Abstract
Development of the brain prenatally is affected by maternal experience and exposure. Prenatal maternal psychological stress changes brain development and results in increased risk for neuropsychiatric disorders. In this review, multiple levels of prenatal stress mechanisms (offspring brain, placenta, and maternal physiology) are discussed and their intersection with cellular stress mechanisms explicated. Heat shock factors and oxidative stress are closely related to each other and converge with the inflammation, hormones, and cellular development that have been more deeply explored as the basis of prenatal stress risk. Increasing evidence implicates cellular stress mechanisms in neuropsychiatric disorders associated with prenatal stress including affective disorders, schizophrenia, and child-onset psychiatric disorders. Heat shock factors and oxidative stress also have links with the mechanisms involved in other kinds of prenatal stress including external exposures such as environmental toxicants and internal disruptions such as preeclampsia. Integrative understanding of developmental neurobiology with these cellular and physiological mechanisms is necessary to reduce risks and promote healthy brain development.
Keywords: prenatal stress, oxidative stress, heat shock, neurodevelopment, embryonic brain, neuropsychiatric disorders
1. Introduction
Prenatal brain development occurs via a complex interplay between maternal, fetal, and environmental signals. Although it has been recognized for centuries that environmental influences on a pregnant mother relate to offspring health and development, intentional manipulation of the maternal environment with the goal of benefitting child development is relatively new [14]. In the past few decades, vitamin supplementation, psychotherapy, and good relationships with obstetric health professionals have been used to increase positive outcomes for mothers and their offspring [88]. Today, we take for granted that attentive prenatal care is essential for optimal embryonic and fetal development. However, current efforts do not address many other poorly understood aspects of the maternal-fetal interaction. Of particular interest are the effects of maternal psychological stress on prenatal neurodevelopment [118].
Critically, the physiologic state created by stress during pregnancy can have adverse effects on offspring brain development. Prenatal perturbations in neurodevelopment due to stress have been demonstrated to increase the risk for psychiatric disorders including attention deficit hyperactivity disorder (ADHD), autism spectrum disorder (ASD), major depressive disorder, bipolar disorder, and schizophrenia [57, 112, 149, 199], with male offspring being at higher risk than females [57, 112, 128]. To this point, pregnant women who experience bereavement [94, 112], war [193], natural disasters [99, 106], or high daily stress [57] have offspring with higher rates of these disorders. Additionally, stress may arise from poorly managed mood disorders during pregnancy, and mood disorder severity is linked to altered neurodevelopmental outcomes in offspring [26, 36, 38, 57, 112, 128].
Though poorly understood, a complex interplay of molecular and cellular mechanisms contributes to the aberrant and pathological outcomes induced by prenatal maternal stress. Stress hormones— glucocorticoids—are an obvious area of inquiry, but it has become clear that other mechanisms are involved including inflammation, hypoxia, and growth-factor signaling [157]. These physiological mechanisms induce oxidative stress and interact at the cellular level with the heat shock response, complicating how prenatal stress contributes risk for psychiatric disorders.
To address these complex risks, there is a critical need to better understand the cellular and molecular mechanisms by which the maternal physiologic stress response alters neurodevelopment. For new interventions to be developed and current guidelines refined, an understanding of neurodevelopmental mechanisms, placental intermediaries, and maternal physiology is needed. Physiological stress responses are linked through multiple mechanisms to cellular stress responses [75]. In this review, we will examine the roles of heat shock systems and oxidative stress in prenatal stress and its effects on neurodevelopment, as well as how these cellular systems may interact with maternal stress physiology. Elucidating the relationship between these processes is essential for understanding and targeting these mechanisms to buffer the effects of prenatal stress on offspring neurodevelopment.
2. Brain development and prenatal stress
2.1. Neurodevelopmental processes affected by prenatal stress
There are numerous neurodevelopmental processes that may be impacted by maternal stress, many of which may interact with the heat shock response (HSR) and oxidative stress. Maternal, placental, and embryonic factors, as well as the interactions between these systems, may impact neurodevelopmental processes during prenatal stress. We will explore these interactions in subsequent sections of this review, but we will first examine mechanisms in the developing brain that are susceptible to prenatal stress. Among these affected processes are changes in offspring GABAergic systems, neuroimmune activation, and hypothalamic-pituitary-adrenal (HPA) axis development [180].
Development of the GABAergic neuron system is particularly vulnerable to prenatal stress. During embryonic development, GABAergic neuronal progenitors are born in the medial and caudal ganglionic eminences, from which they migrate tangentially into the developing neocortex [72, 152, 184]. GABAergic progenitors differentiate into interneurons as they migrate and integrate into the neocortex, ultimately forming the inhibitory neural circuitry required for cognition, learning, and motor tasks [7, 56]. Prenatal stress late in mouse gestation reduces GABAergic progenitor production [190], while prenatal stress early in gestation delays the tangential migration in GABAergic interneurons [180]. Multiple components of the maternal physiologic stress response have effects on GABAergic neuron development. Our group and others have found that these embryonic brain processes may be sensitive to maternal inflammation [71, 142] and oxidative stress [28], for example. In addition, embryonic glucocorticoid exposure impedes pyramidal neuron radial migration in the developing cortex [62]. Thus, maternal physiologic stress response may impede multiple aspects of neuronal migration events in the embryonic forebrain. Impaired heat shock systems may be a means by which cell migration is affected, due to interactions with the proteins necessary for cell movement [52, 74, 129].
Furthermore, the effects of prenatal stress on GABAergic interneuron populations persist well into adulthood. For instance, protein levels of the GABA-synthesizing enzyme GAD67 are decreased in the frontal cortex with prenatal stress, and the maturation of postnatal GABAergic neuron development is markedly delayed [119, 126, 190]. GABAergic neuron development may be linked to both oxidative stress and heat shock signaling. Postanatal parvalbumin neuron development, for instance, is sensitive to redox imbalance [50]. Intracellular heat shock protein (HSP) production promotes GABAergic interneuron genesis in vitro [92].
Recent evidence suggests that the maternal stress response elicits changes to the embryonic immune system, which in turn can impact neurodevelopment. Microglia are resident macrophages of the central nervous system that play important roles in brain development. Derived from yolk-sac monocytes, microglia migrate from the vasculature into the developing brain by embryonic day 9 in mice [186]. Like other phagocytic cells, microglia are innate immune cells that regulate inflammatory responses, though they also have a parallel role in sculpting neurodevelopment. For instance, microglia regulate circuity in the forebrain by modulating axon growth [179]. As in other immune cells, cellular stress mechanisms like oxidative stress and the HSR are intrinsically linked to microglia function [84, 183]. This is critical, as microglia control neural progenitor cell populations during embryonic development by either promoting cell survival or reducing the progenitor pool via phagocytosis [124, 187].
Prenatal stress affects microglia populations in both the embryonic and adult brain [49]. During normal development, microglia transition from an amoeboid state to progressively more ramified morphologies, and maternal stress appears to accelerate this process in offspring brain [177]. In addition, prenatal stress increases multivacuolated microglia density in the cortical plate at embryonic day 14 in mice [71]. Furthermore, these changes in microglial activation appear to persist into adulthood, as adult rodents exposed to prenatal stress exhibit less ramified and more readily-activated microglia populations in the cortex and hippocampus [49, 71]. These observed changes in microglia proliferation and maturation suggest that prenatal stress may induce a lasting deleterious, pro-inflammatory environment in the offspring brain.
The growth and maturation of the hypothalamic-pituitary-adrenal (HPA) axis is another neurodevelopmental process that is affected by prenatal stress. In the hypothalamus, for instance, early increases in apoptotic activity is observed in response to prenatal stress [61]. Prenatal stress may have lasting deleterious effects on postnatal HPA functioning. For example, animal studies suggest a hyperactive (higher baseline activity) or hypersensitive (greater response to stressful stimuli) HPA phenotype in prenatally stressed offspring. Adult rat offspring from prenatally-stressed litters exhibit greater HPA sensitivity to stressful stimuli, and prenatally-stressed rhesus monkeys have been found to exhibit higher baseline cortisol and ACTH plasma levels during adulthood [43, 78, 198]. Female offspring tend to show greater susceptibility to the effects of prenatal stress on HPA axis development, but there is some discrepancy between studies regarding sex-specific effects [93]. Changes in offspring HPA functioning following prenatal stress also involve the hippocampus, which regulates the HPA axis via glucocorticoid receptor (GR)-induced feedback inhibition [60]. Decreased hippocampal volume and GR levels, for example, are observed in non-human primates [66]. The effects of prenatal stress on the developing HPA axis are evidently sufficient to produce permanent maladaptive changes to this neuroendocrine system.
More recent evidence suggests an epigenetic mechanism underlying changes in HPA functioning associated with prenatal stress. Prenatal stress increases promoter methylation of the GR gene NR3C1 in infants, and NR3C1 methylation levels in turn predicts infant cortisol reactivity [66, 178]. Due to the heritability of DNA methylation patterns, prenatal stress may have cascading negative impacts on multiple generations [127]. Furthermore, higher cortisol reactivity may predispose female offspring to experience heightened stress during their own pregnancies, facilitating a cycle of stress and adverse neurologic and psychiatric outcomes. GABAergic neurons in the developing hypothalamus and pituitary gland are particularly susceptible to heat stress during pregnancy [204], however little is known about the direct role of heat shock signaling in mediating altered HPA development in prenatally stressed offspring.
The effects of prenatal stress on the formation of overall brain structure and function has also been studied. In humans, structural and functional changes in multiple fore- and midbrain regions have been identified using imaging techniques (see Van den Bergh et al. 2018 for a more extensive review) [192]. The cellular and molecular processes underlying these large-scale changes are not fully understood, but likely involve alterations to neuron migration, microglial activation, and HPA axis development among others. Interestingly, the cellular changes associated with inflammatory and glucocorticoid-mediators of stress have significant links to heat shock signaling [108, 134]. As such, more research is needed to address whether heat shock signaling functions as a protective mechanism against these processes, or instead acts as a critical intermediary of prenatal stress effects on neurodevelopment.
2.2. Heat Shock mechanisms, prenatal stress, and neurodevelopmental processes
Heat shock factors are basic components of cellular function. The proteins induced in the heat shock system, HSPs, are protein chaperones that re-fold denatured proteins in response to high heat, pro-oxidants, and other forms of proteotoxic stress [3]. In the absence of significant stressors, HSPs play a role in normal protein synthesis and maintenance to allow for normal processes of cell growth and survival [111]. Developmentally, they are particularly important during periods of high cell growth due to the increased demand for proteins in synthesizing cellular structures [41]. HSPs also help mitigate harmful intracellular protein aggregation [44].
Importantly, HSPs play significant roles in neurodevelopmental processes. For instance, they aid in supporting somal growth, cell migration, and outgrowth of axons and dendrites [129]. Apoptosis, which is an imperative, controlled process during embryonic brain development, is dependent on HSPs through the ubiquitin proteasome system [98]. The relatively low oxygen levels of the embryonic environment may also induce the HSR, which in turn may promote angiogenesis necessary for oxygenation of the brain [89]. For a review of the neurodevelopmental roles of heat shock systems, see [130].
Given the important role of heat shock factors in the cellular stress response and their additional roles in facilitating neurodevelopmental processes, HSPs are likely involved in the mechanisms by which prenatal stress alters embryonic neurodevelopment. Alterations in protein folding and stability can dramatically change many of the cellular processes that are critical for proper embryonic brain development, many of which are affected by prenatal stress. Possible effects of prenatal stress involving protein stability include permanent changes in neuronal and glial number, location and growth of neurons and glia, extension of neural processes that underlie functional circuits, mitochondrial functioning, and the epigenome [1]. Maternal prenatal stress could theoretically alter embryonic brain proteins in this way, and the protective effects of the HSR could play a corrective role, although there are currently no studies of prenatal stress that demonstrate this. For example, induction of the HSR in the embryonic brain in response to prenatal stress could aid in restoring homeostasis, as seen for the protection of differentiating neurons after arsenite exposure by conditioning heat shock [201] allowing for normal development to progress. The protection of embryonic brain by HSR is also demonstrated by greater vulnerability of cortical neuron production to maternal mercury, alcohol, or seizure exposure in mice lacking heat shock factor 1 (HSF1) [74]. However, given the critical roles of HSPs in facilitating neurodevelopmental processes, HSR induction by prenatal stress may alternatively induce significant changes the developmental trajectory of the brain. For example, HSF1 and HSF2 impair normal neuronal radial migration gene transcription and post-translational modification in the setting of prenatal alcohol exposure [52]; inhibitory neuron tangential migration, impaired after prenatal stress [180], involves the same genes (e.g. Cdk5) which may indicate a role for HSF1 and HSF2 in prenatal stress. In either case, the HSR may, in part, provide an explanation for why some individuals are resilient against the effects of prenatal stress, while others are more susceptible.
