link_type=DOI) 86. Thorsteinsson EB, James JE. A Meta-analysis of the effects of experimental manipulations of social support during laboratory stress. Psychol Health 1999;14:869–86. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1080/08870449908407353&link_type=DOI) [Web of Science](http://jpn.ca/lookup/external-ref?access_num=000083823500007&link_type=ISI) 87. Yang YC, Schorpp K, Harris KM. Social support, social strain and inflammation: Evidence from a national longitudinal study of U.S. adults. Soc Sci Med 2014;107:124–35. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1016/j.socscimed.2014.02.013&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=24607674&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) 88. McCormick CM, Smith K, Baumbach JL, et al. Adolescent social instability stress leads to immediate and lasting sex-specific changes in the neuroendocrine-immune-gut axis in rats. Horm Behav 2020;126:104845. 89. Gibb J, Hayley S, Gandhi R, et al. Synergistic and additive actions of a psychosocial stressor and endotoxin challenge: circulating and brain cytokines, plasma corticosterone and behavioral changes in mice. Brain Behav Immun 2008;22:573–89. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1016/j.bbi.2007.12.001&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=18191534&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) [Web of Science](http://jpn.ca/lookup/external-ref?access_num=000255724800018&link_type=ISI) 90. Lee W, Milewski TM, Dwortz MF, et al. Distinct immune and transcriptomic profiles in dominant versus subordinate males in mouse social hierarchies. Brain Behav Immun 2022;103:130–44. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1016/j.bbi.2022.04.015&link_type=DOI) 91. VanRyzin JW, Marquardt AE, Argue KJ, et al. Microglial Phagocytosis of newborn cells is induced by endocannabinoids and sculpts sex differences in juvenile rat social play. Neuron 2019;102:435–49.e6. 92. VanRyzin JW, Marquardt AE, Pickett LA, et al. Microglia and sexual differentiation of the developing brain: a focus on extrinsic factors. Glia 2020;68:1100–13. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1002/glia.23740&link_type=DOI) 93. Li H, Tofigh AM, Amirfakhraei A, et al. Modulation of astrocyte activity and improvement of oxidative stress through blockage of NO/NMDAR pathway improve posttraumatic stress disorder (PTSD)-like behavior induced by social isolation stress. Brain Behav 2022;12:e2620. 94. Watanabe S, Omran AA, Shao AS, et al. Dihydromyricetin improves social isolation-induced cognitive impairments and astrocytic changes in mice. Sci Rep 2022;12:5899. 95. Dion-Albert L, Dudek KA, Russo SJ, et al. Neurovascular adaptations modulating cognition, mood, and stress responses. Trends Neurosci 2023 Feb. 17. doi: 10.1016/j.tins.2023.01.005 [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1016/j.tins.2023.01.005&link_type=DOI) 96. Schiavone S, Mhillaj E, Neri M, et al. Early loss of blood-brain barrier integrity precedes *NOX2* elevation in the prefrontal cortex of an animal model of psychosis. Mol Neurobiol 2017;54:2031–44. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1007/s12035-016-9791-8&link_type=DOI) 97. Hoxha V, Lama C, Chang PL, et al. Sex-specific signaling in the blood-brain barrier is required for male courtship in *Drosophila.* PLoS Genet 2013;9:e1003217. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1371/journal.pgen.1003217&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=23359644&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) 98. Reiter RJ, Mayo JC, Tan D-X, et al. Melatonin as an antioxidant: under promises but over delivers. J Pineal Res 2016;61:253–78. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1111/jpi.12360&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=27500468&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) 99. Dudek KA, Dion-Albert L, Lebel M, et al. Molecular adaptations of the blood-brain barrier promote stress resilience vs. depression. Proc Natl Acad Sci U S A 2020;117:3326–36. [Abstract/FREE Full Text](http://jpn.ca/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NDoicG5hcyI7czo1OiJyZXNpZCI7czoxMDoiMTE3LzYvMzMyNiI7czo0OiJhdG9tIjtzOjE5OiIvanBuLzQ4LzMvRTE5MC5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 100.Vanuytsel T, van Wanrooy S, Vanheel H, et al. Psychological stress and corticotropin-releasing hormone increase intestinal permeability in humans by a mast cell-dependent mechanism. Gut 2014;63:1293–9. [Abstract/FREE Full Text](http://jpn.ca/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NjoiZ3V0am5sIjtzOjU6InJlc2lkIjtzOjk6IjYzLzgvMTI5MyI7czo0OiJhdG9tIjtzOjE5OiIvanBuLzQ4LzMvRTE5MC5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 101.Kim C-S, Shin G-E, Cheong Y, et al. Experiencing social exclusion changes gut microbiota composition. Transl Psychiatry 2022;12:254. 102.Tillisch K, Mayer EA, Gupta A, et al. Brain structure and response to emotional stimuli as related to gut microbial profiles in healthy women. Psychosom Med 2017;79:905–13. [PubMed](http://jpn.ca/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) 103.Liu D, Jiang X-Y, Zhou L-S. Enriched environment on the intestinal mucosal barrier and brain-gut axis in rats with colorectal cancer. Exp Biol Med (Maywood) 2018;243:1185–98. 104.Donovan M, Mackey CS, Platt GN, et al. Social isolation alters behavior, the gut-immune-brain axis, and neurochemical circuits in male and female prairie voles. Neurobiol Stress 2020;13:100278. 105.Szyszkowicz JK, Wong A, Anisman H, et al. Implications of the gut microbiota in vulnerability to the social avoidance effects of chronic social defeat in male mice. Brain Behav Immun 2017;66:45–55. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1016/j.bbi.2017.06.009&link_type=DOI) 106.Wang T, Xu J, Xu Y, et al. Gut microbiota shapes social dominance through modulating HDAC2 in the medial prefrontal cortex. Cell Rep 2022;38:110478. 107.Argandoña EG, Bengoetxea H, Lafuente JV. Lack of experience-mediated differences in the immunohistochemical expression of blood-brain barrier markers (EBA and GluT-1) during the postnatal development of the rat visual cortex. Brain Res Dev Brain Res 2005;156:158–66. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1016/j.devbrainres.2005.02.007&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=16099302&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) 108.Moriguchi Y, Shinohara I. Socioeconomic disparity in prefrontal development during early childhood. Sci Rep 2019;9:2585. 109.Brenes Sáenz JC, Villagra OR, Fornaguera Trías J. Factor analysis of Forced Swimming test, Sucrose Preference test and Open Field test on enriched, social and isolated reared rats. Behav Brain Res 2006;169:57–65. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1016/j.bbr.2005.12.001&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=16414129&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) [Web of Science](http://jpn.ca/lookup/external-ref?access_num=000236136800007&link_type=ISI) 110.Perez JA, Clinton SM, Turner CA, et al. A new role for FGF2 as an endogenous inhibitor of anxiety. J Neurosci 2009;29:6379–87. [Abstract/FREE Full Text](http://jpn.ca/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6Njoiam5ldXJvIjtzOjU6InJlc2lkIjtzOjEwOiIyOS8xOS82Mzc5IjtzOjQ6ImF0b20iO3M6MTk6Ii9qcG4vNDgvMy9FMTkwLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 111.Meshi D, Drew MR, Saxe M, et al. Hippocampal neurogenesis is not required for behavioral effects of environmental enrichment. Nat Neurosci 2006;9:729–31. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1038/nn1696&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=16648847&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) [Web of Science](http://jpn.ca/lookup/external-ref?access_num=000237895200009&link_type=ISI) 112.Scala F, Nenov MN, Crofton EJ, et al. Environmental enrichment and social isolation mediate neuroplasticity of medium spiny neurons through the GSK3 pathway. Cell Rep 2018;23:555–67. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1016/j.celrep.2018.03.062&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=29642012&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) 113.van Praag H, Kempermann G, Gage FH. Neural consequences of enviromental enrichment. Nat Rev Neurosci 2000;1:191–8. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1038/35044558&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=11257907&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) [Web of Science](http://jpn.ca/lookup/external-ref?access_num=000165764700017&link_type=ISI) 114.McQuaid RJ, Dunn R, Jacobson-Pick S, et al. Post-weaning environmental enrichment in male CD-1 mice: impact on social behaviors, corticosterone levels and prefrontal cytokine expression in adulthood. Front Behav Neurosci 2018;12:145. 115.Cadoret A, Dion-Albert L, Amrani S, et al. Environmental conditions of recognition memory testing induce neurovascular changes in the hippocampus in a sex-specific manner in mice. Behav Brain Res 2023;448:114443. 116.Kerr AL, Steuer EL, Pochtarev V, et al. Angiogenesis but not neurogenesis is critical for normal learning and memory acquisition. Neuroscience 2010;171:214–26. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1016/j.neuroscience.2010.08.008&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=20804819&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) [Web of Science](http://jpn.ca/lookup/external-ref?access_num=000284016300021&link_type=ISI) 117.Ortuzar N, Rico-Barrio I, Bengoetxea H, et al. VEGF reverts the cognitive impairment induced by a focal traumatic brain injury during the development of rats raised under environmental enrichment. Behav Brain Res 2013;246:36–46. 118.Wang S, Chen L, Zhang L, et al. Effects of long-term exercise on spatial learning, memory ability, and cortical capillaries in aged rats. Med Sci Monit 2015;21:945–54. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.12659/MSM.893935&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=25828032&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) 119.He C, Tsipis CP, LaManna JC, et al. Environmental enrichment induces increased cerebral capillary density and improved cognitive function in mice. Adv Exp Med Biol 2017;977:175–81. 120.Salois G, Smith JS. Housing complexity alters GFAP-immunoreactive astrocyte morphology in the rat dentate gyrus. Neural Plast 2016;2016:3928726. 121.Jones TA, Greenough WT. Ultrastructural evidence for increased contact between astrocytes and synapses in rats reared in a complex environment. Neurobiol Learn Mem 1996;65:48–56. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1006/nlme.1996.0005&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=8673406&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) [Web of Science](http://jpn.ca/lookup/external-ref?access_num=A1996TP83700005&link_type=ISI) 122.Pusic KM, Pusic AD, Kemme J, et al. Spreading depression requires microglia and is decreased by their M2a polarization from environmental enrichment. Glia 2014;62:1176–94. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1002/glia.22672&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=24723305&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) [Web of Science](http://jpn.ca/lookup/external-ref?access_num=000336001100012&link_type=ISI) 123.Bekinschtein P, Oomen CA, Saksida LM, et al. Effects of environmental enrichment and voluntary exercise on neurogenesis, learning and memory, and pattern separation: BDNF as a critical variable? Semin Cell Dev Biol 2011;22:536–42. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1016/j.semcdb.2011.07.002&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=21767656&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) 124.Gómez-Pinilla F, So V, Kesslak JP. Spatial learning and physical activity contribute to the induction of fibroblast growth factor: neural substrates for increased cognition associated with exercise. Neuroscience 1998;85:53–61. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1016/S0306-4522(97)00576-9&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=9607702&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) [Web of Science](http://jpn.ca/lookup/external-ref?access_num=000073098300005&link_type=ISI) 125.Seo JH, Yu JH, Suh H, et al. Fibroblast growth factor-2 induced by enriched environment enhances angiogenesis and motor function in chronic hypoxic-ischemic brain injury. PLoS One 2013;8:e74405. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1371/journal.pone.0074405&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=24098645&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) 126.Zhan Y, Li M-Z, Yang L, et al. The three-phase enriched environment paradigm promotes neurovascular restorative and prevents learning impairment after ischemic stroke in rats. Neurobiol Dis 2020;146:105091. 127.Beauquis J, Roig P, De Nicola AF, et al. Short-term environmental enrichment enhances adult neurogenesis, vascular network and dendritic complexity in the hippocampus of type 1 diabetic mice. PLoS One 2010;5:e13993. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1371/journal.pone.0013993&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=21085588&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) 128.Birch AM, Kelly ÁM. Lifelong environmental enrichment in the absence of exercise protects the brain from age-related cognitive decline. Neuropharmacology 2019;145:59–74. 129.Qu C, Xu L, Shen J, et al. Protection of blood-brain barrier as a potential mechanism for enriched environments to improve cognitive impairment caused by chronic cerebral hypoperfusion. Behav Brain Res 2020;379:112385. 130.Song MK, Kim YJ, Lee J-M, et al. Neurovascular integrative effects of long-term environmental enrichment on chronic cerebral hypoperfusion rat model. Brain Res Bull 2020;163:160–9. 131.Xu L, Qu C, Qu C, et al. Improvement of autophagy dysfunction as a potential mechanism for environmental enrichment to protect blood-brain barrier in rats with vascular cognitive impairment. Neurosci Lett 2020;739:135437. 132.Jin X, Li T, Zhang L, et al. Environmental enrichment improves spatial learning and memory in vascular dementia rats with activation of Wnt/β-catenin signal pathway. Med Sci Monit 2017;23:207–15. 133.Marrocco F, Delli Carpini M, Garofalo S, et al. Short-chain fatty acids promote the effect of environmental signals on the gut microbiome and metabolome in mice. Commun Biol 2022;5:517. 134.Gubert C, Love CJ, Kodikara S, et al. Gene-environment-gut interactions in Huntington’s disease mice are associated with environmental modulation of the gut microbiome. iScience 2021;25:103687. 135.Bice BD, Stephens MR, Georges SJ, et al. Environmental enrichment induces pericyte and IgA-dependent wound repair and lifespan extension in a colon tumor model. Cell Rep 2017;19:760–73. 136.Singh Y, El-Hadidi M, Admard J, et al. Enriched environmental conditions modify the gut microbiome composition and fecal markers of inflammation in Parkinson’s disease. Front Neurosci 2019;13:1032. 137.Dishman RK, Berthoud H-R, Booth FW, et al. Neurobiology of exercise. Obesity (Silver Spring) 2006;14:345–56. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1038/oby.2006.46&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=16648603&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) 138.Petersen AMW, Pedersen BK. The anti-inflammatory effect of exercise. J Appl Physiol (1985) 2005;98:1154–62. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1152/japplphysiol.00164.2004&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=15772055&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) [Web of Science](http://jpn.ca/lookup/external-ref?access_num=000227665400002&link_type=ISI) 139.Greenwood BN, Foley TE, Le TV, et al. Long-term voluntary wheel running is rewarding and produces plasticity in the mesolimbic reward pathway. Behav Brain Res 2011;217:354–62. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1016/j.bbr.2010.11.005&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=21070820&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) [Web of Science](http://jpn.ca/lookup/external-ref?access_num=000286698300012&link_type=ISI) 140.Cohen DA, McKenzie TL, Sehgal A, et al. Contribution of public parks to physical activity. Am J Public Health 2007;97:509–14. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.2105/AJPH.2005.072447&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=17267728&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) [Web of Science](http://jpn.ca/lookup/external-ref?access_num=000244636500028&link_type=ISI) 141.Parks SE, Housemann RA, Brownson RC. Differential correlates of physical activity in urban and rural adults of various socioeconomic backgrounds in the United States. J Epidemiol Community Health 2003;57:29–35. [Abstract/FREE Full Text](http://jpn.ca/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NDoiamVjaCI7czo1OiJyZXNpZCI7czo3OiI1Ny8xLzI5IjtzOjQ6ImF0b20iO3M6MTk6Ii9qcG4vNDgvMy9FMTkwLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 142.Manzanares G, Brito-da-Silva G, Gandra PG. Voluntary wheel running: patterns and physiological effects in mice. Braz J Med Biol Res 2018;52:e7830. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1590/1414-431X20187830&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=30539969&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) 143.Novak CM, Burghardt PR, Levine JA. The use of a running wheel to measure activity in rodents: relationship to energy balance, general activity, and reward. Neurosci Biobehav Rev 2012;36:1001–14. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1016/j.neubiorev.2011.12.012&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=22230703&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) 144.Bastioli G, Arnold JC, Mancini M, et al. Voluntary exercise boosts striatal dopamine release: evidence for the necessary and sufficient role of BDNF. J Neurosci 2022;42:4725–36. [Abstract/FREE Full Text](http://jpn.ca/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6Njoiam5ldXJvIjtzOjU6InJlc2lkIjtzOjEwOiI0Mi8yMy80NzI1IjtzOjQ6ImF0b20iO3M6MTk6Ii9qcG4vNDgvMy9FMTkwLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 145.Steensberg A, Fischer CP, Keller C, et al. IL-6 enhances plasma IL-1ra, IL-10, and cortisol in humans. Am J Physiol Endocrinol Metab 2003;285:E433–7. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1152/ajpendo.00074.2003&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=12857678&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) [Web of Science](http://jpn.ca/lookup/external-ref?access_num=000184205400024&link_type=ISI) 146.Steensberg A, Keller C, Starkie RL, et al. IL-6 and TNF-α expression in, and release from, contracting human skeletal muscle. Am J Physiol Endocrinol Metab 2002;283:E1272–8. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1152/ajpendo.00255.2002&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=12388119&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) [Web of Science](http://jpn.ca/lookup/external-ref?access_num=000179080300021&link_type=ISI) 147.Mailing LJ, Allen JM, Buford TW, et al. Exercise and the gut microbiome: a review of the evidence, potential mechanisms, and implications for human health. Exerc Sport Sci Rev 2019;47:75–85. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1249/JES.0000000000000183&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) 148.Matsumoto M, Inoue R, Tsukahara T, et al. Voluntary running exercise alters microbiota composition and increases n-butyrate concentration in the rat cecum. Biosci Biotechnol Biochem 2008;72:572–6. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1271/bbb.70474&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=18256465&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) [Web of Science](http://jpn.ca/lookup/external-ref?access_num=000253892800039&link_type=ISI) 149.Tharp GD. The role of glucocorticoids in exercise. Med Sci Sports 1975;7:6–11. [PubMed](http://jpn.ca/lookup/external-ref?access_num=1143055&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) [Web of Science](http://jpn.ca/lookup/external-ref?access_num=A1975AB17000003&link_type=ISI) 150.Duclos M, Gouarne C, Bonnemaison D. Acute and chronic effects of exercise on tissue sensitivity to glucocorticoids. J Appl Physiol 1985 2003;94:869–75. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1152/japplphysiol.00108.2002&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=12433870&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) [Web of Science](http://jpn.ca/lookup/external-ref?access_num=000180827400004&link_type=ISI) 151.Svensson M, Rosvall P, Boza-Serrano A, et al. Forced treadmill exercise can induce stress and increase neuronal damage in a mouse model of global cerebral ischemia. Neurobiol Stress 2016;5:8–18. 152.Chen C, Nakagawa S, An Y, et al. The exercise-glucocorticoid paradox: how exercise is beneficial to cognition, mood, and the brain while increasing glucocorticoid levels. Front Neuroendocrinol 2017;44:83–102. 153.Amat J, Baratta MV, Paul E, et al. Medial prefrontal cortex determines how stressor controllability affects behavior and dorsal raphe nucleus. Nat Neurosci 2005;8:365–71. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1038/nn1399&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=15696163&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) [Web of Science](http://jpn.ca/lookup/external-ref?access_num=000227354600022&link_type=ISI) 154.Sandi C. Stress and cognition. Wiley Interdiscip Rev Cogn Sci 2013;4:245–61. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1002/wcs.1222)&link_type=DOI) 155.Adkins AM, Wellman LL, Sanford LD. Controllable and uncontrollable stress differentially impact fear conditioned alterations in sleep and neuroimmune signaling in mice. Life (Basel) 2022;12:1320. 156.Querido JS, Sheel AW. Regulation of cerebral blood flow during exercise. Sports Med 2007;37:765–82. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.2165/00007256-200737090-00002&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=17722948&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) [Web of Science](http://jpn.ca/lookup/external-ref?access_num=000249839500002&link_type=ISI) 157.Salzman T, Dupuy O, Fraser SA. Effects of cardiorespiratory fitness on cerebral oxygenation in healthy adults: a systematic review. Front Physiol 2022;13:838450. 158.Willie CK, Cowan EC, Ainslie PN, et al. Neurovascular coupling and distribution of cerebral blood flow during exercise. J Neurosci Methods 2011;198:270–3. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1016/j.jneumeth.2011.03.017&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=21459113&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) [Web of Science](http://jpn.ca/lookup/external-ref?access_num=000292435900015&link_type=ISI) 159.Brooks S, Branyan KW, DeVallance E, et al. Psychological stress-induced cerebrovascular dysfunction: the role of metabolic syndrome and exercise. Exp Physiol 2018;103:761–76. 160.Morland C, Andersson KA, Haugen ØP, et al. Exercise induces cerebral VEGF and angiogenesis via the lactate receptor HCAR1. Nat Commun 2017;8:15557. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1038/ncomms15557&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=28534495&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) 161.Saur L, Baptista PPA, de Senna PN, et al. Physical exercise increases GFAP expression and induces morphological changes in hippocampal astrocytes. Brain Struct Funct 2014;219:293–302. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1007/s00429-012-0500-8&link_type=DOI) 162.Lundquist AJ, Parizher J, Petzinger GM, et al. Exercise induces region-specific remodeling of astrocyte morphology and reactive astrocyte gene expression patterns in male mice. J Neurosci Res 2019;97:1081–94. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1002/jnr.24430&link_type=DOI) 163.Brockett AT, LaMarca EA, Gould E. Physical exercise enhances cognitive flexibility as well as astrocytic and synaptic markers in the medial prefrontal cortex. PLoS One 2015;10:e0124859. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1371/journal.pone.0124859&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=25938418&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) 164.Nagelhus EA, Ottersen OP. Physiological roles of aquaporin-4 in brain. Physiol Rev 2013;93:1543–62. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1152/physrev.00011.2013&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=24137016&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) 165.He X-F, Liu D-X, Zhang Q, et al. Voluntary exercise promotes glymphatic clearance of amyloid beta and reduces the activation of astrocytes and microglia in aged mice. Front Mol Neurosci 2017;10:144. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.3389/fnmol.2017.00144&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=28579942&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) 166.Kiuchi T, Lee H, Mikami T. Regular exercise cures depression-like behavior via VEGF-Flk-1 signaling in chronically stressed mice. Neuroscience 2012;207:208–17. 167.Rich B, Scadeng M, Yamaguchi M, et al. Skeletal myofiber vascular endothelial growth factor is required for the exercise training-induced increase in dentate gyrus neuronal precursor cells: skeletal myofiber VEGF-dependent cerebral neurogenesis. J Physiol 2017;595:5931–43. 168.Wolff G, Davidson SJ, Wrobel JK, et al. Exercise maintains blood-brain barrier integrity during early stages of brain metastasis formation. Biochem Biophys Res Commun 2015;463:811–7. 169.de Senna PN, Xavier LL, Bagatini PB, et al. Physical training improves non-spatial memory, locomotor skills and the blood brain barrier in diabetic rats. Brain Res 2015;1618:75–82. 170.Souza PS, Gonçalves ED, Pedroso GS, et al. Physical exercise attenuates experimental autoimmune encephalomyelitis by inhibiting peripheral immune response and blood-brain barrier disruption. Mol Neurobiol 2017;54:4723–37. [PubMed](http://jpn.ca/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) 171.Isla AG, Vázquez-Cuevas FG, Peña-Ortega F. Exercise prevents amyloid-β-induced hippocampal network disruption by inhibiting GSK3β activation. J Alzheimers Dis 2016;52:333–43. 172.Ramirez SH, Fan S, Dykstra H, et al. Inhibition of glycogen synthase kinase 3β promotes tight junction stability in brain endothelial cells by half-life extension of occludin and claudin-5. PLoS One 2013;8:e55972. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1371/journal.pone.0055972&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=23418486&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) 173.Ramirez SH, Fan S, Zhang M, et al. Inhibition of glycogen synthase kinase 3β (GSK3β) decreases inflammatory responses in brain endothelial cells. Am J Pathol 2010;176:881–92. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.2353/ajpath.2010.090671&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=20056834&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) [Web of Science](http://jpn.ca/lookup/external-ref?access_num=000274111400036&link_type=ISI) 174.De Miguel Z, Khoury N, Betley MJ, et al. Exercise plasma boosts memory and dampens brain inflammation via clusterin. Nature 2021;600:494–9. 175.Sinha RK, Yang XV, Fernández JA, et al. Apolipoprotein E receptor 2 mediates activated protein C-induced endothelial Akt activation and endothelial barrier stabilization. Arterioscler Thromb Vasc Biol 2016;36:518–24. [Abstract/FREE Full Text](http://jpn.ca/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NzoiYXR2YmFoYSI7czo1OiJyZXNpZCI7czo4OiIzNi8zLzUxOCI7czo0OiJhdG9tIjtzOjE5OiIvanBuLzQ4LzMvRTE5MC5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 176.Yang XV, Banerjee Y, Fernández JA, et al. Activated protein C ligation of ApoER2 (LRP8) causes Dab1-dependent signaling in U937 cells. Proc Natl Acad Sci U S A 2009;106:274–9. [Abstract/FREE Full Text](http://jpn.ca/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NDoicG5hcyI7czo1OiJyZXNpZCI7czo5OiIxMDYvMS8yNzQiO3M6NDoiYXRvbSI7czoxOToiL2pwbi80OC8zL0UxOTAuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 177.Ribeiro FM, Petriz B, Marques G, et al. Is there an exercise-intensity threshold capable of avoiding the leaky gut? Front Nutr 2021;8:627289. 178.van Wijck K, Lenaerts K, van Loon LJC, et al. Exercise-Induced Splanchnic Hypoperfusion Results in Gut Dysfunction in Healthy Men. PLoS One 2011;6:e22366. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1371/journal.pone.0022366&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=21811592&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) 179.Walter E, Watt PW, Gibson OR, et al. Exercise hyperthermia induces greater changes in gastrointestinal permeability than equivalent passive hyperthermia. Physiol Rep 2021;9:e14945. [PubMed](http://jpn.ca/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) 180.Hoffman-Goetz L, Pervaiz N, Packer N, et al. Freewheel training decreases pro- and increases anti-inflammatory cytokine expression in mouse intestinal lymphocytes. Brain Behav Immun 2010; 24:1105–15. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1016/j.bbi.2010.05.001&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=20510350&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) 181.Hoffman-Goetz L, Pervaiz N, Guan J. Voluntary exercise training in mice increases the expression of antioxidant enzymes and decreases the expression of TNF-alpha in intestinal lymphocytes. Brain Behav Immun 2009;23:498–506. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1016/j.bbi.2009.01.015&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=19486647&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) 182.Lancaster GI, Khan Q, Drysdale P, et al. The physiological regulation of toll-like receptor expression and function in humans. J Physiol 2005;563:945–55. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1113/jphysiol.2004.081224&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=15661814&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) [Web of Science](http://jpn.ca/lookup/external-ref?access_num=000227924500023&link_type=ISI) 183.Moitinho-Silva L, Wegener M, May S, et al. Short-term physical exercise impacts on the human holobiont obtained by a randomised intervention study. BMC Microbiol 2021;21:162. 184.Clarke SF, Murphy EF, O’Sullivan O, et al. Exercise and associated dietary extremes impact on gut microbial diversity. Gut 2014;63:1913–20. [Abstract/FREE Full Text](http://jpn.ca/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NjoiZ3V0am5sIjtzOjU6InJlc2lkIjtzOjEwOiI2My8xMi8xOTEzIjtzOjQ6ImF0b20iO3M6MTk6Ii9qcG4vNDgvMy9FMTkwLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 185.Bycura D, Santos AC, Shiffer A, et al. Impact of different exercise modalities on the human gut microbiome. Sports (Basel) 2021;9:14. 