Background: From the original studies investigating the effects of adrenal gland secretion to modern high-throughput multidimensional analyses, stress research has been a topic of scientific interest spanning just over a century. Summary: The objective of this review was to provide historical context for influential discoveries, surprising findings, and preclinical models in stress-related neuroimmune research. Furthermore, we summarize this work and present a current understanding of the stress pathways and their effects on the immune system and behavior. We focus on recent work demonstrating stress-induced immune changes within the brain and highlight studies investigating stress effects on microglia. Lastly, we conclude with potential areas for future investigation concerning microglia heterogeneity, bone marrow niches, and sex differences. Key Messages: Stress is a phenomenon that ties together not only the central and peripheral nervous system, but the immune system as well. The cumulative effects of stress can enhance or suppress immune function, based on the intensity and duration of the stressor. These stress-induced immune alterations are associated with neurobiological changes, including structural remodeling of neurons and decreased neurogenesis, and these contribute to the development of behavioral and cognitive deficits. As such, research in this field has revealed important insights into neuroimmune communication as well as molecular and cellular mediators of complex behaviors relevant to psychiatric disorders.

The wonder of the human mind – that which governs the rest of the body – has enchanted and puzzled philosophers and scientists for millennia. One experience of great interest is stress – the familiar feeling one gets when faced with a challenge. Despite the widespread public use of the term “stress,” it is usually described vaguely and ambiguously. For example, it can refer to the event (stressor) or the response (stress response) [1]. As once described by Hans Selye, the man who coined the term, “everybody knows what stress is and nobody knows what it is” [2]. To start our historical review, it is best for us to start at the turn of the 20th century. Many of the important events discussed in this review are summarized in a timeline presented in Figure 1.

Fig. 1.

Timeline of prominent events and findings in stress research (1879–present).

Fig. 1.

Timeline of prominent events and findings in stress research (1879–present).

Close modal

In 1884, an article by the American philosopher and psychologist William James posed an interesting question: “Do we run from a bear because we’re afraid or are we afraid because we run?” [3]. James poses the idea that physiological changes directly follow the perception of external stimuli, and the sum of these internal changes is the experienced emotion. This is the basis of the James-Lange theory of emotion [4]. This assertion was met with much scrutiny by the American physiologist Walter Cannon, who through his experiments on severing afferent nerves of the sympathetic division and assessing emotional response argued that the feeling of emotion and the physiological changes that occur in response to external stimuli are independent of one another [5]. In the same work, Cannon coined the term “fight-or-flight” to describe the state an animal goes into when challenged with a threat. This state is marked by physiological changes such as increased heart rate, ventilation, piloerection, and decreased digestion. Cannon later went on to establish the idea of homeostasis [6], in large part based on Claude Bernard’s work describing the constancy of the “milieu interior” or “internal environment” of the body (first event on Fig. 1 timeline) [7]. In this article, Cannon emphasizes the role of the autonomic nervous system (ANS) in keeping constant the factors of the interior environment and how changes in the external environment excite reactions in this system and lead to disturbances.

In the 1930s, the Hungarian-Canadian endocrinologist Hans Selye was conducting experiments to identify female sex hormones. These studies involved the injection of various extracts into female mice. Curiously, all the extracts produced the same results: adrenal gland growth, thymic involution, and peptic ulcers in the stomach and duodenum. Selye continued experiments on female mice but instead of injecting them with various extracts, he exposed them to stressful situations such as cold exposure or forced running on a wheel. These additional experiments yielded the same effects, leading Selye to conclude that this syndrome arises from the animal attempting to adapt to the changing conditions. He called these combined effects the general adaptation syndrome (also referred to as Selye’s syndrome) and organized it into three distinct phases (see top-right corner of Fig. 1): the alarm phase, the phase of resistance, and the phase of exhaustion [8]. He later renamed general adaptation syndrome as the “stress response,” or more simply “stress,” borrowing the term from physics and inserting it into the medical lexicon [9]. Selye continued to make further advancements in the newly established field of stress research by describing information “mediators” between the brain and peripheral organs [10], comparing the differences between healthy stress (“eustress”) and pathogenic stress (“distress”) [11], and began to link stress biological pathways with immunology [12].

The hormones secreted from the adrenal glands were known to be necessary in facilitating physiological responses as far back as Cannon’s work [13, 14], but the steroid compounds were not formally isolated from the adrenal cortex until the 1930s through the efforts of competing scientists and eventual 1950 Nobel laureates, Edward Kendall and Tadeusz Reichstein (Fig. 1) [15]. Cortisol, called “compound F” by Kendall, was synthesized a decade later [16]. All steroids secreted by the adrenal gland would come to be known as corticosteroids, which itself was broken into two classes: glucocorticoids (GCs) and mineralocorticoids, also named by Selye [15, 17]. Of note, it was Selye who emphasized that the stress response is not solely due to the systemic release of adrenaline and noradrenaline (catecholamines) from the adrenal medulla but also the systemic release of cortisol (GC) from the adrenal cortex that plays a role in the stress response [18]. Nearly 40 years later, GC release was found to be controlled by communication between the hypothalamus, pituitary gland, and adrenal gland. These insights established the field of neuroendocrinology and initial conceptualization of the brain-body connection known as the hypothalamic-pituitary-adrenal (HPA) axis [19‒21].

Later, the American neuroscientist Bruce McEwen connected the stress response to brain regions previously associated with memory and cognition. His seminal work in the 1960s (see middle section of Fig. 1), through the use of radioactive corticosterone, showed a high presence of glucocorticoid receptors (GRs) in the rat hippocampus [22]. He and his colleagues would go on to show further retention of GCs in other rat brain regions [23], as well as the hippocampus of nonhuman primates [24]. Around this same time, estrogen (another steroid hormone) was found to bind to intracellular receptors and enact transcriptional changes in various tissues leading to morphological changes [25]. These breakthroughs led to an exciting question – could steroid hormones such as GCs produced during stress influence the structure and function of neurons, and consequently entire brain regions?

A plethora of studies looking into how stress and GC signaling affect neuronal structure within multiple brain regions were published in the 1990s–2000s (see Fig. 1). Early studies linking GC levels and memory loss in hippocampus [26] directed investigators to analyze the structure of pyramidal neurons in the CA1, CA3, and dentate gyrus [27‒29]. These studies revealed that stress caused dendritic retractions in pyramidal neurons of the CA1 and CA3 regions and reduced cell counts within the dentate gyrus – demonstrating a reduction in adult neurogenesis. Intriguingly, antidepressants were found to increase neurogenesis in the same region, linking depression and stress with overlapping neurobiological alterations [30]. Other studies showed that patients with psychiatric disorders such as depression or post-traumatic stress disorder (PTSD) exhibit reduced hippocampal volume [31, 32] which incorporated the hippocampus into brain regions associated with mood disorders. Further studies revealed that the prefrontal cortex (PFC) also had high levels of GRs [33]. As such, exposure to chronic stress caused dendritic retraction of PFC pyramidal neurons as well [34‒36]. Dendritic spines were also shown to decrease on pyramidal neurons of the PFC following stress exposure, with the largest deficits rising in the most distal sections of the apical dendrites residing in layer I [37]. In contrast, the amygdala, which is reciprocally connected to the hippocampus and PFC [38], exhibited an expansion of dendrites following stress exposure [39]. The orbitofrontal cortex also exhibited dendritic expansion following repeated stress [35]. One potential cause in the differential stress response between these regions may be differences in expression of neurotrophic factors such as brain-derived neurotrophic factor following stress exposure [40].

The impact of stress responses on the immune system and disease was recognized by the American psychologist Robert Ader and American microbiologist Nicholas Cohen. Before their work, it was widely believed that the immune system was autonomous – strictly independent of the nervous system [41]. In a study that was initially investigating taste aversion, rats were subjugated to injections of cyclophosphamide, an immunosuppressing agent, after they drank from saccharine-water solution to condition them to avoid the sweetened water [42]. After sufficient conditioning, Ader and Cohen force-fed the rats the saccharine-water without injection of cyclophosphamide to simply complete the experiment properly with extinction trials. What they did not expect was the sugary water alone elicited a rather severe reaction leading to the death of multiple rats. Ader and Cohen hypothesized that avoidance was not the only thing conditioned but also the immunosuppressive effects of cyclophosphamide, effectively establishing a functional connection between the brain with the immune system. This work was foundational for the beginning of an emerging new field called psychoneuroimmunology – a term coined by Ader himself who would first use it in a speech to the American Psychosomatic Society in 1980 (Fig. 1) [41]. This paved the way for breakthroughs in the late 1970s and 1980s that showed inflammation could alter neuronal signaling in the hypothalamus [43] and bidirectional communication between the brain and the immune system [44, 45]. Further studies showed how stress could alter the immune system [46, 47] and increase susceptibility to physical illness [48]. Specifically, cytokines can engage in neuronal communication regulating GC release, and GCs can subdue further cytokine release [49]. Concurrently, other studies were finding immune abnormalities within patients diagnosed with major depressive disorder (MDD) [48, 50] or schizophrenia [46], laying the groundwork for further studies investigating the relationship between the immune system and complex brain disorders.

To study stress and its associated cluster of stress-related psychiatric disorders, a multitude of preclinical rodent models have been developed [51]. They can be categorized based on the type of stressors they employ (physiological, psychological, or both) and/or the duration of the model (acute vs. chronic). Examples of commonly used rodent stress models are the social defeat paradigm, chronic unpredictable stress (CUS), restraint stress, and early-life stress (ELS).

The social defeat paradigm is a physiological and psychological model that involves an intruder Sprague-Dawley rat being attacked (defeated) by resident Long Evans rats over the course of multiple days [52]. After being defeated, intruder rats are confined in the same cage as their attackers with only perforated plexiglass separating them, allowing for intense visual, olfactory, and auditory stimuli to affect the defeated rat. This paradigm is proposed to mimic human stressors such as aggression, bullying, and chronic subordination that contribute to the development of PTSD [53]. Social defeat stress has been shown to promote social avoidance and induce increases in pro-inflammatory cytokines and blood-brain barrier dysfunction in mice, which recapitulate some features of psychiatric conditions [54, 55].

CUS uses multiple randomized physiological and psychological stressors to avoid acclimation. When first developed in the early 1980s (as shown in Fig. 1), experimenters primarily used physiologically related stressors involving pain (shock exposure), hunger (food deprivation), thirst (water deprivation), and exposure to noxious conditions such as extreme heat [56]. In 1987, modifications to include more natural and milder stressors were used to better simulate the continued mild stress in which humans might endure [57]. These stressors included by Willner were wet (soiled) bedding, tilted cage, radio noise, constant lighting, and strobe light exposure. Nonetheless, it is important to note to the reader that laboratories often employ unique combinations of these stressors and for various timespans [58]. Other names have been given to this paradigm such as chronic variable stress [59] and chronic mild stress [58] – but it should be stated clearly that these all describe the same practical experimental set-up [60]. CUS is model aspects of psychiatric disorders as the regiment has been shown to induce passive coping behavior, working memory deficits, and synaptic loss [61, 62].

Restraint stress, originally devised in France to study interactions between the nervous system and gastrointestinal tract (see Fig. 1) [63, 64], is a stressor in which the animal is restricted to a small space in which they are unable to move freely for a set period of time and this duration varies from experiment to experiment [65]. Intriguingly, anxiety-like and depressive-like behaviors are more repeatably and reliably observed in chronic restraint paradigms (>6 days) [66]. This regimen was used to show that hippocampal CA3 neurons undergo atrophy of their apical dendrites following stress [67]. Of note, repeated stress was used to discover circuit-specific effects of stress on neuronal morphology in the infralimbic region of the PFC, emphasizing that neurons with different efferent projections can have divergent stress effects [68]. Interestingly, shortened versions of this regimen (5 min daily) have been shown to reverse stress-induced behavioral deficits from longer versions (2 h daily), showing the potential adaptive features of acute stress [69].

All the models listed previously involve stress exposure in adulthood. However, data have shown that exposure to stressful events early in life can have long-lasting consequences, such as higher risk for development of depression, anxiety, and cognitive impairments [70, 71]. Also, clinical data suggest that exposure to ELS (i.e., childhood trauma) increases circulation of inflammatory biomarkers [72]. In order to model childhood adversity or trauma, most ELS models employ a method where parental/maternal care is reduced, because the parent-child relationship is the source of most early-life interactions [73]. Two models that manipulate this relationship are intermittent maternal separation (MS) [74, 75] and limited nesting and bedding material [76, 77]. MS involves keeping mice separated from their mothers and their nest for 3 or more hours a day over the first two postnatal weeks [74] which leads to increases in depressive-like and anxiety-like behaviors [78] and alterations in HPA axis activity [79]. Intriguingly, “handling” or shorter bouts of separation from the mother (15 min daily) result in opposite effects of MS [78]. Limited nesting and bedding involves limiting nesting material for the mother to build a nest for her pups [76] which leads to a myriad of stress-related effects across many mouse lines [80], including elevated corticosterone levels [81] and altered exploratory and coping behaviors in various tests [82, 83].

