Next Article in Journal
The Role of the RNA Helicase DDX3X in Medulloblastoma Progression
Previous Article in Journal
Biomarkers in Cerebrospinal Fluid for the Diagnosis and Monitoring of Gliomas
Previous Article in Special Issue
Increasing CB2 Receptor Activity after Early Life Stress Prevents Depressive Behavior in Female Rats
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Early Life Adversity, Microbiome, and Inflammatory Responses

by
Eléonore Beurel
1,2 and
Charles B. Nemeroff
3,4,*
1
Department of Psychiatry and Behavioral Sciences, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
2
Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
3
Department of Psychiatry and Behavioral Sciences, Mulva Clinic for Neurosciences, University of Texas (UT) Dell Medical School, Austin, TX 78712, USA
4
Mulva Clinic for Neurosciences, UT Austin Dell Medical School, Austin, TX 78712, USA
*
Author to whom correspondence should be addressed.
Biomolecules 2024, 14(7), 802; https://doi.org/10.3390/biom14070802
Submission received: 29 April 2024 / Revised: 28 June 2024 / Accepted: 1 July 2024 / Published: 6 July 2024

Abstract

:
Early life adversity has a profound impact on physical and mental health. Because the central nervous and immune systems are not fully mature at birth and continue to mature during the postnatal period, a bidirectional interaction between the central nervous system and the immune system has been hypothesized, with traumatic stressors during childhood being pivotal in priming individuals for later adult psychopathology. Similarly, the microbiome, which regulates both neurodevelopment and immune function, also matures during childhood, rendering this interaction between the brain and the immune system even more complex. In this review, we provide evidence for the role of the immune response and the microbiome in the deleterious effects of early life adversity, both in humans and rodent models.

1. Early Life Adversity

Early life adversity affects 1 out of 10 children (under 18) to the point that these children will experience long-term physical and mental health consequences [1,2], including an increased risk for psychiatric disorders [3], and many other medical disorders including asthma, diabetes, stroke and cardiovascular disease [4]; and an overall marked increased risk of mortality by age 50 [5,6]. The impact of early life adversity likely results from multifactorial mechanisms. The prevailing theory of how early life adversity increases risk for physical and mental illnesses revolves around the body’s response to stress and the idea that there are possible sensitive periods when early life adversity impacts neurodevelopment the most. Physiological changes after acute stress include the release of cortisol and norepinephrine [7], which prepare the body to maximize the response to the threat while minimizing harm and mobilizing the immune system. Early life adversity includes childhood physical or sexual abuse, neglect, and other negative life events (e.g., poverty, loss of a parent, parental psychopathology, bullying, natural disaster, etc.) [8,9]. The high frequency or magnitude of exposure to the stressors during early life adversity dictates the degree and chronicity of subsequent inflammation [10]. Persistent inflammation has a multitude of detrimental effects on the body over time [11,12,13,14,15]. In summary, although the pathophysiological mechanisms responsible for the many detrimental effects of early life adversity on health remain poorly understood, many studies have posited the immune system to be a major mediator. Thus, the release of norepinephrine under threat conditions, mobilizes the immune system to produce cytokines in the damaged tissues to help clear the pathogens and promote wound healing [16]. Simultaneously, the adrenal glands increase the production of cortisol and epinephrine, which initially reinforce the proinflammatory actions of norepinephrine, but after the threat wanes, in contrast, they counteract the effects of norepinephrine [17,18]. From an evolutionary point of view, stress is thought to prepare the body to clear pathogens and enhance tissue healing. Although stress and inflammation are considered culprits during early life adversity, their exact role in critical periods of neurodevelopment remains to be fully elucidated and will not be discussed in this review.

2. Immune System

The main function of the immune system is to protect the organism against pathogens (e.g., microbes, toxins, etc.). There are two major immune arms: the innate immune system, which is already active at birth, but weaker than later in life, and the adaptive immune system, which develops as the child is exposed to antigens.

2.1. Innate Immune System

The innate immune system comprises neutrophils, monocytes/macrophages, dendritic cells, which react to pathogen invasion by phagocytosing them. Children secrete lower levels of cytokines responsible for the priming of T helper (Th) 1 and CD8 T cell responses, known to eliminate pathogens, compared to adults, whereas their production of cytokines required for Th17 responses, which are required for some bacterial and fungal infection clearance is similar to adults’ function (for review [19]). This suggests an increased susceptibility of children to certain pathogens, which will be discussed later in the review. In summary, the innate immune system is muted at birth to allow all the remodeling during development and to tolerate maternal antigens, and this puts the child at higher risks for bacterial and viral infections and vulnerability to stressors associated with early life adversity.

2.2. Adaptive Immune System

If the innate immune system is not able to clear the pathogen, it recruits by producing cytokines and by presenting antigens to the adaptive immune system, whose function is to help the innate immune system clear the pathogen. The adaptive immune system comprises T and B cells. With the help of T cells, B cells produce antibodies to clear pathogens. The first encounter with bacteria is during the passage through the birth canal and then the contact of the skin and lungs with the exterior world. During the child’s life, the adaptive immune system changes and forms immunological memory to protect the child against a new encounter of the same pathogen [20], emphasizing the importance of immunizations [21] to help train the adaptive immune system of children. It is remarkable that a mother can transfer to the fetus immunoglobulins from infections she encountered 20–30 years ago, helping the child cope with infections. There are also many asymptomatic viral infections that induce a strong CD4 and CD8 T cell immune responses, such as cytomegalovirus (CMV) or Epstein–Barr virus (EBV), and any immunosuppressive therapy can reactivate viruses and have dramatic negative consequences for the child. Furthermore, naïve B cells in response to a specific antigen undergo somatic hypermutation, class switching and proliferation, creating B cells producing higher affinity-binding immunoglobulins, a process that takes months and that leads to a better immunity over time [22]. Furthermore, maturation of the T and B cells is critical for immunological memory. As an individual gets older, the repertoire of immune cells experiences maturation. This adds to the already high genetic variability between individuals and explains the considerable variation in the immune response between individuals. For example, Th17, regulatory T cells and NK cells are altered in patients with high liability for psychosis and childhood trauma. Thus, Th17 cell number increases in the high liability group with childhood trauma, regulatory T cells increases, and NK cells decreases in the high liability group and predicted stress experience [23]. Th17 cells previously show a trend to increase with early life adversity [24]. This is consistent with the findings that Th17 cell differentiation is already operational in children, similar to adults [19], that Th17 cells are associated with asthma [25,26], which is linked to early life adversity [27,28] and that Th17 cells promote susceptibility to depression after stress and affect neuronal circuits in autism [29].
Altogether, these findings suggest that any perturbations of the immune system maturation can have consequences for the child health and may explain the increased susceptibility of certain children to mental health and physical health disorders.

3. Effects of Early Life Adversity on the Immune System in Humans

Childhood adversity primes immune cells (e.g., monocytes, macrophages) that initiate and sustain inflammation. Thus, maltreated or disadvantaged children are often more exposed to air pollution, high-fat, high-sugar diets or stressors, such as family instability, poverty, and violence [30,31], and these exposures prime monocytes to produce higher levels of cytokines when restimulated ex vivo with microbial products [32,33] and render monocytes less sensitive to inhibition by glucocorticoids [34]. Insensitivity to glucocorticoids has been postulated to be adaptive during acute threats, while in contrast, if sustained, insensitivity to glucocorticoids would promote chronic inflammation involved in emotional and physical health issues [15,35].

3.1. Inflammation

3.1.1. Early Life Stress Is Associated with Inflammation

Longitudinal studies in humans have revealed a correlation between early life stress, exposure to trauma and the presence of inflammatory markers in adulthood, such as the circulating acute phase protein, C-reactive protein (CRP), proinflammatory cytokines (interleukin-6, IL-6; IL-1β; tumor necrosis factor, TNF) [8,36,37,38], and the alteration of the leukocyte count [39]. For example, in the Dunedin Multidisciplinary Health and Development Study [40], during which subjects have been followed from birth to adult life, it was found that cumulative exposure to early life maltreatment was associated with greater inflammatory marker levels later in adulthood when controlling for all possible confounding factors [41]. Other studies have confirmed these findings [8,9]. Similar findings were found whether the maltreatment was perpetrated by adults or peers (e.g., bullying). It has been estimated that ~10% of low-grade inflammation (defined as an elevation of 2–3 fold of interleukin-6, tumor necrosis factor and C-reactive protein [8]) in adults may be due to childhood maltreatment [41,42]. Furthermore, early life stress has profound health consequences, because childhood maltreatment is also associated with impaired acquired immunity [43], an enhanced inflammatory response to stressors [11,14,44,45,46] and an increased risk of physical and mental health disorders [47].
The inflammation seems to be triggered by exposure to early life stress, as the increased inflammation was detectable in maltreated children [38], as evidenced by elevated CRP levels in maltreated children. In addition, increased inflammation is associated with the onset of depression [48], reinforcing the importance of inflammation in priming depressive symptoms. Consistent with this, increased inflammation was found in adolescents exposed to adverse childhood experiences [49] and in children with a Child Protective Service-documented history of maltreatment [50].

3.1.2. Comorbidities Render the Association between Early Life Stress and Inflammation More Complex

Comorbid factors such as obesity also render the association between childhood maltreatment and inflammation complex because obesity increases inflammatory markers [51], and an increased risk of obesity is associated with childhood maltreatment [13], raising questions about whether inflammation or obesity emerges first [8,9]. Additional life stresses in adulthood likely also contribute to the alteration of immune measures.

3.1.3. Stress Affects Inflammation

Increased stress in childhood is also associated with increased hypothalamic–pituitary–adrenal (HPA) axis activity, leading to increased production of cortisol [52,53], and cortisol exerts acute anti-inflammatory property [54,55]. Therefore, the concentration of cortisol might dictate the outcome of the inflammatory response. Consistent with this notion, children with supportive caregivers exhibited attenuated HPA axis activity [56]. In adolescents, the HPA axis seems altered [57,58], and the impact of cortisol on the immune response seems to be lessened [59]. This might explain why cytokines are detected more systematically in older children. There is also evidence of profound hyperactivity of corticotropin-releasing factor (CRF)-containing circuits in the brain after early life adversity, and this system is also known to regulate immune function.

3.1.4. Other Factors Also Contribute to Early Life Stress Outcomes on Inflammation

Other behaviors in adolescence might also explain the change in cytokine production, such as changes in diet, absence of exercise, substance and alcohol abuse, and stress associated with risky behaviors [60,61,62,63]. Overall, there is substantial variability in the measured cytokines depending on the type of stress, the age, and the method of assessment of inflammation. No clear picture has emerged about the functionality of the immune system in early life adversity, but growing evidence suggests an integrated, bidirectional interaction between the cortico-amygdala circuit and inflammation to detect threats to well-being and to promote coping through the propagation of inflammation by the monocytes/macrophages [16,64,65]. In addition, the cortico-basal ganglia circuit and reward-related behaviors are also affected by early life adversity. Thus, maltreatment correlated with later reward-processing deficits on behavioral tasks [66,67], and inflammation blunted reward sensitivity, referred to as sickness behaviors, which belong to a set of adaptations to infection mediated by cytokines [68,69]. Consistent with this, inflammation decreased animals’ sensitivity to reward [70]. In addition, prefrontal executive control deficits contribute to neuroimmune responses. The cerebral prefrontal cortex is particularly vulnerable to early life adversity, and reductions in the gray matter volume, cortical thickness, and white matter tract integrity of the cerebral prefrontal cortex were reported in individuals with early life adversity [60,71,72,73,74,75]. Inflammation also interacts with the prefrontal cortex, with reports of smaller prefrontal cortex volume and white matter integrity associated with increased inflammatory markers [76,77]. In summary, the presence of low-grade chronic inflammation in early life adversity seems to expose children to a plethora of health issues across the lifespan [15,78,79,80]. Focusing on mental health issues, it has been proposed that defects in the cortico-amygdala circuit and inflammation manifest in childhood, while reward and executive functions are affected later in life during adolescence and adulthood [81].

