Fact-checked by Grok 2 weeks ago

Behavioral epigenetics

Behavioral epigenetics is an emerging interdisciplinary field that investigates how environmental experiences, such as stress, nurturing, and social interactions, induce epigenetic modifications—reversible chemical changes to DNA and associated proteins that alter gene expression without changing the underlying genetic sequence—thereby influencing behavior across individuals and potentially generations. This field bridges the classic nature-nurture debate by demonstrating that psychosocial factors dynamically interact with the genome to shape traits like stress resilience, learning, and social behavior, resolving the dichotomy through mechanisms that integrate genetic predispositions with environmental inputs. Central to behavioral epigenetics are key epigenetic mechanisms, including , where methyl groups attach to bases (typically at CpG sites) to silence ; histone modifications, such as or of proteins that regulate accessibility; and noncoding RNAs, like microRNAs, which fine-tune gene activity in response to stimuli. These processes enable the brain to adapt to environmental cues, for instance, by modulating neural plasticity in regions like the and . Landmark studies in have shown that variations in maternal care, such as high levels of licking and grooming, reduce at the (Nr3c1) gene, leading to enhanced stress responses and calmer offspring behavior, effects that can be reversed through cross-fostering. In humans, behavioral epigenetics reveals links between early adversity and long-term outcomes, with childhood abuse associated with hyper of the NR3C1 in the , correlating with altered regulation and increased risk of or . Prenatal maternal depression has been tied to methylation changes in the (SLC6A4) in newborns, potentially predisposing them to anxiety or mood disorders. Preliminary evidence from studies on offspring of shows altered methylation patterns in genes related to regulation, such as NR3C1 or , suggesting intergenerational epigenetic effects, though challenges in replication and confirming transmission persist. The field's implications span and policy, highlighting the malleability of the epigenome throughout the lifespan and the potential for interventions like enriched environments, exercise, or to reverse adverse marks and promote . For example, dietary factors and can induce demethylation in genes linked to metabolic and behavioral , offering avenues for preventing disorders influenced by early-life . Recent as of 2025 has advanced understanding of interventions modulating to enhance behavioral against social . Ongoing challenges include distinguishing causal epigenetic effects from genetic confounders and elucidating intergenerational transmission in humans, yet behavioral epigenetics promises to redefine understandings of , , and therapeutic strategies.

Fundamentals

Definition and Scope

Behavioral epigenetics is the study of stable, heritable changes in gene expression that do not involve alterations to the underlying DNA sequence, with a particular focus on how these modifications influence behavioral outcomes such as learning, stress responses, and social interactions. These epigenetic changes, often induced by environmental experiences, allow organisms to adapt behaviorally without genetic mutations, bridging the gap between nurture and nature. The field emphasizes mechanisms like DNA methylation and histone modifications that regulate gene activity in response to external cues, thereby shaping neural and physiological processes relevant to behavior. The scope of behavioral epigenetics is inherently interdisciplinary, integrating insights from , , , , and to explore how environmental factors modulate and, in turn, . It encompasses applications across diverse species, including humans, such as rats and mice, and social insects like honeybees and , where epigenetic mechanisms underlie differentiation and social roles. This broad reach highlights the field's emphasis on how nurture—through experiences like or social interactions—can persistently shape by altering epigenetic marks on genes involved in behavioral regulation. Behavioral epigenetics emerged in the early as an extension of developmental epigenetics, building on foundational work linking environmental influences to in behavioral contexts. A seminal example is the influence of early maternal care in rats, where variations in pup licking and grooming by mothers lead to differential of the gene promoter in the offspring's , resulting in heightened or reduced stress reactivity that persists into adulthood. This demonstrates how early experiences can induce lasting epigenetic changes that affect behavioral phenotypes, which can influence the next generation through altered parental behavior.

Key Epigenetic Mechanisms

is a fundamental epigenetic mechanism involving the covalent addition of a to the 5-position of residues, predominantly within CpG dinucleotides in promoter regions, which typically results in transcriptional repression by recruiting methyl-binding proteins and altering chromatin accessibility. In neural contexts, this process plays a critical role in silencing genes associated with behavioral regulation, such as (BDNF), where hypermethylation of its promoter in response to diminishes expression and impairs in regions like the . For instance, early-life adversity has been shown to increase BDNF , correlating with altered stress responses and reduced in both peripheral blood and brain tissue. Histone modifications represent another core epigenetic layer, where post-translational alterations to histone tails—such as , which neutralizes positive charges to promote open and activation, or , which can either repress or activate depending on the residue—influence structure and DNA accessibility in neurons. Specifically, lysine 9 (H3K9) , often catalyzed by enzymes like G9a/GLP, induces formation and , but its dynamic regulation is essential for processes in the adult . Inhibition of H3K9 trimethylation () has been demonstrated to enhance and improve contextual fear memory in aged mice by promoting synaptic protein expression and spine formation. Non-coding RNAs, including microRNAs (miRNAs) and long non-coding RNAs (lncRNAs), exert epigenetic control by binding to target mRNAs to modulate their stability, translation, or degradation, thereby fine-tuning gene expression in post-mitotic neurons without altering the DNA sequence. In behavioral epigenetics, miR-124 stands out as a brain-enriched miRNA that promotes neuronal differentiation by repressing non-neuronal transcripts, such as those involved in progenitor proliferation, facilitating the transition to mature neuronal phenotypes critical for circuit formation. This silencing mechanism helps establish cell-type-specific gene profiles in the developing and adult nervous system, influencing behaviors tied to neural identity. Chromatin remodeling involves ATP-dependent protein complexes, such as and ISWI families, that utilize energy from ATP hydrolysis to reposition, evict, or restructure nucleosomes, thereby exposing or concealing DNA segments for transcriptional machinery access in synaptic contexts. These complexes are vital for , where they facilitate activity-induced changes underlying learning-related adaptations, such as in dendritic spine morphogenesis and storage. Disruptions in remodeler function, like mutations in SMARCA2, have been linked to neurodevelopmental disorders affecting behavioral outcomes through impaired dynamics at neuronal enhancers. At a conceptual level, epigenetic in behavioral contexts can be modeled as a integrating the fixed DNA with modifiable epigenetic marks and dynamic environmental signals: Gene = f(DNA + epigenetic marks + environmental signals), where environmental inputs like or experiences trigger mark alterations to adaptively tune neural activity without changes. This framework underscores how bridges and to influence behaviorally relevant transcription in the .

Historical Development

Early Foundations

The foundations of behavioral epigenetics trace back to classical genetics, where the concept of epigenetics emerged as a framework for understanding gene-environment interactions in development. In 1942, British embryologist Conrad Hal Waddington coined the term "epigenetics" to describe the processes bridging genotype and phenotype, emphasizing how environmental factors influence developmental trajectories without altering the DNA sequence itself. Waddington's seminal idea, illustrated later in his 1957 "epigenetic landscape" model, portrayed development as a dynamic interplay where genes and external cues canalize phenotypic outcomes, laying groundwork for later explorations of environmental modulation of behavior. Early links between environment and behavior were further illuminated through twin studies in the 1960s and 1970s, which quantified the relative contributions of heredity and environment to personality traits. These investigations, including analyses of monozygotic twins reared apart or together, revealed that while genetic factors accounted for substantial variance in traits like extraversion and neuroticism (heritability estimates around 40-50%), shared and non-shared environments significantly shaped individual differences. The implications extended to behavioral epigenetics by highlighting how environmental exposures could amplify or suppress genetic potentials, as seen in the Minnesota Study of Twins Reared Apart, initiated in the late 1970s and yielding key data by the 1990s on how divergent rearing environments influenced personality despite identical genotypes. Milestones in the and solidified epigenetic mechanisms' role in behavior through discoveries in disorders, such as Prader-Willi syndrome (PWS). First clinically described in 1956, PWS was linked in 1981 to deletions on the paternal chromosome 15q11-q13, but by 1989, studies identified maternal as an alternative cause, suggesting parent-of-origin-specific via epigenetic marks like . This imprinting defect led to behavioral phenotypes including hyperphagia, cognitive impairments, and obsessive-compulsive tendencies, demonstrating how epigenetic dysregulation could directly manifest in complex behaviors. Foundational animal experiments in the 1990s provided direct evidence of environmental influences on epigenetic regulation of . Michael Meaney's studies on pups showed that variations in maternal and grooming during the first week postpartum stably altered stress responses by modifying hypothalamic-pituitary-adrenal () axis function, with high-care pups exhibiting reduced fearfulness and reactivity compared to low-care counterparts. These effects, observed as early as 1992 in initial reports on maternal care's impact on monoaminergic systems, foreshadowed epigenetic mediation, later confirmed through changes at promoters. Key figures like advanced this by elucidating how learning-induced in involved experience-dependent , essential for formation without genetic mutations. Similarly, Arthur L. Beaudet's work in the 1990s on in imprinting disorders highlighted its role in behavioral phenotypes, as seen in PWS models where errors silenced paternal genes critical for neurodevelopment.

Major Discoveries and Milestones

One of the foundational milestones in behavioral epigenetics occurred in 2003 with the viable yellow (A^vy) led by Randy Jirtle and Robert Waterland, which demonstrated that maternal dietary supplementation with methyl donors like folic acid, , choline, and betaine could alter at the Agouti gene promoter, shifting from obese, yellow-coated phenotypes prone to and cancer to lean, brown-coated, healthier ones with reduced obesity-related behaviors. This experiment provided direct evidence that environmental nutrition influences epigenetic marks, thereby modulating behavioral and metabolic traits without changing the DNA sequence. In human studies, the Dutch Hunger Winter famine of 1944-1945 offered early insights into prenatal nutrition's epigenetic legacy; a 2008 analysis by Bastiaan Heijmans and colleagues revealed persistent hypomethylation at the IGF2 gene in adults exposed , correlating with increased risk and other metabolic disorders, underscoring how famine-induced epigenetic changes link early adversity to adult behavioral vulnerabilities. Building on such findings, a 2011 review in BioScience by Tabitha Powledge titled "Behavioral Epigenetics: How Nurture Shapes Nature" synthesized emerging evidence that environmental experiences, such as and social interactions, induce epigenetic modifications affecting in regions tied to , establishing the field as a bridge between nature and nurture. During the , advances highlighted transgenerational effects; a 2014 study by Brian Dias and Kerry Ressler showed that early-life in male mice led to altered profiles, transmitting fear responses to offspring across generations via epigenetic inheritance in the , without direct exposure. Concurrently, research on social revealed DNA methylation's role in caste determination; a 2010 study by Andrew Foret and colleagues identified over 550 differentially methylated genes in honeybee brains, where royal jelly-induced hypomethylation promoted queen development over worker castes, influencing and reproductive behaviors. Methodological progress included the widespread adoption of ChIP-seq in the for behavioral , enabling genome-wide mapping of modifications in response to environmental cues, as reviewed in 2011 by R. David Hawkins and Bing Ren, which facilitated precise identification of activity-dependent epigenetic changes in neural circuits. Recent developments from 2023 to 2025 have further solidified the field's relevance to and . A 2025 review in Neuropsychopharmacology by C. J. Peña and colleagues detailed how postnatal epigenome maturation, particularly dynamics in neurons, enhances and resilience, integrating early experiences into adaptive behavioral circuits. Complementing this, a 2025 narrative review in the Journal of Laboratory and Precision Medicine by Rosita Gabbianelli examined studies showing that modulates at genes like BDNF and NR3C1, increasing susceptibility to issues including anxiety-like behaviors in adulthood. Additionally, a 2025 JETIR publication by Yash Gawande synthesized evidence linking behavioral epigenetics to outcomes, emphasizing how trauma-induced changes contribute to disorders like through altered response pathways.

Core Mechanisms in Behavior

Gene-Environment Interactions

Gene-environment interactions in behavioral epigenetics describe the dynamic process by which environmental cues, such as or , influence through epigenetic mechanisms, thereby shaping behavioral traits. These interactions occur when environmental signals activate transcription factors that deposit epigenetic marks, like or histone modifications, on genes involved in neural and behavioral regulation. For instance, stressors can alter the epigenome by modulating the activity of enzymes such as DNA methyltransferases, leading to persistent changes in without altering the sequence itself. This framework extends the classic gene-environment (GxE) model, where behavioral phenotypes arise from additive effects of (G) and (E), plus their interaction (G × E) mediated by epigenetic modulation, emphasizing how environmental inputs can amplify or suppress genetic predispositions to specific behaviors. A well-established example involves prenatal stress in rodent models, where maternal stress exposure leads to increased anxiety-like behaviors in via hyper of the gene (Nr3c1) promoter in the . This methylation reduces expression, heightening hypothalamic-pituitary-adrenal () axis reactivity and sensitivity, as demonstrated in studies using rat models exposed to repeated restraint during gestation. In humans, studies from the 2010s and 2020s examining (ACEs) have linked early-life adversity, such as abuse or neglect, to epigenetic alterations associated with adult ; for example, increased methylation in stress-response genes correlates with heightened scores in longitudinal samples of individuals with high ACE scores. These findings highlight how early environmental insults can program long-term behavioral vulnerabilities through epigenetic reprogramming of key regulatory genes. The bidirectional nature of these interactions is evident in how behaviors themselves can feedback to reshape the epigenome. Physical exercise, for instance, promotes and reduces 5 levels at the promoter region of the (BDNF) gene, enhancing BDNF expression and supporting mood improvement by fostering in mood-regulating brain regions like the . This reciprocal dynamic underscores that while environments drive initial epigenetic changes, subsequent behavioral adaptations can reinforce or reverse them, offering potential therapeutic avenues. Recent 2025 research further illustrates protective factors, showing that buffers epigenetic stress marks by attenuating in HPA-axis genes among individuals facing chronic adversity, thereby mitigating behavioral risk profiles.

Molecular Pathways and Transgenerational Effects

Molecular pathways in behavioral epigenetics involve intricate biochemical cascades that link environmental stimuli to lasting changes in gene expression, particularly influencing fear and stress responses. The ERK/MAPK signaling pathway plays a central role in fear memory formation by activating downstream effectors that modify chromatin structure. Specifically, ERK/MAPK activation leads to phosphorylation of histone H3 at serine 10 (H3S10), which facilitates subsequent acetylation at lysine 14 (H3K14), enhancing transcriptional accessibility in the hippocampus during contextual fear conditioning. This histone acetylation promotes the expression of immediate early genes like c-Fos, essential for memory consolidation. In parallel, glucocorticoid signaling within the hypothalamic-pituitary-adrenal (HPA) axis mediates stress-induced epigenetic alterations. Binding of glucocorticoids to their receptors triggers translocation to the nucleus, where they interact with glucocorticoid response elements to modulate DNA methylation and histone modifications in stress-responsive genes, such as FKBP5, thereby altering HPA axis sensitivity and behavioral resilience to chronic stress. These pathways exemplify how acute environmental challenges can induce stable epigenetic marks that influence behavioral phenotypes. Transgenerational inheritance of epigenetic modifications extends these pathways beyond the exposed individual, allowing behavioral traits to persist across generations via . In a seminal 2014 mouse study, olfactory in parental males resulted in heightened odor sensitivity and altered neural structure in and grand-offspring, mediated by changes in sperm microRNAs (miRNAs) that reprogram in the progeny. This demonstrates how fear-associated epigenetic signals can be packaged into gametes and conveyed heritably. In humans, evidence from descendants of reveals intergenerational of effects through altered patterns in PTSD-related genes. For instance, of survivors exhibit lower methylation at specific FKBP5 intronic sites, associated with maternal exposure and increased sensitivity, as documented in studies from the early 2020s. However, human evidence for remains correlational and is subject to debate, with calls for further research to confirm and rule out alternative explanations. These findings highlight the potential for environmental to imprint behavioral vulnerabilities across generations. Key mechanisms underlying transgenerational effects include paramutation-like phenomena and the regulatory role of PIWI-interacting RNAs (piRNAs) in the . Paramutation-like effects in mammals involve heritable epigenetic conversions where one induces a stable, meiotically transmissible change in a homologous , challenging classical and observed in contexts like stress-responsive loci. Such interactions can propagate behavioral adaptations without genetic sequence alterations. piRNAs contribute by guiding and silencing transposable elements in germ cells, selectively retaining environmentally induced marks while erasing others during ; transgenerationally inherited piRNAs trigger de novo piRNA biogenesis from homologous regions, ensuring persistence of adaptive epigenetic states. These processes balance erasure and retention to facilitate inheritance. A for epigenetic posits that the probability of equals the product of retention rate in the and environmental persistence, underscoring how these factors determine the evolutionary utility of transgenerational effects in behavioral . Recent analyses, such as a 2025 review, emphasize "epigenetic echoes" of , integrating , histone modifications, and non-coding RNAs to explain multigenerational behavioral shifts and their implications for in fluctuating environments. This perspective reveals how such mechanisms enable rapid to stressors, influencing population-level behaviors like stress avoidance.

Cognitive Processes

Learning and Memory

Epigenetic modifications play a crucial role in underlying learning and processes, particularly through histone acetylation mediated by the cAMP response element-binding protein (CREB) and its co-activator (CBP). In (LTP), a cellular model of learning, CREB leads to CBP , which acetylates histones at promoters of plasticity-related genes, facilitating opening and transcriptional essential for synaptic strengthening in the . This mechanism is conserved across species and is vital for the induction of late-phase LTP, where deficits in CBP activity impair both synaptic potentiation and formation. In associative learning paradigms such as contextual in mice, epigenetic changes including promote the expression of immediate-early genes critical for encoding. Specifically, training induces active demethylation at the promoter of the gene in hippocampal neurons, enhancing Arc transcription and supporting dendritic spine remodeling necessary for fear . This demethylation is mediated by factors like Gadd45b, which facilitates enzyme activity to convert to hydroxymethylcytosine, thereby derepressing plasticity genes within hours of learning. Such dynamic epigenetic shifts ensure rapid to environmental cues while maintaining memory specificity. Memory consolidation relies on sustained epigenetic regulation, where histone deacetylase (HDAC) inhibitors enhance retention by increasing histone acetylation and in . Systemic or hippocampal administration of HDAC inhibitors like immediately after training improves performance in spatial and fear memory tasks, such as the Morris water maze and contextual , by boosting CREB-dependent transcription of genes like BDNF. In animal models, exercise induces epigenetic changes, such as reduced at hippocampal promoters of genes like BDNF. In humans, aerobic training in older adults correlates with improved recall and elevated BDNF levels, suggesting related mechanisms that enhance . These interventions highlight as a bridge between lifestyle factors and cognitive enhancement. Epigenetic mechanisms differ across memory types, with DNA methylation patterns in the hippocampus governing spatial memory formation, while amygdala modifications support implicit emotional memories. In spatial learning, increased methylation of repressor genes in the dorsal hippocampus stabilizes engrams during navigation tasks, whereas hypomethylation of plasticity genes like BDNF facilitates long-term retention; disruptions in this balance, such as DNMT inhibition, impair Morris water maze performance. Conversely, in implicit fear memory, amygdala histone acetylation and demethylation at promoters of genes like Nr4a consolidate Pavlovian associations, with HDAC inhibition enhancing trace fear conditioning by promoting synaptic strengthening in the lateral amygdala. A 2025 review synthesizes evidence on epigenome maturation during neural , emphasizing how early-life epigenetic marks, such as persistent alterations from stress, predispose individuals to adult learning deficits by constraining hippocampal adaptability. In pathological contexts like models, amyloid-beta oligomers induce aberrant deacetylation and hypermethylation in the , suppressing plasticity genes and causing impairment; reversing these via HDAC inhibitors restores LTP and cognitive function in transgenic mice.

Decision-Making and Executive Function

Epigenetic modifications in the () play a critical role in modulating processes, particularly through alterations in signaling pathways. of genes, such as DRD2, has been linked to increased risk-taking behaviors in , where environmental stressors exacerbate vulnerability by downregulating receptor expression and impairing regulation of . For instance, repeated exposure during reduces DRD2 expression via deacetylation in the , heightening predisposition to addictive and high-risk decisions. These epigenetic changes interact with genetic variants, such as the DRD2 A1 , amplifying novelty-seeking and emotional dysregulation under adverse conditions. Prenatal alcohol exposure induces lasting epigenetic disruptions that compromise executive function, including planning and impulse control, through (HDAC) activity and altered acetylation patterns. Specifically, alcohol enhances HDAC2 expression, leading to reduced and H4 acetylation in key neural regions, which impairs essential for . Studies in models demonstrate that these changes persist into adulthood, correlating with deficits in and , as seen in phenotypes. Such mechanisms highlight how early environmental insults reprogram states to bias toward short-term gains over long-term consequences. In the (), a subregion of the integral to value-based choices, epigenetic factors like microRNA-mediated silencing influence dendritic maturation and contribute to decision biases, such as overvaluation of immediate rewards. Early environmental exposures alter profiles in OFC neurons, shifting excitatory-inhibitory balance and promoting perseverative errors in reversal learning tasks. Human imaging studies further reveal correlations between these epigenetic signatures and functional connectivity patterns during . Recent investigations integrate with to elucidate in conditions like ADHD, where 2024 analyses show correlations between peripheral at dopamine-related loci and fMRI hypoactivation in networks during inhibitory tasks. In gambling contexts, altered histone marks, including increased H3K9 at DRD2 promoters in reward circuits, enhance anticipation-driven by sensitizing ventral striatal responses. A 2025 study in Frontiers in demonstrates that sub-chronic stress induces sex-specific changes in genes like DRD2 and prodynorphin, impairing effort-based under . Conceptual frameworks, such as the valence weighting model, posit that epigenetic tuning of serotonin pathways modulates the relative processing of positive and negative outcomes in decisions. Hypermethylation of the gene (SLC6A4) predicts heightened reactivity to aversive cues, biasing choices toward avoidance and altering reward valuation. These modifications interact with dopamine-serotonin dynamics to fine-tune OFC-PFC circuits, influencing adaptive versus maladaptive executive strategies.

Psychiatric Disorders

Stress and Anxiety Disorders

Epigenetic modifications play a critical role in the dysregulation of the hypothalamic-pituitary-adrenal (HPA) axis, which governs stress responses and contributes to vulnerability for anxiety disorders. In particular, DNA methylation changes in the glucocorticoid receptor gene NR3C1 have been linked to altered HPA axis sensitivity. Studies on maternal care in rodents demonstrate that variations in licking and grooming behaviors lead to increased NR3C1 promoter methylation in offspring, resulting in heightened anxiety-like behaviors and reduced glucocorticoid feedback inhibition. In humans, higher maternal anxiety during pregnancy predicts elevated NR3C1 methylation in offspring, accounting for significant variance in methylation levels at specific CpG sites and associating with increased risk for anxiety vulnerability later in life. Similarly, low maternal-infant interaction quality correlates with higher NR3C1 methylation in children, exacerbating HPA axis hyperactivity and anxiety proneness. In anxiety disorders, epigenome-wide association studies (EWAS) have identified key methylation sites influencing symptom severity. For (PTSD), differential methylation of the FKBP5 gene, a regulator of sensitivity, emerges as a prominent marker; trauma-exposed individuals with FKBP5 demethylation show impaired recovery and heightened PTSD risk, particularly when combined with childhood adversity. In generalized anxiety disorder (GAD), methylation alterations in the serotonin transporter gene (SLC6A4) are associated with reduced transporter expression and persistent anxiety symptoms; higher methylation upstream of SLC6A4 predicts poorer response to in affected children. These findings highlight how epigenetic marks at - and serotonin-related loci mediate the transition from acute to chronic anxiety pathologies. Chronic stress contributes to allostatic load—the cumulative wear from repeated physiological activation—through -induced epigenetic changes, including modifications that alter neuronal . Repeated exposure leads to persistent acetylation and methylation shifts in the , promoting maladaptive responses and anxiety escalation. A 2025 narrative review in the Journal of Laboratory and Precision Medicine synthesizes evidence that social disrupts neuronal , inducing changes in brain regions like the that heighten anxiety vulnerability and allostatic burden. Interventional approaches targeting epigenetics show promise in mitigating anxiety-related marks. In rodent models, early environmental enrichment—providing enhanced sensory, social, and cognitive stimulation—reverses methylation alterations induced by early-life stress, reducing anxiety-like behaviors and restoring HPA axis balance into adulthood. Human cohort studies from 2023 to 2025 further substantiate these links, revealing that epigenetically programs adult anxiety. Longitudinal analyses indicate that physical and emotional maltreatment in childhood accelerates epigenetic aging clocks, with increased at trauma-sensitive loci correlating with elevated adult anxiety scores independent of chronological age. A 2025 study of third-generation found distinct patterns in anxiety- and stress-related genes (e.g., , NR3C1), associating with heightened stress reactivity and subclinical anxiety-related symptoms in adulthood. These findings underscore the enduring impact of early adversity on the epigenome and its role in anxiety persistence.

Mood and Neurodevelopmental Disorders

Behavioral epigenetics has implicated epigenetic modifications in the pathophysiology of mood disorders, where influences patterns of key genes involved in . In (MDD), hypermethylation of the (BDNF) promoter region, particularly exon IV, has been consistently observed in patients exposed to prolonged , leading to reduced BDNF expression and impaired hippocampal function. This methylation change correlates with depressive symptom severity and cognitive deficits, highlighting BDNF as a critical mediator of stress-induced vulnerability. For , modifications, including variants in acetylation and methylation states, show phase-specific alterations, with increased activity during manic episodes contributing to dysregulated in neurons. These epigenetic shifts in mania phases disrupt genes and enhance excitability in limbic circuits, exacerbating mood instability. In neurodevelopmental disorders, epigenome-wide association studies (EWAS) have identified aberrant at the SHANK3 locus in , where hypermethylation of intragenic CpG islands in postmortem tissues reduces SHANK3 protein levels essential for synaptic scaffolding. This epigenetic dysregulation correlates with core symptoms like social deficits and repetitive behaviors, affecting up to 15% of cases with altered SHANK3 expression. Similarly, prenatal famine exposure in the Dutch Hunger Winter study induces persistent hypomethylation of the IGF2 gene in survivors, which has been associated with elevated risk; alterations in IGF2 may contribute to disrupted neurodevelopmental programming and increased incidence (twofold in exposed offspring). Offspring of exposed individuals exhibit a twofold increased incidence of , with these epigenetic marks persisting into adulthood and linking early to dopaminergic pathway abnormalities. Mechanistic insights underscore imprinting disruptions as a core epigenetic factor in mood regulation within neurodevelopmental contexts, as seen in Prader-Willi syndrome (PWS), where paternal deletion or maternal at 15q11.2-q13 leads to loss of imprinted , resulting in hypothalamic dysregulation and mood lability. This imprinting failure silences SNORD116 clusters, promoting hyperphagia-related irritability and emotional outbursts that mimic phenotypes. Recent analyses further demonstrate that epigenetic resilience to mood disorders involves protective demethylation of genes, buffering against in vulnerable populations and potentially mitigating depressive trajectories. However, challenges remain in distinguishing causal epigenetic effects from genetic or environmental confounders in these associations. Therapeutic interventions targeting these epigenomes offer promising avenues; for instance, treatment in induces genome-wide demethylation and acetylation changes, enhancing BDNF expression and stabilizing mood episodes in responders. In ASD models, prenatal valproic acid exposure recapitulates epigenetic signatures like hyperacetylation and MeCP2 dysregulation, producing autism-like behaviors in that inform environmental risk factors. A 2025 review synthesizes these findings, emphasizing how epigenetics intersects with neurodevelopment to shape disorder susceptibility and advocating for biomarker-driven interventions. Transgenerational mood risks may arise from these marks, though detailed inheritance patterns warrant further delineation.

Addiction and Impulse Control Disorders

Epigenetic modifications in the brain's reward pathways play a central role in the development and persistence of , particularly through alterations in and that affect in regions like the . In , chronic exposure induces persistent acetylation of histones associated with the DeltaFosB , a that accumulates in the and maintains elevated levels even after prolonged , thereby sustaining motivational responses to drug cues. Similarly, involves increased at the promoter region of the OPRM1 , which encodes the mu-opioid receptor; this hypermethylation, observed in peripheral blood and brain tissues of users, reduces receptor expression and correlates with heightened to dependence. These changes contribute to the long-term maladaptive underlying compulsive drug-seeking behaviors. In impulse control disorders, epigenetic dysregulation of dopamine-related genes further exacerbates behavioral disinhibition. For attention-deficit/hyperactivity disorder (ADHD), a condition characterized by impaired impulse control, methylation of the DAT1 gene, which codes for the , has been identified as a ; higher levels at specific CpG sites in DAT1 are associated with increased ADHD symptoms and reduced dopamine clearance in prefrontal circuits. Behavioral addictions, such as pathological , involve histone modifications in the , where chronic engagement leads to altered patterns that enhance reward sensitivity and diminish , mirroring molecular changes seen in substance use disorders. Environmental factors during critical developmental windows can induce epigenetic marks that elevate transgenerational risk for and . Adolescent exposure to drugs like or opioids triggers heritable changes in germ cells, increasing addiction susceptibility in offspring through altered expression of reward-related genes, as demonstrated in models where F1 and generations exhibit heightened drug self-administration. Recent 2025 research on highlights its role in , showing that adolescent vaping induces epigenetic silencing of nicotinic receptor genes via promoter hypermethylation, which persists into adulthood and amplifies risk for and impulsive . Therapeutic interventions targeting these epigenetic mechanisms show promise for mitigating addiction and impulse control deficits. (HDAC) inhibitors, such as , have been effective in preclinical models by reversing cocaine- or opioid-induced hypoacetylation in the , thereby reducing reinstatement of drug-seeking behaviors and cue-induced craving without affecting baseline locomotion.

Social and Evolutionary Behaviors

Epigenetics in Social Insects

In eusocial insects such as honeybees (Apis mellifera) and , epigenetic modifications play a pivotal role in determining differentiation and social roles without altering the underlying sequence. These organisms exhibit remarkable behavioral plasticity, where identical genotypes can produce distinct phenotypes like queens, workers, or soldiers based on environmental cues during development. , in particular, has been identified as a key mechanism regulating differences between castes. In honeybees, the discovery of a functional CpG system demonstrated that patterns are conserved and active in social insects, contrasting with many non-social . Subsequent studies revealed widespread differential between queen and worker castes, with over 550 genes showing significant methylation differences in tissue, often silencing genes associated with reproductive traits in workers to prevent queen-like development. Behavioral plasticity in these species is further exemplified by nutritional influences on epigenetic states. In honeybees, larvae fed develop into queens through histone deacetylase inhibitor (HDACi) activity in the jelly, which promotes histone acetylation and alters to favor queen morphology and fertility. This epigenetic switch occurs during a critical larval window, enabling reversible changes in developmental trajectories. In ants, such as Camponotus floridanus, division of labor among workers involves epigenetic reprogramming mediated by the CoREST corepressor complex, which acts as a to transition behaviors from to , influenced by signaling. These mechanisms allow colonies to adapt task allocation dynamically without genetic reconfiguration. Epigenetics also contributes to the evolutionary success of social insects by facilitating rapid to environmental pressures through nongenetic . In eusocial species, and other marks enable quick phenotypic responses, such as shifts in response to colony needs, bypassing slower genetic evolution and promoting . This contrasts with sociality, where similar epigenetic processes influence group behaviors but operate in more genetically diverse systems. In termites like Zootermopsis nevadensis, non-coding RNAs, including microRNAs, regulate soldier differentiation by targeting genes involved in morphological specialization, highlighting RNA-based epigenetics in defensive roles. Recent advances, including CRISPR-Cas9-based epigenome editing, have provided functional validation of these mechanisms; for instance, targeted modifications in honeybee genes have confirmed causal links between patterns and behavioral traits like .

Human Social Behavior and Inheritance Patterns

Behavioral epigenetics examines how environmental influences on shape human social interactions, such as attachment and , through mechanisms like of key receptors. Methylation levels at the (OXTR) have been linked to variations in empathic responses and social bonding; specifically, lower OXTR correlates with enhanced in response to , as observed in studies of healthy adults where reduced methylation in OXTR and the oxytocin (OXT) predicted stronger prosocial behaviors. Similarly, OXTR influences attachment styles, with higher methylation associated with increased attachment anxiety and reduced secure bonding in interpersonal relationships across adulthood. These findings underscore how epigenetic modifications in the oxytocin system modulate normative social behaviors essential for and group cohesion. Twin studies provide evidence for epigenetic contributions to social traits by comparing monozygotic twins, who share identical DNA but may differ in gene expression due to environmental factors. In discordant monozygotic twin pairs, epigenetic differences, particularly in DNA methylation patterns of stress-response genes like MAOA, have been associated with variations in anxious-depressive liability, including social withdrawal and anxiety in interpersonal contexts. Such studies reveal that early social experiences can induce site-specific methylation changes that diverge between twins, influencing social anxiety traits independently of genetic factors and highlighting the role of environment in shaping epigenetic profiles for social adaptation. Transgenerational epigenetic inheritance patterns demonstrate how social can propagate altered behaviors across generations, affecting and social trust. A 2025 review on epigenetic echoes of details how exposure to severe social adversities, such as or collective violence, leads to persistent changes in offspring and grandchildren. For instance, descendants of Dutch Hunger Winter survivors exhibit epigenetic marks at growth-related genes like IGF2. However, transgenerational epigenetic inheritance in humans remains controversial, with ongoing debates about distinguishing true epigenetic effects from genetic, cultural, or environmental confounders. From an evolutionary perspective, epigenetics intersects with human social behaviors by facilitating flexible responses that align with and altruism. Epigenetic regulation of genes, including those in the oxytocin pathway, allows for environmentally tuned expression that promotes prosocial actions toward kin, enhancing without altering the DNA sequence. This epigenetic layer enables rapid behavioral adjustments to social pressures, complementing genetic kin selection by embedding learned cooperative patterns across generations. Maternal influences via placental epigenetics exemplify how prenatal social environments shape offspring social skills. Placental DNA methylation patterns associated with maternal stress or caregiving quality predict infant socioemotional development; for example, hypomethylation at certain loci correlates with better emotional regulation and social engagement in early childhood, as seen in cohorts tracking maternal depressive symptoms. These effects highlight a direct pathway for epigenetic transmission of social competencies, where placental modifications act as a bridge between maternal experiences and child interpersonal abilities, fostering adaptive social behaviors from birth.

Challenges and Future Directions

Methodological Limitations

One major technical challenge in behavioral epigenetics research is tissue specificity, as epigenetic modifications, particularly , exhibit substantial variation across different s, making it difficult to infer brain-specific changes from accessible peripheral samples like or . For instance, studies have shown that fewer than 8% of CpG sites display strong correlations between and methylation patterns, limiting the reliability of peripheral s as proxies for neural processes underlying . This issue is exacerbated in neuropsychiatric contexts, where direct access to is restricted to postmortem samples, which suffer from small sample sizes, incomplete phenotyping, and potential effects that alter epigenetic profiles. Cell-type heterogeneity further complicates epigenomic profiling, as bulk tissue analyses aggregate signals from diverse cell populations, such as neurons, glia, and immune cells in the , potentially masking behaviorally relevant changes in specific subtypes. In complex tissues, shifts in cell-type composition can mimic disease- or environment-induced epigenetic alterations, reducing the sensitivity of epigenome-wide association studies (EWAS) to detect true biomarkers. For example, in EWAS, many differentially methylated sites were attributable to changes in granulocyte-lymphocyte proportions rather than intrinsic disease effects, a confound that similarly affects behavioral studies where cellular diversity in brain regions like the obscures targeted insights. Adjusting for cell-type fractions using deconvolution algorithms is recommended but not universally applied, highlighting the need for single-cell , though these methods remain nascent and resource-intensive. Causality remains a persistent issue in behavioral epigenetics, particularly in EWAS, where observed associations between epigenetic marks and behavioral traits often reflect correlations rather than direct causation, compounded by reverse causation or bidirectional influences. For externalizing behaviors like aggression, DNA methylation differences in peripheral blood may arise as consequences of the behavior itself, such as through chronic stress responses, rather than as predisposing factors, with cross-sectional designs unable to disentangle these directions. Confounding variables, including socioeconomic status, lifestyle, and comorbidities, further obscure causal links, as epigenetic changes are dynamic and responsive to multiple environmental inputs. Mendelian randomization approaches have been proposed to strengthen inference, but their application in behavioral contexts is limited by the polygenic nature of traits and the lack of robust instrumental variables for epigenetic variants. Reproducibility challenges are evident in the field, with replication failures common due to small sample sizes, methodological inconsistencies, and transient epigenetic marks that vary over time or across populations. In stress-related epigenetics, for example, only a of CpG sites associated with childhood adversity at age 7 failed to replicate at age 15, underscoring the instability of findings and the need for longitudinal, large-scale meta-analyses. The translatability of animal models to humans poses additional ethical and practical limitations, as species differences in structure and epigenetic responses hinder extrapolation; overall, over 90% of preclinical findings from fail to translate to human behavioral outcomes, necessitating cautious interpretation of rodent-based insights into human . High-throughput amplifies these issues, where the (FDR)—defined as the proportion of false positives among all positive results—must be controlled to avoid spurious associations, yet underpowered studies often inflate FDR estimates in epigenomic screens.

Emerging Therapies and Research Frontiers

Emerging research in behavioral epigenetics is exploring therapeutic interventions that target epigenetic modifications to address trauma-related disorders. Histone deacetylase (HDAC) inhibitors have shown promise in preclinical models of post-traumatic stress disorder (PTSD) by enhancing fear extinction and reducing symptoms through increased histone acetylation in brain regions like the hippocampus. For instance, systemic administration of HDAC inhibitors in rodent models of PTSD improved spatial memory and memory extinction, suggesting potential for translating these effects to human therapies. Similarly, in addiction research, CRISPR-dCas9-based epigenetic editing tools are being developed for targeted DNA demethylation at loci associated with reward pathways, with preclinical studies demonstrating potential for reversing drug-induced epigenetic changes without altering the underlying DNA sequence. At the research frontiers, single-nucleus is enabling high-resolution mapping of epigenetic states in individual neurons to uncover behavioral diversity, such as variations in stress response across cortical cell types. This approach has revealed how chromatin accessibility influences patterns linked to behavioral traits, providing a foundation for studying complex behaviors like social interaction. Additionally, is integrating with epigenetic data to predict behavioral risks; for example, AI-driven models using clocks can forecast susceptibility to . Transdiagnostic strategies are advancing through epigenetic biomarkers that support personalized , where DNA methylation profiles at genes like NR3C1 predict treatment responses across disorders such as and anxiety. These biomarkers facilitate tailored interventions by identifying individuals likely to benefit from specific therapies, promoting a shift toward precision medicine. Complementing this, evolutionary models highlight how epigenetic mechanisms drive behavioral , such as transgenerational of stress in populations exposed to environmental pressures, offering insights into behavioral . A 2025 review in further emphasized protective factors like that foster epigenetic , reducing behavioral vulnerability through altered patterns in stress-response genes. Looking ahead, longitudinal human studies are prioritizing interventions—such as exercise and —to reverse maladaptive epigenetic marks associated with behavioral disorders, with early trials showing slowed epigenetic aging and improved cognitive outcomes after sustained implementation.

References

  1. [1]
    Behavioral Epigenetics: Perspectives Based on Experience ... - NIH
    Aug 23, 2019 · Behavioral epigenetics is defined as the study of how epigenetic alterations induced by experience and environmental stress may affect animal behavior.2. Epigenetic Regulation... · 2.1. Dna Methylation · 2.2. Histone Modification
  2. [2]
    Epigenetic Consequences of Adversity and Intervention Throughout ...
    Aug 20, 2020 · Behavioral epigenetics posits that both nature and nurture must be considered when determining the etiology of behavior or disease.Abstract · Introduction · Summary And Policy...
  3. [3]
    Behavioral epigenetics - Wiley Interdisciplinary Reviews
    Dec 1, 2016 · Other behaviors also influence our epigenetic states. For example, exercise causes changes in DNA methylation in muscle cells.30 Likewise, what ...Epigenetic Mechanisms · Epigenetic Effects Of... · This Is Not Your Father's...
  4. [4]
    [PDF] "The Emerging Field of Social and Behavioral Epigenetics" in
    Social and behavioral epigenetics is the study of psychosocial factors that impact biol- ogy through an epigenetic mechanism.
  5. [5]
    Epigenetic mechanisms in the development of behavior
    As will be reviewed here, studies have demonstrated that DNA methylation alterations are biological responses to psychological and social-contextual factors.
  6. [6]
    Behavioral epigenetics - PMC - PubMed Central
    Behavioral epigenetics was described as the application of the principles of epigenetics to the study of physiological, genetic, environmental, and ...
  7. [7]
    Eusocial insects as emerging models for behavioural epigenetics
    Sep 9, 2014 · This Review provides insights into the use of eusocial insect species as models to study the role of epigenetic processes in regulating ...Missing: behavioral | Show results with:behavioral
  8. [8]
    Epigenetic programming by maternal behavior - PubMed
    Moshe Szyf, Michael J Meaney. Affiliation. 1 Douglas Hospital Research ... Rats; Receptors, Glucocorticoid / genetics*; Repressor Proteins / metabolism ...Missing: seminal | Show results with:seminal
  9. [9]
    Genetics, Epigenetic Mechanism - StatPearls - NCBI Bookshelf - NIH
    Epigenetics is the study of heritable and stable changes in gene expression that occur through alterations in the chromosome rather than in the DNA sequence.
  10. [10]
    DNA methylation of BDNF as a biomarker of early-life adversity - PNAS
    Nov 10, 2014 · Our data suggest that BDNF DNA methylation in the blood may be used as a predictor of brain BDNF DNA methylation and gene expression as well as behavioral ...
  11. [11]
    Epigenetic Regulation of BDNF Gene in Response to Stress - PMC
    Further studies examining whether stress affects other epigenetic pathways involved in regulation of the BDNF gene, such as DNA methylation, may provide ...
  12. [12]
    The role of histone modifications: from neurodevelopment to ...
    Jul 6, 2022 · A growing body of evidence suggests that epigenetic mechanisms, such as histone modifications, allow the fine-tuning and coordination of spatiotemporal gene ...
  13. [13]
    A focus on the repressive complex GLP/G9a - ScienceDirect
    Dynamic H3K9 histone methyltransferase activity underlies learning and memory. •. H3K9 methylation underlies changes in neuronal circuitry in addiction. •.
  14. [14]
    H3K9me3 Inhibition Improves Memory, Promotes Spine Formation ...
    Mar 23, 2016 · In this study, we demonstrate that epigenetic regulation of key synaptic proteins may be an underlying, yet reversible, cause of this decline.
  15. [15]
    The role of microRNAs in human neural stem cells, neuronal ...
    Aug 30, 2014 · Among the brain-enriched miRNAs, the functions of miR-124 and miR-9 in promoting neuronal differentiation have been extensively studied ( ...Microrna-Mediated... · Micrornas Interact With Gene... · Micrornas As Tools To...
  16. [16]
    A functional study of miR-124 in the developing neural tube - PMC
    We found that neither inhibition nor ectopic expression of miR-124 had a significant effect on neuronal differentiation. However, by repressing laminin γ1 and ...Missing: seminal papers epigenetics
  17. [17]
    An updated role of microRNA-124 in central nervous system disorders
    May 19, 2015 · Besides effects on neuron fate, miR-124 also contributes to the control of neurite outgrowth during neuronal differentiation possibly by ...Missing: seminal papers epigenetics
  18. [18]
    Epigenetic Regulation by ATP-Dependent Chromatin Remodeling ...
    Each remodeling complex contains a single catalytic ATPase that utilizes ATP hydrolysis to achieve the energy-intensive modulation of chromatin (Figure 1, ...
  19. [19]
    Epigenetic regulation and chromatin remodeling in learning ... - Nature
    Jan 13, 2017 · In this review, we will introduce three major epigenetic processes involved in memory regulation: DNA methylation, histone methylation and histone acetylation.
  20. [20]
    The Emerging Role of ATP-Dependent Chromatin Remodeling in ...
    This review examines how histone-modifying enzymes and chromatin remodelers restructure chromatin to facilitate memory formation.
  21. [21]
    Basic concepts of epigenetics: Impact of environmental signals on ...
    Epigenetics is control over how genomic DNA is expressed, involving DNA methylation, histone modifications, and the epigenome, which is non-static.
  22. [22]
    The epigenotype. 1942 - PubMed
    Dr Waddington proposes the name 'epigenotype.' He here describes some of the general characteristics of an epigenotype, with special reference to the fruit-fly ...
  23. [23]
    Epigenetics: The origins and evolution of a fashionable topic
    Aug 1, 2016 · The “epigenetic landscape” as proposed by Conrad Waddington shows a ball rolling down on an inclined surface with a cascade of branching ridges ...
  24. [24]
    [PDF] Major Findings from Twin Studies of Ability, Personality and Interests.
    Sep 4, 1976 · There are several, lines of evidence that allow the separation of the effects of heredity and environment on behavior. These include studies of ...
  25. [25]
    the Minnesota Study of Twins Reared Apart - PubMed - NIH
    Oct 12, 1990 · Like the prior, smaller studies of monozygotic twins reared apart, about 70% of the variance in IQ was found to be associated with genetic variation.Missing: 1960s- 1970s
  26. [26]
    Prader-Willi Syndrome - GeneReviews® - NCBI Bookshelf - NIH
    ... research on Prader-Willi syndrome (PWS) since the late 1980s. He has been a major contributor to the understanding of the genetics of PWS and genomic imprinting ...
  27. [27]
    Maternal care during infancy regulates the development of neural ...
    Maternal care during infancy serves to “program” behavioral responses to stress in the offspring by altering the development of the neural systems that mediate ...
  28. [28]
    [PDF] Eric R. Kandel - Nobel Lecture
    At a given synapse synaptic plasticity can either be short- or long-lived depending on the number of spaced repe- titions of the learning stimulus, and these ...Missing: epigenetics | Show results with:epigenetics
  29. [29]
    targets for early nutritional effects on epigenetic gene regulation
    Early nutrition affects adult metabolism in humans and other mammals, potentially via persistent alterations in DNA methylation. With viable yellow agouti ...Missing: obesity behavior
  30. [30]
    Persistent epigenetic differences associated with prenatal exposure ...
    Nov 4, 2008 · Here we report that periconceptional exposure to famine during the Dutch Hunger Winter is associated with lower methylation of the IGF2 DMR 6 ...
  31. [31]
    The Honey Bee Epigenomes: Differential Methylation of Brain DNA ...
    Nov 2, 2010 · Over 550 genes show significant methylation differences between queens and workers, revealing the intricate dynamics of methylation patterns.
  32. [32]
    Next generation sequencing based approaches to epigenomics
    Jan 22, 2011 · Next generation sequencing has brought epigenomic studies to the forefront of current research. The power of massively parallel sequencing ...
  33. [33]
    Epigenetic regulation of brain development, plasticity, and response ...
    Aug 6, 2025 · Early-life stress accelerates aging of the DNA methylome. c Early-life stress alters DNA methylation at NR3C1, FKBP5, and BDNF in human and ...
  34. [34]
    Impact of social stress on epigenetics: an updated narrative review
    Jul 30, 2025 · Social stressors can perturb neuronal development, epigenetic age and adult health by modulation of DNA methylation at selected genes.
  35. [35]
    [PDF] BEHAVIORAL EPIGENETICS AND MENTAL HEALTH - JETIR.org
    Behavioral epigenetics is a rapidly evolving interdisciplinary field that explores how life experiences—such as trauma, caregiving, stress ...
  36. [36]
    Epigenetics and the biological basis of gene x environment ...
    This review examines epigenetics as a candidate biological mechanism for gene x environment interactions, with a focus on environmental influences that occur ...
  37. [37]
    Fetal glucocorticoid receptor (Nr3c1) deficiency alters the landscape ...
    Jan 17, 2019 · Prenatal stress decreases Bdnf expression and increases methylation of Bdnf exon IV in rats. Epigenetics 9, 437–447 (2014). Article Google ...
  38. [38]
    Epigenetic Modifications in Stress Response Genes Associated With ...
    Adverse childhood experiences and adult risk factors for age-related disease: depression, inflammation, and clustering of metabolic risk markers. Arch ...
  39. [39]
    Neurogenetics and Epigenetics in Impulsive Behaviour: Impact on ...
    These adverse childhood experiences are linked to persistent changes in stress-related systems and brain regions involved in mood, cognition, and behavior.
  40. [40]
    Exercise impacts brain-derived neurotrophic factor plasticity by ...
    We report that exercise affects histone acetylation and DNA methylation localized to the promoter IV region of the Bdnf gene. Exercise also affects the levels ...
  41. [41]
    Epigenetics, Resilience, Protective Factors and Factors Promoting ...
    Oct 20, 2025 · This body of research opens lines of intervention at the social level that can be reflected at the molecular level, thereby mitigating stress ...
  42. [42]
    IQGAP1/ERK regulates fear memory formation via histone ... - PubMed
    IQGAP1-/- mice exhibited impaired fear memory, decreased levels of phosphorylated ERK1/2 and histone H3S10, decreased acetylation of H3K14, and decreased c-Fos ...
  43. [43]
    Gene–Stress–Epigenetic Regulation of FKBP5 - Nature
    Aug 13, 2015 · The FKBP5 gene is regulated via complex interactions among environmental stressors, FKBP5 genetic variants, and epigenetic modifications of glucocorticoid- ...
  44. [44]
    Glucocorticoid Signaling and Epigenetic Alterations in Stress ...
    Aberrant glucocorticoid signaling has been associated with an increased risk for psychiatric and mood disorders, including schizophrenia, post-traumatic stress ...
  45. [45]
    Parental olfactory experience influences behavior and neural ... - NIH
    Dec 1, 2013 · Olfactory fear conditioning to study descendant generations. We examined whether olfactory fear conditioning of the F0 generation leads ...
  46. [46]
    Intergenerational Effects of Maternal Holocaust Exposure on FKBP5 ...
    Apr 21, 2020 · FKBP5 site 6 methylation was significantly lower in Holocaust offspring than in control subjects, an effect associated with maternal Holocaust exposure in ...
  47. [47]
    Genetic control of non-genetic inheritance in mammals
    Jun 11, 2020 · A paramutation is one example of genetically controlled heritable epigenetic variation which defies Mendel's first law of inheritance, according ...
  48. [48]
    Transgenerationally inherited piRNAs trigger piRNA biogenesis by ...
    We found that transgenerationally inherited piRNAs provide the critical trigger for piRNA production from homologous genomic regions in the next generation.
  49. [49]
    HOW STABLE 'SHOULD' EPIGENETIC MODIFICATIONS BE ...
    Nov 26, 2013 · Epigenetic stability both within and across generations will also depend on the degree and predictability of environmental variation, dispersal ...
  50. [50]
    Epigenetic Echoes: Bridging Nature, Nurture, and Healing Across ...
    This review explores the epigenetic mechanisms that have been correlated with the effects of trauma across generations, including DNA methylation, histone ...Epigenetic Echoes: Bridging... · 2. Dna Methylation And... · 3. Histone Modifications
  51. [51]
    NF-κB mediates Gadd45β expression and DNA demethylation in the ...
    Mice were trained to a single trial auditory plus contextual fear conditioning paradigm that consisted of a 1.5 min baseline followed a single tone (100 Hz, 30 ...
  52. [52]
    Genetic Deletion of gadd45b, a Regulator of Active DNA ...
    Nov 28, 2012 · This study found memory-associated expression of gadd45b in the hippocampus and characterized the behavioral phenotype of gadd45b −/− mice.
  53. [53]
    Histone Deacetylase Inhibitors Enhance Memory and Synaptic ...
    Jun 6, 2007 · Our results suggest that HDAC inhibitors enhance memory processes by the activation of key genes regulated by the CREB:CBP transcriptional complex.
  54. [54]
    Exercise Training Improves Memory Performance in Older Adults
    Acute and regular exercise training induces both short and long-term epigenetic regulations, creating a “functional genome” that consequently leads to ...
  55. [55]
    Hippocampal DNA Methylation, Epigenetic Age, and Spatial ...
    Dec 29, 2022 · We assessed age-related changes in DNA methylation (DNAm) landscape in the rat hippocampus and the correlation of spatial memory with hippocampal DNAm age.
  56. [56]
    Epigenetic Alterations Are Critical for Fear Memory Consolidation ...
    Epigenetic mechanisms, including histone acetylation and DNA methylation, have been widely implicated in hippocampal-dependent learning paradigms.
  57. [57]
    Epigenetic Modifications in Alzheimer's Neuropathology and ...
    May 9, 2019 · Alzheimer's disease (AD) is a neurodegenerative disease characterized by significant impairments in neural synapses and deficiencies in memory.
  58. [58]
    Neurogenetic and Epigenetic Correlates of Adolescent ...
    Neurogenetic and Epigenetic Correlates of Adolescent Predisposition to and Risk for Addictive Behaviors as a Function of Prefrontal Cortex Dysregulation.
  59. [59]
  60. [60]
  61. [61]
  62. [62]
  63. [63]
    Advances in the etiology and neuroimaging of children with attention ...
    This review will present current advancements in the field of attention deficit hyperactivity disorder, including genetics, environmental factors, epigenetics, ...
  64. [64]
  65. [65]
  66. [66]
    Serotonin Transporter Epigenetic Modification Predicts Human Brain ...
    Specifically, we found that increased promoter methylation of the serotonin transporter gene predicted increased threat-related amygdala reactivity and ...Missing: valence pathways
  67. [67]
    Dopamine and serotonin interplay for valence-based spatial learning
    Apr 12, 2022 · The interplay between dopamine (DA) and serotonin, or 5-hydroxytryptamine (5-HT), regulates cognitive functions underpinning decision making; ...
  68. [68]
    Epigenetic Modifications Associated with Maternal Anxiety during ...
    Higher maternal anxiety predicted higher NR3C1 methylation level, explaining 6.7% of the variance of methylation level for NR3C1-CpG3, 10.3% for NR3C1-CpG5 ...
  69. [69]
    Maternal–infant interaction quality is associated with child NR3C1 ...
    Feb 13, 2023 · We hypothesized that the social–emotional quality of maternal–infant interactions are associated with methylation of HPA‐axis gene NR3C1 later in childhood.
  70. [70]
    A narrative review of the epigenetics of post-traumatic stress ...
    Nov 6, 2022 · This paper reviews the epigenetic markers associated with PTSD and its treatment, including candidate genes and epigenome-wide studies.Epigenetic markers in post... · Epigenetic mechanisms... · Challenges and future...
  71. [71]
    Serotonin tranporter methylation and response to cognitive ... - Nature
    Sep 16, 2014 · In this study, we measured DNA methylation upstream of SERT in 116 children with an anxiety disorder, before and after receiving CBT. Change ...
  72. [72]
    Regulation of hippocampal H3 histone methylation by acute ... - PNAS
    This study documents dramatic effects of acute stress and changes in three histone marks that are indicative of time dependent effects of repeated stress that ...
  73. [73]
    Environmental enrichment improves lifelong persistent behavioral ...
    Environmental enrichment improves lifelong persistent behavioral and epigenetic changes induced by early-life stress.
  74. [74]
    Childhood Maltreatment and Longitudinal Epigenetic Aging
    Jul 29, 2024 · This cohort study examines whether childhood exposure to physical and emotional abuse and neglect is associated with the rate of epigenetic ...
  75. [75]
    From trauma to resilience: psychological and epigenetic adaptations ...
    Jul 18, 2025 · Our findings revealed that descendants exhibited significantly lower general attachment avoidance, and a DNA methylation pattern associated with ...
  76. [76]
    BDNF genetic variants and methylation: effects on cognition in major ...
    Oct 21, 2019 · Research on methylation of promoter IV has also shown increased methylation levels in patients with depression when compared to those in healthy ...
  77. [77]
    BDNF Unveiled: Exploring Its Role in Major Depression Disorder ...
    Aug 3, 2023 · In this review, we will focus on the role of BDNF in major depression disorder serotonergic imbalance and associated stress conditions.
  78. [78]
    Effect of mood stabilizers on DNA methylation in human ...
    Our comprehensive analysis unexpectedly revealed a profound epigenetic effect of lithium compared with the two other mood stabilizers. In addition, we found ...
  79. [79]
    A bipolar disorder-associated missense variant alters adenylyl ...
    Jul 13, 2024 · A bipolar disorder-associated missense variant alters adenylyl cyclase 2 activity and promotes mania-like behavior. Paromita Sen ORCID: orcid ...<|separator|>
  80. [80]
    Epigenetic dysregulation of SHANK3 in brain tissues from ... - NIH
    This study is the first to find altered methylation patterns in SHANK3 in ASD brain samples. Our finding provides evidence to support an alternative approach.
  81. [81]
    DNA Methylation in Autism Spectrum Disorders: Biomarker or ...
    In 15% of ASD brain tissues, elevated methylation was concentrated in CGI-2 and CGI-4. This indicated that SHANK3 is a useful biomarker for examining the role ...
  82. [82]
    The association between prenatal famine, DNA methylation and ...
    Sep 16, 2023 · One epigenetic mechanism that has been associated with mental disorders and undernutrition is alterations in DNA methylation. The effect of ...
  83. [83]
    Epigenetics in Prader-Willi Syndrome - PMC - PubMed Central
    PWS is both a genetic and epigenetic disorder, mapping the imprinted chromosomal domain of 15q11.2-13.3. Common to all cases of PWS is the absence of an ...
  84. [84]
    Mechanism of EHMT2-mediated genomic imprinting associated with ...
    Jul 3, 2025 · Prader-Willi Syndrome (PWS) is caused by the loss of expression of paternally expressed genes in the human 15q11.2-q13 imprinting domain.Missing: mood | Show results with:mood
  85. [85]
    Genetic, Epigenetic, and Hormonal Regulation of Stress Phenotypes ...
    Mar 26, 2025 · This review explores the crucial roles of stress, genetics, epigenetics, and hormones in shaping susceptibility and resilience to mood disorders.
  86. [86]
    Genetic and Epigenetic Markers of Lithium Response - PMC - NIH
    The mood stabilizer lithium represents a cornerstone in the long term treatment of bipolar disorder (BD), although with substantial interindividual variability ...
  87. [87]
    The valproic acid-induced rodent model of autism - PubMed
    This model might better represent the many cases of idiopathic autism which are of environmental/epigenetic origins than do transgenic models carrying mutations ...
  88. [88]
    The clinical use of epigenetics in psychiatry: a narrative review of ...
    By exploring the role of clinically relevant epigenetic genes, we review the role of the epigenome in the context of psychiatric disorders and human behavior; ...Missing: JETIR | Show results with:JETIR
  89. [89]
    Epigenetic mechanisms in drug addiction - PMC - NIH
    Changes in gene expression in brain reward regions are thought to contribute to the pathogenesis and persistence of drug addiction.
  90. [90]
    ΔFosB Mediates Epigenetic Desensitization of the c-fos Gene After ...
    Jul 16, 2008 · We show that accumulation of ΔFosB in striatum after chronic amphetamine treatment desensitizes c-fos mRNA induction to a subsequent drug dose.
  91. [91]
    Effect of short-term prescription opioids on DNA methylation of the ...
    Jun 3, 2020 · The present study provides evidence that the hypermethylation of the OPRM1 promoter is in response to opioid use and that epigenetic differences in OPRM1 and ...
  92. [92]
    DAT1 Gene Methylation as an Epigenetic Biomarker in Attention ...
    May 11, 2020 · ADHD candidate genes have been hypothesized, including dopamine and serotonin transporters and the dopamine D4 and D5 receptors.
  93. [93]
    Transcriptional and Epigenetic Mechanisms of Addiction - PMC
    histone tail modification, DNA methylation, and microRNAs — and summarize the major ...Missing: anticipation | Show results with:anticipation
  94. [94]
    (PDF) Adolescent Substance Abuse, Transgenerational ...
    Aug 6, 2025 · In addition, epigenetic modifications of the genome following drug exposure will be discussed as emerging evidence of the underlying adverse ...
  95. [95]
    Adolescent nicotine exposure and persistent neurocircuitry changes
    Aug 29, 2025 · Nicotine use, and particularly nicotine use during adolescence is associated with an increased risk of addiction and mood disorders in adulthood ...
  96. [96]
    The Inhibition of Histone Deacetylases Reduces the Reinstatement ...
    In conclusion, our results strongly suggest that treatment aimed at modulating epigenetic regulations, especially the pharmacological inhibition of HDAC ...
  97. [97]
    Functional CpG Methylation System in a Social Insect | Science
    We present a bioinformatic, molecular, and biochemical characterization of a functional DNA methylation system in an insect.
  98. [98]
    DNA methylation is widespread and associated with differential ...
    Jul 7, 2009 · Recent research has identified a functional DNA methylation system in a social insect, the honeybee, Apis mellifera (6). Social insects are ...Missing: 2010s | Show results with:2010s
  99. [99]
    Histone deacetylase inhibitor activity in royal jelly might facilitate ...
    Thus, the epigenetic regulation of queen bee development is likely driven, in part, by HDACi activity in royal jelly.
  100. [100]
    The role of epigenetics, particularly DNA methylation, in the ...
    Apr 19, 2021 · Most social insect species examined have methylation, as evidenced by the presence of Dnmt-coding genes in their genomes, and evidence of ...
  101. [101]
    Regulation of soldier caste differentiation by microRNAs in ...
    Feb 29, 2024 · This study demonstrates that DEmiRs and DEGs form a complex network regulating soldier caste differentiation in termites.Rna Extraction And... · Sequencing Quality · Table 1. Read Counts And...Missing: epigenetics | Show results with:epigenetics
  102. [102]
    CRISPR/Cas9-Based Genome Editing Strategies in Honey Bees
    Sep 18, 2023 · CRISPR/Cas9-based gene editing strategies and current studies in honey bees were compiled in this study.
  103. [103]
    Oxytocin system gene methylation is associated with empathic ...
    Linear mixed effects analyses demonstrated that lower methylation levels in the OXT and OXTR genes were related to enhanced empathic responses. The effect of ...
  104. [104]
    The role of oxytocin receptor gene (OXTR) DNA methylation (DNAm ...
    May 29, 2018 · Oxytocin enhances the experience of attachment ... Oxytocin receptor genetic variation related to empathy and stress reactivity in humans.
  105. [105]
  106. [106]
    Epigenetics of discordant monozygotic twins: implications for disease
    Jul 31, 2014 · Analysis of disease-discordant MZ twins has been successfully used to study epigenetic mechanisms in aging, cancer, autoimmune disease, psychiatric, ...
  107. [107]
    Epigenetic Echoes: Bridging Nature, Nurture, and Healing Across ...
    This review explores the epigenetic mechanisms that have been correlated with the effects of trauma across generations.
  108. [108]
    Transgenerational epigenetic inheritance: a critical perspective
    Aug 29, 2024 · Transgenerational epigenetic inheritance in humans and other vertebrates has been controversial for over 150 years and remains so.
  109. [109]
    Epigenetic modulation of social cognition: exploring the impact of ...
    Jan 27, 2025 · This study investigates how these methylation differences affect social cognition across sexes, focusing on both perceptual and inferential cognitive levels.
  110. [110]
    Epigenetics in Psychiatry: Implications for Genomics, Evolution,
    This article provides a comprehensive overview of the role of epigenetics in psychiatry, discussing its implications for genomics, evolution, inheritance ...
  111. [111]
    Epigenome-Wide Associations of Placental DNA Methylation and ...
    Jul 21, 2023 · Our results further suggest sex-specific associations between placental DNA methylation and child emotional and behavioral difficulties. Further ...2. Results · 2.4. Epigenome-Wide... · 4.3. Placental Dna...
  112. [112]
    Maternal caregiving and DNA methylation in human infants and ...
    Oct 17, 2019 · 17 The seminal work by Michael Meaney et al provided the first ... Broad epigenetic signature of maternal care in the brain of adult rats.