Research in the field of stress has demonstrated that physiological and neurobiological systems are important for adapting to stress. Exposure to stressful stimuli and perception of threat activate two systems within the body, the ANS and the HPA axis [84]. These systems function as survival mechanisms and ready the body to respond to the dangers at hand. McEwen developed the construct of “allostatic load” to describe the long-term effect of the physiological response to stress, applying the concept of allostasis to stress theory. Allostasis is defined as stability through change. He postulated that the accommodation the body makes via the autonomic system and HPA axis to protect the body from internal and external stress can result in wear and tear either through chronic overactivity or underactivity of the aforementioned neurobiological systems [85]. Correlations were found between stressful life events and an increased disposition to develop psychiatric conditions such as depression, PTSD, and anxiety disorders which researchers refer to as “stress”-related psychiatric disorders [85‒87].

Further studies of the body’s stress response have made it apparent that many other systems are affected after exposure, one of them being the immune system [88, 89]. Both the HPA axis and the ANS act as extensions of the nervous system, directly influence the function of the immune system, and affect immune changes throughout the body. Under normal conditions, the body only encounters stressful situations every now and then; however, certain conditions can lead to persistent or chronic stress. Understanding the connection between chronic stress and immune system activation is important because dysregulation of either part can lead to the development of neurobiological deficits associated with psychiatric disorders [90, 91].

The ANS functions to regulate involuntary actions of the nervous system. At the most basic level, this system controls the body’s ability to switch between a state of “alertness and responsivity” and a state of “rest and maintenance.” These two opposing states are mediated by innervating systems called the sympathetic nervous system (SNS) and parasympathetic nervous system (PNS), respectively [92], and they can produce systemic changes through its extensive innervation of most tissues and organ systems in the body [93]. Neurons and nerves within these systems potentiate signals through the release of either noradrenaline or acetylcholine neurotransmitters. Since both systems are tonically activated, input from either can increase or decrease, and often have opposing effects. Simultaneous increase in the activity of one system and decrease in the activity of the other allow the ANS to quickly change the body’s state in response to threats. Therefore, the ANS is the first line of defense in a situation where stress is encountered [94].

One example of the body’s response to stress is a rapid increase in heart rate and blood pressure. This is driven by sympathetic projections that increase activation of cardiovascular tissues such as the heart and blood vessels [93]. Other effects of increased sympathetic activation are sweat secretion, dilation of blood vessels within skeletal muscle, and constriction of blood vessels within the gastrointestinal tract. All these actions work to direct energy to the parts of the body that would need to respond in the event that there is a threat to survival. These effects as described here are carried out by direct projections to the target tissue; however, the SNS can also induce these effects indirectly through projections to the inner part of the adrenal gland called the adrenal medulla. The cells inside the medulla produce adrenaline which is secreted and picked up by the circulating blood. This allows for transmission of adrenaline to the different target tissues throughout the body [94], amplifying the effects of SNS activation.

The HPA axis is an endocrine network in the body that shapes the stress response by optimizing energy utilization. While the ANS and HPA axis have different roles in the stress response, they both ultimately work to prepare the body to respond to the stressor. One difference is that the ANS responds to a stressful situation within seconds and its activation is relatively short, whereas the HPA axis responds more gradually and persists beyond the initial activation [94, 95]. This is due in part to the anatomical location of the components and because the signals must travel between parts of the axis through the bloodstream. In particular, the signaling cascade is initiated in the hypothalamus, an area of the brain that receives input from the brainstem and is activated by signals of homeostatic imbalance. Sympathetic neurons within the brainstem project to and excite neurons in the hypothalamus, leading it to secrete corticotropin-releasing factor (CRF) that acts on the pituitary gland and signals the secretion of adrenocorticotropic hormone (ACTH) [96]. The pituitary is connected to the circulatory system, so once the secreted hormone, ACTH, enters the bloodstream, it is transported throughout the body. Once it reaches the adrenal gland, it works to stimulate the synthesis and release of GCs from the outer adrenal cortex. Interestingly, activation of the HPA axis does not necessarily require stimulus from sympathetic neurons and can be activated by both real and predicted threats. The response to a predicted threat is the body anticipating homoeostatic imbalance and can be triggered by species-specific innate fear or through conditioning and memory [97]. Many of the nodes of the HPA axis can be inhibited or stimulated by other brain regions, such as the hippocampus (see Fig. 1) [98, 99], PFC [100], and amygdala [101, 102]. Generally speaking, the hippocampus and PFC inhibit HPA activation and the amygdala stimulates HPA activation [103] but there are differences within subregions.

Through pharmacological experiments, scientists have shown that HPA axis activation during acute stress and subsequent GC release induces preparation of stored energy reserves for use and inhibition of glucose (energy) use in peripheral tissues [104]. This is important for the body to be prepared to expend energy and act in response to any threats presented by the stressful situation. One of the most important functions of GCs (most notably, cortisol) is regulation of their own secretion at multiple junctures in the HPA axis through negative feedback mechanisms [105]. Cortisol has been shown to inhibit production of both CRF from the hypothalamic paraventricular nucleus (PVN) [106, 107] and ACTH from corticotroph cells of the anterior pituitary [108, 109]. Also, cortisol can act directly on steroidogenic cells within the zona fasciculata of the adrenal cortex. It is been shown both in vitro and in vivo that GC presence in the adrenal gland can limit ACTH-induced cortisol synthesis and release of GCs [110, 111]. GR is present within these cells, suggesting a local feedback mechanism driven by activated GR translocating into the nucleus and modulating expression of genes regulating steroidogenesis and cytoskeleton reorganization [112, 113]. These mechanisms govern the homeostatic balance of HPA axis activity and facilitate the basal vicissitudes of GC secretion in the body.

The circadian rhythm is another important factor that controls HPA axis activation. Similar to other hormones, GC release follows a cyclic pattern with peak GC concentrations observed at the onset of the active period (i.e., early morning for diurnal animals, just after sunset for nocturnal animals) and the trough observed at the onset of the inactive period (i.e., just after sunset for diurnal animals, early morning for nocturnal animals). These rhythms are regulated by “master clock” systems that include light-sensitive neurons in the retina and the suprachiasmatic nucleus (SCN) of the hypothalamus. Neuroendocrine cells in the SCN project to and stimulate the CRF/arginine-vasopressin-containing neurons of the PVN to secrete CRF [114], activating the HPA axis cascade described previously in this review. In addition, SCN neurons can modulate the sensitivity of the adrenal gland to ACTH via its connection to preautonomic neurons of the PVN [115]. Studies transecting splanchnic nerves observed adrenal glands with decreased sensitivity to ACTH, implicating the nerve in this pathway [116].

Reciprocally, cyclic GC release facilitates the synchronization of intrinsic molecular patterns. GCs have been found to regulate transcription of clock proteins within peripheral tissues [117], immune cells [118], and neurons in the PFC [119], hippocampus [120], and amygdala [121]. Furthermore, the time-of-day-dependent fluctuations in GC levels in these brain regions lead to changes in formation and elimination of dendritic spines on neurons [122, 123]. These findings provide evidence that GCs are critical mediators of neuron function and behavior across the circadian cycle [124]. Intriguingly, GC feedback to the SCN appears to be promoted by astrocytes [125], as SCN neurons of adult rodents are some of the few cells that do not express GR [126].

With the integrated physiological actions of circadian rhythm and HPA axis, it may come as no surprise that disruptions of the circadian rhythm are associated with altered HPA axis activity and stress-related psychiatric disorders. For instance, reduced clock gene mRNA was observed in postmortem PFC tissue of patients with MDD [127] and abnormal diurnal cortisol patterns were observed in patients with PTSD [128]. Other studies have linked both knockdown of the clock protein and chronic stress with disruptions in mood-related behaviors [129, 130].

The tie between the nervous system and the immune system most likely arose as an evolutionary mechanism, as historical stressors were likely more physically threatening and may have resulted in injury. In this context, activation of the immune system following stressor exposure allows the body to prepare an immune response to any wounds inflicted and likely would have promoted survival of the organism [131‒133]. To that point, short-term stress can mobilize immune cells to enhance innate and adaptive immune responses. For example, acute stressors have been shown to traffic and redistribute leukocytes from the blood to organs like the skin, lymph nodes, and bone marrow [134, 135], and this enhanced immunity in those organs [136]. Interestingly though, when stress becomes chronic, it has the paradoxical effect of suppressing the immune system. Indeed, a paradigm-shifting study conducted by Dhabhar and McEwen analyzing an immune response in the pinnae of rats after stress exposure showed that while acute stress increased the redeployment of blood lymphocytes to the pinnae, chronic stress produced the opposite effect – a decrease in the number of blood leukocytes trafficked to the pinnae [137]. Reinforcing Selye’s concept of eustress and distress, this study led to a more nuanced understanding of stress effects on immune function in which short-term stress enhances immune reactivity and long-term (chronic) stress diminishes immune responsivity. It was later found that chronic stress also leads to dysregulation of cytokines and related immune responses [138, 139]. So, it has been proposed that the effect of stress on the immune system is dependent on the type and duration of the stressor [137, 140]. In this context, it is important to define how individual components of the stress response (i.e., the ANS and the HPA axis) contribute to differing effects on the peripheral immune system.

Neuroanatomical studies of innervation of the immune system have demonstrated that the ANS directly interacts with immune organs, including the thymus [141], spleen [142], lymph nodes [143], and bone marrow [144]. This is mainly regarding the SNS, as no evidence has been found for PNS innervation of immune organs. Those studies have shown that there are direct sympathetic projections to all primary and secondary immune organs [145], and it is through these conduits that the SNS enacts its effects on the peripheral nervous system via release of catecholaminergic neurotransmitters [146]. Meanwhile, the PNS drives its effects on the peripheral immune system primarily through stimulation of the vagus cranial nerve [147]. When stimulated, acetylcholine is released by the efferent arms of the vagus nerve and binds to cholinergic receptors on macrophages, inhibiting cytokine release [148]. Interestingly, the afferent arms of the vagus nerve respond to fluctuations in peripheral cytokines, such as tumor necrosis factor (TNF), acting as a sensor of peripheral immune changes [149]. The combination of the afferent and efferent vagal fibers creates a vagal-immune connection called the cholinergic anti-inflammatory pathway [150, 151].

Stress has been shown to alter both arms of the ANS. In terms of the SNS, chronic stress increased innervation of the paracortex of lymph nodes and increased acceleration of immunopathogenesis [152]. Bone marrow production of pro-inflammatory monocytes and myeloid-derived suppressor cells has also shown to be upregulated with chronic stress – another SNS-induced stress effect [153, 154]. Acute stress has also been shown to cause changes in immune cell distribution throughout the periphery. One study in mice subjected to experimental autoimmune encephalomyelitis showed a reduction of leukocytes in the lymph nodes as well as an increase of leukocytes in bone marrow following acute stress [155]. When it comes to the PNS, it was found that acute restraint stress was able to protect mouse kidneys from ischemia-reperfusion injury via the cholinergic anti-inflammatory pathway and that C1 neurons of the medulla oblongata were required for this protective effect [156]. These studies highlight not only the wide effects stress can have on the periphery, but the importance that the duration has on the outcome (acute vs. chronic).

Another function of GC release during chronic stress involves regulating aspects of peripheral immunity. Historically, GCs have been characterized as primarily anti-inflammatory in nature because they are capable of inducing apoptosis of some immune cells, and synthetic GCs have been successfully used to treat autoimmune disorders for many years [157]. Some other examples of the suppressive nature of GCs include their ability to inhibit antigen presentation and reduce proliferation of B cells [95]. However, even though evidence has found an anti-inflammatory role for GCs, evidence to the contrary has been found. In some contexts, they can increase the likelihood of survival of some immune cells [95, 134, 158]. Initial findings suggested that GCs suppress maturation (negative selection) of T lymphocytes, but further studies showed that they can also promote maturation of T cells (positive selection) by working in concert with other mechanisms of T-cell selection [159]. As previously mentioned, short-term stress (or short-term increase in GC secretion) can increase immune reactivity, while chronic stress (persistent increase in GC secretion) can dampen immune responses [136, 137]. This is supported by studies looking at varying levels of GCs; it appears that it can have opposing roles depending on the concentration administered and the duration of the administration [95, 160, 161].

GCs mainly enact their effects via binding to GRs or mineralocorticoid receptors (MRs) [162] which are expressed by most cell types in the body [163]. Both GR and MR are nuclear receptors, meaning after they are bound by GCs, they translocate to the nucleus to regulate gene transcription [164]. Activated GR can exert anti-inflammatory effects through transcriptional regulation of immune-related genes containing a GC regulatory element [165]. Another means by which GR can regulate immune-related transcription is by interfering with pro-inflammatory transcription factors such as nuclear factor-κB and activator protein 1 (AP-1) in a process called tethering [166]. It should be noted that not all GC-mediated effects are genomic [167]. Non-genomic effects of GCs such as changes in intracellular calcium and activation of kinase cascades (MAPK, ERK, etc.) are regulated by membrane-associated GRs and MRs [168, 169]. Through these mechanisms, GCs can direct cell-type-specific effects that lead to alterations in both the innate and adaptive immune systems [170, 171].