In fact, very little is understood about how maternal stress physiology affects heat shock mechanisms of the developing brain. The classic stimulus for HSR, thermal changes, have yet to be studied in the embryonic brain as a consequence of maternal psychosocial stress. If such changes were to occur, the induction of HSPs may underlie interesting susceptibilities to psychiatric illness. Indeed, almost 200 genes, including those for HSPs and those associated with ASD and schizophrenia, are altered when neuronal aggregates reflecting embryonic stages of development from human inducible pluripotent stem cells are exposed to high temperature [115]. Regardless of whether thermal changes occur, proteostasis and proteotoxicity have been neglected outcomes in the study of any offspring effects after exposure to prenatal stress. While no changes in HSP25, 32, or 70 are seen in mouse offspring brain after maternal exposure to either restraint stress or radiofrequency fields [58], many other questions remain about other aspects of the HSR and prenatal stress, including whether specific developing cells may be more susceptible based on their stage (e.g. proliferative, differentiating) or subtype (e.g. forebrain or hindbrain, glial or neuron).
Despite limited evidence of heat shock systems’ involvement with prenatal stress effects on embryonic brain, the mechanisms broadly implicated in prenatal stress shed some light on how heat shock systems may be involved. For example, HSP90 may be essential for response of the embryonic brain to GR activation, which we describe in detail later in this review. Embryonic brain GR activation may occur as a consequence of prenatal stress [158] and is critical to HPA axis maturation which is influenced by prenatal stress. While this may implicate a negative impact of HSPs in stress effects on HPA axis maturation and other neurodevelopmental effects, HSP90 and other HSPs may also mitigate physiological stress on cells, which could alternatively be a critical protection for developing neurons. As we will describe later in this review, HSPs may protect cells in pro-inflammatory conditions, a likely result of maternal stress. In fact, Torii and colleagues demonstrated that a pro-inflammatory maternal immune activation protocol in mice activated a heat shock reporter in 25–30% of offspring brains [188].
Because of the nutritional requirements for protein homeostasis, impacts of maternal stress on caloric intake may also impact heat shock systems. For instance, intrauterine growth restriction in rats due to caloric restriction is associated with increased HSP90 [9] and maternal hyperglycemia in mice activates the HSR [188], both in offspring brain. Hypoglycemic conditions may alter expression of factors involved in heat shock in the embryonic brain, however, there is some discrepancy whether and how nutrient levels regulate HSP expression. One study found that low glucose levels induce Hsp7 gene expression in mouse neuroblastoma cell lines by upregulating HSF-1 activity [175]. On the other hand, other studies have shown that high glucose and amino acid levels upregulate HSF-1 through mTOR-mediated phosphorylation, suggesting that nutrient deprivation to the fetal brain may actually lead to negative HSF1 regulation [42]. In support of the latter, amino acid deprivation had been demonstrated to inhibit the DNA binding activity of HSF-1 and downregulate heat shock element (HSE)-controlled genes [79].
In addition to maternal psychological stress, prenatal exposure to environmental chemicals may alter neurodevelopment in children through similar mechanisms involving heat shock signaling. For example, prenatal exposure to sodium arsenite and cadmium have been demonstrated to induce HSP accumulation in the neuroepithelial tissue of mouse embryos [81]. In this regard, it is unclear whether HSR induction occurs as a protective mechanism or if heat shock signaling may play a significant role in mediating the adverse effects of environmental chemical exposures.
In support the theory that the HSR offers protection against the toxicity of environmental chemicals, ethanol and methylmercury at doses below the threshold for acute toxicity activate HSF1 in the murine embryonic cerebral cortex [74]. Interestingly, knockout of HSF1 in addition to ethanol and methylmercury exposure significantly increases cortical dysplasia. Furthermore, reintroduction of exogenous HSP70 protein to HSF1 knockout mice rescued the phenotype observed in response to ethanol and methylmercury treatment, suggesting that HSR activation in embryonic brain protects against prenatal toxicant exposure [74]. Interestingly, the same study demonstrated that neural progenitor cells generated from patients with schizophrenia exhibited significant increases in cell-to-cell variation in heat shock response to ethanol and methylmercury [74]. As such, genetic mutations leading to altered heat shock response may increase the vulnerability of neural progenitors to environmental insults, thus contributing to the development of schizophrenia.
In addition to their roles in protecting the embryonic brain from stress, HSPs also play crucial roles in the regulation of neurodevelopment as discussed previously, rendering them a possible target for toxicant-induced neuronal dysfunction. For example, prenatal exposure to polychlorinated biphenyls (PCBs) in zebrafish induce significant reductions in neuronal levels of HSC70, a member of the heat-shock protein 70 family, alongside significant reductions in neuronal serotonin [102]. HSC70 is constitutively expressed in neuronal synapses and may play a role in synaptic plasticity and vesicle recycling [139, 191]. As such, HSP70’s role in synaptic vesicle cycling may underlie the depletion of neuronal serotonin in the embryonic brain with PCB exposure [102]. Ultimately, empirical evidence linking prenatal stress and the HSR is clearly lacking. However, what is known about embryonic brain outcomes and physiological mechanisms of prenatal stress implicates heat shock systems. In particular, the HSR to oxidative stress is an important consideration, given the significant role oxidative stress may play in mediating the effects of prenatal stress on neurodevelopment [116].
2.3. Oxidative stress mechanisms, prenatal stress, and neurodevelopmental processes
The impact of maternal stress on developmental processes in the embryonic brain may result from molecular mechanisms in response to cellular stress. Oxidative stress results from a range of adverse exposures and physiological changes and is of particular interest due to its close relationship with heat shock signaling. The chief mediators of oxidative stress are reactive oxygen species (ROS), generated largely through the processes of oxidative phosphorylation and aerobic metabolism. ROS are also generated in significant amounts through reactions catalyzed by oxidoreductase enzymes, non-enzymatic reactions catalyzed by endogenous metal ions, and by neutrophils and macrophages via NADPH oxidase. Members of the ROS family include the superoxide anion (O2−), hydroxyl radicals (OH•), hydrogen peroxide (H2O2), and lipid peroxides (LOOH). Excess generation of ROS can result in cellular damage in the forms of lipid peroxidation, protein misfolding, and DNA damage, all of which may significantly interfere with cellular functions. Furthermore, high levels of ROS induce the opening of the mitochondrial permeability transition pore (MPTP), leading to cell death via apoptosis [162, 167].
Importantly, the embryonic brain is particularly susceptible to the effects of oxidative stress, as expression of antioxidant enzymes are relatively low in embryonic brain and the cellular environment has relatively high concentrations of catalytic metals such as Fe2+ and Cu+ that promote the production of hydroxyl radicals from hydrogen peroxide. Furthermore, neural tissue has a relatively high abundance of membrane polyunsaturated fatty acids, which can be readily oxidized by ROS to produce highly reactive byproducts such as 4-hydroxynonenal (4-HNE) and malondialdehyde (MDA), which form stable adducts to protein and DNA nucleophiles [85, 172].
Maintaining cellular redox balance is critical for proper brain development, as both the over- and underproduction of ROS can negatively impact cellular functioning and survival [39]. In developing neurons, ROS play a regulatory role in multiple important cellular pathways. For instance, ROS are implicated in the activation of nuclear factor erythroid-derived 2-like 2 (NRF2) and Redox factor-1 (REF-1) signaling [159]. Both of these pathways are important for astrocyte survival and function, with NRF2 inducing cytoprotective repair mechanisms and REF-1 playing a critical role in controlling inflammation [12, 18]. Additionally, ROS regulate the transcription of genes involved in inflammation, cell proliferation, cell adhesion, and angiogenesis through activation of redox-sensitive transcription factors such as the nuclear factor NF-κB and AP-1 [78].
Neural stem cell proliferation and differentiation also both rely on ROS-mediated processes. For example, NAPDH oxidase 2 (NOX2), an enzyme that generates ROS, regulates neuronal stem cell development by mediating key transcription factor activity [138]. Furthermore, ROS mediate the mitogen-activated protein kinase (MAPK) and phosphoinositide 3-kinase (PI3K) pathways, both which play a role in neural stem cell proliferation [11]. In addition, activation of epidermal growth factor (EGF) receptors by H2O2 plays a critical role in promoting neural progenitor proliferation [86].
ROS levels are also specifically maintained for cell fate determination. Higher levels of intracellular ROS are present in cells that differentiate into neurons, while lower levels are present in those with a glial lineage [189]. As such, the significant roles that ROS play in key neurodevelopmental processes demonstrates how disruptions to the oxidative state of the embryonic brain may disrupt its formation and long-term functioning.
Mediators of the maternal stress response, such as increased inflammation or cortisol production, impact the redox balance in the embryonic environment. In this respect, GSH levels in the embryonic brain decrease in response to both maternal inflammation and restraint stress [105, 120]. Prenatal stress also increases lipid peroxidation in offspring postnatal brain, suggesting that maternal stress during pregnancy may have long-lasting effects on brain redox balance [25].
Recent evidence suggests that ROS play significant roles in mediating the effects of maternal stress on specific neurodevelopmental processes. For example, increased ROS [28] and blockade of mitochondrial oxidative phosphorylation disrupt GABAergic interneuron tangential migration [114] in a similar manner to that seen with maternal restraint stress [180]. The impact of prenatal stress on offspring HPA axis development may result from the susceptibility of embryonic hypothalamic neuronal and microglia progenitors together to redox shifts, as has been shown in response to fetal alcohol exposure [33]. Additionally, application of the antioxidants N-acetylcysteine (NAC) and astaxanthin both block changes in neuronal migration and microglial activation seen with prenatal stress, indicating that oxidative stress is a requirement for the observed phenotypes [28, 29]. Administration of the antioxidant NAC has also been shown to protect against learning and memory deficits in prenatally stressed mice [24].
Taken together, the aforementioned studies support ROS-mediated mechanisms in the effects of prenatal stress on brain growth and development. Furthermore, the efficacy of antioxidants such as NAC in negating the effects of prenatal stress indicate that buffering against oxidative stress may be a potential intervention for preventing adverse neurodevelopmental outcomes in children.
3. Relationships between heat shock and oxidative stress mechanisms
Given the role that HSPs play in protecting cells under stressed conditions, it is not surprising that a significant relationship exists between oxidative stress and heat shock systems. Similar to conditions of heat stress, increased levels of ROS induce protein unfolding through a variety of posttranslational protein modifications. Under conditions of oxidative stress, the most vulnerable protein targets are the sulfur-containing side chains of cysteine and methionine; however other side chains such as histidine, tryptophan, lysine, tyrosine, and arginine can be readily oxidized [23]. These modifications can lead to the loss of a protein’s secondary and tertiary structure, which in turn reduces its solubility, stability, and activity [23]. To protect cells from damage caused by misfolded proteins, heat shock factors demonstrate redox sensitive activation through a variety of mechanisms, including direct induction of heat shock factors by reactive electrophiles, transcriptional regulation via the antioxidant response elements, and production of HSPs as a consequence of ROS-mediated protein unfolding [2].
Redox-mediated activation of heat shock factors
Heat shock factor 1 (HSF1) functions as a sensor for the unfolded protein response and is responsible for the transcription of various HSPs. Under non-stressed conditions, HSF1 monomers are sequestered in the cytoplasm by the HSP90 multichaperone complex consisting of HSP90, HSP40, and HSP70. During cellular stress, increased levels of unfolded proteins compete for binding with these chaperones, leading to the release of HSF1. Upon its release, HSF1 translocates to the nucleus where it trimerizes and binds heat shock elements HSE to induce HSP transcription [132]. As such, redox-mediated activation of HSF1 can be achieved indirectly through an increase in misfolded proteins due to oxidative damage. HSF1 may also be transcriptionally upregulated by oxidative stress via NRF2 interaction with antioxidant response elements (AREs) located upstream of the Hsf1 gene transcription start site [151].
Direct mechanisms of HSF1 activation by oxidative stress have been postulated to be similar to those of the redox sensitive transcription factor NRF2, in which HSF1 is activated via thiol oxidation or sglutathiolation. For example, H2O2 directly activates HSF1 through oxidation of cysteine residues 35 and 105, which upon disulfide bridge formation promote HSF1 trimerization and HSE binding [2, 117]. Endogenous electrophiles produced as a consequence of oxidative stress, such as alpha-beta-unsaturated aldehydes like 4-HNE, also activate HSF1 through similar mechanisms involving the oxidation of certain amino acid residues. In addition, HSF1 activation may be induced indirectly by nitric oxide, which inactivates HSP90 through s-nitrosylation of cysteine 597, in turn promoting the dissociation of HSF1 from the HSP90 chaperone complex [123].