186.Everard A, Belzer C, Geurts L, et al. Cross-talk between *Akkermansia muciniphila* and intestinal epithelium controls diet-induced obesity. Proc Natl Acad Sci U S A 2013;110:9066–71. [Abstract/FREE Full Text](http://jpn.ca/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NDoicG5hcyI7czo1OiJyZXNpZCI7czoxMToiMTEwLzIyLzkwNjYiO3M6NDoiYXRvbSI7czoxOToiL2pwbi80OC8zL0UxOTAuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 187.Quiroga R, Nistal E, Estébanez B, et al. Exercise training modulates the gut microbiota profile and impairs inflammatory signaling pathways in obese children. Exp Mol Med 2020;52:1048–61. 188.Allen JM, Mailing LJ, Niemiro GM, et al. Exercise alters gut microbiota composition and function in lean and obese humans. Med Sci Sports Exerc 2018;50:747–57. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1249/MSS.0000000000001495&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) 189.Peng L, Li Z-R, Green RS, et al. Butyrate enhances the intestinal barrier by facilitating tight junction assembly via activation of AMP-activated protein kinase in Caco-2 cell monolayers. J Nutr 2009;139:1619–25. [Abstract/FREE Full Text](http://jpn.ca/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6OToibnV0cml0aW9uIjtzOjU6InJlc2lkIjtzOjEwOiIxMzkvOS8xNjE5IjtzOjQ6ImF0b20iO3M6MTk6Ii9qcG4vNDgvMy9FMTkwLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 190.Beurel E, Toups M, Nemeroff CB. The bidirectional relationship of depression and inflammation: double trouble. Neuron 2020;107:234–56. 191.Kimura LF, Novaes LS, Picolo G, et al. How environmental enrichment balances out neuroinflammation in chronic pain and comorbid depression and anxiety disorders. Br J Pharmacol 2022;179:1640–60. 192.McCutcheon JE, Marinelli M. Age matters. Eur J Neurosci 2009; 29:997–1014. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1111/j.1460-9568.2009.06648.x&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=19291226&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) [Web of Science](http://jpn.ca/lookup/external-ref?access_num=000263755000014&link_type=ISI) 193.Delaney C, Campbell M. The blood brain barrier: insights from development and ageing. Tissue Barriers 2017;5:e1373897. 194.Weström B, Arévalo Sureda E, Pierzynowska K, et al. The immature gut barrier and its importance in establishing immunity in newborn mammals. Front Immunol 2020;11:1153. 195.Goasdoué K, Miller SM, Colditz PB, et al. Review: The blood-brain barrier; protecting the developing fetal brain. Placenta 2017;54:111–6. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1016/j.placenta.2016.12.005&link_type=DOI) 196.Wang Z, Zhang C, Huang F, et al. Breakthrough of ZrO2 nanoparticles into fetal brains depends on developmental stage of maternal placental barrier and fetal blood-brain-barrier. J Hazard Mater 2021;402:123563. 197.Brockmeyer S, D’Angiulli A. How air pollution alters brain development: the role of neuroinflammation. Transl Neurosci 2016;7:24–30. [PubMed](http://jpn.ca/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) 198.Bloise E, Petropoulos S, Iqbal M, et al. Acute effects of viral exposure on P-glycoprotein function in the mouse fetal blood-brain barrier. Cell Physiol Biochem 2017;41:1044–50. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1159/000461569&link_type=DOI) 199.Shao Q, Herrlinger S, Yang S-L, et al. Zika virus infection disrupts neurovascular development and results in postnatal microcephaly with brain damage. Development 2016;143:4127–36. [Abstract/FREE Full Text](http://jpn.ca/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NzoiZGV2ZWxvcCI7czo1OiJyZXNpZCI7czoxMToiMTQzLzIyLzQxMjciO3M6NDoiYXRvbSI7czoxOToiL2pwbi80OC8zL0UxOTAuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 200.Bordeleau M, Comin CH, Fernández de Cossío L, et al. Maternal high-fat diet in mice induces cerebrovascular, microglial and long-term behavioural alterations in offspring. Commun Biol 2022;5:26. 201.Coe CL, Lubach GR. Prenatal influences on neuroimmune set points in infancy. Ann N Y Acad Sci 2000;917:468–77. [PubMed](http://jpn.ca/lookup/external-ref?access_num=11268374&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) [Web of Science](http://jpn.ca/lookup/external-ref?access_num=000171939400045&link_type=ISI) 202.Gu J-Y, Xu Y-W, Feng L-P, et al. Enriched environment mitigates depressive behavior by changing the inflammatory activation phenotype of microglia in the hippocampus of depression model rats. Brain Res Bull 2021;177:252–62. 203.Han VX, Patel S, Jones HF, et al. Maternal acute and chronic inflammation in pregnancy is associated with common neurodevelopmental disorders: a systematic review. Transl Psychiatry 2021;11:71. 204.Lopes NA, Falkenberg EA, Wiley C, et al. Social isolation stress modulates pregnancy outcomes and the inflammatory profile of rat uterus. Int J Mol Sci 2022;23:6169. 205.Cutuli D, Berretta E, Pasqualini G, et al. Influence of pre-reproductive maternal enrichment on coping response to stress and expression of c-Fos and glucocorticoid receptors in adolescent offspring. Front Behav Neurosci 2017;11:73. 206.Welberg L, Thrivikraman K, Plotsky P. Combined pre- and postnatal environmental enrichment programs the HPA axis differentially in male and female rats. Psychoneuroendocrinology 2006;31:553–64. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1016/j.psyneuen.2005.11.011&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=16434144&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) 207.Schander JA, Marvaldi C, Correa F, et al. Maternal environmental enrichment modulates the immune response against an inflammatory challenge during gestation and protects the offspring. J Reprod Immunol 2021;144:103273. 208.Herring A, Donath A, Yarmolenko M, et al. Exercise during pregnancy mitigates Alzheimer-like pathology in mouse offspring. FASEB J 2012;26:117–28. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1096/fj.11-193193&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=21948247&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) [Web of Science](http://jpn.ca/lookup/external-ref?access_num=000299202200013&link_type=ISI) 209.He N, Kong Q-Q, Wang J-Z, et al. Parental life events cause behavioral difference among offspring: adult pre-gestational restraint stress reduces anxiety across generations. Sci Rep 2016;6:39497. 210.Kajimoto K, Valenzuela CF, Allan AM, et al. Prenatal alcohol exposure alters synaptic activity of adult hippocampal dentate granule cells under conditions of enriched environment. Hippocampus 2016;26:1078–87. 