This review has extensively covered the effects of GCs secreted from the adrenal gland following HPA axis stimulation, but the adrenal gland can release other soluble factors that regulate immune function, namely, cytokines. Early studies revealed that stress elicits the production of interleukin (IL)-18, also known as interferon-ɣ-inducing factor, in the adrenal cortex, which directly impacts innate and adaptive immune responses [172‒174]. The inactive form of IL-18 is constitutively synthesized in a variety of cell types including cortical cells in the zona fasciculata of the adrenal gland [175, 176]. Upon processing primarily done by caspase-1 [177, 178], the mature and biologically active form of IL-18 can be secreted to bind to IL-18 receptors on target cells. IL-18 receptor signaling is primarily pro-inflammatory as it stimulates NK-κB-dependent transcription [179]. After stress exposure, levels of IL-18 and caspase-1 have been shown to increase in the adrenal gland as well as in circulating serum levels, suggesting HPA activation leads to production and secretion of IL-18 from the adrenal gland [180]. Heightened systemic IL-18 levels are not only observed following stress, but in patients diagnosed with MDD as well [181, 182]. These changes in IL-18 signaling may contribute to stress-related changes in cytokines, which will be discussed in subsequent sections.

Alongside the establishment of multiple neuroimmune pathways in the 1990s and 2000s [183], evidence began to show that chronic and severe stress is associated with altered levels of cytokines such as IL-1β, IL-6, TNF alpha, and C-reactive protein (CRP) in blood and cerebrospinal fluid samples [184, 185], and that these changes corresponded with behavioral and cognitive deficits [186]. Multiple studies have shown that many of these same cytokines are shown to be increased in patients with stress-related psychiatric disorders such as depression [187, 188] and PTSD [189, 190].

The first group to study this discovered a correlation between cytokine levels in the blood and reduced hippocampal volume in patients with MDD [191], which is interesting considering that high levels of cortisol or inflammatory cytokines have been shown to impair neuroplasticity in the hippocampus. This work led scientists to perform clinical studies testing anti-inflammatory interventions for MDD treatment. Initial studies using infliximab (anti-TNF) [192] and celecoxib (NSAID) [193] showed modest effects, but post hoc analyses suggested that those with higher levels of CRP had higher response rates. Another more recent clinical study subcategorized the depressive patient group into the “inflamed depression subgroup” and the “uninflamed depression subgroup.” They observed that depression was more severe in the inflamed subgroup and these patients exhibited increased numbers of several immune cell subtypes (i.e., monocytes, CD4+ cells, neutrophils) as well as increased CRP and IL-6 [194]. This evidence all suggests that not only is immune dysfunction connected to chronic stress, but that it may be implicated in the pathophysiology of depression [195].

As discussed above, initial studies examined peripheral immune cells because these samples were readily accessible. However, with advances in preclinical models and experimental methods, the field began to examine stress effects on immune function in the brain. Preclinical studies have found evidence that behavioral and cognitive changes caused by stress can be prevented via block of CNS cytokine activity [196, 197]. This work builds on seminal studies that demonstrated immune responses to infection drive behavioral alterations, namely, “sickness behavior” [198‒200]. These studies have guided emerging theories that link dysregulation of peripheral and central immune systems in the etiology of psychiatric disorders, particularly MDD [91, 201]. These connections provide the impetus to study how stress affects immunity in the brain and determine if it is involved in the link between immune dysfunction and psychiatric disorders.

Stress-induced dysregulation of neuroimmune function is in part due to the effects of GCs in the brain. As stated above, these bind to both GRs and MRs. The fact that GR and MR have opposing actions explains the inverse U shape of GR action in which intermediate (basal) levels of GCs have opposite results from the extreme ends (no GC and high GC). For instance, acute stress levels of GCs were sufficient and necessary to limit damage following LPS infusion into the brain [202] showing that the basal levels of GCs following acute response are anti-inflammatory in nature [203]. Strikingly, this effect is reversed in chronic stress or high GC concentrations as GCs appear to exacerbate cytotoxic inflammation following either physiological stress [204, 205], psychological stress [206], or prolonged exposure to high levels of GCs [207]. Timing can also determine whether GCs promote pro- or anti-inflammatory effects, as it was shown that if stress-like levels of corticosterone are delivered prior to LPS administration, the immune response (e.g., IL-1β, TNF alpha, IL-6 cytokine release) is enhanced in the hippocampus. However, if the stress-like levels of corticosterone are delivered after LPS administration, the immune response is reduced [208].

Microglia are brain-resident macrophages with diverse functions such as surveilling the CNS for pathogens and maintaining homeostasis through interactions with neurons, astrocytes, and oligodendrocytes [209]. Originally thought to switch between a ramified “resting” state and an ameboid “activated” state, recent studies have shown heterogenous phenotypes in various contexts, reflecting the highly dynamic nature of microglia [210‒212]. Stress exposure leads to changes in many aspects that cause microglia to respond as they attempt to maintain homeostasis. Acute stress studies have shown that morphological changes occur in microglia [213], but that they are not solely dependent on GCs [214]. In addition to changes in GC signaling [215], microglia have been found to respond to changes in neuronal activity via neuron-derived adenosine triphosphate signaling [216] and changes in norepinephrine concentration [217], both of which also change in multiple brain regions following stress [218, 219]. Other studies have implicated microglia in local cytokine production as the antibiotic minocycline reduced IL-1β levels in the hippocampus following stress exposure [220‒222].

Contrasting acute stress, chronic stress exposure sees a decline in beneficial aspects. In response to chronic stress, higher activated microglia counts are observed (Fig. 1) [206, 223]. Further studies suppressing microglial activation found that this manipulation could ameliorate stress-induced memory deficits following CUS [224]. Furthermore, microglia engulf more dendritic material following CUS signifying a greater proportion of microglia are taking on a phagocytic state following stress [225]. The mechanisms driving increased phagocytosis are not fully understood, but it is proposed that this may be a response to physiological changes in neurons and an attempt to establish homeostatic brain activity [226].

In the healthy brain, neuron-microglia cross-talk is critical for maintaining the functional surveying state of microglia [227]. Notably, neurons release several soluble factors such as adenosine triphosphate [228], colony-stimulating factor 1 (CSF1) [229], IL-34 [230], and fractalkine (CX3CL1) [231] that bind to receptors expressed by microglia. Disruption of these communication pathways significantly changes stress effects. Pharmacological blockage of P2YR12, a microglial receptor that facilitates microglial process contact with neurons [232], results in limited dendritic spine loss on apical dendrites of pyramidal neurons within the PFC, implicating P2YR12 as a critical mediator of stress-induced morphological changes [233]. Another important signaling molecule involved in this cell-cell communication is CSF1. Exposure to CUS increased neuronal CSF1 levels in the PFC, and this was associated with microglial phagocytosis of neuronal elements and decreased dendritic spine density. Further studies showed that viral-mediated knockdown of CSF1 mitigated the synaptic and behavioral deficits following chronic stress (see Fig. 1) [62].

As previously discussed, chronic stress can result in enhanced myelopoiesis [135, 234] and redistributed immune cells throughout the periphery [154, 235]. Some studies suggest that these peripheral immune alterations promote monocyte trafficking to the brain, but this is observed primarily in the repeated social defeat model [236]. The presence of peripheral monocytes in stress-responsive brain regions such as the amygdala and hippocampus can promote anxiety-like and depressive-like behavioral responses with stress [237, 238]. Recently, a study using a social defeat model found that both circulating monocytes and brain-trafficking monocytes within stress-susceptible mice exhibited increased expression of a myeloid-cell-specific proteinase called matrix metalloproteinase 8 (Mmp8) [239]. More so, they found that Mmp8 deletion attenuated neurophysiological and behavioral changes typically observed following chronic social defeat, further implicating monocytes in the neurobiology of stress. In contrast, other studies report that peripheral monocyte recruitment to the brain is not necessary for the behavioral alterations and microglial morphological changes that accompany social defeat exposure [226, 240]. These varied outcomes may be attributed to differences in stress models as well as experimental approaches.

Historically, microglia were largely regarded as a homogenous cell population in which each cell can shift between a “resting” and “activated” form [241]. However, the past few decades have seen remarkable advancements in microglia research including recent studies uncovering the heterogeneity of microglia across different brain regions [242‒244] and how they take on specific forms in response to various diseases [245, 246]. This is relevant because stress is known to elicit brain region-specific changes in microglial density and morphology [247]. Further experiments using advanced approaches (i.e., scRNA-Seq) are needed to define region-specific microglial responses to stress and to understand their impact on associated neurobiology. In addition to region-specific microglia differences, neuronal circuit-specific stress-induced effects are another growing area of interest. Optogenetic studies stimulating connections between limbic structures have shown increased neuronal activity can reduce anxiolytic and depressive phenotypes induced by chronic stress (see Fig. 1) [248‒250]. Continued studies investigating other connections and cell types (excitatory vs. inhibitory neurons) are required for further elucidation of stress-induced circuit-specific neurobiological changes.

Niches are restricted tissue microenvironments that maintain adult stem cells [251]. The heterogeneity of bone marrow and the study of bone marrow niches has been an emerging area of research for years [252]. Of particular interest to neuroimmunology, there has been increased attention given to the connection between the bone marrow of the skull and the meninges. Recently, with the use of tissue clearing and whole-body immunolabeling methods (vDISCO), a subset of short vascular connections between skull bone marrow and outer surfaces of the meninges were discovered and coined skull-meninges connections [253, 254]. Previously, it was thought that meningeal immune cells, such as myeloid cells and B cells, were supplied by systemic circulation, but recent developments have shown that these cells originate from the bone marrow of the skull [255, 256]. These intriguing results lead to the question, what role could skull bone marrow-derived myeloid cells have in the neurobiological changes associated with stress?

It is critical to point out that many studies highlighted in this review have been conducted in male rodents. There is substantial evidence that there are sex differences in the neurobiological effects of stress [257]. For instance, neuronal morphology of pyramidal neurons of the PFC after restraint stress or CUS has been shown to differ by sex [62, 258]. Microglia transcriptional changes following stress also appear to be sexually dimorphic [259, 260]. Sex-specific studies have ignited interest in investigating how sex hormones act as signaling molecules in the brain [261]. Estradiol has been of particular interest as it was shown that estrogen receptors are widely distributed throughout the brain [262], have neuroprotective effects [263], and reduce stress effects when administered during stress regiments [258]. Also, studies investigating intrinsic connectivity networks subject to stress-induced changes [264] such as the default mode network [265], salience network [266], and executive control network have shown fluctuations in functional connectivity during the menstrual cycle [267], a reoccurring process in which the ovaries secrete varying estradiol levels. However, other estrogens, androgens, and progestogens, along with their active metabolites, remain understudied [268] particularly in neuroimmune and stress contexts.

The goal of this review was to provide historical context for research in the neurobiology of stress and psychoneuroimmunology. The studies discussed here have revealed how neuroimmune systems interface with the brain to affect behavior, but there is still much more to uncover about the function of these dynamic biological systems. Technological advancements in research will lead to new insights, but it is important to consider how our current work can be informed by or related to past studies. The history of stress is a collection of individual observations, some rigorously planned and some serendipitous, but all aid in understanding the physiological and behavioral changes caused by external pressures and internal perceptions. These experiences are a fundamental part of the human condition and validates our existence. In the words of Hans Selye, “The absence of stress is death.”

The authors would like to thank BioRender for their software which allowed us to create our timeline figure. The authors would also like to thank Justin L. Bollinger, Lauren Larke Vollmer, and David T. Dadosky for their input on figure design.

The authors have declared no conflict of interest exists.

This work was enabled by generous support from the University of Cincinnati College of Medicine and funding from the National Institutes of Health (R01 MH123545; MH130399; T32 NS007453-22).

Alexander M. Kuhn: writing, editing, and figure design; Kelly E. Bosis: writing, editing, and figure editing; Eric S. Wohleb, PhD: conceptualization, editing, review, and figure editing.

