<<

Review How Stress Physically Re-shapes the Brain: Impact on Shapes, Numbers and Connections in Psychiatric Disorders

Dominic Kaul1,2, Sibylle G. Schwab1,2, Naguib Mechawar4, Natalie Matosin1,2,3*

1 Illawarra Health and Medical Research Institute, Northfields Ave, Wollongong 2522, Australia

2 Molecular Horizons, School of Chemistry and Molecular Biosciences, University of Wollongong, Northfields Ave, Wollongong 2522, Australia

4 Douglas Mental Health University Institute, 6875 LaSalle blvd, Verdun (Qc), Canada, H4H 1R3

3 Max Planck Institute of Psychiatry, Kraepelinstrasse 2-10, 80804 Munich, Germany

* Corresponding Author

Dr Natalie Matosin Faculty of Science, Medicine and Health University of Wollongong Northfields Avenue Wollongong 2522, Australia Phone: +61 2 4221 5150 Email: [email protected]

Conflict of interest: The authors declare no competing financial interests.

0 ABSTRACT

KAUL, D., S.G. Schwab, N. Mechawar and N. Matosin. How Stress Physically Re-shapes the Brain: Impact on Brain Cell Shapes, Numbers and Connections in Psychiatric Disorders…NEUROSCI BIOBEHAV REV XX(X) XXX-XXX, 2020.-Severe stress is among the most robust risk factors for the development of psychiatric disorders. Imaging studies indicate that life stress is integral to shaping the , especially regions involved in processing the stress response. Although this is likely underpinned by changes to the cytoarchitecture of cellular networks in the brain, we are yet to clearly understand how these define a role for stress in human psychopathology. In this review, we consolidate evidence of macro-structural morphometric changes and the cellular mechanisms that likely underlie them. Focusing on stress-sensitive regions of the brain, we illustrate how stress throughout life may lead to persistent remodelling of the both and in cellular networks and how these may lead to psychopathology. We support that greater translation of cellular alterations to human cohorts will support parsing the psychological sequalae of severe stress and improve our understanding of how stress shapes the human brain. This will remain a critical step for improving treatment interventions and prevention outcomes.

Keywords: Stress, adversity, cytoarchitecture, psychiatric disorder, , glia, , , amygdala, depression, , bipolar disorder

1 1. INTRODUCTION

The brain is centrally responsible for coordinating stress detection and response. In particular, the prefrontal cortex (PFC), hippocampus, and amygdala are areas critical for regulating the systemic effects of stress [1]. The sensitivity of these brain regions to stress may also precipitate changes to brain structure in an effort to adapt to stress and enhance survival by improving the ability to deal with similar future stress [2]. However, this is not always beneficial, as long-term brain adaptations in response to severe stress can have deleterious effects on crucial brain functions, including emotional regulation and cognitive capacity [3].

Stress is among the strongest risk factors for psychotic and mood disorders [4, 5]. This severe stress may be any environmental stress that is perceived to overwhelm the individual’s adaptive capacity [6]. The complexity and inherent individuality of stress and stress vulnerability, particularly in humans [7, 8] but also in rodent models [9, 10], has posed a major challenge in determining how specific psychological stressors contribute or lead to the development of distinct psychopathologies in human populations. Stress vulnerability is highly variable given the complex interaction between genetics and environment [11]. How stress physiologically contributes towards individual disorder trajectory under current nosology has been largely overlooked in much of the psychiatry literature [12]. Refining our understanding of how stress physiologically contributes to symptoms of psychiatric disorders is important to refine targets for prevention and intervention.

In this review, we summarise data from both rodent models and humans examining how stress impacts on the PFC, hippocampus, and amygdala, brain areas which have been the focus of extensive efforts to delineate a role for stress in psychopathology. First, we summarise evidence regarding how stress shapes the brain at the macrostructural level (e.g. entire brain region volume/shape). We then explore how these key regions are impacted at the cytoarchitectural

2 level, across both neurons and glia. With this information, we suggest that dysregulation at the cytoarchitectural level may underlie why distinct stress timings contribute differently towards psychiatric risk and that specific cellular contributions likely shape larger structural and functional deficits.

2. MACROSTRUCTURAL CHANGES TO A STRESSED BRAIN: IMPLICATIONS FOR PSYCHOPATHOLOGY

How stress shapes human brain morphology over the lifespan has by-and-large been conducted through macrostructural studies utilising structural and functional magnetic resonance imaging

(sMRI and fMRI). These studies have suggested significant association between stressful life experiences and altered brain shape. Alterations to the structure and function of specific regions of the brain likely contributes toward a range of emotional and cognitive phenotypes associated with psychopathology. For example, sMRI studies have associated volumetric changes in stress- sensitive regions, including the PFC, hippocampus, and amygdala, with emotional and cognitive symptoms in cases of depression [13-15]. Similarly, an fMRI study indicated that psychosocial stress is associated with reduced centrality, or communicability, of the PFC and hippocampus, and an increased centrality in amygdala connectivity [16], features also observed in psychiatric disorders [17-19]. This is perhaps not surprising given the PFC, hippocampus, and amygdala are strongly influenced by stress [20, 21] and subsequent stress regulation [7, 22]. It is however important to note that although this review focuses on these three brain regions, the impacts of stress are seen across the entire brain. For example, elevated stress is associated with increases in the volumes of some regions such as the fusiform cortex and parahippocampal gyrus, and volumetric reductions in the corpus callosum [21]. Lower volumes in the and insular cortex have also been associated with experiencing adversity throughout life [23-25].

3 2.1. Prefrontal cortex (PFC)

The PFC plays an important role in the top-down control of behaviour, thought, and mood [26].

This regulatory role extends to the stress response. Through connectivity across the cortex and larger subcortical networks, including the hippocampus and hypothalamus, the PFC plays a complex role in the perception and response to stress. In particular, the PFC is important for modulating the neuroendocrine (hypothalamic pituitary adrenal [HPA] axis) stress response [27-

29], which is intimately involved in exerting the effects of stress across nearly every cell in the body [30]. However, the responsiveness of the PFC to stress appears to make the region highly sensitive to stress-induced remodelling. Several sMRI studies have associated psychological stress with reductions in cortical volume across the medial PFC (mPFC) [25, 31], orbitofrontal cortex (OFC) [32, 33], ventromedial PFC (VMPFC) [34, 35], dorsolateral PFC (DLPFC) [36,

37], cingulate cortex [38, 39] and the underlying [40] (Table 1). An extensive review by Savitz and Drevets [41] also notes that the PFC, including the OFC, VMPFC, DLPFC, and cingulate cortex, generally exhibit decreased volume, and reduced functional activation in cases of depression and bipolar disorder. Accordingly, several subsequent neuroimaging studies have reported volumetric reductions in the PFC in several psychiatric disorders, including major depressive disorder, bipolar disorder and schizophrenia [42-44]. In addition, reduced volume has been associated with the speed of disorder progression and symptom severity [45, 46].

Consequently, it is hypothesised that persistent changes in the PFC structure induced by stress may play a role in the development of psychiatric symptoms.

A large number of fMRI studies corroborate the hypothesis that stress-induced deficits in the

PFC contribute to some extent in the dysregulation of functions commonly seen in psychiatric disorders (Table 1). For example, adults exposed to chronic psychosocial stress demonstrate reduced DLPFC function and connectivity [47]. Reduced DLPFC function in fMRI studies has been associated with deficits in behavioural and cognitive scoring in cases of schizophrenia [48].

4 Losses of mPFC volume due to chronic stress have also been associated with reduced synaptic output and reduced capacity to suppress the HPA axis and thus stress responsiveness [49]. Long- term reductions to PFC function may therefore be associated with the development of a range of symptoms expressed in severe psychiatric disorders, including hypercortisolemia [50] and anhedonia [51].

Early-life adversity poses a particularly strong and distinct risk for psychopathology [52].

Moreover, the type of stress during early life further distinguishes the risk towards psychiatric disorder development [52]. Early-life physical and emotional abuse have been closely associated with persistently reduced cortical thickness in the mPFC [53], OFC [31, 33], cingulate cortex

[38, 39], and DLPFC [36]. These effects are distinct from stress experienced in adulthood. In older adults, recent self-perceived stress was associated with reductions in the lateral regions of the PFC, with less significant effects seen in the medial regions [34]. Likewise, the recurrence of stress also contributes to distinct risk for psychopathology [54]. Repeated exposure to stress has been associated with reduced mPFC and cingulate cortex volume [25]. Smaller PFC volume also associates with increased susceptibility to perceived stress, suggesting that accumulative stress exposure is a key factor affecting PFC function, even in older adults [34]. As reduced PFC volume is also associated with impaired stress regulatory pathways [49], reducing the capacity of the PFC to regulate stress responses can sensitise the region to further harmful effects of stress.

Indeed, a recent fMRI study indicates that individuals exposed to childhood stress exhibit significant increases in activation in the DLPFC and decreases in the VMPFC when faced with psychosocial stress in adulthood, compared to those without a history of stress [55].

Consequently, the and timing of stress is intimately involved with the structural remodelling of the cortex, and suggests that further dynamics influence how stress contributes towards cortical psychopathology.

5 Although substantial efforts to resolve how stress shapes the PFC have been undertaken, several reports also suggest that the wider cortex is vulnerable to stress. Cortical thinning associated with childhood adversity is widespread across the cortex [38]. In particular, a number of studies suggest that the type of stress experienced is integral to how the cortex is reshaped. In the somatosensory cortex, thinning in the genital area is specifically associated with sexual assault, whereas emotional abuse thinned regions associated with self-awareness and reflection [38].

Thinning of the visual cortex after witnessing domestic violence during childhood [56] and the auditory association cortex after parental verbal abuse [57] have also been reported.

Consequently, the cortex is highly sensitive to specific kinds of stress, which may preferentially remodel specific circuits that process those stressors. Understanding how these specific circuit vulnerabilities precipitate across the brain is necessary to resolving the pathophysiological contributions of stress.

2.2. Hippocampus

The hippocampus is also intricately involved in regulation of the stress response. It is central to processing and contextualising including experiences of severe stress [58].

Functionally, increased activation of the hippocampus suppresses stress responses through direct projections from the ventral hippocampus (anterior hippocampus in humans) to the paraventricular nucleus of the mPFC, amygdala, and hypothalamus, where the HPA axis hormone cascade initiates [59-61]. Rodent models also indicate that the ventral hippocampus is distinctly vulnerable to stress across volumetric, cellular and molecular studies [62-64]. In humans and rodents, the hippocampus is particularly sensitive to stress mediated by the HPA axis as it contains high levels of corticosteroid receptors [65, 66]. As a result, it is closely tuned to stress responses and one of the most widely examined regions in macrostructural studies of stress.

6 As in the PFC, severe psychological stress is generally associated with reduced hippocampal volume [67-72] (Table 1). These effects are observed across a range of stress timings including both early-life, adolescence, and adulthood stress as well as both acute and chronic stress [67,

73-78], although some studies have also observed no change [79], or increases [80] in volume.

Additionally, the severity of stress (type, persistence, recurrence) during early-life has been an effective predictor of hippocampus volume, including events such as life-threatening illness, parental neglect, and physical, emotional, and sexual abuse [81]. Psychological stress also specifically reduces the total volume of the anterior hippocampus, further supporting that this region is particularly responsive to stress [82]. This is paralleled in functional neuroimaging studies. In an fMRI study, chronic stress associated with reduced connectivity across the CA2,

CA3, and dentate gyrus (DG) of the hippocampus in adults, which was in turn associated with impaired cognitive functions, such as [83]. Conversely, reduced hippocampal volume may confer vulnerability to stress and also stress regulation. Twin studies indicate that reduced hippocampal volume is associated with increased vulnerability to psychological stress, including the development of stress-associated psychopathology [84]. Stress, in all its heterogeneity, consistently impairs the hippocampus, both structurally and functionally.

Reduced hippocampal volume is also commonly observed in cases of psychiatric disorders, including mood and psychotic disorders [85-87], with disorder progression compounding loss of hippocampal volume [88-91]. Deficits in hippocampal morphology have also been strongly associated both the severity of psychotic [92] and depressive [93, 94] symptoms. However, changes to hippocampal volume are not solely associated with the development of psychiatric disorders. Severe stress can reduce hippocampal volume without associating with the development of psychiatric symptoms [95]. Likely, the interaction between the timing of stress exposure, individual susceptibility to stress, and recurrence of stress exposure all differentially impact on hippocampal development and function. Further study into the impact that specific life

7 stressors, such as early-life stress, have both acute and long-term are essential to resolve the role of the hippocampus in mediating the risk of stress in the development of psychiatric disorders.

2.3. Amygdala

The amygdala is another brain region that has become a focus of studies of stress neurobiology, as it has been widely implicated in emotional and memory, as well as fear aversion [96].

Unlike the PFC and hippocampus, the amygdala generally potentiates the stress response [97].

Morphologically, studies of amygdala have provided conflicting accounts how stress affects volume. Some accounts have noted stress in adulthood increases amygdala volume [99] while others report decreased volume [72, 100], as summarised in Table 1. In both contexts, these effects appear to be mostly recoverable after a period with stress-reduction activity [99].

Similarly, after psychological stress, the amygdala sustains increased connectivity [101, 102] and activity [103], and can also be improved with stress-intervention strategies [98]. This confers with evidence from PTSD studies which suggest that the presence of previous trauma did not associate with altered amygdala volume [104]. The amygdala may be sensitive to acute responsiveness to stress during adulthood, yet also responsive to treatment. However, it is unclear what effects may be seen in chronically stressed individuals and whether these effects are also largely recoverable in adulthood. Also unclear is how the short-term response of the amygdala drives dysregulation in other areas.

Early-life stress particularly impacts amygdala, being closely associated with increased amygdala size [105] and reactivity [106]. In pre-adolescence, psychosocial stress also associated with increased amygdala volume in later adolescence [107] adulthood [108]. However, as reviewed by Teicher and Samson [109], several studies in which individuals were exposed to adversity across adolescence display reduced amygdala volume, compared to controls [110-112].

The timing of stress therefore appears to be particularly important in determining how the

8 amygdala is shaped. Initial exposures to stress may sensitise the brain to subsequent stress, and the recurrence of stress throughout adolescence may be important to shaping the amygdala, contributing to a diversity of phenotypes in adulthood. This is particularly relevant as a meta- analysis of adults with a history of childhood adversity found no significant change in amygdala volume [113], although this may be owing to the diversity of stressors in included studies.

The morphological alterations to amygdala in psychiatric disorders has faced similar challenges.

Some studies have found no difference in amygdala volume between healthy human controls and cases of depression [114, 115], while others have identified reductions [116]. Reduced amygdala volume has also been observed in individuals with psychotic symptoms, compared to those without and healthy controls [115, 117]. Despite losses in volume, amygdala hyperactivity has still been noted in cases with psychotic symptoms [118]. Moreover, psychotic symptoms are associated with blunted amygdala-PFC connectivity [119]. The diversity of observations regarding the amygdala have made resolving a role for how the amygdala contributes towards stress risk in these conditions difficult. One hypothesis is that hyperactivity, combined with impaired connectivity, may lead to impaired stress regulation and increased stress vulnerability, which feed forward into regions of the brain where functional and morphological alterations are more persistent [120]. It is crucial to understand how specific stress types and timings contribute to distinct amygdala morphology before its role in shaping how stress contributes to psychopathology can be discerned.

3. CYTOARCHITECTURAL CHANGES INDUCED BY STRESS: DEFININING THE ROADMAP OF PATHOLOGICAL PROGRESSION Although macrostructural neuroimaging studies can shed light on what brain areas are affected by stress, and how their overall physical structure might change following stress (e.g. volume loss or cortical thinning), what these changes represent requires investigation at the cellular

9 level. Several recent efforts have suggested that altered volumes detected by neuroimaging are associated with altered density, in particular and [8, 121-123], suggesting that macrostructural studies may indeed correlate with specific cellular changes.

However, we are only just beginning to explore the changes to local circuits at the level of cell distribution, density, and morphology (referred to collectively as cytoarchitecture) which underlie these larger volumetric changes in humans. Particularly relevant in this sense is determining the cellular contributions driving region-specific vulnerability and resilience to stress and what role this plays in psychopathology. The formidable task of resolving the cellular contributions towards regional alterations is important in enabling psychiatric subtyping, and may provide new avenues of pursuit for prevention, intervention and treatment strategies.

3.1.Stress at the centre of pre-clinical rodent models of psychiatric

disorders

How the brain is shaped by stress and how these stress-induced changes may contribute to psychopathology have been largely examined in pre-clinical rodent models. These models remain an essential tool in understanding fundamental brain functions, and how these functions may be impaired by factors such as genetics and environment [124]. The capacity to model both the specific and longitudinal effects of stress in controlled scenarios holds a distinct advantage in determining how stressful events contribute to the onset of specific symptoms and the cellular and molecular consequences which underlie these states. The focus on pre-clinical rodent models has also been driven by the very limited possibility to study the human brain in the living at a detailed cellular and molecular resolution, relying almost exclusively on postmortem brain samples which are often further limited to late-stage cases, and sometimes, poor quality.

Pre-clinical rodent models of depression principally rely on stress to induce depression-like behaviour in either wild-type rodent lines, such as Sprague-Dawley rats or C57BL/6 mice, or

10 stress-vulnerable rodent lines, such as Wistar-Kyoto rats (for review of such pre-clinical models and their limitations and relevancy to human psychopathology, see Gururajan et al. [125]). These stress paradigms vary from adult chronic restraint and social defeat, to early-life maternal separation, learned helplessness, and corticosterone administration, but all operate based on the principal that overcoming the organism’s ability to cope with adversity will ultimately induce emotional and cognitive dysregulation in vulnerable individuals. Stress has also been used to induce psychosis-like symptoms in rodent lines, however, less often [126]. Importantly, while these models induce similar behavioural and cognitive phenotypes, the nature of these stress paradigms (chronicity, repetition, intensity, age of exposure, type of stress) all likely contribute to underlying variability in the cellular and molecular effects over the life course. For the intent of this review, we will consider stress based on time-point of exposure, either during early life, adolescence, and adulthood, unless otherwise stated. However, underlying variability between models should be weighed when considering these consolidated findings.

When discussing rodent models of psychiatry, it is also important to note that these models are designed to induce certain psychiatric phenotypes, such as social avoidance and cognitive impairments. These models do not comprehensively model neither the symptoms nor the developmental trajectory of psychiatric disorders in humans. They are, however, highly valuable in facilitating the examination of specific stress effects, such as resolving the cell-type specific impacts, and dissecting how these contribute to psychiatric-like phenotypes, which remains difficult to study in humans.

The final caveat in these pre-clinical models is that research has been almost exclusively conducted in males, due to the presumption that hormonal variability over time would introduce extensive variability in results, despite meta-analyses since indicating that this is likely not the case [127, 128]. A growing body of evidence indicates that there are several variations between

11 males and females in response to stress and vulnerability to subsequent psychiatric disorders.

This has been extensively reviewed by Wellman et al. [129], and presents an important consideration as some stress-linked psychiatric disorders are either equally prevalent in females compared to males (e.g. bipolar disorder) [130] or twice as prevalent in females compared to males (e.g. depression) [131]. Consequently, what contributes to these sex-specific differences will remain an important consideration in ensuring equitable and effective prevention and treatment strategies moving forward.

3.2.Excitatory Neurons

A plethora of neuronal subtypes exist across the brain, varying on the basis of their morphological, molecular, and electrophysiological properties [132]. Broadly, the overwhelming majority of neurons can be classified as excitatory (e.g. glutamatergic neurons) or inhibitory (e.g.

GABAergic neurons), however, within these classifications exists enormous diversity. The recent explosion of single-cell transcriptomics studies of the brain indicate that excitatory neurons demonstrate highly diverse transcriptomes both across and within regions [133]. As the most populous excitatory neurons in the cortex, hippocampus, and amygdala, glutamatergic pyramidal neurons play an integral role in the consolidation of inputs and broader integration via projections, often between different regions of the brain [134]. As a result, a broad body of work has begun to unravel how these cells are shaped by stress and how this likely contributes to specific emotional and cognitive symptoms of psychiatry.

3.2.1. Excitatory neurons are highly shaped by stress: evidence from pre-clinical models Several seminal works have documented that chronic stress causes a loss of dendritic complexity across the PFC, particularly in the mPFC of adult rodents (mice and rats) [135-139] (Figure 1).

Similarly, in the hippocampus of adult rodents (mice and rats) and tree shrews, chronic restraint and psychosocial stress reduced dendritic complexity in CA3, despite very few effects being

12 noted in CA1 and the DG [140-144]. This reduced dendritic complexity is also more pronounced in the ventral aspects of CA3, further supporting that the ventral hippocampus is distinctly responsive to stress [143]. The mPFC and CA3 also demonstrate reductions in dendritic complexity in response to chronic corticosteroid administration [145-147], supporting that stress- associated remodelling may be in part associated with altered HPA axis signalling [148]. One hypothesis suggests this occurs in an effort to promote stress reactivity through both the suppression of inhibitory regions, such as the mPFC and hippocampus, as well as to de-centralise processing away from higher-cognitive areas to promote rapid innate responsiveness [149].

However, these alterations may contribute to secondary, unwanted changes in certain brain functions, such as decision making, memory, and behavioural responses [150].

Across rodent models, stress-induced dendritic retractions predominantly impact apical dendrites

[140, 142, 144, 145, 147, 151]. Apical dendrites have distinct inputs (as compared to basal dendrites [152]), being closely involved in the integration of projections from other brain regions, particularly at their distal sites [152, 153]. Impairment of these synapses again suggests that stress is contributing to specific short-term inhibition of communication between regions, such as the PFC and hippocampus, as a compensatory mechanism to increase stress responsiveness [149]. This is supported by findings from Radley and Sawchenko [154] wherein lesions in the rat PFC and hippocampus had additive effects in hyper-activation of HPA axis. In further support of this stress adaptation hypothesis, the retraction of dendrites appears to be mostly recoverable after the removal of the stress despite dendritic regrowth often occurring closer to the [123, 136, 155, 156] (Figure 1).

3.2.2. Early-life stress exerts persistent changes on excitatory neurons Unlike stress during adulthood, stress during early-life may exert more persistent effects on pyramidal neurons (Figure 1). In one study, rats exposed to interrupted maternal care throughout early-life displayed impaired dendritic complexity in CA1 after maturation to adulthood [138].

13 Another study in the rodent Octodon degus examining the OFC found that losses of both basal and apical dendrites were associated with parental neglect, and that these rodents had persisting impairments to apical complexity into adulthood [139]. Further adding to these temporal dynamics, early-life stress can sensitise the brain to subsequent stressors during adulthood [157]. In fact, rats exposed to early-life maternal separation also exposed to restraint stress in adulthood display greater depressive-like behaviour in a forced swim test than either maternal separation or restraint stress individually [158]. One hypothesis is that early-life stress causes persistent impairments to prefrontal and hippocampal circuits, such as seen in volumetric and fMRI studies, which increase the perception and vulnerability to stress in later life [159].

Underlying these changes may be cytoarchitectural changes to the neuropil, including dendrites and synapses [8, 121-123]. The double-hit model suggests that stress during key neurodevelopmental stages may prime the brain for vulnerability to subsequent stress, which persists throughout life [160]. As the brain loses with aging, the primed regions may acquire alterations which are then unable to be recovered, leading to the persistently impaired circuits and thus driving neurodevelopment of psychiatric disorders. This agrees with evidence in aged rodents which are less responsive to dendritic remodelling, but when they are affected, display a diminished ability to recover [139, 161, 162].

Unlike the PFC and hippocampus, the impacts of stress on dendritic morphology in the amygdala have been noted to be variable. Both increases [163-165] and decreases [166, 167] in dendritic complexity have been identified in response to chronic stress. In response to acute corticosterone administration, the amygdala displays dendritic hypertrophy in rats and induces anxiety-like behaviour [168]. In further disparity from the PFC and hippocampus, amygdala dendritic hypertrophy after chronic immobilisation of adult rats is persistent, even after stress recovery [169, 170]. Likewise, chronic maternal separation in early-life, has been shown to induce long-term dendritic hypertrophy in the BLA of rodents, but may be recoverable after

14 environmental enrichment in adulthood [171]. Chronic hyper-activation of the amygdala likely contributes towards both anxiety and emotional alterations associated with psychopathology.

The amygdala is vulnerable to stress-associated neural remodelling in both early-life and adulthood, although the discrimination of how these experiences contribute to psychiatric-like symptoms remains to be evaluated.

