From Autism to Eating Disorders and More: The Role of Oxytocin in Neuropsychiatric Disorders

Adele Romano, Bianca Tempesta, Maria Vittoria Micioni Di Bonaventura, Silvana Gaetani, Adele Romano, Bianca Tempesta, Maria Vittoria Micioni Di Bonaventura, Silvana Gaetani

Abstract

Oxytocin (oxy) is a pituitary neuropeptide hormone synthesized from the paraventricular and supraoptic nuclei within the hypothalamus. Like other neuropeptides, oxy can modulate a wide range of neurotransmitter and neuromodulator activities. Additionally, through the neurohypophysis, oxy is secreted into the systemic circulation to act as a hormone, thereby influencing several body functions. Oxy plays a pivotal role in parturition, milk let-down and maternal behavior and has been demonstrated to be important in the formation of pair bonding between mother and infants as well as in mating pairs. Furthermore, oxy has been proven to play a key role in the regulation of several behaviors associated with neuropsychiatric disorders, including social interactions, social memory response to social stimuli, decision-making in the context of social interactions, feeding behavior, emotional reactivity, etc. An increasing body of evidence suggests that deregulations of the oxytocinergic system might be involved in the pathophysiology of certain neuropsychiatric disorders such as autism, eating disorders, schizophrenia, mood, and anxiety disorders. The potential use of oxy in these mental health disorders is attracting growing interest since numerous beneficial properties are ascribed to this neuropeptide. The present manuscript will review the existing findings on the role played by oxy in a variety of distinct physiological and behavioral functions (Figure 1) and on its role and impact in different psychiatric disorders. The aim of this review is to highlight the need of further investigations on this target that might contribute to the development of novel more efficacious therapies. Figure 1Oxytocin regulatory control of different and complex processes.

Keywords: anxiety; autism; eating disorders; mood disorders; oxytocinergic system.

Figures

Figure 1
Figure 1
Oxytocin regulatory control of different and complex processes.

References

    1. Altemus M. (1995). Neuropeptides in anxiety disorders. Effects of lactation. Ann. N.Y. Acad. Sci. 771, 697–707. 10.1111/j.1749-6632.1995.tb44721.x
    1. Altemus M., Jacobson K. R., Debellis M., Kling M., Pigott T., Murphy D. L., et al. . (1999). Normal CSF oxytocin and NPY levels in OCD. Biol. Psychiatry 45, 931–933. 10.1016/S0006-3223(98)00263-7
    1. Altemus M., Redwine L. S., Leong Y. M., Frye C. A., Porges S. W., Carter C. S. (2001). Responses to laboratory psychosocial stress in postpartum women. Psychosom. Med. 63, 814–821. 10.1097/00006842-200109000-00015
    1. Altirriba J., Poher A. L., Caillon A., Arsenijevic D., Veyrat-Durebex C., Lyautey J., et al. . (2014). Divergent effects of oxytocin treatment of obese diabetic mice on adiposity and diabetes. Endocrinology 155, 4189–41201. 10.1210/en.2014-1466
    1. Alvares G. A., Chen N. T., Balleine B. W., Hickie I. B., Guastella A. J. (2012). Oxytocin selectively moderates negative cognitive appraisals in high trait anxious males. Psychoneuroendocrinology 37, 2022–2031. 10.1016/j.psyneuen.2012.04.018
    1. Amar A. P., Weiss M. H. (2003). Pituitary anatomy and physiology. Neurosurg. Clin. N. Am. 14, 11–23. 10.1016/S1042-3680(02)00017-7
    1. Amico J. A., Challinor S. M., Cameron J. L. (1990). Pattern of oxytocin concentrations in the plasma and cerebrospinal fluid of lactating rhesus monkeys (Macaca mulatta):evidence for functionally independent oxytocinergic pathways in primates. J. Clin. Endocrinol. 71, 1531–1535. 10.1210/jcem-71-6-1531
    1. Amico J. A., Mantella R. C., Vollmer R. R., Li X. (2014). Anxiety and stress responses in female oxytocin deficient mice. J. Neuroendocrinol. 16, 319–324. 10.1111/j.0953-8194.2004.01161.x
    1. Anagnostou E., Soorya L., Chaplin W., Bartz J., Halpern D., Wasserman S., et al. . (2012). Intranasal oxytocin versus placebo in the treatment of adults with autism spectrum disorders: a randomized controlled trial. Mol. Autism. 3:16. 10.1186/2040-2392-3-16
    1. Andari E., Duhamel J. R., Zalla T., Herbrecht E., Leboyer M., Sirigu A. (2010). Promoting social behavior with oxytocin in high-functioning autism spectrum disorders. Proc. Natl. Acad. Sci. U.S.A. 107, 4389–4394. 10.1073/pnas.0910249107
    1. Anderberg U. M., Uvnäs-Moberg K. (2000). Plasma oxytocin levels in female fibromyalgia syndrome patients. Z. Rheumatol. 59, 373–379. 10.1007/s003930070045
    1. Anderson-Hunt M., Dennerstein L. (1994). Increased female sexual response after oxytocin. BMJ 309:929. 10.1136/bmj.309.6959.929
    1. Anderson-Hunt M., Dennerstein L. (1995). Oxytocin and female sexuality. Gynecol. Obstet. Invest. 40, 217–221. 10.1159/000292340
    1. Arletti R., Benelli A., Bertolini A. (1989). Influence of oxytocin on feeding behavior in the rat. Peptides 10, 89–93. 10.1016/0196-9781(89)90082-X
    1. Arletti R., Benelli A., Bertolini A. (1990). Oxytocin inhibits food and fluid intake in rats. Physiol. Behav. 48, 825–830. 10.1016/0031-9384(90)90234-U
    1. Bakharev V. D., Tikhomirov S. M., Lozhkina T. K. (1986). Psychotropic properties of oxytocin. Neurosci. Behav. Physiol. 16, 160–164. 10.1007/BF01186517
    1. Bale T. L., Davis A. M., Auger A. P., Dorsa D. M., McCarthy M. M. (2001). CNS region-specific oxytocin receptor expression: importance in regulation of anxiety and sex behavior. J. Neurosci. 21, 2546–2552. Available online at:
    1. Bales K. L., Perkeybile A. M., Conley O. G., Lee M. H., Guoynes C. D., Downing G. M., et al. . (2013). Chronic intranasal oxytocin causes long-term impairments in partner preference formation in male prairie voles. Biol. Psychiatry 74, 180–188. 10.1016/j.biopsych.2012.08.025
    1. Bartz J. A., Zaki J., Bolger N., Ochsner K. N. (2011). Social effects of oxytocin in humans: context and person matter. Trends Cogn. Sci. 15, 301–309. 10.1016/j.tics.2011.05.002
    1. Baskerville T. A., Allard J., Wayman C., Douglas A. J. (2009). Dopamine-oxytocin interactions in penile erection. Eur. J. Neurosci. 30, 2151–2164. 10.1111/j.1460-9568.2009.06999.x
    1. Beckmann H., Lang R. E., Gattaz W. F. (1985). Vasopressin–oxytocin in cerebrospinal fluid of schizophrenic patients and normal controls. Psychoneuroendocrinology 10, 187–191. 10.1016/0306-4530(85)90056-3
    1. Bell C. J., Nicholson H., Mulder R. T., Luty S. E., Joyce P. R. (2006). Plasma oxytocin levels in depression and their correlation with the temperament dimension of reward dependence. J. Psychopharmacol. 20, 656–660. 10.1177/0269881106060512
    1. Ben-Ari Y., Gaiarsa J. L., Tyzio R., Khazipov R. (2007). GABA: a pioneer transmitter that excites immature neurons and generates primitive oscillations. Physiol. Rev. 87, 1215–1284. 10.1152/physrev.00017.2006
    1. Benelli A., Bertolini A., Poggioli R., Menozzi B., Basaglia R., Arletti R. (1995). Polymodal dose-response curve for oxytocin in the social recognition test. Neuropeptides 28, 251–255. 10.1016/0143-4179(95)90029-2
    1. Bhandari R., van der Veen R., Parsons C. E., Young K. S., Voorthuis A., Bakermans-Kranenburg M. J., et al. . (2014). Effects of intranasal oxytocin administration on memory for infant cues: moderation by childhood emotional maltreatment. Soc. Neurosci. 9, 536–547. 10.1080/17470919.2014.932307
    1. Blanks A. M., Thornton S. (2003). The role of oxytocin in parturition. BJOG 110(Suppl. 20), 46–51. 10.1046/j.1471-0528.2003.00024.x
    1. Blatt G. J., Fatemi S. H. (2011). Alterations in GABAergic biomarkers in the autism brain: research findings and clinical implications. Anat. Rec. (Hoboken) 294, 1646–1452. 10.1002/ar.21252
    1. Blevins J. E., Graham J. L., Morton G. J., Bales K. L., Schwartz M. W., Baskin D. G., et al. . (2015). Chronic oxytocin administration inhibits food intake, increases energy expenditure, and produces weight loss in fructose-fed obese rhesus monkeys. Am. J. Physiol. Regul. Integr. Comp. Physiol. 308, R431–R438. 10.1152/ajpregu.00441.2014
    1. Blevins J. E., Ho J. M. (2013). Role of oxytocin signaling in the regulation of body weight. Rev. Endocr. Metab. Disord. 14, 311–329. 10.1007/s11154-013-9260-x
    1. Blume A., Bosch O. J., Miklos S., Torner L., Wales L., Waldherr M., et al. . (2008). Oxytocin reduces anxiety via ERK1/2 activation: local effect within the rat hypothalamic paraventricular nucleus. Eur. J. Neurosci. 27, 1947–1956. 10.1111/j.1460-9568.2008.06184.x
    1. Bosch O. J., Neumann I. D. (2012). Both oxytocin and vasopressin are mediators of maternal care and aggression in rodents: from central release to sites of action. Horm. Behav. 61, 293–303. 10.1016/j.yhbeh.2011.11.002
    1. Breuil V., Amri E. Z., Panaia-Ferrari P., Testa J., Elabd C., Albert-Sabonnadière C., et al. . (2011). Oxytocin and bone remodelling: relationships with neuropituitary hormones, bone status and body composition. Joint Bone Spine 78, 611–615. 10.1016/j.jbspin.2011.02.002
