Abstract
The early years of life are a critical period for brain development, encompassing high sensitivity to adverse experiences. Early life stress (ELS) is known to “scar” the brain and shape mental health trajectories later in life. Still, a great percentage of children faced with ELS develop adaptive competencies that maintain normal physiological and behavioral function across the lifespan, a process referred to as resilience. Work in humans and rodent models has demonstrated that resilience is an active process mediated largely by the induction of unique molecular, cellular, and circuit adaptations. In this review, we highlight evidence from rodent studies exploring the behavioral, circuit, cellular, and molecular effects of ELS and discuss resilient phenotypes that emerge from specific ELS paradigms. To this end, we focus on models comprising ELS exposure within pre-weening and adolescence. We next address critical factors that influence the effects of ELS, such as behavioral readouts, environmental conditions, or sex differences, and we compare these findings in light of human studies. Finally, we advocate for the use of novel and more sophisticated behavioral tasks for rodents that capture, at least in part, resilient phenotypes observed in humans and that can be directly linked to specific brain circuits.
This is a preview of subscription content, access via your institution
Access options
Subscribe to this journal
Receive 13 print issues and online access
$259.00 per year
only $19.92 per issue
Buy this article
- Purchase on SpringerLink
- Instant access to full article PDF
Prices may be subject to local taxes which are calculated during checkout




Similar content being viewed by others

References
Teicher MH, Samson JA. Annual research review: enduring neurobiological effects of childhood abuse and neglect. J Child Psychol Psychiatry. 2016;57:241–66.
Gee DG. Early adversity and development: parsing heterogeneity and identifying pathways of risk and resilience. Am J Psychiatry. 2021;178:998–1013.
Green JG, McLaughlin KA, Berglund PA, Gruber MJ, Sampson NA, Zaslavsky AM, et al. Childhood adversities and adult psychiatric disorders in the National Comorbidity Survey Replication I: associations with first onset of DSM-IV disorders. Arch Gen Psychiatry. 2010;67:113–23.
Kessler RC, McLaughlin KA, Green JG, Gruber MJ, Sampson NA, Zaslavsky AM, et al. Childhood adversities and adult psychopathology in the WHO World Mental Health Surveys. Br J Psychiatry. 2010;197:378–85.
Scott KM, McLaughlin KA, Smith DA, Ellis PM. Childhood maltreatment and DSM-IV adult mental disorders: comparison of prospective and retrospective findings. Br J Psychiatry. 2012;200:469–75.
Walsh WA, Dawson J, Mattingly MJ. How are we measuring resilience following childhood maltreatment? Is the research adequate and consistent? What is the impact on research, practice, and policy?. Trauma Violence Abuse. 2010;11:27–41.
Yu W, Zhu F, Foo MD, Wiklund J. What does not kill you makes you stronger: entrepreneurs’ childhood adversity, resilience, and career success. J Bus Res. 2022;151:40–55.
Bonanno GA. Loss, trauma, and human resilience: Have we underestimated the human capacity to thrive after extremely aversive events? Psychol Trauma Theory, Res Pract Policy. 2008;S:101–13.
Bonanno GA, Papa A, O’Neill K. Loss and human resilience. Appl Prev Psychol. 2001;10:193–206.
Southwick SM, Charney DS. Resilience: the science of mastering life’s greatest challenges. Cambridge: Cambridge University Press; 2012.
Ryff C, Friedman E, Fuller-Rowell T, Love G, Miyamoto Y, Morozink J, et al. Varieties of resilience in MIDUS. Soc Pers Psychol Compass. 2012;6:792–806.
Cromer KR, Sachs-Ericsson N. The association between childhood abuse, PTSD, and the occurrence of adult health problems: moderation via current life stress. J Trauma Stress. 2006;19:967–71.
Torres-Berrío A, Issler O, Parise EM, Nestler EJ. Unraveling the epigenetic landscape of depression: focus on early life stress. Dialogues Clin Neurosci. 2019;21:341–57.
Baram TZ, Davis EP, Obenaus A, Sandman CA, Small SL, Solodkin A, et al. Fragmentation and unpredictability of early-life experience in mental disorders. Am J Psychiatry. 2012;169:907–15.
Sheridan MA, McLaughlin KA. Dimensions of early experience and neural development: deprivation and threat. Trends Cogn Sci. 2014;18:580–85.
Felitti VJ, Anda RF, Nordenberg D, Williamson DF, Spitz AM, Edwards V. et al. Relationship of childhood abuse and household dysfunction to many of the leading causes of death in adults. The Adverse Childhood Experiences (ACE) study. Am J Prev Med. 1998;14:245–58.
Merrick MT, Ford DC, Ports KA, Guinn AS. Prevalence of adverse childhood experiences From the 2011-2014 behavioral risk factor surveillance system in 23 states. JAMA Pediatr. 2018;172:1038–44.
Hammen C. Adolescent depression: stressful interpersonal contexts and risk for recurrence. Curr Dir Psychol Sci. 2009;18:200–04.
Hammen C, Henry R, Daley SE. Depression and sensitization to stressors among young women as a function of childhood adversity. J Consult Clin Psychol. 2000;68:782–7.
Rudolph KD, Hammen C, Burge D, Lindberg N, Herzberg D, Daley SE. Toward an interpersonal life-stress model of depression: the developmental context of stress generation. Dev Psychopathol. 2000;12:215–34.
Teicher MH, Anderson CM, Ohashi K, Khan A, McGreenery CE, Bolger EA, et al. Differential effects of childhood neglect and abuse during sensitive exposure periods on male and female hippocampus. Neuroimage. 2018;169:443–52.
Dunn EC, Gilman SE, Willett JB, Slopen NB, Molnar BE. The impact of exposure to interpersonal violence on gender differences in adolescent-onset major depression: results from the National Comorbidity Survey Replication (NCS-R). Depress Anxiety. 2012;29:392–99.
Michael C, Gard AM, Tillem S, Hardi FA, Dunn EC, Smith ADAC, et al. Developmental timing of associations among parenting, brain architecture, and mental health. JAMA Pediatr. 2024;178:1326–36.
Jiang T, Yakin S, Crocker J, Way BM. Perceived social support-giving moderates the association between social relationships and interleukin-6 levels in blood. Brain Behav Immun. 2022;100:25–28.
Morozink JA, Friedman EM, Coe CL, Ryff CD. Socioeconomic and psychosocial predictors of interleukin-6 in the MIDUS national sample. Health Psychol. 2010;29:626–35.
Park AT, Tooley UA, Leonard JA, Boroshok AL, McDermott CL, Tisdall MD, et al. Early childhood stress is associated with blunted development of ventral tegmental area functional connectivity. Dev Cogn Neurosci. 2021;47:100909.
Marusak HA, Hatfield JRB, Thomason ME, Rabinak CA. Reduced ventral tegmental area–hippocampal connectivity in children and adolescents exposed to early threat. Biol Psychiatry Cogn Neurosci Neuroimaging. 2017;2:130–37.