1.
McEwen
BS
,
Wingfield
JC
.
The concept of allostasis in biology and biomedicine
.
Horm Behav
.
2003
;
43
(
1
):
2
15
.
2.
Selye
H
.
The evolution of the stress concept
.
Am Sci
.
1973
;
61
(
6
):
692
9
.
3.
James
W
.
What is an Emotion
.
Mind
.
1884
;
9
:
188
205
.
4.
Cannon
WB
.
The james-lange theory of emotions: a critical examination and an alternative theory
.
Am J Psychol
.
1927
;
39
(
1/4
):
106
24
.
5.
Cannon
W
Bodily changes in pain, hunger, fear and rage, an account of recent researches into the function of emotional excitement
;
1915
; p.
1915
.
6.
Cannon
W
.
Organization for physiological homeostasis
.
Physiol Rev
.
1929
;
IX
(
3
).
7.
Bernard
C
.
Leçons sur les phénomènes de la vie communs aux animaux et aux végétaux
.
Librairie J.-B. Baillière et Fils
;
1879
.
8.
Selye
H
.
A syndrome produced by diverse nocuous agents
.
Nature
.
1936
;
138
(
3479
):
32
.
9.
Selye
H
.
The stress of life
.
New York
:
McGraw-Hill
;
1956
.
10.
Selye
H
.
The alarm reaction, the general adaptation syndrome, and the role of stress and of the adaptive hormones in dental medicine
.
Oral Surg Oral Med Oral Pathol
.
1954
;
7
(
4
):
355
67
.
11.
Selye
H
.
Forty years of stress research: principal remaining problems and misconceptions
.
Can Med Assoc J
.
1976
;
115
(
1
):
53
6
.
12.
Leidy
NK
.
A physiological analysis of stress and chronic illness
.
J Adv Nurs
.
1989
;
14
(
10
):
868
76
.
13.
Czubalski
.
Zentralblatt fur Physiologie
. vol.
xxvii
,
1913
. p.
580
.
14.
Starkenstein
E
.
Der Mechanismus der Adrenalinwirkung
.
Z Exp Pathol Ther
.
1911
;
10
(
1
):
78
119
.
15.
Ingle
D
,
Edward
C
.
Kendall: a biographical memoir
.
National Academy of Sciences
;
1975
.
16.
Sarett
LH
.
Partial synthesis of PREGNENE-4-TRIOL-17(β),20(β),21-DIONE-3,11 and PREGNENE-4-DIOL-17(β),21-TRIONE-3,11,20 monoacetate
.
J Biol Chem
.
1946
;
162
(
3
):
601
31
.
17.
Nicolaides
NC
,
Chrousos
GP
.
Glucocorticoid signaling pathway: from bench to bedside
.
Int J Mol Sci
.
2023
;
24
(
13
):
11030
.
18.
Selye
H
.
Thymus and adrenals in the response of the organism to injuries and intoxications
.
Br J Exp Pathol
.
1936
;
17
:
234
48
.
19.
Guillemin
R
.
Peptides in the brain: the new endocrinology of the neuron
.
Science
.
1978
;
202
(
4366
):
390
402
.
20.
Harris
GW
.
Effects of the nervous system on the pituitary-adrenal activity
.
Prog Brain Res
.
1970
;
32
:
86
8
.
21.
Schally
AV
,
Arimura
A
,
Kastin
AJ
.
Hypothalamic regulatory hormones
.
Science
.
1973
;
179
(
4071
):
341
50
.
22.
Mcewen
BS
,
Weiss
JM
,
Schwartz
LS
.
Selective retention of corticosterone by limbic structures in rat brain
.
Nature
.
1968
;
220
(
5170
):
911
2
.
23.
Mcewen
BS
,
Plapinger
L
.
Association of 3H corticosterone-1,2 with macromolecules extracted from brain cell nuclei
.
Nature
.
1970
;
226
(
5242
):
263
5
.
24.
Gerlach
JL
,
McEwen
BS
,
Pfaff
DW
,
Moskovitz
S
,
Ferin
M
,
Carmel
PW
, et al
.
Cells in regions of rhesus monkey brain and pituitary retain radioactive estradiol, corticosterone and cortisol differentially
.
Brain Res
.
1976
;
103
(
3
):
603
12
.
25.
Jensen
EV
.
On the mechanism of estrogen action
.
Perspect Biol Med
.
1962
;
6
(
1
):
47
60
.
26.
Landfield
PW
,
Waymire
JC
,
Lynch
G
.
Hippocampal aging and adrenocorticoids: quantitative correlations
.
Science
.
1978
;
202
(
4372
):
1098
102
.
27.
Brunson
KL
,
Kramár
E
,
Lin
B
,
Chen
Y
,
Colgin
LL
,
Yanagihara
TK
, et al
.
Mechanisms of late-onset cognitive decline after early-life stress
.
J Neurosci
.
2005
;
25
(
41
):
9328
38
.
28.
McEwen
BS
.
Stress and hippocampal plasticity
.
Annu Rev Neurosci
.
1999
;
22
:
105
22
.
29.
Pham
K
,
Nacher
J
,
Hof
PR
,
McEwen
BS
.
Repeated restraint stress suppresses neurogenesis and induces biphasic PSA-NCAM expression in the adult rat dentate gyrus
.
Eur J Neurosci
.
2003
;
17
(
4
):
879
86
.
30.
Malberg
JE
,
Eisch
AJ
,
Nestler
EJ
,
Duman
RS
.
Chronic antidepressant treatment increases neurogenesis in adult rat Hippocampus
.
J Neurosci
.
2000
;
20
(
24
):
9104
10
.
31.
Bremner
JD
,
Randall
P
,
Scott
TM
,
Bronen
RA
,
Seibyl
JP
,
Southwick
SM
, et al
.
MRI-based measurement of hippocampal volume in patients with combat-related posttraumatic stress disorder
.
Am J Psychiatry
.
1995
;
152
(
7
):
973
81
.
32.
Bremner
JD
,
Narayan
M
,
Anderson
ER
,
Staib
LH
,
Miller
HL
,
Charney
DS
.
Hippocampal volume reduction in major depression
.
Aust J Pharm
.
2000
;
157
(
1
):
115
8
.
33.
Sánchez
MM
,
Young
LJ
,
Plotsky
PM
,
Insel
TR
.
Distribution of corticosteroid receptors in the rhesus brain: relative absence of glucocorticoid receptors in the hippocampal formation
.
J Neurosci
.
2000
;
20
(
12
):
4657
68
.
34.
Cook
SC
,
Wellman
CL
.
Chronic stress alters dendritic morphology in rat medial prefrontal cortex
.
J Neurobiol
.
2004
;
60
(
2
):
236
48
.
35.
Liston
C
,
Miller
MM
,
Goldwater
DS
,
Radley
JJ
,
Rocher
AB
,
Hof
PR
, et al
.
Stress-induced alterations in prefrontal cortical dendritic morphology predict selective impairments in perceptual attentional set-shifting
.
J Neurosci
.
2006
;
26
(
30
):
7870
4
.
36.
Radley
JJ
,
Sisti
HM
,
Hao
J
,
Rocher
AB
,
McCall
T
,
Hof
PR
, et al
.
Chronic behavioral stress induces apical dendritic reorganization in pyramidal neurons of the medial prefrontal cortex
.
Neuroscience
.
2004
;
125
:
1
6
.
37.
Radley
JJ
,
Rocher
AB
,
Miller
M
,
Janssen
WGM
,
Liston
C
,
Hof
PR
, et al
.
Repeated stress induces dendritic spine loss in the rat medial prefrontal cortex
.
Cereb Cortex
.
2006
;
16
(
3
):
313
20
.
38.
Šimić
G
,
Tkalčić
M
,
Vukić
V
,
Mulc
D
,
Španić
E
,
Šagud
M
, et al
.
Understanding emotions: origins and roles of the amygdala
.
Biomolecules
.
2021
;
11
(
6
):
823
.
39.
Vyas
A
,
Mitra
R
,
Shankaranarayana Rao
BS
,
Chattarji
S
.
Chronic stress induces contrasting patterns of dendritic remodeling in hippocampal and amygdaloid neurons
.
J Neurosci
.
2002
;
22
(
15
):
6810
8
.
40.
Lakshminarasimhan
H
,
Chattarji
S
.
Stress leads to contrasting effects on the levels of brain derived neurotrophic factor in the hippocampus and amygdala
.
PLoS One
.
2012
;
7
(
1
):
e30481
.
41.
Ader
R
.
On the development of psychoneuroimmunology
.
Eur J Pharmacol
.
2000
;
405
(
1–3
):
167
76
.
42.
Ader
R
,
Cohen
N
.
Behaviorally conditioned immunosuppression
.
Psychosom Med
.
1975
;
37
(
4
):
333
40
.
43.
Besedovsky
H
,
Sorkin
E
,
Felix
D
,
Haas
H
.
Short papers
.
Eur J Immunol
.
1977
;
7
(
5
):
323
5
.
44.
Besedovsky
H
,
del Rey
A
,
Sorkin
E
,
Da Prada
M
,
Burri
R
,
Honegger
C
.
The immune response evokes changes in brain noradrenergic neurons
.
Science
.
1983
;
221
(
4610
):
564
6
.
45.
Riley
V
.
Psychoneuroendocrine influences on immunocompetence and neoplasia
.
Science
.
1981
;
212
(
4499
):
1100
9
.
46.
Ader
R
.
Psychoneuroimmunology
. In:
Ballieux
RE
,
Fielding
JF
,
L’Abbate
A
, editors.
Breakdown in human adaptation to “stress”: towards a multidisciplinary approach volume II
.
Dordrecht
:
Springer Netherlands
;
1984
. p.
653
70
.
47.
Khansari
DN
,
Murgo
AJ
,
Faith
RE
.
Effects of stress on the immune system
.
Immunol Today
.
1990
;
11
:
170
5
.
48.
Kronfol
Z
,
House
JD
,
Silva
J
,
Greden
J
,
Carroll
BJ
.
Depression, urinary free cortisol excretion and lymphocyte function
.
Br J Psychiatry
.
1986
;
148
:
70
3
.
49.
Besedovsky
H
,
del Rey
A
,
Sorkin
E
,
Dinarello
CA
.
Immunoregulatory feedback between interleukin-1 and glucocorticoid hormones
.
Science
.
1986
;
233
(
4764
):
652
4
.
50.
Schleifer
SJ
,
Keller
SE
,
Meyerson
AT
,
Raskin
MJ
,
Davis
KL
,
Stein
M
.
Lymphocyte function in major depressive disorder
.
Arch Gen Psychiatry
.
1984
;
41
(
5
):
484
6
.
51.
Atrooz
F
,
Alkadhi
KA
,
Salim
S
.
Understanding stress: insights from rodent models
.
Curr Res Neurobiol
.
2021
;
2
:
100013
.
52.
Miczek
KA
.
A new test for aggression in rats without aversive stimulation: differential effects of d-amphetamine and cocaine
.
Psychopharmacology
.
1979
;
60
(
3
):
253
9
.
53.
Björkqvist
K
.
Social defeat as a stressor in humans
.
Physiol Behav
.
2001
;
73
(
3
):
435
42
.
54.
Hodes
GE
,
Pfau
ML
,
Leboeuf
M
,
Golden
SA
,
Christoffel
DJ
,
Bregman
D
, et al
.
Individual differences in the peripheral immune system promote resilience versus susceptibility to social stress
.
Proc Natl Acad Sci U S A
.
2014
;
111
(
45
):
16136
41
.
55.
Menard
C
,
Pfau
ML
,
Hodes
GE
,
Kana
V
,
Wang
VX
,
Bouchard
S
, et al
.
Social stress induces neurovascular pathology promoting depression
.
Nat Neurosci
.
2017
;
20
(
12
):
1752
60
.
56.
Katz
RJ
.
Animal model of depression: pharmacological sensitivity of a hedonic deficit
.
Pharmacol Biochem Behav
.
1982
;
16
(
6
):
965
8
.
57.
Willner
P
,
Towell
A
,
Sampson
D
,
Sophokleous
S
,
Muscat
R
.
Reduction of sucrose preference by chronic unpredictable mild stress, and its restoration by a tricyclic antidepressant
.
Psychopharmacology
.
1987
;
93
(
3
):
358
64
.
58.
Strekalova
T
,
Liu
Y
,
Kiselev
D
,
Khairuddin
S
,
Chiu
JLY
,
Lam
J
, et al
.
Chronic mild stress paradigm as a rat model of depression: facts, artifacts, and future perspectives
.
Psychopharmacology
.
2022
;
239
(
3
):
663
93
.
59.
Herman
JP
,
Adams
D
,
Prewitt
C
.
Regulatory changes in neuroendocrine stress-integrative circuitry produced by a variable stress paradigm
.
Neuroendocrinology
.
1995
;
61
(
2
):
180
90
.
60.
Willner
P
.
The Chronic Mild Stress (CMS) model of depression: history, evaluation and usage
.
Neurobiol Stress
.
2017
;
6
:
78
93
.
61.
Elizalde
N
,
Gil-Bea
FJ
,
Ramírez
MJ
,
Aisa
B
,
Lasheras
B
,
Del Rio
J
, et al
.
Long-lasting behavioral effects and recognition memory deficit induced by chronic mild stress in mice: effect of antidepressant treatment
.
Psychopharmacology
.
2008
;
199
:
1
14
.
62.
Wohleb
ES
,
Terwilliger
R
,
Duman
CH
,
Duman
RS
.
Stress-induced neuronal colony stimulating factor 1 provokes microglia-mediated neuronal remodeling and depressive-like behavior
.
Biol Psychiatr
.
2018
;
83
:
38
49
.
63.
Brodie
DA
,
Hanson
HM
.
A study of the factors involved in the production of gastric ulcers by the restraint technique
.
Gastroenterology
.
1960
;
38
(
3
):
353
60
.
64.
Rossi
G
,
Bonfils