3.2. Immune Cell Defect Is Associated with Early Life Stress

Both innate and adaptive immune cell functions are affected by early life stress.

3.2.1. Innate Immunity Cell Defects

At the cellular level, a 20% reduction in granulocyte function (assessed by killing ex vivo Staphylococcus aureus) has been reported in children of separated or divorced parents [82]. Studies have reported low natural killer (NK) cell activity in adolescents with childhood trauma [83] and in women with breast cancer who had experienced early life adversity [84], increased NK activity in children whose parents reported chronic stress [85], and no change in NK activity in sexually abused adolescents [86]. Therefore, although innate immunity through cytokine production by monocytes seems enhanced, granulocyte and NK functions seem to decrease after early life adversity.

3.2.2. Adaptive Immunity Cell Defects

In term of adaptive immunity, IgE and IL-4 productions were increased in predisposed children to asthma, with parents experiencing chronic stress [27,87,88] suggesting hyperreactive peripheral blood mononuclear cells (PBMCs). It has been reported that early life adversity has a profound impact on the thymus, leading to involution or atrophy of the thymus or the spleen, including spleen atrophy [89], providing a potential basis for a T cell defect because T cells are maturing in the thymus and are housed in the spleen and lymph nodes. Furthermore, research on adolescents and young adults demonstrated that early life adversity events during infancy were associated with changes in specific T cell populations, including T cell immunosenescence [37,90,91], and there was an increase in CD8 T cells relative to CD4 T cells [24,91,92,93]. CMV seropositivity was also associated with the long-term shift in T cells [91,92], showing signs of senescence. All together, these findings point to a reduction in the function of T cells in adolescents with childhood trauma, confirming the significant impact of childhood trauma on immune function.

3.3. Early Life Adversity and Increased Susceptibility to Infection

It has been reported that children of parents with psychiatric symptoms are more prone to illnesses [85,94]. Similarly, children who perceive their family as dysfunctional are more susceptible to influenza B infection when compared to children who do not have this perception [95]. Children more sensitive to stress also exhibit higher rates of respiratory infections [96,97], and high parental stress predicts infant wheezing even after controlling for allergen exposure, parental smoking, and susceptibility to respiratory infections [98]. These findings suggest a potential weakening of the immune response in children with high levels of stress.
Lower socioeconomic status has also been associated with an increased risk of infectious disease in adulthood [99,100,101], but this association was absent when the relationship with one parent was supportive [102], reinforcing the importance of a supportive child–parent relationship.
The response to vaccination seems to also be attenuated in children with early life adversity [103,104], consistent with a weakening of the body’s response to vaccines by stress [105]. Although the consequences of a weakened immune response are not clear, it likely contributes to the reactivation of dormant viruses such as the Herpes simplex virus [43,106], Epstein–Barr virus [107,108] and cytomegalovirus [37,94] leading to higher virus-specific antibody titers in children or adolescents with early life adversity. Similarly, sensitization to allergens has been associated with early life adversity [109,110].
In summary, the association between inflammation and early life adversity is complex. Even though there is an increase in proinflammatory cytokines in individuals with early life adversity, it appears that the immune response is less effective than in individuals without early life adversity. This might explain findings of an increased risk of developing depression, cancer, cardiovascular diseases, or other serious illnesses in adulthood of individuals with early life adversity.

3.4. Early Life Adversity Affects the Microbiome

The gut microbiome represents a unique community of trillions of microorganisms cohabiting in the gut, including bacteria, fungi and viruses implicated in both mental and physical health. Each individual microbiome is unique and is influenced by the diet, behaviors, and environment. The gut microbiome is critical during the first years of life and may have long-term consequences for health due to its role in shaping the immune system [111]. Prenatal stress impacts the microbiome [112,113,114,115]. Thus, infants with high stress levels [115] or newborns from stressed pregnant mothers [114] exhibited an increased abundance of Proteobacteria spp. and a decreased abundance of Bifidobacterium spp. Infants with high level of cortisol 3 days postpartum also exhibited increased levels of the Proteobacteria genus Enterobacteriaceae [114], though other studies failed to reproduce these findings [116,117]. Changes in the microbiome of infants who experienced maternal prenatal stress have been suggested to shape changes in neurodevelopment [118], which could predispose individuals to psychopathologies.
Changes in the gut microbiome have also been associated with early life adversity and changes in blood T cell subsets in youth [119]. Beta diversity (e.g., differences between the different bacterial communities) and bacterial counts were reported to be reduced in youth exposed to early life adversity [120]. Bacterial changes were also associated with other early life adversity outcomes, such as the activation of the medial prefrontal cortex in emotional faces. This suggests that early life adversity, caused by remodeling the gut microbiome, affects both the immune system and emotional reactivity. Prevotella, which is associated with diet [121,122,123] or HIV infection [124], Bacteroides and Coprococcus levels are augmented in institutionalized youth [119,120]. Similarly, the oral microbiome was affected in individuals with early life adversity, and changes in taxa (e.g., hierarchical subdivisions of the bacteria species) abundance could be detected 24 years after the early life adversity event. Changes in the taxa abundance were associated with early institutionalization and CMV seropositivity [125]. There was also a link between T cell senescence, NK activation status, and changes in the oral microbiome. Although in humans it is difficult to attribute the changes of microbiome and immune function to early life adversity, it points to an interesting association between institutionalization time in the postnatal period, microbiome, and immune changes in youth.

3.5. Effects of Early Life Adversity on Pregnancy Outcomes

Early life adversity is associated with a 2-fold increased risk of preterm delivery compared to mothers without early life adversity [126]. This elevated risk for preterm labor is in part attributed to risky behaviors such as smoking, alcohol or substance abuse and mental health conditions such as anxiety and depression [6,126,127,128,129]. In addition, the gestation age, and the weight at delivery of the offspring of mothers with early life adversity are lower [130]. These seem associated with increased maternal cortisol [131] and increased basal levels of IL-6 [132] and are consistent with an adverse effect of inflammation on pregnancy and the developing fetus. Furthermore, inflammation, which is associated with adverse childhood experiences, increases the risk of developing depression during pregnancy [133] and other pregnancy complications, such as gestational diabetes and hypertensive disorders [134].
Parenting may also be difficult for individuals with early life adversity [135], although not all parents with early life adversity mistreat their children. Several studies showed a link between allostatic load (e.g., exposure to stress for a long period of time) and poor parenting [136,137], which is consistent with the negative impact of adverse childhood experiences on cognition and social development [136,137].
Altogether, these findings point to a potential intergenerational transmission of stress and adverse health outcomes. Although intergenerational transmission is difficult to establish methodologically, some researchers have reported evidence for intergenerational transmission of neglect and sexual abuse but not physical abuse [138]. It has been proposed that central and peripheral systems, including neural circuits, the HPA axis, and the microbiome, are possible pathways mediating the effects of early life adversity across generations [139,140,141]. The gut microbiome of pregnant mothers shapes the development of the fetal brain and immune system [142,143]. Recently, a large longitudinal study showed that maternal prenatal anxiety or maternal childhood maltreatment and second-generation exposure to stressful life events are sufficient to impact the gut microbiome composition of the second-generation children at 2 years of age, as well as impacting behaviors (e.g., socioemotional functioning) at 2 and 4 years of age [141]. It also seems that the timing of exposure to early life adversity (prenatal vs. postnatal) has a different impact on the gut microbiome composition, but both are associated with increased inflammation-associated taxa [141]. These findings confirm the notion that early life adversity, by affecting the microbiome could consequently affect the immune system, which in turn could interfere with neuronal circuits, and prime individuals to psychopathologies (Figure 1), although this hypothesis will need to be further tested.

4. Modeling Childhood Trauma in Animals and Effects of the Immune System

4.1. Proinflammatory Markers

Initial evidence of the impact of childhood experiences on the immune system was reported more than a half century ago in rodents, showing that rats handled before weaning, which promoted bonding with the mother, exhibited better health outcomes [144,145]. Similar behavioral outcomes were reported in non-human primates subjected to early life stress [146,147,148]. Since then, other models have been used to induce early life stress in rodents, such as maternal separation, maternal deprivation, early weaning, or nursery rearing, and those have generated mixed results in terms of inflammatory consequences [149,150,151]. A clearer picture emerges when focusing on maternal separation studies. Indeed, maternal separation induces macrophage activity [152], monocyte-dependent proinflammatory gene upregulation [153], and increases plasma proinflammatory cytokine concentration [154], which is also associated with increased body temperature [155]. There is a lack of evidence for a change in anti-inflammatory cytokines such as IL-10 after maternal separation [156]. Female mice are more prone to increase cytokines after a stressful event than males [157,158]. In the pup brain, maternal separation is associated with a reduction in proinflammatory mediators in the hippocampus, such as lipopolysaccharide-binding protein [159] and of microglia in the midbrain [160]. In contrast, an increased level of the interleukin 1 receptor on synapses [161], an increased number of microglia [162], and an increased activated state [163] are observed in the adult brains of rodents subjected to maternal separation, suggesting possible time-sensitive windows for the effects of inflammation on neuronal development. Furthermore, maternal separation primes rodents for even more robust responses when encountering subsequent stress or immune challenges. Thus, rodents exposed to maternal separation exhibit a greater increase in core temperature after re-exposure to maternal stress [155], greater cytokine production to a viral infection [164], and greater microglia activation after stress [163].

4.2. Sex Differences

In addition, an increasing body of evidence suggests that male and female rodents are not equally emotionally affected by early life adversity [165,166,167,168,169]. Early life adversity induces social deficits in male offspring but increases anxiety in female offspring [167,170,171,172,173,174,175,176], which seems to correspond to differential microbial and immune changes between males and females. Associated with these differential outcomes in behaviors, males exhibit predominantly a down regulation of overall gene expression in the medial prefrontal cortex, while overall gene expression is upregulated in females [177], suggesting that the prefrontal cortex executive circuit is not affected similarly in males and females after early life adversity.

4.3. Microbiome

Because the stress response is shaped by the microbiome [178,179,180,181] and vice versa, the microbiome is shaped by the stress response [118,182,183,184,185,186], prenatal stress in animals affects the microbiome [187,188] similarly to the findings in humans. In rats, an increased Prevotella level is associated with early life adversity [189]. In male mice, Lachnospiraceae was reported to be highly sensitive to multiple early life stresses, while in female mice, only Lactobacillus and Mucispirillum are affected [177]. Maternal separation increases Bacteroides, Lachnospiraceae and Clostridium XIVa spp. in both mice and rats [184,190,191,192]. Associated with a change in the microbiota composition, there is also the presence of gut dysbiosis and associated gut leakiness in animals exposed to early life stress [186,189,191,193,194,195]. Because the gut microbiome is critical during brain development, it has also been proposed that changes in the gut microbiome could affect brain development during early life adversity [196]. Early life stress affects the medial prefrontal cortex, a brain area involved in the regulation of emotional behavior [197,198]. Expression of immediate early genes such as Arc, Egr4, and Fosb in the medial prefrontal cortex is also regulated by the gut microbiota [190,199,200], suggesting a convergence of effects of early life adversity and the microbiota on the medial prefrontal cortex.
Altogether, in animals, early life stress promotes inflammation and alteration of the microbiome in a sex-dependent manner, leading to long-lasting behavioral changes.