    1. Broadbear J. H., Tunstall B., Beringer K. (2011). Examining the role of oxytocin in the interoceptive effects of 3,4-methylenedioxymethamphetamine (MDMA, “ecstasy”) using a drug discrimination paradigm in the rat. Addict. Biol. 16, 202–214. 10.1111/j.1369-1600.2010.00267.x
    1. Brownstein M. J., Russell J. T., Gainer H. (1980). Synthesis, transport, and release of posterior pituitary hormones. Science 207, 373–378. 10.1126/science.6153132
    1. Burbach J. P., Luckman S. M., Murphy D., Gainer H. (2001). Gene regulation in the magnocellular hypothalamo-neurohypophysial system. Physiol. Rev. 81, 1197–1267. Available online at:
    1. Camerino C. (2009). Low sympathetic tone and obese phenotype in oxytocin-deficient mice. Obesity (Silver Spring) 17, 980–984. 10.1038/oby.2009.12
    1. Campbell D. B., Datta D., Jones S. T., Batey Lee E., Sutcliffe J. S., Hammock E. A., et al. . (2011). Association of oxytocin receptor (OXTR) gene variants with multiple phenotype domains of autism spectrum disorder. J. Neurodev. Disord. 3, 101–112. 10.1007/s11689-010-9071-2
    1. Carmichael M. S., Humbert R., Dixen J., Palmisano G., Greenleaf W., Davidson J. M. (1987). Plasma oxytocin increases in the human sexual response. J. Clin. Endocrinol. Metab. 64, 27–31. 10.1210/jcem-64-1-27
    1. Carson D. S., Cornish J. L., Guastella A. J., Hunt G. E., McGregor I. S. (2010a). Oxytocin decreases methamphetamine self-administration, methamphetamine hyperactivity, and relapse to methamphetamine seeking behaviour in rats. Neuropharmacology 58, 38–43. 10.1016/j.neuropharm.2009.06.018
    1. Carson D. S., Hunt G. E., Guastella A. J., Barber L., Cornish J. L., Arnold J. C., et al. . (2010b). Systemically administered oxytocin decreases methamphetamine activation of the subthalamic nucleus and accumbens core and stimulates oxytocinergic neurons in the hypothalamus. Addict. Biol. 15, 448–463. 10.1111/j.1369-1600.2010.00247.x
    1. Carter C. S. (1992). Oxytocin and sexual behavior. Neurosci. Biobehav. Rev. 16, 131–144. 10.1016/S0149-7634(05)80176-9
    1. Champagne F. A., Chretien P., Stevenson C. W., Zhang T. Y., Gratton A., Meaney M. J. (2004). Variations in nucleus accumbens dopamine associated with individual differences in maternal behavior in the rat. J. Neurosci. 24, 4113–4123. 10.1523/JNEUROSCI.5322-03.2004
    1. Cicutti N. J., Smyth C. E., Rosaeg O. P., Wilkinson M. (1999). Oxytocin receptor binding in rat and human heart. Can. J. Cardiol. 15, 1267–1273.
    1. Condés-Lara M., Maie I. A., Dickenson A. H. (2005). Oxytocin actions on afferent evoked spinal cord neuronal activities in neuropathic but not in normal rats. Brain Res. 1045, 124–133. 10.1016/j.brainres.2005.03.020
    1. Coote J. H. (2005). A role for the paraventricular nucleus of the hypothalamus in the autonomic control of heart and kidney. Exp. Physiol. 90, 169–173. 10.1113/expphysiol.2004.029041
    1. Costa B., Pini S., Gabelloni P., Abelli M., Lari L., Cardini A., et al. . (2009). Oxytocin receptor polymorphisms and adult attachment style in patients with depression. Psychoneuroendocrinology 34, 1506–1514. 10.1016/j.psyneuen.2009.05.006
    1. Crock L. W., Kolber B. J., Morgan C. D., Sadler K. E., Vogt S. K., Bruchas M. R., et al. . (2012). Central amygdala metabotropic glutamate receptor 5 in the modulation of visceral pain. J. Neurosci. 32, 14217–14226. 10.1523/JNEUROSCI.1473-12.2012
    1. Cyranowski J. M., Hofkens T. L., Frank E., Seltman H., Cai H. M., Amico J. A. (2008). Evidence of dysregulated peripheral oxytocin release among depressed women. Psychosom. Med. 70, 967–975. 10.1097/PSY.0b013e318188ade4
    1. Dabrowska J., Hazra R., Ahern T. H., Guo J. D., McDonald A. J., Mascagni F., et al. . (2011). Neuroanatomical evidence for reciprocal regulation of the corticotrophin-releasing factor and oxytocin systems in the hypothalamus and the bed nucleus of the stria terminalis of the rat: Implications for balancing stress and affect. Psychoneuroendocrinology 36, 1312–1326. 10.1016/j.psyneuen.2011.03.003
    1. Dantzer R., Bluthe R. M., Koob G. F., Le Moal M. (1987). Modulation of social memory in male rats by neurohypophyseal peptides. Psychopharmacology (Berl). 91, 363–368. 10.1007/BF00518192
    1. Davis M. C., Green M. F., Lee J., Horan W. P., Senturk D., Clarke A. D., et al. . (2014). Oxytocin-augmented social cognitive skills training in schizophrenia. Neuropsychopharmacology 39, 2070–2077. 10.1038/npp.2014.68
    1. Deblon N., Veyrat-Durebex C., Bourgoin L., Caillon A., Bussier A. L., Petrosino S., et al. . (2011). Mechanisms of the anti-obesity effects of oxytocin in diet-induced obese rats. PLoS ONE 6:e25565. 10.1371/journal.pone.0025565
    1. de Bree F. M. (2000). Trafficking of the vasopressin and oxytocin prohormone through the regulated secretory pathway. J. Neuroendocrinol. 12, 589–594. 10.1046/j.1365-2826.2000.00521.x
    1. Demitrack M. A., Lesem M. D., Listwak S. J., Brandt H. A., Jimerson D. C., Gold P. W. (1990). CSF oxytocin in anorexia nervosa and bulimia nervosa: clinical and pathophysiologic considerations. Am. J. Psychiatry 147, 882–886. 10.1176/ajp.147.7.882
    1. Dogterom J., Van Wimersma Greidanus T. B., Swabb D. F. (1977). Evidence for the release of vasopressin and oxytocin into cerebrospinal fluid: measurements in plasma and CSF of intact and hypophysectomized rats. Neuroendocrinology 24, 108–118. 10.1159/000122702
    1. Dölen G., Darvishzadeh A., Huang K. W., Malenka R. C. (2013). Social reward requires coordinated activity of nucleus accumbens oxytocin and serotonin. Nature 501, 179–184. 10.1038/nature12518
    1. Dombret C., Nguyen T., Schakman O., Michaud J. L., Hardin-Pouzet H., Bertrand M. J., et al. . (2012). Loss of Maged1 results in obesity, deficits of social interactions, impaired sexual behavior and severe alteration of mature oxytocin production in the hypothalamus. Hum. Mol. Genet. 21, 4703–4717. 10.1093/hmg/dds310
    1. Domes G., Heinrichs M., Michel A., Berger C., Herpertz S. C. (2007). Oxytocin improves “mind-reading” in humans. Biol. Psychiatry 61, 731–733. 10.1016/j.biopsych.2006.07.015
    1. Drago F., Contarino A., Busà L. (1999). The expression of neuropeptide-induced excessive grooming behavior in dopamine D1 and D2 receptor-deficient mice. Eur. J. Pharmacol. 365, 125–131. 10.1016/S0014-2999(98)00877-2
    1. Dumont G. J., Sweep F. C., van der Steen R., Hermsen R., Donders A. R., Touw D. J., et al. . (2009). Increased oxytocin concentrations and prosocial feelings in humans after ecstasy (3,4-methylenedioxymethamphetamine) administration. Soc. Neurosci. 4, 359–366. 10.1080/17470910802649470
    1. Dutil J., Moujahidine M., Lemieux C., Jankowski M., Gutkowska J., Deng A. Y. (2001). Chromosomal and comparative mapping of rat oxytocin, oxytocin receptor and vasopressin genes. Cytogenet. Cell Genet. 93, 57–59. 10.1159/000056949
    1. Dykens E. M., Leckman J. F., Cassidy S. B. (1996). Obsessions and compulsions in Prader-Willi syndrome. J. Child Psychol. Psychiatry 37, 995–1002. 10.1111/j.1469-7610.1996.tb01496.x
    1. Engelmann M., Ebner K., Wotjak C. T., Landgraf R. (1998). Endogenous oxytocin is involved in short-term olfactory memory in female rats. Behav. Brain Res. 90, 89–94. 10.1016/S0166-4328(97)00084-3
    1. Evans J. J., Reid R. A., Wakeman S. A., Croft L. B., Benny P. S. (2003). Evidence that oxytocin is a physiological component of LH regulation in non-pregnant women. Hum. Reprod. 18, 1428–1431. 10.1093/humrep/deg291
    1. Favaretto A. L., Ballejo G. O., Albuquerque-Araújo W. I., Gutkowska J., Antunes-Rodrigues J., McCann S. M. (1997). Oxytocin releases atrial natriuretic peptide from rat atria in vitro that exerts negative inotropic and chronotropic action. Peptides 18, 1377–1381. 10.1016/S0196-9781(97)00209-X
    1. Feifel D., Macdonald K., Cobb P., Minassian A. (2012). Adjunctive intranasal oxytocin improves verbal memory in people with schizophrenia. Schizophr. Res. 139, 207–210. 10.1016/j.schres.2012.05.018
    1. Feifel D., Macdonald K., Nguyen A., Cobb P., Warlan H., Galangue B., et al. . (2010). Adjunctive intranasal oxytocin reduces symptoms in schizophrenia patients. Biol. Psychiatry 68, 678–680. 10.1016/j.biopsych.2010.04.039
    1. Ferguson J. N., Young L. J., Hearn E. F., Matzuk M. M., Insel T. R., Winslow J. T. (2000). Social amnesia in mice lacking the oxytocin gene. Nat. Genet. 25, 284–288. 10.1038/77040
    1. Ferguson J. N., Young L. J., Insel T. R. (2002). The neuroendocrine basis of social recognition. Front. Neuroendocrinol. 23, 200–224. 10.1006/frne.2002.0229
    1. Francis S. M., Sagar A., Levin-Decanini T., Liu W., Carter C. S., Jacob S. (2014). Oxytocin and vasopressin systems in genetic syndromes and neurodevelopmental disorders. Brain Res. 1580, 199–218. 10.1016/j.brainres.2014.01.021
    1. Frasch A., Zetzsche T., Steiger A., Jirikowski G. F. (1995). Reduction of plasma oxytocin levels in patients suffering from major depression. Adv. Exp. Med. Biol. 395, 257–258.