S
,
Lieffogh
F
,
Lambling
A
.
Technique nouvelle pour produire des ulcerations gastriques chez le rat blanc: lulcère de contrainte
.
C R Seances Soc Biol Fil
.
1956
;
150
:
2124
6
.
65.
Paré
WP
,
Glavin
GB
.
Restraint stress in biomedical research: a review
.
Neurosci Biobehav Rev
.
1986
;
10
(
3
):
339
70
.
66.
Kim
K-S
,
Han
P-L
.
Optimization of chronic stress paradigms using anxiety- and depression-like behavioral parameters
.
J Neurosci Res
.
2006
;
83
(
3
):
497
507
.
67.
Magariños
AM
,
McEwen
BS
.
Stress-induced atrophy of apical dendrites of hippocampal CA3c neurons: involvement of glucocorticoid secretion and excitatory amino acid receptors
.
Neuroscience
.
1995
;
69
(
1
):
89
98
.
68.
Shansky
RM
,
Hamo
C
,
Hof
PR
,
McEwen
BS
,
Morrison
JH
.
Stress-induced dendritic remodeling in the prefrontal cortex is circuit specific
.
Cereb Cortex
.
2009
;
19
(
10
):
2479
84
.
69.
Lee
E-H
,
Park
J-Y
,
Kwon
H-J
,
Han
P-L
.
Repeated exposure with short-term behavioral stress resolves pre-existing stress-induced depressive-like behavior in mice
.
Nat Commun
.
2021
;
12
(
1
):
6682
.
70.
Bremner
JD
.
Long-term effects of childhood abuse on brain and neurobiology
.
Child Adolesc Psychiatr Clin N Am
.
2003
;
12
(
2
):
271
92
.
71.
Wilson
RS
,
Schneider
JA
,
Boyle
PA
,
Arnold
SE
,
Tang
Y
,
Bennett
DA
.
Chronic distress and incidence of mild cognitive impairment
.
Neurology
.
2007
;
68
(
24
):
2085
92
.
72.
Baumeister
D
,
Akhtar
R
,
Ciufolini
S
,
Pariante
CM
,
Mondelli
V
.
Childhood trauma and adulthood inflammation: a meta-analysis of peripheral C-reactive protein, interleukin-6 and tumour necrosis factor-α
.
Mol Psychiatry
.
2016
;
21
(
5
):
642
9
.
73.
Bolton
JL
,
Molet
J
,
Ivy
A
,
Baram
TZ
.
New insights into early-life stress and behavioral outcomes
.
Curr Opin Behav Sci
.
2017
;
14
:
133
9
.
74.
Millstein
RA
,
Holmes
A
.
Effects of repeated maternal separation on anxiety: and depression-related phenotypes in different mouse strains
.
Neurosci Biobehav Rev
.
2007
;
31
(
1
):
3
17
.
75.
Rombaut
C
,
Roura-Martinez
D
,
Lepolard
C
,
Gascon
E
.
Brief and long maternal separation in C57Bl6J mice: behavioral consequences for the dam and the offspring
.
Front Behav Neurosci
.
2023
;
17
:
1269866
.
76.
Rice
CJ
,
Sandman
CA
,
Lenjavi
MR
,
Baram
TZ
.
A novel mouse model for acute and long-lasting consequences of early life stress
.
Endocrinology
.
2008
;
149
(
10
):
4892
900
.
77.
Walker
C-D
,
Bath
KG
,
Joels
M
,
Korosi
A
,
Larauche
M
,
Lucassen
PJ
, et al
.
Chronic early life stress induced by Limited Bedding and Nesting (LBN) material in rodents: critical considerations of methodology, outcomes and translational potential
.
Stress
.
2017
;
20
(
5
):
421
48
.
78.
Meaney
MJ
.
Maternal care, gene expression, and the transmission of individual differences in stress reactivity across generations
.
Annu Rev Neurosci
.
2001
;
24
(
1
):
1161
92
.
79.
de Kloet
ER
,
Sibug
RM
,
Helmerhorst
FM
,
Schmidt
M
.
Stress, genes and the mechanism of programming the brain for later life
.
Neurosci Biobehav Rev
.
2005
;
29
(
2
):
271
81
.
80.
Molet
J
,
Maras
PM
,
Avishai-Eliner
S
,
Baram
TZ
.
Naturalistic rodent models of chronic early-life stress
.
Dev Psychobiol
.
2014
;
56
(
8
):
1675
88
.
81.
Gilles
EE
,
Schultz
L
,
Baram
TZ
.
Abnormal corticosterone regulation in an immature rat model of continuous chronic stress
.
Pediatr Neurol
.
1996
;
15
(
2
):
114
9
.
82.
Dalle Molle
R
,
Portella
AK
,
Goldani
MZ
,
Kapczinski
FP
,
Leistner-Segal
S
,
Salum
GA
, et al
.
Associations between parenting behavior and anxiety in a rodent model and a clinical sample: relationship to peripheral BDNF levels
.
Transl Psychiatry
.
2012
;
2
(
11
):
e195
.
83.
Raineki
C
,
Cortés
MR
,
Belnoue
L
,
Sullivan
RM
.
Effects of early-life abuse differ across development: infant social behavior deficits are followed by adolescent depressive-like behaviors mediated by the amygdala
.
J Neurosci
.
2012
;
32
(
22
):
7758
65
.
84.
Plotsky
PM
,
Cunningham
ET
Jr
,
Widmaier
EP
.
Catecholaminergic modulation of corticotropin-releasing factor and adrenocorticotropin secretion
.
Endocr Rev
.
1989
;
10
(
4
):
437
58
.
85.
McEwen
BS
.
Protective and damaging effects of stress mediators
.
N Engl J Med Overseas Ed
.
1998
;
338
(
3
):
171
9
.
86.
Heim
C
,
Binder
EB
.
Current research trends in early life stress and depression: review of human studies on sensitive periods, gene–environment interactions, and epigenetics
.
Exp Neurol
.
2012
;
233
(
1
):
102
11
.
87.
Sapolsky
RM
.
Why stress is bad for your brain
.
Science
.
1996
;
273
(
5276
):
749
50
.
88.
Dantzer
R
,
Kelley
KW
.
Stress and immunity: an integrated view of relationships between the brain and the immune system
.
Life Sci
.
1989
;
44
(
26
):
1995
2008
.
89.
McEwen
BS
,
Stellar
E
.
Stress and the individual: mechanisms leading to disease
.
Arch Intern Med
.
1993
;
153
(
18
):
2093
101
.
90.
Capuron
L
,
Dantzer
R
.
Cytokines and depression: the need for a new paradigm
.
Brain Behav Immun
.
2003
;
17
(
Suppl 1
):
119
24
.
91.
Wohleb
ES
,
Franklin
T
,
Iwata
M
,
Duman
RS
.
Integrating neuroimmune systems in the neurobiology of depression
.
Nat Rev Neurosci
.
2016
;
17
(
8
):
497
511
.
92.
Langley
JN
The autonomic nervous system
.
Cambridge
:
W. HEFFER & SONS LTD
;
1921
. p.
10
.
93.
McCorry
LK
.
Physiology of the autonomic nervous system
.
Am J Pharm Educ
.
2007
;
71
(
4
):
78
.
94.
Ulrich-Lai
YM
,
Herman
JP
.
Neural regulation of endocrine and autonomic stress responses
.
Nat Rev Neurosci
.
2009
;
10
(
6
):
397
409
.
95.
Sapolsky
RM
,
Romero
LM
,
Munck
AU
.
How do glucocorticoids influence stress responses? Integrating permissive, suppressive, stimulatory, and preparative actions
.
Endocr Rev
.
2000
;
21
(
1
):
55
89
.
96.
Antoni
FA
.
Hypothalamic control of adrenocorticotropin secretion: advances since the discovery of 41-residue corticotropin-releasing factor
.
Endocr Rev
.
1986
;
7
(
4
):
351
78
.
97.
Herman
JP
,
Figueiredo
H
,
Mueller
NK
,
Ulrich-Lai
Y
,
Ostrander
MM
,
Choi
DC
, et al
.
Central mechanisms of stress integration: hierarchical circuitry controlling hypothalamo–pituitary–adrenocortical responsiveness
.
Front Neuroendocrinol
.
2003
;
24
(
3
):
151
80
.
98.
Herman
JP
,
Schafer
MK
,
Young
EA
,
Thompson
R
,
Douglass
J
,
Akil
H
, et al
.
Evidence for hippocampal regulation of neuroendocrine neurons of the hypothalamo-pituitary-adrenocortical axis
.
J Neurosci
.
1989
;
9
:
3072
82
.
99.
Jacobson
L
,
Sapolsky
R
.
The role of the Hippocampus in feedback regulation of the hypothalamic-pituitary-adrenocortical Axis
.
Endocr Rev
.
1991
;
12
(
2
):
118
34
.
100.
Diorio
D
,
Viau
V
,
Meaney
M
.
The role of the medial prefrontal cortex (cingulate gyrus) in the regulation of hypothalamic-pituitary-adrenal responses to stress
.
J Neurosci
.
1993
;
13
(
9
):
3839
47
.
101.
Matheson
GK
,
Branch
BJ
,
Taylor
AN
.
Effects of amygdaloid stimulation on pituitary-adrenal activity in conscious cats
.
Brain Res
.
1971
;
32
(
1
):
151
67
.
102.
Van de Kar
LD
,
Piechowski
RA
,
Rittenhouse
PA
,
Gray
TS
.
Amygdaloid lesions: differential effect on conditioned stress and immobilization-induced increases in corticosterone and renin secretion
.
Neuroendocrinology
.
1991
;
54
(
2
):
89
95
.
103.
Herman
JP
,
Ostrander
MM
,
Mueller
NK
,
Figueiredo
H
.
Limbic system mechanisms of stress regulation: hypothalamo-pituitary-adrenocortical axis
.
Prog Neuro Psychopharmacol Biol Psychiatr
.
2005
;
29
(
8
):
1201
13
.
104.
Munck
A
,
Guyre
PM
,
Holbrook
NJ
.
Physiological functions of glucocorticoids in stress and their relation to pharmacological actions
.
Endocr Rev
.
1984
;
5
(
1
):
25
44
.
105.
Gjerstad
JK
,
Lightman
SL
,
Spiga
F
.
Role of glucocorticoid negative feedback in the regulation of HPA axis pulsatility
.
Stress
.
2018
;
21
(
5
):
403
16
.
106.
Aguilera
G
,
Kiss
A
,
Liu
Y
,
Kamitakahara
A
.
Negative regulation of corticotropin releasing factor expression and limitation of stress response
.
Stress
.
2007
;
10
(
2
):
153
61
.
107.
Kovács
KJ
,
Makara
GB
.
Corticosterone and dexamethasone act at different brain sites to inhibit adrenalectomy-induced adrenocorticotropin hypersecretion
.
Brain Res
.
1988
;
474
(
2
):
205
10
.
108.
Duncan
PJ
,
Tabak
J
,
Ruth
P
,
Bertram
R
,
Shipston
MJ
.
Glucocorticoids inhibit CRH/AVP-Evoked bursting activity of male murine anterior pituitary corticotrophs
.
Endocrinology
.
2016
;
157
(
8
):
3108
21
.
109.
John
CD
,
Christian
HC
,
Morris
JF
,
Flower
RJ
,
Solito
E
,
Buckingham
JC
.
Annexin 1 and the regulation of endocrine function
.
Trends Endocrinol Metab
.
2004
;
15
(
3
):
103
9
.
110.
Carsia
RV
,
Malamed
S
.
Acute self-suppression of corticosteroidogenesis in isolated adrenocortical cells
.
Endocrinology
.
1979
;
105
(
4
):
911
4
.
111.
Langecker
H
,
Lurie
R
.
Die hemmung der corticotropin-sekretion durch steroide
.
Acta Endocrinol
.
1957
;
25
(
1
):
54
8
.
112.
Buckingham
JC
,
John
CD
,
Solito
E
,
Tierney
T
,
Flower
RJ
,
Christian
H
, et al
.
Annexin 1, glucocorticoids, and the neuroendocrine: immune interface
.
Ann N Y Acad Sci
.
2006
;
1088
(
1
):
396
409
.
113.
Gummow
BM
,
Scheys
JO
,
Cancelli
VR
,
Hammer
GD
.
Reciprocal regulation of a glucocorticoid receptor-steroidogenic factor-1 transcription complex on the dax-1 promoter by glucocorticoids and adrenocorticotropic hormone in the adrenal cortex
.
Mol Endocrinol
.
2006
;
20
(
11
):
2711
23
.
114.
Nader
N
,
Chrousos
GP
,
Kino
T
.
Interactions of the circadian CLOCK system and the HPA Axis
.
Trends Endocrinol Metab
.
2010
;
21
(
5
):
277
86
.
115.
Buijs
RM
,
la Fleur
SE
,
Wortel
J
,
van Heyningen
C
,
Zuiddam
L
,
Mettenleiter
TC
, et al
.
The suprachiasmatic nucleus balances sympathetic and parasympathetic output to peripheral organs through separate preautonomic neurons
.
J Comp Neurol
.
2003
;
464
(
1
):
36
48
.
116.
Ulrich-Lai
YM
,
Arnhold
MM
,
Engeland
WC
.
Adrenal splanchnic innervation contributes to the diurnal rhythm of plasma corticosterone in rats by modulating adrenal sensitivity to ACTH
.
Am J Physiol Regul Integr Comp Physiol
.
2006
;
290
(
4
):
R1128
35
.
117.
Almon
RR
,
Yang
E
,
Lai
W
,
Androulakis
IP
,
Ghimbovschi
S
,
Hoffman
EP
, et al
.
Relationships between circadian rhythms and modulation of gene expression by glucocorticoids in skeletal muscle
.
Am J Physiol Regul Integr Comp Physiol
.
2008
;
295
(
4
):
R1031
47
.
118.
Shimba
A
,
Ikuta
K
.
Glucocorticoids regulate circadian rhythm of innate and adaptive immunity
.
Front Immunol
.
2020
;
11
:
2143
.
119.
Woodruff
ER
,
Chun
LE
,
Hinds
LR
,
Spencer
RL
.
Diurnal corticosterone presence and phase modulate clock gene expression in the male rat prefrontal cortex
.
Endocrinology
.
2016
;
157
(
4
):
1522
34
.
120.
Bering
T
,
Blancas-Velazquez
AS
,
Rath
MF
.
Circadian clock genes are regulated by rhythmic corticosterone at physiological levels in the rat Hippocampus
.
Neuroendocrinology
.
2023
;
113
(
10
):
1076
90
.
121.
Segall
LA
,
Milet
A
,
Tronche
F
,
Amir
S
.
Brain glucocorticoid receptors are necessary for the rhythmic expression of the clock protein, PERIOD2, in the central extended amygdala in mice
.
Neurosci Lett
.
2009
;
457
(
1
):
58
60
.
122.
Ikeda
M
,
Hojo
Y
,
Komatsuzaki
Y
,
Okamoto
M
,
Kato
A
,
Takeda
T
, et al
.
Hippocampal spine changes across the sleep–wake cycle: corticosterone and kinases
.
J Endocrinol
.
2015
;
226
(
2
):
M13
27
.
123.
Perez-Cruz
C
,
Simon
M
,
Flügge
G
,
Fuchs
E
,
Czéh
B
.
Diurnal rhythm and stress regulate dendritic architecture and spine density of pyramidal neurons in the rat infralimbic cortex
.
Behav Brain Res
.
2009
;
205
(
2
):
406
13
.
124.
Miller
AM
,
Daniels
RM
,
Sheng
JA
,
Wu
TJ
,
Handa
RJ
.
Glucocorticoid regulation of diurnal spine plasticity in the murine ventromedial prefrontal cortex
.
J Neuroendocrinol
.
2022
;
34
(
12
):
e13212
.
125.
Lehmann
M
,
Haury
K
,
Oster
H
,
Astiz
M
.
Circadian glucocorticoids throughout development
.
Front Neurosci
.
2023
;
17
:
1165230
.
126.
Rosenfeld
P
,
Van Eekelen
JA
,
Levine
S
,
De Kloet
ER
.
Ontogeny of the type 2 glucocorticoid receptor in discrete rat brain regions: an immunocytochemical study
.
Brain Res
.
1988
;
470
(
1
):
119
27
.
127.
Li
JZ
,
Bunney
BG
,
Meng
F
,
Hagenauer
MH
,
Walsh
DM
,
Vawter
MP
, et al
.
Circadian patterns of gene expression in the human brain and disruption in major depressive disorder
.
Proc Natl Acad Sci U S A
.
2013
;
110
(
24
):
9950
5
.
128.
Yehuda
R
.
Biology of posttraumatic stress disorder
.
J Clin Psychiatry
.
2001
;
62
(
Suppl 17
):
41
6
.
129.
Mukherjee
S
,
Coque
L
,
Cao
J-L
,
Kumar
J
,
Chakravarty
S
,
Asaithamby
A
, et al
.
Knockdown of Clock in the ventral tegmental area through RNA interference results in a mixed state of mania and depression-like behavior
.
Biol Psychiatry
.
2010
;
68
(
6
):
503
11
.
130.
Logan
RW
,
Edgar
N
,
Gillman
AG
,
Hoffman
D
,
Zhu
X
,
McClung
CA
.
Chronic stress induces brain region-specific alterations of molecular rhythms that correlate with depression-like behavior in mice
.
Biol Psychiatry
.
2015
;
78
(
4
):
249
58
.
131.
Dhabhar
FS
.
The short-term stress response: mother nature’s mechanism for enhancing protection and performance under conditions of threat, challenge, and opportunity
.
Front Neuroendocrinol
.
2018
;
49
:
175
92
.
132.
Dhabhar
FS
,
Miller
AH
,
McEwen
BS
,
Spencer
RL
.
Effects of stress on immune cell distribution. Dynamics and hormonal mechanisms
.
J Immunol
.
1995
;
154
(
10
):
5511
27
.
133.
Dhabhar
FS
,
McEwen
BS
.
Stress-induced enhancement of antigen-specific cell-mediated immunity
.
J Immunol
.
1996
;
156
(
7
):
2608
15
.
134.
Dhabhar
FS
.
Enhancing versus suppressive effects of stress on immune function: implications for immunoprotection and immunopathology
.
Neuroimmunomodulation
.
2009
;
16
(
5
):
300
17
.
135.
Engler
H
,
Bailey
MT
,
Engler
A
,
Sheridan
JF
.
Effects of repeated social stress on leukocyte distribution in bone marrow, peripheral blood and spleen
.
J Neuroimmunol
.
2004
;
148
(
1–2
):
106
15
.
136.
Dhabhar
FS
,
McEwen
BS
.
Enhancing versus suppressive effects of stress hormones on skin immune function
.
Proc Natl Acad Sci U S A
.
1999
;
96
(
3
):
1059
64
.
137.
Dhabhar
FS
,
McEwen
BS
.
Acute stress enhances while chronic stress suppresses cell-mediated immunity in vivo: a potential role for leukocyte trafficking
.
Brain Behav Immun
.
1997
;
11
(
4
):
286
306
.
138.
Elenkov
IJ
,
Papanicolaou
DA
,
Wilder
RL
,
Chrousos
GP
.
Modulatory effects of glucocorticoids and catecholamines on human interleukin-12 and interleukin-10 production: clinical implications
.
Proc Assoc Am Physicians
.
1996
;
108
:
374
81
.
139.
Glaser
R
,
MacCallum
RC
,
Laskowski
BF
,
Malarkey
WB
,
Sheridan
JF
,
Kiecolt-Glaser
JK
.
Evidence for a shift in the Th-1 to Th-2 cytokine response associated with chronic stress and aging
.
J Gerontol A Biol Sci Med Sci
.
2001
;
56
(
8
):
M477
82
.
140.
Dhabhar
FS
.
Effects of stress on immune function: the good, the bad, and the beautiful
.
Immunol Res
.
2014
;
58
(
2–3
):
193
210
.
141.
Bulloch
K
,
Moore
RY
.
Innervation of the thymus gland by brain stem and spinal cord in mouse and rat
.
Am J Anat
.
1981
;
162
(
2
):
157
66
.
142.
Nance
DM
,
Burns
J
.
Innervation of the spleen in the rat: evidence for absence of afferent innervation
.
Brain Behav Immun
.
1989
;
3
(
4
):
281
90
.
143.
Felten
DL
,
Felten
SY
,
Carlson
SL
,
Olschowka
JA
,
Livnat
S
.
Noradrenergic and peptidergic innervation of lymphoid tissue
.
J Immunol
.
1985
;
135
(
2
):
755s
65s
.
144.
Bellinger
DL
,
Lorton
D
,
Felten
SY
,
Felten
DL
.
Innervation of lymphoid organs and implications in development, aging, and autoimmunity
.
Int J Immunopharmacol
.
1992
;
14
(
3
):
329
44
.
145.
Nance
DM
,
Sanders
VM
.
Autonomic innervation and regulation of the immune system (1987–2007)
.
Brain Behav Immun
.
2007
;
21
(
6
):
736
45
.
146.
Elenkov
IJ
,
Wilder
RL
,
Chrousos
GP
,
Vizi
ES
.
The sympathetic nerve--an integrative interface between two supersystems: the brain and the immune system
.
Pharmacol Rev
.
2000
;
52
(
4
):
595
638
.
147.
Borovikova
LV
,
Ivanova
S
,
Zhang
M
,
Yang
H
,
Botchkina
GI
,
Watkins
LR
, et al
.
Vagus nerve stimulation attenuates the systemic inflammatory response to endotoxin
.
Nature
.
2000
;
405
(
6785
):
458
62
.
148.
Bernik
TR
,
Friedman
SG
,
Ochani
M
,
DiRaimo
R
,
Ulloa
L
,
Yang
H
, et al
.
Pharmacological stimulation of the cholinergic antiinflammatory pathway
.
J Exp Med
.
2002
;
195
(
6
):
781
8
.
149.
Goehler
LE
,
Gaykema
RPA
,
Hansen
MK
,
Anderson
K
,
Maier
SF
,
Watkins
LR
.
Vagal immune-to-brain communication: a visceral chemosensory pathway
.
Auton Neurosci
.
2000
;
85
(
1–3
):
49
59
.
150.
Kelly
MJ
,
Breathnach
C
,
Tracey
KJ
,
Donnelly
SC
.
Manipulation of the inflammatory reflex as a therapeutic strategy
.
Cell Rep Med
.
2022
;
3
(
7
):
100696
.
151.
Tracey
KJ
.
The inflammatory reflex
.
Nature
.
2002
;
420
(
6917
):
853
9
.
152.
Sloan
EK
,
Capitanio
JP
,
Tarara
RP
,
Mendoza
SP
,
Mason
WA
,
Cole
SW
.
Social stress enhances sympathetic innervation of primate lymph nodes: mechanisms and implications for viral pathogenesis
.
J Neurosci
.
2007
;
27
(
33
):
8857
65
.
153.
Wohleb
ES
,
Powell
ND
,
Godbout
JP
,
Sheridan
JF
.
Stress-induced recruitment of bone marrow-derived monocytes to the brain promotes anxiety-like behavior
.
J Neurosci
.
2013
;
33
(
34
):
13820
33
.
154.
McKim
DB
,
Yin
W
,
Wang
Y
,
Cole
SW
,
Godbout
JP
,
Sheridan
JF
.
Social stress mobilizes hematopoietic stem cells to establish persistent splenic myelopoiesis
.
Cell Rep
.
2018
;
25
(
9
):
2552
62.e3
.
155.
Poller
WC
,
Downey
J
,
Mooslechner
AA
,
Khan
N
,
Li
L
,
Chan
CT
, et al
.
Brain motor and fear circuits regulate leukocytes during acute stress
.
Nature
.
2022
;
607
(
7919
):
578
84
.
156.
Abe
C
,
Inoue
T
,
Inglis
MA
,
Viar
KE
,
Huang
L
,
Ye
H
, et al
.
C1 neurons mediate a stress-induced anti-inflammatory reflex in mice
.
Nat Neurosci
.
2017
;
20
(
5
):
700
7
.
157.
Taves
MD
,
Ashwell
JD
.
Glucocorticoids in T cell development, differentiation and function
.
Nat Rev Immunol
.
2021
;
21
(
4
):
233
43
.
158.
Besedovsky
HO
,
del Rey
A
.
Immune-neuro-endocrine interactions: facts and hypotheses
.
Endocr Rev
.
1996
;
17
(
1
):
64
102
.
159.
Zacharchuk
CM
,
Merćep
M
,
Chakraborti
PK
,
Simons
SS
Jr
,
Ashwell
JD
.
Programmed T lymphocyte death. Cell activation: and steroid-induced pathways are mutually antagonistic
.
J Immunol
.
1990
;
145
(
12
):
4037
45
.
160.
Wiegers
GJ
,
Croiset
G
,
Reul
JM
,
Holsboer
F
,
de Kloet
ER
.
Differential effects of corticosteroids on rat peripheral blood T-lymphocyte mitogenesis in vivo and in vitro
.
Am J Physiol
.
1993
;
265
(
6 Pt 1
):
E825
830
.
161.
Wiegers
GJ
,
Labeur
MS
,
Stec
IE
,
Klinkert
WE
,
Holsboer
F
,
Reul
JM
.
Glucocorticoids accelerate anti-T cell receptor-induced T cell growth
.
J Immunol
.
1995
;
155
(
4
):
1893
902
.
162.
Reul
JM
,
Kloet
ERD
.
Two receptor systems for corticosterone in rat brain: microdistribution and differential occupation
.
Endocrinology
.
1985
;
117
(
6
):
2505
11
.
163.
Kadmiel
M
,
Cidlowski
JA
.
Glucocorticoid receptor signaling in health and disease
.
Trends Pharmacol Sci
.
2013
;
34
(
9
):
518
30
.
164.
Robertson
NM
,
Schulman
G
,
Karnik
S
,
Alnemri
E
,
Litwack
G
.
Demonstration of nuclear translocation of the Mineralocorticoid Receptor (MR) using an anti-MR antibody and confocal laser scanning microscopy
.
Mol Endocrinol
.
1993
;
7
(
9
):
1226
39
.
165.
J M Schaaf
M
,
Meijer
OC
.
Immune modulations by glucocorticoids: from molecular biology to clinical research
.
Cells
.
2022
;
11
(
24
):
4032
.
166.
Ratman
D
,
Vanden Berghe
W
,
Dejager
L
,
Libert
C
,
Tavernier
J
,
Beck
IM
, et al
.
How glucocorticoid receptors modulate the activity of other transcription factors: a scope beyond tethering
.
Mol Cell Endocrinol
.
2013
;
380
(
1–2
):
41
54
.
167.
Tasker
JG
,
Di
S
,
Malcher-Lopes
R
.
Minireview: rapid glucocorticoid signaling via membrane-associated receptors
.
Endocrinology
.
2006
;
147
(
12
):
5549
56
.
168.
Panettieri
RA
,
Schaafsma
D
,
Amrani
Y
,
Koziol-White
C
,
Ostrom
R
,
Tliba
O
.
NON-GENOMIC effects of glucocorticoids: an updated view
.
Trends Pharmacol Sci
.
2019
;
40
(
1
):
38
49
.
169.