3.2.3. Alterations of dendritic spines and post-synaptic sites Dendritic spines are small (1-10 µm) protrusions from neuronal dendrites. Mature spines are sites of the (PSD), containing several important postsynaptic , neurotransmitter, neurotrophic, and hormone receptors [172]. The unique structure, localisation and motility of dendritic spines play a role in the compartmentalisation of neuronal signalling

[173]. Their integral role in synaptic signalling has made these structures a well-documented feature in studies of stress and are hypothesised to significantly contribute to volumetric changes in response to stress [123, 155].

In rodent models of chronic stress, alterations to dendritic spines are among the most consistently identified cellular changes. Significant losses of dendritic spines in response to chronic stress (restraint, corticosterone treatment) in adult rodents (rats and mice) have been identified in the PFC [139, 174-176], with the most striking losses observed on apical dendrites

[137, 177]. In the hippocampus, several studies in rats have also identified reduced density in response to chronic restraint stress, particularly on apical dendrites in CA3 [156,

178, 179], and findings in CA1 have been variable, reporting both increases and decreases in spine density in response to restraint stress [155, 178, 180]. Chronic corticosterone administration has also been shown to cause loss of dendritic spines in the mPFC of adult rats and the CA3 of adult mice [146, 177]. After recovery from stress in adulthood, however, it appears that mPFC and CA3 spine losses are mostly restored [176, 179, 181]. Together, it appears that not only are dendrites retracted by stress, but their constituent post-synaptic sites

15 may be eliminated in regions of the PFC and hippocampus associated with regulating the stress response (Figure 1). This two-fold inhibition of post-synaptic structures (i.e. the post-synaptic sites and the dendrites themselves) greatly reduces the capacity of these dendrites to form efficient synapses, particularly in these distal apical circuits. In agreement, following chronic stress, glutamatergic neurotransmission is impaired across the PFC and hippocampus, despite acute stress inducing transient increases, as reviewed by Popoli et al. [182]. When compared with fMRI data from humans that indicate the centrality of the PFC and hippocampus is reduced in response to stress [16], cytoarchitectural evidence suggests that the communication in pyramidal excitatory circuits in these regions is similarly inhibited, particularly in response to chronic stress.

In the amygdala, chronic stress (immobilisation, social defeat) in adult rodents (rats and mice) has been repeatedly shown to increase dendritic spine density, particularly in the BLA [165, 183-

187]. Similarly, chronic corticosterone administration over a 20 day period in adult mice also induces increases in spine density, although these increases return to normal after a washout period [146]. Moreover, these effects are delayed following traumatic experiences, taking up to

10 days for increases in both total spines and mushroom spines to be identified in rats after a single prolonged shock [188]. Interestingly, after one day, increases in stubby spines were identified, and persisted after 10 days, although their contribution to amygdala connectivity is not well-understood [188]. Increases in dendritic spine density in the amygdala such as those induced in adult rats by chronic immobilisation stress may also increase susceptibility to subsequent stressors through increasing synaptic activity and increasing responsiveness to fear conditioning [165]. The persistent hyperactivity of the amygdala induced by stress may contribute to increases in anxiety and impaired emotional regulation commonly seen across psychiatric disorders [189, 190].

16 Early-life and adolescence appear to be particularly vulnerable developmental stages for psychological stress to cause persistent alterations to dendritic spines. For example, early-life maternal separation reduces total spine density in the whole PFC and CA1 of the hippocampus of rats, which persist to adulthood [191]. Similarly in adolescent mice, social defeat stress reduces the density of stubby spines, but increases that of thin spines in the hippocampal CA1, and is accompanied by depression-like behaviour, as indicated by social interaction and tail suspension tests [192]. Juvenile restraint stress can also reduce dendritic spines in the PFC and

BLA of rats, which persist into adulthood [193]. In the BLA, reduced spine density induced in young rats by chronic social stress contradicts increases seen in adults [194].

These cytoarchitectural changes likely have functional consequences which may be sex-specific.

For example, juvenile male rats exposed to repeated stress display reductions in postsynaptic α- amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) and N-methyl-D-aspartate receptor (NMDA) expression in the PFC, associated with cognitive impairment in temporal order recognition testing; these glutamate receptor changes were also recapitulated with corticosterone treatment in a cell culture model, highlighting that these glutamate receptor changes are mediated through glucocorticoid receptors [195]. Interestingly, female juvenile rats exposed to a repeated restraint stress paradigm did not demonstrate the same cognitive impairments or glutamate receptor reductions [196]. However, by blocking oestrogen receptors with injections of the receptor antagonist ICI182 780, stress-related deficits were again observed [196]. These studies collectively suggest that early-life and juvenile stress sex- specifically induces deficits in the structure and function of excitatory circuitry, specifically via glucocorticoid-induced alterations of post-synaptic glutamate receptors, leading to both cognitive and behavioural consequences.

17 Studies in rodent models have also suggested that restoring impaired dendritic spines may alleviate psychiatric-like symptoms, further implicating glutamatergic signalling in mediating stress in psychopathology. For example, treatment with ketamine appears to recover spine density and restore microcircuits identified by imaging in the PFC following chronic corticosterone administration [175]. Recovery of spines associated with ketamine administration in the PFC also alleviated depression-like symptoms, as indicated by a tail suspension test [175].

The antidepressant desipramine similarly recovers reduced spine populations in the mPFC induced by a single foot shock in rats [197]. Consequently, targeting dendritic spines may prove to be an effective method of alleviating some symptoms associated with psychiatric disorders, however, how stress drives these synaptic alterations remains to be resolved.

3.2.4. Translating stress-induced excitatory neuron remodelling into human populations – implications for psychopathology Although rodent models have been valuable for better understanding how different types and timings of stress may shape principal neurons, it is unclear to what extent these pre-clinical findings translate to humans. Particularly relevant in this context is that stress experienced by humans is extremely variable and likely exerts distinct effects compared to rodents. The remodelling of dendrites and dendritic spines is highly implicated in human pathology across stress-associated psychiatric disorders [198]. Alterations to the structure and function of excitatory neurons in the human brain in psychopathology have been extensively reviewed across disorders including depression [199, 200], bipolar disorder [198], and schizophrenia [201,

202]. A number of trans-diagnostic pathologies have begun to emerge which may be shaped by stress. For example, a reduction in spine density has been observed in the DLPFC’s cortical layer

3 in schizophrenia [203], bipolar disorder [204], and depression [205]. The DLPFC is important both to cognitive functions such as attention and memory as well as in the regulation of negative emotion [206], symptoms associated with these disorders under DSM-5 diagnostics [207].

18 Reduced NMDA receptor binding in the DLPFC is associated with individuals with a history early-life adversity [208], suggesting that these circuits are indeed impaired by a history of stress, and may be associated with specific stress timings. However, the contributions of stress towards the remodelling of excitatory circuits, have been sparsely investigated in human cohorts at the cellular level.

A handful of studies of human cohorts with stress histories have evaluated how glutamatergic circuits are shaped by stress. One of the first human postmortem PTSD cohorts identified specific reductions in mature spines and an increase in stubby spines in the OFC, compared to healthy controls [209], which may indicate and impaired capacity to form effective synapses. As

PTSD shares a high co-morbidity with psychiatric disorders, such as depression [210], these disorders may share related aetiology, including traumatic experiences [211]. Another study noted that dendritic retraction and reduced spine density in CA3 of a non-psychiatric adult cohort were associated with anxiety and depressive symptoms [212]. We have recently identified that dendritic spines in individuals with psychiatric disorders and early life stress demonstrate persistent and pronounced losses of mature dendritic spines in the superficial layers of the OFC.

[213]. Although more moderate, these same effects were seen in individual previously exposed to severe chronic stress in adulthood. Importantly, no significant losses were seen in individuals with psychopathology and no distinct chronic severe stress history [213]. These effects suggest that stress exposures can persistently shape excitatory circuits within psychiatric cases and highlights the importance of assessing stress history in future cohort design. Further studies in larger sample sizes are needed to replicate these findings and to provide more details on how stress exposures can reshape brain cytoarchitecture in other, equally important brain areas.

19 3.3.Inhibitory Neurons

Mounting evidence supports that inhibitory GABAergic neurons are susceptible to stress and mediate contributions of stress towards psychopathology [214]. While GABAergic neurons only account for approximately 10-20% of the neuronal population across the brain [215, 216], the balance between inhibitory and excitatory signalling remains at the crux of regular brain function

[217]. Inhibitory neurons are intimately involved in modulating local circuits and are integral in tuning regional functions such as emotional regulation in the cortex [218].

A significant amount of work has been undertaken to delineate the diversity of within the CNS, which are now recognised to be enormously diverse [219]. Inhibitory neurons are classified largely based on their expression of calcium binding proteins (parvalbumin [PV], calbindin, and calretinin) as well as neuropeptides (neuropeptide Y, vasoactive intestinal

(VIP), somatostatin (SST), cholecystokinin) [220]. Three of the most abundant inhibitory neuron classifications are those that express PV, SST, and VIP [221]. These inhibitory neurons form distinct microcircuits to attenuate signals within and between regions. Within these circuits, PV- expressing interneurons predominantly mediate somatic inhibition of pyramidal neurons and the inhibition of other PV-expressing interneurons [222]. They are important to both feedforward and feedback inhibition and in the regulation of signal propagation [223].

Alternatively, SST-expressing interneurons primarily control the inhibition of inputs to pyramidal neuron apical dendrites [220], although also inhibit other inhibitory neuron types, such as PV-expressing inhibitory neurons [224]. Finally, VIP-expressing interneurons typically inhibit SST-interneurons, which feeds forward to reduce inhibition of other inhibitory neurons

[224]. Although further diversity of inhibitory neurons based on morphology, function and transcriptome has been widely demonstrated at single-cell molecular resolution [133, 221, 225], an extensive analysis of the matching cell morphology has not yet been performed in the context of stress and psychopathology. Using markers derived from the single-cell sequencing studies

20 could provide an important basis for morphologically categorising functional GABAergic neurons, and understanding how inhibitory microcircuits are influenced by stress.

3.3.1. Stress shifts the excitatory/inhibitory balance – evidence from pre-clinical studies Several lines of evidence indicate that inhibitory neurons are sensitive to stress and that these effects modulate broader neural circuitry. In the mouse PFC, GABAergic signalling and metabolic activity are highly dysregulated by social stress and associated with depression-like behaviour, as indicated by decreased sucrose preference and social interaction [226]. Similarly, in the rat mPFC, chronic mild stress greatly impairs GABAergic signalling and is also associated with the development of depression-like anhedonia [227]. Destabilisation of the excitatory/inhibitory (E/I) balance is widely hypothesised to be one pathway through which psychiatric disorders develop [214, 228, 229], and has been hypothesised to underlie disorder trajectories throughout adolescence [230]. Resolving how this balance is impacted by stress may be essential to understanding how distinct stress timings contribute to the aetiology of psychiatric disorders.

A recent review has extensively detailed evidence for the dysregulation of cortical GABAergic signalling by stress in rodent models of depression [231]. The authors note that stress broadly impairs cortical inhibitory neuron GABA production, turnover, and responsiveness as well as reduces markers across several classes of inhibitory neurons including both PV and SST. These deficits also associate with the development of depression-like behaviour and impaired cognition

[231]. This is seen across the PFC, including the mPFC, DLPFC, cingulate cortex, and OFC (see

Fogaça and Duman [231] for this extensive review) (Figure 2). Cortical inhibitory neurons are important in coordinating functions involved in emotional and behavioural processing [217] and losses of this control may be important in the development of associated psychiatric symptoms.

In mice, knockout of cortical PV transcription factors Dlx5 and Dlx6 reduced

21 cognitive flexibility, and optogenetic activation of these same interneurons recovered cognitive flexibility to levels comparable to controls [232]. Similarly, chemogenetic suppression of PV interneurons in the mouse mPFC induces helplessness [233]. Consequently, deficits in cortical inhibitory circuits induced by stress are likely involved in both behavioural and cognitive impairments as are seen in psychiatric disorders.

However, several further dynamics that influence inhibitory circuits are shaped by stress. First, the duration and intensity of stress appears to influence the cytoarchitecture of inhibitory neurons. Both male and female mice exposed to chronic mild stress for 2 or 4 weeks have increased numbers of PV-expressing interneurons across the PFC [234, 235]. Alternatively, male adult rats exposed to mild stress for 9 weeks [227] or more severe social isolation for 3 weeks

[236] exhibited losses of PV-expressing interneurons in the mPFC and displayed anhedonia-like symptoms. It is likely that there is a phasic response to stress, and that short-term increases may be followed by more gradual, yet persistent reductions in inhibitory neurons. Second, sex not only the impacts of stress on interneuron cytoarchitecture, but may contribute to distinct psychiatric-like symptoms. For example, chronic but not acute chemogenetic activation of PV- expressing interneurons in the mPFC was sufficient to induce anxiety-like behaviour in adult female rats, but not in males [235]. Alternatively, male rats exposed to chronic variable stress at postnatal day 60 demonstrated increased inhibitory connectivity between these neurons and excitatory neurons, dampening excitatory potential within PFC networks [49]. These male rats displayed reduced cognitive performance in a delayed spatial win-shift task [49].

Inhibitory circuits in subcortical regions also appear to be vulnerable to stress. In the hippocampal CA3 and DG, chronic social stress in adult tree shrews reduces the number of PV- expressing interneurons [237]. This is similar to chronic restraint of adult rats where reduced PV immunoreactive neurons across the whole hippocampus has been noted [238-240]. In rodents

22 susceptible to stress (displaying depressive-like symptoms), several additional interneuron types, including SST interneurons are also significantly reduced in number across CA1 and CA3 [239].

However, both acute corticosterone administration and chronic restraint stress of rats also increased GABAergic signalling, measured by postsynaptic currents, in the region [240]. In the amygdala chronic stress in a number of adult rodent models (unpredictable, water deprivation, corticosterone administration) broadly increase the inhibitory capacity of GABAergic neurons

(see review by Jie et al [241] for comprehensive detail), however, cytoarchitectural changes to cell populations and morphology are not well characterised in the region.

Evidently, one of the most notable challenges in defining a role for inhibitory neurons in response to stress are the seemingly contrary responses seen in the structure and function of inhibitory circuits. One hypothesis is that although both glutamatergic and GABAergic signalling are suppressed by stress, a net hypoactivity is established, contributing towards functional deficits across the PFC and hippocampus [214]. This may be driven both by specific alterations to the connectivity of inhibitory neuron microcircuits, such as reduced inhibition of

PV-expressing interneurons due to reductions in somatostatin interneuron populations and functions [224], as well as an over-representation of inhibitory signals exerted on excitatory neurons [214]. However, it will be important to resolve structural and functional abnormalities to determine how this informs the E/I balance across regions of the brain.

3.3.2. Excitatory/inhibitory imbalance: the path to resolving the developmental trajectory of psychopathology? One emerging hypothesis regarding how the developmental trajectory of psychiatric disorders is established, reasons that the balance of excitatory/inhibitory balance throughout development is an important factor in the eventual dysregulation of neural circuits [214]. Converging evidence from pre-clinical models suggests that inhibitory neurons are vulnerable to early-life stress, particularly in stress-sensitive regions. In rats, early-life isolation increases the number of PV-

23 expressing interneurons in the adult PFC, despite no changes in the amygdala [242]. Although no difference in total cell numbers were identified, the authors noted that the ratio of excitatory and inhibitory receptors (VLUGT1/VGAT) was decreased in the BLA [242], indicating a tendency towards over-representation of inhibitory signalling and subsequent hypoactivation of this brain area. Impaired early-life maternal care has also been shown to increase the number of PV- expressing interneurons in the hippocampus of male mice [243]. Recent evidence suggests that these changes are timing-dependent. Mice with early life fragmented maternal care have an increased rate of development of PV-immunoreactive neurons in the BLA and hippocampus, but not the mPFC; however by adolescence, no difference is seen in the BLA and hippocampus, yet increased numbers are seen in the mPFC [243, 244]. This is coupled with increased inhibitory postsynaptic currents in the mPFC during adulthood [245]. However, conflicting reports suggest that stress during early-life and adolescence impairs inhibitory signalling, particularly in the mPFC. Early-life maternal separation has been associated with reduced PV mRNA in the mPFC of male rats in adolescence [246], which were not recoverable with social reintegration [247].

Interestingly, maternal separation also reduces PV mRNA in the PFC of female rats, however, at an earlier juvenile time point [248]. In agreement, functional activation of PV inhibitory neurons following unpredictable chronic mild stress in adolescence can prevent the losses of pyramidal dendritic spines and improve behaviour [249], suggesting that improving inhibitory tone throughout development can alleviate stress induced structural deficits. It is likely that this highly vulnerable and plastic window presents an important impasse in stress-associated remodelling. Evidence from pre-clinical rodent models strongly suggests that stress induces deficits in inhibitory neurons and signalling, although these may exhibit phasic changes dependent on the timing of exposure (Figure 3). Understanding how inhibitory circuits are shaped, and how they shape connectivity across the brain, are important considerations in psychiatry.

24 3.3.3. Inhibitory signalling and a role in human psychopathology As mentioned above, the E/I balance has been purported to be involved in the trajectory of psychiatric disorders (as reviewed in [214]), and is quickly gaining traction for its likely contribution towards the development of cognitive and emotional symptoms in psychiatric disorders [214, 228, 229]. Evidence from postmortem studies largely suggests that several classes of interneurons are inhibited in psychiatric disorders and contribute to a loss of cognitive control [250], although matching these effects to those seen in response to stress has been difficult. In major depressive disorder, the density of calbindin-expressing neurons is reduced

DLPFC and OFC compared to healthy controls, despite no changes in the distribution of PV- expressing interneurons [251]. Decreased somatostatin and somatostatin-associated mRNA and proteins have also been reported in the PFC of cases of depression, despite no changes in PV- related expression [252]. Alternatively, in cases of schizophrenia, PV-expressing interneurons are decreased in the DLPFC [253], coupled with reduced expression of GABA synthesizing enzyme, GAD-67 [250, 254]. A reduction in GAD-67 has also been identified in major depression [255]. Likewise the number of PV and somatostatin-positive neurons are reduced across all subfields of the hippocampus in cases of schizophrenia [256, 257]. Although a wide range of evidence suggests that inhibitory neuron populations are decreased and also that their inhibitory functions are impaired in psychiatry, how these are influenced by stress remains to be clearly understood.

One of the most prominent gaps in our knowledge regarding how stress shapes the human brain is how the E/I balance shifts throughout life, both in health and disease. It remains important to resolve what are the consequences of deficits in both inhibitory and excitatory signalling on the function and communication of the brain. Rodent models suggest that stress timing is important to shaping this balance across several brain areas. Resolving how this balance is not only formed throughout life but also how environmental challenges such as stress shift the dynamic E/I

25 balance may provide important critical timings for intervention, treatment and prevention strategies.

3.4.

As the most abundant glia in the human brain, astrocytes are important regulators of the synaptic microenvironment. In the healthy adult brain, they account for 10-20% of total cells and 20-40% of glia in the brain, with this proportion changing between regions [258]. Once thought to be solely supporting cells, they are now known to be integral in many functions including ion/neurotransmitter homeostasis, metabolic support, and synaptic development/stabilisation and transmission [259]. As a consequence of this diversity, the heterogeneity of astrocytes has been widely identified across morphological [260], functional [261, 262], electrophysiological [263], and transcriptomic [264, 265] studies. Astrocytic heterogeneity is also evident across the whole brain [266], within individual regions, and even within local circuits [267]. A consideration that underlies studies of astrocytes is the difficulty in effective methods to both delineate astrocytes from other cellular populations, as well as delineate subpopulations involved in specific functions. For example, the most widely used marker, glial fibrillary acidic protein

(GFAP), is not constitutively expressed in all astrocytes and shows both regional and subregional localisation [268-270]. In the hippocampus, approximately 80% of all astrocytes are GFAP positive [271], whereas only 10-20% are marked in the PFC [272], in which this expression is most pronounced in the superficial layers and white matter [273]. A wide diversity of “pan”- astrocyte markers have also emerged, including, ALDH1L1, GLT-1, SOX9, however, these markers still demonstrate some degree of either regional heterogeneity or subcellular localisation

[274, 275]. One of the primary challenges facing studies of astrocytes is defining what constitutes an astrocyte and how can these populations be effectively, and most importantly, comparatively evaluated across the brain and in disease states.

26 Another major obstacle for discerning a role for astrocytes in stress-mediated psychopathology is that astrocytic complexity is linked with species complexity. Astrocytes in the two most commonly evaluated systems, rodent and human, differ in several metrics with human astrocytes being more morphologically diverse and complex [260, 272, 276], and astrocyte transcriptomic responses to environmental stimuli differing in rodents compared to humans [277]. Although these considerations must be recognised, studies in rodent models have formed the basis of our knowledge regarding how stress shapes astrocytes and still provide an important framework for determining the contributions of stress to shaping astrocytes. Given how vital astrocytes are in maintaining and modulating numerous functions across the brain and their capacity to interact and influence a wide range of cells in the brain, including interacting with the blood-brain barrier, neurons, and in the brain, astrocytes are uniquely positioned at the frontline of systemic stress, while also baring the capacity to transduce responses across cells of the brain.

3.4.1. Astrocytes at the interface of stress: evidence from pre-clinical rodent studies The transcriptional profile of rodent astrocytes is highly responsive to glucocorticoids both in vitro and in vivo [278]. In fact, astrocytes are particularly vulnerable to stress being more transcriptionally responsive to glucocorticoids than neurons [279]. Cytoarchitectural studies also note that astrocytes are vulnerable to glucocorticoids as chronic exposure reduces the number of

GFAP-positive cells both in vitro in primary rodent culture [280] and in vivo in the rat PFC

[281]. In primary hippocampal astrocyte culture, this GFAP reduction is coupled with reduced

S100β, a protein widely expressed by brain astrocytes, however, in vivo, only GFAP is reduced

[282].

In chronic stress models, such as restraint and variable stress, reduced GFAP immunoreactivity in the PFC of adult rodents (rats and mice) is well-documented, despite no changes in total astrocyte number [283, 284]. This is paired with reduced complexity of GFAP-positive processes

27 [283], however, may due to activity-dependent decreases in the protein rather than process retraction [285]. Alternatively, acute shock stress in adult mice causes astrocytic hypertrophy in the marked with ALDH1L1 as an astrocyte marker [286]. This acute stress was sufficient to induce reduced functional coupling between neurons and astrocytes as indicated by reduced astrocyte proteins [286]. The astrocytic gap junction proteins, connexin

(Cx) 30 and 43, are rapidly becoming a focus of attempts to resolve a role for astrocytes in shaping stress responses. These gap junctions are important in cell-cell communication both within the astrocytic syncytium, as well as between astrocytes and other glia and neurons [287].

As with acute stress, chronic unpredictable stress in adult rats reduces Cx30 and Cx43 punctae across the PFC, including the OFC, and ACC [288, 289]. This has also been observed in the mPFC of adult mice exposed to chronic social defeat stress [290] and is associated with reduced synaptic activity [288, 290] and myelination [288]. Reduced neuronal coupling with astrocytes is also sufficient to impair the synaptic plasticity of neurons [286] and suggests that stress-induced deficits in astrocyte function may mediate impaired synaptic plasticity across the PFC seen in response to stress. Indeed, these gap junctions are also important to the development of psychiatric-like symptoms in rodents. Blocking astrocytic gap functions is sufficient to induce depressive-like behaviour in rats, as indicated by decreased sucrose preference [289]. Given the role that astrocytes play in both mediating synaptic transmission and myelination, the vulnerability of these cells to stress is likely important to transducing stress across the brain and in precipitating these effects into psychopathology (Figure 2).

Similar deficits are observed in several subcortical regions, such as the hippocampus and amygdala. In the hippocampus, the density of GFAP immunoreactive astrocytes is typically reduced in adult rats exposed to either acute (foot shock) [291] or chronic stress (unpredictable and repeated shock) [292, 293], although some studies failed to identify changes, such as in response to chronic immobilisation [294]. Likewise, the number of GFAP-positive cells, and

28 their occupied volume, decreases in the amygdala of adult rodents exposed to chronic immobilisation stress [294]. This is paired with decreased volume occupied by astrocytes and reduced neuropil volume [294]. Several of these effects translate into primates. In adult tree shrews, both the number and soma size of GFAP-positive astrocytes were reduced by psychosocial stress in the whole hippocampus [295]. Although in neurogenic regions, such as the

DG, chronic unpredictable stress coupled with social isolation increases number of GFAP and

S100β-positive astrocytes, as well as branching complexity in male mice [296]. Stress, such as chronic social defeat, also reduces Cx30 and Cx43 expression in the hippocampus [290]. Indeed, this is mediated by the HPA axis as chronic corticosterone administration increases the proportion of phosphorylated Cx43 in the hippocampus of adult mice, which associated with anxiety and depression-like behaviour [297]. Consequently, astrocyte stress sensitivity is evident across many regions of the brain, particularly the brain areas closely involved in stress regulation.