211.Duran-Carabali LE, Odorcyk FK, Greggio S, et al. Pre- and early postnatal enriched environmental experiences prevent neonatal hypoxia-ischemia late neurodegeneration via metabolic and neuroplastic mechanisms. J Neurochem 2021;157:1911–29. 212.Murthy S, Gould E. Early life stress in rodents: Animal models of illness or resilience? Front Behav Neurosci 2018;12:157. 213.Smith KE, Pollak SD. Early life stress and development: potential mechanisms for adverse outcomes. J Neurodev Disord 2020;12:34. 214.Li W, Zou J, Shang J, et al. Both the complexity of tight junctions and endothelial transcytosis are increased during BBB postnatal development in rats. Front Neurosci 2022;16:850857. 215.Bengoetxea H, Argandona EG, Lafuente JV. Effects of visual experience on vascular endothelial growth factor expression during the postnatal development of the rat visual cortex. Cereb Cortex 2008;18:1630–9. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1093/cercor/bhm190&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=17986606&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) 216.Workman JL, Fonken LK, Gusfa J, et al. Post-weaning environmental enrichment alters affective responses and interacts with behavioral testing to alter nNOS immunoreactivity. Pharmacol Biochem Behav 2011;100:25–32. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1016/j.pbb.2011.07.008.&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=21777607&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) 217.Collins JM, Caputi V, Manurung S, et al. Supplementation with milk fat globule membrane from early life reduces maternal separation-induced visceral pain independent of enteric nervous system or intestinal permeability changes in the rat. Neuropharmacology 2022;210:109026. 218.Kelly KM, Nadon NL, Morrison JH, et al. The neurobiology of aging. Epilepsy Res 2006;68(Suppl 1): S5–20. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1016/j.eplepsyres.2005.07.015&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=16386406&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) [Web of Science](http://jpn.ca/lookup/external-ref?access_num=000235984000002&link_type=ISI) 219.Mattson MP, Magnus T. Ageing and neuronal vulnerability. Nat Rev Neurosci 2006;7:278–94. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1038/nrn1886&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=16552414&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) [Web of Science](http://jpn.ca/lookup/external-ref?access_num=000236170400014&link_type=ISI) 220.Elahy M, Jackaman C, Mamo JC, et al. Blood-brain barrier dysfunction developed during normal aging is associated with inflammation and loss of tight junctions but not with leukocyte recruitment. Immun Ageing 2015;12:2. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1186/s12979-015-0029-9&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=25784952&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) 221.Montagne A, Barnes SR, Sweeney MD, et al. Blood-brain barrier breakdown in the aging human hippocampus. Neuron 2015;85:296–302. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1016/j.neuron.2014.12.032&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=25611508&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) 222.Vecchio LM, Meng Y, Xhima K, et al. The neuroprotective effects of exercise: maintaining a healthy brain throughout aging. Brain Plast 2018;4:17–52. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.3233/bpl-180069&link_type=DOI) 223.Latimer CS, Searcy JL, Bridges MT, et al. Reversal of glial and neurovascular markers of unhealthy brain aging by exercise in middle-aged female mice. PLoS One 2011;6:e26812. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1371/journal.pone.0026812&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=22046366&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) 224.McMurphy T, Huang W, Queen NJ, et al. Implementation of environmental enrichment after middle age promotes healthy aging. Aging (Albany NY) 2018;10:1698–721. 225.Soto I, Graham LC, Richter HJ, et al. APOE stabilization by exercise prevents aging neurovascular dysfunction and complement induction. PLoS Biol 2015;13:e1002279. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1371/journal.pbio.1002279&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=26512759&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) 226.Furman D, Campisi J, Verdin E, et al. Chronic inflammation in the etiology of disease across the life span. Nat Med 2019;25:1822–32. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1038/s41591-019-0675-0&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=31806905&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) 227.Huang H, Wang Q, Guan X, et al. Effects of enriched environment on depression and anxiety-like behavior induced by early life stress: a comparison between different periods. Behav Brain Res 2021;411:113389. 228.Ilin Y, Richter-Levin G. Enriched environment experience overcomes learning deficits and depressive-like behavior induced by juvenile stress. PLoS One 2009;4:e4329. [PubMed](http://jpn.ca/lookup/external-ref?access_num=19180243&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) 229.Global regional, and national burden of 12 mental disorders in 204 countries and territories 1990–2019: a systematic analysis for the Global Burden of Disease Study 2019. Lancet Psychiatry 2022;9:137–50. [PubMed](http://jpn.ca/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) 230.Krishnan V, Nestler EJ. The molecular neurobiology of depression. Nature 2008;455:894–902. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1038/nature07455&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=18923511&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) [Web of Science](http://jpn.ca/lookup/external-ref?access_num=000260038300037&link_type=ISI) 231.Pettersson E, Lichtenstein P, Larsson H, et al. Genetic influences on eight psychiatric disorders based on family data of 4 408 646 full and half-siblings, and genetic data of 333 748 cases and controls. Psychol Med 2019;49:1166–73. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1017/S0033291718002039&link_type=DOI) 232.Maes M, Kubera M, Leunis J-C. The gut-brain barrier in major depression: intestinal mucosal dysfunction with an increased translocation of LPS from gram negative enterobacteria (leaky gut) plays a role in the inflammatory pathophysiology of depression. Neuroendocrinol Lett 2008;29:117–24. [PubMed](http://jpn.ca/lookup/external-ref?access_num=18283240&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) 233.Niklasson F, Agren H. Brain energy metabolism and blood-brain barrier permeability in depressive patients: analyses of creatine, creatinine, urate, and albumin in CSF and blood. Biol Psychiatry 1984;19:1183–206. [PubMed](http://jpn.ca/lookup/external-ref?access_num=6498242&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) [Web of Science](http://jpn.ca/lookup/external-ref?access_num=A1984TN58900003&link_type=ISI) 234.Kirch DG, Kaufmann CA, Papadopoulos NM, et al. Abnormal cerebrospinal fluid protein indices in schizophrenia. Biol Psychiatry 1985;20:1039–46. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1016/0006-3223(85)90002-2&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=4041510&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) 235.Muscatell KA, Brosso SN, Humphreys KL. Socioeconomic status and inflammation: a meta-analysis. Mol Psychiatry 2020; 25:2189–99. [PubMed](http://jpn.ca/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) 236.Bowyer RCE, Jackson MA, Le Roy CI, et al. Socioeconomic status and the gut microbiome: a TwinsUK cohort study. Microorganisms 2019;7:17. 237.McEwen BS. Stressed or stressed out: What is the difference? J Psychiatry Neurosci 2005;30:315–8. 238.Guidi J, Lucente M, Sonino N, et al. Allostatic load and its impact on health: a systematic review. Psychother Psychosom 2021; 90:11–27. [PubMed](http://jpn.ca/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) 239.1. Fisher S, 2. Reason J Sterling P, Eyer J. Allostasis: a new paradigm to explain arousal pathology. In: Fisher S, Reason J, editors. Handbook of Life Stress, Cognition and Health. John Wiley & Sons; 1988:629–49. 240.Picard M, Juster R-P, McEwen BS. Mitochondrial allostatic load puts the “gluc” back in glucocorticoids. Nat Rev Endocrinol 2014;10:303–10. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1038/nrendo.2014.22&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=24663223&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) 241.Bhatnagar S. Rethinking stress resilience. Trends Neurosci 2021; 44:936–45. 242.1. Cicchetti D Juster R-P, Seeman T, McEwen BS, et al. Social inequalities and the road to allostatic load: from vulnerability to resilience. In: Cicchetti D, editors. Developmental Psychopathology. John Wiley & Sons; 2016: 381–434. Available: [https://onlinelibrary.wiley.com/doi/10.1002/9781119125556.devpsy408](https://onlinelibrary.wiley.com/doi/10.1002/9781119125556.devpsy408) (accessed 2022 Nov. 22). 243.Li Y, Luo Y, Tang J, et al. The positive effects of running exercise on hippocampal astrocytes in a rat model of depression. Transl Psychiatry 2021;11:83. 244.van de Wouw M, Boehme M, Lyte JM, et al. Short-chain fatty acids: microbial metabolites that alleviate stress-induced brain-gut axis alterations. J Physiol 2018;596:4923–44. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1113/JP276431&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=30066368&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) 245.Grundmann M, Bender E, Schamberger J, et al. Pharmacology of free fatty acid receptors and their allosteric modulators. Int J Mol Sci 2021;22:1763. 246.Lee GO, McCormick BJJ, Seidman JC, et al.Mal-Ed Network Investigators. Infant nutritional status, feeding practices, enteropathogen exposure, socioeconomic status, and illness are associated with gut barrier function as assessed by the lactulose mannitol test in the MAL-ED birth cohort. Am J Trop Med Hyg 2017;97:281–90. [PubMed](http://jpn.ca/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) 247.Martin M, Vermeiren S, Bostaille N, et al. Engineered Wnt ligands enable blood-brain barrier repair in neurological disorders. Science 2022;375:eabm4459. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1126/science.abm4459&link_type=DOI) 248.Greene C, Hanley N, Reschke CR, et al. Microvascular stabilization via blood-brain barrier regulation prevents seizure activity. Nat Commun 2022;13:2003. 249.Moffat JG, Vincent F, Lee JA, et al. Opportunities and challenges in phenotypic drug discovery: an industry perspective. Nat Rev Drug Discov 2017;16:531–43. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1038/nrd.2017.111&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=28685762&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) 250.Freland L, Beaulieu J-M. Inhibition of GSK3 by lithium, from single molecules to signaling networks. Front Mol Neurosci 2012;5:14. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.3389/fnmol.2012.00014&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=22363263&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) 251.Costemale-Lacoste JF, Guilloux JP, Gaillard R. The role of GSK-3 in treatment-resistant depression and links with the pharmacological effects of lithium and ketamine: a review of the literature. Encephale 2016;42:156–64. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1016/j.encep.2016.02.003&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=26995153&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) 252.Mohammed AH, Zhu SW, Darmopil S, et al. Environmental enrichment and the brain. Prog Brain Res 2002;138:109–33. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1016/S0079-6123(02)38074-9&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=12432766&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) [Web of Science](http://jpn.ca/lookup/external-ref?access_num=000179669700008&link_type=ISI) 253.Moncek F, Duncko R, Johansson BB, et al. Effect of environmental enrichment on stress related systems in rats. J Neuroendocrinol 2004;16:423–31. [CrossRef](http://jpn.ca/lookup/external-ref?access_num=10.1111/j.1365-2826.2004.01173.x&link_type=DOI) [PubMed](http://jpn.ca/lookup/external-ref?access_num=15117335&link_type=MED&atom=%2Fjpn%2F48%2F3%2FE190.atom) [Web of Science](http://jpn.ca/lookup/external-ref?access_num=000221542600002&link_type=ISI)