5. Conclusions

Overall, early life adversity has a negative impact on health, priming individuals for psychopathologies and other medical disorders. It is now unequivocally recognized as one of the preeminent factors in adult morbidity and mortality. The precise cause of the increased vulnerability remains unclear, although the inflammation has been proposed to be one of the major culprits. In addition, patients with mood disorders, including depression and bipolar disorder, who have childhood maltreatment histories have a more pernicious disease course and are relatively treatment resistant, in part due to increases in inflammatory status. Moreover, children with early life adversity are more susceptible to infections and exhibit immune responses that are disrupted throughout the individual’s life. Although the precise causes of the immune dysregulation remain obscure, some have proposed that infection with CMV could induce immunosenescence, rendering the immune system less efficient at clearing insults. These perturbations in the immune system might also be amplified by a less diverse microbiome due to poorer nutrition or hygiene and a lower socioeconomic environment. Gut dysbiosis and leakiness lead to increased neuroinflammation, which has detrimental effects on various neuronal circuits, such as the cortico-amygdala circuit, cortico-basal ganglia circuit and prefrontal executive control circuit, and may lead to behavioral changes associated with the development of psychopathologies. However, evidence is still lacking to show a causal effect of early life adversity on the immune system and microbiome in humans, warranting additional research on the topic.

Funding

Research in the laboratories of Drs. Beurel and Nemeroff are funded by NIH (MH104656, AA029090 respectively).

Conflicts of Interest

Charles Nemeroff has patent #US 6,375,990B1 licensed to Charles Nemeroff. Charles Nemeroff has patent #US 7,148,027B2 licensed to Charles Nemeroff. Charles B Nemeroff is a compensated board member for Lucy Scientific Discovery; a consultant for Abbvie, AneuroTech (division of Anima BV), Intra-Cellular Therapies, Inc., EMA Wellness, Sage, Silo Pharma, Engrail Therapeutics, GoodCap Pharmaceuticals, Inc., Senseye, Clexio, EmbarkNeuro, Relmada Therapeutics, Galen Mental Health, LLC, and AutoBahn Therapeutics, Inc; a scientific advisory board member for ANeuoTech (division of Anima BV), Brain and Behavior Research Foundation (BBRF), Anxiety and Depression Association of America (ADAA), Skyland Trail, Signant Health, Laureate Institute for Brain Research (LIRB), Inc., and Sage; and owns stock in Corcept Therapeutics EMA Wellness, Precisement Health, Senseye, and Relmada Therapeutics. Dr. Beurel has not conflict of interest.