Importantly, astrocytes are responsive to antidepressants and may be leveraged for the treatment of psychopathology. Across rodent and primate models of depression, stress-induced changes to astrocytes in the PFC and hippocampus during adulthood, such as reduced gap junctions and reduced GFAP-positive cell density, are recoverable with the use of antidepressants, including fluoxetine, duloxetine and clomipramine [295, 297-300]. Reversal of these astrocytic impairments was shown to rescue depression-like phenotypes [289, 295, 298, 299]. Chronic antipsychotics can also increase the density of astrocytes, such as in the rat PFC [301].

Normalising astrocyte populations and function thus provide a putative target for reversing stress-induced psychopathological phenotypes, at least in a subset of psychiatric patients.

Delineating which patients have astrocyte-related pathologies and therefore would benefit from these interventions remains an avenue of exploration.

29 3.4.2. Astrocytes and a role in early-life vulnerability to stress As with neurons, astrocytes are also highly vulnerable to stress in a timing-specific manner.

Octodon degus exposed to maternal separation for the first three postnatal weeks demonstrate an increased total number of S100β-positive astrocytes as well as decreased total GFAP-positive cells across the mPFC, anterior cingulate cortex, and prelimbic cortex [302]. Alternatively, rats exposed to maternal separation in adolescence (14 days postnatal) exhibited increased numbers of GFAP positive astrocytes in the PFC [303], although whether this effect is sustained into adulthood has not been considered. These timing-dependent increases following early-life stress have also been observed in the hippocampus of mice [304]. Following 7 days of postnatal maternal separation, wild-type mice exhibit increased GFAP coverage in the hippocampus, however, by adulthood (10 months), GFAP coverage is reduced to lower than that of controls

[304], in line with adult rodent stress models. Regular astrocyte function is essential to the maintenance of numerous developmental processes including in the development and diversification of synapses [305], regulating neuroinflammation [306], and establishment of myelination [307]. Homeostasis of these functions is essential to the development of a healthy brain and so astrocytes are distinctly positioned to precipitate the effects of environmental cues, such as stress, across the brain. It will be important to determine how these transient shifts in astrocytes contribute to cellular remodelling throughout development. While a consensus on the specific cytoarchitectural changes to astrocytes have not yet been reached, there is robust indication that stress-induced changes to astrocytes are involved in psychiatric disorder pathology contributing to synaptic and network abnormalities, common to psychiatric disorders.

3.4.3. Bridging the gap between astrocyte models and human psychopathology There is little literature translating rodent studies of astrocytes to the human brain. To our knowledge, there have been no studies focusing on assessing the impact of stress on astrocyte numbers or their morphology in the postmortem human brain, although several studies have

30 examined postmortem brain tissues derived from individuals who lived with stress-associated psychiatric disorders. These studies indicate that astrocytes are involved in the pathology of at least a subset of psychiatric patients. For example, alterations to GFAP expression in cases of major depressive disorder are not seen globally across the brain [268]. Reductions in this marker are limited to networks associated with mood disorders, including the caudate nucleus and mediodorsal thalamus [268].

Across the PFC, Cx30 and Cx43, are reduced in cases of depression [308]. In the ACC of depressed suicides, reduced Cx30 and Cx43 are coupled with reduced communication between astrocytes and oligodendrocytes compared to healthy controls, as indicated by reduced gap junction coupling [309]. Gap junctions between astrocytes and oligodendrocytes are essential to the transportation of small ions and metabolites and losses of these junctions have been associated with demyelination [310, 311]. Further studies in mood disorders have indicated that

GFAP reductions are exacerbated in cortical white matter [312], however, morphological studies note astrocytic hypertrophy in the white matter of depressed suicides, as measured by Golgi staining [313]. These regional effects may indicate that astrocyte losses are closely involved in mediating demyelination associated with mood disorders (see section below).

There are also several studies indicating that astrocyte pathology in the PFC of psychiatric disorders exhibits age-dependent effects. Reduced GFAP-positive cells have been reported in the

DLPFC and mPFC of adult (<45 years old) depressed individuals [314, 315]. Likewise, reduced glial density has also been observed in the ACC of depressed individuals [316]. This effect was not observed in cases later in life (>60 years old), as measured by the number GFAP-positive cells [317]. GFAP-positive astrocytes are also reduced in the deeper layers of in the

DLPFC of psychotic cases [318] and may be also in part attributable to age [319]. However, this phenomenon is not seen across all major psychiatric disorders. In cases of bipolar disorder older

31 than 60, reduced astrocytic GFAP immunoreactivity and process complexity has been identified

[317]. It is probable that cortical astrocyte dysregulation is involved in a subset of psychiatric pathology and is age-dependent, yet the role that stress exposure plays in mediating/exacerbating these effects is not clear.

Astrocyte alterations have also been reported in subcortical areas of the postmortem brain in psychiatric cases, although are not as well documented as prefrontal effects. Losses of GFAP positive astrocyte density have been reported in the hippocampus of cases of depression and bipolar disorder [320] and more specifically in CA1 in individuals with no history of antidepressants [321]. Furthermore, the number of GFAP-positive cells is decreased in the CA1 of depressed individuals compared to controls, despite no effects being identified in the CA3 or

DG [322]. A decrease in GFAP-positive astrocytes has also been identified in the amygdala of depressed individuals, although was not observed in cases of bipolar disorder or schizophrenia

[323]. In both in cortical and subcortical regions of the brain central to the processing of stress, a common phenotype in psychiatric disorders is a loss of astrocyte function. Although we have evidence that astrocytes are indeed implicated in psychiatric disorders across the brain, as of yet, we do not know what the contribution of astrocytes in mediating stress within these disorders.

3.5.Microglia

Microglia are the primary immune constituents of the central . In addition to their immune functions, they mediate synaptic plasticity, most notably through regulating synaptic pruning [324, 325], and participate in the pruning of excess synapses throughout neurodevelopment [326]. Microglia are also highly motile cells [327] which facilitates their high responsiveness to changes in the cellular environment as well as their ability to influence the functions of neurons and other glia [328]. As with the other glial cell types, microglia contain a

32 high abundance of glucocorticoid and mineralocorticoid receptors [329], priming them to detect and respond to stress.

3.5.1. Neuroinflammation at the centre of dysregulation in response to stress – evidence from pre-clinical models Rodent models have shown that stress is central to the potentiation of the microglial immune response [330, 331]. Several rodent psychological stress protocols (restraint, social defeat, foot shock) dramatically increase mRNA and protein levels for the activated microglia marker ionised calcium binding adaptor molecule 1 (Iba-1) [332]. This has been observed across the amygdala [333], hippocampus [334], and PFC [335, 336]. Reports are conflicting about whether these changes are associated with increased numbers of reactive cells, or with increases in already reactive populations [337-339], although microglia-mediated neuroinflammation is consistently increased nevertheless. Morphologically, the activation of microglia in response to stress typically increases microglia cell soma size, process thickness, and reactive density of Iba-

1, transitioning to an ameboid-like shape [340, 341]. These morphological changes have been noted in both the PFC and hippocampus in response to social defeat stress in mice and associated with the development of depression-like symptoms [334, 342] (Figure 2). As part of the neuroinflammation response, reactive microglia synthesise and release a number of pro- inflammatory molecules, including cytokines and chemokines [343]. These pro-inflammatory molecules, including Interleukin 1 and Tumour Necrosis Factor α, hold the potential to reshape both neurons and glia by impairing synaptic plasticity [344], myelination [345], and astrocyte functions [346]. Increased reactivity of microglia may assist in driving cellular dysregulation in response to stress.

Microglia are also important facilitators of increasing vulnerability to subsequent stress.

Following recovery from stress, microglia remain primed to subsequent stimuli including psychological stress [347] and facilitate increased reactiveness to repeated stress [334]. In adult

33 mice, primed microglia have been observed in response to social defeat stress 24 hours after exposure [348]. Chronic unpredictable stress in rats/mice adult/teen/juvenile also increases contact between microglia and neuronal processes in both the mPFC [163] and hippocampal

CA1 [349]. Morphologically, these changes prime increased microglial activation to change the cellular environment, facilitating upregulated cytokine release and synaptic remodelling at both the presynaptic and postsynaptic sites [350].

However, not all microglia appear to remain sensitised following stress (Figure 2). Restraint stress paradigms indicate that, although exposure to chronic stress increases Iba-1 reactivity in adult rats/mice, this is coupled with populations of hyper-ramified microglia [336, 337]. Stress- induced hyper-ramification of microglia has been associated with synaptic deficits in the mPFC

[339] and may contribute to depressive-like behaviour [351]. Classically, microglial hyper- ramification has been more closely associated with quiescent states [352], although ramification does not directly correlate to reactivity [353]. It has been suggested that, in response to adversity, populations of microglia are “sensitised” while other become “de-sensitised”, exhibiting hyper- ramification [354], although the consequences of these diverging populations remains unknown.

Exploring the subpopulations of microglia may be important to understanding why certain regions of the brain are more closely affected by stress, and the particular vulnerability that recurrent stress associates with.

3.5.2. Neuroinflammation and early-life vulnerability to stress Psychological stress and the onset of increased activation of microglia is particularly notable during key neurodevelopmental stages, such as in adolescence. Microglial functions, including the maintenance of synaptic pruning, astrocyte maturation, and myelination [355] are essential to typical neural development [324, 356]. Dysregulation of these functions during development can result in atypical and potentially deleterious cellular networks [349] which persist into adulthood

[357]. Moreover, alterations in microglial functions caused by stress have been associated with

34 the development of depression-like symptoms [163, 351, 358], and can be diminished via reversal of microglia-mediated neuroinflammation [359]. It has been hypothesised that alterations in microglial functions throughout early-life and adolescence shift the trajectories of neuronal and glial maturation which lead to persistent dysregulated circuits which underlie psychiatric symptoms [360]. Microglial priming may also underlie why the brain becomes sensitised to subsequent stress after early-life adversity and why stress at this critical time point leads to such a high risk for psychiatric disorder development. Primed microglia hold a greater potential to alter neurons and glia [361] and thus are likely involved in enacting the first line of response to stress in the brain. While we are now beginning to understand how microglia can influence neural circuits in a development-specific manner, we are yet to resolve the contributions of these cells towards the dysregulation of neural circuits which may underlie psychopathology. The next major hurdle will be to resolve exactly how microglia mediate stress and how this influences the developmental trajectories of neurons and other glia, as well as the development of psychiatric symptoms.

3.5.3. Neuroinflammation at the interface of stress and human psychopathology? Although rodent models suggest that microglia may mediate the effects of stress across the brain, evidence supporting a role for microglia in human psychopathology is not widely agreed upon.

Developments in positron emission tomography imaging of the activated microglial marker translocator protein (TSPO), a translocator protein, have facilitated in vivo analysis of microglial quantity and activity. In individuals with a current episode of depression, increased TSPO binding activity indicative of neuroinflammation, are seen in the PFC and ACC [362]. TSPO appears to be particularly high in depressed individuals with suicidal thoughts [363]. Increased activity also persists in recovery periods following depression later in life [364]. These imaging studies, however, do not provide the resolution needed to understand detailed cytoarchitectural

35 changes that might occur beyond general microglial activity, but support that microglia indeed mediate neuropathology of mood disorders, meriting further human research.

In postmortem studies, it has been difficult to disentangle the contributions of suicide facilitation from neuroinflammatory risk in depression. Increased Iba-1 as well as increased microglial priming have been reported in the white matter of the ACC of depressed individuals having died by suicide [365]. Although increased microglial activation is also seen in the grey matter of the

ACC, this may be associated with the risk of suicide within the condition, rather than the condition itself [366]. Indeed, increased microglial density appears to be an important consideration for suicide facilitation in depression [366, 367]. Similarly, postmortem studies of bipolar disorder have had limited success identifying increased microglial populations or activation across the DLPFC, ACC, and hippocampus, owing to small sample sizes, influences of medications, and variability in sample demographics (see systemic review by Giridharan et al.

[368] for limitations of current postmortem bipolar studies).

Microglia-mediated inflammation is also a hypothesised mechanism contributing to abnormal neural connectivity in schizophrenia [369, 370], however, resolving a defined microglial pathology in schizophrenia has been as contentious. Several postmortem studies have identified increased expression of microglial markers as well as number and density across the PFC and in the hippocampus and amygdala, yet almost as many studies have noted the opposite or null trends (as reviewed by Laskaris et al.[371] and Trépanier et al. [372]). However, markers of other functions, such as microglial-associated phagocytosis in synaptic pruning are elevated in cases of schizophrenia [373]. Indeed, a schizophrenia patient-derived cell model demonstrated increased microglial-mediated synaptic pruning [374]. Consequently, microglial pathology in psychotic disorders may be driven by activity-dependent increases in functions such as synaptic pruning, rather than limited to states of increased activated microglia associated with

36 neuroinflammation. Determining how the functions of microglia are influenced within these disorders may be important in resolving why stress is a highly common sequelae for psychiatric disorders.

3.6.Oligodendrocytes

Oligodendrocytes are essential myelinating cells of the that consist of a small cell body and a few branching processes able to myelinate dozens of . They are also important in maintaining axonal integrity [375], metabolic and trophic support of neurons [376] and in the support of the blood-brain barrier [377]. Myelinating oligodendrocytes are largely restricted to white matter [378], while those in the grey matter, considered satellite oligodendrocytes, are less intimately involved in myelination [379]. These functions are important in coordinating neural connectivity, and thus are an important, but often overlooked contributor towards psychiatric disorders [380]. The distribution of oligodendrocytes varies notably between brain regions e.g. low in the cerebellum and higher in the cortex [378] and decreases with age [381].

3.6.1. Stress impairs myelination: evidence from pre-clinical studies As with other glia, oligodendrocytes are highly susceptible to stress. In adult mice, chronic social stress downregulates a number of oligodendrocyte proteins involved in myelination in the amygdala and PFC [382]. This stress paradigm also decreases thickness and internodal length alongside the total number of mature oligodendrocytes in the mouse mPFC [383] (Figure

2). Adult mice more susceptible to the effects of stress, demonstrated by increased social avoidance behaviour, had even greater reductions in myelin thickness, internodal length and number of oligodendrocytes [383]. A reduction in myelination in the mouse PFC also accompanies the development of depression-like symptoms [384]. Conversely, both restraint stress and corticosteroid administration increase oligodendrogenesis in the DG of the hippocampus [385], potentially as a compensatory effect for the loss of oligodendrocyte 37 function. Loss of oligodendrocyte function, especially loss of myelination is intimately involved with neuronal damage [386] and impairment of synaptic signal conductance [387], likely contributing towards deficits in regional functions such as in cognition and behaviour [388].

3.6.2. Persistent shaping of oligodendrocytes by early-life stress As seen for other glia, the timing of stress exposure is important to the long-term effects on oligodendrocyte cytoarchitecture. Exposing adult mice to social isolation for eight weeks reduces myelination in the PFC [389]. Upon social reintegration for four weeks following this adversity, myelination is recoverable to levels similar to controls, suggesting a robust level of glial plasticity [389]. However, when exposed during juvenile stages, myelination is not recoverable even after maturation to adulthood is completed [390]. This may contribute towards degraded neuronal signalling in the PFC and may help explain why the effects of stress in early life are not only persistent, but notably damaging for the brain. Both astrocytes and microglia are important facilitators of myelination, and affect the migration, proliferation and mature functions of these cells [391]. For example, microglial activation stimulates the production of a number of pro- inflammatory molecules (e.g. reactive oxygen species, cytokines, chemokines) which oligodendrocytes are particularly sensitive to [392]. In fact, activation of microglia by lipopolysaccharide injection into rats is sufficient enough to delay the development of oligodendrocytes and induce hypomyelination [393]. As discussed in previous sections, both astrocytes and microglia are closely involved in timing-dependent vulnerability to stress and may contribute to myelin and oligodendrocyte impairment throughout development. This may facilitate the persistent dysregulation of neuronal connectivity and thus may be important in resolving how different stress histories contribute towards the dysregulation of brain functions.

3.6.3. Oligodendrocytes are sensitive to stress in the human brain: cause or effect? It is not yet clear whether oligodendrocytes are impacted as a consequence of stress, or as effectors of stress and how this contributes towards psychopathology. At the cytoarchitectural

38 level, markers of apoptosis and necrosis of oligodendrocytes are seen in major depressive disorder, bipolar disorder, and schizophrenia [394, 395]. These findings indicate that oligodendrocytes and their capacity to myelinate other cells are both reduced in these disorders.

This would suggest that oligodendrocytes are impacted downstream in the progress of cellular dysregulation. However, recent single-nucleus RNA sequencing study of the DLPFC implicated oligodendrocyte precursors, alongside excitatory neurons, as being the most transcriptionally dysregulated cellular population in depression [396]. In psychiatric disorders, alterations in gene expression associated with myelination are most pronounced in the hippocampus and PFC [397,

398], and favour that oligodendrocytes are particularly vulnerable in stress responsive regions. In further support that oligodendrocytes are actively involved in stress responsiveness, in the human ACC early-life adversity has a robust and lasting effect on the oligodendrocyte transcriptome, being associated with decreased myelination of fine axons [399]. This adversity also impairs the maturation of oligodendrocytes in the VMPFC [400]. However, this still bares the question of whether these changes are induced by stress or by changes in the cellular environment. As mentioned previously, regular myelination and communication between oligodendrocytes and neurons remains central to the maintenance of a healthy brain [401]. The communicability of oligodendrocytes with both neurons and other glia suggests that these deficits may be driven by complex interactions with the cellular environment, for example, reduced metabolic support from astrocytes, and increased neuroinflammation.

39 4. CONCLUSIONS AND FUTURE DIRECTIONS

An extensive body of work has contributed to our understanding of how stress shapes the brain and how this likely contributes to psychopathology, yet we are still a distance away from understanding these effects with clarity. Progress has largely been achieved through rodent preclinical models. These models remain essential to advancing our understanding of the neurobiology of stress, particularly with de-convoluting the complexity of stress in a controlled manner (often not possible in human studies where stress exposures are varied and complex).

Animal model studies have particularly provided valuable knowledge about how different types, timings and durations of stress shape the cellular environment through a complex inter- relationship between cells, as all major cell types are in some regard affected by stress and also associated with psychopathology. Although the data provided by rodent models have provided fundamental understanding and insight into the effects of stress, a challenge in the field has been translating the effects identified in pre-clinical rodent models into comparable effects in humans, as very little consideration has been given to stress history in human cohorts. To further progress this research area, alongside rodent models, the study of both living (molecular brain imaging methods such as PET) and postmortem (cellular and molecular) human cohorts is important.

At the macrostructural level, human studies have offered insight into how the brain is shaped by stress and potential roles this may play in psychopathology. However, several questions remain to be answered by future studies:

Firstly, how do individual experiences of stress contribute to individual psychiatric disorder trajectories? Macrostructural studies highlight that the recurrence [54], timing, and chronicity of stress are an important consideration as to how the brain is shaped [402], yet these factors are often not considered in clinical psychiatry research despite stress being a leading risk factor for psychopathology. Curation of specialised cohorts that consider the individual sequalae shaping

40 psychopathology – accounting for stress history, treatment strategies and disorder progression – may be crucial to accurately understand the complex trajectory of how psychiatric disorders develop.

Secondly, how does stress differentially contribute to structural changes in the brain throughout development? Determining which aspects of stress cause persistent effects and influence developmental trajectories is important in being able to identify at-risk individuals and design the interventions that prevent them from developing psychiatric disorders. Finally, what are the molecular and cellular processes that underlie these macrostructural changes? Characterising the molecular and cellular impacts of stress and how these influence psychopathology will enable development of new targets for prevention and treatment.

At the cytoarchitectural level, several additional questions concerning the temporal sequence in which morphological and functional changes occur in response to stress are important for future studies to address. For example, is there a domino effect (in which case, what is the first cell type to “fall”?), or are all cell types affected equally in parallel? To what extent are cellular changes recoverable and when do cellular deficits begin to contribute to the development of behavioural and cognitive dysfunction? We suggest that, while stress affects cellular populations at any life stage, brain-cell changes induced during periods when cellular population trajectories are being established, could lead to sustained atypical connectivity contributing to psychopathology. This is likely compounded by an increased responsiveness of different brain cells to stress, as well as alterations to stress responsiveness through cell-to-cell communication. Thus persistent dysregulation of these circuits via these mechanisms during early-life and adolescence could increase the risk of developing behavioural and cognitive symptoms throughout development.

Another important question is, why do certain brain areas appear to be more vulnerable to stress, and what are the cellular and molecular drivers of this vulnerability? The recent explosion of

41 single-cell sequencing and high-resolution molecule/cell visualisation techniques being applied to the brain indicate that the brain consists of an enormously diverse profile of cells that varies according to brain region [403]. It is likely that subpopulations of cells are uniquely vulnerable to stress [265], and this precipitates specific effects across cellular networks that persistently impact on brain circuitry. Exploring this possibility in cases of psychopathology, especially in the context of stress exposure or history, will assist in improving current understandings of cellular pathologies in psychiatry that are induced by stress. This approach can also assist in the identification of specific patient subgroups characterised by similar underlying pathology, the biomarkers for identification of these patient subgroups, and the biological targets for their treatment.

42 Acknowledgements: This work was supported by an NHMRC Early Career Fellowship

(APP1105445) awarded to Dr Matosin and grants from the Brain Behavior Research Foundation

(BBRF/NARSAD Young Investigator Grant) as well as the Rebecca L. Cooper Medical

Research Foundation. Dr Mechawar’s research is supported by grants from CIHR, HBHL and

ERA-NET NEURON.

43 REFERENCES

[1] B.S. McEwen, Neurobiological and systemic effects of chronic stress, Chronic Stress 1 (2017) 1-11. [2] B.S. McEwen, N.P. Bowles, J.D. Gray, M.N. Hill, R.G. Hunter, I.N. Karatsoreos, C. Nasca, Mechanisms of stress in the brain, Nat Neurosci 18(10) (2015) 1353-1363. [3] P. Kim, G.W. Evans, M. Angstadt, S.S. Ho, C.S. Sripada, J.E. Swain, I. Liberzon, K.L. Phan, Effects of childhood poverty and chronic stress on emotion regulatory brain function in adulthood, Proc Natl Acad Sci USA 110(46) (2013) 18442-18447. [4] L. Shanahan, W. Copeland, E. Jane Costello, A. Angold, Specificity of putative psychosocial risk factors for psychiatric disorders in children and adolescents, J Child Psychol Psychiatry 49(1) (2008) 34- 42. [5] N. Breslau, Epidemiologic studies of trauma, posttraumatic stress disorder, and other psychiatric disorders, Can J Psychiatry 47 (2002) 923-929. [6] S. Cohen, D. Janicki-Deverts, G.E. Miller, Psychological stress and disease, Jama 298(14) (2007) 1685-1687. [7] B.S. McEwen, P.J. Gianaros, Central role of the brain in stress and adaptation: links to socioeconomic status, health, and disease, Ann NY Acad Sci 1186 (2010) 190. [8] Bruce S. McEwen, John H. Morrison, The brain on stress: vulnerability and plasticity of the prefrontal cortex over the life course, Neuron 79(1) (2013) 16-29. [9] F. Hollis, F. Duclot, A. Gunjan, M. Kabbaj, Individual differences in the effect of social defeat on anhedonia and histone acetylation in the rat hippocampus, Horm Behav 59(3) (2011) 331-337. [10] A. Du Preez, T. Law, D. Onorato, Y.M. Lim, P. Eiben, K. Musaelyan, M. Egeland, A. Hye, P.A. Zunszain, S. Thuret, C.M. Pariante, C. Fernandes, The type of stress matters: repeated injection and permanent social isolation stress in male mice have a differential effect on anxiety- and depressive-like behaviours, and associated biological alterations, Transl Psychiatry 10(1) (2020) 325. [11] E.B. Binder, Dissecting the molecular mechanisms of gene x environment interactions: implications for diagnosis and treatment of stress-related psychiatric disorders, Eur J Psychotraumatol 8 (sup5) (2017) 1412745. [12] R. Uher, A. Zwicker, Etiology in psychiatry: embracing the reality of poly‐‐ gene environmental causation of mental illness, World Psychiatry 16(2) (2017) 121-129. [13] W.C. Drevets, Orbitofrontal cortex function and structure in depression, Ann NY Acad Sci 1121 (2007) 499-527. [14] A. Saleh, G.G. Potter, D.R. McQuoid, B. Boyd, R. Turner, J.R. MacFall, W.D. Taylor, Effects of early life stress on depression, cognitive performance and brain morphology, Psychol Med 47(1) (2017) 171-181. [15] S. Daftary, E. Van Enkevort, A. Kulikova, M. Legacy, E.S. Brown, Relationship between depressive symptom severity and amygdala volume in a large community-based sample, Psychiatry Res Neuroimaging 283 (2019) 77-82. [16] M.D. Wheelock, D. Rangaprakash, N.G. Harnett, K.H. Wood, T.R. Orem, S. Mrug, D.A. Granger, G. Deshpande, D.C. Knight, Psychosocial stress reactivity is associated with decreased whole-brain network efficiency and increased amygdala centrality, Behav Neurosci 132(6) (2018) 561-572. [17] J.T. Baker, D.G. Dillon, L.M. Patrick, J.L. Roffman, R.O. Brady, D.A. Pizzagalli, D. Öngür, A.J. Holmes, Functional connectomics of affective and psychotic pathology, Proc Natl Acad Sci USA 116(18) (2019) 9050-9059. [18] N.V. Kraguljac, D.M. White, N. Hadley, J.A. Hadley, L. Ver Hoef, E. Davis, A.C. Lahti, Aberrant Hippocampal Connectivity in Unmedicated Patients With Schizophrenia and Effects of Antipsychotic Medication: A Longitudinal Resting State Functional MRI Study, Schizophr Bull 42(4) (2016) 1046- 1055. [19] T.T. Yang, A.N. Simmons, S.C. Matthews, S.F. Tapert, G.K. Frank, J.E. Max, A. Bischoff-Grethe, A.E. Lansing, G. Brown, I.A. Strigo, J. Wu, M.P. Paulus, Adolescents with major depression demonstrate increased amygdala activation, J Am Acad Child Adolesc Psychiatry 49(1) (2010) 42-51.