Groeneweg
FL
,
Karst
H
,
de Kloet
ER
,
Joëls
M
.
Mineralocorticoid and glucocorticoid receptors at the neuronal membrane, regulators of nongenomic corticosteroid signalling
.
Mol Cell Endocrinol
.
2012
;
350
(
2
):
299
309
.
170.
Cain
DW
,
Cidlowski
JA
.
Immune regulation by glucocorticoids
.
Nat Rev Immunol
.
2017
;
17
(
4
):
233
47
.
171.
Rocamora-Reverte
L
,
Villunger
A
,
Wiegers
GJ
.
Cell-specific immune regulation by glucocorticoids in murine models of infection and inflammation
.
Cells
.
2022
;
11
(
14
):
2126
.
172.
Ihim
SA
,
Abubakar
SD
,
Zian
Z
,
Sasaki
T
,
Saffarioun
M
,
Maleknia
S
, et al
.
Interleukin-18 cytokine in immunity, inflammation, and autoimmunity: biological role in induction, regulation, and treatment
.
Front Immunol
.
2022
;
13
:
919973
.
173.
Kaplanski
G
.
Interleukin-18: biological properties and role in disease pathogenesis
.
Immunol Rev
.
2018
;
281
(
1
):
138
53
.
174.
Conti
B
,
Sugama
S
,
Kim
Y
,
Tinti
C
,
Kim
H
,
Baker
H
, et al
.
Modulation of IL-18 production in the adrenal cortex following acute ACTH or chronic corticosterone treatment
.
Neuroimmunomodulation
.
2000
;
8
:
1
7
.
175.
Conti
B
,
Jahng
JW
,
Tinti
C
,
Son
JH
,
Joh
TH
.
Induction of interferon-γ inducing factor in the adrenal cortex
.
J Biol Chem
.
1997
;
272
(
4
):
2035
7
.
176.
Sugama
S
,
Wang
N
,
Shimokawa
N
,
Koibuchi
N
,
Fujita
M
,
Hashimoto
M
, et al
.
The adrenal gland is a source of stress-induced circulating IL-18
.
J Neuroimmunol
.
2006
;
172
(
1–2
):
59
65
.
177.
Ghayur
T
,
Banerjee
S
,
Hugunin
M
,
Butler
D
,
Herzog
L
,
Carter
A
, et al
.
Caspase-1 processes IFN-gamma-inducing factor and regulates LPS-induced IFN-gamma production
.
Nature
.
1997
;
386
(
6625
):
619
23
.
178.
Akita
K
,
Ohtsuki
T
,
Nukada
Y
,
Tanimoto
T
,
Namba
M
,
Okura
T
, et al
.
Involvement of caspase-1 and caspase-3 in the production and processing of mature human interleukin 18 in monocytic THP.1 cells
.
J Biol Chem
.
1997
;
272
(
42
):
26595
603
.
179.
Matsumoto
S
,
Tsuji-Takayama
K
,
Aizawa
Y
,
Koide
K
,
Takeuchi
M
,
Ohta
T
, et al
.
Interleukin-18 activates NF-κB in murine T helper type 1 cells
.
Biochem Biophys Res Commun
.
1997
;
234
(
2
):
454
7
.
180.
Sekiyama
A
,
Ueda
H
,
Kashiwamura
S
,
Sekiyama
R
,
Takeda
M
,
Rokutan
K
, et al
.
A stress-induced, superoxide-mediated caspase-1 activation pathway causes plasma IL-18 upregulation
.
Immunity
.
2005
;
22
(
6
):
669
77
.
181.
Al-Hakeim
HK
,
Al-Rammahi
DA
,
Al-Dujaili
AH
.
IL-6, IL-18, sIL-2R, and TNFα proinflammatory markers in depression and schizophrenia patients who are free of overt inflammation
.
J Affect Disord
.
2015
;
182
:
106
14
.
182.
Prossin
AR
,
Koch
AE
,
Campbell
PL
,
McInnis
MG
,
Zalcman
SS
,
Zubieta
J-K
.
Association of plasma interleukin-18 levels with emotion regulation and μ-opioid neurotransmitter function in major depression and healthy volunteers
.
Biol Psychiatry
.
2011
;
69
(
8
):
808
12
.
183.
Quan
N
,
Banks
WA
.
Brain-immune communication pathways
.
Brain Behav Immun
.
2007
;
21
(
6
):
727
35
.
184.
Tursich
M
,
Neufeld
RWJ
,
Frewen
PA
,
Harricharan
S
,
Kibler
JL
,
Rhind
SG
, et al
.
Association of trauma exposure with proinflammatory activity: a transdiagnostic meta-analysis
.
Transl Psychiatry
.
2014
;
4
(
7
):
e413
.
185.
Zou
W
,
Feng
R
,
Yang
Y
.
Changes in the serum levels of inflammatory cytokines in antidepressant drug-naïve patients with major depression
.
PLoS One
.
2018
;
13
(
6
):
e0197267
.
186.
Kessler
RC
,
McLaughlin
KA
,
Green
JG
,
Gruber
MJ
,
Sampson
NA
,
Zaslavsky
AM
, et al
.
Childhood adversities and adult psychopathology in the WHO world mental health surveys
.
Br J Psychiatry
.
2010
;
197
(
5
):
378
85
.
187.
Miller
AH
,
Raison
CL
.
The role of inflammation in depression: from evolutionary imperative to modern treatment target
.
Nat Rev Immunol
.
2016
;
16
(
1
):
22
34
.
188.
Miller
AH
,
Maletic
V
,
Raison
CL
.
Inflammation and its discontents: the role of cytokines in the pathophysiology of major depression
.
Biol Psychiatry
.
2009
;
65
(
9
):
732
41
.
189.
Hori
H
,
Kim
Y
.
Inflammation and post-traumatic stress disorder
.
Psychiatry Clin Neurosci
.
2019
;
73
(
4
):
143
53
.
190.
Sun
Y
,
Qu
Y
,
Zhu
J
.
The relationship between inflammation and post-traumatic stress disorder
.
Front Psychiatry
.
2021
;
12
:
707543
.
191.
Frodl
T
,
Carballedo
A
,
Hughes
MM
,
Saleh
K
,
Fagan
A
,
Skokauskas
N
, et al
.
Reduced expression of glucocorticoid-inducible genes GILZ and SGK-1: high IL-6 levels are associated with reduced hippocampal volumes in major depressive disorder
.
Transl Psychiatry
.
2012
;
2
(
3
):
e88
.
192.
Raison
CL
,
Rutherford
RE
,
Woolwine
BJ
,
Shuo
C
,
Schettler
P
,
Drake
DF
, et al
.
A randomized controlled trial of the tumor necrosis factor antagonist infliximab for treatment-resistant depression: the role of baseline inflammatory biomarkers
.
JAMA Psychiatry
.
2013
;
70
(
1
):
31
41
.
193.
Majd
M
,
Hashemian
F
,
Hosseini
SM
,
Vahdat Shariatpanahi
M
,
Sharifi
A
.
A randomized, double-blind, placebo-controlled trial of celecoxib augmentation of sertraline in treatment of drug-naive depressed women: a pilot study
.
Iran J Pharm Res
.
2015
;
14
(
3
):
891
9
.
194.
Lynall
M-E
,
Turner
L
,
Bhatti
J
,
Cavanagh
J
,
de Boer
P
,
Mondelli
V
, et al
.
Peripheral blood cell: stratified subgroups of inflamed depression
.
Biol Psychiatry
.
2020
;
88
(
2
):
185
96
.
195.
Otte
C
,
Gold
SM
,
Penninx
BW
,
Pariante
CM
,
Etkin
A
,
Fava
M
, et al
.
Major depressive disorder
.
Nat Rev Dis Primers
.
2016
;
2
:
16065
20
.
196.
Barrientos
RM
,
Sprunger
DB
,
Campeau
S
,
Higgins
EA
,
Watkins
LR
,
Rudy
JW
, et al
.
Brain-derived neurotrophic factor mRNA downregulation produced by social isolation is blocked by intrahippocampal interleukin-1 receptor antagonist
.
Neuroscience
.
2003
;
121
(
4
):
847
53
.
197.
Koo
JW
,
Duman
RS
.
IL-1beta is an essential mediator of the antineurogenic and anhedonic effects of stress
.
Proc Natl Acad Sci U S A
.
2008
;
105
(
2
):
751
6
.
198.
Dantzer
R
.
Evolutionary aspects of infections: inflammation and sickness behaviors
. In:
Savitz
J
,
Yolken
RH
, editors.
Microorganisms and mental Health
.
Cham
:
Springer International Publishing
;
2023
. p.
1
14
.
199.
Dantzer
R
,
O’Connor
JC
,
Freund
GG
,
Johnson
RW
,
Kelley
KW
.
From inflammation to sickness and depression: when the immune system subjugates the brain
.
Nat Rev Neurosci
.
2008
;
9
(
1
):
46
56
.
200.
Hart
BL
.
Biological basis of the behavior of sick animals
.
Neurosci Biobehav Rev
.
1988
;
12
(
2
):
123
37
.
201.
Leonard
BE
.
The concept of depression as a dysfunction of the immune system
.
Curr Immunol Rev
.
2010
;
6
(
3
):
205
12
.
202.
Nadeau
S
,
Rivest
S
.
Glucocorticoids play a fundamental role in protecting the brain during innate immune response
.
J Neurosci
.
2003
;
23
(
13
):
5536
44
.
203.
Rivest
S
.
Molecular insights on the cerebral innate immune system
.
Brain Behav Immun
.
2003
;
17
(
1
):
13
9
.
204.
Dinkel
K
,
MacPherson
A
,
Sapolsky
RM
.
Novel glucocorticoid effects on acute inflammation in the CNS
.
J Neurochem
.
2003
;
84
(
4
):
705
16
.
205.
Sapolsky
RM
,
Pulsinelli
WA
.
Glucocorticoids potentiate ischemic injury to neurons: therapeutic implications
.
Science
.
1985
;
229
(
4720
):
1397
400
.
206.
de Pablos
RM
,
Villarán
RF
,
Argüelles
S
,
Herrera
AJ
,
Venero
JL
,
Ayala
A
, et al
.
Stress increases vulnerability to inflammation in the rat prefrontal cortex
.
J Neurosci
.
2006
;
26
(
21
):
5709
19
.
207.
Sapolsky
RM
.
A mechanism for glucocorticoid toxicity in the hippocampus: increased neuronal vulnerability to metabolic insults
.
J Neurosci
.
1985
;
5
:
1228
32
.
208.
Frank
MG
,
Miguel
ZD
,
Watkins
LR
,
Maier
SF
.
Prior exposure to glucocorticoids sensitizes the neuroinflammatory and peripheral inflammatory responses to E. coli lipopolysaccharide
.
Brain Behav Immun
.
2010
;
24
(
1
):
19
30
.
209.
Borst
K
,
Dumas
AA
,
Prinz
M
.
Microglia: immune and non-immune functions
.
Immunity
.
2021
;
54
(
10
):
2194
208
.
210.
Hammond
TR
,
Dufort
C
,
Dissing-Olesen
L
,
Giera
S
,
Young
A
,
Wysoker
A
, et al
.
Single-cell RNA sequencing of microglia throughout the mouse lifespan and in the injured brain reveals complex cell-state changes
.
Immunity
.
2019
;
50
(
1
):
253
71.e6
.
211.
Paolicelli
RC
,
Sierra
A
,
Stevens
B
,
Tremblay
M-E
,
Aguzzi
A
,
Ajami
B
, et al
.
Microglia states and nomenclature: a field at its crossroads
.
Neuron
.
2022
;
110
(
21
):
3458
83
.
212.
Vidal-Itriago
A
,
Radford
RAW
,
Aramideh
JA
,
Maurel
C
,
Scherer
NM
,
Don
EK
, et al
.
Microglia morphophysiological diversity and its implications for the CNS
.
Front Immunol
.
2022
;
13
:
997786
.
213.
Sugama
S
,
Fujita
M
,
Hashimoto
M
,
Conti
B
.
Stress induced morphological microglial activation in the rodent brain: involvement of interleukin-18
.
Neuroscience
.
2007
;
146
(
3
):
1388
99
.
214.
Sugama
S
,
Takenouchi
T
,
Fujita
M
,
Kitani
H
,
Conti
B
,
Hashimoto
M
.
Corticosteroids limit microglial activation occurring during acute stress
.
Neuroscience
.
2013
;
232
:
13
20
.
215.
Carrillo-de Sauvage
,
Maatouk
L
,
Arnoux
I
,
Pasco
M
,
Sanz Diez
A
,
Delahaye
M
, et al
.
Potent and multiple regulatory actions of microglial glucocorticoid receptors during CNS inflammation
.
Cell Death Differ
.
2013
;
20
(
11
):
1546
57
.
216.
Badimon
A
,
Strasburger
HJ
,
Ayata
P
,
Chen
X
,
Nair
A
,
Ikegami
A
, et al
.
Negative feedback control of neuronal activity by microglia
.
Nature
.
2020
;
586
(
7829
):
417
23
.
217.
Mercan
D
,
Heneka
MT
.
Norepinephrine as a modulator of microglial dynamics
.
Nat Neurosci
.
2019
;
22
(
11
):
1745
6
.
218.
McEwen
BS
,
Nasca
C
,
Gray
JD
.
Stress effects on neuronal structure: Hippocampus, amygdala, and prefrontal cortex
.
Neuropsychopharmacology
.
2016
;
41
(
1
):
3
23
.
219.
Bouras
NN
,
Mack
NR
,
Gao
W-J
.
Prefrontal modulation of anxiety through a lens of noradrenergic signaling
.
Front Syst Neurosci
.
2023
;
17
:
1173326
.
220.
Blecharz-Lang
KG
,
Patsouris
V
,
Nieminen-Kelhä
M
,
Seiffert
S
,
Schneider
UC
,
Vajkoczy
P
.
Minocycline attenuates microglia/macrophage phagocytic activity and inhibits SAH-induced neuronal cell death and inflammation
.
Neurocrit Care
.
2022
;
37
(
2
):
410
23
.
221.
Yrjänheikki
J
,
Keinänen
R
,
Pellikka
M
,
Hökfelt
T
,
Koistinaho
J
.
Tetracyclines inhibit microglial activation and are neuroprotective in global brain ischemia
.
Proc Natl Acad Sci U S A
.
1998
;
95
(
26
):
15769
74
.
222.
Blandino
P
,
Barnum
CJ
,
Deak
T
.
The involvement of norepinephrine and microglia in hypothalamic and splenic IL-1beta responses to stress
.
J Neuroimmunol
.
2006
;
173
(
1–2
):
87
95
.
223.
Nair
A
,
Bonneau
RH
.
Stress-induced elevation of glucocorticoids increases microglia proliferation through NMDA receptor activation
.
J Neuroimmunol
.
2006
;
171
(
1–2
):
72
85
.
224.
Hinwood
M
,
Morandini
J
,
Day
TA
,
Walker
FR
.
Evidence that microglia mediate the neurobiological effects of chronic psychological stress on the medial prefrontal cortex
.
Cereb Cortex
.
2012
;
22
(
6
):
1442
54
.
225.
Woodburn
SC
,
Bollinger
JL
,
Wohleb
ES
.
Synaptic and behavioral effects of chronic stress are linked to dynamic and sex-specific changes in microglia function and astrocyte dystrophy
.
Neurobiol Stress
.
2021
;
14
:
100312
.
226.
Woodburn
SC
,
Bollinger
JL
,
Wohleb
ES
.
The semantics of microglia activation: neuroinflammation, homeostasis, and stress
.
J Neuroinflammation
.
2021
;
18
(
1
):
258
.
227.
Szepesi
Z
,
Manouchehrian
O
,
Bachiller
S
,
Deierborg
T
.
Bidirectional microglia: neuron communication in health and disease
.
Front Cell Neurosci
.
2018
;
12
:
323
.
228.
Davalos
D
,
Grutzendler
J
,
Yang
G
,
Kim
JV
,
Zuo
Y
,
Jung
S
, et al
.
ATP mediates rapid microglial response to local brain injury in vivo
.
Nat Neurosci
.
2005
;
8
(
6
):
752
8
.
229.
Elmore
MRP
,
Najafi
AR
,
Koike
MA
,
Dagher
NN
,
Spangenberg
EE
,
Rice
RA
, et al
.
Colony-stimulating factor 1 receptor signaling is necessary for microglia viability, unmasking a microglia progenitor cell in the adult brain
.
Neuron
.
2014
;
82
(
2
):
380
97
.
230.
Easley-Neal
C
,
Foreman
O
,
Sharma
N
,
Zarrin
AA
,
Weimer
RM
.
CSF1R ligands IL-34 and CSF1 are differentially required for microglia development and maintenance in white and gray matter brain regions
.
Front Immunol
.
2019
;
10
:
2199
.
231.
Cardona
AE
,
Pioro
EP
,
Sasse
ME
,
Kostenko
V
,
Cardona
SM
,
Dijkstra
IM
, et al
.
Control of microglial neurotoxicity by the fractalkine receptor
.
Nat Neurosci
.
2006
;
9
(
7
):
917
24
.
232.
Haynes
SE
,
Hollopeter
G
,
Yang
G
,
Kurpius
D
,
Dailey
ME
,
Gan
W-B
, et al
.
The P2Y12 receptor regulates microglial activation by extracellular nucleotides
.
Nat Neurosci
.
2006
;
9
(
12
):
1512
9
.
233.
Bollinger
JL
,
Dadosky
DT
,
Flurer
JK
,
Rainer
IL
,
Woodburn
SC
,
Wohleb
ES
.
Microglial P2Y12 mediates chronic stress-induced synapse loss in the prefrontal cortex and associated behavioral consequences
.
Neuropsychopharmacol
.
2023
;
48
(
9
):
1347
57
.
234.
Heidt
T
,
Sager
HB
,
Courties
G
,
Dutta
P
,
Iwamoto
Y
,
Zaltsman
A
, et al
.
Chronic variable stress activates hematopoietic stem cells
.
Nat Med
.
2014
;
20
(
7
):
754
8
.
235.
Stefanski
V
.
Social stress in laboratory rats: hormonal responses and immune cell distribution
.
Psychoneuroendocrinology
.
2000
;
25
(
4
):
389
406
.
236.
Wohleb
ES
,
McKim
DB
,
Sheridan
JF
,
Godbout
JP
.
Monocyte trafficking to the brain with stress and inflammation: a novel axis of immune-to-brain communication that influences mood and behavior
.
Front Neurosci
.
2014
;
8
:
447
.
237.
Hu
H
,
Yang
X
,
He
Y
,
Duan
C
,
Sun
N
.
Psychological stress induces depressive-like behavior associated with bone marrow-derived monocyte infiltration into the hippocampus independent of blood-brain barrier disruption
.
J Neuroinflammation
.
2022
;
19
(
1
):
208
.
238.
Sawada
A
,
Niiyama
Y
,
Ataka
K
,
Nagaishi
K
,
Yamakage
M
,
Fujimiya
M
.
Suppression of bone marrow-derived microglia in the amygdala improves anxiety-like behavior induced by chronic partial sciatic nerve ligation in mice
.
Pain
.
2014
;
155
(
9
):
1762
72
.
239.
Cathomas
F
,
Lin
H-Y
,
Chan
KL
,
Li
L
,
Parise
LF
,
Alvarez
J
, et al
.
Circulating myeloid-derived MMP8 in stress susceptibility and depression
.
Nature
.
2024
;
626
(
8001
):
1108
15
.
240.
Lehmann
ML
,
Cooper
HA
,
Maric
D
,
Herkenham
M
.
Social defeat induces depressive-like states and microglial activation without involvement of peripheral macrophages
.
J Neuroinflammation
.
2016
;
13
(
1
):
224
.
241.
Tan
Y-L
,
Yuan
Y
,
Tian
L
.
Microglial regional heterogeneity and its role in the brain
.
Mol Psychiatry
.
2020
;
25
(
2
):
351
67
.
242.
Ayata
P
,
Badimon
A
,
Strasburger
HJ
,
Duff
MK
,
Montgomery
SE
,
Loh
Y-HE
, et al
.
Epigenetic regulation of brain region-specific microglia clearance activity
.
Nat Neurosci
.
2018
;
21
(
8
):
1049
60
.
243.
Böttcher
C
,
Schlickeiser
S
,
Sneeboer
MAM
,
Kunkel
D
,
Knop
A
,
Paza
E
, et al
.
Human microglia regional heterogeneity and phenotypes determined by multiplexed single-cell mass cytometry
.
Nat Neurosci
.
2019
;
22
(
1
):
78
90
.
244.
Grabert
K
,
Michoel
T
,
Karavolos
MH
,
Clohisey
S
,
Baillie
JK
,
Stevens
MP
, et al
.
Microglial brain region−dependent diversity and selective regional sensitivities to aging
.
Nat Neurosci
.
2016
;
19
(
3
):
504
16
.
245.
Keren-Shaul
H
,
Spinrad
A
,
Weiner
A
,
Matcovitch-Natan
O
,
Dvir-Szternfeld
R
,
Ulland
TK
, et al
.
A unique microglia type associated with restricting development of alzheimer’s disease
.
Cell
.
2017
;
169
(
7
):
1276
90.e17
.
246.
Silvin
A
,
Uderhardt
S
,
Piot
C
,
Da Mesquita
S
,
Yang
K
,
Geirsdottir
L
, et al
.
Dual ontogeny of disease-associated microglia and disease inflammatory macrophages in aging and neurodegeneration
.
Immunity
.
2022
;
55
(
8
):
1448
65.e6
.
247.
Tynan
RJ
,
Naicker
S
,
Hinwood
M
,
Nalivaiko
E
,
Buller
KM
,
Pow
DV
, et al
.
Chronic stress alters the density and morphology of microglia in a subset of stress-responsive brain regions
.
Brain Behav Immun
.
2010
;
24
(
7
):
1058
68
.
248.
Covington
HE
,
Lobo
MK
,
Maze
I
,
Vialou
V
,
Hyman
JM
,
Zaman
S
, et al
.
Antidepressant effect of optogenetic stimulation of the medial prefrontal cortex
.
J Neurosci
.
2010
;
30
(
48
):
16082
90
.
249.
Vialou
V
,
Bagot
RC
,
Cahill
ME
,
Ferguson
D
,
Robison
AJ
,
Dietz
DM
, et al
.
Prefrontal cortical circuit for depression- and anxiety-related behaviors mediated by cholecystokinin: role of ΔFosB
.
J Neurosci
.
2014
;
34
(
11
):
3878
87
.
250.
Liu
Q
,
Zhang
Z
,
Zhang
W
.
Optogenetic dissection of neural circuits underlying stress-induced mood disorders
.
Front Psychol
.
2021
;
12
:
600999
.
251.
Morrison
SJ
,
Spradling
AC
.
Stem cells and niches: mechanisms that promote stem cell maintenance throughout life
.
Cell
.
2008
;
132
(
4
):
598
611
.
252.
Morrison
SJ
,
Scadden
DT
.
The bone marrow niche for haematopoietic stem cells
.
Nature
.
2014
;
505
(
7483
):
327
34
.
253.
Cai
R
,
Pan
C
,
Ghasemigharagoz
A
,
Todorov
MI
,
Förstera
B
,
Zhao
S
, et al
.
Panoptic imaging of transparent mice reveals whole-body neuronal projections and skull: meninges connections
.
Nat Neurosci
.
2019
;
22
(
2
):
317
27
.
254.
Herisson
F
,
Frodermann
V
,
Courties
G
,
Rohde
D
,
Sun
Y
,
Vandoorne
K
, et al
.
Direct vascular channels connect skull bone marrow and the brain surface enabling myeloid cell migration
.
Nat Neurosci
.
2018
;
21
(
9
):
1209
17
.
255.
Brioschi
S
,
Wang
W-L
,
Peng
V
,
Wang
M
,
Shchukina
I
,
Greenberg
ZJ
, et al
.
Heterogeneity of meningeal B cells reveals a lymphopoietic niche at the CNS borders
.
Science
.
2021
;
373
(
6553
):
eabf9277
.
256.
Cugurra
A
,
Mamuladze
T
,
Rustenhoven
J
,
Dykstra
T
,
Beroshvili
G
,
Greenberg
ZJ
, et al
.
Skull and vertebral bone marrow are myeloid cell reservoirs for the meninges and CNS parenchyma
.
Science
.
2021
;
373
(
6553
):
eabf7844
.
257.
Shansky
RM
,
Morrison
JH
.
Stress-induced dendritic remodeling in the medial prefrontal cortex: effects of circuit, hormones and rest
.
Brain Res
.
2009
;
1293
:
108
13
.
258.
Garrett
JE
,
Wellman
CL
.
Chronic stress effects on dendritic morphology in medial prefrontal cortex: sex differences and estrogen dependence
.
Neuroscience
.
2009
;
162
(
1
):
195
207
.
259.
Bollinger
JL
.
Uncovering microglial pathways driving sex-specific neurobiological effects in stress and depression
.
Brain Behav Immun Health
.
2021
;
16
:
100320
.
260.
Bollinger
JL
,
Bergeon Burns
CM
,
Wellman
CL
.
Differential effects of stress on microglial cell activation in male and female medial prefrontal cortex
.
Brain Behav Immun
.
2016
;
52
:
88
97
.
261.
Zárate
S
,
Stevnsner
T
,
Gredilla
R
.
Role of estrogen and other sex hormones in brain aging. Neuroprotection and DNA repair
.
Front Aging Neurosci
.
2017
;
9
.
262.
Hara
Y
,
Waters
EM
,
McEwen
BS
,
Morrison
JH
.
Estrogen effects on cognitive and synaptic health over the lifecourse
.
Physiol Rev
.
2015
;
95
(
3
):
785
807
.
263.
Hoffman
GE
,
Merchenthaler
I
,
Zup
SL
.
Neuroprotection by ovarian hormones in animal models of neurological disease
.
Endocrine
.
2006
;
29
(
2
):
217
31
.
264.
Zhang
W
,
Kaldewaij
R
,
Hashemi
MM
,
Koch
SBJ
,
Smit
A
,
van Ast
VA
, et al
.
Acute-stress-induced change in salience network coupling prospectively predicts post-trauma symptom development
.
Transl Psychiatry
.
2022
;
12
:
63
8
.
265.
Mueller
JM
,
Pritschet
L
,
Santander
T
,
Taylor
CM
,
Grafton
ST
,
Jacobs
EG
, et al
.
Dynamic community detection reveals transient reorganization of functional brain networks across a female menstrual cycle
.
Netw Neurosci
.
2021
;
5
(
1
):
125
44
.
266.
Andreano
JM
,
Touroutoglou
A
,
Dickerson
B
,
Barrett
LF
.
Hormonal cycles, brain network connectivity, and windows of vulnerability to affective disorder
.
Trends Neurosci
.
2018
;
41
(
10
):
660
76
.
267.
Hidalgo-Lopez
E
,
Zeidman
P
,
Harris
T
,
Razi
A
,
Pletzer
B
.
Spectral dynamic causal modelling in healthy women reveals brain connectivity changes along the menstrual cycle
.
Commun Biol
.
2021
;
4
(
1
):
954
.
268.
Petersen
N
.
Spotlighting SHAPERS: sex hormones associated with psychological and endocrine roles
.
Neuropsychopharmacology
.
2024
;
49
(
5
):
772
3
.