Protective effects of heat shock proteins against oxidative stress
In regard to the antioxidant effects of heat shock proteins, several unique modes of action have been proposed. For example, HSP27 contributes to intracellular glutathione reduction by increasing the activity of glutathione reductase (GSR). This increase in activity may result from the stabilizing effect that HSP27 chaperones have on the enzyme G6PDH, which leads to increased levels of the GSR cofactor NADPH. Additionally, although the underlying mechanism remains unclear, HSP27 reduces intracellular iron levels, further protecting cellular structures from oxidative stress associated with Fe2+-mediated generation of hydroxyl radicals [10].
In addition to the direct antioxidant effects of HSPs, protection from oxidative stress is also accomplished through the stabilization or degradation of misfolded proteins. Similar to other heat shock proteins, HSP70 identifies misfolded proteins by recognizing exposed hydrophobic residues and utilizing ATP-dependent mechanisms to refold them. In addition, co-chaperones such as Bag-1 and CHIP can modify the effects of HSPs on misfolded proteins, allowing for protein degradation in cases of significant damage. For example, CHIP inhibits the ATPase activity of HSP70 and utilizes its E3 ubiquitin ligase activity to ubiquitinate oxidized proteins and direct them to the proteasome for degradation [45].
HSPs also play a protective role in oxidative stress-induced apoptosis through interactions with apoptotic proteins. For example, HSP70 and HSP90 bind Apaf-1 and inhibit its downstream apoptotic effects [164]. In addition, HSP27 inhibits cytochrome C-mediated apoptosis, either through direct binding to cytochrome C or by inhibiting its release from mitochondria [69, 147]. Heat shock proteins also counteract activation of inflammatory pathways following oxidative stress. For instance, HSP70 suppresses both the upregulation of Cox-2 and the production of nitric oxide in response to oxidative stress by inhibiting NF-κB gene activation [76, 196].
In sum, HSPs are activated by redox status both directly and indirectly and may protect from oxidative stress via antioxidant properties, correction of misfolded proteins, and their anti-apoptotic effects in the context of cellular stress. With growing appreciation for the role of oxidative stress in prenatal stress effects on offspring brain [28], HSP activation in developing neural cells may be similarly implicated.
4. Mechanisms of prenatal stress outside of the developing brain
4.1. Maternal hormonal and inflammatory factors in prenatal stress
While embryonic brain is a potential site of heat shock mechanisms in prenatal stress effects, HSPs and oxidative stress also have a role in the complex physiology of maternal stress factors that constitute a major waypoint in prenatal stress effects. Glucocorticoids in maternal circulation are critically elevated with prenatal stress [166, 170, 200], and there is considerable interest in the complex crosstalk between glucocorticoid and heat shock signaling pathways. GR-mediated gene expression may occur in multiple maternal tissues and is significantly enhanced by HSF-1, suggesting a potential for synergy between these two pathways in response to stress [91]. Consistent with this notion, HSP70 and HSP90 facilitate the assembly of GR heterocomplexes and ensure proper conformation for steroid binding [48, 133]. Additionally, HSF1 upregulates FK506-binding protein-52, which helps translocate GR to the nucleus following ligand binding [91]. On the other hand, GR activation inhibits the transcriptional activity of HSF1 in response to both heat and chemical shock by preventing the binding of HSF1 to Hsp promotors [109, 110]. Evidently, these two pathways exhibit reciprocal regulation, wherein heat shock proteins act to potentiate the effects of the GR under low-glucocorticoid concentration conditions while high concentrations of glucocorticoids act to suppress the heat shock response [108]. Furthermore, maternal cortisol release depends on maternal adrenal redox signaling [6] and may be influenced by the response of HSPs to pro-oxidants. Lastly, GR activation suppresses NRF2 transcriptional activity [4], one of the major mediators of endogenous antioxidant upregulation. Therefore, influences of maternal glucocorticoids during prenatal stress that ultimately affect the embryonic brain may be intimately affected by heat shock responses and may influence oxidative stress response. These may involve the interaction of GR activation, HSR, and antioxidants in other maternal cells that have a significant role to play in the cascade of prenatal stress effects (e.g. immune cells, maternal brain, vasculature). Consequences may also result from direct passage of maternal glucocorticoids to the embryonic brain or through effects of glucocorticoids on the placenta, as discussed in detail in the next section.
Psychological stress is also associated with immune activation and increased levels of pro-inflammatory cytokines such as TNF-α and IL-6 [47]. Furthermore, both stress and inflammation can result in offspring with alterations in brain function [136]. The immune component of maternal physiology during stress may depend on HSR systems. Maternal immune cells may be activated by extracellular HSPs and use heat shock proteins and redox signaling intracellularly in the process of cytokine release [206].
Preeclampsia, a pregnancy disorder characterized by hypertension and proteinuria, is associated with adverse neurodevelopmental outcomes in offspring [141] like maternal psychological stress. In women with preeclampsia, heat shock systems have been highly implicated [83, 144, 163]. Screenings aimed at identifying markers associated with preeclampsia have identified differential expression of HSPs in maternal blood as a common characteristic among patients [82]. In mothers with pregnancies complicated by preeclampsia, circulating levels of HSP70 are significantly correlated with increased pro-inflammatory cytokines, further confirming the link between heat shock systems and inflammation [131].
Overall, maternal factors involved in prenatal stress effects on the developing brain may be both a product of and result in heat shock and oxidative stress. The role for these cellular stress systems in the maternal response to psychological stress suggests more potential for translatable interventions, given the more immediate access of maternal physiology to intervention. However, maternal physiology cannot be considered in isolation; its close partner in supporting embryonic development, the placenta, must be considered in translational efforts as well.
4.2. Placenta as a mediator of prenatal stress
The placenta’s role in regulating maternal-fetal interactions makes it a prime target for understanding the mechanisms by which maternal stress alters neurodevelopment, including the role of HSPs and oxidative stress. The placenta has numerous functions, including the transfer of nutrients and oxygen to the fetus, protection of the fetus from maternal factors, and the secretion of growth factors and hormones that regulate fetal growth [63]. Consistent with these roles, maternal stress is associated with reduced birth weight and altered nutrient transfer, suggesting that altered placental functioning may underlie the mechanisms responsible for altered neurodevelopment [34, 121, 140]. Furthermore, given that the placenta consists of cells of both embryonic and maternal origin, sex differences in placental function are present throughout gestation, mediating sex-specific responses to maternal stress [13, 161].
Maternal stress has been associated with increased placental production of proinflammatory cytokines, which may upregulate heat shock protein expression in the embryonic brain. For example, rodent models of prenatal stress have demonstrated significant increases in the placental production of IL-6, IL-1β, and TNF-α [35, 135]. Placental production of cytokines in response to stress has been directly linked to altered neurodevelopment, as demonstrated by the protective effects of anti-inflammatory treatments on offspring neurodevelopment [35]. Consistent with this observation, proinflammatory cytokines have been demonstrated to upregulate the expression of placental HSPs [17, 155]. Placental regulation of embryonic inflammation, therefore, may depend on HSPs. Oskvig and colleagues demonstrated that increased maternal cytokine levels after lipopolysaccharide (LPS) administration, a model of bacterial infection, did not change expression of inflammatory mediators such as IL-6, IL-1B, and TNF-a in the embryonic brain. Furthermore, cytokine level changes in this model diminished in magnitude from maternal serum to amniotic fluid to embryonic brain, indicating that maternal cytokine fluctuations are buffered, perhaps due to placental actions [142] that may be influenced by the HSR.
Active placental mechanisms maintain amniotic and fetal cortisol levels lower than maternal cortisol [21, 65]. 11β-Hydroxysteroid dehydrogenases (11β-HSD) regulate glucocorticoids by converting inactive cortisone into cortisol and vice versa. The principal isozyme expressed in the placenta is 11β-HSD2, which serves to protect the embryo from maternal fluctuations in cortisol [5, 22, 37, 182, 202]. Placental 11β-HSD2 expression and activity, which is an oxidative process, increases with gestational age [168, 171]. Importantly, IL-1β and TNF-α, which increase with maternal stress, appear to contribute to the regulation of 11β-HSD proteins [46, 53, 122]. While these pro-inflammatory cytokines augment the expression and reductase activity of 11β-HSD1 [46, 53], they negatively regulate the expression and activity of 11β-HSD2 in a dose-dependent manner [46]. Maternal anxiety, depression, and psychological stress in humans, as well as chronic maternal stress in rodents, have been demonstrated to reduce placental expression and activity of 11β-HSD2, in turn leading to fetal glucocorticoid overexposure [121, 148, 153]. On the other hand, maternal exposure to cold temperature in rats increases placental 11β-HSD2 expression [113], which is associated with placental HSP70 production, suggesting that placental HSR may regulate fetal glucocorticoid exposure.
Maternal stress is also associated with restricted umbilical artery blood flow, which can impair the transfer of flow-limited substrates such as oxygen and carbon dioxide between maternal and fetal circulation [137, 176]. Stress-induced elevations in maternal serotonin, a vasoconstrictor, may contribute to reduced blood flow [31, 32]. Placental hypoxia promotes oxidative stress and is linked to adverse neurodevelopmental effects seen in animal models of prenatal stress, such as abnormal neuronal migration and reduced myelination [16, 104]. Hypoxia in late pregnancy can affect signaling molecules in the fetal hippocampus, resulting in impaired neuronal migration, similar to the migration delays seen with prenatal stress or exposure to pro-inflammatory cytokines and glucocorticoids [68, 181]. Placental hypoxia during stress stimulates the release of prostanoids and cytokines, including IL-1β and TNF-α [122, 160], but depending on developmental timing, may alternatively cause no change to placental cytokine production [68]. Consistent with the role of HSPs in mitigating oxidative stress, Hsp70 is upregulated alongside biomarkers of oxidative stress in placental tissues of patients with preeclampsia [15, 80, 143]. In addition, upregulation of Hsp27 in the placentas of preeclamptic patients has also been noted in several studies [64, 174]. The links between hypoxia, inflammation, the HSR, and oxidative stress as outlined in previous sections suggest a significant role for targeting these cellular mechanisms in the placenta to protect against prenatal stress which may be similar to these demonstrated effects of hypoxia and preeclampsia.
In addition to reduced overall placental blood flow, restrictions to glucose, lipid, and amino acid transfer have been observed in animal models of prenatal stress [121]. For example, altered activity of the placental glucose transporter GLUT-1 is found in both animal models of prenatal stress and humans [135]. Glutamate transporters in other tissues are induced through the HSR [70]. Thus, placental nutrient transporters may also be linked to the actions of HSPs. The impacts of prenatal stress also may precipitate through changes in placental oxygenation and oxidative state, both of which have demonstrated impacts on placental nutrient transport [8].
The placenta also has critical endocrine functions that participate in fetal neurodevelopment. In this regard, maternal stress alters placental production of neurotransmitters, growth hormones, neurotrophins, and cytokines, all of which have critical neurodevelopmental functions. The production of these diffusible factors—the placental secretome—depends on the viability and functioning of trophoblasts and other cells, which can be disrupted through oxidative stress and HSR mechanisms. The placental secretome also has direct impacts on embryonic brain where the HSR and ROS mediate responses. For example, maternal IL-1β decreases placental production of BDNF specifically in female offspring of rodent models of prenatal stress [73]. Given that previous studies have demonstrated a suppressive role of BNDF in the stress-induced upregulation of embryonic brain HSPs, decreased placental BDNF may enhance the heat shock response to hypoxia in fetal brain [103]. Animal models of prenatal stress have also demonstrated alterations in placental production of IGF-1 and IGF-2, which are crucial regulators of fetal growth and neurodevelopment [195]. In addition to its role in regulating fetal growth, IGF signaling has also been implicated in regulating expression of heat shock factors through an mTOR-mediated mechanism [42].
Maternal stress is also associated with increased serotonin, norepinephrine, and dopamine in maternal circulation [90]. These increases in circulating neurotransmitters are coupled with reduced MAO-A and increased serotonin transporters in the placentas of stressed mothers [30, 153]. Placental serotonin processing may be a critical mediator of maternal physiological impacts on embryonic brain [67, 194] and may be influenced by cellular stress mechanisms such as HSR and ROS. Serotonin can trigger the HSR in C. elegans via a temperature independent mechanism in some cell types [185] which may also occur in placenta and/or embryonic brain.