References

  1. Felitti, V.J.; Anda, R.F.; Nordenberg, D.; Williamson, D.F.; Spitz, A.M.; Edwards, V.; Koss, M.P.; Marks, J.S. Relationship of childhood abuse and household dysfunction to many of the leading causes of death in adults. The Adverse Childhood Experiences (ACE) Study. Am. J. Prev. Med. 1998, 14, 245–258. [Google Scholar] [CrossRef] [PubMed]
  2. Kalmakis, K.A.; Chandler, G.E. Health consequences of adverse childhood experiences: A systematic review. J. Am. Assoc. Nurse Pract. 2015, 27, 457–465. [Google Scholar] [CrossRef]
  3. Kessler, R.C.; McLaughlin, K.A.; Green, J.G.; Gruber, M.J.; Sampson, N.A.; Zaslavsky, A.M.; Aguilar-Gaxiola, S.; Alhamzawi, A.O.; Alonso, J.; Angermeyer, M.; et al. Childhood adversities and adult psychopathology in the WHO World Mental Health Surveys. Br. J. Psychiatry 2010, 197, 378–385. [Google Scholar] [CrossRef] [PubMed]
  4. Korkeila, J.; Vahtera, J.; Korkeila, K.; Kivimaki, M.; Sumanen, M.; Koskenvuo, K.; Koskenvuo, M. Childhood adversities as predictors of incident coronary heart disease and cerebrovascular disease. Heart 2010, 96, 298–303. [Google Scholar] [CrossRef]
  5. Chen, E.; Turiano, N.A.; Mroczek, D.K.; Miller, G.E. Association of Reports of Childhood Abuse and All-Cause Mortality Rates in Women. JAMA Psychiatry 2016, 73, 920–927. [Google Scholar] [CrossRef]
  6. Kelly-Irving, M.; Lepage, B.; Dedieu, D.; Bartley, M.; Blane, D.; Grosclaude, P.; Lang, T.; Delpierre, C. Adverse childhood experiences and premature all-cause mortality. Eur. J. Epidemiol. 2013, 28, 721–734. [Google Scholar] [CrossRef] [PubMed]
  7. Bremner, J.D. Traumatic stress: Effects on the brain. Dialogues Clin. Neurosci. 2006, 8, 445–461. [Google Scholar] [CrossRef]
  8. Baumeister, D.; Akhtar, R.; Ciufolini, S.; Pariante, C.M.; Mondelli, V. Childhood trauma and adulthood inflammation: A meta-analysis of peripheral C-reactive protein, interleukin-6 and tumour necrosis factor-alpha. Mol. Psychiatry 2016, 21, 642–649. [Google Scholar] [CrossRef]
  9. Coelho, R.; Viola, T.W.; Walss-Bass, C.; Brietzke, E.; Grassi-Oliveira, R. Childhood maltreatment and inflammatory markers: A systematic review. Acta Psychiatr. Scand. 2014, 129, 180–192. [Google Scholar] [CrossRef]
  10. Boyce, W.T.; Levitt, P.; Martinez, F.D.; McEwen, B.S.; Shonkoff, J.P. Genes, Environments, and Time: The Biology of Adversity and Resilience. Pediatrics 2021, 147, e20201651. [Google Scholar] [CrossRef]
  11. Carpenter, L.L.; Gawuga, C.E.; Tyrka, A.R.; Lee, J.K.; Anderson, G.M.; Price, L.H. Association between plasma IL-6 response to acute stress and early-life adversity in healthy adults. Neuropsychopharmacology 2010, 35, 2617–2623. [Google Scholar] [CrossRef] [PubMed]
  12. Chen, E.; Miller, G.E.; Yu, T.; Brody, G.H. Unsupportive parenting moderates the effects of family psychosocial intervention on metabolic syndrome in African American youth. Int. J. Obes. 2018, 42, 634–640. [Google Scholar] [CrossRef] [PubMed]
  13. Danese, A.; Tan, M. Childhood maltreatment and obesity: Systematic review and meta-analysis. Mol. Psychiatry 2014, 19, 544–554. [Google Scholar] [CrossRef] [PubMed]
  14. Gouin, J.P.; Glaser, R.; Malarkey, W.B.; Beversdorf, D.; Kiecolt-Glaser, J.K. Childhood abuse and inflammatory responses to daily stressors. Ann. Behav. Med. 2012, 44, 287–292. [Google Scholar] [CrossRef] [PubMed]
  15. Miller, G.E.; Chen, E.; Parker, K.J. Psychological stress in childhood and susceptibility to the chronic diseases of aging: Moving toward a model of behavioral and biological mechanisms. Psychol. Bull. 2011, 137, 959–997. [Google Scholar] [CrossRef] [PubMed]
  16. Irwin, M.R.; Cole, S.W. Reciprocal regulation of the neural and innate immune systems. Nat. Rev. Immunol. 2011, 11, 625–632. [Google Scholar] [CrossRef] [PubMed]
  17. Sapolsky, R.M.; Romero, L.M.; Munck, A.U. How do glucocorticoids influence stress responses? Integrating permissive, suppressive, stimulatory, and preparative actions. Endocr. Rev. 2000, 21, 55–89. [Google Scholar] [CrossRef]
  18. Sternberg, E.M. Neural regulation of innate immunity: A coordinated nonspecific host response to pathogens. Nat. Rev. Immunol. 2006, 6, 318–328. [Google Scholar] [CrossRef] [PubMed]
  19. Simon, A.K.; Hollander, G.A.; McMichael, A. Evolution of the immune system in humans from infancy to old age. Proc. Biol. Sci. 2015, 282, 20143085. [Google Scholar] [CrossRef]
  20. Hayward, A.C.; Fragaszy, E.B.; Bermingham, A.; Wang, L.; Copas, A.; Edmunds, W.J.; Ferguson, N.; Goonetilleke, N.; Harvey, G.; Kovar, J.; et al. Comparative community burden and severity of seasonal and pandemic influenza: Results of the Flu Watch cohort study. Lancet Respir. Med. 2014, 2, 445–454. [Google Scholar] [CrossRef]
  21. CDC. Immunization Schedules. 2015. Available online: http://www.cdc.gov/vaccines/schedules (accessed on 7 April 2024).
  22. Klein, U.; Dalla-Favera, R. Germinal centres: Role in B-cell physiology and malignancy. Nat. Rev. Immunol. 2008, 8, 22–33. [Google Scholar] [CrossRef] [PubMed]
  23. Counotte, J.; Drexhage, H.A.; Wijkhuijs, J.M.; Pot-Kolder, R.; Bergink, V.; Hoek, H.W.; Veling, W. Th17/T regulator cell balance and NK cell numbers in relation to psychosis liability and social stress reactivity. Brain Behav. Immun. 2018, 69, 408–417. [Google Scholar] [CrossRef] [PubMed]
  24. Elwenspoek, M.M.C.; Hengesch, X.; Leenen, F.A.D.; Schritz, A.; Sias, K.; Schaan, V.K.; Meriaux, S.B.; Schmitz, S.; Bonnemberger, F.; Schachinger, H.; et al. Proinflammatory T Cell Status Associated with Early Life Adversity. J. Immunol. 2017, 199, 4046–4055. [Google Scholar] [CrossRef]
  25. Chakir, J.; Shannon, J.; Molet, S.; Fukakusa, M.; Elias, J.; Laviolette, M.; Boulet, L.P.; Hamid, Q. Airway remodeling-associated mediators in moderate to severe asthma: Effect of steroids on TGF-beta, IL-11, IL-17, and type I and type III collagen expression. J. Allergy Clin. Immunol. 2003, 111, 1293–1298. [Google Scholar] [CrossRef] [PubMed]
  26. Molet, S.; Hamid, Q.; Davoine, F.; Nutku, E.; Taha, R.; Page, N.; Olivenstein, R.; Elias, J.; Chakir, J. IL-17 is increased in asthmatic airways and induces human bronchial fibroblasts to produce cytokines. J. Allergy Clin. Immunol. 2001, 108, 430–438. [Google Scholar] [CrossRef]
  27. Chen, E.; Hanson, M.D.; Paterson, L.Q.; Griffin, M.J.; Walker, H.A.; Miller, G.E. Socioeconomic status and inflammatory processes in childhood asthma: The role of psychological stress. J. Allergy Clin. Immunol. 2006, 117, 1014–1020. [Google Scholar] [CrossRef]
  28. Schreier, H.M.C.; Chen, E.; Miller, G.E. Child maltreatment and pediatric asthma: A review of the literature. Asthma Res. Pract. 2016, 2, 7. [Google Scholar] [CrossRef] [PubMed]
  29. Beurel, E.; Medina-Rodriguez, E.M.; Jope, R.S. Targeting the Adaptive Immune System in Depression: Focus on T Helper 17 Cells. Pharmacol. Rev. 2022, 74, 373–386. [Google Scholar] [CrossRef]
  30. Cicchetti, D.; Toth, S.L. Child maltreatment. Annu. Rev. Clin. Psychol. 2005, 1, 409–438. [Google Scholar] [CrossRef]
  31. Evans, G.W. The environment of childhood poverty. Am. Psychol. 2004, 59, 77–92. [Google Scholar] [CrossRef]
  32. Azad, M.B.; Lissitsyn, Y.; Miller, G.E.; Becker, A.B.; HayGlass, K.T.; Kozyrskyj, A.L. Influence of socioeconomic status trajectories on innate immune responsiveness in children. PLoS ONE 2012, 7, e38669. [Google Scholar] [CrossRef] [PubMed]
  33. Wright, R.J.; Visness, C.M.; Calatroni, A.; Grayson, M.H.; Gold, D.R.; Sandel, M.T.; Lee-Parritz, A.; Wood, R.A.; Kattan, M.; Bloomberg, G.R.; et al. Prenatal maternal stress and cord blood innate and adaptive cytokine responses in an inner-city cohort. Am. J. Respir. Crit. Care Med. 2010, 182, 25–33. [Google Scholar] [CrossRef] [PubMed]
  34. Schreier, H.M.; Roy, L.B.; Frimer, L.T.; Chen, E. Family chaos and adolescent inflammatory profiles: The moderating role of socioeconomic status. Psychosom. Med. 2014, 76, 460–467. [Google Scholar] [CrossRef] [PubMed]
  35. Raison, C.L.; Miller, A.H. When not enough is too much: The role of insufficient glucocorticoid signaling in the pathophysiology of stress-related disorders. Am. J. Psychiatry 2003, 160, 1554–1565. [Google Scholar] [CrossRef] [PubMed]
  36. Tursich, M.; Neufeld, R.W.; Frewen, P.A.; Harricharan, S.; Kibler, J.L.; Rhind, S.G.; Lanius, R.A. Association of trauma exposure with proinflammatory activity: A transdiagnostic meta-analysis. Transl. Psychiatry 2014, 4, e413. [Google Scholar] [CrossRef] [PubMed]
  37. Elwenspoek, M.M.C.; Kuehn, A.; Muller, C.P.; Turner, J.D. The effects of early life adversity on the immune system. Psychoneuroendocrinology 2017, 82, 140–154. [Google Scholar] [CrossRef] [PubMed]
  38. Kuhlman, K.R.; Horn, S.R.; Chiang, J.J.; Bower, J.E. Early life adversity exposure and circulating markers of inflammation in children and adolescents: A systematic review and meta-analysis. Brain Behav. Immun. 2020, 86, 30–42. [Google Scholar] [CrossRef] [PubMed]
  39. Surtees, P.; Wainwright, N.; Day, N.; Luben, R.; Brayne, C.; Khaw, K.T. Association of depression with peripheral leukocyte counts in EPIC-Norfolk--role of sex and cigarette smoking. J. Psychosom. Res. 2003, 54, 303–306. [Google Scholar] [CrossRef] [PubMed]
  40. Poulton, R.; Moffitt, T.E.; Silva, P.A. The Dunedin Multidisciplinary Health and Development Study: Overview of the first 40 years, with an eye to the future. Soc. Psychiatry Psychiatr. Epidemiol. 2015, 50, 679–693. [Google Scholar] [CrossRef]
  41. Danese, A.; Pariante, C.M.; Caspi, A.; Taylor, A.; Poulton, R. Childhood maltreatment predicts adult inflammation in a life-course study. Proc. Natl. Acad. Sci. USA 2007, 104, 1319–1324. [Google Scholar] [CrossRef]
  42. Speer, K.; Upton, D.; Semple, S.; McKune, A. Systemic low-grade inflammation in post-traumatic stress disorder: A systematic review. J. Inflamm. Res. 2018, 11, 111–121. [Google Scholar] [CrossRef] [PubMed]
  43. Shirtcliff, E.A.; Coe, C.L.; Pollak, S.D. Early childhood stress is associated with elevated antibody levels to herpes simplex virus type 1. Proc. Natl. Acad. Sci. USA 2009, 106, 2963–2967. [Google Scholar] [CrossRef] [PubMed]
  44. Danese, A.; Moffitt, T.E.; Pariante, C.M.; Ambler, A.; Poulton, R.; Caspi, A. Elevated inflammation levels in depressed adults with a history of childhood maltreatment. Arch. Gen. Psychiatry 2008, 65, 409–415. [Google Scholar] [CrossRef] [PubMed]
  45. Kiecolt-Glaser, J.K.; Gouin, J.P.; Weng, N.P.; Malarkey, W.B.; Beversdorf, D.Q.; Glaser, R. Childhood adversity heightens the impact of later-life caregiving stress on telomere length and inflammation. Psychosom. Med. 2011, 73, 16–22. [Google Scholar] [CrossRef] [PubMed]