44 [20] S. Wang, Y. Zhao, L. Zhang, X. Wang, X. Wang, B. Cheng, K. Luo, Q. Gong, Stress and the brain: Perceived stress mediates the impact of the superior frontal gyrus spontaneous activity on depressive symptoms in late adolescence, Hum Brain Mapp 40(17) (2019) 4982-4993. [21] H. Li, W. Li, D. Wei, Q. Chen, T. Jackson, Q. Zhang, J. Qiu, Examining brain structures associated with perceived stress in a large sample of young adults via voxel-based morphometry, NeuroImage 92 (2014) 1-7. [22] S.J. Lupien, B.S. McEwen, M.R. Gunnar, C. Heim, Effects of stress throughout the lifespan on the brain, behaviour and cognition, Nat Neurosci 10 (2009) 434. [23] S.J. Brooks, V. Naidoo, A. Roos, J.P. Fouché, C. Lochner, D.J. Stein, Early-life adversity and orbitofrontal and cerebellar volumes in adults with obsessive-compulsive disorder: voxel-based morphometry study, Br J Psychiatry 208(1) (2016) 34-41. [24] L. Baldaçara, J.G. Fiorani Borgio, C. Araújo, F. Nery-Fernandes, A.L. Taveres Lacerda, W.A. dos Santos Moraes, M.B.M. Macedo Montaño, M. Rocha, L.C. Quarantini, A. Schoedl, Relationship between structural abnormalities in the cerebellum and dementia, posttraumatic stress disorder and bipolar disorder, Dement Neuropsychol 6(4) (2012) 203-211. [25] E.B. Ansell, K. Rando, K. Tuit, J. Guarnaccia, R. Sinha, Cumulative Adversity and Smaller Gray Matter Volume in Medial Prefrontal, Anterior Cingulate, and Insula Regions, Biol Psychiatry 72(1) (2012) 57-64. [26] E.K. Miller, J.D. Cohen, An integrative theory of prefrontal cortex function, Annu Rev Neurosci 24 (2001) 167-202. [27] M.S. Weinberg, D.C. Johnson, A.P. Bhatt, R.L. Spencer, Medial prefrontal cortex activity can disrupt the expression of stress response habituation, Neuroscience 168(3) (2010) 744-756. [28] J.J. Radley, C.M. Arias, P.E. Sawchenko, Regional differentiation of the medial prefrontal cortex in regulating adaptive responses to acute emotional stress, The Journal of neuroscience : the official journal of the Society for Neuroscience 26(50) (2006) 12967-12976. [29] D. Diorio, V. Viau, M.J. Meaney, The role of the medial prefrontal cortex (cingulate gyrus) in the regulation of hypothalamic-pituitary-adrenal responses to stress, The Journal of neuroscience : the official journal of the Society for Neuroscience 13(9) (1993) 3839-3847. [30] L.D. Godoy, M.T. Rossignoli, P. Delfino-Pereira, N. Garcia-Cairasco, E.H. De Lima Umeoka, A Comprehensive Overview on Stress Neurobiology: Basic Concepts and Clinical Implications, Front Behav Neurosci 12 (2018) 127. [31] J.L. Hanson, M.K. Chung, B.B. Avants, E.A. Shirtcliff, J.C. Gee, R.J. Davidson, S.D. Pollak, Early stress is associated with alterations in the orbitofrontal cortex: a tensor-based morphometry investigation of brain structure and behavioral risk, The Journal of neuroscience : the official journal of the Society for Neuroscience 30(22) (2010) 7466-7472. [32] N.E. Holz, R. Boecker, E. Hohm, K. Zohsel, A.F. Buchmann, D. Blomeyer, C. Jennen-Steinmetz, S. Baumeister, S. Hohmann, I. Wolf, M.M. Plichta, G. Esser, M. Schmidt, A. Meyer-Lindenberg, T. Banaschewski, D. Brandeis, M. Laucht, The long-term impact of early life poverty on orbitofrontal cortex volume in adulthood: results from a prospective study over 25 years, Neuropsychopharmacology 40(4) (2015) 996-1004. [33] M. Monninger, E.J. Kraaijenvanger, T.M. Pollok, R. Boecker-Schlier, C. Jennen-Steinmetz, S. Baumeister, G. Esser, M. Schmidt, A. Meyer-Lindenberg, M. Laucht, D. Brandeis, T. Banaschewski, N.E. Holz, The long-term impact of early life stress on orbitofrontal cortical thickness, Cerebral cortex (New York, N.Y. : 1991) 3(3) (2020) 1307-1317. [34] G.L. Moreno, J. Bruss, N.L. Denburg, Increased perceived stress is related to decreased prefrontal cortex volumes among older adults, Journal of clinical and experimental neuropsychology 39(4) (2017) 313-325. [35] A.L. Gold, M.A. Sheridan, M. Peverill, D.S. Busso, H.K. Lambert, S. Alves, D.S. Pine, K.A. McLaughlin, Childhood abuse and reduced cortical thickness in brain regions involved in emotional processing, J Child Psychol Psychiatry 57(10) (2016) 1154-1164. [36] M.D. Underwood, M.J. Bakalian, T. Escobar, S. Kassir, J.J. Mann, V. Arango, Early-Life Adversity, but Not Suicide, Is Associated With Less Prefrontal Cortex Gray Matter in Adulthood, Int J Neuropsychopharmacol 22(5) (2019) 349-357. [37] C. Hoffmann, T.E. Van Rheenen, S.G. Mancuso, A. Zalesky, J. Bruggemann, R.K. Lenroot, S. Sundram, C.S. Weickert, T.W. Weickert, C. Pantelis, V. Cropley, C.A. Bousman, Exploring the

45 moderating effects of dopaminergic polymorphisms and childhood adversity on brain morphology in schizophrenia-spectrum disorders, Psychiatry Res Neuroimaging 281 (2018) 61-68. [38] C.M. Heim, H.S. Mayberg, T. Mletzko, C.B. Nemeroff, J.C. Pruessner, Decreased cortical representation of genital somatosensory field after childhood sexual abuse, Am J Psychiatry 170(6) (2013) 616-623. [39] P.J. Gianaros, J.A. Horenstein, S. Cohen, K.A. Matthews, S.M. Brown, J.D. Flory, H.D. Critchley, S.B. Manuck, A.R. Hariri, Perigenual anterior cingulate morphology covaries with perceived social standing, Social cognitive and affective neuroscience 2(3) (2007) 161-173. [40] P.J. Gianaros, A.L. Marsland, L.K. Sheu, K.I. Erickson, T.D. Verstynen, Inflammatory pathways link socioeconomic inequalities to white matter architecture, Cerebral cortex (New York, N.Y. : 1991) 23(9) (2013) 2058-2071. [41] J. Savitz, W.C. Drevets, Bipolar and major depressive disorder: Neuroimaging the developmental- degenerative divide, Neurosci Biobehav Rev 33(5) (2009) 699-771. [42] T. Wise, J. Radua, E. Via, N. Cardoner, O. Abe, T.M. Adams, F. Amico, Y. Cheng, J.H. Cole, C. de Azevedo Marques Périco, D.P. Dickstein, T.F.D. Farrow, T. Frodl, G. Wagner, I.H. Gotlib, O. Gruber, B.J. Ham, D.E. Job, M.J. Kempton, M.J. Kim, P.C.M.P. Koolschijn, G.S. Malhi, D. Mataix-Cols, A.M. McIntosh, A.C. Nugent, J.T. O'Brien, S. Pezzoli, M.L. Phillips, P.S. Sachdev, G. Salvadore, S. Selvaraj, A.C. Stanfield, A.J. Thomas, M.J. van Tol, N.J.A. van der Wee, D.J. Veltman, A.H. Young, C.H. Fu, A.J. Cleare, D. Arnone, Common and distinct patterns of grey-matter volume alteration in major depression and bipolar disorder: evidence from voxel-based meta-analysis, Mol Psychiatry 22 (2016) 1455. [43] D.P. Hibar, L.T. Westlye, N.T. Doan, N. Jahanshad, J.W. Cheung, C.R.K. Ching, A. Versace, A.C. Bilderbeck, A. Uhlmann, B. Mwangi, B. Kramer, B. Overs, C.B. Hartberg, C. Abe, D. Dima, D. Grotegerd, E. Sprooten, E. Boen, E. Jimenez, F.M. Howells, G. Delvecchio, H. Temmingh, J. Starke, J.R.C. Almeida, J.M. Goikolea, J. Houenou, L.M. Beard, L. Rauer, L. Abramovic, M. Bonnin, M.F. Ponteduro, M. Keil, M.M. Rive, N. Yao, N. Yalin, P. Najt, P.G. Rosa, R. Redlich, S. Trost, S. Hagenaars, S.C. Fears, S. Alonso-Lana, T.G.M. van Erp, T. Nickson, T.M. Chaim-Avancini, T.B. Meier, T. Elvsashagen, U.K. Haukvik, W.H. Lee, A.H. Schene, A.J. Lloyd, A.H. Young, A. Nugent, A.M. Dale, A. Pfennig, A.M. McIntosh, B. Lafer, B.T. Baune, C.J. Ekman, C.A. Zarate, C.E. Bearden, C. Henry, C. Simhandl, C. McDonald, C. Bourne, D.J. Stein, D.H. Wolf, D.M. Cannon, D.C. Glahn, D.J. Veltman, E. Pomarol-Clotet, E. Vieta, E.J. Canales-Rodriguez, F.G. Nery, F.L.S. Duran, G.F. Busatto, G. Roberts, G.D. Pearlson, G.M. Goodwin, H. Kugel, H.C. Whalley, H.G. Ruhe, J.C. Soares, J.M. Fullerton, J.K. Rybakowski, J. Savitz, K.T. Chaim, M. Fatjo-Vilas, M.G. Soeiro-de-Souza, M.P. Boks, M.V. Zanetti, M.C.G. Otaduy, M.S. Schaufelberger, M. Alda, M. Ingvar, M.L. Phillips, M.J. Kempton, M. Bauer, M. Landen, N.S. Lawrence, N.E.M. van Haren, N.R. Horn, N.B. Freimer, O. Gruber, P.R. Schofield, P.B. Mitchell, R.S. Kahn, R. Lenroot, R. Machado-Vieira, R.A. Ophoff, S. Sarro, S. Frangou, T.D. Satterthwaite, T. Hajek, U. Dannlowski, U.F. Malt, V. Arolt, W.F. Gattaz, W.C. Drevets, X. Caseras, I. Agartz, P.M. Thompson, O.A. Andreassen, Cortical abnormalities in bipolar disorder: an MRI analysis of 6503 individuals from the ENIGMA Bipolar Disorder Working Group, Mol Psychiatry 23(4) (2018) 932- 942. [44] U.K. Haukvik, C.B. Hartberg, I. Agartz, Schizophrenia--what does structural MRI show?, Tidsskrift for den Norske laegeforening : tidsskrift for praktisk medicin, ny raekke 133(8) (2013) 850-853. [45] M.T. Treadway, M.L. Waskom, D.G. Dillon, A.J. Holmes, M.T.M. Park, M.M. Chakravarty, S.J. Dutra, F.E. Polli, D.V. Iosifescu, M. Fava, J.D.E. Gabrieli, D.A. Pizzagalli, Illness progression, recent stress, and morphometry of hippocampal subfields and medial prefrontal cortex in major depression, Biol Psychiatry 77(3) (2015) 285-294. [46] N. Dusi, M. Bellani, C. Perlini, L. Squarcina, V. Marinelli, L. Finos, C.A. Altamura, M. Ruggeri, P. Brambilla, Progressive disability and prefrontal shrinkage in schizophrenia patients with poor outcome: A 3-year longitudinal study, Schizophrenia research 179 (2017) 104-111. [47] C. Liston, B.S. McEwen, B. Casey, Psychosocial stress reversibly disrupts prefrontal processing and attentional control, Proc Natl Acad Sci USA 106(3) (2009) 912-917. [48] J.H. Yoon, M.J. Minzenberg, S. Ursu, B.S. Ryan Walter, C. Wendelken, J.D. Ragland, C.S. Carter, Association of dorsolateral prefrontal cortex dysfunction with disrupted coordinated brain activity in schizophrenia: relationship with impaired cognition, behavioral disorganization, and global function, Am J Psychiatry 165(8) (2008) 1006-1014.

46 [49] J.M. McKlveen, R.L. Morano, M. Fitzgerald, S. Zoubovsky, S.N. Cassella, J.R. Scheimann, S. Ghosal, P. Mahbod, B.A. Packard, B. Myers, Chronic stress increases prefrontal inhibition: a mechanism for stress-induced prefrontal dysfunction, Biol Psychiatry 80(10) (2016) 754-764. [50] V.G. Carrion, C.F. Weems, K. Richert, B.C. Hoffman, A.L. Reiss, Decreased prefrontal cortical volume associated with increased bedtime cortisol in traumatized youth, Biol Psychiatry 68(5) (2010) 491-493. [51] M.T. Mitterschiffthaler, V. Kumari, G.S. Malhi, R.G. Brown, V.P. Giampietro, M.J. Brammer, J. Suckling, L. Poon, A. Simmons, C. Andrew, T. Sharma, Neural response to pleasant stimuli in anhedonia: an fMRI study, Neuroreport 14(2) (2003) 177-182. [52] C.P. Carr, C.M. Martins, A.M. Stingel, V.B. Lemgruber, M.F. Juruena, The role of early life stress in adult psychiatric disorders: a systematic review according to childhood trauma subtypes, The Journal of nervous and mental disease 201(12) (2013) 1007-1020. [53] A.-L. van Harmelen, M.-J. van Tol, N.J.A. van der Wee, D.J. Veltman, A. Aleman, P. Spinhoven, M.A. van Buchem, F.G. Zitman, B.W.J.H. Penninx, B.M. Elzinga, Reduced Medial Prefrontal Cortex Volume in Adults Reporting Childhood Emotional Maltreatment, Biol Psychiatry 68(9) (2010) 832-838. [54] B.S. McEwen, P.J. Gianaros, Stress- and allostasis-induced brain plasticity, Annu Rev Med 62 (2011) 431-445. [55] X. Zhong, Q. Ming, D. Dong, X. Sun, C. Cheng, G. Xiong, C. Li, X. Zhang, S. Yao, Childhood Maltreatment Experience Influences Neural Response to Psychosocial Stress in Adults: An fMRI Study, Front Psychol 10 (2020) 2961-2961. [56] A. Tomoda, A. Polcari, C.M. Anderson, M.H. Teicher, Reduced visual cortex gray matter volume and thickness in young adults who witnessed domestic violence during childhood, PLoS One 7(12) (2012) e52528. [57] A. Tomoda, Y.S. Sheu, K. Rabi, H. Suzuki, C.P. Navalta, A. Polcari, M.H. Teicher, Exposure to parental verbal abuse is associated with increased gray matter volume in superior temporal gyrus, Neuroimage 54 Suppl 1 (2011) S280-6. [58] R.D. Rubin, P.D. Watson, M.C. Duff, N.J. Cohen, The role of the hippocampus in flexible cognition and social behavior, Front Hum Neurosci 8 (2014) 742-742. [59] J.P. Herman, W.E. Cullinan, M.I. Morano, H. Akil, S.J. Watson, Contribution of the ventral subiculum to inhibitory regulation of the hypothalamo-pituitary-adrenocortical axis, Journal of neuroendocrinology 7(6) (1995) 475-482. [60] J.D. Dunn, S.E. Orr, Differential plasma corticosterone responses to hippocampal stimulation, Experimental brain research 54(1) (1984) 1-6. [61] L. Tannenholz, J.C. Jimenez, M.A. Kheirbek, Local and regional heterogeneity underlying hippocampal modulation of cognition and mood, Front Behav Neurosci 8 (2014) 147. [62] M.S. Fanselow, H.-W. Dong, Are the dorsal and ventral hippocampus functionally distinct structures?, Neuron 65(1) (2010) 7-19. [63] T.J. Schoenfeld, H.C. McCausland, H.D. Morris, V. Padmanaban, H.A. Cameron, Stress and loss of adult neurogenesis differentially reduce hippocampal volume, Biol Psychiatry 82(12) (2017) 914-923. [64] A. Floriou-Servou, L. von Ziegler, L. Stalder, O. Sturman, M. Privitera, A. Rassi, A. Cremonesi, B. Thöny, J. Bohacek, Distinct proteomic, transcriptomic, and epigenetic stress responses in dorsal and ventral hippocampus, Biol Psychiatry 84(7) (2018) 531-541. [65] A. Sarrieau, M. Dussaillant, R.M. Sapolsky, D.H. Aitken, A. Olivier, S. Lal, W.H. Rostene, R. Quirion, M.J. Meaney, Glucocorticoid binding sites in human temporal cortex, Brain research 442(1) (1988) 157-160. [66] J.M. Reul, E.R. de Kloet, Two receptor systems for corticosterone in rat brain: microdistribution and differential occupation, Endocrinology 117(6) (1985) 2505-2511. [67] F.L. Woon, S. Sood, D.W. Hedges, Hippocampal volume deficits associated with exposure to psychological trauma and posttraumatic stress disorder in adults: A meta-analysis, Prog Neuropsychopharmacol Biol Psychiatry 34(7) (2010) 1181-1188. [68] A.S. Zannas, D.R. McQuoid, M.E. Payne, D.C. Steffens, J.R. MacFall, A. Ashley-Koch, W.D. Taylor, Negative life stress and longitudinal hippocampal volume changes in older adults with and without depression, Journal of psychiatric research 47(6) (2013) 829-834.

47 [69] R. Admon, D. Leykin, G. Lubin, V. Engert, J. Andrews, J. Pruessner, T. Hendler, Stress-induced reduction in hippocampal volume and connectivity with the ventromedial prefrontal cortex are related to maladaptive responses to stressful military service, Hum Brain Mapp 34(11) (2013) 2808-2816. [70] D.G. Weissman, H.K. Lambert, A.M. Rodman, M. Peverill, M.A. Sheridan, K.A. McLaughlin, Reduced hippocampal and amygdala volume as a mechanism underlying stress sensitization to depression following childhood trauma, Depression and anxiety 37(9) (2020) 916-925. [71] G.S. Malhi, P. Das, T. Outhred, L. Irwin, D. Gessler, Z. Bwabi, R. Bryant, Z. Mannie, The effects of childhood trauma on adolescent hippocampal subfields, Aust N Z J Psychiatry 53(5) (2019) 447-457. [72] P. Butterworth, N. Cherbuin, P. Sachdev, K.J. Anstey, The association between financial hardship and amygdala and hippocampal volumes: results from the PATH through life project, Social cognitive and affective neuroscience 7(5) (2012) 548-556. [73] L.R. Piccolo, K.G. Noble, N. Pediatric Imaging, S. Genetics, Perceived stress is associated with smaller hippocampal volume in adolescence, Psychophysiology 55(5) (2018) e13025-e13025. [74] M.H. Teicher, C.M. Anderson, A. Polcari, Childhood maltreatment is associated with reduced volume in the hippocampal subfields CA3, dentate gyrus, and subiculum, Proc Natl Acad Sci USA 109(9) (2012) E563-E572. [75] R. Colle, T. Segawa, M. Chupin, M.N.T.K. Tran Dong, P. Hardy, B. Falissard, O. Colliot, D. Ducreux, E. Corruble, Early life adversity is associated with a smaller hippocampus in male but not female depressed in-patients: a case–control study, BMC Psychiatry 17(1) (2017) 71. [76] K.L. Humphreys, L.S. King, M.D. Sacchet, M.C. Camacho, N.L. Colich, S.J. Ordaz, T.C. Ho, I.H. Gotlib, Evidence for a sensitive period in the effects of early life stress on hippocampal volume, Dev Sci 22(3) (2019) e12775. [77] B. Dahmen, V.B. Puetz, W. Scharke, G.G. von Polier, B. Herpertz-Dahlmann, K. Konrad, Effects of Early-Life Adversity on Hippocampal Structures and Associated HPA Axis Functions, Dev Neurosci 40(1) (2018) 13-22. [78] A.M. LoPilato, K. Goines, J. Addington, C.E. Bearden, K.S. Cadenhead, T.D. Cannon, B.A. Cornblatt, D.H. Mathalon, T.H. McGlashan, L. Seidman, D.O. Perkins, M.T. Tsuang, S.W. Woods, E.F. Walker, Impact of childhood adversity on corticolimbic volumes in youth at clinical high-risk for psychosis, Schizophrenia research 213 (2019) 48-55. [79] T. Frodl, D. Janowitz, L. Schmaal, L. Tozzi, H. Dobrowolny, D.J. Stein, D.J. Veltman, K. Wittfeld, T.G.M. van Erp, N. Jahanshad, A. Block, K. Hegenscheid, H. Völzke, J. Lagopoulos, S.N. Hatton, I.B. Hickie, E.M. Frey, A. Carballedo, S.J. Brooks, D. Vuletic, A. Uhlmann, I.M. Veer, H. Walter, K. Schnell, D. Grotegerd, V. Arolt, H. Kugel, E. Schramm, C. Konrad, B. Zurowski, B.T. Baune, N.J.A. van der Wee, M.J. van Tol, B. Penninx, P.M. Thompson, D.P. Hibar, U. Dannlowski, H.J. Grabe, Childhood adversity impacts on brain subcortical structures relevant to depression, Journal of psychiatric research 86 (2017) 58-65. [80] P. Mikolas, L. Tozzi, K. Doolin, C. Farrell, V. O'Keane, T. Frodl, Effects of early life adversity and FKBP5 genotype on hippocampal subfields volume in major depression, J Affect Disord 252 (2019) 152- 159. [81] U. Rao, L.A. Chen, A.S. Bidesi, M.U. Shad, M.A. Thomas, C.L. Hammen, Hippocampal changes associated with early-life adversity and vulnerability to depression, Biol Psychiatry 67(4) (2010) 357-364. [82] P.R. Szeszko, J.D. Betensky, C. Mentschel, H. Gunduz-Bruce, T. Lencz, M. Ashtari, A.K. Malhotra, R.M. Bilder, Increased stress and smaller anterior hippocampal volume, Neuroreport 17(17) (2006) 1825- 1828. [83] J. Chen, Z. Wei, H. Han, L. Jin, C. Xu, D. Dong, J. Lu, G. Wan, Z. Peng, An Effect of Chronic Stress on Prospective Memory via Alteration of Resting-State Hippocampal Subregion Functional Connectivity, Sci Rep 9(1) (2019) 19698. [84] M.W. Gilbertson, M.E. Shenton, A. Ciszewski, K. Kasai, N.B. Lasko, S.P. Orr, R.K. Pitman, Smaller hippocampal volume predicts pathologic vulnerability to psychological trauma, Nat Neurosci 5(11) (2002) 1242-1247. [85] B. Cao, I.C. Passos, B. Mwangi, H. Amaral-Silva, J. Tannous, M.J. Wu, G.B. Zunta-Soares, J.C. Soares, Hippocampal subfield volumes in mood disorders, Mol Psychiatry 22(9) (2017) 1352-1358. [86] J.D. Bremner, M. Narayan, E.R. Anderson, L.H. Staib, H.L. Miller, D.S. Charney, Hippocampal volume reduction in major depression, Am J Psychiatry 157(1) (2000) 115-118.

