Open access peer-reviewed chapter

Epigenetic

Written By

Mehmet Ünal

Submitted: 07 November 2020 Reviewed: 18 August 2021 Published: 03 September 2021

DOI: 10.5772/intechopen.99964

From the Edited Volume

Epigenetics to Optogenetics - A New Paradigm in the Study of Biology

Edited by Mumtaz Anwar, Zeenat Farooq, Riyaz Ahmad Rather, Mohammad Tauseef and Thomas Heinbockel

Chapter metrics overview

349 Chapter Downloads

View Full Metrics

Abstract

Lately, a brand-new studies agenda emphasizing interactions between societal elements and wellness has emerged. The phrase social determinant of health and fitness typically refers to any nonmedical element directly effecting health, including behaviors, knowledge, attitudes, and values. Status of health is adversely and strongly impacted throughout the life span by social disadvantages. Epigenetic mechanisms are implicated in the processes through which social stressors erode health in humans and other animals. Research in epigenetics suggests that alterations in DNA methylation might offer a temporary link between interpersonal adversity and wellness disparity. Likewise, accelerated loss in telomeres is extremely correlated not only with chronic and social stress but also aging. Therefore, it may provide a link between the various physiological events associated with health inequalities. Research in epigenetics indicates that alterations in DNA methylation may provide a causal link between social adversity and health disparity. Additionally, these experimental paradigms have yielded insights into the potential role of epigenetic mechanisms in mediating the effects of the environment on human development and indicate that consideration of the sensitivity of laboratory animals to environmental cues may be an important factor in predicting long-term health and welfare.

Keywords

  • stress
  • epigenetic
  • DNA
  • social
  • environment

1. Introduction

Absence of early life stress has great effects on both health and well-being. This particular topic focuses on the availability or quality of food sources, exposure to toxic effects, community-based events, and the presence of stressors or threats in the ecosystem. In humans, years of extensive researches have revealed some sort of correlation among variations within being exposed in early life era and long-lasting risks of psychiatric and physical illness of maturity [1].

While it is expected that the effects of exposure to chemical substances besides deprivation or stress hormones of a vital vitamin or energy supply, they might have biological impacts on human depending on the specification of the exposure and availability. Personal and social experiences carry the partly same features with biochemical, cellular, and neurobiological changes and this case is amplified with psychosocial expertise executed on animal models in the light of finding answers to these questions.

Effects of social experiences and aggravating events taken place in life leads to the experimental study which has mainly focused on types of lab rodents (usually including mice and rats), albeit several numbers of primate studies can be obtained to maintain additional data in the interest of powerful impacts of encounters taken place in the initial stages of life [2, 3]. In these designs, impact of prenatal nervousness, absence of a mother role and also disengagement variance in maternal treatment, adolescent cultural enhancement, isolation and mature cultural anxiety. When these have been investigated, results indicate that the quality of emotional stress experience or social experiences may cause neuroendocrine consequences which effects social and reproductive behavior. While it is the evidence of there is a phase of increased sensitivity to these eco-induced factors during prenatal as well as first postnatal development, there may also be plasticity that extends into adulthood and adolescence after infancy. The long-term negative impact of the first latter life events on human brain’s region-specific gene expression part would be an important finding within these studies. Substantial evidence supports the existence of longterm effects on HPA axis functioning following early life stress; these effects persist into adulthood and are accompanied by lasting behavioral changes. In clinical studies, early life stress has been shown to be a strong predictor of ACTH responsiveness [4, 5]. Further analysis of the molecular mechanisms that could mediate this long-lasting effect involved in gene regulation have been led by these findings.

Epigenetic processes are effective events in changing gene expression and the long-term effects of events experienced in the early stages of life on gene expression are the subject of biologically based research [6]. Across species, it is obvious that a selection of experiences, such as the aspects of interactions experienced socially as well as being exposed to the stressors, can trigger epigenetic consequences. In addition, these progressive outcomes may be carried over descendants in certain cases, resulting in behavioral and neurobiological disturbance of the offspring and even of the grand offspring [7, 8].

Advertisement

2. Epigenetic effects of stress experienced in early life

The crucial role of epigenetic machinery in the biological embedding of stressful exposures in early life has been demonstrated in a number of rodent models, where considerable variations in both DNA methylation and histone modification have been reported in offspring exposed to different prenatal stresses, inappropriate maternal care, maternal deprivation/separation, as well as to juvenile social enrichment/isolation [9, 10, 11]. It was found out by the rodent tests that mothers’ offspring with comparatively flat postnatal maternal hygiene grades increased fearfulness, anxiety, and stress-reactivity compared to the mothers’ offspring with caregiving of normal PH grades [12]. These behavioral anomalies were followed by diminished exposition of the glucocorticoid receptor (GR) encoding NR3C1 gene within the hippocampus along with increased methylation of CpG dinucleotides in the NR3C1 promoter and additionally great levels of activation of the hypothalamic–pituitary–adrenal (HPA) axis and serum glucocorticoids [13, 14, 15]. Maternally deprived offspring developed irritability, anxiety, depressive symptoms, interruption of socially received interactions, genome-wide changes and high reactivity of DNA methylation transcription [16, 17, 18].

Depriving the organization and maternal attention of some other family members of juvenile rhesus macaques and infants also induced stress and depression related symptoms followed by protracted activation of the HPA axis. Consecutively, changed genome-wide modifications and gene transcription to DNA methylation patterns are within the mental faculties and in the peripheral T lymphocyte [19, 20, 21, 22, 23, 24]. The amygdala, hippocampus, HPA axis, and medial prefrontal cortex form the areas affected by reduced maternal care in rodents [25]. Within the very first week of postnatal existence, maltreatment of mothers breastfeeding new-born children has shown to cause frequent vocalized distress calls in offspring and shifts in patterns of methylcytosine last long and its hydroxymethyl cytosine derivative at many spots such as the BDNF locus, in the amygdala, hippocampus, and in exposed offspring’s medial prefrontal cortex, all at the time of exposure [26, 27, 28]. Early-life stress as in maternal separation-induced in the paraventricular nucleus of the hypothalamus increased AVP and POMPC expression and decreased DNA methylation at these loci [29]. Experienced early-life stress increased NR3CR1 transcription in the paraventricular nucleus and rendered CRH transcription refractory to maturity chronical stress after stress experienced in early life, suggesting that it jeopardizes CRH transcriptional responsiveness in the hypothalamus to later chronic stressors, probably through a GR-dependent mechanism [30].

The long-lasting effects of prenatal maternal pressure on the neural genes as well as offspring behavioral word was showed more by a research analyzing the effects on adult offspring of prenatal exposure to predator odor during fetal gestation. In offspring of whose mothers suffered being exposed to the odor of predator in the time of pregnancy and in female (yet no male) offspring, stated endocrine and behavioral modifications were followed by BDNF’s diminished expression and improved DNA methylation of BDNF promoter sequences of the hippocampus, as well as bb promoter sequences of the hippocampus, odor avoidance, addition to corticosterone production as a response to being exposed to the odor, were enhanced. Mentioned studies show that not only prenatal but also postnatal stressors may result in long-lasting alterations in epigenetic control, neuroendocrine function, and neural gene transcription activity that continuing through adulthood [31].

Research one on humans have advanced and expanded the results of animal studies on the effects of early-life strains [32]. So, proof states that adolescence, infancy, and fetal gestation are actually delicate stages during which epigenetic, psychological, and behavioral changes continue through adulthood which may be caused by exposing to cultural adversity. A recent systematic review reported, forty studies which were betwixt 2004 and 2014, that listed NR3C1 methylation changes in response to early life adversity, parental anxiety, and psychopathology studies, of which twenty-seven were human studies [33]. While in these papers a variety of different NR3C1 sequences are actually involved as reglementary targets, perhaps the most stable finding is actually a highly related expansion in exon 1F methylation in the NR3C1 gene of the individual (or maybe the analogous exon seventeen in the thirteen animal studies) as well as early life adversity experience. Exon 1F/17 has a portion of the DNA sequence encoding a methylation-sensitive binding website for the NGF1A/EGR1 [34] controlled transcription activator for neural activity. The decreased NR3C1 expression caused by increased methylation of this unique binding website, consecutively decreases the means of providing bad feedback to the hypothalamus and pituitary mediated by glucocorticoid, resulting in continuous activation of the HPA axis, as well as the ensuing disorders. Improved NR3C1 promoter methylation has been associated with a variety of experiences highly appropriate for inducing prenatal tension, such as near partner aggression, or perhaps maternal exposure to genocidal war [35, 36]. In addition, neglect, being abused in childhood, and destitute were also concluded to be related with enhanced methylation of the NR3C1 promoter [34, 37, 38, 39]. Recently prepared reports give further backing for a correlation among early life adversity, improved methylation of the promoter, and reduced NR3C1 transcription [38, 40, 41, 42, 43, 44]. These mixed scientific studies indicate that as a consequence of allostatic overload caused by a range of stressful interactions, attenuation of the NR3C1 phrase is actually an aspect of the task leading to elevation of HPA axis operation. Despite such insights, it remains unclear how NR3C1 is specifically targeted for epigenetic silencing, and whether methylation of the NGF1A/EGR1 binding site, which prevents NGF1A/EGR1 recruitment to NR3C1, is accompanied by other changes that attenuate NR3C1 expression in the brain. As inhibitors of NR3C1 protein run [45, 46], noncoding RNAs such as the lncRNA GAS5 have been involved, as well as miRNAs such as MIR 124 could as well control the durability of NR3C1 transcripts [47, 48]. Though, several different early life adversity response studies including DNA methylation changes linked to several extra-human genes, such as SLC17A3, PM20D1, KITLG, SLC6A4, BDNF, MORC1, LGI1/LGI2, FKBP5, CRHBP, CRH, and MAOA [49, 50, 51, 52, 53, 54]. Additional studies to explain the purposeful interrelationships of genes with NR3C1, along with a greater comprehension of exactly how these genes are actually controlled, possibly make it possible to reveal precisely how prenatal stress, childhood neglect, and parental hygiene deprivation are biologically embedded and have long-lasting effects that dwell on across. In addition, an in-depth study of the procedures may explicate the biological base of resistance to anxiety as well as provide an evidence base for successful interventions.

Advertisement

3. Epigenetic plasticity in adulthood as well as adolescence

While plasticity carrying epigenetic pathways was originally considered to be confined for the premature steps of embryogenesis, now it is becoming more and more apparent to that epigenetic variation can be caused by experiences occurring during the lifespan. In addition, a key facet of memory and learning from infancy to adulthood may be the guiding source to modify DNA methylation and histone tails [55]. Variation of epigenetic, similarly, has been related with the alterations in phenotype and gene expression in the mean time of the latter stages of advancement in the form of studies on the effect of social experiences & stressors. In addition to adult mice with genetically mediated memory dysfunction, being exposed to complicated housing environments for four weeks were found to be associated with enhanced hippocampal histone acetylation as well as memory enhancement and cortex [56]. Surprisingly, these enrichment-induced effects on histones, memory and comprehending, can as well be obtained with pharmacological remedies in non-memory-impaired mice that promote histone acetylation. Histone modifications (particularly histone methylation) are also observed in mice in enriched settings in the BDNF III, IV, and VI promoter regions [57].

Chronic stress is actually linked to decreases in the expression of this specific gene compared to environmental enrichment, which has been shown to advance stages of BDNF, an epigenetic base might be an issue for such consequences. In rats, immobilization pressure was found to cause great rises in hippocampal DNA methylation in the BDNF gene, coupled with exposure to predator odor [58]. Reductions in BDNF are actually found within the version of social defeat after a month of being exposed to what is known as social stressor, then hippocampal histone demethylation at the promoter of BDNF might take credit for such specific impact [59]. Histone acetylation is temporarily diminished, after that shows elongated risings on mice that is rejected socially. Therefore, such specific impact may be related with long-lasting reductions in histone deacetylase amounts induced by stress [60]. Similarly, increased histone acetylation in rats continues to be observed for up to twenty-four hours following the experience of repeated population-based rejection [61]. In addition, the behavior-based outcomes of interpersonal loss, such as diminished social-based activity, can be pharmacologically turned around thanks to a medication that inhibits histone deacetylases [60]. It is concluded that a group of mice were resistance to our stressor. And the other conclusion is that discomfiture between humans results in consequences which would last long. Stress-susceptible mice were found in a recent study to have elevated levels of CRF mRNA in the PVN and also diminished methylation of DNA in the CRF gene [62]. In comparison, stress-resilient mice were found to undergo no changes in the CRF mRNA or perhaps DNA methylation of this particular gene. In fact, differential sensitivity to the effects of stress is a key thing to consider in these studies, and as a consequence of anxiety, there is growing evidence for strain or perhaps genotype-specific epigenetic consequences [63].

Advertisement

4. Transgenerational effect of the social stress and environment

The continuous epigenetic effects of environmental events were presented in the previous sections. In influencing behavioral and neurobiological effects, these consequences seem to play a crucial role. However, it may also be the case that these eco-induced influences are capable of persisting over centuries. One mechanism by which this transmission occurs [64] may be transgenerational continuity in maternal behavior. There is evidence in rodents that coercive caregiving as well as variance in maternal LG can change the enhancement of female offspring so that each offspring as well as grand offspring can also find these maternal characteristics. In the medial preoptic position of the hypothalamus of female offspring, maternal LG tends to alter the DNA methylation as well as the Esr1 phrase [65]. Throughout the postnatal period, during which adulthood continues, these implications emerge. ER-alpha quantities possess a crucial part in deciding the responsiveness of females to circulating oestrogens for late gestation, and stated specific responsiveness predicts the quality as well as the number of interactions between postnatal mother babies. Individual differences in maternal LG are actually transferred from mother to offspring (generation F1) as well as to grand offspring (generation F2) as a result of the epigenetic changes [64, 66].

In studies of child violence, variance of behavioral segmentations is found across species. In rats, both the variation of DNA methylation and the BDNF phrase in the prefrontal cortex can be correlated with the transgenerational continuity of violence. Females’ BDNF term tends to be decreased if experienced violence in childhood and methylation of BDNF IV promoter DNA in the prefrontal cortex is to be improved by this situation as well. The offspring of these females likewise have increased Bdnf IV promoter DNA methylation in the prefrontal cortex. Addition to these, offspring of stated females is likely to have enhanced methylation of BDNF IV promoter DNA in the prefrontal cortex [67]. Surprisingly, scientific studies on cross-fostering recommend that prenatal elements may be linked to the transmission of violence and also the epigenetic variance related to this specific phenotype rather than postnatal events with the dam. In fact, the direct inheritance of epigenetic change is another path by which perinatal epigenetic, as well as behavioral effects, may persist over generations. Although it has long been believed that during the first stages of embryogenesis, a complete erasure of epigenetic disruption to the genome is at hand. Revelation of imprinted genes (genes that are expressed depending on the parent from whom they are inherited) has led to widely believed speculation that the previous generation’s epigenetic “memory” is actually retained and transmitted to the genome [68].

The epigenetic inheritance of the toxicological exposure effect has been studied and also suggests the existence of the patriline effects for many decades [69]. More recently, in offspring exposed to separation (F1), the offspring of separated males (F2), and the grand offspring of separated males (F3), the transgenerational impact of maternal separation have been investigated. With this paradigm, during the postnatal era, mice were exposed to unforeseen separation from the dam. Depressive-like behaviors that were activated in the offspring of the F1 model were found to be contained in both F2 and F3 generation mice [70]. DNA methylation patterns caused by separation have been shown to be contained in the sperm and brains of F1 male mice and also in the brains of F2 males. Hypermethylation of the Mecp2 gene as well as hypomethylation of the Crfr2 gene resulted in these epigenetic consequences. As a result, it appears that the epigenetic variation that is created with the early life environment’s aspect could be encoded within the germ cells, leaving traces of stress and social environment on the next descendants through transgenerational approach.

Advertisement

5. Human health’s social-based determinants

A distinguishing figure in inequal cultures is the fact that in neighborhoods and neighborhoods with lower socioeconomic mobility (SES) [71, 72], community problems and persistent wellbeing appear to be more prevalent. In lower SES populations, the risks of illnesses such as cardiovascular disease, stroke, diabetes, obesity, and mental conditions are highest. It as well needs to be remembered, however, that the prevalence of such chronic, non-communicable diseases is actually classified across social classes, with the lowest incidence in higher SES categories. Expectancy of life across social classes is equally ranked, being higher in high SES categories. The prospect of designing evidence-based health and policy that covers social subjects strategies is to elucidate the biological and social roots of the social gradient in health that could increase well-being and health for everyone while providing additional benefit to those with higher needs. Good knowledge of the biochemical mechanisms by which wellbeing is impacted by the social gradient will also help to identify relevant intervention or mitigation goals and sustain biomarkers for which to track effects.

Salary inequality can be seen as sensitive, quantitative relative place measures among a larger hierarchy of socioeconomic status that indicate disparities in access to, along with economic resources, a number of forms of social, educational and cultural capital [73, 74, 75]. It has been speculated that social rank is essentially a result of the amount of entrance obtained to stated and differentiated forms of resources in extremely unequal environments that competing social experiences for such access are actually mental stressors that can contribute to uncertainty about ranking. Many scientific researches confirm the theory of status anxiety, connecting not so high social expectations to nervousness, guilt, depression and harming oneself [76, 77, 78, 79]. In addition, status-based nervousness and its not only mental but also cognitive outcomes are potentially possible contributors to some kinds of social adversity, childhood era deprivation for example, limited autonomy on making decisions which even consider matter of life events, reduced social connectivity, and decreased levels of interest in certain different parts and members of the community. Such theories align with the findings of hierarchies of animal domination, yet in which situation that dominant individuals are literally prohibited from ensuring preferential entrance to group services provided in scarce supplies, such as meals, water, accommodation, as well as companions, and access to subordinates. As previously stated, the hierarchy of dominance is currently an evolutionarily and strongly maintained form of social organization. Undoubtedly, social rank understanding evolves very quickly in members of humanity, as it is a prevalent conception for infants [hundred] and is used by children from two years of age to create relationships of dominance [80, 81].

As potential causes of chronic behavioral tension and allostatic load, inducing hypercortisolaemia and improved levels of inflammatory biomarkers in the blood, the pervasive experiences of status tournaments, in which rank is actually guided and controlled by oneself and others, have been labeled [82, 83]. Moreover, epidemiological data suggest that low SES increases allostatic load and increases blood inflammatory biomarkers [84, 85]. Therefore, the tendencies of too many social challenges to erode group harmony, degrade social networking sites and impede mutual assistance, such as persistently competitive activities and violence, may place restrictions on social practices that decide the fundamental characteristics of human well-being.

Emerging research recognizes social networking platforms as critical factors in health security [86, 87, 88, 89, 90] and supports the theory that loss of social capital accentuates the very poor health outcomes of low SES communities by weakening or even diminishing social networks. In order to minimize allostatic burden and buffer the immune system against inflammatory stimuli associated with very low SES [91, 92, 93, 94], increased parental assistance has been confirmed. Close comparisons can be drawn between the mechanisms of action of animal and human social buffering treatments, general themes of which include reducing the function of the HPA axis, attenuating inflammation and increasing the development of oxytocin [95]. Via enhancing parenting skills, strengthening family relationships, or even developing capacities for young people, social interventions that promote group buffering have all been found to decrease pro-inflammatory biomarkers, indicating a preventive impact, while some aspects of resilience-building may be much more durable relative to others. Additional analyses of these and other human cohorts would enable the biological mechanisms and psychosocial processes by which such strategies accomplish their buffering effects to be explained in greater detail [93, 94].

Advertisement

6. The epigenetic effects of persistent interpersonal stress in humane communities

In particular being long-lasting exposed to the social-based stressors associated with low SES over the course of life are recognized to impact the likelihood of chronical illness by using their effects on a broad variety of physiological processes affecting the nervous, hemopoietic, cardiovascular, and endocrine systems. Most documented studies of the effects of SES on the epigenome showed that DNA methylation patterns in samples taken from tissues collected, such as whole blood, fractionated white blood cells, or perhaps buccal swabs were altered. Numerous scientific works indicate that SES is actually connected to differences in patterns of genomic DNA methylation [96, 97]. Childhood SES was shown to be specifically correlated with differential methylation of 1252 gene promoters in forty individuals from the 1957 British Birth Cohort in one of several experiments using promoter microarrays so that increased methylation was related with lower childhood SES for 586 promoters and superior childhood SES for the remaining 666 promoters [96]. A review of 239 participants of the Glasgow pSoBid cohort, which indicates a particularly steep social gradient in wellbeing, found with an alternative technique of DNA methylation analysis that average DNA methylation levels across the entire genome were more or less seventeen percent lower in the most deprived group than in perhaps the least deprived group [98]. It remains to be clarified if this low SES-related hypomethylation of whole genomic DNA is specifically directed at gene bodies, cis-regulatory elements, intergenic areas, and/or repetitive DNA sequences. Recent studies have reported that genes whose transcription is actually involved in neuroendocrine and inflammatory responses to low SES also show epigenetic SES sensitive modifications, which are actually in line with previous studies involving process dysregulation in low SES individuals [99, 100].

In more unequal environments, possibility of mental wellness problems such as nervousness, alcohol, and anxiety are actually much higher. Consecutively, these disorders are certainly easier to come across in low-SES communities [101]. Predictive connections between lower SES, differential methylation of the SLC6A4 serotonin transporter gene promoter, greater amygdala activity, as well as signs of depression have been reported in a recently published analysis [102]. Previously, differential SLC6A4 methylation has been shown to be exclusively linked to molestation of child [103], low SES [104], stress-related depression [105], as well as enhanced amygdala reactivity to frightening stimuli. In addition, increased fear reactivity of the amygdala in puberty has been shown to become a likely biomarker indicative of adult stress and tension [106, 107, 108]. These findings were expanded to include a possible analysis of depression development within a cohort of teenagers tested on 3 occasions at 11–15,13–18 and 14–19 years of age, in order [102]. Using blood and saliva samples for DNA methylation analysis, low SES at age 11–15 years was discovered to be predictive of increased methylation of SLC6A4 at age 13–18 years, which was predictive of improved amygdala reactivity to a fearful stimulus (detected by fMRI) over exactly the same time, and which subsequently was connected with an elevated threat of depression between ages of 14–19 for adolescents with an optimistic depression-based family history. Such outcomes, therefore, propose a plausible biological path through which low SES, by methylation of SLC6A4, may attenuate expression of the serotonin transporter encoded by that stated particular gene, forwarding to elevations not only in amygdala reactivity but also in liability of depression. Additionally, such indications pinpoint possible biomarkers for building and evaluating preventive or maybe treatment-based interventions that may buffer the effects of low SES on liability of depression in the means of lifetime.

Advertisement

7. Post-traumatic stress and anxiety

The intense stress caused by imposed to stressful experiences, battle for example, genocide, starvation, elevates the liability of psychiatric wellbeing problems, including PTSD, schizophrenia, depression, even killing oneself [36, 37]. Similarly, US fighting veterans with an analysis of moderate PTSD showed hypomethylation on the regulatory portion of the NR3C1 promoter 1F and decreased HPA axis actions [109]. As a result, in the meantime NR3C1 is a very typical target for epigenetic modifications as a response upon these distinctive stress forms, in addition to qualitative discrepancies in the mechanisms as well as the context of traumatic events themselves, the particular variations within the patterns of NR3C1 methylation could represent disparities in the timings as well as times of coverage for trauma.

Traumatic stress and PTSD-associated variance in DNA methylation at the SKA2 locus have been reported in recently made studies on epigenetic alterations among war vets suffering extreme PTSD [110, 111]. SKA2 encodes a protein that is rather to serve as a chaperone or possibly a GR activity regulator, allowing negative feedback to the HPA axis mediated by cortisol dependent GR. SKA2 was recognized as a hypermethylated, under expressed locus of suicide completers in post-mortem cortical tissue, and variance in SKA methylation was also correlated with suicidal activities in PTSD individuals [112, 113]. Although the discovery of altered DNA methylation taken from those who tried to commit suicide and who suffers PTSD at this locus in tissue indicates possible functions for SKA2 in the control of traumatic stress response, it is presently uncertain if these modifications are directly linked to the multiple psychopathological behaviors under investigation.

In intergenerational epigenetic responses to stress, research on the effects of PTSD suffered by those who survived Holocaust through their offspring includes modified HPA axis actions, and even more especially dysregulation of the GR along with its auxiliary components. In a similar study, methylation with a CpG dinucleotide inside a GR binding website inserted within an intron of this gene encoding the GR regulator FKBP5 was shown to be increased within the blood cells of Holocaust survivors, as well as lower in their offspring, relative to the quantity of this gene encoding the GR regulator FKBP5 [114]. These findings show that intergenerational transfer of trauma-related DNA methylation modifications has taken place between Holocaust survivors and descendants of theirs in at least a variety of cases. In response to a new, moderately demanding exposure, diminished behavioral regulation was recognized as diminished latency to type in unknown places, indicative of probably elevated resistance or impulsivity. Additionally, DNA methylation switches at candidate gene loci transmitted via the germline along with mediated, transmitted behavioral anomalies from the MSUS. The applicant genes include Nr3c1, which demonstrated decreased methylation of the promoter and increased transcription of this progeny of MSUS treated mice in the hippocampus. In addition, traumatized men’s sperm contained microinjection and trauma-induced miRNAs of distilled RNA derived from their traumatized adult men’s sperm recapitulated MSUS-induced behavioral abnormalities, suggesting functions within the intergenerational transmission of trauma-induced phenotypes for these very short noncoding RNAs. Interestingly, the intergenerational transfer of MSUS mediated altered behavioral reactions to moderately challenging stimuli, increased Nr3c1 methylation, and decreased transcription of this specific gene within the hippocampus was improved by environmental enrichment. In a related study, corticosterone administration to adult male mice caused behavioral phenotypes that indicated hyper nervousness within their male F1 progeny, as well as reduced anxiety levels, but elevated depressed characteristics within their F2 progeny. In addition, both the F1 and F2 phenotypes have been identified with the expression within the paternal sperm of some corticosterone mediated miRNAs. Taken together, these animal experiments suggest that ameliorative and negative interactions modulate behavior, the effects of which could be delivered among one descendant to the others, possibly by epigenetic modulation of neuroendocrine reaction systems involving miRNAs [115].

Advertisement

8. Conclusion

The pervasive influence of social stressors on well-being and health are recorded in detailed literature. Emerging data shows that the biological embedding of psychosocial messages throughout the body potentially includes epigenetic pathways, influencing cellular mechanisms that could influence health hazards over the life cycle. Many vertebrates display social activities that represent similar operating concepts of hierarchical domination, like teleost fish, non-human primates, and humans, and elicit stress responses that are actually followed by impacts on the epigenome. In hierarchies, social differences can act as persistent behavioral stressors and limit access to services, leading to a social gradient in health and expectancy of life in humans. Epigenetic changes are triggered by a broad scope of behavior-based stressors, traumatically suffered experiences included which present promise as biomarkers of disease risk and can likely be reversed by intervference to lessen the health-based effects of distress.

Advertisement

Conflict of interest

The authors state no conflict of interest.

References

  1. 1. Tomalski P, Johnson MH. The effects of early adversity on the adult and developing brain. Curr Opin Psychiatry. 2010;23(3):233-238
  2. 2. Harlow HF, Dodsworth RO, Harlow MK. "Total social isolation in monkeys." Proceedings of the National Academy of Sciences of the United States of America 54.1 (1965): 90
  3. 3. Suomi SJ, et al. Effects of maternal and peer separations on young monkeys. J Child Psychol Psychiatry. 1976;17(2):101-112
  4. 4. Ladd CO, et al. Long-term adaptations in glucocorticoid receptor and mineralocorticoid receptor mRNA and negative feedback on the hypothalamo-pituitary-adrenal axis following neonatal maternal separation. Biol Psychiatry. 2004;55(4):367-375
  5. 5. Maccari, Stefania, et al. "Prenatal stress and long-term consequences: implications of glucocorticoid hormones." Neuroscience & Biobehavioral Reviews 27.1-2 (2003): 119-127
  6. 6. Curley JP, et al. Social influences on neurobiology and behavior: epigenetic effects during development. Psychoneuroendocrinology. 2011;36(3):352-371
  7. 7. Champagne FA. Epigenetic mechanisms and the transgenerational effects of maternal care. Front Neuroendocrinol. 2008;29(3):386-397
  8. 8. Franklin TB, et al. Epigenetic transmission of the impact of early stress across generations. Biol Psychiatry. 2010;68(5):408-415
  9. 9. Anacker, Christoph, Kieran J. O'Donnell, and Michael J. Meaney. "Early life adversity and the epigenetic programming of hypothalamic-pituitary-adrenal function." Dialogues in clinical neuroscience 16.3 (2014): 321
  10. 10. Maccari S, et al. The consequences of early-life adversity: neurobiological, behavioural and epigenetic adaptations. J Neuroendocrinol. 2014;26(10):707-723
  11. 11. Kundakovic M, Champagne FA. Early-life experience, epigenetics, and the developing brain. Neuropsychopharmacology. 2015;40(1):141-153
  12. 12. Weaver IC, Cervoni N, Champagne FA, et al. Epigenetic programming by maternal behavior. Nat Neurosci. 2004;7(8):847-854
  13. 13. Weaver IC, Meaney MJ, Szyf M. Maternal care effects on the hippocampal transcriptome and anxiety-mediated behaviors in the offspring that are reversible in adulthood. Proc Natl Acad Sci USA. 2006;103(9):3480-3485
  14. 14. McGowan PO, Suderman M, Sasaki A, et al. Broad epigenetic signature of maternal care in the brain of adult rats. PLoS One. 2011;6(2):e14739
  15. 15. Suderman M, McGowan PO, Sasaki A, et al. Conserved epigenetic sensitivity to early life experience in the rat and human hippocampus. Proc Natl Acad Sci USA. 2012;109 Suppl. 2:17266-17272
  16. 16. Murgatroyd C, Patchev AV, Wu Y, et al. Dynamic DNA methylation programs persistent adverse effects of early-life stress. Nat Neurosci. 2009;(12):1559-1566
  17. 17. Franklin TB, Russig H, Weiss IC, et al. Epigenetic transmission of the impact of early stress across generations. Biol Psychiatry. 2010;68(5):408-415
  18. 18. Franklin TB, Saab BJ, Mansuy IM. Neural mechanisms of stress resilience and vulnerability. Neuron. 2012;75(5):747-761
  19. 19. Provencal N, Suderman MJ, Guillemin C, et al. The signature of maternal rearing in the methylome in rhesus macaque prefrontal cortex and T cells. J Neurosci. 2012;32(44):15626-15642
  20. 20. Cole SW, Conti G, Arevalo JM, Ruggiero AM, Heckman JJ, Suomi SJ. Transcriptional modulation of the developing immune system by early life social adversity. Proc Natl Acad Sci USA. 2012;109(50):20578-20583
  21. 21. Suomi SJ. Risk, resilience, and gene-environment interplay in primates. J Can Acad Child Adolesc Psychiatry. 2011;20(4):289-297
  22. 22. Dettmer AM, Suomi SJ. Nonhuman primate models of neuropsychiatric disorders: influences of early rearing, genetics, and epigenetics. ILAR J. 2014;55(2):361-370
  23. 23. Kinnally EL. Epigenetic plasticity following early stress predicts long-term health outcomes in rhesus macaques. Am J Phys Anthropol. 2014;155(2):192-199
  24. 24. Nieratschker V, Massart R, Gilles M, et al. MORC1 exhibits cross-species differential methylation in association with early life stress as well as genome-wide association with MDD. Transl Psychiatry. 2014;4:e429
  25. 25. McEwen BS, Nasca C, Gray JD. Stress effects on neuronal structure: hippocampus, amygdala, and prefrontal cortex. Neuropsychopharmacology. 2016;41(1):3-23
  26. 26. Blaze J, Scheuing L, Roth TL. Differential methylation of genes in the medial prefrontal cortex of developing and adult rats following exposure to maltreatment or nurturing care during infancy. Dev Neurosci. 2013;35(4):306-316
  27. 27. Roth TL, Matt S, Chen K, Blaze J. Bdnf DNA methylation modifications in the hippocampus and amygdala of male and female rats exposed to different caregiving environments outside the homecage. Dev Psychobiol. 2014;56(8):1755-1763
  28. 28. Doherty TS, Forster A, Roth TL. Global and gene-specific DNA methylation alterations in the adolescent amygdala and hippocampus in an animal model of caregiver maltreatment. Behav. Brain Res. 298(Pt A), 55-61 (2016)
  29. 29. Wu Y, Patchev AV, Daniel G, Almeida OF, Spengler D. Early-life stress reduces DNA methylation of the Pomc gene in male mice. Endocrinology. 2014;155(5):1751-1762
  30. 30. Bockmuhl Y, Patchev AV, Madejska A, et al. Methylation at the CpG island shore region upregulates Nr3c1 promoter activity after early-life stress. Epigenetics. 2015;10(3):247-257
  31. 31. St-Cyr S, McGowan PO. Programming of stress-related behavior and epigenetic neural gene regulation in mice offspring through maternal exposure to predator odor. Front. Behav. Neurosci. 9, 145 (2015). 27 Andersen SL. Exposure to early adversity: points of crossspecies translation that can lead to improved understanding of depression. Dev. Psychopathol.(2), 477-491 (2015)
  32. 32. Turecki G, Meaney MJ. Effects of the social environment and stress on glucocorticoid receptor gene methylation: a systematic review. Biol Psychiatry. 2016;79(2):87-96
  33. 33. A comprehensive review of the literature describing experience-dependent changes in DNA methylation patterns at the NR3C1 locus in humans and model organisms
  34. 34. McGowan PO, Sasaki A, D’Alessio AC, et al. Epigenetic regulation of the glucocorticoid receptor in human brain associates with childhood abuse. Nat Neurosci. 2009;12(3):342-348
  35. 35. Radtke KM, Ruf M, Gunter HM, et al. Transgenerational impact of intimate partner violence on methylation in the promoter of the glucocorticoid receptor. Transl Psychiatry. 2011;1:e21
  36. 36. Mulligan CJ, D’Errico NC, Stees J, Hughes DA. Methylation changes at NR3C1 in newborns associate with maternal prenatal stress exposure and newborn birth weight. Epigenetics. 2012;7(8):853-857
  37. 37. Labonte B, Yerko V, Gross J, et al. Differential glucocorticoid receptor exon 1(B), 1(C), and 1(H) expression and methylation in suicide completers with a history of childhood abuse. Biol Psychiatry. 2012;72(1):41-48
  38. 38. Tyrka AR, Parade SH, Eslinger NM, et al. Methylation of exons 1D, 1F, and 1H of the glucocorticoid receptor gene promoter and exposure to adversity in preschool-aged children. Dev Psychopathol. 2015;27(2):577-585
  39. 39. Weder N, Zhang H, Jensen K, et al. Child abuse, depression, and methylation in genes involved with stress, neural plasticity, and brain circuitry. J Am Acad Child Adolesc Psychiatry. 2014;53(4):417-424.e415
  40. 40. van der Knaap LJ, Riese H, Hudziak JJ, et al. Glucocorticoid receptor gene (NR3C1) methylation following stressful events between birth and adolescence. The TRAILS study. Transl Psychiatry. 2014;4:e381
  41. 41. Braithwaite EC, Kundakovic M, Ramchandani PG, Murphy SE, Champagne FA. Maternal prenatal depressive symptoms predict infant NR3C1 1F and BDNF IV DNA methylation. Epigenetics. 2015;10(5):408-417
  42. 42. Murgatroyd C, Quinn JP, Sharp HM, Pickles A, Hill J. Effects of prenatal and postnatal depression, and maternal stroking, at the glucocorticoid receptor gene. Transl Psychiatry. 2015;5:e560
  43. 43. Radtke KM, Schauer M, Gunter HM, et al. Epigenetic modifications of the glucocorticoid receptor gene are associated with the vulnerability to psychopathology in childhood maltreatment. Transl Psychiatry. 2015;5:e571
  44. 44. Kertes DA, Kamin HS, Hughes DA, Rodney NC, Bhatt S, Mulligan CJ. Prenatal maternal stress predicts methylation of genes regulating the hypothalamic–pituitary–adrenocortical system in mothers and newborns in the Democratic Republic of Congo. Child Dev. 2016;87(1):61-72
  45. 45. Kino T, Hurt DE, Ichijo T, Nader N, Chrousos GP. Noncoding RNA gas5 is a growth arrest- and starvationassociated repressor of the glucocorticoid receptor. Sci Signal. 2010;3(107):ra8
  46. 46. Hudson WH, Pickard MR, de Vera IM, et al. Conserved sequence-specific lincRNA-steroid receptor interactions drive transcriptional repression and direct cell fate. Nat Commun. 2014;5:5395
  47. 47. Vreugdenhil E, Verissimo CS, Mariman R, et al. MicroRNA 18 and 124a down-regulate the glucocorticoid receptor: implications for glucocorticoid responsiveness in the brain. Endocrinology. 2009;150(5):2220-2228
  48. 48. Pan-Vazquez A, Rye N, Ameri M, et al. Impact of voluntary exercise and housing conditions on hippocampal glucocorticoid receptor, miR-124 and anxiety. Mol Brain. 2015;8:40
  49. 49. Melas PA, Wei Y, Wong CC, et al. Genetic and epigenetic associations of MAOA and NR3C1 with depression and childhood adversities. Int J Neuropsychopharmacol. 2013;16(7):1513-1528
  50. 50. Cao-Lei L, Massart R, Suderman MJ, et al. DNA methylation signatures triggered by prenatal maternal stress exposure to a natural disaster: Project Ice Storm. PLoS One. 2014;9(9):e107653
  51. 51. Khulan B, Manning JR, Dunbar DR, et al. Epigenomic profiling of men exposed to early-life stress reveals DNA methylation differences in association with current mental state. Transl Psychiatry. 2014;4:e448
  52. 52. Non AL, Hollister BM, Humphreys KL, et al. DNA methylation at stress-related genes is associated with exposure to early life institutionalization. Am J Phys Anthropol. 2016;161(1):84-93
  53. 53. Houtepen LC, Vinkers CH, Carrillo-Roa T, et al. Genomewide DNA methylation levels and altered cortisol stress reactivity following childhood trauma in humans [•• Evidence is presented which indicates that increased DNA methylation at the KITLG locus is a mediator of childhood trauma-induced blunting of the cortisol response to stress in adulthood.]. Nat Commun. 2016;7:10967
  54. 54. Suderman M, Borghol N, Pappas JJ, et al. Childhood abuse is associated with methylation of multiple loci in adult DNA. BMC Med Genomics. 2014;7:13
  55. 55. Miller CA, Sweatt JD. Covalent modification of DNA regulates memory formation. Neuron. 2007;53:857-869
  56. 56. Fischer A, Sananbenesi F, Wang X, Dobbin M, Tsai LH. Recovery of learning and memory is associated with chromatin remodelling. Nature. 2007;447:178-182
  57. 57. Kuzumaki N, Ikegami D, Tamura R, Hareyama N, Imai S, Narita M, et al. Hippocampal epigenetic modification at the brainderived neurotrophic factor gene induced by an enriched environment. Hippocampus. 2011;21:127-132
  58. 58. Roth TL, Zoladz PR, Sweatt JD, Diamond DM. Epigenetic modification of hippocampal Bdnf DNA in adult rats in an animal model of posttraumatic stress disorder. J Psychiatr Res. 2011;45:919-926
  59. 59. sankova NM, Berton O, Renthal W, Kumar A, Neve RL, Nestler EJ. Sustained hippocampal chromatin regulation in a mouse model of depression and antidepressant action. Nat Neurosci. 2006;9:519-25
  60. 60. Covington HE 3rd, Maze I, LaPlant QC, Vialou VF, Ohnishi YN, Berton O, et al. Antidepressant actions of histone deacetylase inhibitors. J Neurosci. 2009;29:11451-11460
  61. 61. Hollis F, Wang H, Dietz D, Gunjan A, Kabbaj M. The effects of repeated social defeat on long-term depressive-like behavior and shortterm histone modifications in the hippocampus in male Sprague-Dawley rats. Psychopharmacology (Berl). 2010;211:69-77
  62. 62. Elliott E, Ezra-Nevo G, Regev L, Neufeld-Cohen A, Chen A. Resilience to social stress coincides with functional DNA methylation of the Crf gene in adult mice. Nat Neurosci. 2010;13:1351-1353
  63. 63. Uchida S, Hara K, Kobayashi A, Otsuki K, Yamagata H, Hobara T, et al. Epigenetic status of Gdnf in the ventral striatum determines susceptibility and adaptation to daily stressful events. Neuron. 2011;69:359-372
  64. 64. Champagne FA. Epigenetic mechanisms and the transgenerational effects of maternal care. Front Neuroendocrinol. 2008;29:386-397
  65. 65. Champagne FA, Meaney MJ. Stress during gestation alters postpartum maternal care and the development of the offspring in a rodent model. Biol Psychiatry. 2006;59:1227-1235
  66. 66. Champagne FA, Meaney MJ. Transgenerational effects of social environment on variations in maternal care and behavioral response to novelty. Behav Neurosci. 2007;121:1353-1363
  67. 67. Roth TL, Lubin FD, Funk AJ, Sweatt JD. Lasting epigenetic influence of early-life adversity on the BDNF gene. Biol Psychiatry. 2009;65:760-769
  68. 68. Weaver JR, Susiarjo M, Bartolomei MS. Imprinting and epigenetic changes in the early embryo. Mamm Genome. 2009;20:532-543
  69. 69. Anway MD, Cupp AS, Uzumcu M, Skinner MK. Epigenetic transgenerational actions of endocrine disruptors and male fertility. Science. 2005;308:1466-1469
  70. 70. Franklin TB, Russig H, Weiss IC, Graff J, Linder N, Michalon A, et al. Epigenetic transmission of the impact of early stress across generations. Biol Psychiatry. 2010;68:408-415
  71. 71. Wilkinson R, Pickett K. The Spirit Level: Why Equality is Better for Everyone. London, UK: Penguin; 2010
  72. 72. Marmot M. The Health Gap: the Challenge of an Unequal World. London, UK: Bloomsbury Publishing; 2015
  73. 73. Bourdieu P. The Forms Of Capital. In: Handbook Of Theory And Research For The Sociology Of Education. Greenwood, New York, NY, USA, 241-258 (1986)
  74. 74. Coleman J. Foundations of Social Theory. Cambridge, USA: Harvard University Press; 1994
  75. 75. Putnam RD. Bowling Alone: The Collapse and Revival of American Community. NY, USA: Simon and Schuster; 2001
  76. 76. Dickerson SS, Kemeny ME. Acute stressors and cortisol responses: a theoretical integration and synthesis of laboratory research. Psychol Bull. 2004;130(3):355-391
  77. 77. Gilbert P, McEwan K, Bellew R, Mills A, Gale C. The dark side of competition: how competitive behaviour and striving to avoid inferiority are linked to depression, anxiety, stress and self-harm. Psychol Psychother. 2009;82(Pt 2):123-136
  78. 78. Layte R. The association between income inequality and mental health: testing status anxiety, social capital, and neo-materialist explanations. Eur Sociol Rev. 2011;28(4):498-511
  79. 79. Layte R, Whelan CT. Who feels inferior? A test of the status anxiety hypothesis of social inequalities in health. Eur Sociol Rev. 2014;30(4):525-535
  80. 80. Mascaro O, Csibra G. Representation of stable social dominance relations by human infants. Proc Natl Acad Sci USA. 2012;109(18):6862-6867
  81. 81. Frankel D, Arbel T. Group formation by two-year olds. Int J Behav Dev. 1980;3(3):287-298
  82. 82. Dickerson SS, Gable SL, Irwin MR, Aziz N, Kemeny ME. Social-evaluative threat and proinflammatory cytokine regulation: an experimental laboratory investigation. Psychol Sci. 2009;20(10):1237-1244
  83. 83. Chiang JJ, Eisenberger NI, Seeman TE, Taylor SE. Negative and competitive social interactions are related to heightened proinflammatory cytokine activity. Proc Natl Acad Sci USA. 2012;109(6):1878-1882
  84. 84. Packard CJ, Bezlyak V, McLean JS, et al. Early life socioeconomic adversity is associated in adult life with chronic inflammation, carotid atherosclerosis, poorer lung function and decreased cognitive performance: a cross-sectional, population-based study. BMC Public Health. 2011;11:42
  85. 85. Kumari M, Shipley M, Stafford M, Kivimaki M. Association of diurnal patterns in salivary cortisol with all-cause and cardiovascular mortality: findings from the Whitehall II study. J Clin Endocrinol Metab. 2011;96(5):1478-1485
  86. 86. Kiecolt-Glaser JK, Gouin JP, Hantsoo L. Close relationships, inflammation, and health. Neurosci Biobehav Rev. 2010;35(1):33-38
  87. 87. Aslund C, Starrin B, Nilsson KW. Social capital in relation to depression, musculoskeletal pain, and psychosomatic symptoms: a cross-sectional study of a large population-based cohort of Swedish adolescents. BMC Public Health. 2010;10:715
  88. 88. Aslund C, Starrin B, Nilsson KW. Psychosomatic symptoms and low psychological well-being in relation to employment status: the influence of social capital in a large cross-sectional study in Sweden. Int J Equity Health. 2014;13:22
  89. 89. Youm Y, Laumann EO, Ferraro KF, et al. Social network properties and self-rated health in later life: comparisons from the Korean social life, health, and aging project and the national social life, health and aging project. BMC Geriatr. 2014;14:102
  90. 90. Uphoff EP, Pickett KE, Cabieses B, Small N, Wright J. A systematic review of the relationships between social capital and socioeconomic inequalities in health: a contribution to understanding the psychosocial pathway of health inequalities. Int J Equity Health. 2013;12:54
  91. 91. Fisher PA, Gunnar MR, Chamberlain P, Reid JB. Preventive intervention for maltreated preschool children: impact on children’s behavior, neuroendocrine activity, and foster parent functioning. J Am Acad Child Adolesc Psychiatry. 2000;39(11):1356-1364
  92. 92. Evans GW, Kim P, Ting AH, Tesher HB, Shannis D. Cumulative risk, maternal responsiveness, and allostatic load among young adolescents. Dev Psychol. 2007;43(2):341-351
  93. 93. Chen E, Miller GE, Kobor MS, Cole SW. Maternal warmth buffers the effects of low early-life socioeconomic status on pro-inflammatory signaling in adulthood. Mol Psychiatry. 2011;16(7):729-737
  94. 94. Miller GE, Brody GH, Yu T, Chen E. A family-oriented psychosocial intervention reduces inflammation in low-SES African American youth. Proc Natl Acad Sci USA. 2014;111(31):11287-11292
  95. 95. Hostinar CE, Sullivan RM, Gunnar MR. Psychobiological mechanisms underlying the social buffering of the hypothalamic-pituitary-adrenocortical axis: a review of animal models and human studies across development. Psychol Bull. 2014;140(1):256-282
  96. 96. Borghol N, Suderman M, McArdle W, et al. Associations with early-life socio-economic position in adult DNA methylation. Int J Epidemiol. 2012;41(1):62-74
  97. 97. Lam LL, Emberly E, Fraser HB, et al. Factors underlying variable DNA methylation in a human community cohort. Proc Natl Acad Sci USA. 2012;109 Suppl. 2:17253-17260
  98. 98. McGuinness D, McGlynn LM, Johnson PC, et al. Socioeconomic status is associated with epigenetic differences in the pSoBid cohort. Int J Epidemiol. 2012;41(1):151-160
  99. 99. Needham BL, Smith JA, Zhao W, et al. Life course socioeconomic status and DNA methylation in genes related to stress reactivity and inflammation: the multi-ethnic study of atherosclerosis. Epigenetics. 2015;10(10):958-969
  100. 100. Stringhini S, Polidoro S, Sacerdote C, et al. Life-course socioeconomic status and DNA methylation of genes regulating inflammation. Int J Epidemiol. 2015;44(4):1320-1330
  101. 101. Fryers T, Melzer D, Jenkins R, Brugha T. The distribution of the common mental disorders: social inequalities in Europe. Clin Pract Epidemiol Ment Health. 2005;1:14
  102. 102. Swartz JR, Hariri AR, Williamson DE. An epigenetic mechanism links socioeconomic status to changes in depression-related brain function in high-risk adolescents. Mol Psychiatry. 2016; https://doi.org/10.1038/mp.2016.82.; Epub ahead of print
  103. 103. Beach SR, Brody GH, Todorov AA, Gunter TD, Philibert RA. Methylation at SLC6A4 is linked to family history of child abuse: an examination of the Iowa Adoptee sample. Am J Med Genet B Neuropsychiatr Genet. 2010;153b(2):710-3
  104. 104. Beach SR, Brody GH, Lei MK, Kim S, Cui J, Philibert RA. Is serotonin transporter genotype associated with epigenetic susceptibility or vulnerability? Examination of the impact of socioeconomic status risk on African American youth. Dev Psychopathol. 2014;26(2):289-304
  105. 105. Zhao J, Goldberg J, Bremner JD, Vaccarino V. Association between promoter methylation of serotonin transporter gene and depressive symptoms: a monozygotic twin study. Psychosom Med. 2013;75(6):523-529
  106. 106. Nikolova YS, Koenen KC, Galea S, et al. Beyond genotype: serotonin transporter epigenetic modification predicts human brain function. Nat Neurosci. 2014;17(9):1153-1155
  107. 107. Swartz JR, Knodt AR, Radtke SR, Hariri AR. A neural biomarker of psychological vulnerability to future life stress. Neuron. 2015;85(3):505-511
  108. 108. Swartz JR, Williamson DE, Hariri AR. Developmental change in amygdala reactivity during adolescence: effects of family history of depression and stressful life events. Am J Psychiatry. 2015;172(3):276-283
  109. 109. Yehuda R, Flory JD, Bierer LM, et al. Lower methylation of glucocorticoid receptor gene promoter 1F in peripheral blood of veterans with post-traumatic stress disorder. Biol Psychiatry. 2015;77(4):356-364
  110. 110. Sadeh N, Spielberg JM, Logue MW, et al. SKA2 methylation is associated with decreased prefrontal cortical thickness and greater PTSD severity among trauma-exposed veterans. Mol Psychiatry. 2016;21(3):357-363
  111. 111. Boks MP, Rutten BP, Geuze E, et al. SKA2 methylation is involved in cortisol stress reactivity and predicts the development of post-traumatic stress disorder (PTSD) after military deployment. Neuropsychopharmacology. 2016;41(5):1350-1356
  112. 112. Guintivano J, Brown T, Newcomer A, et al. Identification and replication of a combined epigenetic and genetic biomarker predicting suicide and suicidal behaviors. Am J Psychiatry. 2014;171(12):1287-1296
  113. 113. Kaminsky Z, Wilcox HC, Eaton WW, et al. Epigenetic and genetic variation at SKA2 predict suicidal behavior and posttraumatic stress disorder. Transl Psychiatry. 2015;5:e627
  114. 114. Yehuda R, Daskalakis NP, Bierer LM, et al. Holocaust exposure induced intergenerational effects on FKBP5 methylation. Biol Psychiatry. 2016;80(5):372-380
  115. 115. Cunliffe VT. The epigenetic impacts of social stress: how does social adversity become biologically embedded? Epigenomics. 2016;8(12):1653-1669

Written By

Mehmet Ünal

Submitted: 07 November 2020 Reviewed: 18 August 2021 Published: 03 September 2021