  46. Pace, T.W.; Mletzko, T.C.; Alagbe, O.; Musselman, D.L.; Nemeroff, C.B.; Miller, A.H.; Heim, C.M. Increased stress-induced inflammatory responses in male patients with major depression and increased early life stress. Am. J. Psychiatry 2006, 163, 1630–1633. [Google Scholar] [CrossRef] [PubMed]
  47. Zagaria, A.; Fiori, V.; Vacca, M.; Lombardo, C.; Pariante, C.M.; Ballesio, A. Inflammation as a mediator between adverse childhood experiences and adult depression: A meta-analytic structural equation model. J. Affect. Disord. 2024, 357, 85–96. [Google Scholar] [CrossRef] [PubMed]
  48. Danese, A.; Caspi, A.; Williams, B.; Ambler, A.; Sugden, K.; Mika, J.; Werts, H.; Freeman, J.; Pariante, C.M.; Moffitt, T.E.; et al. Biological embedding of stress through inflammation processes in childhood. Mol. Psychiatry 2011, 16, 244–246. [Google Scholar] [CrossRef] [PubMed]
  49. Slopen, N.; Kubzansky, L.D.; McLaughlin, K.A.; Koenen, K.C. Childhood adversity and inflammatory processes in youth: A prospective study. Psychoneuroendocrinology 2013, 38, 188–200. [Google Scholar] [CrossRef] [PubMed]
  50. Cicchetti, D.; Handley, E.D.; Rogosch, F.A. Child maltreatment, inflammation, and internalizing symptoms: Investigating the roles of C-reactive protein, gene variation, and neuroendocrine regulation. Dev. Psychopathol. 2015, 27, 553–566. [Google Scholar] [CrossRef]
  51. Pang, Y.; Kartsonaki, C.; Lv, J.; Fairhurst-Hunter, Z.; Millwood, I.Y.; Yu, C.; Guo, Y.; Chen, Y.; Bian, Z.; Yang, L.; et al. Associations of Adiposity, Circulating Protein Biomarkers, and Risk of Major Vascular Diseases. JAMA Cardiol. 2021, 6, 276–286. [Google Scholar] [CrossRef]
  52. Kuhlman, K.R.; Repetti, R.L.; Reynolds, B.M.; Robles, T.F. Change in parent-child conflict and the HPA-axis: Where should we be looking and for how long? Psychoneuroendocrinology 2016, 68, 74–81. [Google Scholar] [CrossRef]
  53. Kuhlman, K.R.; Repetti, R.L.; Reynolds, B.M.; Robles, T.F. Interparental conflict and child HPA-axis responses to acute stress: Insights using intensive repeated measures. J. Fam. Psychol. 2018, 32, 773–782. [Google Scholar] [CrossRef]
  54. Barnes, P.J. Anti-inflammatory actions of glucocorticoids: Molecular mechanisms. Clin. Sci. 1998, 94, 557–572. [Google Scholar] [CrossRef]
  55. Waage, A.; Slupphaug, G.; Shalaby, R. Glucocorticoids inhibit the production of IL6 from monocytes, endothelial cells and fibroblasts. Eur. J. Immunol. 1990, 20, 2439–2443. [Google Scholar] [CrossRef] [PubMed]
  56. Gunnar, M.R.; Donzella, B. Social regulation of the cortisol levels in early human development. Psychoneuroendocrinology 2002, 27, 199–220. [Google Scholar] [CrossRef]
  57. Gunnar, M.; Quevedo, K. The neurobiology of stress and development. Annu. Rev. Psychol. 2007, 58, 145–173. [Google Scholar] [CrossRef] [PubMed]
  58. Tarullo, A.R.; Gunnar, M.R. Child maltreatment and the developing HPA axis. Horm. Behav. 2006, 50, 632–639. [Google Scholar] [CrossRef]
  59. Miller, G.E.; Chen, E. Harsh family climate in early life presages the emergence of a proinflammatory phenotype in adolescence. Psychol. Sci. 2010, 21, 848–856. [Google Scholar] [CrossRef] [PubMed]
  60. Hanson, J.L.; Nacewicz, B.M.; Sutterer, M.J.; Cayo, A.A.; Schaefer, S.M.; Rudolph, K.D.; Shirtcliff, E.A.; Pollak, S.D.; Davidson, R.J. Behavioral problems after early life stress: Contributions of the hippocampus and amygdala. Biol. Psychiatry 2015, 77, 314–323. [Google Scholar] [CrossRef] [PubMed]
  61. Lovallo, W.R. Early life adversity reduces stress reactivity and enhances impulsive behavior: Implications for health behaviors. Int. J. Psychophysiol. 2013, 90, 8–16. [Google Scholar] [CrossRef]
  62. Rudolph, K.D.; Hammen, C. Age and gender as determinants of stress exposure, generation, and reactions in youngsters: A transactional perspective. Child. Dev. 1999, 70, 660–677. [Google Scholar] [CrossRef] [PubMed]
  63. Rudolph, K.D.; Hammen, C.; Burge, D.; Lindberg, N.; Herzberg, D.; Daley, S.E. Toward an interpersonal life-stress model of depression: The developmental context of stress generation. Dev. Psychopathol. 2000, 12, 215–234. [Google Scholar] [CrossRef] [PubMed]
  64. Miller, A.H.; Haroon, E.; Raison, C.L.; Felger, J.C. Cytokine targets in the brain: Impact on neurotransmitters and neurocircuits. Depress. Anxiety 2013, 30, 297–306. [Google Scholar] [CrossRef] [PubMed]
  65. Slavich, G.M.; Irwin, M.R. From stress to inflammation and major depressive disorder: A social signal transduction theory of depression. Psychol. Bull. 2014, 140, 774–815. [Google Scholar] [CrossRef] [PubMed]
  66. Dillon, D.G.; Holmes, A.J.; Birk, J.L.; Brooks, N.; Lyons-Ruth, K.; Pizzagalli, D.A. Childhood adversity is associated with left basal ganglia dysfunction during reward anticipation in adulthood. Biol. Psychiatry 2009, 66, 206–213. [Google Scholar] [CrossRef] [PubMed]
  67. Guyer, A.E.; Kaufman, J.; Hodgdon, H.B.; Masten, C.L.; Jazbec, S.; Pine, D.S.; Ernst, M. Behavioral alterations in reward system function: The role of childhood maltreatment and psychopathology. J. Am. Acad. Child. Adolesc. Psychiatry 2006, 45, 1059–1067. [Google Scholar] [CrossRef] [PubMed]
  68. Maier, S.F.; Watkins, L.R. Cytokines for psychologists: Implications of bidirectional immune-to-brain communication for understanding behavior, mood, and cognition. Psychol. Rev. 1998, 105, 83–107. [Google Scholar] [CrossRef] [PubMed]
  69. Miller, A.H.; Maletic, V.; Raison, C.L. Inflammation and its discontents: The role of cytokines in the pathophysiology of major depression. Biol. Psychiatry 2009, 65, 732–741. [Google Scholar] [CrossRef] [PubMed]
  70. Dantzer, R.; O’Connor, J.C.; Freund, G.G.; Johnson, R.W.; Kelley, K.W. From inflammation to sickness and depression: When the immune system subjugates the brain. Nat. Rev. Neurosci. 2008, 9, 46–56. [Google Scholar] [CrossRef]
  71. Edmiston, E.E.; Wang, F.; Mazure, C.M.; Guiney, J.; Sinha, R.; Mayes, L.C.; Blumberg, H.P. Corticostriatal-limbic gray matter morphology in adolescents with self-reported exposure to childhood maltreatment. Arch. Pediatr. Adolesc. Med. 2011, 165, 1069–1077. [Google Scholar] [CrossRef]
  72. Gianaros, P.J.; Marsland, A.L.; Sheu, L.K.; Erickson, K.I.; Verstynen, T.D. Inflammatory pathways link socioeconomic inequalities to white matter architecture. Cereb. Cortex 2013, 23, 2058–2071. [Google Scholar] [CrossRef] [PubMed]
  73. Hanson, J.L.; Hair, N.; Shen, D.G.; Shi, F.; Gilmore, J.H.; Wolfe, B.L.; Pollak, S.D. Family poverty affects the rate of human infant brain growth. PLoS ONE 2013, 8, e80954. [Google Scholar] [CrossRef] [PubMed]
  74. Lawson, G.M.; Duda, J.T.; Avants, B.B.; Wu, J.; Farah, M.J. Associations between children’s socioeconomic status and prefrontal cortical thickness. Dev. Sci. 2013, 16, 641–652. [Google Scholar] [CrossRef] [PubMed]
  75. Luby, J.; Belden, A.; Botteron, K.; Marrus, N.; Harms, M.P.; Babb, C.; Nishino, T.; Barch, D. The effects of poverty on childhood brain development: The mediating effect of caregiving and stressful life events. JAMA Pediatr. 2013, 167, 1135–1142. [Google Scholar] [CrossRef] [PubMed]
  76. Marsland, A.L.; Gianaros, P.J.; Abramowitch, S.M.; Manuck, S.B.; Hariri, A.R. Interleukin-6 covaries inversely with hippocampal grey matter volume in middle-aged adults. Biol. Psychiatry 2008, 64, 484–490. [Google Scholar] [CrossRef] [PubMed]
  77. Satizabal, C.L.; Zhu, Y.C.; Mazoyer, B.; Dufouil, C.; Tzourio, C. Circulating IL-6 and CRP are associated with MRI findings in the elderly: The 3C-Dijon Study. Neurology 2012, 78, 720–727. [Google Scholar] [CrossRef] [PubMed]
  78. Danese, A.; McEwen, B.S. Adverse childhood experiences, allostasis, allostatic load, and age-related disease. Physiol. Behav. 2012, 106, 29–39. [Google Scholar] [CrossRef] [PubMed]
  79. Shonkoff, J.P.; Boyce, W.T.; McEwen, B.S. Neuroscience, molecular biology, and the childhood roots of health disparities: Building a new framework for health promotion and disease prevention. JAMA 2009, 301, 2252–2259. [Google Scholar] [CrossRef] [PubMed]
  80. Teicher, M.H.; Samson, J.A. Childhood maltreatment and psychopathology: A case for ecophenotypic variants as clinically and neurobiologically distinct subtypes. Am. J. Psychiatry 2013, 170, 1114–1133. [Google Scholar] [CrossRef]
  81. Nusslock, R.; Miller, G.E. Early-Life Adversity and Physical and Emotional Health Across the Lifespan: A Neuroimmune Network Hypothesis. Biol. Psychiatry 2016, 80, 23–32. [Google Scholar] [CrossRef]
  82. Bartlett, J.A.; Demetrikopoulos, M.K.; Schleifer, S.J.; Keller, S.E. Phagocytosis and killing of Staphylococcus aureus: Effects of stress and depression in children. Clin. Diagn. Lab. Immunol. 1997, 4, 362–366. [Google Scholar] [CrossRef] [PubMed]
  83. Birmaher, B.; Rabin, B.S.; Garcia, M.R.; Jain, U.; Whiteside, T.L.; Williamson, D.E.; al-Shabbout, M.; Nelson, B.C.; Dahl, R.E.; Ryan, N.D. Cellular immunity in depressed, conduct disorder, and normal adolescents: Role of adverse life events. J. Am. Acad. Child. Adolesc. Psychiatry 1994, 33, 671–678. [Google Scholar] [CrossRef] [PubMed]
  84. Witek Janusek, L.; Tell, D.; Albuquerque, K.; Mathews, H.L. Childhood adversity increases vulnerability for behavioral symptoms and immune dysregulation in women with breast cancer. Brain Behav. Immun. 2013, 30, S149–S162. [Google Scholar] [CrossRef] [PubMed]
  85. Wyman, P.A.; Moynihan, J.; Eberly, S.; Cox, C.; Cross, W.; Jin, X.; Caserta, M.T. Association of family stress with natural killer cell activity and the frequency of illnesses in children. Arch. Pediatr. Adolesc. Med. 2007, 161, 228–234. [Google Scholar] [CrossRef] [PubMed]
  86. Ayaydin, H.; Abali, O.; Akdeniz, N.O.; Kok, B.E.; Gunes, A.; Yildirim, A.; Deniz, G. Immune system changes after sexual abuse in adolescents. Pediatr. Int. 2016, 58, 105–112. [Google Scholar] [CrossRef] [PubMed]
  87. Chen, E.; Chim, L.S.; Strunk, R.C.; Miller, G.E. The role of the social environment in children and adolescents with asthma. Am. J. Respir. Crit. Care Med. 2007, 176, 644–649. [Google Scholar] [CrossRef]
  88. Wright, R.J.; Finn, P.; Contreras, J.P.; Cohen, S.; Wright, R.O.; Staudenmayer, J.; Wand, M.; Perkins, D.; Weiss, S.T.; Gold, D.R. Chronic caregiver stress and IgE expression, allergen-induced proliferation, and cytokine profiles in a birth cohort predisposed to atopy. J. Allergy Clin. Immunol. 2004, 113, 1051–1057. [Google Scholar] [CrossRef]
  89. Fukunaga, T.; Mizoi, Y.; Yamashita, A.; Yamada, M.; Yamamoto, Y.; Tatsuno, Y.; Nishi, K. Thymus of abused/neglected children. Forensic Sci. Int. 1992, 53, 69–79. [Google Scholar] [CrossRef] [PubMed]
  90. Holland, J.F.; Khandaker, G.M.; Dauvermann, M.R.; Morris, D.; Zammit, S.; Donohoe, G. Effects of early life adversity on immune function and cognitive performance: Results from the ALSPAC cohort. Soc. Psychiatry Psychiatr. Epidemiol. 2020, 55, 723–733. [Google Scholar] [CrossRef]