Sinha R, Lacadie CM, Constable RT, Seo D. Dynamic neural activity during stress signals resilient coping. Proc Natl Acad Sci USA. 2016;113:8837–42.
McEwen BS, Gray J, Nasca C. Recognizing resilience: learning from the effects of stress on the brain. Neurobiol Stress. 2015;1:1–11.
Norbury A, Seeley SH, Perez-Rodriguez MM, Feder A. Functional neuroimaging of resilience to trauma: convergent evidence and challenges for future research. Psychol Med. 2023;53:3293–305.
Libé-Philippot B, Vanderhaeghen P. Cellular and molecular mechanisms linking human cortical development and evolution. Annu Rev Genet. 2021;55:555–81.
Zeiss CJ. Comparative milestones in rodent and human postnatal central nervous system development. Toxicol Pathol. 2021;49:1368–73.
Wallace JL, Pollen AA. Human neuronal maturation comes of age: cellular mechanisms and species differences. Nat Rev Neurosci. 2024;25:7–29.
Prince GS, Reynolds M, Martina V, Sun H. Gene-environmental regulation of the postnatal post-mitotic neuronal maturation. Trends Genet. 2024;40:480–94.
Knudsen EI. Sensitive periods in the development of the brain and behavior. J Cogn Neurosci. 2004;16:1412–25.
Peña CJ. Early-life stress sensitizes response to future stress: evidence and mechanisms. Neurobiol Stress. 2025;35:100716.
Dobbing J, Sands J. Comparative aspects of the brain growth spurt. Early Hum Dev. 1979;3:79–83.
Sengupta P. The laboratory rat: relating its age with human’s. Int J Prev Med. 2013;4:624–30.
Berry KP, Nedivi E. Spine dynamics: Are they all the same? Neuron. 2017;96:43–55.
Kalemaki K, Velli A, Christodoulou O, Denaxa M, Karagogeos D, Sidiropoulou K. The developmental changes in intrinsic and synaptic properties of prefrontal neurons enhance local network activity from the second to the third postnatal weeks in mice. Cereb Cortex. 2021;32:3633–50.
Rurak GM, Simard S, Freitas-Andrade M, Lacoste B, Charih F, Van Geel A, et al. Sex differences in developmental patterns of neocortical astroglia: a mouse translatome database. Cell Rep. 2022;38:110310.
Farhy-Tselnicker I, Allen NJ. Astrocytes, neurons, synapses: a tripartite view on cortical circuit development. Neural Dev. 2018;13:7.
Matcovitch-Natan O, Winter DR, Giladi A, Vargas Aguilar S, Spinrad A, Sarrazin S, et al. Microglia development follows a stepwise program to regulate brain homeostasis. Science. 2016;353:aad8670.
Li Q, Cheng Z, Zhou L, Darmanis S, Neff NF, Okamoto J, et al. Developmental Heterogeneity of microglia and brain myeloid cells revealed by deep single-cell RNA sequencing. Neuron. 2019;101:207–23.e10.
Kessaris N, Fogarty M, Iannarelli P, Grist M, Wegner M, Richardson WD. Competing waves of oligodendrocytes in the forebrain and postnatal elimination of an embryonic lineage. Nat Neurosci. 2006;9:173–9.
Teissier A, Le Magueresse C, Olusakin J, Andrade da Costa BLS, De Stasi AM, Bacci A, et al. Early-life stress impairs postnatal oligodendrogenesis and adult emotional behaviour through activity-dependent mechanisms. Mol Psychiatry. 2020;25:1159–74.
Wang C, Xue Y, Markovic T, Li H, Wang S, Zhong Y, et al. Blood–brain-barrier-crossing lipid nanoparticles for mRNA delivery to the central nervous system. Nat Mater. 2025 https://doi.org/10.1038/s41563-024-02114-5.
Solarz A, Majcher-Maślanka I, Chocyk A. Effects of early-life stress and sex on blood–brain barrier permeability and integrity in juvenile and adult rats. Dev Neurobiol. 2021;81:861–76.
Milbocker KA, Campbell TS, Collins N, Kim S, Smith IF, Roth TL, et al. Glia-driven brain circuit refinement is altered by early-life adversity: behavioral outcomes. Front Behav Neurosci. 2021;15:786234.
Nestler EJ, Russo SJ. Neurobiological basis of stress resilience. Neuron. 2024;112:1911–29.
McEwen BS, Bowles NP, Gray JD, Hill MN, Hunter RG, Karatsoreos IN, et al. Mechanisms of stress in the brain. Nat Neurosci. 2015;18:1353–63.
Schmidt M, Enthoven L, van der Mark M, Levine S, de Kloet ER, Oitzl MS. The postnatal development of the hypothalamic–pituitary–adrenal axis in the mouse. Int J Dev Neurosci. 2003;21:125–32.
Bolton JL, Short AK, Othy S, Kooiker CL, Shao M, Gunn BG, et al. Early stress-induced impaired microglial pruning of excitatory synapses on immature CRH-expressing neurons provokes aberrant adult stress responses. Cell Rep. 2022;38:110600.
Bolton JL, Short AK, Simeone KA, Daglian J, Baram TZ. Programming of stress-sensitive neurons and circuits by early-life experiences. Front Behav Neurosci. 2019;13:30.
Anderson KN, Swedo EA, Trinh E, Ray CM, Krause KH, Verlenden JV, et al. Adverse childhood experiences during the COVID-19 pandemic and associations with poor mental health and suicidal behaviors among high school students—Adolescent Behaviors and Experiences Survey, United States, January-June 2021. MMWR Morb Mortal Wkly Rep. 2022;71:1301–05.
Kessler RC, Amminger GP, Aguilar-Gaxiola S, Alonso J, Lee S, Üstün TB. Age of onset of mental disorders: a review of recent literature. Curr Opin Psychiatry. 2007;20:359-64
Reynolds LM, Yetnikoff L, Pokinko M, Wodzinski M, Epelbaum JG, Lambert LC, et al. Early adolescence is a critical period for the maturation of inhibitory behavior. Cereb Cortex. 2018;29:3676–86.
Hoops D, Flores C. Making dopamine connections in adolescence. Trends Neurosci. 2017;40:709–19.
Floresco SB. The nucleus accumbens: an interface between cognition, emotion, and action. Annu Rev Psychol. 2015;66:25–52.
Brenhouse HC, Andersen SL. Developmental trajectories during adolescence in males and females: a cross-species understanding of underlying brain changes. Neurosci Biobehav Rev. 2011;35:1687–703.
Somerville LH, Hare T, Casey BJ. Frontostriatal maturation predicts cognitive control failure to appetitive cues in adolescents. J Cogn Neurosci. 2011;23:2123–34.