48 [87] S.J.M. Arnold, E.I. Ivleva, T.A. Gopal, A.P. Reddy, H. Jeon-Slaughter, C.B. Sacco, A.N. Francis, N. Tandon, A.S. Bidesi, B. Witte, G. Poudyal, G.D. Pearlson, J.A. Sweeney, B.A. Clementz, M.S. Keshavan, C.A. Tamminga, Hippocampal volume is reduced in schizophrenia and schizoaffective disorder but not in psychotic bipolar I disorder demonstrated by both manual tracing and automated parcellation (FreeSurfer), Schizophr Bull 41(1) (2015) 233-249. [88] K.T. Kronmuller, J. Pantel, S. Kohler, D. Victor, F. Giesel, V.A. Magnotta, C. Mundt, M. Essig, J. Schroder, Hippocampal volume and 2-year outcome in depression, Br J Psychiatry 192(6) (2008) 472- 473. [89] W.D. Taylor, D.R. McQuoid, M.E. Payne, A.S. Zannas, J.R. MacFall, D.C. Steffens, Hippocampus atrophy and the longitudinal course of late-life depression, The American journal of geriatric psychiatry : official journal of the American Association for Geriatric Psychiatry 22(12) (2014) 1504-1512. [90] M. Serra-Blasco, J. de Diego-Adelino, Y. Vives-Gilabert, J. Trujols, D. Puigdemont, M. Carceller- Sindreu, V. Perez, E. Alvarez, M.J. Portella, Naturalistic course of major depressive disorder predicted by clinical and structural neuroimaging data: A 5-year follow up, Depression and anxiety 33(11) (2016) 1055-1064. [91] T.S. Frodl, N. Koutsouleris, R. Bottlender, C. Born, M. Jager, I. Scupin, M. Reiser, H.J. Moller, E.M. Meisenzahl, Depression-related variation in brain morphology over 3 years: effects of stress?, Archives of general psychiatry 65(10) (2008) 1156-1165. [92] S.A. Schobel, N.M. Lewandowski, C.M. Corcoran, H. Moore, T. Brown, D. Malaspina, S.A. Small, Differential targeting of the CA1 subfield of the hippocampal formation by schizophrenia and related psychotic disorders, Archives of general psychiatry 66(9) (2009) 938-946. [93] K. Vakili, S.S. Pillay, B. Lafer, M. Fava, P.F. Renshaw, C.M. Bonello-Cintron, D.A. Yurgelun- Todd, Hippocampal volume in primary unipolar major depression: a magnetic resonance imaging study, Biol Psychiatry 47(12) (2000) 1087-1090. [94] C. Saylam, H. Üçerler, Ö. Kitiş, E. Ozand, A.S. Gönül, Reduced hippocampal volume in drug-free depressed patients, Surg Radiol Anat 28(1) (2006) 82-87. [95] S.A. Papagni, S. Benetti, S. Arulanantham, E. McCrory, P. McGuire, A. Mechelli, Effects of stressful life events on human brain structure: a longitudinal voxel-based morphometry study, Stress (Amsterdam, Netherlands) 14(2) (2011) 227-232. [96] B.W. Balleine, S. Killcross, Parallel incentive processing: an integrated view of amygdala function, Trends Neurosci 29(5) (2006) 272-279. [97] K.J. Ressler, Amygdala activity, fear, and anxiety: modulation by stress, Biol Psychiatry 67(12) (2010) 1117-1119. [98] A.A. Taren, P.J. Gianaros, C.M. Greco, E.K. Lindsay, A. Fairgrieve, K.W. Brown, R.K. Rosen, J.L. Ferris, E. Julson, A.L. Marsland, J.K. Bursley, J. Ramsburg, J.D. Creswell, Mindfulness meditation training alters stress-related amygdala resting state functional connectivity: a randomized controlled trial, Social cognitive and affective neuroscience 10(12) (2015) 1758-1768. [99] B.K. Hölzel, J. Carmody, K.C. Evans, E.A. Hoge, J.A. Dusek, L. Morgan, R.K. Pitman, S.W. Lazar, Stress reduction correlates with structural changes in the amygdala, Social cognitive and affective neuroscience 5(1) (2010) 11-17. [100] M.E. Sublette, H.C. Galfalvy, M.A. Oquendo, C.P. Bart, N. Schneck, V. Arango, J.J. Mann, Relationship of recent stress to amygdala volume in depressed and healthy adults, J Affect Disord 203 (2016) 136-142. [101] H.J.F. van Marle, E.J. Hermans, S. Qin, G. Fernández, Enhanced resting-state connectivity of amygdala in the immediate aftermath of acute psychological stress, Neuroimage 53(1) (2010) 348-354. [102] J.D. Bremner, Functional neuroanatomical correlates of traumatic stress revisited 7 years later, this time with data, Psychopharmacol bull 37(2) (2003) 6-25. [103] D. Seo, K.A. Tsou, E.B. Ansell, M.N. Potenza, R. Sinha, Cumulative adversity sensitizes neural response to acute stress: association with health symptoms, Neuropsychopharmacology 39(3) (2014) 670- 680. [104] F.L. Woon, D.W. Hedges, Amygdala Volume in Adults with Posttraumatic Stress Disorder: A Meta-Analysis, J Neuropsychiatry Clin Neurosci 21(1) (2009) 5-12. [105] R. Cacciaglia, F. Nees, O. Grimm, S. Ridder, S.T. Pohlack, S.J. Diener, C. Liebscher, H. Flor, Trauma exposure relates to heightened stress, altered amygdala morphology and deficient extinction learning: Implications for psychopathology, Psychoneuroendocrinology 76 (2017) 19-28.

49 [106] T. Yamamoto, S. Toki, G.J. Siegle, M. Takamura, Y. Takaishi, S. Yoshimura, G. Okada, T. Matsumoto, T. Nakao, H. Muranaka, Y. Kaseda, T. Murakami, Y. Okamoto, S. Yamawaki, Increased amygdala reactivity following early life stress: a potential resilience enhancer role, BMC Psychiatry 17(1) (2017) 27. [107] S. Whittle, M. Dennison, N. Vijayakumar, J.G. Simmons, M. Yücel, D.I. Lubman, C. Pantelis, N.B. Allen, Childhood Maltreatment and Psychopathology Affect Brain Development During Adolescence, J Am Acad Child Adolesc Psychiatry 52(9) (2013) 940-952. [108] P. Pechtel, K. Lyons-Ruth, C.M. Anderson, M.H. Teicher, Sensitive periods of amygdala development: the role of maltreatment in preadolescence, Neuroimage 97 (2014) 236-244. [109] M.H. Teicher, J.A. Samson, Annual Research Review: Enduring neurobiological effects of childhood abuse and neglect, J Child Psychol Psychiatry 57(3) (2016) 241-266. [110] M. Driessen, J. Herrmann, K. Stahl, M. Zwaan, S. Meier, A. Hill, M. Osterheider, D. Petersen, Magnetic resonance imaging volumes of the hippocampus and the amygdala in women with borderline personality disorder and early traumatization, Archives of general psychiatry 57(12) (2000) 1115-1122. [111] C.G. Schmahl, E. Vermetten, B.M. Elzinga, J. Douglas Bremner, Magnetic resonance imaging of hippocampal and amygdala volume in women with childhood abuse and borderline personality disorder, Psychiatry research 122(3) (2003) 193-198. [112] A. Oshri, J.C. Gray, M.M. Owens, S. Liu, E.B. Duprey, L.H. Sweet, J. MacKillop, Adverse Childhood Experiences and Amygdalar Reduction: High-Resolution Segmentation Reveals Associations With Subnuclei and Psychiatric Outcomes, Child Maltreat 24(4) (2019) 400-410. [113] M. Calem, K. Bromis, P. McGuire, C. Morgan, M.J. Kempton, Meta-analysis of associations between childhood adversity and hippocampus and amygdala volume in non-clinical and general population samples, Neuroimage Clin 14 (2017) 471-479. [114] M.A. Munn, J. Alexopoulos, T. Nishino, C.M. Babb, L.A. Flake, T. Singer, J.T. Ratnanather, H. Huang, R.D. Todd, M.I. Miller, K.N. Botteron, Amygdala volume analysis in female twins with major depression, Biol Psychiatry 62(5) (2007) 415-422. [115] J. Keller, L. Shen, R.G. Gomez, A. Garrett, H.B. Solvason, A. Reiss, A.F. Schatzberg, Hippocampal and amygdalar volumes in psychotic and nonpsychotic unipolar depression, Am J Psychiatry 165(7) (2008) 872-880. [116] J. Sacher, J. Neumann, T. Fünfstück, A. Soliman, A. Villringer, M.L. Schroeter, Mapping the depressed brain: A meta-analysis of structural and functional alterations in major depressive disorder, J Affect Disord 140(2) (2012) 142-148. [117] A.M. Rich, Y.T. Cho, Y. Tang, A. Savic, J.H. Krystal, F. Wang, K. Xu, A. Anticevic, Amygdala volume is reduced in early course schizophrenia, Psychiatry Res Neuroimaging 250 (2016) 50-60. [118] A.E. Pinkham, P. Liu, H. Lu, M. Kriegsman, C. Simpson, C. Tamminga, Amygdala hyperactivity at rest in paranoid individuals with schizophrenia, Am J Psychiatry 172(8) (2015) 784-792. [119] J.-L. Yue, P. Li, L. Shi, X. Lin, H.-Q. Sun, L. Lu, Enhanced temporal variability of amygdala- frontal functional connectivity in patients with schizophrenia, Neuroimage Clin 18 (2018) 527-532. [120] X. Zhang, T.T. Ge, G. Yin, R. Cui, G. Zhao, W. Yang, Stress-induced functional alterations in amygdala: Implications for neuropsychiatric diseases, Front Neurosci 12 (2018) 367-367. [121] C.A. Stockmeier, G.J. Mahajan, L.C. Konick, J.C. Overholser, G.J. Jurjus, H.Y. Meltzer, H.B.M. Uylings, L. Friedman, G. Rajkowska, Cellular changes in the postmortem hippocampus in major depression, Biol Psychiatry 56(9) (2004) 640-650. [122] A.P. Mallya, A.Y. Deutch, (Micro)Glia as Effectors of Cortical Volume Loss in Schizophrenia, Schizophr Bull 44(5) (2018) 948-957. [123] O.P. Keifer, Jr., R.C. Hurt, D.A. Gutman, S.D. Keilholz, S.L. Gourley, K.J. Ressler, Voxel-based morphometry predicts shifts in dendritic spine density and morphology with auditory fear conditioning, Nature communications 6 (2015) 7582. [124] B. Ellenbroek, J. Youn, Rodent models in neuroscience research: is it a rat race?, Dis Models Mech 9(10) (2016) 1079-1087. [125] A. Gururajan, A. Reif, J.F. Cryan, D.A. Slattery, The future of rodent models in depression research, Nat Rev Neurosci 20(11) (2019) 686-701. [126] B. Vafadari, S. Mitra, M. Stefaniuk, L. Kaczmarek, Psychosocial Stress Induces Schizophrenia- Like Behavior in Mice With Reduced MMP-9 Activity, Front Behav Neurosci 13 (2019) 195.

50 [127] J.B. Becker, B.J. Prendergast, J.W.J.B.o.s.d. Liang, Female rats are not more variable than male rats: a meta-analysis of neuroscience studies, Biol Sex Differ 7(1) (2016) 1-7. [128] B.J. Prendergast, K.G. Onishi, I. Zucker, Female mice liberated for inclusion in neuroscience and biomedical research, Neurosci Biobehav Rev 40 (2014) 1-5. [129] C.L. Wellman, D.A. Bangasser, J.L. Bollinger, L. Coutellier, M.L. Logrip, K.M. Moench, K.R. Urban, Sex Differences in Risk and Resilience: Stress Effects on the Neural Substrates of Emotion and Motivation, The Journal of neuroscience : the official journal of the Society for Neuroscience 38(44) (2018) 9423-9432. [130] A. Diflorio, I. Jones, Is sex important? Gender differences in bipolar disorder, International review of psychiatry (Abingdon, England) 22(5) (2010) 437-452. [131] P.R. Albert, Why is depression more prevalent in women?, J Psychiatry Neurosci 40(4) (2015) 219- 221. [132] T.O. Sharpee, Toward functional classification of neuronal types, Neuron 83(6) (2014) 1329-1334. [133] B. Tasic, V. Menon, T.N. Nguyen, T.K. Kim, T. Jarsky, Z. Yao, B. Levi, L.T. Gray, S.A. Sorensen, T. Dolbeare, D. Bertagnolli, J. Goldy, N. Shapovalova, S. Parry, C. Lee, K. Smith, A. Bernard, L. Madisen, S.M. Sunkin, M. Hawrylycz, C. Koch, H. Zeng, Adult mouse cortical cell taxonomy revealed by single cell transcriptomics, Nat Neurosci 19(2) (2016) 335-346. [134] N. Spruston, Pyramidal neurons: dendritic structure and synaptic integration, Nat Rev Neurosci 9(3) (2008) 206-221. [135] J.J. Radley, H.M. Sisti, J. Hao, A.B. Rocher, T. McCall, P.R. Hof, B.S. McEwen, J.H. Morrison, Chronic behavioral stress induces apical dendritic reorganization in pyramidal neurons of the medial prefrontal cortex, Neuroscience 125(1) (2004) 1-6. [136] J.J. Radley, A.B. Rocher, M. Miller, W.G.M. Janssen, C. Liston, P.R. Hof, B.S. McEwen, J.H. Morrison, Repeated stress induces dendritic spine loss in the rat medial prefrontal cortex, Cerebral cortex (New York, N.Y. : 1991) 16(3) (2005) 313-320. [137] C. Liston, M.M. Miller, D.S. Goldwater, J.J. Radley, A.B. Rocher, P.R. Hof, J.H. Morrison, B.S. McEwen, Stress-induced alterations in prefrontal cortical dendritic morphology predict selective impairments in perceptual attentional set-shifting, The Journal of neuroscience : the official journal of the Society for Neuroscience 26(30) (2006) 7870-7874. [138] K.L. Brunson, E. Kramar, B. Lin, Y. Chen, L.L. Colgin, T.K. Yanagihara, G. Lynch, T.Z. Baram, Mechanisms of late-onset cognitive decline after early-life stress, The Journal of neuroscience : the official journal of the Society for Neuroscience 25(41) (2005) 9328-9338. [139] C. Helmeke, K. Seidel, G. Poeggel, T.W. Bredy, A. Abraham, K. Braun, Paternal deprivation during infancy results in dendrite- and time-specific changes of dendritic development and spine formation in the orbitofrontal cortex of the biparental rodent Octodon degus, Neuroscience 163(3) (2009) 790-798. [140] A.M. Magariños, B.S. McEwen, G. Flügge, E. Fuchs, Chronic psychosocial stress causes apical dendritic atrophy of hippocampal CA3 pyramidal neurons in subordinate tree shrews, The Journal of neuroscience : the official journal of the Society for Neuroscience 16(10) (1996) 3534-3540. [141] K. McLaughlin, S. Baran, R. Wright, C. Conrad, Chronic stress enhances spatial memory in ovariectomized female rats despite CA3 dendritic retraction: possible involvement of CA1 neurons, Neuroscience 135(4) (2005) 1045-1054. [142] Y. Watanabe, E. Gould, B.S. McEwen, Stress induces atrophy of apical dendrites of hippocampal CA3 pyramidal neurons, Brain research 588(2) (1992) 341-345. [143] K.M. Christian, A.D. Miracle, C.L. Wellman, K. Nakazawa, Chronic stress-induced hippocampal dendritic retraction requires CA3 NMDA receptors, Neuroscience 174 (2011) 26-36. [144] C.R. McKittrick, A.M. Magariños, D.C. Blanchard, R.J. Blanchard, B.S. McEwen, R.R. Sakai, Chronic social stress reduces dendritic arbors in CA3 of hippocampus and decreases binding to serotonin transporter sites, 36(2) (2000) 85-94. [145] C.L. Wellman, Dendritic reorganization in pyramidal neurons in medial prefrontal cortex after chronic corticosterone administration, J Neurobiol 49(3) (2001) 245-253. [146] S.L. Gourley, A.M. Swanson, A.J. Koleske, Corticosteroid-induced neural remodeling predicts behavioral vulnerability and resilience, The Journal of neuroscience : the official journal of the Society for Neuroscience 33(7) (2013) 3107-3112.

51 [147] C.S. Woolley, E. Gould, B.S. McEwen, Exposure to excess glucocorticoids alters dendritic morphology of adult hippocampal pyramidal neurons, Brain research 531(1-2) (1990) 225-231. [148] S.J. Lupien, B.S. McEwen, M.R. Gunnar, C. Heim, Effects of stress throughout the lifespan on the brain, behaviour and cognition, Nature reviews neuroscience 10(6) (2009) 434-445. [149] J.E. Reser, Chronic stress, cortical plasticity and neuroecology, Behav Processes 129 (2016) 105- 115. [150] I. Negrón-Oyarzo, F. Aboitiz, P. Fuentealba, Impaired Functional Connectivity in the Prefrontal Cortex: A Mechanism for Chronic Stress-Induced Neuropsychiatric Disorders, Neural Plast 2016 (2016) 7539065. [151] S.C. Cook, C.L. Wellman, Chronic stress alters dendritic morphology in rat medial prefrontal cortex, J Neurobiol 60(2) (2004) 236-248. [152] L. Petreanu, T. Mao, S.M. Sternson, K. Svoboda, The subcellular organization of neocortical excitatory connections, Nature 457(7233) (2009) 1142-1145. [153] T. Vu, R. Gugustea, L.S. Leung, Long-term potentiation of the nucleus reuniens and entorhinal cortex to CA1 distal dendritic synapses in mice, Brain Struct Funct 225(6) (2020) 1817-1838. [154] J.J. Radley, P.E. Sawchenko, A common substrate for prefrontal and hippocampal inhibition of the neuroendocrine stress response, The Journal of neuroscience : the official journal of the Society for Neuroscience 31(26) (2011) 9683-9695. [155] M.S. Kassem, J. Lagopoulos, T. Stait-Gardner, W.S. Price, T.W. Chohan, J.C. Arnold, S.N. Hatton, M.R. Bennett, Stress-induced grey matter loss determined by MRI is primarily due to loss of dendrites and their synapses, Molecular neurobiology 47(2) (2013) 645-661. [156] C.D. Conrad, J.E. LeDoux, A.M. Magariños, B.S. McEwen, Repeated restraint stress facilitates fear conditioning independently of causing hippocampal CA3 dendritic atrophy, Behav Neurosci 113(5) (1999) 902-913. [157] L. Eiland, B.S. McEwen, Early life stress followed by subsequent adult chronic stress potentiates anxiety and blunts hippocampal structural remodeling, Hippocampus 22(1) (2012) 82-91. [158] M.K. Seo, N.N. Ly, C.H. Lee, H.Y. Cho, C.M. Choi, L.H. Nhu, J.G. Lee, B.J. Lee, G.-M. Kim, B.J. Yoon, S.W. Park, Y.H. Kim, Early life stress increases stress vulnerability through BDNF gene epigenetic changes in the rat hippocampus, Neuropharmacology 105 (2016) 388-397. [159] K.A. McLaughlin, M.A. Sheridan, A.L. Gold, A. Duys, H.K. Lambert, M. Peverill, C. Heleniak, T. Shechner, Z. Wojcieszak, D.S. Pine, Maltreatment Exposure, Brain Structure, and Fear Conditioning in Children and Adolescents, Neuropsychopharmacology 41(8) (2016) 1956-1964. [160] N.P. Daskalakis, R.C. Bagot, K.J. Parker, C.H. Vinkers, E.R. de Kloet, The three-hit concept of vulnerability and resilience: toward understanding adaptation to early-life adversity outcome, Psychoneuroendocrinology 38(9) (2013) 1858-1873. [161] E.B. Bloss, W.G. Janssen, D.T. Ohm, F.J. Yuk, S. Wadsworth, K.M. Saardi, B.S. McEwen, J.H. Morrison, Evidence for reduced experience-dependent dendritic spine plasticity in the aging prefrontal cortex, The Journal of neuroscience : the official journal of the Society for Neuroscience 31(21) (2011) 7831-7839. [162] E.S. Rosenzweig, C.A. Barnes, Impact of aging on hippocampal function: plasticity, network dynamics, and cognition, Prog Neurobiol 69(3) (2003) 143-179. [163] E.S. Wohleb, R. Terwilliger, C.H. Duman, R.S. Duman, Stress-Induced Neuronal Colony Stimulating Factor 1 Provokes Microglia-Mediated Neuronal Remodeling and Depressive-like Behavior, Biol Psychiatry 83(1) (2018) 38-49. [164] H. Qiao, M.-X. Li, C. Xu, H.-B. Chen, S.-C. An, X.-M. Ma, Dendritic spines in depression: what we learned from animal models, Neural Plast 2016 (2016) 8056370. [165] A. Suvrathan, S. Bennur, S. Ghosh, A. Tomar, S. Anilkumar, S. Chattarji, Stress enhances fear by forming new synapses with greater capacity for long-term potentiation in the amygdala, Philosophical transactions of the Royal Society of London. Series B, Biological sciences 369(1633) (2014) 20130151. [166] C.A. Grillo, M. Risher, V.A. Macht, A.L. Bumgardner, A. Hang, C. Gabriel, E. Mocaër, G.G. Piroli, J.R. Fadel, L.P. Reagan, Repeated restraint stress-induced atrophy of glutamatergic pyramidal neurons and decreases in glutamatergic efflux in the rat amygdala are prevented by the antidepressant agomelatine, Neuroscience 284 (2015) 430-443.

52 [167] M. Maroun, P.J. Ioannides, K.L. Bergman, A. Kavushansky, A. Holmes, C.L. Wellman, Fear extinction deficits following acute stress associate with increased spine density and dendritic retraction in basolateral amygdala neurons, Eur J Neurosci 38(4) (2013) 2611-2620. [168] R. Mitra, R.M. Sapolsky, Acute corticosterone treatment is sufficient to induce anxiety and amygdaloid dendritic hypertrophy, Proc Natl Acad Sci USA 105(14) (2008) 5573-5578. [169] H. Lakshminarasimhan, S. Chattarji, Stress leads to contrasting effects on the levels of brain derived neurotrophic factor in the hippocampus and amygdala, PLoS One 7(1) (2012) e30481. [170] A. Vyas, A. Pillai, S. Chattarji, Recovery after chronic stress fails to reverse amygdaloid neuronal hypertrophy and enhanced anxiety-like behavior, Neuroscience 128(4) (2004) 667-673. [171] A.S. Koe, A. Ashokan, R. Mitra, Short environmental enrichment in adulthood reverses anxiety and basolateral amygdala hypertrophy induced by maternal separation, Transl Psychiatry 6(2) (2016) e729. [172] J. Herms, M.M. Dorostkar, Dendritic spine pathology in neurodegenerative diseases, Annu Rev Pathol 11 (2016) 221-250. [173] E.A. Nimchinsky, B.L. Sabatini, K. Svoboda, Structure and function of dendritic spines, Annual review of physiology 64 (2002) 313-353. [174] C. Yang, Y. Shirayama, J.-c. Zhang, Q. Ren, K. Hashimoto, Regional differences in brain-derived neurotrophic factor levels and dendritic spine density confer resilience to inescapable stress, Int J Neuropsychopharmacol 18(7) (2015) 121. [175] R.N. Moda-Sava, M.H. Murdock, P.K. Parekh, R.N. Fetcho, B.S. Huang, T.N. Huynh, J. Witztum, D.C. Shaver, D.L. Rosenthal, E.J. Alway, K. Lopez, Y. Meng, L. Nellissen, L. Grosenick, T.A. Milner, K. Deisseroth, H. Bito, H. Kasai, C. Liston, Sustained rescue of prefrontal circuit dysfunction by antidepressant-induced spine formation, Science 364(6436) (2019) eaat8078. [176] D.S. Goldwater, C. Pavlides, R.G. Hunter, E.B. Bloss, P.R. Hof, B.S. McEwen, J.H. Morrison, Structural and functional alterations to rat medial prefrontal cortex following chronic restraint stress and recovery, Neuroscience 164(2) (2009) 798-808. [177] R.M. Anderson, R.M. Glanz, S.B. Johnson, M.M. Miller, S.A. Romig-Martin, J.J. Radley, Prolonged corticosterone exposure induces dendritic spine remodeling and attrition in the rat medial prefrontal cortex, J Comp Neurol 524(18) (2016) 3729-3746. [178] D. Orlowski, B. Elfving, H.K. Müller, G. Wegener, C.R. Bjarkam, Wistar rats subjected to chronic restraint stress display increased hippocampal spine density paralleled by increased expression levels of synaptic scaffolding proteins, Stress (Amsterdam, Netherlands) 15(5) (2012) 514-523. [179] M.G. Stewart, H.A. Davies, C. Sandi, I.V. Kraev, V.V. Rogachevsky, C.J. Peddie, J.J. Rodriguez, M.I. Cordero, H.S. Donohue, P.L. Gabbott, Stress suppresses and learning induces plasticity in CA3 of rat hippocampus: a three-dimensional ultrastructural study of thorny excrescences and their postsynaptic densities, Neuroscience 131(1) (2005) 43-54. [180] A. Magarinos, C. Li, J. Gal Toth, K. Bath, D. Jing, F. Lee, B. McEwen, Effect of brain‐ derived neurotrophic factor haploinsufficiency on stress‐ induced remodeling of hippocampal neurons, Hippocampus 21(3) (2011) 253-264. [181] C. Sandi, H.A. Davies, M.I. Cordero, J.J. Rodriguez, V.I. Popov, M.G. Stewart, Rapid reversal of stress induced loss of synapses in CA3 of rat hippocampus following water maze training, Eur J Neurosci 17(11) (2003) 2447-2456. [182] M. Popoli, Z. Yan, B.S. McEwen, G. Sanacora, The stressed synapse: the impact of stress and glucocorticoids on glutamate transmission, Nat Rev Neurosci 13(1) (2011) 22-37. [183] M. Qin, Z. Xia, T. Huang, C.B. Smith, Effects of chronic immobilization stress on anxiety-like behavior and basolateral amygdala morphology in Fmr1 knockout mice, Neuroscience 194 (2011) 282- 290. [184] M.N. Hill, S.A. Kumar, S.B. Filipski, M. Iverson, K.L. Stuhr, J.M. Keith, B.F. Cravatt, C.J. Hillard, S. Chattarji, B.S. McEwen, Disruption of fatty acid amide hydrolase activity prevents the effects of chronic stress on anxiety and amygdalar microstructure, Mol Psychiatry 18(10) (2013) 1125-1135. [185] A. Vyas, S. Jadhav, S. Chattarji, Prolonged behavioral stress enhances synaptic connectivity in the basolateral amygdala, Neuroscience 143(2) (2006) 387-393. [186] R. Mitra, S. Jadhav, B.S. McEwen, A. Vyas, S. Chattarji, Stress duration modulates the spatiotemporal patterns of spine formation in the basolateral amygdala, Proc Natl Acad Sci USA 102(26) (2005) 9371-9376.