  91. Reid, B.M.; Coe, C.L.; Doyle, C.M.; Sheerar, D.; Slukvina, A.; Donzella, B.; Gunnar, M.R. Persistent skewing of the T-cell profile in adolescents adopted internationally from institutional care. Brain Behav. Immun. 2019, 77, 168–177. [Google Scholar] [CrossRef]
  92. Elwenspoek, M.M.C.; Sias, K.; Hengesch, X.; Schaan, V.K.; Leenen, F.A.D.; Adams, P.; Meriaux, S.B.; Schmitz, S.; Bonnemberger, F.; Ewen, A.; et al. T Cell Immunosenescence after Early Life Adversity: Association with Cytomegalovirus Infection. Front. Immunol. 2017, 8, 1263. [Google Scholar] [CrossRef] [PubMed]
  93. Esposito, E.A.; Jones, M.J.; Doom, J.R.; MacIsaac, J.L.; Gunnar, M.R.; Kobor, M.S. Differential DNA methylation in peripheral blood mononuclear cells in adolescents exposed to significant early but not later childhood adversity. Dev. Psychopathol. 2016, 28, 1385–1399. [Google Scholar] [CrossRef] [PubMed]
  94. Caserta, M.T.; O’Connor, T.G.; Wyman, P.A.; Wang, H.; Moynihan, J.; Cross, W.; Tu, X.; Jin, X. The associations between psychosocial stress and the frequency of illness, and innate and adaptive immune function in children. Brain Behav. Immun. 2008, 22, 933–940. [Google Scholar] [CrossRef] [PubMed]
  95. Clover, R.D.; Abell, T.; Becker, L.A.; Crawford, S.; Ramsey, C.N., Jr. Family functioning and stress as predictors of influenza B infection. J. Fam. Pract. 1989, 28, 535–539. [Google Scholar] [PubMed]
  96. Boyce, W.T.; Chesney, M.; Alkon, A.; Tschann, J.M.; Adams, S.; Chesterman, B.; Cohen, F.; Kaiser, P.; Folkman, S.; Wara, D. Psychobiologic reactivity to stress and childhood respiratory illnesses: Results of two prospective studies. Psychosom. Med. 1995, 57, 411–422. [Google Scholar] [CrossRef] [PubMed]
  97. Meyer, R.J.; Haggerty, R.J. Streptococcal infections in families. Factors altering individual susceptibility. Pediatrics 1962, 29, 539–549. [Google Scholar] [CrossRef] [PubMed]
  98. Wright, R.J.; Cohen, S.; Carey, V.; Weiss, S.T.; Gold, D.R. Parental stress as a predictor of wheezing in infancy: A prospective birth-cohort study. Am. J. Respir. Crit. Care Med. 2002, 165, 358–365. [Google Scholar] [CrossRef]
  99. Cohen, S.; Doyle, W.J.; Turner, R.B.; Alper, C.M.; Skoner, D.P. Childhood socioeconomic status and host resistance to infectious illness in adulthood. Psychosom. Med. 2004, 66, 553–558. [Google Scholar] [CrossRef] [PubMed]
  100. Miller, G.E.; Chen, E.; Fok, A.K.; Walker, H.; Lim, A.; Nicholls, E.F.; Cole, S.; Kobor, M.S. Low early-life social class leaves a biological residue manifested by decreased glucocorticoid and increased proinflammatory signaling. Proc. Natl. Acad. Sci. USA 2009, 106, 14716–14721. [Google Scholar] [CrossRef]
  101. Ziol-Guest, K.M.; Duncan, G.J.; Kalil, A.; Boyce, W.T. Early childhood poverty, immune-mediated disease processes, and adult productivity. Proc. Natl. Acad. Sci. USA 2012, 109 (Suppl. 2), 17289–17293. [Google Scholar] [CrossRef]
  102. Cohen, S.; Chiang, J.J.; Janicki-Deverts, D.; Miller, G.E. Good Relationships with Parents During Childhood as Buffers of the Association between Childhood Disadvantage and Adult Susceptibility to the Common Cold. Psychosom. Med. 2020, 82, 538–547. [Google Scholar] [CrossRef] [PubMed]
  103. Hayward, S.E.; Dowd, J.B.; Fletcher, H.; Nellums, L.B.; Wurie, F.; Boccia, D. A systematic review of the impact of psychosocial factors on immunity: Implications for enhancing BCG response against tuberculosis. SSM Popul. Health 2020, 10, 100522. [Google Scholar] [CrossRef] [PubMed]
  104. O’Connor, T.G.; Wang, H.; Moynihan, J.A.; Wyman, P.A.; Carnahan, J.; Lofthus, G.; Quataert, S.A.; Bowman, M.; Burke, A.S.; Caserta, M.T. Observed parent-child relationship quality predicts antibody response to vaccination in children. Brain Behav. Immun. 2015, 48, 265–273. [Google Scholar] [CrossRef] [PubMed]
  105. Madison, A.A.; Shrout, M.R.; Renna, M.E.; Kiecolt-Glaser, J.K. Psychological and Behavioral Predictors of Vaccine Efficacy: Considerations for COVID-19. Perspect. Psychol. Sci. 2021, 16, 191–203. [Google Scholar] [CrossRef] [PubMed]
  106. Stowe, R.P.; Peek, M.K.; Perez, N.A.; Yetman, D.L.; Cutchin, M.P.; Goodwin, J.S. Herpesvirus reactivation and socioeconomic position: A community-based study. J. Epidemiol. Community Health 2010, 64, 666–671. [Google Scholar] [CrossRef] [PubMed]
  107. McDade, T.W.; Stallings, J.F.; Angold, A.; Costello, E.J.; Burleson, M.; Cacioppo, J.T.; Glaser, R.; Worthman, C.M. Epstein-Barr virus antibodies in whole blood spots: A minimally invasive method for assessing an aspect of cell-mediated immunity. Psychosom. Med. 2000, 62, 560–567. [Google Scholar] [CrossRef] [PubMed]
  108. Schmeer, K.K.; Ford, J.L.; Browning, C.R. Early childhood family instability and immune system dysregulation in adolescence. Psychoneuroendocrinology 2019, 102, 189–195. [Google Scholar] [CrossRef] [PubMed]
  109. Lendor, C.; Johnson, A.; Perzanowski, M.; Chew, G.L.; Goldstein, I.F.; Kelvin, E.; Perera, F.; Miller, R.L. Effects of winter birth season and prenatal cockroach and mouse allergen exposure on indoor allergen-specific cord blood mononuclear cell proliferation and cytokine production. Ann. Allergy Asthma Immunol. 2008, 101, 193–199. [Google Scholar] [CrossRef] [PubMed]
  110. Rowe, J.; Kusel, M.; Holt, B.J.; Suriyaarachchi, D.; Serralha, M.; Hollams, E.; Yerkovich, S.T.; Subrata, L.S.; Ladyman, C.; Sadowska, A.; et al. Prenatal versus postnatal sensitization to environmental allergens in a high-risk birth cohort. J. Allergy Clin. Immunol. 2007, 119, 1164–1173. [Google Scholar] [CrossRef]
  111. Backhed, F. Programming of host metabolism by the gut microbiota. Ann. Nutr. Metab. 2011, 58 (Suppl. 2), 44–52. [Google Scholar] [CrossRef]
  112. Aatsinki, A.K.; Keskitalo, A.; Laitinen, V.; Munukka, E.; Uusitupa, H.M.; Lahti, L.; Kortesluoma, S.; Mustonen, P.; Rodrigues, A.J.; Coimbra, B.; et al. Maternal prenatal psychological distress and hair cortisol levels associate with infant fecal microbiota composition at 2.5 months of age. Psychoneuroendocrinology 2020, 119, 104754. [Google Scholar] [CrossRef] [PubMed]
  113. Grant-Beurmann, S.; Jumare, J.; Ndembi, N.; Matthew, O.; Shutt, A.; Omoigberale, A.; Martin, O.A.; Fraser, C.M.; Charurat, M. Dynamics of the infant gut microbiota in the first 18 months of life: The impact of maternal HIV infection and breastfeeding. Microbiome 2022, 10, 61. [Google Scholar] [CrossRef] [PubMed]
  114. Jahnke, J.R.; Roach, J.; Azcarate-Peril, M.A.; Thompson, A.L. Maternal precarity and HPA axis functioning shape infant gut microbiota and HPA axis development in humans. PLoS ONE 2021, 16, e0251782. [Google Scholar] [CrossRef] [PubMed]
  115. Zijlmans, M.A.; Korpela, K.; Riksen-Walraven, J.M.; de Vos, W.M.; de Weerth, C. Maternal prenatal stress is associated with the infant intestinal microbiota. Psychoneuroendocrinology 2015, 53, 233–245. [Google Scholar] [CrossRef] [PubMed]
  116. Hermes, G.D.A.; Eckermann, H.A.; de Vos, W.M.; de Weerth, C. Does entry to center-based childcare affect gut microbial colonization in young infants? Sci. Rep. 2020, 10, 10235. [Google Scholar] [CrossRef] [PubMed]
  117. Keskitalo, A.; Aatsinki, A.K.; Kortesluoma, S.; Pelto, J.; Korhonen, L.; Lahti, L.; Lukkarinen, M.; Munukka, E.; Karlsson, H.; Karlsson, L. Gut microbiota diversity but not composition is related to saliva cortisol stress response at the age of 2.5 months. Stress 2021, 24, 551–560. [Google Scholar] [CrossRef] [PubMed]
  118. Jasarevic, E.; Howerton, C.L.; Howard, C.D.; Bale, T.L. Alterations in the Vaginal Microbiome by Maternal Stress Are Associated with Metabolic Reprogramming of the Offspring Gut and Brain. Endocrinology 2015, 156, 3265–3276. [Google Scholar] [CrossRef] [PubMed]
  119. Reid, B.M.; Horne, R.; Donzella, B.; Szamosi, J.C.; Coe, C.L.; Foster, J.A.; Gunnar, M.R. Microbiota-immune alterations in adolescents following early life adversity: A proof of concept study. Dev. Psychobiol. 2021, 63, 851–863. [Google Scholar] [CrossRef]
  120. Callaghan, B.L.; Fields, A.; Gee, D.G.; Gabard-Durnam, L.; Caldera, C.; Humphreys, K.L.; Goff, B.; Flannery, J.; Telzer, E.H.; Shapiro, M.; et al. Mind and gut: Associations between mood and gastrointestinal distress in children exposed to adversity. Dev. Psychopathol. 2020, 32, 309–328. [Google Scholar] [CrossRef]
  121. De Filippo, C.; Cavalieri, D.; Di Paola, M.; Ramazzotti, M.; Poullet, J.B.; Massart, S.; Collini, S.; Pieraccini, G.; Lionetti, P. Impact of diet in shaping gut microbiota revealed by a comparative study in children from Europe and rural Africa. Proc. Natl. Acad. Sci. USA 2010, 107, 14691–14696. [Google Scholar] [CrossRef]
  122. Wu, G.D.; Chen, J.; Hoffmann, C.; Bittinger, K.; Chen, Y.Y.; Keilbaugh, S.A.; Bewtra, M.; Knights, D.; Walters, W.A.; Knight, R.; et al. Linking long-term dietary patterns with gut microbial enterotypes. Science 2011, 334, 105–108. [Google Scholar] [CrossRef] [PubMed]
  123. Wu, G.D.; Compher, C.; Chen, E.Z.; Smith, S.A.; Shah, R.D.; Bittinger, K.; Chehoud, C.; Albenberg, L.G.; Nessel, L.; Gilroy, E.; et al. Comparative metabolomics in vegans and omnivores reveal constraints on diet-dependent gut microbiota metabolite production. Gut 2016, 65, 63–72. [Google Scholar] [CrossRef]
  124. Dillon, S.M.; Lee, E.J.; Kotter, C.V.; Austin, G.L.; Gianella, S.; Siewe, B.; Smith, D.M.; Landay, A.L.; McManus, M.C.; Robertson, C.E.; et al. Gut dendritic cell activation links an altered colonic microbiome to mucosal and systemic T-cell activation in untreated HIV-1 infection. Mucosal Immunol. 2016, 9, 24–37. [Google Scholar] [CrossRef] [PubMed]
  125. Charalambous, E.G.; Meriaux, S.B.; Guebels, P.; Muller, C.P.; Leenen, F.A.D.; Elwenspoek, M.M.C.; Thiele, I.; Hertel, J.; Turner, J.D. Early-Life Adversity Leaves Its Imprint on the Oral Microbiome for More Than 20 Years and Is Associated with Long-Term Immune Changes. Int. J. Mol. Sci. 2021, 22, 12682. [Google Scholar] [CrossRef] [PubMed]
  126. Wajid, A.; van Zanten, S.V.; Mughal, M.K.; Biringer, A.; Austin, M.P.; Vermeyden, L.; Kingston, D. Adversity in childhood and depression in pregnancy. Arch. Women’s Ment. Health 2020, 23, 169–180. [Google Scholar] [CrossRef] [PubMed]
  127. Dunkel Schetter, C.; Tanner, L. Anxiety, depression and stress in pregnancy: Implications for mothers, children, research, and practice. Curr. Opin. Psychiatry 2012, 25, 141–148. [Google Scholar] [CrossRef]
  128. Olsen, J.M. Integrative Review of Pregnancy Health Risks and Outcomes Associated with Adverse Childhood Experiences. J. Obstet. Gynecol. Neonatal Nurs. 2018, 47, 783–794. [Google Scholar] [CrossRef] [PubMed]