5. Heat shock systems, downstream mechanisms, and neuropsychiatric disorders
While it is unclear whether there is a direct connection between prenatal stress, the HSR, and eventual psychopathology in offspring, HSPs have been implicated in the etiology of several psychiatric disorders [19, 20, 40, 95, 97, 101, 107, 146, 169]. As previously discussed, HSPs are anti-inflammatory, anti-apoptotic, and attenuate the effects of oxidative stress [55, 203]. The HSR, perhaps because of elevated HSP70, prevents activation of NF-κB and downstream inflammatory response [77]. Indeed, hyperthermia-induced HSR protects against inflammation caused by combined injection of LPS, IL-1β, and IFNγ in mice [77]. NF-κB is also a mediator of oxidative stress, and induced HSP70 attenuates the effects of this system by reducing NF-κB translocation to the nucleus [55]. These links suggest that HSPs may have a role as biomarkers for these other contributing mechanisms.
Important to psychiatric disorders, the HSR causes nuclear translocation of GRs [165], and HSPs play a critical role in GR folding, transport, and function, as previously described. For this reason, HSP dysfunction is implicated in the etiology of many stress-related psychopathologies, particularly affective disorders.
5.1. Affective disorders
A higher risk for bipolar disorder is associated with prenatal stress [100]. Peripheral blood lymphocytes in individuals with bipolar disorder have lower HSP70 and high GR-HSP70 heterocomplexes; these same cells had low cytosolic BAX, suggesting a susceptiblility to apoptosis [19, 20]. In addition, Cheng and colleagues found elevated serum HSP60 and HSP70 in a cohort of hospitalized patients with bipolar disorder [40]. Gene expression for HSP40, which interacts with HSP70, is also elevated in brain and peripheral blood cells of individuals with bipolar disorder [87]. An integrated transcriptome and methylome analysis of peripheral blood cells also implicated HSPA1L (HS70 protein-1 like) in youth with or at risk for bipolar disorder [59].
Major depressive disorder is associated with prenatal stress [156] and HSP70 abnormalities [173]. Pae and colleagues found that HSP70 single nucleotide polymorphisms are associated with worse symptomology and poorer response to antidepressant treatment [146], suggesting lower HSP70 expression and less induction of HSP70 upon treatment. However, higher baseline serum HSP70 in women predicted the development of depression over a three year period [150]. In mice, knockout of HSF1, which may affect HSP70 induction, results in depressive-like phenotypes [190]. There is significant interest in further understanding the role of HSPs in the stress susceptibility that contributes to bipolar and major depressive illness.
A particularly well-studied HSR protein that may play a role in stress susceptibility and neuropsychiatric disorders is FK506 binding protein 51 (FKPB5). FKBP5 is a co-chaperone of HSP90, which regulates GR sensitivity. Others have reviewed in detail how dysregulation of FKBP5 may disrupt normal cortisol signaling, contributing to the development of affective disorders [27, 125]. In fact, a meta-analysis found that three specific polymorphisms in the FKBP5 gene confer susceptibility to early life stress, causing an increased risk for developing PTSD or depression in response to stressful events [197]. This suggests that early life stressors (including prenatal stress) and genetic predisposition may work synergistically to promote mood and anxiety disorder development [125].
5.2. Schizophrenia
Prenatal stress contributes to risk for schizophrenia [56], and the pathophysiology of schizophrenia may result from reduced protective capacity of HSPs during inflammatory and oxidative challenges to neurodevelopment. Consistent with this, antibodies to HSP70 and HSP90, which may disrupt their function, are elevated in individuals with schizophrenia [96, 169]. Polymorphisms in the HSP70 gene are also associated with schizophrenia in multiple populations of different ethnic backgrounds [97, 101, 145]. HSPA12A, a member of the HSP70 family expressed highly in neurons, particularly in the frontal cortex, is significantly reduced in the dorsolateral prefrontal cortex in individuals with schizophrenia [154]. Interestingly, induced pluripotent stem cells derived from patients with schizophrenia show greater variability in Hsp70 transcription when exposed to methylmercury or ethanol [74] which may suggest dysfunctional upstream regulation in response to stress, perhaps by HSF1. As previously described, HSF1 is an upstream regulator of the HSR and HSP70 in particular [205]. Environmental insults including ethanol, methylmercury, and induced seizure exposure increase HSF1 binding to the Hsp70 promoter in rodent embryonic cortex and human fetal cortical cells [74]. HSF1 knockout disrupts normal cortical development and increases vulnerability to seizures from these exposures, most likely due to cell cycle arrest or premature exit, as well as increased apoptotic activity [74]. These mechanisms may contribute to the risk developing schizophrenia from prenatal adversity.
5.3. Childhood disorders
ASD and ADHD are child onset neuropsychiatric disorders with links to prenatal stress [1]. However, few studies have examined the heat shock system in peripheral tissues, genetics, or postmortem brain of individuals with ADHD or ASD. One finding that does address similar links to those found in schizophrenia is that antibodies to HSP90 are observed in some children with ASD [54]. Additionally, HSP70 is also elevated in some children with ASD, similar to individuals with mood disorders [51]. Deeper investigation of the links between heat shock systems and these early onset disorders may yield not only important biomarkers but also potential pathophysiological mechanisms that could be targeted in treatment development.
6. Conclusion
Intrinsic biological responses that protect against cellular stress are prime candidates for elucidating both how adversity impacts prenatal brain development and how resultant disruptions could be prevented. These include cellular responses to oxidative stress, notably heat shock systems. As a result, HSPs and antioxidants are targets for drug development to treat diseases of the brain and many other systems. Interestingly, dysregulation of heat shock and redox regulation systems may contribute to the incidence of neuropsychiatric disease. On the other hand, heat shock and oxidative stress may also be indirectly induced during pathophysiological processes because of their important connections with multiple cellular functions. This latter possibility highlights how these cellular mechanisms have the capacity to protect the brain. To consider how targeting HSPs and antioxidants could prevent neurodevelopmental disruptions of prenatal stress, substantial progress is necessary in understanding the role of heat shock systems in specific cell subtypes and processes in embryonic brain. In addition, the complexities of the interacting systems in prenatal brain development – from maternal physiology to placenta function to embryonic growth – also require coordinated discovery of the distinct roles of the heat shock and oxidative stress responses in each system.
Highlights.
Prenatal maternal stress increases risk for neuropsychiatric disorders in offspring
Heat shock factor and oxidative stress mechanisms are implicated in prenatal stress
These cellular stress mechanisms may be targets for neuropsychiatric interventions
Acknowledgements
The authors would like to thank all members of the Stevens lab for their helpful discussion and acknowledge that this work was supported by the following sources: Roy J. Carver Charitible Trust, Ida P. Haller Chair in Child Psychiatry, Nellie Ball Trust, and the National Institute for Environmental Health Sciences through the University of Iowa Environmental Health Sciences Research Center, NIEHS/NIH P30 ES005605.
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- [1].Abbott PW, Gumusoglu SB, Bittle J, Beversdorf DQ, Stevens HE, Prenatal stress and genetic risk: How prenatal stress interacts with genetics to alter risk for psychiatric illness, Psychoneuroendocrinology 90 (2018) 9–21. [DOI] [PubMed] [Google Scholar]
- [2].Ahn SG, Thiele DJ, Redox regulation of mammalian heat shock factor 1 is essential for Hsp gene activation and protection from stress, Genes Dev 17 (2003) 516–528. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [3].Akerfelt M, Morimoto RI, Sistonen L, Heat shock factors: integrators of cell stress, development and lifespan, Nat Rev Mol Cell Biol 11 (2010) 545–555. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [4].Alam MM, Okazaki K, Nguyen LTT, Ota N, Kitamura H, Murakami S, Shima H, Igarashi K, Sekine H, Motohashi H, Glucocorticoid receptor signaling represses the antioxidant response by inhibiting histone acetylation mediated by the transcriptional activator NRF2, The Journal of biological chemistry 292 (2017) 7519–7530. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [5].Alfaidy N, Gupta S, DeMarco C, Caniggia I, Challis JRG, Oxygen Regulation of Placental 11β-Hydroxysteroid Dehydrogenase 2: Physiological and Pathological Implications, J Clin Endocrinol Metab 87 (2002) 4797–4805. [DOI] [PubMed] [Google Scholar]
- [6].Almeida OFX, Condé GL, Crochemore C, Demeneix BA, Fischer D, Hassan AHS, Meyer M, Holsboer F, Michaelidis TM, Subtle shifts in the ratio between pro- and antiapoptotic molecules after activation of corticosteroid receptors decide neuronal fate, The FASEB Journal 14 (2000) 779–790. [DOI] [PubMed] [Google Scholar]
- [7].Ansen-Wilson LJ, Lipinski RJ, Gene-environment interactions in cortical interneuron development and dysfunction: A review of preclinical studies, Neurotoxicology 58 (2017) 120–129. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [8].Araujo JR, Correia-Branco A, Pereira AC, Pinho MJ, Keating E, Martel F, Oxidative stress decreases uptake of neutral amino acids in a human placental cell line (BeWo cells), Reprod Toxicol 40 (2013) 76–81. [DOI] [PubMed] [Google Scholar]
- [9].Aravidou E, Eleftheriades M, Malamitsi-Puchner A, Anagnostopoulos AK, Aravantinos L, Dontas I, Aravidis C, Creatsas G, Tsangaris G, Chrousos GP, Protein expression in the brain of rat offspring in relation to prenatal caloric restriction, J Matern Fetal Neonatal Med 29 (2016) 2707–2714. [DOI] [PubMed] [Google Scholar]
- [10].Arrigo AP, Virot S, Chaufour S, Firdaus W, Kretz-Remy C, Diaz-Latoud C, Hsp27 consolidates intracellular redox homeostasis by upholding glutathione in its reduced form and by decreasing iron intracellular levels, Antioxid Redox Signal 7 (2005) 414–422. [DOI] [PubMed] [Google Scholar]
- [11].Bae YS, Oh H, Rhee SG, Yoo YD, Regulation of reactive oxygen species generation in cell signaling, Mol Cells 32 (2011) 491–509. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [12].Baek H, Lim CS, Byun HS, Cho HS, Lee YR, Shin YS, Kim HW, Jeon BH, Kim DW, Hong J, Hur GM, Park JB, The anti-inflammatory role of extranuclear apurinic/apyrimidinic endonuclease 1/redox effector factor-1 in reactive astrocytes, Mol Brain 9 (2016) 99. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [13].Bale TL, The placenta and neurodevelopment: sex differences in prenatal vulnerability, Dialogues in clinical neuroscience 18 (2016) 459–464. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [14].Barker DJ, Fetal origins of coronary heart disease, Br Heart J 69 (1993) 195–196. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [15].Barut F, Barut A, Dogan Gun B, Kandemir NO, Aktunc E, Harma M, Harma MI, Ozdamar SO, Expression of heat shock protein 70 and endothelial nitric oxide synthase in placental tissue of preeclamptic and intrauterine growth-restricted pregnancies, Pathology, research and practice 206 (2010) 651–656. [DOI] [PubMed] [Google Scholar]
- [16].Basilious A, Yager J, Fehlings MG, Neurological outcomes of animal models of uterine artery ligation and relevance to human intrauterine growth restriction: a systematic review, 57 (2015) 420–430. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [17].Bausero MA, Gastpar R, Multhoff G, Asea A, Alternative mechanism by which IFN-gamma enhances tumor recognition: active release of heat shock protein 72, Journal of immunology (Baltimore, Md. : 1950) 175 (2005) 2900–2912. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [18].Baxter PS, Hardingham GE, Adaptive regulation of the brain’s antioxidant defences by neurons and astrocytes, Free Radic Biol Med 100 (2016) 147–152. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [19].Bei E, Salpeas V, Pappa D, Anagnostara C, Alevizos V, Moutsatsou P, Phosphorylation status of glucocorticoid receptor, heat shock protein 70, cytochrome c and Bax in lymphocytes of euthymic, depressed and manic bipolar patients, Psychoneuroendocrinology 34 (2009) 1162–1175. [DOI] [PubMed] [Google Scholar]
- [20].Bei ES, Salpeas V, Alevizos B, Anagnostara C, Pappa D, Moutsatsou P, Pattern of heat shock factor and heat shock protein expression in lymphocytes of bipolar patients: Increased HSP70-glucocorticoid receptor heterocomplex, Journal of Psychiatric Research 47 (2013) 1725–1736. [DOI] [PubMed] [Google Scholar]
- [21].Beitins IZ, Bayard F, Ances IG, Kowarski A, Migeon CJ, The Metabolic Clearance Rate, Blood Production, Interconversion and Transplacental Passage of Cortisol and Cortisone in Pregnancy Near Term, Pediatr. Res 7 (1973) 509–519. [DOI] [PubMed] [Google Scholar]
- [22].Benediktsson R, Calder AA, Edwards CRW, Seckl JR, Placental 11β-hydroxysteroid dehydrogenase: a key regulator of fetal glucocorticoid exposure, Clinical Endocrinology 46 (1997) 161–166. [DOI] [PubMed] [Google Scholar]
- [23].Berlett BS, Stadtman ER, Protein oxidation in aging, disease, and oxidative stress, The Journal of biological chemistry 272 (1997) 20313–20316. [DOI] [PubMed] [Google Scholar]
- [24].Bernhardt LK, Bairy KL, Madhyastha S, Neuroprotective Role of N-acetylcysteine against Learning Deficits and Altered Brain Neurotransmitters in Rat Pups Subjected to Prenatal Stress, Brain Sci 8 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [25].Bernhardt LK, Madhyastha S, Bairy L, Kishore A, Status of the brain antioxidant system at different growing periods after prenatal stress and N -acetyl cysteine administration, Folia Neuropathol 55 (2017) 38–48. [DOI] [PubMed] [Google Scholar]
- [26].Betts KS, Williams GM, Najman JM, Alati R, The Relationship Between Maternal Depressive, Anxious, and Stress Symptoms During Pregnancy and Adult Offspring Behavioral and Emotional Problems, Depression and Anxiety 32 (2015) 82–90. [DOI] [PubMed] [Google Scholar]
- [27].Binder EB, The role of FKBP5, a co-chaperone of the glucocorticoid receptor in the pathogenesis and therapy of affective and anxiety disorders, Psychoneuroendocrinology 34 (2009) S186–S195. [DOI] [PubMed] [Google Scholar]
- [28].Bittle J, Menezes EC, McCormick ML, Spitz DR, Dailey M, Stevens HE, The Role of Redox Dysregulation in the Effects of Prenatal Stress on Embryonic Interneuron Migration, Cerebral cortex (New York, N.Y.: 1991) (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [29].Bittle J, Stevens HE, The role of glucocorticoid, interleukin-1beta, and antioxidants in prenatal stress effects on embryonic microglia, J Neuroinflammation 15 (2018) 44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [30].Blakeley PM, Capron LE, Jensen AB, O’Donnell KJ, Glover V, Maternal prenatal symptoms of depression and down regulation of placental monoamine oxidase A expression, Journal of Psychosomatic Research 75 (2013) 341–345. [DOI] [PubMed] [Google Scholar]
- [31].Bolte AC, van Geijn HP, Dekker GA, Pathophysiology of preeclampsia and the role of serotonin, Eur J Obstet Gynecol Reprod Biol 95 (2001) 12–21. [DOI] [PubMed] [Google Scholar]
- [32].Bonnin A, Levitt P, Fetal, maternal, and placental sources of serotonin and new implications for developmental programming of the brain, Neuroscience 197 (2011) 1–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [33].Boyadjieva NI, Sarkar DK, Microglia play a role in ethanol-induced oxidative stress and apoptosis in developing hypothalamic neurons, Alcohol Clin Exp Res 37 (2013) 252–262. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [34].Brett KE, Ferraro ZM, Yockell-Lelievre J, Gruslin A, Adamo KB, Maternal-fetal nutrient transport in pregnancy pathologies: the role of the placenta, International journal of molecular sciences 15 (2014) 16153–16185. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [35].Bronson SL, Bale TL, Prenatal Stress-Induced Increases in Placental Inflammation and Offspring Hyperactivity Are Male-Specific and Ameliorated by Maternal Antiinflammatory Treatment, Endocrinology 155 (2014) 2635–2646. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [36].Brown AS, van Os J, Driessens C, Hoek HW, Susser ES, Further Evidence of Relation Between Prenatal Famine and Major Affective Disorder, AJP 157 (2000) 190–195. [DOI] [PubMed] [Google Scholar]
- [37].Brown RW, Chapman KE, Edwards CR, Seckl JR, Human placental 11 beta-hydroxysteroid dehydrogenase: evidence for and partial purification of a distinct NAD-dependent isoform, Endocrinology 132 (1993) 2614–2621. [DOI] [PubMed] [Google Scholar]
- [38].Buitelaar JK, Huizink AC, Mulder EJ, de Medina PGR, Visser GHA, Prenatal stress and cognitive development and temperament in infants, Neurobiology of Aging 24 (2003) S53–S60. [DOI] [PubMed] [Google Scholar]
- [39].Castagne V, Relationships between neuronal death and the cellular redox status. Focus on the developing nervous system, Progress in Neurobiology 59 (1999) 397–423. [DOI] [PubMed] [Google Scholar]
- [40].Cheng Y, Li Z, He S, Tian Y, He F, Li W, Elevated heat shock proteins in bipolar disorder patients with hypothalamic pituitary adrenal axis dysfunction, Medicine (Baltimore) 97 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [41].Choe YJ, Park SH, Hassemer T, Korner R, Vincenz-Donnelly L, Hayer-Hartl M, Hartl FU, Failure of RQC machinery causes protein aggregation and proteotoxic stress, Nature 531 (2016) 191–195. [DOI] [PubMed] [Google Scholar]
- [42].Chou SD, Prince T, Gong J, Calderwood SK, mTOR is essential for the proteotoxic stress response, HSF1 activation and heat shock protein synthesis, PLoS One 7 (2012) e39679. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [43].Clarke AS, Wittwer DJ, Abbott DH, Schneider ML, Long-term effects of prenatal stress on HPA axis activity in juvenile rhesus monkeys, Dev Psychobiol 27 (1994) 257–269. [DOI] [PubMed] [Google Scholar]
- [44].Cohen E, Bieschke J, Perciavalle RM, Kelly JW, Dillin A, Opposing activities protect against age-onset proteotoxicity, Science 313 (2006) 1604–1610. [DOI] [PubMed] [Google Scholar]
- [45].Connell P, Ballinger CA, Jiang J, Wu Y, Thompson LJ, Höhfeld J, Patterson C, The co-chaperone CHIP regulates protein triage decisions mediated by heat-shock proteins, Nature Cell Biology 3 (2000) 93. [DOI] [PubMed] [Google Scholar]
- [46].Cooper MS, Bujalska I, Rabbitt E, Walker EA, Bland R, Sheppard MC, Hewison M, Stewart PM, Modulation of 11β-Hydroxysteroid Dehydrogenase Isozymes by Proinflammatory Cytokines in Osteoblasts: An Autocrine Switch from Glucocorticoid Inactivation to Activation, Journal of Bone and Mineral Research 16 (2001) 1037–1044. [DOI] [PubMed] [Google Scholar]
- [47].Coussons-Read ME, Okun ML, Schmitt MP, Giese S, Prenatal stress alters cytokine levels in a manner that may endanger human pregnancy, Psychosom Med 67 (2005) 625–631. [DOI] [PubMed] [Google Scholar]
- [48].Dittmar KD, Pratt WB, Folding of the glucocorticoid receptor by the reconstituted Hsp90-based chaperone machinery. The initial hsp90.p60.hsp70-dependent step is sufficient for creating the steroid binding conformation, The Journal of biological chemistry 272 (1997) 13047–13054. [DOI] [PubMed] [Google Scholar]
- [49].Diz-Chaves Y, Pernia O, Carrero P, Garcia-Segura LM, Prenatal stress causes alterations in the morphology of microglia and the inflammatory response of the hippocampus of adult female mice, J Neuroinflammation 9 (2012) 71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [50].Do KQ, Cabungcal JH, Frank A, Steullet P, Cuenod M, Redox dysregulation, neurodevelopment, and schizophrenia, Curr Opin Neurobiol 19 (2009) 220–230. [DOI] [PubMed] [Google Scholar]
- [51].El-Ansary A, Al-Ayadhi L, Neuroinflammation in autism spectrum disorders, J Neuroinflammation 9 (2012) 265. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [52].El Fatimy R, Miozzo F, Le Mouël A, Abane R, Schwendimann L, Sabéran-Djoneidi D, de Thonel A, Massaoudi I, Paslaru L, Hashimoto-Torii K, Christians E, Rakic P, Gressens P, Mezger V, Heat shock factor 2 is a stress-responsive mediator of neuronal migration defects in models of fetal alcohol syndrome, EMBO Mol Med 6 (2014) 1043–1061. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [53].Escher G, Galli I, Vishwanath BS, Frey BM, Frey FJ, Tumor Necrosis Factor α and Interleukin 1β Enhance the Cortisone/Cortisol Shuttle, J Exp Med 186 (1997) 189–198. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [54].Evers M, Cunningham-Rundles C, Hollander E, Heat shock protein 90 antibodies in autism, Mol Psychiatry 7 Suppl 2 (2002) S26–28. [DOI] [PubMed] [Google Scholar]
- [55].Filipović D, Todorović N, Bernardi RE, Gass P, Oxidative and nitrosative stress pathways in the brain of socially isolated adult male rats demonstrating depressive- and anxiety-like symptoms, Brain Struct Funct 222 (2017) 1–20. [DOI] [PubMed] [Google Scholar]
- [56].Fine R, Zhang J, Stevens HE, Prenatal stress and inhibitory neuron systems: implications for neuropsychiatric disorders, Mol Psychiatry 19 (2014) 641–651. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [57].Fineberg AM, Ellman LM, Schaefer CA, Maxwell SD, Shen L, Chaudhury NH, Cook AL, Bresnahan MA, Susser ES, Brown AS, Fetal exposure to maternal stress and risk for schizophrenia spectrum disorders among offspring: Differential influences of fetal sex, Psychiatry Research 236 (2016) 91–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [58].Finnie JW, Chidlow G, Blumbergs PC, Manavis J, Cai Z, Heat shock protein induction in fetal mouse brain as a measure of stress after whole of gestation exposure to mobile telephony radiofrequency fields, Pathology 41 (2009) 276–279. [DOI] [PubMed] [Google Scholar]
- [59].Fries GR, Quevedo J, Zeni CP, Kazimi IF, Zunta-Soares G, Spiker DE, Bowden CL, Walss-Bass C, Soares JC, Integrated transcriptome and methylome analysis in youth at high risk for bipolar disorder: a preliminary analysis, Transl Psychiatry 7 (2017) e1059. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [60].Frodl T, O’Keane V, How does the brain deal with cumulative stress? A review with focus on developmental stress, HPA axis function and hippocampal structure in humans, Neurobiol Dis 52 (2013) 24–37. [DOI] [PubMed] [Google Scholar]
- [61].Fujioka T, Sakata Y, Yamaguchi K, Shibasaki T, Kato H, Nakamura S, The effects of prenatal stress on the development of hypothalamic paraventricular neurons in fetal rats, Neuroscience 92 (1999) 1079–1088. [DOI] [PubMed] [Google Scholar]
- [62].Fukumoto K, Morita T, Mayanagi T, Tanokashira D, Yoshida T, Sakai A, Sobue K, Detrimental effects of glucocorticoids on neuronal migration during brain development, Mol Psychiatry 14 (2009) 1119–1131. [DOI] [PubMed] [Google Scholar]
- [63].Garnica AD, Chan WY, The role of the placenta in fetal nutrition and growth, Journal of the American College of Nutrition 15 (1996) 206–222. [DOI] [PubMed] [Google Scholar]
- [64].Geisler JP, Manahan KJ, Geisler HE, Tammela JE, Rose SL, Hiett AK, Miller GA, Wiemann MC, Zhou Z, Heat shock protein 27 in the placentas of women with and without severe preeclampsia, Clinical and experimental obstetrics & gynecology 31 (2004) 12–14. [PubMed] [Google Scholar]
- [65].Glover V, Bergman K, Sarkar P, O’Connor TG, Association between maternal and amniotic fluid cortisol is moderated by maternal anxiety, Psychoneuroendocrinology 34 (2009) 430–435. [DOI] [PubMed] [Google Scholar]
- [66].Glover V, O’Connor TG, O’Donnell K, Prenatal stress and the programming of the HPA axis, Neurosci Biobehav Rev 35 (2010) 17–22. [DOI] [PubMed] [Google Scholar]
- [67].Goeden N, Velasquez J, Arnold KA, Chan Y, Lund BT, Anderson GM, Bonnin A, Maternal Inflammation Disrupts Fetal Neurodevelopment via Increased Placental Output of Serotonin to the Fetal Brain, J Neurosci 36 (2016) 6041–6049. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [68].Golan MH, Mane R, Molczadzki G, Zuckerman M, Kaplan-Louson V, Huleihel M, Perez-Polo JR, Impaired migration signaling in the hippocampus following prenatal hypoxia, Neuropharmacology 57 (2009) 511–522. [DOI] [PubMed] [Google Scholar]
- [69].Gorman AM, Szegezdi E, Quigney DJ, Samali A, Hsp27 inhibits 6-hydroxydopamine-induced cytochrome c release and apoptosis in PC12 cells, Biochem Biophys Res Commun 327 (2005) 801–810. [DOI] [PubMed] [Google Scholar]
- [70].Goto A, Egawa T, Sakon I, Oshima R, Ito K, Serizawa Y, Sekine K, Tsuda S, Goto K, Hayashi T, Heat stress acutely activates insulin-independent glucose transport and 5’-AMP-activated protein kinase prior to an increase in HSP72 protein in rat skeletal muscle, Physiol Rep 3 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [71].Gumusoglu SB, Fine RS, Murray SJ, Bittle JL, Stevens HE, The role of IL-6 in neurodevelopment after prenatal stress, Brain Behav Immun 65 (2017) 274–283. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [72].Guo J, Anton ES, Decision making during interneuron migration in the developing cerebral cortex, Trends Cell Biol 24 (2014) 342–351. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [73].Gur TL, Shay L, Palkar AV, Fisher S, Varaljay VA, Dowd S, Bailey MT, Prenatal stress affects placental cytokines and neurotrophins, commensal microbes, and anxiety-like behavior in adult female offspring, Brain, Behavior, and Immunity 64 (2017) 50–58. [DOI] [PubMed] [Google Scholar]
- [74].Hashimoto-Torii K, Torii M, Fujimoto M, Nakai A, El Fatimy R, Mezger V, Min J Ju, S. Ishii, S.-h. Chao, Kristen J. Brennand, Fred H. Gage, P. Rakic, Roles of Heat Shock Factor 1 in Neuronal Response to Fetal Environmental Risks and Its Relevance to Brain Disorders, Neuron 82 (2014) 560–572. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [75].Hayashi T, Conversion of psychological stress into cellular stress response: roles of the sigma-1 receptor in the process, Psychiatry Clin Neurosci 69 (2015) 179–191. [DOI] [PubMed] [Google Scholar]
- [76].Heneka MT, Sharp A, Klockgether T, Gavrilyuk V, Feinstein DL, The heat shock response inhibits NF-kappaB activation, nitric oxide synthase type 2 expression, and macrophage/microglial activation in brain, Journal of cerebral blood flow and metabolism : official journal of the International Society of Cerebral Blood Flow and Metabolism 20 (2000) 800–811. [DOI] [PubMed] [Google Scholar]
- [77].Heneka MT, Sharp A, Klockgether T, Gavrilyuk V, Feinstein DL, The Heat Shock Response Inhibits NF-κB Activation, Nitric Oxide Synthase Type 2 Expression, and Macrophage/Microglial Activation in Brain, Journal of Cerebral Blood Flow & Metabolism 20 (2000) 800–811. [DOI] [PubMed] [Google Scholar]
- [78].Henry C, Kabbaj M, Simon H, Le Moal M, Maccari S, Prenatal stress increases the hypothalamo-pituitary-adrenal axis response in young and adult rats, J Neuroendocrinol 6 (1994) 341–345. [DOI] [PubMed] [Google Scholar]
- [79].Hensen SM, Heldens L, van Enckevort CM, van Genesen ST, Pruijn GJ, Lubsen NH, Heat shock factor 1 is inactivated by amino acid deprivation, Cell stress & chaperones 17 (2012) 743–755. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [80].Hnat MD, Meadows JW, Brockman DE, Pitzer B, Lyall F, Myatt L, Heat shock protein-70 and 4-hydroxy-2-nonenal adducts in human placental villous tissue of normotensive, preeclamptic and intrauterine growth restricted pregnancies, American journal of obstetrics and gynecology 193 (2005) 836–840. [DOI] [PubMed] [Google Scholar]
- [81].Honda K, Hatayama T, Takahashi K, Yukioka M, Heat shock proteins in human and mouse embryonic cells after exposure to heat shock or teratogenic agents, Teratogenesis, carcinogenesis, and mutagenesis 11 (1991) 235–244. [DOI] [PubMed] [Google Scholar]
- [82].Hromadnikova I, Dvorakova L, Kotlabova K, Kestlerova A, Hympanova L, Novotna V, Doucha J, Krofta L, Assessment of placental and maternal stress responses in patients with pregnancy related complications via monitoring of heat shock protein mRNA levels, Molecular biology reports 42 (2015) 625–637. [DOI] [PubMed] [Google Scholar]
- [83].Hromadnikova I, Dvorakova L, Kotlabova K, Kestlerova A, Hympanova L, Novotna V, Doucha J, Krofta L, Circulating heat shock protein mRNA profile in gestational hypertension, preeclampsia & foetal growth restriction, The Indian journal of medical research 144 (2016) 229–237. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [84].Huang C, Lu X, Tong L, Wang J, Zhang W, Jiang B, Yang R, Requirement for endogenous heat shock factor 1 in inducible nitric oxide synthase induction in murine microglia, J Neuroinflammation 12 (2015) 189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [85].Ikonomidou C, Kaindl AM, Neuronal death and oxidative stress in the developing brain, Antioxid Redox Signal 14 (2011) 1535–1550. [DOI] [PubMed] [Google Scholar]
- [86].Iqbal MA, Eftekharpour E, Regulatory Role of Redox Balance in Determination of Neural Precursor Cell Fate, Stem Cells Int 2017 (2017) 9209127. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [87].Iwamoto K, Kakiuchi C, Bundo M, Ikeda K, Kato T, Molecular characterization of bipolar disorder by comparing gene expression profiles of postmortem brains of major mental disorders, Mol Psychiatry 9 (2004) 406–416. [DOI] [PubMed] [Google Scholar]
- [88].Javert CT, Prevention of Habitual Spontaneous Abortion with Early Prenatal Care, Bull N Y Acad Med 34 (1958) 747–756. [PMC free article] [PubMed] [Google Scholar]
- [89].Jo DH, An H, Chang DJ, Baek YY, Cho CS, Jun HO, Park SJ, Kim JH, Lee HY, Kim KW, Lee J, Park HJ, Kim YM, Suh YG, Kim JH, Hypoxia-mediated retinal neovascularization and vascular leakage in diabetic retina is suppressed by HIF-1alpha destabilization by SH-1242 and SH-1280, novel hsp90 inhibitors, J Mol Med (Berl) 92 (2014) 1083–1092. [DOI] [PubMed] [Google Scholar]
- [90].Joëls M, Baram TZ, The neuro-symphony of stress, Nature Reviews Neuroscience 10 (2009) 459. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [91].Jones TJ, Li D, Wolf IM, Wadekar SA, Periyasamy S, Sanchez ER, Enhancement of glucocorticoid receptor-mediated gene expression by constitutively active heat shock factor 1, Molecular endocrinology (Baltimore, Md.) 18 (2004) 509–520. [DOI] [PubMed] [Google Scholar]
- [92].Kaneta H, Ukai W, Tsujino H, Furuse K, Kigawa Y, Tayama M, Ishii T, Hashimoto E, Kawanishi C, Antipsychotics promote GABAergic interneuron genesis in the adult rat brain: Role of heat-shock protein production, J Psychiatr Res 92 (2017) 108–118. [DOI] [PubMed] [Google Scholar]
- [93].Kapoor A, Dunn E, Kostaki A, Andrews MH, Matthews SG, Fetal programming of hypothalamo-pituitary-adrenal function: prenatal stress and glucocorticoids, J Physiol 572 (2006) 31–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [94].Khashan AS, Abel KM, McNamee R, Pedersen MG, Webb RT, Baker PN, Kenny LC, Mortensen PB, Higher risk of offspring schizophrenia following antenatal maternal exposure to severe adverse life events, Arch. Gen. Psychiatry 65 (2008) 146–152. [DOI] [PubMed] [Google Scholar]
- [95].Kim JJ, Lee SJ, Toh KY, Lee CU, Lee C, Paik IH, Identification of antibodies to heat shock proteins 90 kDa and 70 kDa in patients with schizophrenia, Schizophrenia Research 52 (2001) 127–135. [DOI] [PubMed] [Google Scholar]
- [96].Kim JJ, Lee SJ, Toh KY, Lee CU, Lee C, Paik IH, Identification of antibodies to heat shock proteins 90 kDa and 70 kDa in patients with schizophrenia, Schizophrenia Research (2001) 9. [DOI] [PubMed] [Google Scholar]
- [97].Kim JJ, Mandelli L, Lim S, Lim HK, Kwon OJ, Pae CU, Serretti A, Nimgaonkar VL, Paik I-H, Jun T-Y, Association analysis of heat shock protein 70 gene polymorphisms in schizophrenia, European Archives of Psychiatry and Clinical Neuroscience 258 (2008) 239–244. [DOI] [PubMed] [Google Scholar]
- [98].Kinet MJ, Malin JA, Abraham MC, Blum ES, Silverman MR, Lu Y, Shaham S, HSF-1 activates the ubiquitin proteasome system to promote non-apoptotic developmental cell death in C. elegans, Elife 5 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [99].Kinney DK, Miller AM, Crowley DJ, Huang E, Gerber E, Autism prevalence following prenatal exposure to hurricanes and tropical storms in Louisiana, J Autism Dev Disord 38 (2008) 481–488. [DOI] [PubMed] [Google Scholar]
- [100].Kleinhaus K, Harlap S, Perrin M, Manor O, Margalit-Calderon R, Opler M, Friedlander Y, Malaspina D, Prenatal stress and affective disorders in a population birth cohort, Bipolar Disord 15 (2013) 92–99. [DOI] [PubMed] [Google Scholar]
- [101].Kowalczyk M, Owczarek A, Suchanek R, Paul-Samojedny M, Fila-Danilow A, Borkowska P, Kucia K, Kowalski J, Heat shock protein 70 gene polymorphisms are associated with paranoid schizophrenia in the Polish population, Cell stress & chaperones 19 (2014) 205–215. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [102].Kreiling JA, Creton R, Reinisch C, Early embryonic exposure to polychlorinated biphenyls disrupts heat-shock protein 70 cognate expression in zebrafish, Journal of toxicology and environmental health. Part A 70 (2007) 1005–1013. [DOI] [PubMed] [Google Scholar]
- [103].Krueger-Naug AMR, Emsley JG, Myers TL, Currie RW, Clarke DB, Administration of brain-derived neurotrophic factor suppresses the expression of heat shock protein 27 in rat retinal ganglion cells following axotomy, Neuroscience 116 (2003) 49–58. [DOI] [PubMed] [Google Scholar]
- [104].Lane RH, Ramirez RJ, Tsirka AE, Kloesz JL, McLaughlin MK, Gruetzmacher EM, Devaskar SU, Uteroplacental insufficiency lowers the threshold towards hypoxia-induced cerebral apoptosis in growth-retarded fetal rats, Brain Res 895 (2001) 186–193. [DOI] [PubMed] [Google Scholar]
- [105].Lante F, Meunier J, Guiramand J, Maurice T, Cavalier M, de Jesus Ferreira MC, Aimar R, Cohen-Solal C, Vignes M, Barbanel G, Neurodevelopmental damage after prenatal infection: role of oxidative stress in the fetal brain, Free Radic Biol Med 42 (2007) 1231–1245. [DOI] [PubMed] [Google Scholar]
- [106].Laplante DP, Barr RG, Brunet A, Galbaud du Fort G, Meaney ML, Saucier J-F, Zelazo PR, King S, Stress during pregnancy affects general intellectual and language functioning in human toddlers, Pediatr. Res 56 (2004) 400–410. [DOI] [PubMed] [Google Scholar]
- [107].Leonidas M, The Distribution of the Stress Protein HSP70 in the Cerebellum of Patients with Schizophrenia, Neuroscience and Medicine 03 (2012) 368–373. [Google Scholar]
- [108].Li D, Sanchez ER, Glucocorticoid receptor and heat shock factor 1: novel mechanism of reciprocal regulation, Vitamins and hormones 71 (2005) 239–262. [DOI] [PubMed] [Google Scholar]
- [109].Li D, Wadekar SA, Periyasamy S, E.R. Sánchez, Inhibition of Heat Shock Transcription Factor by GR, Molecular Endocrinology 15 (2001) 1396–1410. [DOI] [PubMed] [Google Scholar]
- [110].Li D, Wadekar SA, E.R. Sánchez, Agonist-Activated Glucocorticoid Receptor Inhibits Binding of Heat Shock Factor 1 to the Heat Shock Protein 70 Promoter in Vivo, Molecular Endocrinology 18 (2004) 500–508. [DOI] [PubMed] [Google Scholar]
- [111].Li J, Labbadia J, Morimoto RI, Rethinking HSF1 in Stress, Development, and Organismal Health, Trends Cell Biol 27 (2017) 895–905. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [112].Li J, Olsen J, Vestergaard M, Obel C, Attention-deficit/hyperactivity disorder in the offspring following prenatal maternal bereavement: a nationwide follow-up study in Denmark, Eur Child Adolesc Psychiatry 19 (2010) 747–753. [DOI] [PubMed] [Google Scholar]
- [113].Lian S, Guo J, Wang L, Li W, Wang J, Ji H, Kong F, Xu B, Li S, Yang H, Impact of prenatal cold stress on placental physiology, inflammatory response, and apoptosis in rats, Oncotarget 8 (2017) 115304–115314. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [114].Lin-Hendel EG, McManus MJ, Wallace DC, Anderson SA, Golden JA, Differential Mitochondrial Requirements for Radially and Non-radially Migrating Cortical Neurons: Implications for Mitochondrial Disorders, Cell Rep 15 (2016) 229–237. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [115].Lin M, Zhao D, Hrabovsky A, Pedrosa E, Zheng D, Lachman HM, Heat shock alters the expression of schizophrenia and autism candidate genes in an induced pluripotent stem cell model of the human telencephalon, PLoS One 9 (2014) e94968. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [116].Londono Tobon A, Diaz Stransky A, Ross DA, Stevens HE, Effects of Maternal Prenatal Stress: Mechanisms, Implications, and Novel Therapeutic Interventions, Biol Psychiatry 80 (2016) e85–e87. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [117].Lu M, Kim HE, Li CR, Kim S, Kwak IJ, Lee YJ, Kim SS, Moon JY, Kim CH, Kim DK, Kang HS, Park JS, Two distinct disulfide bonds formed in human heat shock transcription factor 1 act in opposition to regulate its DNA binding activity, Biochemistry 47 (2008) 6007–6015. [DOI] [PubMed] [Google Scholar]
- [118].Lupien SJ, McEwen BS, Gunnar MR, Heim C, Effects of stress throughout the lifespan on the brain, behaviour and cognition, Nat Rev Neurosci 10 (2009) 434–445. [DOI] [PubMed] [Google Scholar]
- [119].Lussier SJ, Stevens HE, Delays in GABAergic interneuron development and behavioral inhibition after prenatal stress, Dev Neurobiol 76 (2016) 1078–1091. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [120].Madhyastha S, Sahu SS, Rao G, Resveratrol for prenatal-stress-induced oxidative damage in growing brain and its consequences on survival of neurons, J Basic Clin Physiol Pharmacol 25 (2014) 63–72. [DOI] [PubMed] [Google Scholar]
- [121].Mairesse J, Lesage J, Breton C, Breant B, Hahn T, Darnaudery M, Dickson SL, Seckl J, Blondeau B, Vieau D, Maccari S, Viltart O, Maternal stress alters endocrine function of the feto-placental unit in rats, Am J Physiol Endocrinol Metab 292 (2007) E1526–1533. [DOI] [PubMed] [Google Scholar]
- [122].Malek A, Sager R, Schneider H, Effect of Hypoxia, Oxidative Stress and Lipopolysaccharides on the Release of Prostaglandins and Cytokines from Human Term Placental Explants, Placenta 22 (2001) S45–S50. [DOI] [PubMed] [Google Scholar]
- [123].Martinez-Ruiz A, Villanueva L, Gonzalez de Orduna C, Lopez-Ferrer D, Higueras MA, Tarin C, Rodriguez-Crespo I, Vazquez J, Lamas S, S-nitrosylation of Hsp90 promotes the inhibition of its ATPase and endothelial nitric oxide synthase regulatory activities, Proc Natl Acad Sci U S A 102 (2005) 8525–8530. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [124].Matarredona ER, Talaveron R, Pastor AM, Interactions Between Neural Progenitor Cells and Microglia in the Subventricular Zone: Physiological Implications in the Neurogenic Niche and After Implantation in the Injured Brain, Front Cell Neurosci 12 (2018) 268. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [125].Matosin N, Halldorsdottir T, Binder EB, Understanding the Molecular Mechanisms Underpinning Gene by Environment Interactions in Psychiatric Disorders: The FKBP5 Model, Biological Psychiatry 83 (2018) 821–830. [DOI] [PubMed] [Google Scholar]
- [126].Matrisciano F, Tueting P, Dalal I, Kadriu B, Grayson DR, Davis JM, Nicoletti F, Guidotti A, Epigenetic modifications of GABAergic interneurons are associated with the schizophrenia-like phenotype induced by prenatal stress in mice, Neuropharmacology 68 (2013) 184–194. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [127].Matthews SG, Phillips DI, Transgenerational inheritance of stress pathology, Exp Neurol 233 (2012) 95–101. [DOI] [PubMed] [Google Scholar]
- [128].Maxwell SD, Fineberg AM, Drabick DA, Murphy SK, Ellman LM, Maternal Prenatal Stress and Other Developmental Risk Factors for Adolescent Depression: Spotlight on Sex Differences, Journal of Abnormal Child Psychology 46 (2018) 381–397. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [129].Miller DJ, Fort PE, Heat Shock Proteins Regulatory Role in Neurodevelopment, Front Neurosci 12 (2018) 821. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [130].Miller DJ, Fort PE, Heat Shock Proteins Regulatory Role in Neurodevelopment, Frontiers in neuroscience 12 (2018) 821-821. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [131].Molvarec A, Szarka A, Walentin S, Bekő G, I. Karádi, Z. Prohászka, J. Rigó, Serum heat shock protein 70 levels in relation to circulating cytokines, chemokines, adhesion molecules and angiogenic factors in women with preeclampsia, Clinica Chimica Acta 412 (2011) 1957–1962. [DOI] [PubMed] [Google Scholar]
- [132].Morimoto RI, Santoro MG, Stress-inducible responses and heat shock proteins: new pharmacologic targets for cytoprotection, Nat Biotechnol 16 (1998) 833–838. [DOI] [PubMed] [Google Scholar]
- [133].Morishima Y, Murphy PJ, Li DP, Sanchez ER, Pratt WB, Stepwise assembly of a glucocorticoid receptor.hsp90 heterocomplex resolves two sequential ATP-dependent events involving first hsp70 and then hsp90 in opening of the steroid binding pocket, The Journal of biological chemistry 275 (2000) 18054–18060. [DOI] [PubMed] [Google Scholar]
- [134].Moseley PL, Heat Shock Proteins and the Inflammatory Response, Annals of the New York Academy of Sciences 856 (1998) 206–213. [DOI] [PubMed] [Google Scholar]
- [135].Mueller BR, Bale TL, Sex-specific programming of offspring emotionality after stress early in pregnancy, J Neurosci 28 (2008) 9055–9065. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [136].Murphy SK, Fineberg AM, Maxwell SD, Alloy LB, Zimmermann L, Krigbaum NY, Cohn BA, Drabick DAG, Ellman LM, Maternal infection and stress during pregnancy and depressive symptoms in adolescent offspring, Psychiatry Research 257 (2017) 102–110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [137].Myatt L, Placental adaptive responses and fetal programming, J Physiol 572 (2006) 25–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [138].Nayernia Z, Colaianna M, Robledinos-Anton N, Gutzwiller E, Sloan-Bena F, Stathaki E, Hibaoui Y, Cuadrado A, Hescheler J, Stasia MJ, Saric T, Jaquet V, Krause KH, Decreased neural precursor cell pool in NADPH oxidase 2-deficiency: From mouse brain to neural differentiation of patient derived iPSC, Redox Biol 13 (2017) 82–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [139].Nedivi E, Hevroni D, Naot D, Israeli D, Citri Y, Numerous candidate plasticity-related genes revealed by differential cDNA cloning, Nature 363 (1993) 718–722. [DOI] [PubMed] [Google Scholar]
- [140].Nkansah-Amankra S, Luchok KJ, Hussey JR, Watkins K, Liu X, Effects of maternal stress on low birth weight and preterm birth outcomes across neighborhoods of South Carolina, 2000–2003, Maternal and child health journal 14 (2010) 215–226. [DOI] [PubMed] [Google Scholar]
- [141].Nomura Y, John RM, Janssen AB, Davey C, Finik J, Buthmann J, Glover V, Lambertini L, Neurodevelopmental consequences in offspring of mothers with preeclampsia during pregnancy: underlying biological mechanism via imprinting genes, Archives of gynecology and obstetrics 295 (2017) 1319–1329. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [142].Oskvig DB, Elkahloun AG, Johnson KR, Phillips TM, Herkenham M, Maternal immune activation by LPS selectively alters specific gene expression profiles of interneuron migration and oxidative stress in the fetus without triggering a fetal immune response, Brain Behav Immun 26 (2012) 623–634. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [143].Padmini E, Lavanya S, Uthra V, Preeclamptic placental stress and over expression of mitochondrial HSP70, Clinical chemistry and laboratory medicine 47 (2009) 1073–1080. [DOI] [PubMed] [Google Scholar]
- [144].Padmini E, Venkatraman U, Srinivasan L, Mechanism of JNK signal regulation by placental HSP70 and HSP90 in endothelial cell during preeclampsia, Toxicol Mech Methods 22 (2012) 367–374. [DOI] [PubMed] [Google Scholar]
- [145].Pae C-U, Kim T-S, Kwon O-J, Artioli P, Serretti A, Lee C-U, Lee S-J, Lee C, Paik I-H, Kim J-J, Polymorphisms of heat shock protein 70 gene (HSPA1A, HSPA1B and HSPA1L) and schizophrenia, Neuroscience Research 53 (2005) 8–13. [DOI] [PubMed] [Google Scholar]
- [146].Pae C-U, Mandelli L, Serretti A, Patkar AA, Kim J-J, Lee C-U, Lee S-J, Lee C, De Ronchi D, Paik I-H, Heat-shock protein-70 genes and response to antidepressants in major depression, Progress in Neuro-Psychopharmacology and Biological Psychiatry 31 (2007) 1006–1011. [DOI] [PubMed] [Google Scholar]
- [147].Pandey P, Saleh A, Nakazawa A, Kumar S, Srinivasula SM, Kumar V, Weichselbaum R, Nalin C, Alnemri ES, Kufe D, Kharbanda S, Negative regulation of cytochrome c-mediated oligomerization of Apaf-1 and activation of procaspase-9 by heat shock protein 90, EMBO J 19 (2000) 4310–4322. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [148].Pankevich DE, Mueller BR, Brockel B, Bale TL, Prenatal stress programming of offspring feeding behavior and energy balance begins early in pregnancy, Physiology & behavior 98 (2009) 94–102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [149].Park M, Brain U, Grunau RE, Diamond A, Oberlander TF, Maternal depression trajectories from pregnancy to 3 years postpartum are associated with children’s behavior and executive functions at 3 and 6 years, Archives of Women’s Mental Health (2018). [DOI] [PubMed] [Google Scholar]
- [150].Pasquali MA, Harlow BL, Soares CN, Otto MW, Cohen LS, Minuzzi L, Gelain DP, Moreira JCF, Frey BN, A longitudinal study of neurotrophic, oxidative, and inflammatory markers in first-onset depression in midlife women, Eur Arch Psychiatry Clin Neurosci 268 (2018) 771–781. [DOI] [PubMed] [Google Scholar]
- [151].Paul S, Ghosh S, Mandal S, Sau S, Pal M, NRF2 transcriptionally activates the heat shock factor 1 promoter under oxidative stress and affects survival and migration potential of MCF7 cells, The Journal of biological chemistry 293 (2018) 19303–19316. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [152].Polleux F, Whitford KL, Dijkhuizen PA, Vitalis T, Ghosh A, Control of cortical interneuron migration by neurotrophins and PI3-kinase signaling, Development 129 (2002) 3147–3160. [DOI] [PubMed] [Google Scholar]
- [153].Ponder KL, Salisbury A, McGonnigal B, Laliberte A, Lester B, Padbury JF, Maternal depression and anxiety are associated with altered gene expression in the human placenta without modification by antidepressant use: Implications for fetal programming, 53 (2011) 711–723. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [154].Pongrac JL, Middleton FA, Peng L, Lewis DA, Levitt P, Mirnics K, Heat shock protein 12A shows reduced expression in the prefrontal cortex of subjects with schizophrenia, Biol Psychiatry 56 (2004) 943–950. [DOI] [PubMed] [Google Scholar]
- [155].Prohászka Z, Füst G, Immunological aspects of heat-shock proteins—the optimum stress of life, Molecular Immunology 41 (2004) 29–44. [DOI] [PubMed] [Google Scholar]
- [156].Pugliese V, Bruni A, Carbone EA, Calabro G, Cerminara G, Sampogna G, Luciano M, Steardo L Jr., Fiorillo A, Garcia CS, De Fazio P, Maternal stress, prenatal medical illnesses and obstetric complications: Risk factors for schizophrenia spectrum disorder, bipolar disorder and major depressive disorder, Psychiatry Res 271 (2019) 23–30. [DOI] [PubMed] [Google Scholar]
- [157].Rakers F, Rupprecht S, Dreiling M, Bergmeier C, Witte OW, Schwab M, Transfer of maternal psychosocial stress to the fetus, Neurosci Biobehav Rev (2017). [DOI] [PubMed] [Google Scholar]
- [158].Ratman D, Vanden Berghe W, Dejager L, Libert C, Tavernier J, Beck IM, De Bosscher K, How glucocorticoid receptors modulate the activity of other transcription factors: a scope beyond tethering, Mol Cell Endocrinol 380 (2013) 41–54. [DOI] [PubMed] [Google Scholar]
- [159].Ray PD, Huang BW, Tsuji Y, Reactive oxygen species (ROS) homeostasis and redox regulation in cellular signaling, Cell Signal 24 (2012) 981–990. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [160].Rong G, Weijian H, Yafeng D, Zhiyong Y, Stites J, Weiner CP, Brain Injury Caused by Chronic Fetal Hypoxemia Is Mediated by Inflammatory Cascade Activation, Reproductive Sciences 17 (2010) 540–548. [DOI] [PubMed] [Google Scholar]
- [161].Rosenfeld CS, Sex-Specific Placental Responses in Fetal Development, Endocrinology 156 (2015) 3422–3434. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [162].Ryter SW, Kim HP, Hoetzel A, Park JW, Nakahira K, Wang X, Choi AM, Mechanisms of cell death in oxidative stress, Antioxid Redox Signal 9 (2007) 49–89. [DOI] [PubMed] [Google Scholar]
- [163].Sahin H, Gunel T, Benian A, Onay Ucar E, Guralp O, Kilic A, Genomic and proteomic investigation of preeclampsia, Exp Ther Med 10 (2015) 711–716. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [164].Saleh A, Srinivasula SM, Balkir L, Robbins PD, Alnemri ES, Negative regulation of the Apaf-1 apoptosome by Hsp70, Nat Cell Biol 2 (2000) 476–483. [DOI] [PubMed] [Google Scholar]
- [165].Sanchez ER, Heat shock induces translocation to the nucleus of the unliganded glucocorticoid receptor, J. Biol. Chem 267 (1992) 17–20. [PubMed] [Google Scholar]
- [166].Sarkar P, Bergman K, Fisk NM, Glover V, Maternal anxiety at amniocentesis and plasma cortisol, Prenatal Diagnosis 26 (2006) 505–509. [DOI] [PubMed] [Google Scholar]
- [167].Schieber M, Chandel NS, ROS function in redox signaling and oxidative stress, Curr Biol 24 (2014) R453–462. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [168].Schoof E, Girstl M, Frobenius W, Kirschbaum M, Repp R, Knerr I, Rascher W, Dotsch J, Course of placental 11beta-hydroxysteroid dehydrogenase type 2 and 15-hydroxyprostaglandin dehydrogenase mRNA expression during human gestation, European Journal of Endocrinology 145 (2001) 187–192. [DOI] [PubMed] [Google Scholar]
- [169].Schwarz MJ, Riedel M, Gruber R, Ackenheil M, Muller N, Antibodies to heat shock proteins in schizophrenic patients: implications for the mechanism of the disease, The American journal of psychiatry 156 (1999) 1103–1104. [DOI] [PubMed] [Google Scholar]
- [170].Seckl JR, Holmes MC, Mechanisms of Disease: glucocorticoids, their placental metabolism and fetal ‘programming’ of adult pathophysiology, Nature Reviews Endocrinology 3 (2007) 479–488. [DOI] [PubMed] [Google Scholar]
- [171].Shams M, Kilby MD, Somerset DA, Howie AJ, Gupta A, Wood PJ, Afnan M, Stewart PM, 11Beta-hydroxysteroid dehydrogenase type 2 in human pregnancy and reduced expression in intrauterine growth restriction, Hum. Reprod 13 (1998) 799–804. [DOI] [PubMed] [Google Scholar]
- [172].Shim SY, Kim HS, Oxidative stress and the antioxidant enzyme system in the developing brain, Korean J Pediatr 56 (2013) 107–111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [173].Shimizu S, Nomura K, Ujihara M, Sakamoto K, Shibata H, Suzuki T, Demura H, An allel-specific abnormal transcript of the heat shock protein 70 gene in patients with major depression, Biochem Biophys Res Commun 219 (1996) 745–752. [DOI] [PubMed] [Google Scholar]
- [174].Shin JK, Baek JC, Kang MY, Park JK, Lee SA, Lee JH, Choi WS, Paik WY, Proteomic analysis reveals an elevated expression of heat shock protein 27 in preeclamptic placentas, Gynecologic and obstetric investigation 71 (2011) 151–157. [DOI] [PubMed] [Google Scholar]
- [175].Shyu W-C, Chen C-P, Saeki K, Kubosaki A, Matusmoto Y, Onodera T, Ding D-C, Chiang M-F, Lee Y-J, Lin S-Z, Li H, Hypoglycemia enhances the expression of prion protein and heat-shock protein 70 in a mouse neuroblastoma cell line, 80 (2005) 887–894. [DOI] [PubMed] [Google Scholar]
- [176].Sjostrom K, Valentin L, Thelin T, Marsal K, Maternal anxiety in late pregnancy and fetal hemodynamics, Eur J Obstet Gynecol Reprod Biol 74 (1997) 149–155. [DOI] [PubMed] [Google Scholar]
- [177].Slusarczyk J, Trojan E, Glombik K, Budziszewska B, Kubera M, Lason W, Popiolek-Barczyk K, Mika J, Wedzony K, Basta-Kaim A, Prenatal stress is a vulnerability factor for altered morphology and biological activity of microglia cells, Front Cell Neurosci 9 (2015) 82. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [178].Sosnowski DW, Booth C, York TP, Amstadter AB, Kliewer W, Maternal prenatal stress and infant DNA methylation: A systematic review, Dev Psychobiol 60 (2018) 127–139. [DOI] [PubMed] [Google Scholar]
- [179].Squarzoni P, Oller G, Hoeffel G, Pont-Lezica L, Rostaing P, Low D, Bessis A, Ginhoux F, Garel S, Microglia modulate wiring of the embryonic forebrain, Cell Rep 8 (2014) 1271–1279. [DOI] [PubMed] [Google Scholar]
- [180].Stevens HE, Su T, Yanagawa Y, Vaccarino FM, Prenatal stress delays inhibitory neuron progenitor migration in the developing neocortex, Psychoneuroendocrinology 38 (2013) 509–521. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [181].Stevens HE, Su T, Yanagawa Y, Vaccarino FM, Prenatal stress delays inhibitory neuron progenitor migration in the developing neocortex, Psychoneuroendocrinology 38 (2013) 509–521. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [182].Stewart PM, Murry BA, Mason JI, Type 2 11 beta-hydroxysteroid dehydrogenase in human fetal tissues, J Clin Endocrinol Metab 78 (1994) 1529–1532. [DOI] [PubMed] [Google Scholar]
- [183].Swaroop S, Mahadevan A, Shankar SK, Adlakha YK, Basu A, HSP60 critically regulates endogenous IL-1beta production in activated microglia by stimulating NLRP3 inflammasome pathway, J Neuroinflammation 15 (2018) 177. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [184].Tanaka DH, Nakajima K, GABAergic interneuron migration and the evolution of the neocortex, Dev Growth Differ 54 (2012) 366–372. [DOI] [PubMed] [Google Scholar]
- [185].Tatum MC, Ooi FK, Chikka MR, Chauve L, Martinez-Velazquez LA, Steinbusch HWM, Morimoto RI, Prahlad V, Neuronal serotonin release triggers the heat shock response in C. elegans in the absence of temperature increase, Curr Biol 25 (2015) 163–174. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [186].Tay TL, Savage JC, Hui CW, Bisht K, Tremblay ME, Microglia across the lifespan: from origin to function in brain development, plasticity and cognition, J Physiol 595 (2017) 1929–1945. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [187].Tong CK, Vidyadaran S, Role of microglia in embryonic neurogenesis, Exp Biol Med (Maywood) 241 (2016) 1669–1675. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [188].Torii M, Sasaki M, Chang YW, Ishii S, Waxman SG, Kocsis JD, Rakic P, Hashimoto-Torii K, Detection of vulnerable neurons damaged by environmental insults in utero, Proc Natl Acad Sci U S A 114 (2017) 2367–2372. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [189].Tsatmali M, Walcott EC, Makarenkova H, Crossin KL, Reactive oxygen species modulate the differentiation of neurons in clonal cortical cultures, Mol Cell Neurosci 33 (2006) 345–357. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [190].Uchida T, Furukawa T, Iwata S, Yanagawa Y, Fukuda A, Selective loss of parvalbumin-positive GABAergic interneurons in the cerebral cortex of maternally stressed Gad1-heterozygous mouse offspring, Transl Psychiatry 4 (2014) e371. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [191].Ungewickell E, The 70-kd mammalian heat shock proteins are structurally and functionally related to the uncoating protein that releases clathrin triskelia from coated vesicles, The EMBO journal 4 (1985) 3385–3391. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [192].van den Bergh BRH, Dahnke R, Mennes M, Prenatal stress and the developing brain: Risks for neurodevelopmental disorders, Dev Psychopathol 30 (2018) 743–762. [DOI] [PubMed] [Google Scholar]
- [193].van Os J, Selten JP, Prenatal exposure to maternal stress and subsequent schizophrenia. The May 1940 invasion of The Netherlands, Br J Psychiatry 172 (1998) 324–326. [DOI] [PubMed] [Google Scholar]
- [194].Velasquez JC, Goeden N, Bonnin A, Placental serotonin: implications for the developmental effects of SSRIs and maternal depression, Frontiers in cellular neuroscience 7 (2013) 47–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [195].Viltart O, Vanbesien-Mailliot CCA, Impact of Prenatal Stress on Neuroendocrine Programming %J TheScientificWorldJOURNAL, 7 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [196].Wang C-H, Chou P-C, Chung F-T, Lin H-C, Huang K-H, Kuo H-P, Heat shock protein70 is implicated in modulating NF-κB activation in alveolar macrophages of patients with active pulmonary tuberculosis, Scientific Reports 7 (2017) 1214. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [197].Wang Q, Shelton RC, Dwivedi Y, Interaction between early-life stress and FKBP5 gene variants in major depressive disorder and post-traumatic stress disorder: A systematic review and meta-analysis, Journal of Affective Disorders 225 (2018) 422–428. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [198].Weinstock M, Matlina E, Maor GI, Rosen H, McEwen BS, Prenatal stress selectively alters the reactivity of the hypothalamic-pituitary adrenal system in the female rat, Brain Research 595 (1992) 195–200. [DOI] [PubMed] [Google Scholar]
- [199].Wolford E, Lahti M, Tuovinen S, Lahti J, Lipsanen J, Savolainen K, Heinonen K, Hämäläinen E, Kajantie E, Pesonen A-K, Villa PM, Laivuori H, Reynolds RM, Räikkönen K, Maternal depressive symptoms during and after pregnancy are associated with attention-deficit/hyperactivity disorder symptoms in their 3- to 6-year-old children, PLOS ONE 12 (2017) e0190248. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [200].Wyrwoll C, Keith M, Noble J, Stevenson PL, Bombail V, Crombie S, Evans LC, Bailey MA, Wood E, Seckl JR, Holmes MC, Fetal brain 11β-hydroxysteroid dehydrogenase type 2 selectively determines programming of adult depressive-like behaviors and cognitive function, but not anxiety behaviors in male mice, Psychoneuroendocrinology 59 (2015) 59–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [201].Yang J, Oza J, Bridges K, Chen KY, Liu AY, Neural differentiation and the attenuated heat shock response, Brain Res 1203 (2008) 39–50. [DOI] [PubMed] [Google Scholar]
- [202].Yang K, Placental 11 beta-hydroxysteroid dehydrogenase: barrier to maternal glucocorticoids, Rev. Reprod 2 (1997) 129–132. [DOI] [PubMed] [Google Scholar]
- [203].Yenari MA, Antiapoptotic and Anti-inflammatory Mechanisms of Heat-Shock Protein Protection, Annals of the New York Academy of Sciences 1053 (2005) 74–83. [DOI] [PubMed] [Google Scholar]
- [204].Yin F, Chen Z, Z.-w. Li, J. Tang, Influence of acute heat stress on the development of GABAergic neurons in HPA-axies of mouse embryos, Journal of Thermal Biology 36 (2011) 486–491. [Google Scholar]
- [205].Zhu X, Peng M, Cheng M, Xiao X, Yi J, Yao S, Zhang X, Hyperthermia protects mice against chronic unpredictable stress-induced anxiety-like behaviour and hippocampal CA3 cell apoptosis, Int J Hyperthermia 27 (2011) 573–581. [DOI] [PubMed] [Google Scholar]
- [206].Zininga T, Ramatsui L, Shonhai A, Heat Shock Proteins as Immunomodulants, Molecules (Basel, Switzerland) 23 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]