53 [187] D. Patel, S. Anilkumar, S. Chattarji, B. Buwalda, Repeated social stress leads to contrasting patterns of structural plasticity in the amygdala and hippocampus, Behavioural brain research 347 (2018) 314-324. [188] H.-H. Zhang, S.-Q. Meng, X.-Y. Guo, J.-L. Zhang, W. Zhang, Y.-Y. Chen, L. Lu, J.-L. Yang, Y.-X. Xue, Traumatic stress produces delayed alterations of synaptic plasticity in basolateral amygdala, Front Psychol 10 (2019) 2394-2394. [189] X. Zhang, T.T. Ge, G. Yin, R. Cui, G. Zhao, W. Yang, Stress-Induced Functional Alterations in Amygdala: Implications for Neuropsychiatric Diseases, Frontiers in Neuroscience 12 (2018). [190] C.M. Schumann, M.D. Bauman, D.G. Amaral, Abnormal structure or function of the amygdala is a common component of neurodevelopmental disorders, Neuropsychologia 49(4) (2011) 745-759. [191] E. Monroy, E. Hernández-Torres, G. Flores, Maternal separation disrupts dendritic morphology of neurons in prefrontal cortex, hippocampus, and in male rat offspring, Journal of chemical neuroanatomy 40(2) (2010) 93-101. [192] S.D. Iñiguez, A. Aubry, L.M. Riggs, J.B. Alipio, R.M. Zanca, F.J. Flores-Ramirez, M.A. Hernandez, S.J. Nieto, D. Musheyev, P.A. Serrano, Social defeat stress induces depression-like behavior and alters spine morphology in the hippocampus of adolescent male C57BL/6 mice, Neurobiol Stress 5 (2016) 54-64. [193] C. Pinzón-Parra, B. Vidal-Jiménez, I. Camacho-Abrego, A.A. Flores-Gómez, A. Rodríguez- Moreno, G. Flores, Juvenile stress causes reduced locomotor behavior and dendritic spine density in the prefrontal cortex and basolateral amygdala in Sprague-Dawley rats, Synapse 73(1) (2019) e22066. [194] S.-F. Tsai, T.-Y. Huang, C.-Y. Chang, Y.-C. Hsu, S.-J. Chen, L. Yu, Y.-M. Kuo, C.J. Jen, Social instability stress differentially affects amygdalar neuron adaptations and memory performance in adolescent and adult rats, Front Behav Neurosci 8 (2014) 27. [195] E.Y. Yuen, J. Wei, W. Liu, P. Zhong, X. Li, Z. Yan, Repeated stress causes cognitive impairment by suppressing glutamate receptor expression and function in prefrontal cortex, Neuron 73(5) (2012) 962- 977. [196] J. Wei, E.Y. Yuen, W. Liu, X. Li, P. Zhong, I.N. Karatsoreos, B.S. McEwen, Z. Yan, Estrogen protects against the detrimental effects of repeated stress on glutamatergic transmission and cognition, Mol Psychiatry 19(5) (2014) 588-598. [197] N. Nava, G. Treccani, N. Liebenberg, F. Chen, M. Popoli, G. Wegener, J.R. Nyengaard, Chronic desipramine prevents acute stress-induced reorganization of medial prefrontal cortex architecture by blocking glutamate vesicle accumulation and increase, Int J Neuropsychopharmacol 18(3) (2014) pyu085. [198] M.P. Forrest, E. Parnell, P. Penzes, Dendritic structural plasticity and neuropsychiatric disease, Nat Rev Neurosci 19(4) (2018) 215-234. [199] C.H. Duman, R.S. Duman, Spine synapse remodeling in the pathophysiology and treatment of depression, Neuroscience letters 601 (2015) 20-29. [200] W. Liu, T. Ge, Y. Leng, Z. Pan, J. Fan, W. Yang, R. Cui, The role of neural plasticity in depression: from hippocampus to prefrontal cortex, Neural Plast 2017 (2017) 6871089. [201] J.R. Glausier, D.A. Lewis, Dendritic spine pathology in schizophrenia, Neuroscience 251 (2013) 90-107. [202] A. Berdenis van Berlekom, C.H. Muflihah, G.J.L.J. Snijders, H.D. MacGillavry, J. Middeldorp, E.M. Hol, R.S. Kahn, L.D. de Witte, Synapse pathology in schizophrenia: A meta-analysis of postsynaptic elements in postmortem brain studies, Schizophr Bull 46(2) (2019) 374-386. [203] L.A. Glantz, D.A. Lewis, Decreased dendritic spine density on prefrontal cortical pyramidal neurons in schizophrenia, Archives of general psychiatry 57(1) (2000) 65-73. [204] G.T. Konopaske, N. Lange, J.T. Coyle, F.M. Benes, Prefrontal cortical dendritic spine pathology in schizophrenia and bipolar disorder, JAMA psychiatry 71(12) (2014) 1323-1331. [205] H.J. Kang, B. Voleti, T. Hajszan, G. Rajkowska, C.A. Stockmeier, P. Licznerski, A. Lepack, M.S. Majik, L.S. Jeong, M. Banasr, H. Son, R.S. Duman, Decreased expression of synapse-related genes and loss of synapses in major depressive disorder, Nat Med 18(9) (2012) 1413-1417. [206] M. Koenigs, J. Grafman, The functional neuroanatomy of depression: Distinct roles for ventromedial and dorsolateral prefrontal cortex, Behavioural brain research 201(2) (2009) 239-243. [207] A.P. Association, Diagnostic and statistical manual of mental disorders (DSM-5®), American Psychiatric Pub2013.

54 [208] M.D. Underwood, M.J. Bakalian, V.L. Johnson, S.A. Kassir, S.P. Ellis, J.J. Mann, V. Arango, Less NMDA Receptor Binding in Dorsolateral Prefrontal Cortex and Anterior Cingulate Cortex Associated With Reported Early-Life Adversity but Not Suicide, Int J Neuropsychopharmacol 23(5) (2020) 311-318. [209] K.A. Young, P.M. Thompson, D.A. Cruz, D.E. Williamson, L.D. Selemon, BA11 FKBP5 expression levels correlate with dendritic spine density in postmortem PTSD and controls, Neurobiol Stress 2 (2015) 67-72. [210] J.D. Flory, R. Yehuda, Comorbidity between post-traumatic stress disorder and major depressive disorder: alternative explanations and treatment considerations, Dialogues Clin Neurosci 17(2) (2015) 141-150. [211] M.L. O’Donnell, M. Creamer, P. Pattison, Posttraumatic Stress Disorder and Depression Following Trauma: Understanding Comorbidity, Am J Psychiatry 161(8) (2004) 1390-1396. [212] A. Soetanto, R.S. Wilson, K. Talbot, A. Un, J.A. Schneider, M. Sobiesk, J. Kelly, S. Leurgans, D.A. Bennett, S.E. Arnold, Association of Anxiety and Depression With -Associated Protein 2– and Synaptopodin-Immunolabeled Dendrite and Spine Densities in Hippocampal CA3 of Older Humans, Archives of general psychiatry 67(5) (2010) 448-457. [213] D. Kaul, C.C. Smith, J. Stevens, A.S. Fröhlich, E.B. Binder, N. Mechawar, S.G. Schwab, N. Matosin, Severe childhood and adulthood stress associates with neocortical layer-specific reductions of mature spines in psychiatric disorders, Neurobiology of Stress 13 (2020) 100270. [214] C.E. Page, L. Coutellier, Prefrontal excitatory/inhibitory balance in stress and emotional disorders: Evidence for over-inhibition, Neurosci Biobehav Rev 105 (2019) 39-51. [215] B. Clancy, T.J. Teague-Ross, R. Nagarajan, Cross-species analyses of the cortical GABAergic and subplate neural populations, Front Neuroanat 3 (2009) 20-20. [216] J. DeFelipe, Chapter 17 Cortical interneurons: from Cajal to 2001, Prog Brain Res, Elsevier2002, pp. 215-238. [217] B.R. Ferguson, W.-J. Gao, PV interneurons: critical regulators of E/I balance for prefrontal cortex- dependent behavior and psychiatric disorders, Front Neural Circuits 12 (2018) 37-37. [218] O.K. Swanson, A. Maffei, From Hiring to Firing: Activation of Inhibitory Neurons and Their Recruitment in Behavior, Front Mol Neurosci 12 (2019) 168-168. [219] Z.J. Huang, A. Paul, The diversity of GABAergic neurons and neural communication elements, Nat Rev Neurosci 20(9) (2019) 563-572. [220] H. Markram, M. Toledo-Rodriguez, Y. Wang, A. Gupta, G. Silberberg, C. Wu, Interneurons of the neocortical inhibitory system, Nat Rev Neurosci 5(10) (2004) 793-807. [221] B. Rudy, G. Fishell, S. Lee, J. Hjerling-Leffler, Three groups of interneurons account for nearly 100% of neocortical GABAergic neurons, Dev Neurobiol 71(1) (2011) 45-61. [222] L. Hertäg, H. Sprekeler, Amplifying the redistribution of somato-dendritic inhibition by the interplay of three interneuron types, PLOS Comput Biol 15(5) (2019) e1006999. [223] H. Hu, J. Gan, P. Jonas, Fast-spiking, parvalbumin+ GABAergic interneurons: From cellular design to microcircuit function, Science 345(6196) (2014) 1255263. [224] C.K. Pfeffer, M. Xue, M. He, Z.J. Huang, M. Scanziani, Inhibition of inhibition in visual cortex: the logic of connections between molecularly distinct interneurons, Nat Neurosci 16(8) (2013) 1068-1076. [225] A.J. McDonald, F. Mascagni, Colocalization of calcium-binding proteins and GABA in neurons of the rat basolateral amygdala, Neuroscience 105(3) (2001) 681-693. [226] P. Veeraiah, J.M. Noronha, S. Maitra, P. Bagga, N. Khandelwal, S. Chakravarty, A. Kumar, A.B. Patel, Dysfunctional glutamatergic and γ-aminobutyric acidergic activities in prefrontal cortex of mice in social defeat model of depression, Biol Psychiatry 76(3) (2014) 231-238. [227] B. Czéh, I. Vardya, Z. Varga, F. Febbraro, D. Csabai, L.S. Martis, K. Højgaard, K. Henningsen, E.V. Bouzinova, A. Miseta, K. Jensen, O. Wiborg, Long-Term Stress Disrupts the Structural and Functional Integrity of GABAergic Neuronal Networks in the Medial Prefrontal Cortex of Rats, Front Cell Neurosci 12 (2018) 148. [228] S. Ghosal, B.D. Hare, R.S. Duman, Prefrontal cortex GABAergic deficits and circuit dysfunction in the pathophysiology and treatment of chronic stress and depression, Curr Opin Behav Sci 14 (2017) 1-8. [229] M. Selten, H. van Bokhoven, N. Nadif Kasri, Inhibitory control of the excitatory/inhibitory balance in psychiatric disorders, F1000Res 7 (2018) 23. [230] V.S. Sohal, J.L.R. Rubenstein, Excitation-inhibition balance as a framework for investigating mechanisms in neuropsychiatric disorders, Mol Psychiatry 24(9) (2019) 1248-1257.

55 [231] M.V. Fogaça, R.S. Duman, Cortical GABAergic Dysfunction in Stress and Depression: New Insights for Therapeutic Interventions, Front Cell Neurosci 13 (2019) 87. [232] K.K.A. Cho, R. Hoch, A.T. Lee, T. Patel, J.L.R. Rubenstein, V.S. Sohal, Gamma rhythms link prefrontal interneuron dysfunction with cognitive inflexibility in Dlx5/6(+/-) mice, Neuron 85(6) (2015) 1332-1343. [233] Z. Perova, K. Delevich, B. Li, Depression of excitatory synapses onto parvalbumin interneurons in the medial prefrontal cortex in susceptibility to stress, The Journal of neuroscience : the official journal of the Society for Neuroscience 35(7) (2015) 3201-3206. [234] R. Shepard, C.E. Page, L. Coutellier, Sensitivity of the prefrontal GABAergic system to chronic stress in male and female mice: Relevance for sex differences in stress-related disorders, Neuroscience 332 (2016) 1-12. [235] C.E. Page, R. Shepard, K. Heslin, L. Coutellier, Prefrontal parvalbumin cells are sensitive to stress and mediate anxiety-related behaviors in female mice, Sci Rep 9(1) (2019) 19772. [236] N. Todorović, B. Mićić, M. Schwirtlich, M. Stevanović, D. Filipović, Subregion-specific Protective Effects of Fluoxetine and Clozapine on Parvalbumin Expression in Medial Prefrontal Cortex of Chronically Isolated Rats, Neuroscience 396 (2019) 24-35. [237] B. Czeh, M. Simon, M.G.C. van der Hart, B. Schmelting, M.B. Hesselink, E. Fuchs, Chronic stress decreases the number of parvalbumin-immunoreactive interneurons in the hippocampus: Prevention by treatment with a substance P receptor (NK1) antagonist, Neuropsychopharmacology 30(1) (2005) 67-79. [238] A.C. Rossetti, M.S. Paladini, M. Colombo, P. Gruca, M. Lason-Tyburkiewicz, K. Tota-Glowczyk, M. Papp, M.A. Riva, R. Molteni, Chronic Stress Exposure Reduces Parvalbumin Expression in the Rat Hippocampus through an Imbalance of Redox Mechanisms: Restorative Effect of the Antipsychotic Lurasidone, Int J Neuropsychopharmacol 21(9) (2018) 883-893. [239] B. Czéh, Z.K. Varga, K. Henningsen, G.L. Kovács, A. Miseta, O. Wiborg, Chronic stress reduces the number of GABAergic interneurons in the adult rat hippocampus, dorsal-ventral and region-specific differences, Hippocampus 25(3) (2015) 393-405. [240] W. Hu, M. Zhang, B. Czéh, G. Flügge, W. Zhang, Stress impairs GABAergic network function in the hippocampus by activating nongenomic glucocorticoid receptors and affecting the integrity of the parvalbumin-expressing neuronal network, Neuropsychopharmacology 35(8) (2010) 1693-1707. [241] F. Jie, G. Yin, W. Yang, M. Yang, S. Gao, J. Lv, B. Li, Stress in regulation of GABA amygdala system and relevance to neuropsychiatric diseases, Front Neurosci 12 (2018) 562-562. [242] E. Castillo-Gómez, M. Pérez-Rando, M. Bellés, J. Gilabert-Juan, J.V. Llorens, H. Carceller, C. Bueno-Fernández, C. García-Mompó, B. Ripoll-Martínez, Y. Curto, N. Sebastiá-Ortega, M.D. Moltó, J. Sanjuan, J. Nacher, Early Social Isolation Stress and Perinatal NMDA Receptor Antagonist Treatment Induce Changes in the Structure and Neurochemistry of Inhibitory Neurons of the Adult Amygdala and Prefrontal Cortex, eNeuro 4(2) (2017) 0034. [243] K.G. Bath, G. Manzano-Nieves, H. Goodwill, Early life stress accelerates behavioral and neural maturation of the hippocampus in male mice, Horm Behav 82 (2016) 64-71. [244] G. Manzano Nieves, M. Bravo, S. Baskoylu, K.G. Bath, Early life adversity decreases pre- adolescent fear expression by accelerating amygdala PV cell development, eLife 9 (2020) e55263. [245] L.O. Franco, M.J. Carvalho, J. Costa, P.A. Ferreira, J.R. Guedes, R. Sousa, M. Edfawy, C.M. Seabra, A.L. Cardoso, J. Peça, Social subordination induced by early life adversity rewires inhibitory control of the prefrontal cortex via enhanced Npy1r signaling, Neuropsychopharmacology 45(9) (2020) 1438-1447. [246] H.C. Brenhouse, S.L. Andersen, Nonsteroidal Anti-Inflammatory Treatment Prevents Delayed Effects of Early Life Stress in Rats, Biol Psychiatry 70(5) (2011) 434-440. [247] C.H. do Prado, T. Narahari, F.H. Holland, H.N. Lee, S.K. Murthy, H.C. Brenhouse, Effects of early adolescent environmental enrichment on cognitive dysfunction, prefrontal cortex development, and inflammatory cytokines after early life stress, Developmental psychobiology 58(4) (2016) 482-491. [248] F.H. Holland, P. Ganguly, D.N. Potter, E.H. Chartoff, H.C. Brenhouse, Early life stress disrupts social behavior and prefrontal cortex parvalbumin interneurons at an earlier time-point in females than in males, Neuroscience letters 566 (2014) 131-136. [249] C.C. Chen, J. Lu, R. Yang, J.B. Ding, Y. Zuo, Selective activation of parvalbumin interneurons prevents stress-induced synapse loss and perceptual defects, Mol Psychiatry 23(7) (2018) 1614-1625.

56 [250] D.A. Lewis, A.A. Curley, J.R. Glausier, D.W. Volk, Cortical parvalbumin interneurons and cognitive dysfunction in schizophrenia, Trends Neurosci 35(1) (2012) 57-67. [251] G. Rajkowska, G. O'Dwyer, Z. Teleki, C.A. Stockmeier, J.J. Miguel-Hidalgo, GABAergic neurons immunoreactive for calcium binding proteins are reduced in the prefrontal cortex in major depression, Neuropsychopharmacology 32(2) (2007) 471-482. [252] E. Sibille, H.M. Morris, R.S. Kota, D.A. Lewis, GABA-related transcripts in the dorsolateral prefrontal cortex in mood disorders, Int J Neuropsychopharmacol 14(6) (2011) 721-734. [253] C.L. Beasley, G.P. Reynolds, Parvalbumin-immunoreactive neurons are reduced in the prefrontal cortex of schizophrenics, Schizophrenia research 24(3) (1997) 349-355. [254] S. Akbarian, H.-S. Huang, Molecular and cellular mechanisms of altered GAD1/GAD67 expression in schizophrenia and related disorders, Brain Res Rev 52(2) (2006) 293-304. [255] B. Karolewicz, D. Maciag, G. O'Dwyer, C.A. Stockmeier, A.M. Feyissa, G. Rajkowska, Reduced level of glutamic acid decarboxylase-67 kDa in the prefrontal cortex in major depression, Int J Neuropsychopharmacol 13(04) (2010) 411. [256] Z. Zhang, G. Reynolds, A selective deficit in the relative density of parvalbumin-immunoreactive neurons in the hippocampus in schizophrenia, Schizophrenia research, ELSEVIER SCIENCE BV PO BOX 211, 1000 AE AMSTERDAM, NETHERLANDS, 2001, pp. 65-65. [257] C. Konradi, C.K. Yang, E.I. Zimmerman, K.M. Lohmann, P. Gresch, H. Pantazopoulos, S. Berretta, S. Heckers, Hippocampal interneurons are abnormal in schizophrenia, Schizophrenia research 131(1-3) (2011) 165-173. [258] S. Herculano-Houzel, The glia/neuron ratio: how it varies uniformly across brain structures and species and what that means for brain physiology and evolution, Glia 62(9) (2014) 1377-1391. [259] A. Verkhratsky, R. Zorec, V. Parpura, Stratification of astrocytes in healthy and diseased brain, Brain Pathol 27(5) (2017) 629-644. [260] N.A. Oberheim, T. Takano, X. Han, W. He, J.H. Lin, F. Wang, Q. Xu, J.D. Wyatt, W. Pilcher, J.G. Ojemann, Uniquely hominid features of adult human astrocytes, The Journal of neuroscience : the official journal of the Society for Neuroscience 29(10) (2009) 3276-3287. [261] A.Y.-S. Huang, J. Woo, D. Sardar, B. Lozzi, N.A. Bosquez Huerta, C.-C.J. Lin, D. Felice, A. Jain, A. Paulucci-Holthauzen, B. Deneen, Region-Specific Transcriptional Control of Astrocyte Function Oversees Local Circuit Activities, Neuron 106(6) (2020) 992-1008.e9. [262] X. Hu, S. Qin, X. Huang, Y. Yuan, Z. Tan, Y. Gu, X. Cheng, D. Wang, X.F. Lian, C. He, Z. Su, Region-Restrict Astrocytes Exhibit Heterogeneous Susceptibility to Neuronal Reprogramming, Stem Cell Rep 12(2) (2019) 290-304. [263] S. Götz, A. Bribian, L. López‐ Mascaraque, M. Götz, B. Grothe, L. Kunz, Heterogeneity of astrocytes: Electrophysiological properties of juxtavascular astrocytes before and after brain injury [published online August 18, 2020], Glia (2020) glia.23900. [264] M.Y. Batiuk, A. Martirosyan, J. Wahis, F. de Vin, C. Marneffe, C. Kusserow, J. Koeppen, J.F. Viana, J.F. Oliveira, T. Voet, C.P. Ponting, T.G. Belgard, M.G. Holt, Identification of region-specific astrocyte subtypes at single cell resolution, Nature communications 11(1) (2020) 1220. [265] Y.E. Wu, L. Pan, Y. Zuo, X. Li, W. Hong, Detecting Activated Cell Populations Using Single-Cell RNA-Seq, Neuron 96(2) (2017) 313-329.e6. [266] D. Keller, C. Erö, H. Markram, Cell Densities in the Mouse Brain: A Systematic Review, Front Neuroanat 12 (2018) 83. [267] F. Vasile, E. Dossi, N. Rouach, Human astrocytes: structure and functions in the healthy brain, Brain Struct Funct 222(5) (2017) 2017-2029. [268] S.G. Torres-Platas, C. Nagy, M. Wakid, G. Turecki, N. Mechawar, Glial fibrillary acidic protein is differentially expressed across cortical and subcortical regions in healthy brains and downregulated in the thalamus and caudate nucleus of depressed suicides, Mol Psychiatry 21(4) (2016) 509-515. [269] M.J. Webster, M.B. Knable, N. Johnston-Wilson, K. Nagata, M. Inagaki, R.H. Yolken, Immunohistochemical Localization of Phosphorylated Glial Fibrillary Acidic Protein in the Prefrontal Cortex and Hippocampus from Patients with Schizophrenia, Bipolar Disorder, and Depression, Brain Behav Immun 15(4) (2001) 388-400. [270] L.A. O’Leary, M.A. Davoli, C. Belliveau, A. Tanti, J.C. Ma, W.T. Farmer, G. Turecki, K.K. Murai, N. Mechawar, Characterization of vimentin-immunoreactive astrocytes in the human brain, Front Neuroanat 14 (2020) 2020.00031.