  129. Dachew, B.A.; Mamun, A.; Maravilla, J.C.; Alati, R. Association between hypertensive disorders of pregnancy and the development of offspring mental and behavioural problems: A systematic review and meta-analysis. Psychiatry Res. 2018, 260, 458–467. [Google Scholar] [CrossRef] [PubMed]
  130. Smith, M.V.; Gotman, N.; Yonkers, K.A. Early Childhood Adversity and Pregnancy Outcomes. Matern. Child. Health J. 2016, 20, 790–798. [Google Scholar] [CrossRef]
  131. Gillespie, S.L.; Christian, L.M.; Alston, A.D.; Salsberry, P.J. Childhood stress and birth timing among African American women: Cortisol as biological mediator. Psychoneuroendocrinology 2017, 84, 32–41. [Google Scholar] [CrossRef]
  132. Miller, G.E.; Culhane, J.; Grobman, W.; Simhan, H.; Williamson, D.E.; Adam, E.K.; Buss, C.; Entringer, S.; Kim, K.Y.; Felipe Garcia-Espana, J.; et al. Mothers’ childhood hardship forecasts adverse pregnancy outcomes: Role of inflammatory, lifestyle, and psychosocial pathways. Brain Behav. Immun. 2017, 65, 11–19. [Google Scholar] [CrossRef] [PubMed]
  133. Angerud, K.; Annerback, E.M.; Tyden, T.; Boddeti, S.; Kristiansson, P. Adverse childhood experiences and depressive symptomatology among pregnant women. Acta Obstet. Gynecol. Scand. 2018, 97, 701–708. [Google Scholar] [CrossRef] [PubMed]
  134. Mamun, A.; Biswas, T.; Scott, J.; Sly, P.D.; McIntyre, H.D.; Thorpe, K.; Boyle, F.M.; Dekker, M.N.; Doi, S.; Mitchell, M.; et al. Adverse childhood experiences, the risk of pregnancy complications and adverse pregnancy outcomes: A systematic review and meta-analysis. BMJ Open 2023, 13, e063826. [Google Scholar] [CrossRef] [PubMed]
  135. Conn, A.M.; Szilagyi, M.A.; Jee, S.H.; Manly, J.T.; Briggs, R.; Szilagyi, P.G. Parental perspectives of screening for adverse childhood experiences in pediatric primary care. Fam. Syst. Health 2018, 36, 62–72. [Google Scholar] [CrossRef] [PubMed]
  136. Connell, A.M.; Goodman, S.H. The association between psychopathology in fathers versus mothers and children’s internalizing and externalizing behavior problems: A meta-analysis. Psychol. Bull. 2002, 128, 746–773. [Google Scholar] [CrossRef] [PubMed]
  137. Howell, B.R.; Sanchez, M.M. Understanding behavioral effects of early life stress using the reactive scope and allostatic load models. Dev. Psychopathol. 2011, 23, 1001–1016. [Google Scholar] [CrossRef] [PubMed]
  138. Widom, C.S.; Czaja, S.J.; DuMont, K.A. Intergenerational transmission of child abuse and neglect: Real or detection bias? Science 2015, 347, 1480–1485. [Google Scholar] [CrossRef] [PubMed]
  139. Bowers, M.E.; Yehuda, R. Intergenerational Transmission of Stress in Humans. Neuropsychopharmacology 2016, 41, 232–244. [Google Scholar] [CrossRef] [PubMed]
  140. Fisher, P.A.; Beauchamp, K.G.; Roos, L.E.; Noll, L.K.; Flannery, J.; Delker, B.C. The Neurobiology of Intervention and Prevention in Early Adversity. Annu. Rev. Clin. Psychol. 2016, 12, 331–357. [Google Scholar] [CrossRef]
  141. Querdasi, F.R.; Enders, C.; Karnani, N.; Broekman, B.; Yap Seng, C.; Gluckman, P.D.; Mary Daniel, L.; Yap, F.; Eriksson, J.G.; Cai, S.; et al. Multigenerational adversity impacts on human gut microbiome composition and socioemotional functioning in early childhood. Proc. Natl. Acad. Sci. USA 2023, 120, e2213768120. [Google Scholar] [CrossRef]
  142. Macpherson, A.J.; de Aguero, M.G.; Ganal-Vonarburg, S.C. How nutrition and the maternal microbiota shape the neonatal immune system. Nat. Rev. Immunol. 2017, 17, 508–517. [Google Scholar] [CrossRef] [PubMed]
  143. Moog, N.K.; Entringer, S.; Rasmussen, J.M.; Styner, M.; Gilmore, J.H.; Kathmann, N.; Heim, C.M.; Wadhwa, P.D.; Buss, C. Intergenerational Effect of Maternal Exposure to Childhood Maltreatment on Newborn Brain Anatomy. Biol. Psychiatry 2018, 83, 120–127. [Google Scholar] [CrossRef] [PubMed]
  144. Ader, R.; Friedman, S.B. Differential Early Experiences and Susceptibility to Transplanted Tumor in the Rat. J. Comp. Physiol. Psychol. 1965, 59, 361–364. [Google Scholar] [CrossRef] [PubMed]
  145. Solomon, G.F.; Levine, S.; Kraft, J.K. Early experience and immunity. Nature 1968, 220, 821–822. [Google Scholar] [CrossRef] [PubMed]
  146. Corcoran, C.A.; Pierre, P.J.; Haddad, T.; Bice, C.; Suomi, S.J.; Grant, K.A.; Friedman, D.P.; Bennett, A.J. Long-term effects of differential early rearing in rhesus macaques: Behavioral reactivity in adulthood. Dev. Psychobiol. 2012, 54, 546–555. [Google Scholar] [CrossRef] [PubMed]
  147. Howell, B.R.; Grand, A.P.; McCormack, K.M.; Shi, Y.; LaPrarie, J.L.; Maestripieri, D.; Styner, M.A.; Sanchez, M.M. Early adverse experience increases emotional reactivity in juvenile rhesus macaques: Relation to amygdala volume. Dev. Psychobiol. 2014, 56, 1735–1746. [Google Scholar] [CrossRef] [PubMed]
  148. Schino, G.; Speranza, L.; Troisi, A. Early maternal rejection and later social anxiety in juvenile and adult Japanese macaques. Dev. Psychobiol. 2001, 38, 186–190. [Google Scholar] [CrossRef]
  149. Ganguly, P.; Brenhouse, H.C. Broken or maladaptive? Altered trajectories in neuroinflammation and behavior after early life adversity. Dev. Cogn. Neurosci. 2015, 11, 18–30. [Google Scholar] [CrossRef] [PubMed]
  150. Hennessy, M.B.; Deak, T.; Schiml-Webb, P.A. Early attachment-figure separation and increased risk for later depression: Potential mediation by proinflammatory processes. Neurosci. Biobehav. Rev. 2010, 34, 782–790. [Google Scholar] [CrossRef]
  151. Shanks, N.; Lightman, S.L. The maternal-neonatal neuro-immune interface: Are there long-term implications for inflammatory or stress-related disease? J. Clin. Investig. 2001, 108, 1567–1573. [Google Scholar] [CrossRef]
  152. Coe, C.L.; Rosenberg, L.T.; Levine, S. Effect of maternal separation on the complement system and antibody responses in infant primates. Int. J. Neurosci. 1988, 40, 289–302. [Google Scholar] [CrossRef] [PubMed]
  153. Cole, S.W.; Conti, G.; Arevalo, J.M.; Ruggiero, A.M.; Heckman, J.J.; Suomi, S.J. Transcriptional modulation of the developing immune system by early life social adversity. Proc. Natl. Acad. Sci. USA 2012, 109, 20578–20583. [Google Scholar] [CrossRef] [PubMed]
  154. Wieck, A.; Andersen, S.L.; Brenhouse, H.C. Evidence for a neuroinflammatory mechanism in delayed effects of early life adversity in rats: Relationship to cortical NMDA receptor expression. Brain Behav. Immun. 2013, 28, 218–226. [Google Scholar] [CrossRef] [PubMed]
  155. Hennessy, M.B.; Deak, T.; Schiml-Webb, P.A.; Carlisle, C.W.; O’Brien, E. Maternal separation produces, and a second separation enhances, core temperature and passive behavioral responses in guinea pig pups. Physiol. Behav. 2010, 100, 305–310. [Google Scholar] [CrossRef] [PubMed]
  156. Lumertz, F.S.; Kestering-Ferreira, E.; Orso, R.; Creutzberg, K.C.; Tractenberg, S.G.; Stocchero, B.A.; Viola, T.W.; Grassi-Oliveira, R. Effects of early life stress on brain cytokines: A systematic review and meta-analysis of rodent studies. Neurosci. Biobehav. Rev. 2022, 139, 104746. [Google Scholar] [CrossRef] [PubMed]
  157. Bekhbat, M.; Neigh, G.N. Sex differences in the neuro-immune consequences of stress: Focus on depression and anxiety. Brain Behav. Immun. 2018, 67, 1–12. [Google Scholar] [CrossRef]
  158. Engler, H.; Benson, S.; Wegner, A.; Spreitzer, I.; Schedlowski, M.; Elsenbruch, S. Men and women differ in inflammatory and neuroendocrine responses to endotoxin but not in the severity of sickness symptoms. Brain Behav. Immun. 2016, 52, 18–26. [Google Scholar] [CrossRef]
  159. Wei, L.; Simen, A.; Mane, S.; Kaffman, A. Early life stress inhibits expression of a novel innate immune pathway in the developing hippocampus. Neuropsychopharmacology 2012, 37, 567–580. [Google Scholar] [CrossRef]
  160. Chocyk, A.; Dudys, D.; Przyborowska, A.; Majcher, I.; Mackowiak, M.; Wedzony, K. Maternal separation affects the number, proliferation and apoptosis of glia cells in the substantia nigra and ventral tegmental area of juvenile rats. Neuroscience 2011, 173, 1–18. [Google Scholar] [CrossRef]
  161. Viviani, B.; Boraso, M.; Valero, M.; Gardoni, F.; Marco, E.M.; Llorente, R.; Corsini, E.; Galli, C.L.; Di Luca, M.; Marinovich, M.; et al. Early maternal deprivation immunologically primes hippocampal synapses by redistributing interleukin-1 receptor type I in a sex dependent manner. Brain Behav. Immun. 2014, 35, 135–143. [Google Scholar] [CrossRef]
  162. Takatsuru, Y.; Nabekura, J.; Ishikawa, T.; Kohsaka, S.; Koibuchi, N. Early-life stress increases the motility of microglia in adulthood. J. Physiol. Sci. 2015, 65, 187–194. [Google Scholar] [CrossRef] [PubMed]
  163. Bachiller, S.; Paulus, A.; Vazquez-Reyes, S.; Garcia-Dominguez, I.; Deierborg, T. Maternal separation leads to regional hippocampal microglial activation and alters the behavior in the adolescence in a sex-specific manner. Brain Behav. Immun. Health 2020, 9, 100142. [Google Scholar] [CrossRef]
  164. Avitsur, R.; Hunzeker, J.; Sheridan, J.F. Role of early stress in the individual differences in host response to viral infection. Brain Behav. Immun. 2006, 20, 339–348. [Google Scholar] [CrossRef]
  165. Farrell, M.R.; Holland, F.H.; Shansky, R.M.; Brenhouse, H.C. Sex-specific effects of early life stress on social interaction and prefrontal cortex dendritic morphology in young rats. Behav. Brain Res. 2016, 310, 119–125. [Google Scholar] [CrossRef] [PubMed]
  166. Foley, K.A.; Ossenkopp, K.P.; Kavaliers, M.; Macfabe, D.F. Pre- and neonatal exposure to lipopolysaccharide or the enteric metabolite, propionic acid, alters development and behavior in adolescent rats in a sexually dimorphic manner. PLoS ONE 2014, 9, e87072. [Google Scholar] [CrossRef]
  167. Monte, A.S.; Mello, B.S.F.; Borella, V.C.M.; da Silva Araujo, T.; da Silva, F.E.R.; Sousa, F.C.F.; de Oliveira, A.C.P.; Gama, C.S.; Seeman, M.V.; Vasconcelos, S.M.M.; et al. Two-hit model of schizophrenia induced by neonatal immune activation and peripubertal stress in rats: Study of sex differences and brain oxidative alterations. Behav. Brain Res. 2017, 331, 30–37. [Google Scholar] [CrossRef] [PubMed]
  168. Mourlon, V.; Baudin, A.; Blanc, O.; Lauber, A.; Giros, B.; Naudon, L.; Dauge, V. Maternal deprivation induces depressive-like behaviours only in female rats. Behav. Brain Res. 2010, 213, 278–287. [Google Scholar] [CrossRef] [PubMed]
  169. Slotten, H.A.; Kalinichev, M.; Hagan, J.J.; Marsden, C.A.; Fone, K.C. Long-lasting changes in behavioural and neuroendocrine indices in the rat following neonatal maternal separation: Gender-dependent effects. Brain Res. 2006, 1097, 123–132. [Google Scholar] [CrossRef] [PubMed]
  170. Fernandez de Cossio, L.; Guzman, A.; van der Veldt, S.; Luheshi, G.N. Prenatal infection leads to ASD-like behavior and altered synaptic pruning in the mouse offspring. Brain Behav. Immun. 2017, 63, 88–98. [Google Scholar] [CrossRef]