Klune CB, Goodpaster CM, Gongwer MW, Gabriel CJ, An J, Chen R, et al. Developmentally distinct architectures in top–down pathways controlling threat avoidance. Nat Neurosci. 2025;28:823–35.
Nelson EE, Jarcho JM, Guyer AE. Social re-orientation and brain development: an expanded and updated view. Dev Cogn Neurosci. 2016;17:118–27.
Chein J, Albert D, O’Brien L, Uckert K, Steinberg L. Peers increase adolescent risk taking by enhancing activity in the brain’s reward circuitry. Dev Sci. 2011;14:F1–10.
McCutcheon JE, Marinelli M. Age matters. Eur J Neurosci. 2009;29:997–1014.
Ghasemi A, Jeddi S, Kashfi K. The laboratory rat: age and body weight matter. EXCLI J. 2021;20:1431–45.
Sturman DA, Moghaddam B. The neurobiology of adolescence: changes in brain architecture, functional dynamics, and behavioral tendencies. Neurosci Biobehav Rev. 2011;35:1704–12.
Sisk CL, Zehr JL. Pubertal hormones organize the adolescent brain and behavior. Front Neuroendocrinol. 2005;26:163–74.
Sisk CL, Foster DL. The neural basis of puberty and adolescence. Nat Neurosci. 2004;7:1040–47.
Lewis EM, Barnett JF Jr., Freshwater L, Hoberman AM, Christian MS. Sexual maturation data for Crl Sprague-Dawley rats: criteria and confounding factors. Drug Chem Toxicol. 2002;25:437–58.
Semple BD, Blomgren K, Gimlin K, Ferriero DM, Noble-Haeusslein LJ. Brain development in rodents and humans: Identifying benchmarks of maturation and vulnerability to injury across species. Prog Neurobiol. 2013;106-107:1–16.
Yang Y, Cheng Z, Tang H, Jiao H, Sun X, Cui Q, et al. Neonatal maternal separation impairs prefrontal cortical myelination and cognitive functions in rats through activation of Wnt signaling. Cereb Cortex. 2017;27:2871–84.
Casey BJ, Getz S, Galvan A. The adolescent brain. Dev Rev. 2008;28:62–77.
Gogtay N, Giedd JN, Lusk L, Hayashi KM, Greenstein D, Vaituzis AC, et al. Dynamic mapping of human cortical development during childhood through early adulthood. Proc Natl Acad Sci USA. 2004;101:8174–79.
Kolk SM, Rakic P. Development of prefrontal cortex. Neuropsychopharmacology. 2022;47:41–57.
Chad JA, Lebel C. Can gray matter loss in early adolescence be explained by white matter growth? Hum Brain Mapp. 2024;45:e26758.
Sowell ER, Peterson BS, Thompson PM, Welcome SE, Henkenius AL, Toga AW. Mapping cortical change across the human life span. Nat Neurosci. 2003;6:309–15.
Manitt C, Mimee A, Eng C, Pokinko M, Stroh T, Cooper HM, et al. The netrin receptor DCC is required in the pubertal organization of mesocortical dopamine circuitry. J Neurosci. 2011;31:8381–94.
Cuesta S, Nouel D, Reynolds LM, Morgunova A, Torres-Berrío A, White A, et al. Dopamine axon targeting in the nucleus accumbens in adolescence requires Netrin-1. Front Cell Dev Biol. 2020;8:487.
Vassilev P, Pantoja-Urban AH, Giroux M, Nouel D, Hernandez G, Orsini T, et al. Unique effects of social defeat stress in adolescent male mice on the Netrin-1/DCC pathway, prefrontal cortex dopamine and cognition. eNeuro. 2021;8:0045-21:ENEURO.0045-21.
Majcher-Maślanka I, Solarz A, Wędzony K, Chocyk A. The effects of early-life stress on dopamine system function in adolescent female rats. Int J Dev Neurosci. 2017;57:24–33.
Chocyk A, Przyborowska A, Dudys D, Majcher I, Maćkowiak M, Wędzony K. The impact of maternal separation on the number of tyrosine hydroxylase-expressing midbrain neurons during different stages of ontogenesis. Neuroscience. 2011;182:43–61.
Tseng KY, O’Donnell P. Dopamine modulation of prefrontal cortical interneurons changes during adolescence. Cereb Cortex. 2007;17:1235–40.
Tseng KY, O’Donnell P. Dopamine–glutamate interactions controlling prefrontal cortical pyramidal cell excitability involve multiple signaling mechanisms. J Neurosci. 2004;24:5131–39.
Murthy S, Gould E. Early life stress in rodents: animal models of illness or resilience? Front Behav Neurosci. 2018;12:157.
Demaestri C, Gallo M, Mazenod E, Hong AT, Arora H, Short AK, et al. Resource scarcity but not maternal separation provokes unpredictable maternal care sequences in mice and both upregulate Crh-associated gene expression in the amygdala. Neurobiol Stress. 2022;20:100484.
Demaestri C, Pan T, Critz M, Ofray D, Gallo M, Bath KG. Type of early life adversity confers differential, sex-dependent effects on early maturational milestones in mice. Horm Behav. 2020;124:104763.
Waters RC, Gould E. Early life adversity and neuropsychiatric disease: differential outcomes and translational relevance of rodent models. Front Syst Neurosci. 2022;16:860847.
Ader R, Tatum R, Beels CC. Social factors affecting emotionality and resistance to disease in animals: I. Age of separation from the mother and susceptibility to gastric ulcers in the rat. J Comp Physiol Psychol. 1960;53:446–54.
Raineki C, Moriceau S, Sullivan RM. Developing a neurobehavioral animal model of infant attachment to an abusive caregiver. Biol Psychiatry. 2010;67:1137–45.
Moriceau S, Shionoya K, Jakubs K, Sullivan RM. Early-life stress disrupts attachment learning: the role of amygdala corticosterone, locus ceruleus corticotropin releasing hormone, and olfactory bulb norepinephrine. J Neurosci. 2009;29:15745–55.
Moriceau S, Sullivan RM. Maternal presence serves as a switch between learning fear and attraction in infancy. Nat Neurosci. 2006;9:1004–6.
Raineki C, Cortés MR, Belnoue L, Sullivan RM. Effects of early-life abuse differ across development: infant social behavior deficits are followed by adolescent depressive-like behaviors mediated by the amygdala. J Neurosci. 2012;32:7758–65.
George ED, Bordner KA, Elwafi HM, Simen AA. Maternal separation with early weaning: a novel mouse model of early life neglect. BMC Neurosci. 2010;11:123.
Gruss M, Braun K, Frey JU, Korz V. Maternal separation during a specific postnatal time window prevents reinforcement of hippocampal long-term potentiation in adolescent rats. Neuroscience. 2008;152:1–7.
Authement ME, Kodangattil JN, Gouty S, Rusnak M, Symes AJ, Cox BM, et al. Histone deacetylase inhibition rescues maternal deprivation-induced GABAergic metaplasticity through restoration of AKAP signaling. Neuron. 2015;86:1240–52.
Sullivan RM, Landers M, Yeaman B, Wilson DA. Good memories of bad events in infancy. Nature. 2000;407:38–9.
Peña CJ, Kronman HG, Walker DM, Cates HM, Bagot RC, Purushothaman I, et al. Early life stress confers lifelong stress susceptibility in mice via ventral tegmental area OTX2. Science. 2017;356:1185–88.
Peña CJ, Nestler EJ, Bagot RC. Environmental programming of susceptibility and resilience to stress in adulthood in male mice. Front Behav Neurosci. 2019;13:40.
Huot RL, Gonzalez ME, Ladd CO, Thrivikraman KV, Plotsky PM. Foster litters prevent hypothalamic-pituitary-adrenal axis sensitization mediated by neonatal maternal separation. Psychoneuroendocrinology. 2004;29:279–89.
Gilles EE, Schultz L, Baram TZ. Abnormal corticosterone regulation in an immature rat model of continuous chronic stress. Pediatr Neurol. 1996;15:114–19.
Ivy AS, Brunson KL, Sandman C, Baram TZ. Dysfunctional nurturing behavior in rat dams with limited access to nesting material: a clinically relevant model for early-life stress. Neuroscience. 2008;154:1132–42.
Rice CJ, Sandman CA, Lenjavi MR, Baram TZ. A novel mouse model for acute and long-lasting consequences of early life stress. Endocrinology. 2008;149:4892–900.
Birnie MT, Baram TZ. The evolving neurobiology of early-life stress. Neuron. 2025;113:1474–90
Molet J, Heins K, Zhuo X, Mei YT, Regev L, Baram TZ, et al. Fragmentation and high entropy of neonatal experience predict adolescent emotional outcome. Transl Psychiatry. 2016;6:e702.
Eck SR, Ardekani CS, Salvatore M, Luz S, Kim ED, Rogers CM, et al. The effects of early life adversity on growth, maturation, and steroid hormones in male and female rats. Eur J Neurosci. 2020;52:2664–80.
Sanchez EO, Bavley CC, Deutschmann AU, Carpenter R, Peterson DR, Karbalaei R. et al. “Early life adversity promotes resilience to opioid addiction-related phenotypes in male rats and sex-specific transcriptional changes”: Correction. Proc Natl Acad Sci USA. 2022;119:1
Peña CJ, Smith M, Ramakrishnan A, Cates HM, Bagot RC, Kronman HG, et al. Early life stress alters transcriptomic patterning across reward circuitry in male and female mice. Nat Commun. 2019;10:5098.
Kronman H, Torres-Berrío A, Sidoli S, Issler O, Godino A, Ramakrishnan A, et al. Long-term behavioral and cell-type-specific molecular effects of early life stress are mediated by H3K79me2 dynamics in medium spiny neurons. Nat Neurosci. 2021;24:667–76.
Balouek JA, McLain CA, Minerva AR, Rashford RL, Bennett SN, Rogers FD, et al. Reactivation of early-life stress-sensitive neuronal ensembles contributes to lifelong stress hypersensitivity. J Neurosci. 2023;43:5996–6009.
Bennett SN, Chang AB, Rogers FD, Jones P, Peña CJ. Thyroid hormones mediate the impact of early-life stress on ventral tegmental area gene expression and behavior. Horm Behav. 2024;159:105472.
Parel ST, Bennett SN, Cheng CJ, Timmermans OC, Fiori LM, Turecki G, et al. Transcriptional signatures of early-life stress and antidepressant treatment efficacy. Proc Natl Acad Sci USA. 2023;120:e2305776120.
Guayasamin M, Depaauw-Holt LR, Adedipe II, Ghenissa O, Vaugeois J, Duquenne M, et al. Early-life stress induces persistent astrocyte dysfunction associated with fear generalisation. eLife. 2025;13:RP99988.
Torres-Berrío A, Estill M, Patel V, Ramakrishnan A, Kronman H, Minier-Toribio A, et al. Mono-methylation of lysine 27 at histone 3 confers lifelong susceptibility to stress. Neuron. 2024;112:2973–89.e10.
Curley JP, Champagne FA. Influence of maternal care on the developing brain: mechanisms, temporal dynamics and sensitive periods. Front Neuroendocrinol. 2016;40:52–66.
Champagne FA, Francis DD, Mar A, Meaney MJ. Variations in maternal care in the rat as a mediating influence for the effects of environment on development. Physiol Behav. 2003;79:359–71.
Lapp HE, Salazar M, Champagne FA. Postnatal rearing environment alters pup cues for caregiver-offspring interactions. Horm Behav. 2024;165:105630.
van der Zee YY, Eijssen LMT, Mews P, Ramakrishnan A, Alvarez K, Lardner CK, et al. Blood miR-144-3p: a novel diagnostic and therapeutic tool for depression. Mol Psychiatry. 2022;27:4536–49.
Jensen Peña C, Champagne FA. Implications of temporal variation in maternal care for the prediction of neurobiological and behavioral outcomes in offspring. Behav Neurosci. 2013;127:33–46.
Hisey EE, Fritsch EL, Newman EL, Ressler KJ, Kangas BD, Carlezon WA. Early life stress in male mice blunts responsiveness in a translationally-relevant reward task. Neuropsychopharmacology. 2023;48:1752–59.
Pantoja-Urbán AH, Richer S, Mittermaier A, Giroux M, Nouel D, Hernandez G, et al. Gains and losses: resilience to social defeat stress in adolescent female mice. Biol Psychiatry. 2024;95:37–47.
Mayeaux MR, Newman EL, Ressler KJ, Hisey EE. Chronic social defeat stress in early adolescent male mice. J Vis Exp. 2025;215:e67113.
Krishnan V, Han M-H, Graham DL, Berton O, Renthal W, Russo SJ, et al. Molecular adaptations underlying susceptibility and resistance to social defeat in brain reward regions. Cell. 2007;131:391–404.
Golden SA, Covington HE, Berton O, Russo SJ. A standardized protocol for repeated social defeat stress in mice. Nat Protoc. 2011;6:1183–91.
Pantoja-Urbán AH, Richer S, Giroux M, Nouel D, Flores C. Social defeat stress model for adolescent C57BL/6 male and female mice. J Vis Exp. 2024;205:e66455.
Shen M, Song Z, Wang J-H. microRNA and mRNA profiles in the amygdala are associated with stress-induced depression and resilience in juvenile mice. Psychopharmacology. 2019;236:2119–42.
Gyles TM, Nestler EJ, Parise EM. Advancing preclinical chronic stress models to promote therapeutic discovery for human stress disorders. Neuropsychopharmacology. 2024;49:215–26.
Stedenfeld KA, Clinton SM, Kerman IA, Akil H, Watson SJ, Sved AF. Novelty-seeking behavior predicts vulnerability in a rodent model of depression. Physiol Behav. 2011;103:210–6.
Porsolt RD, Bertin A, Jalfre M. Behavioural despair” in rats and mice: strain differences and the effects of imipramine. Eur J Pharmacol. 1978;51:291–94.
Samuels BA, Hen R. Novelty-suppressed feeding in the mouse. In: Gould TD, editor. Mood and anxiety related phenotypes in mice: characterization using behavioral tests, Vol. II. Totowa, NJ: Humana Press; 2011. p. 107–21.
Blasco-Serra A, González-Soler EM, Cervera-Ferri A, Teruel-Martí V, Valverde-Navarro AA. A standardization of the Novelty-Suppressed Feeding Test protocol in rats. Neurosci Lett. 2017;658:73–8.
Kulkarni SK, Singh K, Bishnoi M. Elevated zero maze: a paradigm to evaluate antianxiety effects of drugs. Methods Find Exp Clin Pharmacol. 2007;29:343–8.
Walf AA, Frye CA. The use of the elevated plus maze as an assay of anxiety-related behavior in rodents. Nat Protoc. 2007;2:322–28.
Bogdanova OV, Kanekar S, D’Anci KE, Renshaw PF. Factors influencing behavior in the forced swim test. Physiol Behav. 2013;118:227–39.
Berton O, McClung CA, DiLeone RJ, Krishnan V, Renthal W, Russo SJ, et al. Essential role of BDNF in the mesolimbic dopamine pathway in social defeat stress. Science. 2006;311:864–68.
Durand-de Cuttoli R, Martínez-Rivera FJ, Li L, Minier-Toribio A, Holt LM, Cathomas F, et al. Distinct forms of regret linked to resilience versus susceptibility to stress are regulated by region-specific CREB function in mice. Sci Adv. 2022;8:eadd5579.
Durand-de Cuttoli R, Martínez-Rivera FJ, Li L, Minier-Toribio A, Dong Z, Cai DJ, et al. A double hit of social and economic stress in mice precipitates changes in decision-making strategies. Biol Psychiatry. 2024;96:67–78.
Li L, Durand-de Cuttoli R, Aubry AV, Burnett CJ, Cathomas F, Parise LF, et al. Social trauma engages lateral septum circuitry to occlude social reward. Nature. 2023;613:696–703.
Kangas BD, Ang Y-S, Short AK, Baram TZ, Pizzagalli DA. Computational modeling differentiates learning rate from reward sensitivity deficits produced by early-life adversity in a rodent touchscreen probabilistic reward task. Biol Psychiatry Glob Open Sci. 2024;4:100362.
Kangas BD, Short AK, Luc OT, Stern HS, Baram TZ, Pizzagalli DA. A cross-species assay demonstrates that reward responsiveness is enduringly impacted by adverse, unpredictable early-life experiences. Neuropsychopharmacology. 2022;47:767–75.
Diehl MM, Bravo-Rivera C, Quirk GJ. The study of active avoidance: a platform for discussion. Neurosci Biobehav Rev. 2019;107:229–37.
Bolton JL, Molet J, Regev L, Chen Y, Rismanchi N, Haddad E, et al. Anhedonia following early-life adversity involves aberrant interaction of reward and anxiety circuits and is reversed by partial silencing of amygdala corticotropin-releasing hormone gene. Biol Psychiatry. 2018;83:137–47.
Demaestri C, Pisciotta M, Altunkeser N, Berry G, Hyland H, Breton J, et al. Central amygdala CRF+ neurons promote heightened threat reactivity following early life adversity in mice. Nat Commun. 2024;15:5522.
Lepack AE, Bagot RC, Peña CJ, Loh Y-HE, Farrelly LA, Lu Y, et al. Aberrant H3.3 dynamics in NAc promote vulnerability to depressive-like behavior. Proc Natl Acad Sci. 2016;113:12562–67.
Geiger LT, Balouek J-A, Farrelly LA, Chen AS, Tang M, Bennett SN, et al. Early-life stress alters chromatin modifications in VTA to prime stress sensitivity. bioRxiv [Preprint]. 2024. https://www.biorxiv.org/content/10.1101/2024.03.14.584631v1.
Rashford RL, DeBerardine M, Kim HJJ, Arzouni N, Hirschfield LW, Oke OM, et al. Early-life stress alters postnatal chromatin development in the nucleus accumbens. bioRxiv [Preprint]. 2024. https://www.biorxiv.org/content/10.1101/2024.04.12.589272v1.
Parel ST, Peña CJ. Genome-wide signatures of early-life stress: influence of sex. Biol Psychiatry. 2022;91:36–42.
Singh-Taylor A, Molet J, Jiang S, Korosi A, Bolton JL, Noam Y, et al. NRSF-dependent epigenetic mechanisms contribute to programming of stress-sensitive neurons by neonatal experience, promoting resilience. Mol Psychiatry. 2018;23:648–57.
Shi D-D, Zhang Y-D, Ren Y-Y, Peng S-Y, Yuan T-F, Wang Z. Predictable maternal separation confers adult stress resilience via the medial prefrontal cortex oxytocin signaling pathway in rats. Mol Psychiatry. 2021;26:7296–307.
Hegde A, Suresh S, Mitra R. Early-life short-term environmental enrichment counteracts the effects of stress on anxiety-like behavior, brain-derived neurotrophic factor and nuclear translocation of glucocorticoid receptors in the basolateral amygdala. Sci Rep. 2020;10:14053.
Borba LA, Broseghini LDR, Manosso LM, de Moura AB, Botelho MEM, Arent CO, et al. Environmental enrichment improves lifelong persistent behavioral and epigenetic changes induced by early-life stress. J Psychiatr Res. 2021;138:107–16.
Stairs DJ, Chacho NM, Wunsch C, Pipitone L, Dravid SM. Environmental enrichment increases cue-dependent freezing and behavioral despair but decreases anxiety-like behavior in rats. Pharmacol Biochem Behav. 2020;196:172979.
Xu H, Li B, Li L, Fan Z, Gong X, Wu L, et al. Environmental enrichment mitigates PTSD-like behaviors in adult male rats exposed to early life stress by regulating histone acetylation in the hippocampus and amygdala. J Psychiatr Res. 2022;155:120–36.
Connors EJ, Migliore MM, Pillsbury SL, Shaik AN, Kentner AC. Environmental enrichment models a naturalistic form of maternal separation and shapes the anxiety response patterns of offspring. Psychoneuroendocrinology. 2015;52:153–67.
Rule L, Yang J, Watkin H, Hall J, Brydges NM. Environmental enrichment rescues survival and function of adult-born neurons following early life stress. Mol Psychiatry. 2021;26:1898–908.
Soares RO, Rorato RC, Padovan D, Lachat J-J, Antunes-Rodrigues J, Elias LLK, et al. Environmental enrichment reverses reduction in glucocorticoid receptor expression in the hippocampus of and improves behavioral responses of anxiety in early malnourished rats. Brain Res. 2015;1600:32–41.
de Lima RMS, da Mata MJ, Santos JCPD, Costa L, Marques VHM, Bento LVDS, et al. Exploring the role of environmental enrichment and early life adversity on emotional development. Behav Brain Res. 2024;472:115147.
Ma Y-N, Yang C-J, Zhang C-C, Sun Y-X, Yao X-D, Liu X, et al. Prefrontal parvalbumin interneurons mediate CRHR1-dependent early-life stress-induced cognitive deficits in adolescent male mice. Mol Psychiatry. 2024;30:2407–26.
Joushi S, Esmaeilpour K, Masoumi-Ardakani Y, Esmaeili-Mahani S, Sheibani V. Effects of short environmental enrichment on early-life adversity induced cognitive alternations in adolescent rats. J Neurosci Res. 2021;99:3373–91.
Francis DD, Diorio J, Plotsky PM, Meaney MJ. Environmental enrichment reverses the effects of maternal separation on stress reactivity. J Neurosci. 2002;22:7840–43.
Sadeghi M, Peeri M, Hosseini MJ. Adolescent voluntary exercise attenuated hippocampal innate immunity responses and depressive-like behaviors following maternal separation stress in male rats. Physiol Behav. 2016;163:177–83.
Wearick-Silva LE, Marshall P, Viola TW, Centeno-Silva A, de Azeredo LA, Orso R, et al. Running during adolescence rescues a maternal separation-induced memory impairment in female mice: potential role of differential exon-specific BDNF expression. Dev Psychobiol. 2017;59:268–74.
Fuentes IM, Jones BM, Brake AD, Pierce AN, Eller OC, Supple RM, et al. Voluntary wheel running improves outcomes in an early life stress-induced model of urologic chronic pelvic pain syndrome in male mice. Pain. 2021;162:1681–91.
Masrour FF, Peeri M, Azarbayjani MA, Hosseini M-J. Voluntary exercise during adolescence mitigated negative the effects of maternal separation stress on the depressive-like behaviors of adult male rats: role of NMDA receptors. Neurochem Res. 2018;43:1067–74.
Abbink MR, Naninck EFG, Lucassen PJ, Korosi A. Early-life stress diminishes the increase in neurogenesis after exercise in adult female mice. Hippocampus. 2017;27:839–44.
Huang H, Wang Q, Guan X, Zhang X, Zhang Y, Cao J, et al. Effects of enriched environment on depression and anxiety-like behavior induced by early life stress: a comparison between different periods. Behav Brain Res. 2021;411:113389.
Khalil MH. Environmental enrichment: a systematic review on the effect of a changing spatial complexity on hippocampal neurogenesis and plasticity in rodents, with considerations for translation to urban and built environments for humans. Front Neurosci. 2024;18:1368411.
Koe AS, Ashokan A, Mitra R. Short environmental enrichment in adulthood reverses anxiety and basolateral amygdala hypertrophy induced by maternal separation. Transl Psychiatry. 2016;6:e729
Ke Z, Yip SP, Li L, Zheng XX, Tong KY. The effects of voluntary, involuntary, and forced exercises on brain-derived neurotrophic factor and motor function recovery: a rat brain ischemia model. PLoS ONE. 2011;6:e16643.
Campbell EJ, Mitchell CS, Adams CD, Yeoh JW, Hodgson DM, Graham BA, et al. Chemogenetic activation of the lateral hypothalamus reverses early life stress-induced deficits in motivational drive. Eur J Neurosci. 2017;46:2285–96.
Gildawie KR, Wang K, Budge KE, Byrnes EM. Effects of maternal separation on effort-based responding for sucrose are associated with c-Fos expression in the nucleus accumbens core. Neuroscience. 2024;537:174–88.
Pizzagalli DA, Iosifescu D, Hallett LA, Ratner KG, Fava M. Reduced hedonic capacity in major depressive disorder: evidence from a probabilistic reward task. J Psychiatr Res. 2008;43:76–87.
Pechtel P, Lyons-Ruth K, Anderson CM, Teicher MH. Sensitive periods of amygdala development: the role of maltreatment in preadolescence. Neuroimage. 2014;97:236–44.
Frodl T, Reinhold E, Koutsouleris N, Reiser M, Meisenzahl EM. Interaction of childhood stress with hippocampus and prefrontal cortex volume reduction in major depression. J Psychiatr Res. 2010;44:799–807.
Samplin E, Ikuta T, Malhotra AK, Szeszko PR, Derosse P. Sex differences in resilience to childhood maltreatment: effects of trauma history on hippocampal volume, general cognition and subclinical psychosis in healthy adults. J Psychiatr Res. 2013;47:1174–9.
Everaerd D, Gerritsen L, Rijpkema M, Frodl T, van Oostrom I, Franke B, et al. Sex modulates the interactive effect of the serotonin transporter gene polymorphism and childhood adversity on hippocampal volume. Neuropsychopharmacology. 2012;37:1848–55.
Underwood MD, Bakalian MJ, Escobar T, Kassir S, Mann JJ, Arango V. Early-life adversity, but not suicide, is associated with less prefrontal cortex gray matter in adulthood. Int J Neuropsychopharmacol. 2019;22:349–57.
Andersen SL, Tomada A, Vincow ES, Valente E, Polcari A, Teicher MH. Preliminary evidence for sensitive periods in the effect of childhood sexual abuse on regional brain development. J Neuropsychiatry Clin Neurosci. 2008;20:292–301.
Admon R, Milad MR, Hendler T. A causal model of post-traumatic stress disorder: disentangling predisposed from acquired neural abnormalities. Trends Cogn Sci. 2013;17:337–47.
Yan CG, Rincón-Cortés M, Raineki C, Sarro E, Colcombe S, Guilfoyle DN. et al. Aberrant development of intrinsic brain activity in a rat model of caregiver maltreatment of offspring. Transl Psychiatry. 2017;7:e1005
Johnson FK, Delpech J-C, Thompson GJ, Wei L, Hao J, Herman P, et al. Amygdala hyper-connectivity in a mouse model of unpredictable early life stress. Transl Psychiatry. 2018;8:49.
Honeycutt JA, Demaestri C, Peterzell S, Silveri MM, Cai X, Kulkarni P, et al. Altered corticolimbic connectivity reveals sex-specific adolescent outcomes in a rat model of early life adversity. eLife. 2020;9:e52651.
Ishikawa J, Nishimura R, Ishikawa A. Early-life stress induces anxiety-like behaviors and activity imbalances in the medial prefrontal cortex and amygdala in adult rats. Eur J Neurosci. 2015;41:442–53.
Cody CR, de la Villarmois EA, Fernandez AM, Lardizabal J, McKnight C, Tseng K, et al. Effects of early life adversity and adolescent basolateral amygdala activity on corticolimbic connectivity and anxiety behaviors. bioRxiv [Preprint]. 2024. https://www.biorxiv.org/content/10.1101/2024.03.26.586708v1.full.
Hultman R, Ulrich K, Sachs BD, Blount C, Carlson DE, Ndubuizu N, et al. Brain-wide electrical spatiotemporal dynamics encode depression vulnerability. Cell. 2018;173:166–80.e14.
Birnie MT, Short AK, de Carvalho GB, Taniguchi L, Gunn BG, Pham AL, et al. Stress-induced plasticity of a CRH/GABA projection disrupts reward behaviors in mice. Nat Commun. 2023;14:1088.
Spyrka J, Gugula A, Rak A, Tylko G, Hess G, Blasiak A. Early life stress-induced alterations in the activity and morphology of ventral tegmental area neurons in female rats. Neurobiol Stress. 2020;13:100250.
Buthmann JL, Miller JG, Uy JP, Coury SM, Jo B, Gotlib IH. Early life stress predicts trajectories of emotional problems and hippocampal volume in adolescence. Eur Child Adolesc Psychiatry. 2024;33:2331–42.
Ruiz R, Roque A, Pineda E, Licona-Limón P, José Valdéz-Alarcón J, Lajud N. Early life stress accelerates age-induced effects on neurogenesis, depression, and metabolic risk. Psychoneuroendocrinology. 2018;96:203–11.
Dixon R, Malave L, Thompson R, Wu S, Li Y, Sadik N, et al. Sex-specific and developmental effects of early life adversity on stress reactivity are rescued by postnatal knockdown of 5-HT1A autoreceptors. Neuropsychopharmacology. 2025;50:507–18.
Kos A, Lopez JP, Bordes J, de Donno C, Dine J, Brivio E, et al. Early life adversity shapes social subordination and cell type-specific transcriptomic patterning in the ventral hippocampus. Sci Adv. 2023;9:eadj3793.
Allis CD, Jenuwein T. The molecular hallmarks of epigenetic control. Nat Rev Genet. 2016;17:487–500.
Baram TZ, Birnie MT. Enduring memory consequences of early-life stress/adversity: structural, synaptic, molecular and epigenetic mechanisms. Neurobiol Stress. 2024;33:100669.
Peña CJ. Epigenetic regulation of developmental plasticity and response to stress. Neuropsychopharmacology. 2026.
Yim YY, Teague CD, Nestler EJ. In vivo locus-specific editing of the neuroepigenome. Nat Rev Neurosci. 2020;21:471–84.
Walker DM, Zhou X, Cunningham AM, Lipschultz AP, Ramakrishnan A, Cates HM, et al. Sex-specific transcriptional changes in response to adolescent social stress in the brain’s reward circuitry. Biol Psychiatry. 2022;91:118–28.
Walker DM, Zhou X, Cunningham AM, Ramakrishnan A, Cates HM, Lardner CK, et al. Crystallin Mu in medial amygdala mediates the effect of social experience on cocaine seeking in males but not in females. Biol Psychiatry. 2022;92:895–906.
Francis TC, Chandra R, Friend DM, Finkel E, Dayrit G, Miranda J, et al. Nucleus accumbens medium spiny neuron subtypes mediate depression-related outcomes to social defeat stress. Biol Psychiatry. 2015;77:212–22.
Rahman MF, McGowan PO. Cell-type-specific epigenetic effects of early life stress on the brain. Transl Psychiatry. 2022;12:326.
Orso R, Creutzberg KC, Lumertz FS, Kestering-Ferreira E, Stocchero BA, Perrone MK, et al. A systematic review and multilevel meta-analysis of the prenatal and early life stress effects on rodent microglia, astrocyte, and oligodendrocyte density and morphology. Neurosci Biobehav Rev. 2023;150:105202.
Schafer DP, Lehrman EK, Kautzman AG, Koyama R, Mardinly AR, Yamasaki R, et al. Microglia sculpt postnatal neural circuits in an activity and complement-dependent manner. Neuron. 2012;74:691–705.
Dayananda KK, Ahmed S, Wang D, Polis B, Islam R, Kaffman A. Early life stress impairs synaptic pruning in the developing hippocampus. Brain Behav Immun. 2023;107:16–31.
Hoeijmakers L, Ruigrok SR, Amelianchik A, Ivan D, van Dam AM, Lucassen PJ, et al. Early-life stress lastingly alters the neuroinflammatory response to amyloid pathology in an Alzheimer’s disease mouse model. Brain Behav Immun. 2017;63:160–75.
Depaauw-Holt LR, Hamane S, Peyrard S, Rogers B, Fulton S, Bosson A, et al. Astrocyte glucocorticoid receptors mediate sex-specific changes in activity following stress. bioRxiv [Preprint]. 2024. https://www.biorxiv.org/content/10.1101/2024.09.17.613499v1.
Tanti A, Kim JJ, Wakid M, Davoli MA, Turecki G, Mechawar N. Child abuse associates with an imbalance of oligodendrocyte-lineage cells in ventromedial prefrontal white matter. Mol Psychiatry. 2018;23:2018–28.
Wu X, Ding Z, Fan T, Wang K, Li S, Zhao J, et al. Childhood social isolation causes anxiety-like behaviors via the damage of blood-brain barrier in amygdala in female mice. Front Cell Dev Biol. 2022;10:943067.
Menard C, Pfau ML, Hodes GE, Kana V, Wang VX, Bouchard S, et al. Social stress induces neurovascular pathology promoting depression. Nat Neurosci. 2017;20:1752–60.
Shirtcliff EA, Allison AL, Armstrong JM, Slattery MJ, Kalin NH, Essex MJ. Longitudinal stability and developmental properties of salivary cortisol levels and circadian rhythms from childhood to adolescence. Dev Psychobiol. 2012;54:493–502.
Owens M, Herbert J, Jones PB, Sahakian BJ, Wilkinson PO, Dunn VJ, et al. Elevated morning cortisol is a stratified population-level biomarker for major depression in boys only with high depressive symptoms. Proc Natl Acad Sci USA. 2014;111:3638–43.
Khandaker GM, Stochl J, Zammit S, Goodyer I, Lewis G, Jones PB. Childhood inflammatory markers and intelligence as predictors of subsequent persistent depressive symptoms: a longitudinal cohort study. Psychol Med. 2018;48:1514–22.
Chu AL, Stochl J, Lewis G, Zammit S, Jones PB, Khandaker GM. Longitudinal association between inflammatory markers and specific symptoms of depression in a prospective birth cohort. Brain Behav Immun. 2019;76:74–81.
Iob E, Kirschbaum C, Steptoe A. Persistent depressive symptoms, HPA-axis hyperactivity, and inflammation: the role of cognitive-affective and somatic symptoms. Mol Psychiatry. 2020;25:1130–40.
Colich NL, Kircanski K, Foland-Ross LC, Gotlib IH. HPA-axis reactivity interacts with stage of pubertal development to predict the onset of depression. Psychoneuroendocrinology. 2015;55:94–101.
King LS, Colich NL, LeMoult J, Humphreys KL, Ordaz SJ, Price AN, et al. The impact of the severity of early life stress on diurnal cortisol: the role of puberty. Psychoneuroendocrinology. 2017;77:68–74.
Humphreys KL, Moore SR, Davis EG, MacIsaac JL, Lin DTS, Kobor MS, et al. DNA methylation of HPA-axis genes and the onset of major depressive disorder in adolescent girls: a prospective analysis. Transl Psychiatry. 2019;9:245.
Yousufzai MIuA, Harmatz ES, Shah M, Malik MO, Goosens KA. Ghrelin is a persistent biomarker for chronic stress exposure in adolescent rats and humans. Transl Psychiatry. 2018;8:74.
Biltz RG, Sawicki CM, Sheridan JF, Godbout JP. The neuroimmunology of social-stress-induced sensitization. Nat Immunol. 2022;23:1527–35.
Carboni L, Becchi S, Piubelli C, Mallei A, Giambelli R, Razzoli M, et al. Early-life stress and antidepressants modulate peripheral biomarkers in a gene–environment rat model of depression. Prog Neuro Psychopharmacol Biol Psychiatry. 2010;34:1037–48.
Short AK, Weber R, Kamei N, Wilcox Thai C, Arora H, Mortazavi A, et al. Individual longitudinal changes in DNA-methylome identify signatures of early-life adversity and correlate with later outcome. Neurobiol Stress. 2024;31:100652.
Lussier AA, Smith BJ, Fisher J, Luo M, Cerutti J, Schneper L, et al. DNA methylation mediates the link between adversity and depressive symptoms. Nat Ment Health. 2024;2:1476–85.
Kaufman J, Wymbs NF, Montalvo-Ortiz JL, Orr C, Albaugh MD, Althoff R, et al. Methylation in OTX2 and related genes, maltreatment, and depression in children. Neuropsychopharmacology. 2018;43:2204–11.
Issler O, Haramati S, Paul Evan D, Maeno H, Navon I, Zwang R, et al. MicroRNA 135 is essential for chronic stress resiliency, antidepressant efficacy, and intact serotonergic activity. Neuron. 2014;83:344–60.
Lopez JP, Fiori LM, Cruceanu C, Lin R, Labonte B, Cates HM, et al. MicroRNAs 146a/b-5 and 425-3p and 24-3p are markers of antidepressant response and regulate MAPK/Wnt-system genes. Nat Commun. 2017;8:15497.
Torres-Berrío A, Lopez JP, Bagot RC, Nouel D, Dal Bo G, Cuesta S, et al. DCC confers susceptibility to depression-like behaviors in humans and mice and is regulated by miR-218. Biol Psychiatry. 2017;81:306–15.
Torres-Berrío A, Morgunova A, Giroux M, Cuesta S, Nestler EJ, Flores C. miR-218 in adolescence predicts and mediates vulnerability to stress. Biol Psychiatry. 2021;89:911–19.
Torres-Berrío A, Nouel D, Cuesta S, Parise EM, Restrepo-Lozano JM, Larochelle P, et al. MiR-218: a molecular switch and potential biomarker of susceptibility to stress. Mol Psychiatry. 2020;25:951–64.
Muir J, Iyer ES, Tse Y-C, Sorensen J, Wu S, Eid RS, et al. Sex-biased neural encoding of threat discrimination in nucleus accumbens afferents drives suppression of reward behavior. Nat Neurosci. 2024;27:1966–76.
Giovanniello JR, Paredes N, Wiener A, Ramírez-Armenta K, Oragwam C, Uwadia HO, et al. A dual-pathway architecture for stress to disrupt agency and promote habit. Nature. 2025;640:722–31.
Mathis A, Mamidanna P, Cury KM, Abe T, Murthy VN, Mathis MW, et al. DeepLabCut: markerless pose estimation of user-defined body parts with deep learning. Nat Neurosci. 2018;21:1281–89.
Parise EM, Gyles TM, Godino A, Sial OK, Browne CJ, Parise LF, et al. Sex-specific regulation of stress susceptibility by the astrocytic gene Htra1. bioRxiv [Preprint]. 2024. https://pubmed.ncbi.nlm.nih.gov/38659771/.
Sardar D, Cheng Y-T, Woo J, Choi D-J, Lee Z-F, Kwon W, et al. Induction of astrocytic Slc22a3 regulates sensory processing through histone serotonylation. Science. 2023;380:eade0027.
Fulton SL, Bendl J, Di Salvo G, Fullard JF, Al-Kachak A, Lepack AE, et al. Major-depressive-disorder-associated dysregulation of ZBTB7A in orbitofrontal cortex promotes astrocyte-mediated stress susceptibility. Neuron. 2025 https://doi.org/10.1016/j.neuron.2025.05.023.
Badimon A, Strasburger HJ, Ayata P, Chen X, Nair A, Ikegami A, et al. Negative feedback control of neuronal activity by microglia. Nature. 2020;586:417–23.
Chan KL, Poller WC, Swirski FK, Russo SJ. Central regulation of stress-evoked peripheral immune responses. Nat Rev Neurosci. 2023;24:591–604.
Acknowledgements
We thank Nicola Hawes, Carlos Torres-Berrío, and Jill Gregory at the Icahn School of Medicine at Mount Sinai for assistance with figure preparation.
Funding
This work was supported by grants from the National Institute of Mental Health (R01MH129306 to EJN and R01MH129643 to CJP), the Hope for Depression Research Foundation (to EJN), the Charles Hood Foundation (to ATB), and the New York Stem Cell Foundation (to CJP). CJP is a New York Stem Cell Foundation Robertson Investigator. ATB is supported by the Massachusetts General Hospital Lurie Center for Autism, Department of Pediatrics, and Center for Diversity and Inclusion.
Author information
Authors and Affiliations
Contributions
ATB and EJN designed the review timeline. ATB, AB, CJP, and EJN wrote the manuscript. All authors discussed, commented on, and edited the paper.
Corresponding authors
Ethics declarations
Competing interests
The authors declare no competing interests.
Additional information
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Rights and permissions
Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.
About this article
Cite this article
Torres-Berrío, A., Bortolami, A., Peña, C.J. et al. Neurobiology of resilience to early life stress. Neuropsychopharmacol. (2025). https://doi.org/10.1038/s41386-025-02158-4
Received:
Revised:
Accepted:
Published:
DOI: https://doi.org/10.1038/s41386-025-02158-4
This article is cited by
-
A divergent astrocytic response to stress alters activity patterns via distinct mechanisms in male and female mice
Nature Communications (2025)
-
Epigenetic regulation of brain development, plasticity, and response to early-life stress
Neuropsychopharmacology (2025)