57 [271] Z. Zhang, Z. Ma, W. Zou, H. Guo, M. Liu, Y. Ma, L. Zhang, The Appropriate Marker for Astrocytes: Comparing the Distribution and Expression of Three Astrocytic Markers in Different Mouse Cerebral Regions, BioMed Res Int 2019 (2019) 9605265. [272] H.K. Kimelberg, The problem of astrocyte identity, Neurochemistry International 45(2-3) (2004) 191-202. [273] L.A. O’Leary, M.A. Davoli, C. Belliveau, A. Tanti, J.C. Ma, W.T. Farmer, G. Turecki, K.K. Murai, N. Mechawar, Characterization of vimentin-immunoreactive astrocytes in the human brain, Front Neuroanat 14 (2020) 31. [274] X. Hu, S. Qin, X. Huang, Y. Yuan, Z. Tan, Y. Gu, X. Cheng, D. Wang, X.-F. Lian, C. He, Region- restrict astrocytes exhibit heterogeneous susceptibility to neuronal reprogramming, Stem Cell Rep 12(2) (2019) 290-304. [275] W. Sun, A. Cornwell, J. Li, S. Peng, M.J. Osorio, N. Aalling, S. Wang, A. Benraiss, N. Lou, S.A. Goldman, M. Nedergaard, SOX9 Is an Astrocyte-Specific Nuclear Marker in the Adult Brain Outside the Neurogenic Regions, The Journal of neuroscience : the official journal of the Society for Neuroscience 37(17) (2017) 4493-4507. [276] Y. Zhang, S.A. Sloan, L.E. Clarke, C. Caneda, C.A. Plaza, P.D. Blumenthal, H. Vogel, G.K. Steinberg, M.S. Edwards, G. Li, Purification and characterization of progenitor and mature human astrocytes reveals transcriptional and functional differences with mouse, Neuron 89(1) (2016) 37-53. [277] J. Li, L. Pan, M.I. Godoy, W.G. Pembroke, J.E. Rexach, M.C. Condro, A.G. Alvarado, M. Harteni, Y.-W. Chen, L. Stiles, A.Y. Chen, I.B. Wanner, X. Yang, S.A. Goldman, D.H. Geschwind, H.I. Kornblum, Y. Zhang, Conservation and divergence of vulnerability and responses to stressors between human and mouse astrocytes, bioRxiv (2020) 2020.04.17.044222. [278] B.S. Carter, D.E. Hamilton, R.C. Thompson, Acute and chronic glucocorticoid treatments regulate astrocyte-enriched mRNAs in multiple brain regions in vivo, Front Neurosci 7 (2013) 139. [279] M. Piechota, M. Korostynski, S. Golda, J. Ficek, D. Jantas, Z. Barbara, R. Przewlocki, Transcriptional signatures of steroid hormones in the striatal neurons and astrocytes, BMC Neurosci 18(1) (2017) 37. [280] H. Zhang, Y. Zhao, Z. Wang, Chronic corticosterone exposure reduces hippocampal astrocyte structural plasticity and induces hippocampal atrophy in mice, Neuroscience letters 592 (2015) 76-81. [281] K. Unemura, T. Kume, M. Kondo, Y. Maeda, Y. Izumi, A. Akaike, Glucocorticoids decrease astrocyte numbers by reducing glucocorticoid receptor expression in vitro and in vivo, J Pharmacol Sci 119(1) (2012) 30-39. [282] J.P. O'Callaghan, R.E. Brinton, B.S. McEwen, Glucocorticoids regulate the synthesis of glial fibrillary acidic protein in intact and adrenalectomized rats but do not affect its expression following brain injury, Journal of neurochemistry 57(3) (1991) 860-869. [283] R.J. Tynan, S.B. Beynon, M. Hinwood, S.J. Johnson, M. Nilsson, J.J. Woods, F.R. Walker, Chronic stress-induced disruption of the astrocyte network is driven by structural atrophy and not loss of astrocytes, Acta Neuropathol 126(1) (2013) 75-91. [284] S. Simard, G. Coppola, C.A. Rudyk, S. Hayley, R.J. McQuaid, N. Salmaso, Profiling changes in cortical astroglial cells following chronic stress, Neuropsychopharmacology 43(9) (2018) 1961-1971. [285] J. Ridet, A. Privat, S. Malhotra, F. Gage, Reactive astrocytes: cellular and molecular cues to biological function, Trends Neurosci 20(12) (1997) 570-577. [286] C. Murphy-Royal, A.D. Johnston, A.K.J. Boyce, B. Diaz-Castro, A. Institoris, G. Peringod, O. Zhang, R.F. Stout, D.C. Spray, R.J. Thompson, B.S. Khakh, J.S. Bains, G.R. Gordon, Stress gates an astrocytic energy reservoir to impair synaptic plasticity, Nature communications 11(1) (2020) 2014-2014. [287] O.F. Sánchez, A.V. Rodríguez, J.M. Velasco-España, L.C. Murillo, J.-J. Sutachan, S.-L. Albarracin, Role of Connexins 30, 36, and 43 in Brain Tumors, Neurodegenerative Diseases, and Neuroprotection, Cells 9(4) (2020) 846. [288] J.J. Miguel-Hidalgo, M. Moulana, P.H. Deloach, G. Rajkowska, Chronic Unpredictable Stress Reduces Immunostaining for Connexins 43 and 30 and Myelin Basic Protein in the Rat Prelimbic and Orbitofrontal Cortices, Chronic Stress 2 (2018) 2470547018814186. [289] J.D. Sun, Y. Liu, Y.H. Yuan, J. Li, N.H. Chen, Gap junction dysfunction in the prefrontal cortex induces depressive-like behaviors in rats, Neuropsychopharmacology 37(5) (2012) 1305-20.

58 [290] D. Huang, C. Li, W. Zhang, J. Qin, W. Jiang, C. Hu, Dysfunction of astrocytic connexins 30 and 43 in the medial prefrontal cortex and hippocampus mediates depressive-like behaviours, Behavioural brain research 372 (2019) 111950. [291] L. Saur, P.B. Bagatini, L.T. Neves, R.M. de Oliveira, S.P. Vaz, K. Ferreira, S.A. Machado, R.G. Mestriner, L.L. Xavier, Experimental post-traumatic stress disorder decreases astrocyte density and changes astrocytic polarity in the CA1 hippocampus of male rats, Neurocheml Res 41(4) (2016) 892-904. [292] Y. Zhao, Q. Zhang, X. Shao, L. Ouyang, X. Wang, K. Zhu, L. Chen, Decreased glycogen content might contribute to chronic stress-induced atrophy of hippocampal astrocyte volume and depression-like behavior in rats, Sci Rep 7 (2017) 43192. [293] L. Xia, M. Zhai, L. Wang, D. Miao, X. Zhu, W. Wang, FGF2 blocks PTSD symptoms via an astrocyte-based mechanism, Behavioural brain research 256 (2013) 472-480. [294] S. Naskar, S. Chattarji, Stress elicits contrasting effects on the structure and number of astrocytes in the amygdala versus hippocampus, eNeuro 6(1) (2019) ENEURO.0338-18.2019. [295] B. Czéh, M. Simon, B. Schmelting, C. Hiemke, E. Fuchs, Astroglial plasticity in the hippocampus is affected by chronic psychosocial stress and concomitant fluoxetine treatment, Neuropsychopharmacology 31(8) (2006) 1616-1626. [296] A. Du Preez, D. Onorato, I. Eiben, K. Musaelyan, M. Egeland, P.A. Zunszain, C. Fernandes, S. Thuret, C.M. Pariante, Chronic stress followed by social isolation promotes depressive-like behaviour, alters microglial and astrocyte biology and reduces hippocampal neurogenesis in male mice (in press), Brain Behav Immun (2020). [297] G. Quesseveur, B. Portal, J.-A. Basile, P. Ezan, A. Mathou, H. Halley, C. Leloup, X. Fioramonti, N. Déglon, C. Giaume, C. Rampon, B.P. Guiard, Attenuated Levels of Hippocampal Connexin 43 and its Phosphorylation Correlate with Antidepressant- and Anxiolytic-Like Activities in Mice, Front Cell Neurosci 9 (2015) 490-490. [298] Q. Liu, B. Li, H.-Y. Zhu, Y.-Q. Wang, J. Yu, G.-C. Wu, Clomipramine treatment reversed the glial pathology in a chronic unpredictable stress-induced rat model of depression, Eur Neuropsychopharmacol 19(11) (2009) 796-805. [299] M. Banasr, G. Chowdhury, R. Terwilliger, S. Newton, R. Duman, K. Behar, G. Sanacora, Glial pathology in an animal model of depression: reversal of stress-induced cellular, metabolic and behavioral deficits by the glutamate-modulating drug riluzole, Mol Psychiatry 15(5) (2010) 501-511. [300] G. Quesseveur, B. Portal, J.-A. Basile, P. Ezan, A. Mathou, H. Halley, C. Leloup, X. Fioramonti, N. Déglon, C. Giaume, Attenuated levels of hippocampal connexin 43 and its phosphorylation correlate with antidepressant-and anxiolytic-like activities in mice, Front Cell Neurosci 9 (2015) 490. [301] A.C. Vernon, W.R. Crum, J.P. Lerch, W. Chege, S. Natesan, M. Modo, J.D. Cooper, S.C.R. Williams, S. Kapur, Reduced Cortical Volume and Elevated Astrocyte Density in Rats Chronically Treated With Antipsychotic Drugs—Linking Magnetic Resonance Imaging Findings to Cellular Pathology, Biol Psychiatry 75(12) (2014) 982-990. [302] K. Braun, R. Antemano, C. Helmeke, M. Büchner, G. Poeggel, Juvenile separation stress induces rapid region- and layer-specific changes in S100ß- and glial fibrillary acidic protein–immunoreactivity in astrocytes of the rodent medial prefrontal cortex, Neuroscience 160(3) (2009) 629-638. [303] H.R. Kwak, J.W. Lee, K.-J. Kwon, C.D. Kang, I.Y. Cheong, W. Chun, S.-S. Kim, H.J. Lee, Maternal social separation of adolescent rats induces hyperactivity and anxiolytic behavior, Korean J Physiol Pharmacol 13(2) (2009) 79-83. [304] M.R. Abbink, J.M. Kotah, L. Hoeijmakers, A. Mak, G. Yvon-Durocher, B. van der Gaag, P.J. Lucassen, A. Korosi, Characterization of astrocytes throughout life in wildtype and APP/PS1 mice after early-life stress exposure, J Neuroinflammation 17(1) (2020) 1-16. [305] I. Farhy-Tselnicker, N.J. Allen, Astrocytes, neurons, synapses: a tripartite view on cortical circuit development, Neural Dev 13(1) (2018) 7. [306] E. Colombo, C. Farina, Astrocytes: key regulators of neuroinflammation, Trends Immunol 37(9) (2016) 608-620. [307] I. Molina-Gonzalez, V.E. Miron, Astrocytes in myelination and remyelination, Neuroscience letters 713 (2019) 134532. [308] C. Nagy, S.G. Torres-Platas, N. Mechawar, G. Turecki, Repression of astrocytic connexins in cortical and subcortical brain regions and prefrontal enrichment of H3K9me3 in depression and suicide, Int J Neuropsychopharmacol 20(1) (2016) 50-57.

59 [309] A. Tanti, P.-E. Lutz, J. Kim, L. O’Leary, J.-F. Théroux, G. Turecki, N. Mechawar, Evidence of decreased gap junction coupling between astrocytes and oligodendrocytes in the anterior cingulate cortex of depressed suicides, Neuropsychopharmacology 44(12) (2019) 2099-2111. [310] E. Nutma, D. Van Gent, S. Amor, L.A.N. Peferoen, Astrocyte and Oligodendrocyte Cross-Talk in the Central Nervous System, Cells 9(3) (2020) 600. [311] K. Masaki, Early disruption of glial communication via connexin gap junction in multiple sclerosis, Baló's disease and neuromyelitis optica, Neuropathology 35(5) (2015) 469-480. [312] R.A. Gittins, P.J. Harrison, A morphometric study of glia and neurons in the anterior cingulate cortex in mood disorder, J Affect Disord 133(1-2) (2011) 328-332. [313] S.G. Torres-Platas, C. Hercher, M.A. Davoli, G. Maussion, B. Labonté, G. Turecki, N. Mechawar, Astrocytic hypertrophy in anterior cingulate white matter of depressed suicides, Neuropsychopharmacology 36(13) (2011) 2650-2658. [314] J.J. Miguel-Hidalgo, C. Baucom, G. Dilley, J.C. Overholser, H.Y. Meltzer, C.A. Stockmeier, G. Rajkowska, Glial fibrillary acidic protein immunoreactivity in the prefrontal cortex distinguishes younger from older adults in major depressive disorder, Biol Psychiatry 48(8) (2000) 861-873. [315] X. Si, J.J. Miguel-Hidalgo, G. O'Dwyer, C.A. Stockmeier, G. Rajkowska, Age-dependent reductions in the level of glial fibrillary acidic protein in the prefrontal cortex in major depression, Neuropsychopharmacology 29(11) (2004) 2088-2096. [316] D. Ongur, W.C. Drevets, J.L. Price, Glial reduction in the subgenual prefrontal cortex in mood disorders, Proc Natl Acad Sci USA 95(22) (1998) 13290-13295. [317] X.-R. Qi, W. Kamphuis, L. Shan, Astrocyte changes in the prefrontal cortex From aged non- suicidal depressed patients, Front Cell Neurosci 13 (2019) 503-503. [318] G. Rajkowska, J.J. Miguel-Hidalgo, Z. Makkos, H. Meltzer, J. Overholser, C. Stockmeier, Layer- specific reductions in GFAP-reactive astroglia in the dorsolateral prefrontal cortex in schizophrenia, Schizophrenia research 57(2-3) (2002) 127-138. [319] A. Schmitt, C. Steyskal, H.G. Bernstein, T. Schneider-Axmann, E. Parlapani, E.L. Schaeffer, W.F. Gattaz, B. Bogerts, C. Schmitz, P. Falkai, Stereologic investigation of the posterior part of the hippocampus in schizophrenia, Acta Neuropathol 117(4) (2009) 395-407. [320] T. Gos, M.L. Schroeter, W. Lessel, H.-G. Bernstein, H. Dobrowolny, K. Schiltz, B. Bogerts, J. Steiner, S100B-immunopositive astrocytes and oligodendrocytes in the hippocampus are differentially afflicted in unipolar and bipolar depression: a postmortem study, Journal of psychiatric research 47(11) (2013) 1694-1699. [321] J.A. Cobb, K. O’Neill, J. Milner, G.J. Mahajan, T.J. Lawrence, W.L. May, J. Miguel-Hidalgo, G. Rajkowska, C.A. Stockmeier, Density of GFAP-immunoreactive astrocytes is decreased in left hippocampi in major depressive disorder, Neuroscience 316 (2016) 209-220. [322] M.B. Müller, P.J. Lucassen, A. Yassouridis, W.J.G. Hoogendijk, F. Holsboer, D.F. Swaab, Neither major depression nor glucocorticoid treatment affects the cellular integrity of the human hippocampus, Eur J Neurosci 14(10) (2001) 1603-1612. [323] L.L. Altshuler, O.A. Abulseoud, L. Foland-Ross, G. Bartzokis, S. Chang, J. Mintz, G. Hellemann, H.V. Vinters, Amygdala astrocyte reduction in subjects with major depressive disorder but not bipolar disorder, Bipolar Disord 12(5) (2010) 541-549. [324] R.C. Paolicelli, G. Bolasco, F. Pagani, L. Maggi, M. Scianni, P. Panzanelli, M. Giustetto, T.A. Ferreira, E. Guiducci, L. Dumas, D. Ragozzino, C.T. Gross, Synaptic pruning by microglia is necessary for normal brain development, Science 333(6048) (2011) 1456-1458. [325] C.N. Parkhurst, G. Yang, I. Ninan, J.N. Savas, J.R. Yates, 3rd, J.J. Lafaille, B.L. Hempstead, D.R. Littman, W.-B. Gan, Microglia promote learning-dependent synapse formation through brain-derived neurotrophic factor, Cell 155(7) (2013) 1596-1609. [326] Dorothy P. Schafer, Emily K. Lehrman, Amanda G. Kautzman, R. Koyama, Alan R. Mardinly, R. Yamasaki, Richard M. Ransohoff, Michael E. Greenberg, Ben A. Barres, B. Stevens, Microglia Sculpt Postnatal Neural Circuits in an Activity and Complement-Dependent Manner, Neuron 74(4) (2012) 691- 705. [327] A. Nimmerjahn, F. Kirchhoff, F. Helmchen, Resting microglial cells are highly dynamic surveillants of brain parenchyma in vivo, Science 308(5726) (2005) 1314-1318.

60 [328] H.S. Domingues, C.C. Portugal, R. Socodato, J.B. Relvas, Oligodendrocyte, astrocyte, and microglia crosstalk in myelin development, damage, and repair, Front Cell Dev Biol 4 (2016) 71. [329] A. Sierra, A. Gottfried-Blackmore, T.A. Milner, B.S. McEwen, K. Bulloch, Steroid hormone receptor expression and function in microglia, Glia 56(6) (2008) 659-674. [330] A.M. Espinosa-Oliva, R.M. de Pablos, R.F. Villarán, S. Argüelles, J.L. Venero, A. Machado, J. Cano, Stress is critical for LPS-induced activation of microglia and damage in the rat hippocampus, Neurobiology of aging 32(1) (2011) 85-102. [331] M.G. Frank, Z.D. Miguel, L.R. Watkins, S.F. Maier, Prior exposure to glucocorticoids sensitizes the neuroinflammatory and peripheral inflammatory responses to E. coli lipopolysaccharide, Brain Behav Immun 24(1) (2010) 19-30. [332] M.A. Calcia, D.R. Bonsall, P.S. Bloomfield, S. Selvaraj, T. Barichello, O.D. Howes, Stress and neuroinflammation: a systematic review of the effects of stress on microglia and the implications for mental illness, Psychopharmacology (Berl) 233(9) (2016) 1637-1650. [333] E.S. Wohleb, A.M. Fenn, A.M. Pacenta, N.D. Powell, J.F. Sheridan, J.P. Godbout, Peripheral innate immune challenge exaggerated microglia activation, increased the number of inflammatory CNS macrophages, and prolonged social withdrawal in socially defeated mice, Psychoneuroendocrinology 37(9) (2012) 1491-1505. [334] E.S. Wohleb, M.L. Hanke, A.W. Corona, N.D. Powell, L.M. Stiner, M.T. Bailey, R.J. Nelson, J.P. Godbout, J.F. Sheridan, β-Adrenergic receptor antagonism prevents anxiety-like behavior and microglial reactivity induced by repeated social defeat, The Journal of neuroscience : the official journal of the Society for Neuroscience 31(17) (2011) 6277-6288. [335] Y. Bian, Z. Pan, Z. Hou, C. Huang, W. Li, B. Zhao, Learning, memory, and glial cell changes following recovery from chronic unpredictable stress, Brain research bulletin 88(5) (2012) 471-476. [336] M. Hinwood, R.J. Tynan, J.L. Charnley, S.B. Beynon, T.A. Day, F.R. Walker, Chronic stress induced remodeling of the prefrontal cortex: structural re-organization of microglia and the inhibitory effect of minocycline, Cerebral cortex (New York, N.Y. : 1991) 23(8) (2013) 1784-1797. [337] B.L. Kopp, D. Wick, J.P. Herman, Differential effects of homotypic vs. heterotypic chronic stress regimens on microglial activation in the prefrontal cortex, Physiol Behav 122 (2013) 246-252. [338] R.J. Tynan, S. Naicker, M. Hinwood, E. Nalivaiko, K.M. Buller, D.V. Pow, T.A. Day, F.R. Walker, Chronic stress alters the density and morphology of microglia in a subset of stress-responsive brain regions, Brain Behav Immun 24(7) (2010) 1058-1068. [339] K.L. Smith, M.S. Kassem, D.J. Clarke, M.P. Kuligowski, M.A. Bedoya-Pérez, S.M. Todd, J. Lagopoulos, M.R. Bennett, J.C. Arnold, Microglial cell hyper-ramification and neuronal dendritic spine loss in the hippocampus and medial prefrontal cortex in a mouse model of PTSD, Brain Behav Immun 80 (2019) 889-899. [340] E.A. Ling, Y.K. Ng, C.H. Wu, C. Kaur, Microglia: its development and role as a neuropathology sensor, Prog Brain Res 132 (2001) 61-79. [341] S. Sugama, M. Fujita, M. Hashimoto, B. Conti, Stress induced morphological microglial activation in the rodent brain: Involvement of interleukin-18, Neuroscience 146(3) (2007) 1388-1399. [342] D.B. McKim, A. Niraula, A.J. Tarr, E.S. Wohleb, J.F. Sheridan, J.P. Godbout, Neuroinflammatory Dynamics Underlie Memory Impairments after Repeated Social Defeat, The Journal of neuroscience : the official journal of the Society for Neuroscience 36(9) (2016) 2590-2604. [343] E.N. Benveniste, Role of macrophages/microglia in multiple sclerosis and experimental allergic encephalomyelitis, J Mol Med 75(3) (1997) 165-173. [344] R.A. Khairova, R. Machado-Vieira, J. Du, H.K. Manji, A potential role for pro-inflammatory cytokines in regulating synaptic plasticity in major depressive disorder, Int J Neuropsychopharmacol 12(04) (2009) 561. [345] L. Peferoen, M. Kipp, P. Van Der Valk, J.M. Van Noort, S. Amor, Oligodendrocyte-microglia cross-talk in the central nervous system, Immunology 141(3) (2014) 302-313. [346] A. Matejuk, R.M. Ransohoff, Crosstalk Between Astrocytes and Microglia: An Overview, Front Immunol 11 (2020) 1416. [347] C. Cunningham, Microglia and neurodegeneration: the role of systemic inflammation, Glia 61(1) (2013) 71-90.

61 [348] K. Ramirez, D.T. Shea, D.B. McKim, B.F. Reader, J.F. Sheridan, Imipramine attenuates neuroinflammatory signaling and reverses stress-induced social avoidance, Brain Behav Immun 46 (2015) 212-220. [349] G. Milior, C. Lecours, L. Samson, K. Bisht, S. Poggini, F. Pagani, C. Deflorio, C. Lauro, S. Alboni, C. Limatola, I. Branchi, M.-E. Tremblay, L. Maggi, Fractalkine receptor deficiency impairs microglial and neuronal responsiveness to chronic stress, Brain Behav Immun 55 (2016) 114-125. [350] L. Weinhard, G. di Bartolomei, G. Bolasco, P. Machado, N.L. Schieber, U. Neniskyte, M. Exiga, A. Vadisiute, A. Raggioli, A. Schertel, Y. Schwab, C.T. Gross, Microglia remodel synapses by presynaptic trogocytosis and spine head filopodia induction, Nature communications 9(1) (2018) 1228. [351] S. Hellwig, S. Brioschi, S. Dieni, L. Frings, A. Masuch, T. Blank, K. Biber, Altered microglia morphology and higher resilience to stress-induced depression-like behavior in CX3CR1-deficient mice, Brain Behav Immun 55 (2016) 126-137. [352] D.J. Stein, M.F. Vasconcelos, L. Albrechet-Souza, K.M.M. Ceresér, R.M.M. de Almeida, Microglial Over-Activation by Social Defeat Stress Contributes to Anxiety- and Depressive-Like Behaviors, Front Behav Neurosci 11 (2017) 207. [353] A. Karperien, H. Ahammer, H. Jelinek, Quantitating the subtleties of microglial morphology with fractal analysis, Front Cell Neurosci 7 (2013) 3. [354] J.J. Neher, C. Cunningham, Priming Microglia for Innate Immune Memory in the Brain, Trends Immunol 40(4) (2019) 358-374. [355] G.J. Harry, A.D. Kraft, Microglia in the developing brain: A potential target with lifetime effects, NeuroToxicology 33(2) (2012) 191-206. [356] J.C. Delpech, L. Wei, J. Hao, X. Yu, C. Madore, O. Butovsky, A. Kaffman, Early life stress perturbs the maturation of microglia in the developing hippocampus, Brain Behav Immun 57 (2016) 79- 93. [357] Y. Zhan, R.C. Paolicelli, F. Sforazzini, L. Weinhard, G. Bolasco, F. Pagani, A.L. Vyssotski, A. Bifone, A. Gozzi, D. Ragozzino, C.T. Gross, Deficient neuron-microglia signaling results in impaired functional brain connectivity and social behavior, Nat Neurosci 17(3) (2014) 400-406. [358] Y.-L. Wang, Q.-Q. Han, W.-Q. Gong, D.-H. Pan, L.-Z. Wang, W. Hu, M. Yang, B. Li, J. Yu, Q. Liu, Microglial activation mediates chronic mild stress-induced depressive- and anxiety-like behavior in adult rats, J Neuroinflammation 15(1) (2018) 21. [359] M. Hinwood, J. Morandini, T.A. Day, F.R. Walker, Evidence that microglia mediate the neurobiological effects of chronic psychological stress on the medial prefrontal cortex, Cerebral cortex (New York, N.Y. : 1991) 22(6) (2012) 1442-1454. [360] F.K. Johnson, A. Kaffman, Early life stress perturbs the function of microglia in the developing rodent brain: New insights and future challenges, Brain Behav Immun 69 (2018) 18-27. [361] I.B. Hickie, R. Banati, C.H. Stewart, A.R. Lloyd, Are common childhood or adolescent infections risk factors for schizophrenia and other psychotic disorders?, Med J Aust 190(S4) (2009) S17-S21. [362] E. Setiawan, A.A. Wilson, R. Mizrahi, P.M. Rusjan, L. Miler, G. Rajkowska, I. Suridjan, J.L. Kennedy, P.V. Rekkas, S. Houle, J.H. Meyer, Role of translocator protein density, a marker of neuroinflammation, in the brain during major depressive episodes, JAMA psychiatry 72(3) (2015) 268- 275. [363] S.E. Holmes, R. Hinz, S. Conen, C.J. Gregory, J.C. Matthews, J.M. Anton-Rodriguez, A. Gerhard, P.S. Talbot, Elevated Translocator Protein in Anterior Cingulate in Major Depression and a Role for Inflammation in Suicidal Thinking: A Positron Emission Tomography Study, Biol Psychiatry 83(1) (2018) 61-69. [364] L. Su, Y.O. Faluyi, Y.T. Hong, T.D. Fryer, E. Mak, S. Gabel, L. Hayes, S. Soteriades, G.B. Williams, R. Arnold, L. Passamonti, P.V. Rodríguez, A. Surendranathan, R.W. Bevan-Jones, J. Coles, F. Aigbirhio, J.B. Rowe, J.T. O'Brien, Neuroinflammatory and morphological changes in late-life depression: the NIMROD study, Br J Psychiatry 209(6) (2016) 525-526. [365] S.G. Torres-Platas, C. Cruceanu, G.G. Chen, G. Turecki, N. Mechawar, Evidence for increased microglial priming and macrophage recruitment in the dorsal anterior cingulate white matter of depressed suicides, Brain Behav Immun 42 (2014) 50-59. [366] J. Steiner, H. Bielau, R. Brisch, P. Danos, O. Ullrich, C. Mawrin, H.-G. Bernstein, B. Bogerts, Immunological aspects in the neurobiology of suicide: elevated microglial density in schizophrenia and depression is associated with suicide, Journal of psychiatric research 42(2) (2008) 151-157.

62 [367] R. Brisch, J. Steiner, C. Mawrin, M. Krzyżanowska, Z. Jankowski, T. Gos, Microglia in the dorsal raphe nucleus plays a potential role in both suicide facilitation and prevention in affective disorders, Eur Arch Psychiatry Clin Neurosci 267(5) (2017) 403-415. [368] V.V. Giridharan, P. Sayana, O.F. Pinjari, N. Ahmad, M.I. da Rosa, J. Quevedo, T. Barichello, Postmortem evidence of brain inflammatory markers in bipolar disorder: a systematic review, Mol Psychiatry 25(1) (2020) 94-113. [369] N.A. Munn, Microglia dysfunction in schizophrenia: an integrative theory, Medical hypotheses 54(2) (2000) 198-202. [370] A. Monji, T.A. Kato, Y. Mizoguchi, H. Horikawa, Y. Seki, M. Kasai, Y. Yamauchi, S. Yamada, S. Kanba, Neuroinflammation in schizophrenia especially focused on the role of microglia, Prog Neuropsychopharmacol Biol Psychiatry 42 (2013) 115-121. [371] L.E. Laskaris, M.A. Di Biase, I. Everall, G. Chana, A. Christopoulos, E. Skafidas, V.L. Cropley, C. Pantelis, Microglial activation and progressive brain changes in schizophrenia, Br J Pharmacol 173(4) (2016) 666-680. [372] M.O. Trépanier, K.E. Hopperton, R. Mizrahi, N. Mechawar, R.P. Bazinet, Postmortem evidence of cerebral inflammation in schizophrenia: a systematic review, Mol Psychiatry 21(8) (2016) 1009-1026. [373] A. Jenkins, A. Locker, D. Volk, Increased mRNA levels of microglial markers of phagocytosis in schizophrenia, Biol Psychiatry 85(10) (2019) S278-S279. [374] C.M. Sellgren, J. Gracias, B. Watmuff, J.D. Biag, J.M. Thanos, P.B. Whittredge, T. Fu, K. Worringer, H.E. Brown, J. Wang, A. Kaykas, R. Karmacharya, C.P. Goold, S.D. Sheridan, R.H. Perlis, Increased synapse elimination by microglia in schizophrenia patient-derived models of synaptic pruning, Nat Neurosci 22(3) (2019) 374-385. [375] U. Fünfschilling, L.M. Supplie, D. Mahad, S. Boretius, A.S. Saab, J. Edgar, B.G. Brinkmann, C.M. Kassmann, I.D. Tzvetanova, W. Möbius, F. Diaz, D. Meijer, U. Suter, B. Hamprecht, M.W. Sereda, C.T. Moraes, J. Frahm, S. Goebbels, K.-A. Nave, Glycolytic oligodendrocytes maintain myelin and long-term axonal integrity, Nature 485(7399) (2012) 517-521. [376] X. Dai, L.D. Lercher, P.M. Clinton, Y. Du, D.L. Livingston, C. Vieira, L. Yang, M.M. Shen, C.F. Dreyfus, The trophic role of oligodendrocytes in the basal forebrain, The Journal of neuroscience : the official journal of the Society for Neuroscience 23(13) (2003) 5846-5853. [377] J.H. Seo, T. Maki, M. Maeda, N. Miyamoto, A.C. Liang, K. Hayakawa, L.-D.D. Pham, F. Suwa, A. Taguchi, T. Matsuyama, Oligodendrocyte precursor cells support blood-brain barrier integrity via TGF-β signaling, PLoS One 9(7) (2014) e103174. [378] B. Valério-Gomes, D.M. Guimarães, D. Szczupak, R. Lent, The Absolute Number of Oligodendrocytes in the Adult Mouse Brain, Front Neuroanat 12 (2018) 90. [379] S. Szuchet, J.A. Nielsen, G. Lovas, M.S. Domowicz, J.M. de Velasco, D. Maric, L.D. Hudson, The genetic signature of perineuronal oligodendrocytes reveals their unique phenotype, Eur J Neurosci 34(12) (2011) 1906-1922. [380] V. Haroutunian, P. Katsel, P. Roussos, K. Davis, L. Altshuler, G. Bartzokis, Myelination, oligodendrocytes, and serious mental illness, Glia 62(11) (2014) 1856-1877. [381] D.P. Pelvig, H. Pakkenberg, A.K. Stark, B. Pakkenberg, Neocortical glial cell numbers in human brains, Neurobiology of aging 29(11) (2008) 1754-1762. [382] F. Cathomas, D. Azzinnari, G. Bergamini, H. Sigrist, M. Buerge, V. Hoop, B. Wicki, L. Goetze, S. Soares, D. Kukelova, E. Seifritz, S. Goebbels, K.A. Nave, M.S. Ghandour, C. Seoighe, T. Hildebrandt, G. Leparc, H. Klein, E. Stupka, B. Hengerer, C.R. Pryce, Oligodendrocyte gene expression is reduced by and influences effects of chronic social stress in mice, Genes, brain, and behavior 18(1) (2019) e12475. [383] V. Bonnefil, K. Dietz, M. Amatruda, M. Wentling, A.V. Aubry, J.L. Dupree, G. Temple, H.-J. Park, N.S. Burghardt, P. Casaccia, J. Liu, Region-specific myelin differences define behavioral consequences of chronic social defeat stress in mice, eLife 8 (2019) e40855. [384] M.L. Lehmann, T.K. Weigel, A.G. Elkahloun, M. Herkenham, Chronic social defeat reduces myelination in the mouse medial prefrontal cortex, Sci Rep 7 (2017) 46548. [385] S. Chetty, A.R. Friedman, K. Taravosh-Lahn, E.D. Kirby, C. Mirescu, F. Guo, D. Krupik, A. Nicholas, A. Geraghty, A. Krishnamurthy, M.K. Tsai, D. Covarrubias, A. Wong, D. Francis, R.M. Sapolsky, T.D. Palmer, D. Pleasure, D. Kaufer, Stress and glucocorticoids promote oligodendrogenesis in the adult hippocampus, Mol Psychiatry 19(12) (2014) 1275-1283.

63 [386] R. Dutta, B.D. Trapp, Mechanisms of neuronal dysfunction and degeneration in multiple sclerosis, Prog Neurobiol 93(1) (2011) 1-12. [387] P.A. Felts, T.A. Baker, K.J. Smith, Conduction in segmentally demyelinated mammalian central axons, The Journal of neuroscience : the official journal of the Society for Neuroscience 17(19) (1997) 7267-7277. [388] N. Edgar, E. Sibille, A putative functional role for oligodendrocytes in mood regulation, Transl Psychiatry 2(5) (2012) e109-e109. [389] J. Liu, K. Dietz, J.M. DeLoyht, X. Pedre, D. Kelkar, J. Kaur, V. Vialou, M.K. Lobo, D.M. Dietz, E.J. Nestler, J. Dupree, P. Casaccia, Impaired adult myelination in the prefrontal cortex of socially isolated mice, Nat Neurosci 15(12) (2012) 1621-1623. [390] M. Makinodan, K.M. Rosen, S. Ito, G. Corfas, A for Social Experience–Dependent Oligodendrocyte Maturation and Myelination, Science 337(6100) (2012) 1357. [391] E. Traiffort, A. Kassoussi, A. Zahaf, Y. Laouarem, Astrocytes and Microglia as Major Players of Myelin Production in Normal and Pathological Conditions, Front Cell Neurosci 14 (2020) 79-79. [392] L. Peferoen, M. Kipp, P. van der Valk, J.M. van Noort, S. Amor, Oligodendrocyte‐‐ microglia cross talk in the central nervous system, Immunology 141(3) (2014) 302-313. [393] Y. Pang, Z. Cai, P.G. Rhodes, Disturbance of oligodendrocyte development, hypomyelination and white matter injury in the neonatal rat brain after intracerebral injection of lipopolysaccharide, Dev Brain Res 140(2) (2003) 205-214. [394] N.A. Uranova, V.M. Vostrikov, D.D. Orlovskaya, V.I. Rachmanova, Oligodendroglial density in the prefrontal cortex in schizophrenia and mood disorders: a study from the Stanley Neuropathology Consortium, Schizophrenia research 67(2-3) (2004) 269-275. [395] N. Uranova, D. Orlovskaya, O. Vikhreva, I. Zimina, N. Kolomeets, V. Vostrikov, V. Rachmanova, Electron microscopy of oligodendroglia in severe mental illness, Brain research bulletin 55(5) (2001) 597-610. [396] C. Nagy, M. Maitra, A. Tanti, M. Suderman, J.F. Théroux, M.A. Davoli, K. Perlman, V. Yerko, Y.C. Wang, S.J. Tripathy, P. Pavlidis, N. Mechawar, J. Ragoussis, G. Turecki, Single-nucleus transcriptomics of the prefrontal cortex in major depressive disorder implicates oligodendrocyte precursor cells and excitatory neurons, Nat Neurosci (2020) 771-781. [397] P. Katsel, K.L. Davis, V. Haroutunian, Variations in myelin and oligodendrocyte-related gene expression across multiple brain regions in schizophrenia: a gene ontology study, Schizophrenia research 79(2-3) (2005) 157-173. [398] S.N. Mitkus, T.M. Hyde, R. Vakkalanka, B. Kolachana, D.R. Weinberger, J.E. Kleinman, B.K. Lipska, Expression of oligodendrocyte-associated genes in dorsolateral prefrontal cortex of patients with schizophrenia, Schizophrenia research 98(1-3) (2008) 129-138. [399] P.E. Lutz, A. Tanti, A. Gasecka, S. Barnett-Burns, J.J. Kim, Y. Zhou, G.G. Chen, M. Wakid, M. Shaw, D. Almeida, M.A. Chay, J. Yang, V. Larivière, M.N. M'Boutchou, L.C. van Kempen, V. Yerko, J. Prud'homme, M.A. Davoli, K. Vaillancourt, J.F. Théroux, A. Bramoullé, T.Y. Zhang, M.J. Meaney, C. Ernst, D. Côté, N. Mechawar, G. Turecki, Association of a history of child abuse with impaired myelination in the anterior cingulate cortex: convergent epigenetic, transcriptional, and morphological evidence, Am J Psychiatry 174(12) (2017) 1185-1194. [400] A. Tanti, J.J. Kim, M. Wakid, M.A. Davoli, G. Turecki, N. Mechawar, Child abuse associates with an imbalance of oligodendrocyte-lineage cells in ventromedial prefrontal white matter, Mol Psychiatry 23(10) (2018) 2018-2028. [401] A. Alizadeh, S.M. Dyck, S. Karimi-Abdolrezaee, Myelin damage and repair in pathologic CNS: challenges and prospects, Front Mol Neurosci 8 (2015) 35. [402] R.A. Cowell, D. Cicchetti, F.A. Rogosch, S.L. Toth, Childhood maltreatment and its effect on neurocognitive functioning: Timing and chronicity matter, Development and psychopathology 27(2) (2015) 521-533. [403] Q. Mu, Y. Chen, J. Wang, Deciphering Brain Complexity Using Single-cell Sequencing, Genomics Proteomics Bioinformatics 17(4) (2019) 344-366. [404] L.G. Goetschius, T.C. Hein, C. Mitchell, N.L. Lopez-Duran, V.C. McLoyd, J. Brooks-Gunn, S.S. McLanahan, L.W. Hyde, C.S. Monk, Childhood violence exposure and social deprivation predict adolescent amygdala-orbitofrontal cortex white matter connectivity, Dev Cogn Neurosci 45 (2020) 100849.

64 FIGURES

Figure 1. Early-life/juvenile stress vs. adulthood stress impacts on pyramidal neurons. Stress-induced changes are highlighted by the dotted outline. Stress in adulthood causes a two- phase retraction; both of the dendrites themselves and of their constituent dendritic spines. With time, these changes are largely recoverable, albeit with regrowth occurring closer to the soma. However, if stress is experienced during early-life/adolescence, retraction of dendrites and dendritic spines can persist into adulthood. This is seen in both the PFC and hippocampus.

65 Figure 2. Model of cellular impacts of stress based off of evidence from the prefrontal cortex. Stress-induced changes are highlighted by the dotted outline. In control, non-stressed individuals, excitatory and inhibitory balance is maintained through attenuation of inhibitory and excitatory neurons. Glia provide important and diverse roles and form a closely communicative and tightly coordinated network which maintains homeostatic control. Following severe psychological stress, broad structural and functional consequences are seen across cellular networks (highlighted by dotted outlines). Changes to morphology are seen after several different stress exposures but are most substantial in stress-susceptible individuals, during key neurodevelopmental stages (e.g. early-life). Chronic dysregulation of cell-to-cell communication due to cell morphology changes can contribute to the development of cognitive and emotional impairments, which are hallmark symptoms of psychiatric disorders. Specifically: i) Excitatory (pyramidal) neurons display decreased synaptic number and activity as well as reduced process branching, impairing top-down regulation of other brain areas with their projections, as well as integrative capacity of the PFC; ii) Although parvalbumin (PV)-expressing interneurons display

66 decreased branching, they maintain increased number and connectivity with excitatory neurons, dampening the capacity for top-down control of the prefrontal cortex (PFC); iii) Somatostatin (SST)-expressing interneurons display decreased branching and also reduced connectivity with neurons and other interneurons, promoting greater inhibitory actions; iv) Astrocytes display impaired capacity to modulate synaptic environments, further impairing neuronal function and can also impair the function of other glia, such as oligodendrocytes; v) Chronic neuroinflammation caused by increased reactivity and potential reactive capacity of microglia, in conjunction with increased interaction with other cells, contributes to impairments seen in other cellular populations. A population of microglia are also hyper-ramified following stress; vi) Reduced myelination via oligodendrocyte changes further impairs the potential for neuronal , as well increases susceptibility of neurons to damage. This may be due to both direct effects of stress on oligodendrocytes as well an impairment of astrocytes and microglia which are important to oligodendrocyte maintenance.

Figure 3. Model of excitatory/inhibitory balance shifts over the life course in response to stress. During key developmental periods, such as early life and adolescence (left panels, x- axis), the excitatory/inhibitory balance (y-axis) is sensitive to stress (A). Persisting stress, such as neglect or trauma, then establishes shifts across the cellular network including altering microglial

67 and astrocyte functions in synapse regulation, increasing neuroinflammation, decreasing myelination and establishing a net over-inhibition (B). These changes to the cytoarchitecture and function of neurons and glia induced by stress likely impact the developmental trajectory of circuits, with lasting consequences across the life-course (C). In adulthood (right panel, x-axis), the excitatory/inhibitory balance is also sensitive to stress, but the consequences are less persistent and can often be restored to homeostasis if the stress is removed for a period of time (D). The exception appears when stress is experienced earlier in life, and again in adulthood (E), with the earlier stress priming microglia and astrocytes to subsequent stress (F). This likely precipitates dysregulation across excitatory/inhibitory cells, leading to persistent dysfunction of circuits, as seen in (B/C).

68 Table 1. Summary of structural and functional magnetic resonance imaging studies evaluating the structure and function of the prefrontal cortex, hippocampus, and amygdala following stress exposure. Studies were detected in a PudMed search using the terms “adversity”, “stress”, “MRI”, “magnetic resonance imaging”, combined with either “prefrontal cortex”, “hippocampus” or “amygdala”. Unless otherwise stated, stress is compared to healthy controls without a history of stress or adversity. Studies of cohorts limited to cases of PTSD compared to healthy no stress controls were not included in this summary.

Referenc Type of Stress Timing of Stress Time of Measurement Method Impact of stress e Prefrontal Cortex [32] Severe poverty Early life Adulthood sMRI ↓ OFC volume [33] Chronic adversity (multimodal e.g. Infancy/childhood/adolescence Adulthood sMRI ↓ right OFC volume (infancy) socioeconomic status, familial stress) [31] Physical abuse Childhood Childhood sMRI ↓ mPFC volume ↓ OFC volume ↓ DLPFC volume ↓ VMPFC volume [35] Abuse Childhood Adolescence sMRI ↓ VMPFC volume ↓ OFC volume [53] Emotional maltreatment (various) Childhood Adolescence sMRI ↓ mPFC volume [38] Sexual assault Childhood Adulthood sMRI ↓ ACC volume (emotional abuse) Emotional abuse ↓ PCC volume (emotional abuse) [37] Childhood adversity Childhood Adulthood1 sMRI ↓ left DLPFC volume [103] Cumulative adversity Childhood through to Adulthood fMRI ↑ lateral PFC activity adulthood [39] Self-perceived social status Adulthood Adulthood sMRI ↓ ACC volume [25] Cumulative adversity Adulthood Adulthood sMRI ↓ mPFC volume ↓ ACC volume [47] Self-perceived stress Adulthood Adulthood fMRI ↓ DLPFC functional connectivity with frontoparietal network

[34] Self-perceived stress Older adulthood (65-90 years) Adulthood sMRI ↓ lateral PFC volume (dorsolateral and ventrolateral)

Hippocampus [77] Early life adversity Early life Childhood sMRI ↓ left and right hippocampus volume ↓ CA1 volume ↓ CA3 volume ↓ DG volume

0 [76] Early life adversity Early life Childhood sMRI ↓ in total hippocampus volume (only in cases where stress onset during early childhood (<5 years)) [81] Early life adversity (e.g. neglect/abuse), Early life/adolescence Adolescence sMRI ↓ left and right hippocampus volume adolescent chronic stress (socioeconomic status, social, health) [74] Childhood maltreatment Early life Adulthood sMRI ↓ left CA2 volume ↓ left CA3 volume ↓ DG volume [78] Threat (e.g. abuse) Childhood Adolescent/Adulthood sMRI ↓ right hippocampus volume Deprivation (e.g. neglect) (deprivation) ↔ hippocampus volume (threat) [70] Childhood violence Childhood Childhood/Adolescence sMRI ↓ total hippocampus volume (8-17 years) [80] Childhood adversity Childhood Adulthood2 sMRI ↑ CA3 volume [37] Childhood adversity Childhood Adulthood1 sMRI ↓ left and right hippocampus volume [75] Abuse/maltreatment Childhood Adulthood sMRI ↓ left, right and total hippocampus volume ↓ only in men (not women) [79] Childhood adversity Childhood Adulthood sMRI ↔ hippocampus volume [113] Childhood adversity (meta-analysis) Childhood Adulthood sMRI ↓ hippocampus volume [67] Meta-analysis (including Childhood/adulthood Adulthood sMRI ↓ left, right, and total hippocampus childhood/adulthood abuse and combat volume across 12 studies trauma) [103] Cumulative adversity throughout life Childhood through to Adulthood fMRI ↑ hippocampus activity adulthood [73] Self-perceived stress Adolescence Adolescence sMRI ↓ left and total hippocampus volume [71] Emotional trauma Adolescence Adolescence sMRI ↓ CA3 volume [83] Recent life stress Young Adulthood Young Adulthood fMRI ↓ CA2/CA3/DG connectivity with the caudate [68] Self-perceived stress Adulthood Older Adulthood sMRI ↓ left and right hippocampus volume Number of negative stressful life events [72] Financial hardship Adulthood Adulthood sMRI ↓ hippocampus volume [69] Stressful military service Adulthood Adulthood sMRI/fMRI ↓ total hippocampus volume in vulnerable individuals ↓ connectivity with VMPFC [82] Self-perceived stress Adulthood Adulthood sMRI ↓ anterior hippocampus volume 1 Amygdala [106] Early life stress Early life Adulthood fMRI ↑ reactivity in response to mood change [100] Sexual/Physical abuse Early life Adulthood sMRI ↓ left amygdala volume (recent life Recent life stress Adulthood stress) ↔ volume (childhood abuse)

[404] Childhood violence Childhood Adolescence fMRI ↔ amygdala PFC connectivity Social deprivation independently ↓ amygdala-PFC connectivity when exposed to both stress [107] Childhood trauma Childhood Adolescence sMRI ↓ left amygdala development (early to late adolescence)

[70] Childhood violence Childhood Childhood/Adolescence sMRI ↓ total amygdala volume (8-17 years) [108] Childhood adversity Childhood Adulthood sMRI ↑ amygdala volume ↑ right amygdala volume with adversity severity ↑ right amygdala volume with verbal abuse, emotional abuse, and witnessing sibling assault [110] Childhood trauma Childhood Adulthood sMRI ↓ amygdala volume [111] Childhood trauma Childhood Adulthood sMRI ↓ amygdala volume [113] Childhood adversity (meta-analysis) Childhood Adulthood sMRI ↔ amygdala volume [112] Childhood adversity Childhood Adulthood sMRI ↓ right amygdala volume ↓ BLA volume [103] Cumulative adversity throughout life Childhood through to Adulthood fMRI ↑ amygdala activity adulthood [98] Financial stress (unemployment) Adulthood Adulthood fMRI ↑ amygdala functional coupling to ACC

[99] Self-perceived stress Adulthood Adulthood sMRI ↑ right amygdala volume (reduced PSS over 8-week period associated with reduced volume) [72] Financial hardship Adulthood Adulthood sMRI ↓ amygdala volume [105] Various (childhood abuse, domestic Various Adulthood sMRI ↑ left lateral amygdala volume 2 violence, loss of loved one) Abbreviations: fMRI, functional magnetic resonance imaging; sMRI, structural magnetic resonance imaging PFC, prefrontal cortex; mPFC, medial prefrontal cortex; OFC, orbitofrontal cortex; DLFPC, dorsolateral prefrontal cortex; VMPFC ventromedial prefrontal cortex; ACC anterior cingulate cortex; PCC, posterior cingulate cortex, CA, Cornu Ammonis; DG, dentate gyrus; BLA, basolateral amygdala.

1cases diagnosed with schizophrenia 2cases diagnosed with major depressive disorder

3