  171. Hsiao, E.Y.; McBride, S.W.; Hsien, S.; Sharon, G.; Hyde, E.R.; McCue, T.; Codelli, J.A.; Chow, J.; Reisman, S.E.; Petrosino, J.F.; et al. Microbiota modulate behavioral and physiological abnormalities associated with neurodevelopmental disorders. Cell 2013, 155, 1451–1463. [Google Scholar] [CrossRef]
  172. Kim, S.; Kim, H.; Yim, Y.S.; Ha, S.; Atarashi, K.; Tan, T.G.; Longman, R.S.; Honda, K.; Littman, D.R.; Choi, G.B.; et al. Maternal gut bacteria promote neurodevelopmental abnormalities in mouse offspring. Nature 2017, 549, 528–532. [Google Scholar] [CrossRef] [PubMed]
  173. Rincel, M.; Lepinay, A.L.; Delage, P.; Fioramonti, J.; Theodorou, V.S.; Laye, S.; Darnaudery, M. Maternal high-fat diet prevents developmental programming by early-life stress. Transl. Psychiatry 2016, 6, e966. [Google Scholar] [CrossRef] [PubMed]
  174. Shin, S.Y.; Han, S.H.; Woo, R.S.; Jang, S.H.; Min, S.S. Adolescent mice show anxiety- and aggressive-like behavior and the reduction of long-term potentiation in mossy fiber-CA3 synapses after neonatal maternal separation. Neuroscience 2016, 316, 221–231. [Google Scholar] [CrossRef] [PubMed]
  175. Shin Yim, Y.; Park, A.; Berrios, J.; Lafourcade, M.; Pascual, L.M.; Soares, N.; Yeon Kim, J.; Kim, S.; Kim, H.; Waisman, A.; et al. Reversing behavioural abnormalities in mice exposed to maternal inflammation. Nature 2017, 549, 482–487. [Google Scholar] [CrossRef] [PubMed]
  176. Tsuda, M.C.; Yamaguchi, N.; Ogawa, S. Early life stress disrupts peripubertal development of aggression in male mice. Neuroreport 2011, 22, 259–263. [Google Scholar] [CrossRef] [PubMed]
  177. Rincel, M.; Aubert, P.; Chevalier, J.; Grohard, P.A.; Basso, L.; Monchaux de Oliveira, C.; Helbling, J.C.; Levy, E.; Chevalier, G.; Leboyer, M.; et al. Multi-hit early life adversity affects gut microbiota, brain and behavior in a sex-dependent manner. Brain Behav. Immun. 2019, 80, 179–192. [Google Scholar] [CrossRef] [PubMed]
  178. Foster, J.A.; McVey Neufeld, K.A. Gut-brain axis: How the microbiome influences anxiety and depression. Trends Neurosci. 2013, 36, 305–312. [Google Scholar] [CrossRef]
  179. Foster, J.A.; Rinaman, L.; Cryan, J.F. Stress & the gut-brain axis: Regulation by the microbiome. Neurobiol. Stress 2017, 7, 124–136. [Google Scholar] [CrossRef] [PubMed]
  180. Medina-Rodriguez, E.M.; Cruz, A.A.; De Abreu, J.C.; Beurel, E. Stress, inflammation, microbiome and depression. Pharmacol. Biochem. Behav. 2023, 227–228, 173561. [Google Scholar] [CrossRef]
  181. Sherwin, E.; Dinan, T.G.; Cryan, J.F. Recent developments in understanding the role of the gut microbiota in brain health and disease. Ann. N. Y. Acad. Sci. 2018, 1420, 5–25. [Google Scholar] [CrossRef]
  182. Bailey, M.T.; Dowd, S.E.; Galley, J.D.; Hufnagle, A.R.; Allen, R.G.; Lyte, M. Exposure to a social stressor alters the structure of the intestinal microbiota: Implications for stressor-induced immunomodulation. Brain Behav. Immun. 2011, 25, 397–407. [Google Scholar] [CrossRef] [PubMed]
  183. Bharwani, A.; Mian, M.F.; Foster, J.A.; Surette, M.G.; Bienenstock, J.; Forsythe, P. Structural & functional consequences of chronic psychosocial stress on the microbiome & host. Psychoneuroendocrinology 2016, 63, 217–227. [Google Scholar] [CrossRef] [PubMed]
  184. De Palma, G.; Blennerhassett, P.; Lu, J.; Deng, Y.; Park, A.J.; Green, W.; Denou, E.; Silva, M.A.; Santacruz, A.; Sanz, Y.; et al. Microbiota and host determinants of behavioural phenotype in maternally separated mice. Nat. Commun. 2015, 6, 7735. [Google Scholar] [CrossRef] [PubMed]
  185. Golubeva, A.V.; Crampton, S.; Desbonnet, L.; Edge, D.; O’Sullivan, O.; Lomasney, K.W.; Zhdanov, A.V.; Crispie, F.; Moloney, R.D.; Borre, Y.E.; et al. Prenatal stress-induced alterations in major physiological systems correlate with gut microbiota composition in adulthood. Psychoneuroendocrinology 2015, 60, 58–74. [Google Scholar] [CrossRef] [PubMed]
  186. O’Mahony, S.M.; Marchesi, J.R.; Scully, P.; Codling, C.; Ceolho, A.M.; Quigley, E.M.; Cryan, J.F.; Dinan, T.G. Early life stress alters behavior, immunity, and microbiota in rats: Implications for irritable bowel syndrome and psychiatric illnesses. Biol. Psychiatry 2009, 65, 263–267. [Google Scholar] [CrossRef] [PubMed]
  187. Jasarevic, E.; Howard, C.D.; Misic, A.M.; Beiting, D.P.; Bale, T.L. Stress during pregnancy alters temporal and spatial dynamics of the maternal and offspring microbiome in a sex-specific manner. Sci. Rep. 2017, 7, 44182. [Google Scholar] [CrossRef] [PubMed]
  188. Zhang, Z.; Li, N.; Chen, R.; Lee, T.; Gao, Y.; Yuan, Z.; Nie, Y.; Sun, T. Prenatal stress leads to deficits in brain development, mood related behaviors and gut microbiota in offspring. Neurobiol. Stress 2021, 15, 100333. [Google Scholar] [CrossRef] [PubMed]
  189. Pusceddu, M.M.; El Aidy, S.; Crispie, F.; O’Sullivan, O.; Cotter, P.; Stanton, C.; Kelly, P.; Cryan, J.F.; Dinan, T.G. N-3 Polyunsaturated Fatty Acids (PUFAs) Reverse the Impact of Early-Life Stress on the Gut Microbiota. PLoS ONE 2015, 10, e0139721. [Google Scholar] [CrossRef]
  190. Garcia-Rodenas, C.L.; Bergonzelli, G.E.; Nutten, S.; Schumann, A.; Cherbut, C.; Turini, M.; Ornstein, K.; Rochat, F.; Corthesy-Theulaz, I. Nutritional approach to restore impaired intestinal barrier function and growth after neonatal stress in rats. J. Pediatr. Gastroenterol. Nutr. 2006, 43, 16–24. [Google Scholar] [CrossRef]
  191. Murakami, T.; Kamada, K.; Mizushima, K.; Higashimura, Y.; Katada, K.; Uchiyama, K.; Handa, O.; Takagi, T.; Naito, Y.; Itoh, Y. Changes in Intestinal Motility and Gut Microbiota Composition in a Rat Stress Model. Digestion 2017, 95, 55–60. [Google Scholar] [CrossRef]
  192. Zhou, X.Y.; Li, M.; Li, X.; Long, X.; Zuo, X.L.; Hou, X.H.; Cong, Y.Z.; Li, Y.Q. Visceral hypersensitive rats share common dysbiosis features with irritable bowel syndrome patients. World J. Gastroenterol. 2016, 22, 5211–5227. [Google Scholar] [CrossRef] [PubMed]
  193. Amini-Khoei, H.; Haghani-Samani, E.; Beigi, M.; Soltani, A.; Mobini, G.R.; Balali-Dehkordi, S.; Haj-Mirzaian, A.; Rafieian-Kopaei, M.; Alizadeh, A.; Hojjati, M.R.; et al. On the role of corticosterone in behavioral disorders, microbiota composition alteration and neuroimmune response in adult male mice subjected to maternal separation stress. Int. Immunopharmacol. 2019, 66, 242–250. [Google Scholar] [CrossRef] [PubMed]
  194. Bailey, M.T.; Coe, C.L. Maternal separation disrupts the integrity of the intestinal microflora in infant rhesus monkeys. Dev. Psychobiol. 1999, 35, 146–155. [Google Scholar] [CrossRef]
  195. Moya-Perez, A.; Perez-Villalba, A.; Benitez-Paez, A.; Campillo, I.; Sanz, Y. Bifidobacterium CECT 7765 modulates early stress-induced immune, neuroendocrine and behavioral alterations in mice. Brain Behav. Immun. 2017, 65, 43–56. [Google Scholar] [CrossRef] [PubMed]
  196. O’Mahony, S.M.; Clarke, G.; Dinan, T.G.; Cryan, J.F. Early-life adversity and brain development: Is the microbiome a missing piece of the puzzle? Neuroscience 2017, 342, 37–54. [Google Scholar] [CrossRef] [PubMed]
  197. Arnsten, A.F. Stress signalling pathways that impair prefrontal cortex structure and function. Nat. Rev. Neurosci. 2009, 10, 410–422. [Google Scholar] [CrossRef] [PubMed]
  198. Ulrich-Lai, Y.M.; Herman, J.P. Neural regulation of endocrine and autonomic stress responses. Nat. Rev. Neurosci. 2009, 10, 397–409. [Google Scholar] [CrossRef] [PubMed]
  199. Hoban, A.E.; Stilling, R.M.; Gerard, M.M.; Moloney, R.D.; Shanahan, F.; Dinan, T.G.; Cryan, J.F.; Clarke, G. Microbial regulation of microRNA expression in the amygdala and prefrontal cortex. Microbiome 2017, 5, 102. [Google Scholar] [CrossRef]
  200. Hoban, A.E.; Stilling, R.M.; Ryan, F.J.; Shanahan, F.; Dinan, T.G.; Claesson, M.J.; Clarke, G.; Cryan, J.F. Regulation of prefrontal cortex myelination by the microbiota. Transl. Psychiatry 2016, 6, e774. [Google Scholar] [CrossRef]
Figure 1. Speculative integrative view of the role of early life adversity on the microbiome and the immune response to mediate behaviors. Early life adversity triggers changes in the microbiome and in the immune system. It is likely that cortisol and norepinephrine mediate some of the effects of early life adversity on the microbiome and the immune system. Alteration of the microbiota composition is also associated with a leaky gut, possibly contributing to the activation of immune cells to produce cytokines in the lamina propria. Increased sustained cytokine production, in conjunction with bacterial encounters due to leaky gut, might be responsible for the differentiation of T helper cells (e.g., Th17 cells or Tregs). Th17 cells are known to promote susceptibility to depressive-like behaviors. Furthermore, the constant presence of antigens might contribute to immune senescence and the increased susceptibility to infections in children with early life adversity. These interactions are speculative, and further research is required to fully elucidate the effects of early life adversity on immunity.
Figure 1. Speculative integrative view of the role of early life adversity on the microbiome and the immune response to mediate behaviors. Early life adversity triggers changes in the microbiome and in the immune system. It is likely that cortisol and norepinephrine mediate some of the effects of early life adversity on the microbiome and the immune system. Alteration of the microbiota composition is also associated with a leaky gut, possibly contributing to the activation of immune cells to produce cytokines in the lamina propria. Increased sustained cytokine production, in conjunction with bacterial encounters due to leaky gut, might be responsible for the differentiation of T helper cells (e.g., Th17 cells or Tregs). Th17 cells are known to promote susceptibility to depressive-like behaviors. Furthermore, the constant presence of antigens might contribute to immune senescence and the increased susceptibility to infections in children with early life adversity. These interactions are speculative, and further research is required to fully elucidate the effects of early life adversity on immunity.
Biomolecules 14 00802 g001
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Beurel, E.; Nemeroff, C.B. Early Life Adversity, Microbiome, and Inflammatory Responses. Biomolecules 2024, 14, 802. https://doi.org/10.3390/biom14070802

AMA Style

Beurel E, Nemeroff CB. Early Life Adversity, Microbiome, and Inflammatory Responses. Biomolecules. 2024; 14(7):802. https://doi.org/10.3390/biom14070802

Chicago/Turabian Style

Beurel, Eléonore, and Charles B. Nemeroff. 2024. "Early Life Adversity, Microbiome, and Inflammatory Responses" Biomolecules 14, no. 7: 802. https://doi.org/10.3390/biom14070802

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop