Skip to main content

Advertisement

Log in

The Role of Oxytocin in Early Life Adversity and Later Psychopathology: a Review of Preclinical and Clinical Studies

  • Mood Disorders (C Nemeroff and J Newport, Section Editors)
  • Published:
Current Treatment Options in Psychiatry Aims and scope Submit manuscript

A Correction to this article was published on 10 January 2019

This article has been updated

Abstract

Purpose

Oxytocin is a peptide hormone integral in parturition and milk let-down, and is increasingly recognized as an important regulator of human social behaviors including bonding, trust, fear, and stress. There is an increasing evidence that oxytocin is intricately involved in a broad array of neurophysiological functions and may be a common system implicated in multiple psychiatric disorders. This review examines the putative role of oxytocin in early life adversity-mediated risk for later development of subtypes of psychiatric conditions. Several lines of evidence are reviewed including oxytocin levels, response to exogenous oxytocin administration, and genetic studies.

Recent findings

To date, most studies report lower levels of peripheral and central oxytocin in a dose-response manner in adults exposed to early life adversity. Individuals exposed to early life adversity seem to have a differential response to exogenous oxytocin administration, sometimes with negative outcomes. Several polymorphisms in the oxytocin receptor and emerging epigenetic studies point to a link between oxytocinergic systems and psychiatric disorders.

Summary

Specific limitations of the studies in the field are highlighted, and areas for future research are described. Considerably more research is needed to understand the complex role of the oxytocin system in early life adversity and its consequences.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Institutional subscriptions

Similar content being viewed by others

Change history

  • 10 January 2019

    The original publication of this article is missing a disclosure statement for Dr. Amalia Londono Tobon.

  • 10 January 2019

    The original publication of this article is missing a disclosure statement for Dr. Amalia Londono Tobon.

References and Recommended Reading

Papers of particular interest, published recently, have been highlighted as: • Of importance •• Of major importance

  1. Butchart A, Mikton C. Global status report on violence prevention, 2014. 2014.

  2. Child abuse and neglect fatalities 2016: Statistics and interventions. Washington, DC: U.S. Department of Health & Human Services AfCaF2018.

  3. 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.

    CAS  PubMed  Google Scholar 

  4. Anda RF, Felitti VJ, Bremner JD, Walker JD, Whitfield C, Perry BD, et al. The enduring effects of abuse and related adverse experiences in childhood. Eur Arch Psy Clin N N. 2006;256:174–86.

    Google Scholar 

  5. Heim C, Binder EB. Current research trends in early life stress and depression: review of human studies on sensitive periods, gene–environment interactions, and epigenetics. Exp Neurol. 2012;233:102–11.

    PubMed  Google Scholar 

  6. Heim C, Nemeroff CB. The role of childhood trauma in the neurobiology of mood and anxiety disorders: preclinical and clinical studies. Biol Psychiat. 2001;49:1023–39.

    CAS  PubMed  Google Scholar 

  7. Nemeroff CB. Neurobiological consequences of childhood trauma. J Clin Psychiat. 2004;65(Suppl 1):18–28.

    CAS  Google Scholar 

  8. Heim C, Newport DJ, Mletzko T, Miller AH, Nemeroff CB. The link between childhood trauma and depression: insights from HPA axis studies in humans. Psychoneuroendocrino. 2008;33:693–710.

    CAS  Google Scholar 

  9. Neumann ID, Landgraf R. Balance of brain oxytocin and vasopressin: implications for anxiety, depression, and social behaviors. Trends Neurosci. 2012;35:649–59.

    CAS  PubMed  Google Scholar 

  10. Meltzer-Brody S, Boschloo L, Jones I, Sullivan PF, Penninx BW. The EPDS-Lifetime: assessment of lifetime prevalence and risk factors for perinatal depression in a large cohort of depressed women. Arch Women Ment Hlth. 2013;16:465–73.

    Google Scholar 

  11. Meltzer-Brody S, Bledsoe-Mansori SE, Johnson N, Killian C, Hamer RM, Jackson C, et al. A prospective study of perinatal depression and trauma history in pregnant minority adolescents. Am J Obstet Gynecol. 2013;208:211.e1-e7.

    PubMed  Google Scholar 

  12. Maccari S, Krugers H, Morley-Fletcher S, Szyf M, Brunton P. The consequences of early-life adversity: neurobiological, behavioural and epigenetic adaptations. J Neuroendocrinol. 2014;26:707–23.

    CAS  PubMed  Google Scholar 

  13. Londono Tobon A, Diaz Stransky A, Ross DA, Stevens HE. Effects of maternal prenatal stress: mechanisms, implications, and novel therapeutic interventions. Biol Psychiat. 2016;80:e85–7.

    PubMed  Google Scholar 

  14. Dief AE, Sivukhina EV, Jirikowski GF. Oxytocin and stress response. Open J Endo Metab Dis. 2018;8. https://doi.org/10.4236/ojemd.2018.83010.

  15. Neumann ID. Involvement of the brain oxytocin system in stress coping: interactions with the hypothalamo-pituitary-adrenal axis. Prog Brain Res. 2002;139:147–62.

    CAS  PubMed  Google Scholar 

  16. Insel TR. The challenge of translation in social neuroscience: a review of oxytocin, vasopressin, and affiliative behavior. Neuron. 2010;65:768–79.

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Rao VG, Löffler C, Battey J, Hansmann I. The human gene for oxytocin-neurophysin I (OXT) is physically mapped to chromosome 20p13 by in situ hybridization. Cytogenet Genome Res. 1992;61:271–3.

    CAS  Google Scholar 

  18. Donaldson ZR, Young LJ. Oxytocin, vasopressin, and the neurogenetics of sociality. Science. 2008;322(5903):900–4.

    CAS  PubMed  Google Scholar 

  19. Neumann ID. Brain oxytocin: a key regulator of emotional and social behaviours in both females and males. J Neuroendocrinol. 2008;20(6):858–65.

    CAS  PubMed  Google Scholar 

  20. Guldenaar S, Pickering B. Immunocytochemical evidence for the presence of oxytocin in rat testis. Cell Tissue Res. 1985;240(2):485–7.

    CAS  PubMed  Google Scholar 

  21. Gauquelin G, Geelen G, Louis F, Allevard A, Meunier C, Cuisinaud G, et al. Presence of vasopressin, oxytocin and neurophysin in the retina of mammals, effect of light and darkness, comparison with the neuropeptide content of the neurohypophysis and the pineal gland. Peptides. 1983;4:509–15.

    CAS  PubMed  Google Scholar 

  22. Ang V, Jenkis J. Neurohypophysial hormones in the adrenal medulla. J Clin Endocrinol Metab. 1984;58:688–91.

    CAS  PubMed  Google Scholar 

  23. Geenen V, Legros J-J, Franchimont P, Baudrihaye M, Defresne M-P, Boniver J. The neuroendocrine thymus: coexistence of oxytocin and neurophysin in the human thymus. Science. 1986;232(4749):508–11.

    CAS  PubMed  Google Scholar 

  24. Amico JA, Finn FM, Haldar J. Oxytocin and vasopressin are present in human and rat pancreas. Am J Med Sci. 1988;296:303–7.

    CAS  PubMed  Google Scholar 

  25. Boccia M, Petrusz P, Suzuki K, Marson L, Pedersen C. Immunohistochemical localization of oxytocin receptors in human brain. Neuroscience. 2013;253:155–64.

    CAS  PubMed  Google Scholar 

  26. • Freeman SM, Smith AL, Goodman MM, Bales KL. Selective localization of oxytocin receptors and vasopressin 1a receptors in the human brainstem. Soc Neurosci. 2017;12:113–23 This study is an important step towards development of oxytocin receptor ligands to open the door to conducting functional braining imaging studies of oxytocin activity.

    PubMed  Google Scholar 

  27. Meynen G, Unmehopa U, Hofman M, Swaab D, Hoogendijk W. Hypothalamic oxytocin mRNA expression and melancholic depression. Molecular Psychiatr. 2007;12:118.

    CAS  Google Scholar 

  28. Purba JS, Hoogendijk WJ, Hofman MA, Swaab DF. Increased number of vasopressin-and oxytocin-expressing neurons in the paraventricular nucleus of the hypothalamus in depression. Arch Gen Psychiat. 1996;53:137–43.

    CAS  PubMed  Google Scholar 

  29. Smith AL, Freeman SM, Barnhart TE, Abbott DH, Ahlers EO, Kukis DL, et al. Initial investigation of three selective and potent small molecule oxytocin receptor PET ligands in New World monkeys. Bioorgan Med Chem Lett. 2016;26:3370–5.

    CAS  Google Scholar 

  30. Lee H-J, Macbeth AH, Pagani JH, Young WS 3rd. Oxytocin: the great facilitator of life. Prog Neurobiol. 2009;88:127–51.

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Carter CS. Neuroendocrine perspectives on social attachment and love. Psychoneuroendocrinol. 1998;23:779–818.

    CAS  Google Scholar 

  32. Van Ijzendoorn MH, Bakermans-Kranenburg MJ. A sniff of trust: meta-analysis of the effects of intranasal oxytocin administration on face recognition, trust to in-group, and trust to out-group. Psychoneuroendocrinol. 2012;37:438–43.

    Google Scholar 

  33. Guastella AJ, Carson DS, Dadds MR, Mitchell PB, Cox RE. Does oxytocin influence the early detection of angry and happy faces? Psychoneuroendocrinol. 2009;34:220–5.

    CAS  Google Scholar 

  34. Heinrichs M, Baumgartner T, Kirschbaum C, Ehlert U. Social support and oxytocin interact to suppress cortisol and subjective responses to psychosocial stress. Biol Psychiat. 2003;54:1389–98.

    CAS  PubMed  Google Scholar 

  35. MacDonald K, MacDonald TM. The peptide that binds: a systematic review of oxytocin and its prosocial effects in humans. Harvard Rev. Psychiat. 2010;18:1–21.

    Google Scholar 

  36. Cochran D, Fallon D, Hill M, Frazier JA. The role of oxytocin in psychiatric disorders: a review of biological and therapeutic research findings. Harvard Rev. Psychiat. 2013;21:219.

    Google Scholar 

  37. Bakermans-Kranenburg M, van Ijzendoorn M. Sniffing around oxytocin: review and meta-analyses of trials in healthy and clinical groups with implications for pharmacotherapy. Transl Psychiat. 2013;3258.

  38. Schmidt MV, Wang X-D, Meijer OC. Early life stress paradigms in rodents: potential animal models of depression? Psychopharmacology. 2011;214:131–40.

    CAS  PubMed  Google Scholar 

  39. Lyons DM, Parker KJ, Schatzberg AF. Animal models of early life stress: implications for understanding resilience. Dev Psychobiol. 2010;52:616–24.

    PubMed  Google Scholar 

  40. Gould E, Murthy S. Early life stress in rodents: animal models of illness or resilience? Front Behav Neurosci. 2018;12:157.

    PubMed  PubMed Central  Google Scholar 

  41. Francis DD, Champagne FC, Meaney MJ. Variations in maternal behaviour are associated with differences in oxytocin receptor levels in the rat. J Neuroendocrinol. 2000;12:1145–8.

    CAS  PubMed  Google Scholar 

  42. Francis DD, Young LJ, Meaney M, Insel TR. Naturally occurring differences in maternal care are associated with the expression of oxytocin and vasopressin (V1a) receptors: gender differences. J Neuroendocrinol. 2002;14:349–53.

    CAS  PubMed  Google Scholar 

  43. Champagne F, Diorio J, Sharma S, Meaney MJ. Naturally occurring variations in maternal behavior in the rat are associated with differences in estrogen-inducible central oxytocin receptors. P Natl Acad Sci USA. 2001;98:12736–41.

    CAS  Google Scholar 

  44. 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(6343):1185–8.

    PubMed  PubMed Central  Google Scholar 

  45. Kojima S, Stewart R, Demas G, Alberts J. Maternal contact differentially modulates central and peripheral oxytocin in rat pups during a brief regime of mother–pup interaction that induces a filial huddling preference. J Neuroendocrinol. 2012;24:831–40.

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Winslow JT, Noble PL, Lyons CK, Sterk SM, Insel TR. Rearing effects on cerebrospinal fluid oxytocin concentration and social buffering in rhesus monkeys. Neuropsychopharmacology. 2003;28:910.

    CAS  PubMed  Google Scholar 

  47. Hill KT, Warren M, Roth TL. The influence of infant–caregiver experiences on amygdala Bdnf, OXTr, and NPY expression in developing and adult male and female rats. Behav Brain Res. 2014;272:175–80.

    CAS  PubMed  PubMed Central  Google Scholar 

  48. • Beery AK, McEwen LM, MacIsaac JL, Francis DD, Kobor MS. Natural variation in maternal care and cross-tissue patterns of oxytocin receptor gene methylation in rats. Horm Behav. 2016;77:42–52 This animal study demonstrates that parenting behavior produces epigenetic alterations in the oxytocin system.

    CAS  PubMed  Google Scholar 

  49. Champagne FA. Epigenetic mechanisms and the transgenerational effects of maternal care. Front Neuroendocrinol. 2008;29:386–97.

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Arletti R, Bertolini A. Oxytocin acts as an antidepressant in two animal models of depression. Life Sci. 1987;41:1725–30.

    CAS  PubMed  Google Scholar 

  51. Nowakowska E, Kus K, Bobkiewicz-Kozlowska T, Hertmanowska H. Role of neuropeptides in antidepressant and memory improving effects of venlafaxine. Pol J Pharmacol. 2002;54:605–14.

    CAS  PubMed  Google Scholar 

  52. Slattery DA, Neumann ID. Oxytocin and major depressive disorder: experimental and clinical evidence for links to aetiology and possible treatment. Pharmaceuticals. 2010;3:702–24.

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Ring RH, Schechter LE, Leonard SK, Dwyer JM, Platt BJ, Graf R, et al. Receptor and behavioral pharmacology of WAY-267464, a non-peptide oxytocin receptor agonist. Neuropharmacology. 2010;58:69–77.

    CAS  PubMed  Google Scholar 

  54. • Ji H, Su W, Zhou R, Feng J, Lin Y, Zhang Y, et al. Intranasal oxytocin administration improves depression-like behaviors in adult rats that experienced neonatal maternal deprivation. Behav Pharmacol. 2016;27:689–96 This study in adult animals demonstrates that exogenous oxytocin may reverse lasting effects of early life adversity.

    CAS  PubMed  Google Scholar 

  55. McCullough ME, Churchland PS, Mendez AJ. Problems with measuring peripheral oxytocin: can the data on oxytocin and human behavior be trusted? Neurosci Biobehav Rev. 2013;37:1485–92.

    CAS  PubMed  Google Scholar 

  56. Kagerbauer S, Martin J, Schuster T, Blobner M, Kochs E, Landgraf R. Plasma oxytocin and vasopressin do not predict neuropeptide concentrations in human cerebrospinal fluid. J Neuroendocrinol. 2013;25:668–73.

    CAS  PubMed  Google Scholar 

  57. Szeto A, McCabe PM, Nation DA, Tabak BA, Rossetti MA, McCullough ME, et al. Evaluation of enzyme immunoassay and radioimmunoassay methods for the measurement of plasma oxytocin. Psychosom Med. 2011;73:393.

    CAS  PubMed  PubMed Central  Google Scholar 

  58. •• Valstad M, Alvares GA, Egknud M, Matziorinis AM, Andreassen OA, Westlye LT, et al. The correlation between central and peripheral oxytocin concentrations: a systematic review and meta-analysis. Neurosci Biobehav Rev. 2017;78:117–24 This meta-analysis offers important insights for understanding the utility and limitations of using peripheral measures of oxytocin as an index of central nervous system oxytocin activity.

    CAS  PubMed  Google Scholar 

  59. Heim C, Young LJ, Newport DJ, Mletzko T, Miller AH, Nemeroff CB. Lower CSF oxytocin concentrations in women with a history of childhood abuse. Mol Psychiatr. 2009;14:954.

    CAS  Google Scholar 

  60. Opacka-Juffry J, Mohiyeddini C. Experience of stress in childhood negatively correlates with plasma oxytocin concentration in adult men. Stress. 2012;15:1–10.

    CAS  PubMed  Google Scholar 

  61. Mohiyeddini C, Opacka-Juffry J, Gross JJ. Emotional suppression explains the link between early life stress and plasma oxytocin. Anxiety Stress Coping. 2014;27:466–75.

    PubMed  Google Scholar 

  62. Mizuki R, Fujiwara T. Association of oxytocin level and less severe forms of childhood maltreatment history among healthy Japanese adults involved with child care. Front Behav Neurosci. 2015;9:138.

    PubMed  PubMed Central  Google Scholar 

  63. Mielke EL, Neukel C, Bertsch K, Reck C, Möhler E, Herpertz SC. Alterations of brain volumes in women with early life maltreatment and their associations with oxytocin. Horm Behav. 2018;97:128–36.

    CAS  PubMed  Google Scholar 

  64. Fries ABW, Ziegler TE, Kurian JR, Jacoris S, Pollak SD. Early experience in humans is associated with changes in neuropeptides critical for regulating social behavior. P Natl Acad Sci USA. 2005;102:17237–40.

    CAS  Google Scholar 

  65. Mizushima SG, Fujisawa TX, Takiguchi S, Kumazaki H, Tanaka S, Tomoda A. Effect of the nature of subsequent environment on oxytocin and cortisol secretion in maltreated children. Front Psychiatr. 2015;6:173.

    Google Scholar 

  66. •• Julian MM, Rosenblum KL, Doom JR, Leung CY, Lumeng JC, Cruz MG, et al. Oxytocin and parenting behavior among impoverished mothers with low vs. high early life stress. Arch Women Ment Hlth. 2018;21:375–82 This study suggests that the role of oxytocin in maternal parenting behavior may be altered in those with a previous history of childhood trauma.

    Google Scholar 

  67. Apter-Levy Y, Feldman M, Vakart A, Ebstein RP, Feldman R. Impact of maternal depression across the first 6 years of life on the child’s mental health, social engagement, and empathy: the moderating role of oxytocin. Am J Psychiat. 2013;170:1161–8.

    PubMed  Google Scholar 

  68. Pierrehumbert B, Torrisi R, Laufer D, Halfon O, Ansermet F, Popovic MB. Oxytocin response to an experimental psychosocial challenge in adults exposed to traumatic experiences during childhood or adolescence. Neuroscience. 2010;166:168–77.

    CAS  PubMed  Google Scholar 

  69. Seltzer LJ, Ziegler T, Connolly MJ, Prososki AR, Pollak SD. Stress-induced elevation of oxytocin in maltreated children: evolution, neurodevelopment, and social behavior. Child Dev. 2014;85:501–12.

    PubMed  Google Scholar 

  70. Scantamburlo G, Hansenne M, Fuchs S, Pitchot W, Marechal P, Pequeux C, et al. Plasma oxytocin levels and anxiety in patients with major depression. Psychoneuroendocrinol. 2007;32:407–10.

    CAS  Google Scholar 

  71. Parker KJ, Kenna HA, Zeitzer JM, Keller J, Blasey CM, Amico JA, et al. Preliminary evidence that plasma oxytocin levels are elevated in major depression. Psychiat Res. 2010;178:359–62.

    CAS  Google Scholar 

  72. Kendall-Tackett KA, Williams LM, Finkelhor D. Impact of sexual abuse on children: a review and synthesis of recent empirical studies. Psychol Bull. 1993;113:164.

    CAS  PubMed  Google Scholar 

  73. Striepens N, Kendrick KM, Hanking V, Landgraf R, Wüllner U, Maier W, et al. Elevated cerebrospinal fluid and blood concentrations of oxytocin following its intranasal administration in humans. Sci Rep-UK. 2013;3:3440.

    Google Scholar 

  74. Talegaonkar S, Mishra P. Intranasal delivery: an approach to bypass the blood brain barrier. Indian J Pharmacol. 2004;36:140.

    CAS  Google Scholar 

  75. Churchland PS, Winkielman P. Modulating social behavior with oxytocin: how does it work? What does it mean? Horm Behav. 2012;61:392–9.

    CAS  PubMed  Google Scholar 

  76. Graustella AJ, MacLeod C. A critical review of the influence of oxytocin nasal spray on social cognition in humans: evidence and future directions. Horm Behav. 2012;61:410–8.

    CAS  PubMed  Google Scholar 

  77. Bartz JA, Zaki J, Bolger N, Ochsner KN. Social effects of oxytocin in humans: context and person matter. Trends Cogn Sci. 2011;15:301–9.

    CAS  PubMed  Google Scholar 

  78. Riem MM, van IJzendoorn MH, Tops M, Boksem MA, Rombouts SA, Bakermans-Kranenburg MJ. Oxytocin effects on complex brain networks are moderated by experiences of maternal love withdrawal. Eur Neuropsychopharmacol. 2013;23:1288–95.

    CAS  PubMed  Google Scholar 

  79. van Ijzendoorn MH, Huffmeijer R, Alink LR, Bakermans-Kranenburg MJ, Tops M. The impact of oxytocin administration on charitable donating is moderated by experiences of parental love-withdrawal. Front Psychol. 2011;2:258.

    PubMed  PubMed Central  Google Scholar 

  80. Bakermans-Kranenburg MJ, van IJzendoorn MH, Riem MM, Tops M, Alink LR. Oxytocin decreases handgrip force in reaction to infant crying in females without harsh parenting experiences. Soc Cogn Affect Neurosci. 2011;7:951–7.

    PubMed  PubMed Central  Google Scholar 

  81. Meinlschmidt G, Heim C. Sensitivity to intranasal oxytocin in adult men with early parental separation. Biol Psychiat. 2007;61:1109–11.

    CAS  PubMed  Google Scholar 

  82. Grimm S, Pestke K, Feeser M, Aust S, Weigand A, Wang J, et al. Early life stress modulates oxytocin effects on limbic system during acute psychosocial stress. Soc Cogn Affect Neurosci. 2014;9:1828–35.

    PubMed  PubMed Central  Google Scholar 

  83. Fan Y, Herrera-Melendez AL, Pestke K, Feeser M, Aust S, Otte C, et al. Early life stress modulates amygdala-prefrontal functional connectivity: implications for oxytocin effects. Hum Brain Mapp. 2014;35:5328–39.

    PubMed  Google Scholar 

  84. Naber F, van Ijzendoorn MH, Deschamps P, van Engeland H, Bakermans-Kranenburg MJ. Intranasal oxytocin increases fathers’ observed responsiveness during play with their children: a double-blind within-subject experiment. Psychoneuroendocrinol. 2010;35:1583–6.

    CAS  Google Scholar 

  85. Weisman O, Zagoory-Sharon O, Feldman R. Oxytocin administration to parent enhances infant physiological and behavioral readiness for social engagement. Biol Psychiat. 2012;72:982–9.

    CAS  PubMed  Google Scholar 

  86. Mah BL, van Ijzendoorn MH, Smith R, Bakermans-Kranenburg MJ. Oxytocin in postnatally depressed mothers: its influence on mood and expressed emotion. Progr Neuro-Psychopharmacol Biol Psychiat. 2013;40:267–72.

    CAS  Google Scholar 

  87. Kimura T, Tanizawa O, Mori K, Brownstein MJ, Okayama H. Structure and expression of a human oxytocin receptor. Nature. 1992;356(6369):526.

    CAS  PubMed  Google Scholar 

  88. Gimpl G, Fahrenholz F. The oxytocin receptor system: structure, function, and regulation. Physiol Rev. 2001;81:629–83.

    CAS  PubMed  Google Scholar 

  89. •• Smearman EL, Almli LM, Conneely KN, Brody GH, Sales JM, Bradley B, et al. Oxytocin receptor genetic and epigenetic variations: association with child abuse and adult psychiatric symptoms. Child Dev. 2016;87:122–34 This study of oxytocin receptor gene DNA methylation suggests that oxytocin-related epigenetic changes may underlie the association between child abuse history and adult psychiatric illness.

    PubMed  PubMed Central  Google Scholar 

  90. Reiner I, van IJzendoorn M, Bakermans-Kranenburg M, Bleich S, Beutel M, Frieling H. Methylation of the oxytocin receptor gene in clinically depressed patients compared to controls: The role of OXTR rs53576 genotype. J Psychiatr Res. 2015;65:9–15.

    CAS  PubMed  Google Scholar 

  91. Thompson SM, Hammen C, Starr LR, Najman JM. Oxytocin receptor gene polymorphism (rs53576) moderates the intergenerational transmission of depression. Psychoneuroendocrinol. 2014;43:11–9.

    CAS  Google Scholar 

  92. Wang J, Qin W, Liu B, Zhou Y, Wang D, Zhang Y, et al. Neural mechanisms of oxytocin receptor gene mediating anxiety-related temperament. Brain Struct Funct. 2014;219:1543–54.

    CAS  PubMed  Google Scholar 

  93. Sippel LM, Han S, Watkins LE, Harpaz-Rotem I, Southwick SM, Krystal JH, et al. Oxytocin receptor gene polymorphisms, attachment, and PTSD: results from the national health and resilience in veterans study. J Psychiatr Res. 2017;94:139–47.

    PubMed  PubMed Central  Google Scholar 

  94. Li J, Zhao Y, Li R, Broster LS, Zhou C, Yang S. Association of oxytocin receptor gene (OXTR) rs53576 polymorphism with sociality: a meta-analysis. PLoS One. 2015;10:e0131820.

    PubMed  PubMed Central  Google Scholar 

  95. Truzzi A, Poquerusse J, Setoh P, Shinohara K, Bornstein MH, Esposito G. Oxytocin receptor gene polymorphism (rs53576) and early paternal care sensitize males to distressing female vocalizations. Dev Psychobiol. 2018;60:333–9.

    CAS  PubMed  Google Scholar 

  96. Bradley B, Westen D, Mercer KB, Binder EB, Jovanovic T, Crain D, et al. Association between childhood maltreatment and adult emotional dysregulation in a low-income, urban, African American sample: moderation by the oxytocin receptor gene. Dev Psychopathol. 2011;23:439–52.

    PubMed  PubMed Central  Google Scholar 

  97. McQuaid RJ, McInnis OA, Stead JD, Matheson K, Anisman H. A paradoxical association of an oxytocin receptor gene polymorphism: early-life adversity and vulnerability to depression. Front Neurosci. 2013;7:128.

    PubMed  PubMed Central  Google Scholar 

  98. Hostinar CE, Cicchetti D, Rogosch FA. Oxytocin receptor gene polymorphism, perceived social support, and psychological symptoms in maltreated adolescents. Dev Psychopathol. 2014;26:465–77.

    PubMed  PubMed Central  Google Scholar 

  99. Cicchetti D, Rogosch FA. Gene× environment interaction and resilience: effects of child maltreatment and serotonin, corticotropin releasing hormone, dopamine, and oxytocin genes. Dev Psychopathol. 2012;24:411–27.

    PubMed  PubMed Central  Google Scholar 

  100. Dannlowski U, Kugel H, Grotegerd D, Redlich R, Opel N, Dohm K, et al. Disadvantage of social sensitivity: interaction of oxytocin receptor genotype and child maltreatment on brain structure. Biol Psychiat. 2016;80:398–405.

    CAS  PubMed  Google Scholar 

  101. Tollenaar MS, Molendijk ML, Penninx BW, Milaneschi Y, Antypa N. The association of childhood maltreatment with depression and anxiety is not moderated by the oxytocin receptor gene. Eur Arch Psy Clin N. 2017;267:517–26.

    Google Scholar 

  102. Myers AJ, Williams L, McAuley-Clark EZ, Dobson-Stone C, Schofield PR, Nemeroff CB. Variation in the oxytocin receptor gene is associated with increased risk for anxiety, stress and depression in individuals with a history of exposure to early life stress. J Psychiatr Res. 2014;59:93–100.

    PubMed  PubMed Central  Google Scholar 

  103. Cammack AL, Hogue CJ, Drews-Botsch CD, Kramer MR, Pearce BD, Knight B, et al. An exploratory study of whether pregnancy outcomes influence maternal self-reported history of child maltreatment. Child Abuse Negl. 2018. https://doi.org/10.1016/j.chiabu.2018.01.022.

  104. •• Toepfer P, Heim C, Entringer S, Binder E, Wadhwa P, Buss C. Oxytocin pathways in the intergenerational transmission of maternal early life stress. Neurosci Biobehav Rev. 2017;73:293–308 This excellent review examines several biological pathways, including oxytocin, pertinent to the parent-to-child transmission of vulnerability due to early life adversity.

    CAS  PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Charles B. Nemeroff MD, PhD.

Ethics declarations

Conflict of interest

Dr. Londono Tobon has nothing to disclose.

Dr. Newport has received research support from Eli Lilly, Glaxo SmithKline (GSK), Janssen, National Alliance for Research on Schizophrenia and Depression (NARSAD), National Institutes of Health (NIH), Takeda Pharmaceuticals, and Wyeth. He has served on speakers’ bureaus and/or received honoraria from Astra-Zeneca, Eli Lilly, GSK, Pfizer, and Wyeth. He has served on advisory boards for GSK, Janssen, and Sage Therapeutics. He has never served as a consultant to any biomedical or pharmaceutical corporations. Neither he nor family members have ever held equity positions in biomedical or pharmaceutical corporations.

Dr. Nemeroff has received research support from the National Institutes of Health (NIH) and Stanley Medical Research Institute. He has served on scientific advisory boards for the American Foundation for Suicide Prevention (AFSP), Anxiety and Depression Association of America (ADAA), Bracket (Clintara), Brain and Behavior Research Foundation, Laureate Institute for Brain Research (LIBR), Skyland Trail, and Xhale, on the board of directors for AFSP, ADAA, and Gratitude America, and as a consultant (within the last 3 years) to Bracket (Clintara), Fortress Biotech, Intra-Cellular Therapies Inc., Janssen Research & Development LLC, Magstim Inc., Navitor Pharmaceuticals, Sumitomo Dainippon Pharma, Sunovion Pharmaceuticals, Taisho Pharmaceutical Inc., Takeda Pharmaceuticals, TC MSO Inc., and Xhale. He has stock holdings with Abbvie, Antares, BI Gen Holdings Inc., Celgene, Corcept Therapeutics Pharmaceuticals Company, OPKO Health Inc., Seattle Genetics, and Xhale. He has received honoraria, royalties, and expert witness fees from various sources. His income sources or equity of $10,000 or more include American Psychiatric Publishing, Bracket (Clintara), CME Outfitters, Intra-Cellular Therapies Inc., Magstim, Takeda Pharmaceuticals, and Xhale. He holds patents for Method and devices for transdermal delivery of lithium (US 6,375,990B1), method of assessing antidepressant drug therapy via transport inhibition of monoamine neurotransmitters by ex vivo assay (US 7,148,027B2), and Compounds, Compositions, Methods of Synthesis, and Methods of Treatment (CRF Receptor Binding Ligand) (US 8,551, 996 B2).

Human and animal rights and informed consent

This article does not contain any studies with human or animal subjects performed by any of the authors.

Additional information

This article is part of the Topical Collection on Mood Disorders

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Londono Tobon, A., Newport, D.J. & Nemeroff, C.B. The Role of Oxytocin in Early Life Adversity and Later Psychopathology: a Review of Preclinical and Clinical Studies. Curr Treat Options Psych 5, 401–415 (2018). https://doi.org/10.1007/s40501-018-0158-9

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s40501-018-0158-9

Keywords

Navigation