    1. Gaida W., Lang R. E., Kraft K., Unger T., Ganten D. (1985). Altered neuropeptide concentrations in spontaneously hypertensive rats: cause or consequence? Clin. Sci. 68, 35–43. 10.1042/cs0680035
    1. Gerdeman G. L., Lovinger D. M. (2003). Emerging roles for endocannabinoids in long-term synaptic plasticity. Br. J. Pharmacol. 140, 781–789. 10.1038/sj.bjp.0705466
    1. Gibson C. M., Penn D. L., Smedley K. L., Leserman J., Elliott T., Pedersen C. A. (2014). A pilot six-week randomized controlled trial of oxytocin on social cognition and social skills in schizophrenia. Schizophr. Res. 156, 261–265. 10.1016/j.schres.2014.04.009
    1. Gigliucci V., Leonzino M., Busnelli M., Luchetti A., Palladino V. S., D'Amato F. R., et al. . (2014). Region specific up-regulation of oxytocin receptors in the opioid oprm1 (-/-) mouse model of autism. Front. Pediatr. 2:91. 10.3389/fped.2014.00091
    1. Gil M., Bhatt R., Picotte K. B., Hull E. M. (2011). Oxytocin in the medial preoptic area facilitates male sexual behavior in the rat. Horm. Behav. 59, 435–443. 10.1016/j.yhbeh.2010.12.012
    1. Gimpl G., Fahrenholz F. (2001). The oxytocin receptor system: structure, function, and regulation. Physiol. Rev. 81, 629–683. Available online at:
    1. Glovinsky D., Kalogeras K. T., Kirch D. G., Suddath R., Wyatt R. J. (1994). Cerebrospinal fluid oxytocin concentration in schizophrenic patients does not differ from control subjects and is not changed by neuroleptic medication. Schizophr. Res. 11, 273–276. 10.1016/0920-9964(94)90021-3
    1. Gogolla N., Leblanc J. J., Quast K. B., Südhof T. C., Fagiolini M., Hensch T. K. (2009). Common circuit defect of excitatory-inhibitory balance in mouse models of autism. J. Neurodev. Disord. 2, 172–181. 10.1007/s11689-009-9023-x
    1. Goldman M. B., Gomes A. M., Carter C. S., Lee R. (2011). Divergent effects of two different doses of intranasal oxytocin on facial affect discrimination in schizophrenic patients with and without polydipsia. Psychopharmacology (Berl). 216, 101–110. 10.1007/s00213-011-2193-8
    1. Goldman M., Marlow-O'Connor M., Torres I., Carter C. S. (2008). Diminished plasma oxytocin in schizophrenic patients with neuroendocrine dysfunction and emotional deficits. Schizophr. Res. 98, 247–255. 10.1016/j.schres.2007.09.019
    1. Green L., Fein D., Modahl C., Feinstein C., Waterhouse L., Morris M. (2001). Oxytocin and autistic disorder: alterations in peptide forms. Biol. Psychiatry 50, 609–613. 10.1016/S0006-3223(01)01139-8
    1. Gregory S. G., Connelly J. J., Towers A. J., Johnson J., Biscocho D., Markunas C. A., et al. . (2009). Genomic and epigenetic evidence for oxytocin receptor deficiency in autism. BMC Med. 7:62. 10.1186/1741-7015-7-62
    1. Gu X. L., Yu L. C. (2007). Involvement of opioid receptors in oxytocin-induced antinociception in the nucleus accumbens of rats. J. Pain 8, 85–90. 10.1016/j.jpain.2006.07.001
    1. Guastella A. J., Einfeld S. L., Gray K. M., Rinehart N. J., Tonge B. J., Lambert T. J., et al. . (2010). Intranasal oxytocin improves emotion recognition for youth with autism spectrum disorders. Biol. Psychiatry 67, 692–694. 10.1016/j.biopsych.2009.09.020
    1. Guastella A. J., Howard A. L., Dadds M. R., Mitchell P., Carson D. S. (2009). A randomized controlled trial of intranasal oxytocin as an adjunct to exposure therapy for social anxiety disorder. Psychoneuroendocrinology 34, 917–923. 10.1016/j.psyneuen.2009.01.005
    1. Gutkowska J., Jankowski M., Lambert C., Mukaddam-Daher S., Zingg H. H., McCann S. M. (1997). Oxytocin releases atrial natriuretic peptide by combining with oxytocin receptors in the heart. Proc. Natl. Acad. Sci. U.S.A. 94, 11704–11709. 10.1073/pnas.94.21.11704
    1. Hammock E. A., Levitt P. (2013). Oxytocin receptor ligand binding in embryonic tissue and postnatal brain development of the C57BL/6J mouse. Front. Behav. Neurosci. 7:195. 10.3389/fnbeh.2013.00195
    1. Han Y., Yu L. C. (2009). Involvement of oxytocin and its receptor in nociceptive modulation in the central nucleus of amygdala of rats. Neurosci. Lett. 454, 101–104. 10.1016/j.neulet.2009.02.062
    1. Harris M. C., Jones P. M., Robinson I. C. (1981). Differences in the release of oxytocin into the blood and cerebrospinal fluid following hypothalamic and pituitary stimulation in rats. J. Physiol. 320, 109–110.
    1. Hoge E. A., Lawson E. A., Metcalf C. A., Keshaviah A., Zak P. J., Pollack M. H., et al. . (2012). Plasma oxytocin immunoreactive products and response to trust in patients with social anxiety disorder. Depress. Anxiety. 29, 924–930. 10.1002/da.21973
    1. Hoge E. A., Pollack M. H., Kaufman R. E., Zak P. J., Simon N. M. (2008). Oxytocin levels in social anxiety disorder. CNS Neurosci. Ther. 14, 165–170. 10.1111/j.1755-5949.2008.00051.x
    1. Holden K. L., McLaughlin E. J., Reilly E. L., Overall J. E. (1988). Accelerated mental aging in alcoholic patients. J. Clin. Psychol. 44, 286–292.
    1. Holder J. L., Jr., Butte N. F., Zinn A. R. (2000). Profound obesity associated with a balanced translocation that disrupts the SIM1 gene. Hum. Mol. Genet. 9, 101–108. 10.1093/hmg/9.1.101
    1. Hollander E., Bartz J., Chaplin W., Phillips A., Sumner J., Soorya L., et al. . (2007). Oxytocin increases retention of social cognition in autism. Biol. Psychiatry 61, 498–503. 10.1016/j.biopsych.2006.05.030
    1. Hollander E., Novotny S., Hanratty M., Yaffe R., DeCaria C. M., Aronowitz B. R., et al. . (2003). Oxytocin infusion reduces repetitive behaviors in adults with autistic and Asperger's disorders. Neuropsychopharmacology 1, 193–198. 10.1038/sj.npp.1300021
    1. Holzer J. C., Goodman W. K., McDougle C. J., Baer L., Boyarsky B. K., Leckman J. F., et al. . (1994). Obsessive-compulsive disorder with and without a chronic tic disorder. A comparison of symptoms in 70 patients. Br. J. Psychiatry 164, 469–473. 10.1192/bjp.164.4.469
    1. Huang H., Michetti C., Busnelli M., Managò F., Sannino S., Scheggia E., et al. . (2014). Chronic and acute intranasal oxytocin produce divergent social effects in mice. Neuropsychopharmacology 39, 1102–1114. 10.1038/npp.2013.310
    1. Ibragimov R., Kovács G. L., Szabó G., Telegdy G. (1987). Microinjection of oxytocin into limbic-mesolimbic brain structures disrupts heroin self-administration behavior: a receptor-mediated event? Life Sci. 41, 1265–1271. 10.1016/0024-3205(87)90205-0
    1. Insel T. R., Fernald R. D. (2004). How the brain processes social information: searching for the social brain. Annu. Rev. Neurosci. 27, 697–722. 10.1146/annurev.neuro.27.070203.144148
    1. Insel T. R., Hulihan T. J. (1995). A gender-specific mechanism for pair bonding: oxytocin and partner preference formation in monogamous voles. Behav. Neurosci. 109, 782–789. 10.1037/0735-7044.109.4.782
    1. Insel T. R., O'Brien D. J., Leckman J. F. (1999). Oxytocin, vasopressin, and autism: is there a connection? Biol. Psychiatry 45, 145–157. 10.1016/S0006-3223(98)00142-5
    1. Insel T. R., Young L. J. (2001). The neurobiology of attachment. Nat. Rev. Neurosci. 2, 129–136. 10.1038/35053579
    1. Ivell R., Richter D. (1984). Structure and comparison of the oxytocin and vasopressin genes from rat. Proc. Natl. Acad. Sci. U.S.A. 81, 2006–2010. 10.1073/pnas.81.7.2006
    1. Iwasaki Y., Maejima Y., Suyama S., Yoshida M., Arai T., Katsurada K., et al. . (2015). Peripheral oxytocin activates vagal afferent neurons to suppress feeding in normal and leptin-resistant mice: a route forameliorating hyperphagia and obesity. Am. J. Physiol. Regul. Integr. Comp. Physiol. 308, R360–R369. 10.1152/ajpregu.00344.2014
    1. Jacob S., Brune C. W., Carter C. S., Leventhal B. L., Lord C., Cook E. H., et al. . (2007). Association of the oxytocin receptor gene (OXTR) in caucasian children and adolescents with autism. Neurosci. Lett. 417, 6–9. 10.1016/j.neulet.2007.02.001
    1. Jankowski M., Hajjar F., Kawas S. A., Mukaddam-Daher S., Hoffman G., McCann S. M., et al. . (1998). Rat heart: a site of oxytocin production and action. Proc. Natl. Acad. Sci. U.S.A. 95, 14558–14563. 10.1073/pnas.95.24.14558
    1. Jankowski M., Wang D., Hajjar F., Mukaddam-Daher S., McCann S. M., Gutkowska J. (2000). Oxytocin and its receptors are synthesized in the rat vasculature. Proc. Natl. Acad. Sci. U.S.A. 97, 6207–6211. 10.1073/pnas.110137497
    1. Jansen L. M., Gispen-de Wied C. C., Wiegant V. M., Westenberg H. G., Lahuis B. E., van Engeland H. (2006). Autonomic and neuroendocrine responses to a psychosocial stressor in adults with autistic spectrum disorder. J. Autism Dev. Disord. 36, 891–899. 10.1007/s10803-006-0124-z
    1. Jenike M. A. (1990). Obsessive–Compulsive Disorders in Pregnancy and Childbirth. Littleton: Year Book Medical.
    1. Johnstone L. E., Fong T. M., Leng G. (2006). Neuronal activation in the hypothalamus and brainstem during feeding in rats. Cell Metab. 4, 313–321. 10.1016/j.cmet.2006.08.003
    1. Jones P. M., Robinson I. C. (1982). Differential clearance of neurophysin and neurohypophysial peptides from the cerebrospinal fluid in conscious guinea pigs. Neuroendocrinology 34, 297–302. 10.1159/000123316
    1. Kasama S., Furuya M., Toyama T., Ichikawa S., Kurabayashi M. (2008). Effect of atrial natriuretic peptide on left ventricular remodelling in patients with acute myocardial infarction. Eur. Heart J. 29, 1485–1494. 10.1093/eurheartj/ehn206
    1. Kimura T., Tanizawa O., Mori K., Brownstein M. J., Okayama H. (1992). Structure and expression of a human oxytocin receptor. Nature 356, 526–529. 10.1038/356526a0
    1. Kirsch P., Esslinger C., Chen Q., Mier D., Lis S., Siddhanti S., et al. . (2005). Oxytocin modulates neural circuitry for social cognition and fear in humans. J. Neurosci. 25, 11489–11493. 10.1523/JNEUROSCI.3984-05.2005
    1. Kiss A., Bundzikova J., Pirnik Z., Mikkelsen J. D. (2010). Different antipsychotics elicit different effects on magnocellular oxytocinergic and vasopressinergic neurons as revealed by Fos immunohistochemistry. J. Neurosci. Res. 88, 677–685. 10.1002/jnr.22226
    1. Klein D. F., Skrobala A. M., Garfinkel R. S. (1994–1995). Preliminary look at the effects of pregnancy on the course of panic disorder. Anxiety 1, 227–232.
    1. Knobloch H. S., Charlet A., Hoffmann L. C., Eliava M., Khrulev S., Cetin A. H., et al. . (2012). Evoked axonal oxytocin release in the central amygdala attenuates fear response. Neuron 73, 553–566. 10.1016/j.neuron.2011.11.030
    1. Kosaka H., Munesue T., Ishitobi M., Asano M., Omori M., Sato M., et al. . (2012). Long-term oxytocin administration improves social behaviors in a girl with autistic disorder. BMC Psychiatry 12:110. 10.1186/1471-244X-12-110
    1. Kovács C. L., Van Ree J. M. (1985). Behaviorally active oxytocin fragments simultaneously attenuate heroin self-administration and tolerance in rats. Life Sci. 37, 1895–1900. 10.1016/0024-3205(85)90007-4
    1. Kovács G. L., Borthaiser Z., Telegdy G. (1985). Oxytocin reduces intravenous heroin self-administration in heroin-tolerant rats. Life Sci. 37, 17–26. 10.1016/0024-3205(85)90620-4
    1. Kovàcs G. L., Sarnyai Z., Barbarczi E., Szabó G., Telegdy G. (1990). The role of oxytocin-dopamine interactions in cocaine-induced locomotor hyperactivity. Neuropharmacology 29, 365–368. 10.1016/0028-3908(90)90095-9
    1. Kovács G. L., Sarnyai Z., Szabó G. (1998). Oxytocin and addiction: a review. Psychoneuroendocrinology 23, 945–962. 10.1016/S0306-4530(98)00064-X
    1. Kovács G. L., Telegdy G. (1987). Beta-endorphin tolerance is inhibited by oxytocin. Pharmacol. Biochem. Behav. 26, 57–60. 10.1016/0091-3057(87)90533-8
    1. Kublaoui B. M., Gemelli T., Tolson K. P., Wang Y., Zinn A. R. (2008). Oxytocin deficiency mediates hyperphagic obesity of Sim1 haploinsufficient mice. Mol. Endocrinol. 22, 1723–1734. 10.1210/me.2008-0067
    1. Labuschagne I., Phan K. L., Wood A., Angstadt M., Chua P., Heinrichs M., et al. . (2010). Oxytocin attenuates amygdala reactivity to fear in generalized social anxiety disorder. Neuropsychopharmacology 35, 2403–2413. 10.1038/npp.2010.123
    1. Landgraf R., Neumann I. D. (2004). Vasopressin and oxytocin release within the brain: a dynamic concept of multiple and variable modes of neuropeptide communication. Front. Neuroendocrinol. 25, 150–176. 10.1016/j.yfrne.2004.05.001
    1. László K., Kovács A., Zagoracz O., Ollmann T., Péczely L., Kertes E., et al. . (2015). Positive reinforcing effect of oxytocin microinjection in the rat central nucleus of amygdala. Behav. Brain Res. 296, 279–285. 10.1016/j.bbr.2015.09.021
    1. Lawson E. A., Donoho D. A., Blum J. I., Meenaghan E. M., Misra M., Herzog D. B., et al. . (2011). Decreased nocturnal oxytocin levels in anorexia nervosa are associated with low bone mineral density and fat mass. J. Clin. Psychiatry 72, 1546–1551. 10.4088/JCP.10m06617
    1. Lazzari V. M., Becker R. O., de Azevedo M. S., Morris M., Rigatto K., Almeida S., et al. . (2013). Oxytocin modulates social interaction but is not essential for sexual behavior in male mice. Behav. Brain Res. 1, 130–136. 10.1016/j.bbr.2013.01.025
    1. Leckman J. F., Goodman W. K., North W. G., Chappell P. B., Price L. H., Pauls D. L., et al. . (1994). Elevated cerebrospinal fluid levels of oxytocin in obsessive-compulsive disorder. Comparison with Tourette's syndrome and healthy controls. Arch. Gen. Psychiatry 51, 782–792. 10.1001/archpsyc.1994.03950100030003
    1. Leckman J. F., Grice D. E., Barr L. C., de Vries A. L., Martin C., Cohen D. J., et al. . (1994–1995). Tic-related vs. non-tic-related obsessive compulsive disorder. Anxiety 1, 208–215.
    1. Legros J. J., Gazzotti C., Carvelli T., Franchimont P., Timsit-Berthier M., von Frenckell R., et al. . (1992). Apomorphine stimulation of vasopressin- and oxytocin-neurophysins. Evidence for increased oxytocinergic and decreased vasopressinergic function in schizophrenics. Psychoneuroendocrinology 17, 611–617. 10.1016/0306-4530(92)90019-4
    1. Legros J. J., Geenen V. (1996). Neurophysins in central diabetes insipidus. Horm. Res. 45, 182–186. 10.1159/000184784
    1. Legros J. J., Geenen V., Linkowski P., Mendlewicz J. (1983). Increased neurophysin I and II cerbrospinal fluid concentration from bipolar versus unipolar depressed patients. Neuroendocrinol. Lett. 5, 201–205.
    1. Leng G., Caquineau C., Sabatier N. (2005). Regulation of oxytocin secretion. Vitam. Horm. 71, 27–58. 10.1016/S0083-6729(05)71002-5
    1. Lerer E., Levi S., Salomon S., Darvasi A., Yirmiya N., Ebstein R. P. (2008). Association between the oxytocin receptor (OXTR) gene and autism: Relationship to vineland adaptive behavior scales and cognition. Mol. Psychiatry 13, 980–988. 10.1038/sj.mp.4002087
    1. Linkowski P., Geenen V., Kerkhofs M., Mendlewicz J., Legros J. J. (1984). Cerebrospinal fluid neurophysins in affective illness and in schizophrenia. Eur. Arch. Psychiatry Neurol. Sci. 234, 162–165. 10.1007/BF00461555
    1. Lord C., Rutter M., Le Couteur A. (1994). Autism Diagnostic Interview-Revised: a revised version of a diagnostic interview for caregivers of individuals with possible pervasive developmental disorders. J. Autism Dev. Disord. 24, 659–685. 10.1007/BF02172145
    1. Love T. M., Enoch M. A., Hodgkinson C. A., Peciña M., Mickey B., Koeppe R. A., et al. . (2012). Oxytocin gene polymorphisms influence human dopaminergic function in a sex-dependent manner. Biol. Psychiatry 72, 198–206. 10.1016/j.biopsych.2012.01.033
    1. Ludwig M. (1998). Dendritic release of vasopressin and oxytocin. J. Neuroendocrinol. 10, 881–895. 10.1046/j.1365-2826.1998.00279.x
    1. Ludwig M., Leng G. (2006). Dendritic peptide release and peptidedependentbehaviours. Nat. Rev. Neurosci. 7, 126–136. 10.1038/nrn1845
    1. Lukas M., Toth I., Veenema A. H., Neumann I. D. (2013). Oxytocin mediates rodent social memory within the lateral septum and the medial amygdala depending on the relevance of the social stimulus: male juvenile versus female adult conspecifics. Psychoneuroendocrinology 38, 916–926. 10.1016/j.psyneuen.2012.09.018
    1. Lundeberg T., Meister B., Björkstrand E., Uvnas-Moberg K. (1993). Oxytocin modulates the effects of galanin in carrageenan-induced hyperalgesia in rats. Brain Res. 608, 181–185. 10.1016/0006-8993(93)91456-3
    1. Macdonald K., Feifel D. (2012). Oxytocin in schizophrenia: a review of evidence for its therapeutic effects. Acta Neuropsychiatr. 24, 130–146. 10.1111/j.1601-5215.2011.00634.x
    1. Maejima Y., Iwasaki Y., Yamahara Y., Kodaira M., Sedbazar U., Yada T. (2011). Peripheral oxytocin treatment ameliorates obesity by reducing food intake and visceral fat mass. Aging (Albany NY) 3, 1169–1177. 10.1155/2010/289645
    1. Maejima Y., Sedbazar U., Suyama S., Kohno D., Onaka T., Takano E., et al. . (2009). Nesfatin-1-regulated oxytocinergic signaling in the paraventricular nucleus causes anorexia through a leptin independent melanocortin pathway. Cell Metab. 10, 355–365. 10.1016/j.cmet.2009.09.002
    1. Maes M., Monteleone P., Bencivenga R., Goossens F., Maj M., van West D., et al. . (2001). Lower serum activity of prolyl endopeptidase in anorexia and bulimia nervosa. Psychoneuroendocrinology 26, 17–26. 10.1016/S0306-4530(00)00032-9
    1. Maguire S., O'Dell A., Touyz L., Russell J. (2013). Oxytocin and anorexia nervosa: a review of the emerging literature. Eur. Eat. Disord. Rev. 21, 475–478. 10.1002/erv.2252
    1. Marazziti D., Catena Dell'osso M. (2008). The role of oxytocin in neuropsychiatric disorders. Curr. Med. Chem. 15, 698–704. 10.2174/092986708783885291
    1. Marchesi C., Chiodera P., Brusamonti E., Volpi R., Coiro V. (1997). Abnormal plasma oxytocin and beta-endorphin levels in alcoholics after short and long term abstinence. Prog. Neuropsychopharmacol. Biol. Psychiatry 21, 797–807. 10.1016/S0278-5846(97)00080-8
    1. Marlin B. J., Mitre M., D'amour J. A., Chao M. V., Froemke R. C. (2015). Oxytocin enables maternal behaviour by balancing cortical inhibition. Nature 520, 499–504. 10.1038/nature14402
    1. Matsuura K., Nagai T., Nishigaki N., Oyama T., Nishi J., Wada H., et al. . (2004). Adult cardiac Sca-1-positive cells differentiate into beating cardiomyocytes. J. Biol. Chem. 279, 11384–11391. 10.1074/jbc.M310822200
    1. Mazzuca M., Minlebaev M., Shakirzyanova A., Tyzio R., Taccola G., Janackova S., et al. . (2011). Newborn analgesia mediated by oxytocin during delivery. Front. Cell. Neurosci. 5:3. 10.3389/fncel.2011.00003
    1. McCann M. J., Rogers R. C. (1990). Oxytocin excites gastric-related neurones in rat dorsal vagal complex. J. Physiol. (Lond). 428, 95–108. 10.1113/jphysiol.1990.sp018202
    1. McCarthy M. M., McDonald C. H., Brooks P. J., Goldman D. (1996). An anxiolytic action of oxytocin is enhanced by estrogen in the mouse. Physiol. Behav. 60, 1209–1215. 10.1016/S0031-9384(96)00212-0
    1. McDougle C. J., Barr L. C., Goodman W. K., Price L. H. (1999). Possible role of neuropeptides in obsessive compulsive disorder. Psychoneuroendocrinology 24, 1–24. 10.1016/S0306-4530(98)00046-8
    1. Melis M. R., Melis T., Cocco C., Succu S., Sanna F., Pillolla G., et al. . (2007). Oxytocin injected into the ventral tegmental area induces penile erection and increases extracellular dopamine in the nucleus accumbens and paraventricular nucleus of the hypothalamus of male rats. Eur. J. Neurosci. 26, 1026–1035. 10.1111/j.1460-9568.2007.05721.x
    1. Mens W. B., Laczi F., Tonnaer J. A., de Kloet E. R., van Wimersma Greidanus T. B. (1983). Vasopressin and oxytocin content in cerebrospinal fluid and in various brain areas after administration of histamine and pentylenetetrazol. Pharmacol. Biochem. Behav. 19, 587–591. 10.1016/0091-3057(83)90332-5
    1. Meyer C., Freund-Mercier M. J., Guerné Y., Richard P. (1987). Relationship between oxytocin release and amplitude of oxytocin cell neurosecretory bursts during suckling in the rat. J. Endocrinol. 114, 263–270. 10.1677/joe.0.1140263
    1. Meynen G., Unmehopa U. A., Hofman M. A., Swaab D. F., Hoogendijk W. J. (2007). Hypothalamic oxytocin mRNA expression and melancholic depression. Mol. Psychiatry 12, 118–119. 10.1038/sj.mp.4001911
    1. Meziane H., Schaller F., Bauer S., Villard C., Matarazzo V., Riet F., et al. . (2014). An early postnatal oxytocin treatment prevents social and learning deficits in adult mice deficient for Magel2, a gene involved in Prader-Willi Syndrome and autism. Biol Psychiatry 78, 85–94. 10.1016/j.biopsych.2014.11.010
    1. Mizuno A., Cherepanov S. M., Kikuchi Y., Fakhrul A. A., Akther S., Deguchi K., et al. . (2015). Lipo-oxytocin-1, a novel oxytocin analog conjugated with two palmitoyl groups, has long-lasting effects on anxiety-related behavior and social avoidance in CD157 knockout mice. Brain Sci. 5, 3–13. 10.3390/brainsci5010003
    1. Modabbernia A., Rezaei F., Salehi B., Jafarinia M., Ashrafi M., Tabrizi M., et al. . (2013). Intranasal oxytocin as an adjunct to risperidone in patients with schizophrenia: an 8-week, randomized, double-blind, placebo-controlled study. CNS Drugs 27, 57–65. 10.1007/s40263-012-0022-1
    1. Modahl C., Green L., Fein D., Morris M., Waterhouse L., Feinstein C., et al. . (1998). Plasma oxytocin levels in autistic children. Biol. Psychiatry 43, 270–277. 10.1016/S0006-3223(97)00439-3
    1. Modell J. G., Mountz J. M., Curtis G. C., Greden J. F. (1989). Neurophysiologic dysfunction in basal ganglia/limbic striatal and thalamocortical circuits as a pathogenetic mechanism of obsessive-compulsive disorder. J. Neuropsychiatry Clin. Neurosci. 1, 27–36. 10.1176/jnp.1.1.27
    1. Montag C., Brockmann E. M., Bayerl M., Rujescu D., Müller D. J., Gallinat J. (2013). Oxytocin and oxytocin receptor gene polymorphisms and risk for schizophrenia: a case-control study. World J. Biol. Psychiatry 14, 500–508. 10.3109/15622975.2012.677547
    1. Morris J. F., Pow D. V. (1991). Widespread release of peptides in the central nervous system: quantitation of tannic acid-captured exocytoses. Anat. Rec. 231, 437–445. 10.1002/ar.1092310406
    1. Morton G. J., Thatcher B. S., Reidelberger R. D., Ogimoto K., WoldenHanson T., Baskin D. G., et al. . (2012). Peripheral oxytocin suppresses food intake and causes weight loss in diet-induced obese rats. Am. J. Physiol. Endocrinol. Metab. 302, E134–E144. 10.1152/ajpendo.00296.2011
    1. Moy S. S., Nadler J. J., Young N. B., Perez A., Holloway L. P., Barbaro R. P., et al. . (2007). Mouse behavioural tasks relevant to autism: phenotypes of 10 inbred strains. Behav. Brain Res. 176, 4–20. 10.1016/j.bbr.2006.07.030
    1. Muchmore D. B., Little S. A., de Haën C. (1981). A dual mechanism of action of ocytocin in rat epididymal fat cells. J. Biol. Chem. 256, 365–372.
    1. Muhle R., Trentacoste S. V., Rapin I. (2004). The genetics of autism. Pediatrics 113, e472–e486. 10.1542/peds.113.5.e472
    1. Mühlethaler M., Charpak S., Dreifuss J. J. (1984). Contrasting effects of neurohypophysial peptides on pyramidal and non-pyramidal neurones in the rat hippocampus. Brain Res. 308, 97–107. 10.1016/0006-8993(84)90921-1
    1. Muscatelli F., Abrous D. N., Massacrier A., Boccaccio I., Le Moal M., Cau P., et al. . (2000). Disruption of the mouse Necdin gene results in hypothalamic and behavioural alterations reminiscent of the human Prader-Willi syndrome. Hum. Mol. Genet. 9, 3101–3110. 10.1093/hmg/9.20.3101
    1. Myers A. J., Williams L., Gatt J. M., McAuley-Clark E. Z., Dobson-Stone C., Schofield P. R., et al. . (2014). 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. 59, 93–100. 10.1016/j.jpsychires.2014.08.021
    1. Nelson E. E., Alberts J. R., Tian Y., Verbalis J. G. (1998). Oxytocin is elevated in plasma of 10-day-old rats following gastric distension. Brain Res. Dev. Brain Res. 111, 301–303. 10.1016/S0165-3806(98)00147-3
    1. Neumann I., Douglas A. J., Pittman Q. J., Russell J. A., Landgraf R. (1996). Oxytocin released within the supraoptic nucleus of the rat brain by positive feedback action is involved in parturition-related events. J. Neuroendocrinol. 8, 227–233. 10.1046/j.1365-2826.1996.04557.x
    1. Neumann I. D., Torner L., Wigger A. (2000). Brain oxytocin: differential inhibition of neuroendocrine stress responses and anxiety-related behaviour in virgin, pregnant and lactating rats. Neuroscience 95, 567–575. 10.1016/S0306-4522(99)00433-9
    1. Neziroglu F., Anemone R., Yaryura-Tobias J. A. (1992). Onset of obsessive-compulsive disorder in pregnancy. Am. J. Psychiatry 149, 947–950. 10.1176/ajp.149.7.947
    1. Nissen E., Gustavsson P., Widström A. M., Uvnäs-Moberg K. (1998). Oxytocin, prolactin, milk production and their relationship with personality traits in women after vaginal delivery or Cesarean section. J. Psychosom. Obstet. Gynaecol. 19, 49–58. 10.3109/01674829809044221
    1. Olff M., Frijling J. L., Kubzansky L. D., Bradley B., Ellenbogen M. A., Cardoso C., et al. . (2013). The role of oxytocin in social bonding, stress regulation and mental health: an update on the moderating effects of context and interindividual differences. Psychoneuroendocrinology 38, 1883–1894. 10.1016/j.psyneuen.2013.06.019
    1. Olson B. R., Drutarosky M. D., Chow M. S., Hruby V. J., Stricker E. M., Verbalis J. G. (1991). Oxytocin and an oxytocin agonist administered centrally decrease food intake in rats. Peptides 199, 113–118. 10.1016/0196-9781(91)90176-P
    1. Olson B. R., Hoffman G. E., Sved A. F., Stricker E. M., Verbalis J. G. (1992). Cholecystokinin induces c-fos expression in hypothalamic oxytocinergic neurons projecting to the dorsal vagal complex. Brain Res. 569, 238–248. 10.1016/0006-8993(92)90635-M
    1. Onaka T. (2004). Neural pathways controlling central and peripheral oxytocin release during stress. J. Neuroendocrinol. 16, 308–312. 10.1111/j.0953-8194.2004.01186.x
    1. Ondrejcakova M., Ravingerova T., Bakos J., Pancza D., Jezova D. (2009). Oxytocin exerts protective effects on in vitro myocardial injury induced by ischemia and reperfusion. Can. J. Physiol. Pharmacol. 87, 137–142. 10.1139/Y08-108
    1. Ott V., Finlayson G., Lehnert H., Heitmann B., Heinrichs M., Born J., et al. . (2013). Oxytocin reduces reward-driven food intake in humans. Diabetes 62, 3418–3425. 10.2337/db13-0663
    1. Owen S. F., Tuncdemir S. N., Bader P. L., Tirko N. N., Fishell G., Tsien R. W. (2013). Oxytocin enhances hippocampal spike transmission by modulating fast-spiking interneurons. Nature 500, 458–462. 10.1038/nature12330
    1. Oyama T., Nagai T., Wada H., Naito A. T., Matsuura K., Iwanaga K., et al. . (2007). Cardiac side population cells have a potential to migrate and differentiate into cardiomyocytes in vitro and in vivo. J. Cell Biol. 176, 329–341. 10.1083/jcb.200603014
    1. Ozsoy S., Esel E., Kula M. (2009). Serum oxytocin levels in patients with depression and the effects of gender and antidepressant treatment. Psychiatry Res. 169, 249–252. 10.1016/j.psychres.2008.06.034
    1. Page S. R., Ang V. T., Jackson R., White A., Nussey S. S., Jenkins J. S. (1990). The effect of oxytocin infusion on adenohypophyseal function in man. Clin. Endocrinol. (Oxf). 32, 307–313. 10.1111/j.1365-2265.1990.tb00871.x
    1. Peñagarikano O., Lázaro M. T., Lu X. H., Gordon A., Dong H., Lam H. A., et al. . (2015). Exogenous and evoked oxytocin restores social behavior in the Cntnap2 mouse model of autism. Sci. Transl. Med. 7:271ra8. 10.1126/scitranslmed.3010257
    1. Petersson M., Alster P., Lundeberg T., Uvnäs-Moberg K. (1996). Oxytocin causes a long-term decrease of blood pressure in female and male rats. Physiol. Behav. 60, 1311–1315. 10.1016/S0031-9384(96)00261-2
    1. Pierce R. C., Kumaresan V. (2006). The mesolimbic dopamine system: the final common pathway for the reinforcing effect of drugs of abuse? Neurosci. Biobehav. Rev. 30, 215–238. 10.1016/j.neubiorev.2005.04.016
    1. Pierrehumbert B., Torrisi R., Laufer D., Halfon O., Ansermet F., Beck Popovic M. (2010). Oxytocin response to an experimental psychosocial challenge in adults exposed to traumatic experiences during childhood or adolescence. Neuroscience 166, 168–177. 10.1016/j.neuroscience.2009.12.016
    1. Pittman Q. J., Blume H. W., Renaud L. P. (1981). Connections of the hypothalamic paraventricular nucleus with the neurohypophysis, median eminence, amygdala, lateral septum and midbrain periaqueductal gray: an electrophysiological study in the rat. Brain Res. 215, 15–28. 10.1016/0006-8993(81)90488-1
    1. Pitts A. F., Samuelson S. D., Meller W. H., Bissette G., Nemeroff C. B., Kathol R. G. (1995). Cerebrospinal fluid corticotropin-releasing hormone, vasopressin, and oxytocin concentrations in treated patients with major depression and controls. Biol. Psychiatry 38, 330–335. 10.1016/0006-3223(95)00229-A
    1. Purba J. S., Hoogendijk W. J., Hofman M. A., Swaab D. F. (1996). Increased number of vasopressin-and oxytocin-expressing neurons in the paraventricular nucleus of the hypothalamus in depression. Arch. Gen. Psychiatry 53, 137–143. 10.1001/archpsyc.1996.01830020055007
    1. Qin J., Feng M., Wang C., Ye Y., Wang P. S., Liu C. (2009). Oxytocin receptor expressed on the smooth muscle mediates the excitatory effect of oxytocin on gastric motility in rats. Neurogastroenterol. Motil. 21, 430–438. 10.1111/j.1365-2982.2009.01282.x
    1. Rapoport J. L., Wise S. P. (1988). Obsessive-compulsive disorder: evidence for basal ganglia dysfunction. Psychopharmacol. Bull. 24, 380–384.
    1. Rault J. L., Carter C. S., Garner J. P., Marchant-Forde J. N., Richert B. T., Lay D. C., et al. . (2013). Repeated intranasal oxytocin administration in early life dysregulates the HPA axis and alters social behavior. Physiol. Behav. 112–113, 40–48. 10.1016/j.physbeh.2013.02.007
    1. Reeta K. h., Mediratta P. K., Rathi N., Jain H., Chugh C., Sharma K. K. (2006). Role of kappa- and delta-opioid receptors in the antinociceptive effect of oxytocin in formalin-induced pain response in mice. Regul. Pept. 135, 85–90. 10.1016/j.regpep.2006.04.004
    1. Renaud L. P., Tang M., McCann M. J., Stricker E. M., Verbalis J. G. (1987). Cholecystokinin and gastric distension activate oxytocinergic cells in rat hypothalamus. Am. J. Physiol. 253, R661–R665.
    1. Rhodes C. H., Morrell J. I., Pfaff D. W. (1981). Immunohistochemical analysis of magnocellular elements in rat hypothalamus: distribution and numbers of cells containing neurophysin, oxytocin, and vasopressin. J. Comp. Neurol. 198, 45–64. 10.1002/cne.901980106
    1. Richard P., Moos F., Dayanithi G., Gouzènes L., Sabatier N. (1997). Rhythmic activities of hypothalamic magnocellular neurons: autocontrol mechanisms. Biol. Cell. 89, 555–560. 10.1111/j.1768-322X.1997.tb01032.x
    1. Rinaman L. (1998). Oxytocinergic inputs to the nucleus of the solitary tract and dorsal motor nucleus of the vagus in neonatal rats. J. Comp. Neurol. 399, 101–109.
    1. Robbe D., Kopf M., Remaury A., Bockaert J., Manzoni O. J. (2002). Endogenous cannabinoids mediate long-term synaptic depression in the nucleus accumbens. Proc. Natl. Acad. Sci. U.S.A. 99, 8384–8388. 10.1073/pnas.122149199
    1. Robinson G., Evans J. J. (1990). Oxytocin has a role in gonadotrophin regulation in rats. J. Endocrinol. 125, 425–432. 10.1677/joe.0.1250425
    1. Rogers R. C., Hermann G. E. (1987). Oxytocin, oxytocin antagonist, TRH, and hypothalamic paraventricular nucleus stimulation effects on gastric motility. Peptides 8, 505–513. 10.1016/0196-9781(87)90017-9
    1. Ross H. E., Young L. J. (2009). Oxytocin and the neural mechanisms regulating social cognition and affiliative behavior. Front. Neuroendocrinol. 30, 534–547. 10.1016/j.yfrne.2009.05.004
    1. Rubenstein J. L., Merzenich M. M. (2003). Model of autism: increased ratio of excitation/inhibition in key neural systems. Genes Brain Behav. 5, 255–267. 10.1034/j.1601-183X.2003.00037.x
    1. Russell J. A., Leng G., Douglas A. J. (2003). The magnocellular oxytocin system, the fount of maternity: adaptations in pregnancy. Front. Neuroendocrinol. 24, 27–61. 10.1016/S0091-3022(02)00104-8
    1. Ryan B. C., Young N. B., Crawley J. N., Bodfish J. W., Moy S. S. (2009). Social deficits, stereotypy and early emergence of repetitive behavior in the C58/J inbred mouse strain. Behav. Brain Res. 208, 178–188. 10.1016/j.bbr.2009.11.031
    1. Salonia A., Nappi R. E., Pontillo M., Daverio R., Smeraldi A., Briganti A., et al. . (2005). Menstrual cycle-related changes in plasma oxytocin are relevant to normal sexual function in healthy women. Horm. Behav. 47, 164–169. 10.1016/j.yhbeh.2004.10.002
    1. Sanders G., Freilicher J., Lightman S. L. (1990). Psychological stress of exposure to uncontrollable noise increases plasma oxytocin in high emotionality women. Psychoneuroendocrinology 15, 47–58. 10.1016/0306-4530(90)90046-C
    1. Sarnyai Z., Bíró E., Babarczy E., Vecsernyés M., Laczi F., Szabó G., et al. . (1992a). Oxytocin modulates behavioural adaptation to repeated treatment with cocaine in rats. Neuropharmacology 31, 593–598. 10.1016/0028-3908(92)90192-R
    1. Sarnyai Z., Kovács G. L. (1994). Role of oxytocin in the neuroadaptation to drugs of abuse. Psychoneuroendocrinology 19, 85–117. 10.1016/0306-4530(94)90062-0
    1. Sarnyai Z., Szabó G., Kovács G. L., Telegdy G. (1992b). Opposite actions of oxytocin and vasopressin in the development of cocaine-induced behavioral sensitization in mice. Pharmacol. Biochem. Behav. 43, 491–494. 10.1016/0091-3057(92)90182-F
    1. Sasayama D., Hattori K., Teraishi T., Hori H., Ota M., Yoshida S., et al. . (2012). Negative correlation between cerebrospinal fluid oxytocin levels and negative symptoms of male patients with schizophrenia. Schizophr. Res. 139, 201–206. 10.1016/j.schres.2012.06.016
    1. Scantamburlo G., Ansseau M., Geenen V., Legros J. J. (2011). Intranasal oxytocin as an adjunct to escitalopram in major depression. J. Neuropsychiatry Clin. Neurosci. 23:E5. 10.1176/jnp.23.2.jnpe5
    1. Scantamburlo G., Hansenne M., Fuchs S., Pitchot W., Maréchal P., Pequeux C., et al. . (2007). Plasma oxytocin levels and anxiety in patients with major depression. Psychoneuroendocrinology 32, 407–410. 10.1016/j.psyneuen.2007.01.009
    1. Scantamburlo G., Hansenne M., Fuchs S., Pitchot W., Pinto E., Reggers J., et al. . (2005). AVP- and OT-neurophysins response to apomorphine and clonidine in major depression. Psychoneuroendocrinology 30, 839–845. 10.1016/j.psyneuen.2005.04.015
    1. Schaller F., Watrin F., Sturny R., Massacrier A., Szepetowski P., Muscatelli F. (2010). A single postnatal injection of oxytocin rescues the lethal feeding behaviour in mouse newborns deficient for the imprinted Magel2 gene. Hum. Mol. Genet. 19, 4895–4905. 10.1093/hmg/ddq424
    1. Scott A. I., Shering P. A., Legros J. J., Whalley L. J. (1991). Improvement in depressive illness is not associated with altered release of neurophysins over a course of ECT. Psychiatry Res. 36, 65–73. 10.1016/0165-1781(91)90118-9
    1. Scott A. I., Whalley L. J., Bennie J., Bowler G. (1986). Oestrogen-stimulated neurophysin and outcome after electroconvulsive therapy. Lancet 1, 1411–1414. 10.1016/S0140-6736(86)91557-6
    1. Scott A. I., Whalley L. J., Legros J. J. (1989). Treatment outcome, seizure duration, and the neurophysin response to ECT. Biol. Psychiatry 25, 585–597. 10.1016/0006-3223(89)90219-9
    1. Shahrokh D. K., Zhang T. Y., Diorio J., Gratton A., Meaney M. J. (2010). Oxytocin-dopamine interactions mediate variations in maternal behavior in the rat. Endocrinology 151, 2276–2286. 10.1210/en.2009-1271
    1. Shin N. Y., Park H. Y., Jung W. H., Park J. W., Yun J. Y., Jang J. H., et al. . (2015). Effects of oxytocin on neural response to facial expressions in patients with Schizophrenia. Neuropsychopharmacology 40, 1919–1927. 10.1038/npp.2015.78
    1. Sichel D. A., Cohen L. S., Dimmock J. A., Rosenbaum J. F. (1993). Postpartum obsessive compulsive disorder: a case series. J. Clin. Psychiatry 54, 156–159.
    1. Smith J. E., Williams K., Burkett S., Glue P., Nutt D. J. (1990). Oxytocin and vasopressin responses to ECT. Psychiatry Res. 32, 201–202. 10.1016/0165-1781(90)90087-L
    1. Smith J., Williams K., Birkett S., Nicholson H., Glue P., Nutt D. J. (1994). Neuroendocrine and clinical effects of electroconvulsive therapy and their relationship to treatment outcome. Psychol. Med. 24, 547–555. 10.1017/S0033291700027707
    1. South M., Larson M. J., White S. E., Dana J., Crowley M. J. (2011). Better fear conditioning is associated with reduced symptom severity in autism spectrum disorders. Autism Res. 4, 412–421. 10.1002/aur.221
    1. Souza R. P., de Luca V., Meltzer H. Y., Lieberman J. A., Kennedy J. L. (2010). Schizophrenia severity and clozapine treatment outcome association with oxytocinergic genes. Int. J. Neuropsychopharmacol. 13, 793–798. 10.1017/S1461145710000167
    1. Strauss G. P., Keller W. R., Koenig J. I., Gold J. M., Frost K. H., Buchanan R. W. (2015). Plasma oxytocin levels predict social cue recognition in individuals with schizophrenia. Schizophr. Res. 162, 47–51. 10.1016/j.schres.2015.01.034
    1. Swaab D. F., Pool C. W., Nijveldt F. (1975). Immunofluorescence of vasopressin and oxytocin in the rat hypothalamo-neurohypophypopseal system. J. Neural Transm. 36, 195–215. 10.1007/BF01253126
    1. Swaab D. F., Purba J. S., Hofman M. A. (1995). Alterations in the hypothalamic paraventricular nucleus and its oxytocin neurons (putative satiety cells) in Prader-Willi syndrome: a study of five cases. J. Clin. Endocrinol. Metab. 80, 573–579.
    1. Swanson L. W., Kuypers H. G. (1980). The paraventricular nucleus of the hypothalamus: cytoarchitectonic subdivisions and organization of projections to the pituitary, dorsal vagal complex, and spinal cord as demonstrated by retrograde fluorescence double-labeling methods. J. Comp. Neurol. 194, 555–570. 10.1002/cne.901940306
    1. Swarbrick M. M., Evans D. S., Valle M. I., Favre H., Wu S. H., Njajou O. T., et al. . (2011). Replication and extension of association between common genetic variants in SIM1 and human adiposity. Obesity (Silver Spring) 19, 2394–2403. 10.1038/oby.2011.79
    1. Szabó G., Kovács G. L., Telegdy G. (1989). Intraventricular administration of neurohypophyseal hormones interferes with the development of tolerance to ethanol. Acta Physiol. Hung. 73, 97–103.
    1. Tachibana M., Kagitani-Shimono K., Mohri I., Yamamoto T., Sanefuji W., Nakamura A., et al. . (2013). Long-term administration of intranasal oxytocin is a safe and promising therapy for early adolescent boys with autism spectrum disorders. J. Child Adolesc. Psychopharmacol. 23, 123–127. 10.1089/cap.2012.0048
    1. Takayanagi Y., Kasahara Y., Onaka T., Takahashi N., Kawada T., Nishimori K. (2008). Oxytocin receptor-deficient mice developed late-onset obesity. Neuroreport 19, 951–955. 10.1097/WNR.0b013e3283021ca9
    1. Takayanagi Y., Yoshida M., Bielsky I. F., Ross H. E., Kawamata M., Onaka T., et al. . (2005). Pervasive social deficits, but normal parturition, in oxytocin receptor-deficient mice. Proc. Natl. Acad. Sci. U.S.A. 102, 16096–16101. 10.1073/pnas.0505312102
    1. Tang M., Zhang J., Xu L., Chen J. D. (2006). Implantable gastric stimulation alters expression of oxytocin- and orexin-containing neurons in the hypothalamus of rats. Obes. Surg. 16, 762–769. 10.1381/096089206777346745
    1. Tansey K. E., Brookes K. J., Hill M. J., Cochrane L. E., Gill M., Skuse D., et al. . (2010). Oxytocin receptor (OXTR) does not play a major role in the aetiology of autism: Genetic and molecular studies. Neurosci. Lett. 474, 163–167. 10.1016/j.neulet.2010.03.035
    1. Tauber M., Mantoulan C., Copet P., Jauregui J., Demeer G., Diene G., et al. . (2011). Oxytocin may be useful to increase trust in others and decrease disruptive behaviours in patients with Prader-Willi syndrome: a randomised placebo-controlled trial in 24 patients. Orphanet J. Rare Dis. 6:47. 10.1186/1750-1172-6-47
    1. Teltsh O., Kanyas-Sarner K., Rigbi A., Greenbaum L., Lerer B., Kohn Y. (2012). Oxytocin and vasopressin genes are significantly associated with schizophrenia in a large Arab-Israeli pedigree. Int. J. Neuropsychopharmacol. 15, 309–319. 10.1017/S1461145711001374
    1. Teng B. L., Nonneman R. J., Agster K. L., Nikolova V. D., Davis T. T., Riddick N. V., et al. . (2013). Prosocial effects of oxytocin in two mouse models of autism spectrum disorders. Neuropharmacology 72, 187–196. 10.1016/j.neuropharm.2013.04.038
    1. Thompson M. R., Callaghan P. D., Hunt G. E., Cornish J. L., McGregor I. S. (2007). A role for oxytocin and 5-HT(1A) receptors in the prosocial effects of 3,4 methylenedioxymethamphetamine (“ecstasy”). Neuroscience 146, 509–514. 10.1016/j.neuroscience.2007.02.032
    1. Tóth Z. E., Gallatz K., Fodor M., Palkovits M. (1999). Decussations of the descending paraventricular pathways to the brainstem and spinal cord autonomic centers. J. Comp. Neurol. 414, 255–266.
    1. Tyzio R., Cossart R., Khalilov I., Minlebaev M., Hübner C. A., Represa A., et al. . (2006). Maternal oxytocin triggers a transient inhibitory switch in GABA signaling in the fetal brain during delivery. Science 314, 1788–1792. 10.1126/science.1133212
    1. Tyzio R., Nardou R., Ferrari D. C., Tsintsadze T., Shahrokhi A., Eftekhari S., et al. . (2014). Oxytocin-mediated GABA inhibition during delivery attenuates autism pathogenesis in rodent offspring. Science 343, 675–679. 10.1126/science.1247190
    1. Ueta Y., Kannan H., Higuchi T., Negoro H., Yamaguchi K., Yamashita H. (2000). Activation of gastric afferents increases noradrenaline release in the paraventricular nucleus and plasma oxytocin level. J. Auton. Nerv. Syst. 78, 69–76. 10.1016/S0165-1838(99)00049-1
    1. Uvnäs-Moberg K., Bruzelius G., Alster P., Lundeberg T. (1993). The antinociceptive effect of non-noxious sensory stimulation is mediated partly through oxytocinergic mechanisms. Acta Physiol. Scand. 149, 199–204. 10.1111/j.1748-1716.1993.tb09612.x
    1. Van Erp A. M., Kruk M. R., Semple D. M., Verbeet D. W. (1993). Initiation of self-grooming in resting rats by local PVH infusion of oxytocin but not a -MSH. Brain Res. 607, 108–112. 10.1016/0006-8993(93)91494-D
    1. van Londen L., Goekoop J. G., van Kempen G. M., Frankhuijzen-Sierevogel A. C., Wiegant V. M., van der Velde E. A., et al. . (1997). Plasma levels of arginine vasopressin elevated in patients with major depression. Neuropsychopharmacology 17, 284–292. 10.1016/S0893-133X(97)00054-7
    1. Vannucchi G., Masi G., Toni C., Dell'Osso L., Marazziti D., Perugi G. (2014). Clinical features, developmental course, and psychiatric comorbidity of adult autism spectrum disorders. CNS Spectr. 19, 157–164. 10.1017/S1092852913000941
    1. van Tol H. H., van den Buuse M., de Jong W., Burbach J. P. (1988). Vasopressin and oxytocin gene expression in the supraoptic and paraventricular nucleus of the spontaneously hypertensive rat (SHR) during development of hypertension. Brain Res. 464, 303–311. 10.1016/0169-328X(88)90039-3
    1. Viero C., Shibuya I., Kitamura N., Verkhratsky A., Fujihara H., Katoh A., et al. . (2010). Oxytocin: crossing the bridge between basic science and pharmacotherapy. CNS Neurosci. Ther. 16, e138–e156. 10.1111/j.1755-5949.2010.00185.x
    1. Viviani D., Charlet A., van den Burg E., Robinet C., Hurni N., Abatis M., et al. . (2011). Oxytocin selectively gates fear responses through distinct outputs from the central amygdala. Science 333, 104–107. 10.1126/science.1201043
    1. von Eggelkraut-Gottanka R., Beck-Sickinger A. G. (2004). Biosynthesis of peptide hormones derived from precursor sequences. Curr. Med. Chem. 11, 2651–2665. 10.2174/0929867043364405
    1. Wei D., Lee D., Cox C. D., Karsten C. A., Peñagarikano O., Geschwind D. H., et al. . (2015). Endocannabinoid signaling mediates oxytocin-driven social reward. Proc. Natl. Acad. Sci. U.S.A. 112, 14084–14089. 10.1073/pnas.1509795112
    1. Whalley L. J., Eagles J. M., Bowler G. M., Bennie J. G., Dick H. R., McGuire R. J., et al. . (1987). Selective effects of ECT on hypothalamic-pituitary activity. Psychol. Med. 17, 319–328. 10.1017/S0033291700024855
    1. Whalley L. J., Rosie R., Dick H., Levy G., Watts A. G., Sheward W. J., et al. . (1982). Immediate increases in plasma prolactin and neurophysin but not other hormones after electroconvulsive therapy. Lancet 2, 1064–1068. 10.1016/S0140-6736(82)90004-6
    1. Wharton R. H., Bresnan M. J. (1989). Neonatal respiratory depression and delay in diagnosis in Prader-Willi syndrome. Dev. Med. Child Neurol. 31, 231–236. 10.1111/j.1469-8749.1989.tb03983.x
    1. Williams J. R., Insel T. R., Harbaugh C. R., Carter C. S. (1994). Oxytocin administered centrally facilitates formation of a partner preference in female prairie voles (Microtus ochrogaster). J. Neuroendocrinol. 6, 247–250. 10.1111/j.1365-2826.1994.tb00579.x
    1. Williams P. D., Bock M. G., Evans B. E., Freidinger R. M., Pettibone D. J. (1998). Progress in the development of oxytocin antagonists for use in preterm labor. Adv. Exp. Med. Biol. 449, 473–479. 10.1007/978-1-4615-4871-3_61
    1. Witt D. M., Carter C. S., Walton D. M. (1990). Central and peripheral effects of oxytocin administration in prairie voles (Microtus ochrogaster). Pharmacol. Biochem. Behav. 37, 63–69. 10.1016/0091-3057(90)90042-G
    1. Wolff K., Tsapakis E. M., Winstock A. R., Hartley D., Holt D., Forsling M. L., et al. . (2006). Vasopressin and oxytocin secretion in response to the consumption of ecstasy in a clubbing population. J. Psychopharmacol. 20, 400–410. 10.1177/0269881106061514
    1. Wu C. L., Doong M. L., Wang P. S. (2008). Involvement of cholecystokinin receptor in the inhibition of gastrointestinal motility by oxytocin in ovariectomized rats. Eur. J. Pharmacol. 580, 407–415. 10.1016/j.ejphar.2007.11.024
    1. Wu C. L., Hung C. R., Chang F. Y., Pau K. Y., Wang P. S. (2003). Pharmacological effects of oxytocin on gastric emptying and intestinal transit of a non-nutritive liquid meal in female rats. Naunyn Schmiedebergs. Arch. Pharmacol. 367, 406–413. 10.1007/s00210-003-0690-y
    1. Wu S., Jia M., Ruan Y., Liu J., Guo Y., Shuang M., et al. . (2005). Positive association of the oxytocin receptor gene (OXTR) with autism in the Chinese Han population. Biol. Psychiatry 58, 74–77. 10.1016/j.biopsych.2005.03.013
    1. Xi D., Gandhi N., Lai M., Kublaoui B. M. (2012). Ablation of Sim1 neurons causes obesity through hyperphagia and reduced energy expenditure. PLoS ONE 7:e36453. 10.1371/journal.pone.0036453
    1. Xu X. J., Wiesenfeld-Hallin Z. (1994). Intrathecal oxytocin facilitates the spinal nociceptive flexor reflex in the rat. Neuroreport 5, 750–752. 10.1097/00001756-199403000-00003
    1. Yang J. (1976). Intrathecal administration of oxytocin induces analgesia in low back pain involving the endogenous opiate peptide system. Spine (Phila Pa 1976) 19, 867–871. 10.1097/00007632-199404150-00001
    1. Yang J., Yang Y., Chen J. M., Liu W. Y., Wang C. H., Lin B. C. (2007b). Effect of oxytocin on acupuncture analgesia in the rat. Neuropeptides 41, 285–292. 10.1016/j.npep.2007.05.004
    1. Yang J., Yang Y., Chen J. M., Liu W. Y., Wang C. H., Lin B. C. (2007a). Central oxytocin enhances antinociception in the rat. Peptides 28, 1113–1119. 10.1016/j.peptides.2007.03.003
    1. Young K. A., Gobrogge K. L., Liu Y., Wang Z. (2011). The neurobiology of pair bonding: insights from a socially monogamous rodent. Front. Neuroendocrinol. 32, 53–69. 10.1016/j.yfrne.2010.07.006
    1. Young K. A., Liu Y., Wang Z. (2008). The neurobiology of social attachment: A comparative approach to behavioral, neuroanatomical, and neurochemical studies. Comp. Biochem. Physiol. C. Toxicol. Pharmacol. 148, 401–410. 10.1016/j.cbpc.2008.02.004
    1. Yrigollen C. M., Han S. S., Kochetkova A., Babitz T., Chang J. T., Volkmar F. R., et al. . (2008). Genes controlling affiliative behavior as candidate genes for autism. Biol. Psychiatry 63, 911–916. 10.1016/j.biopsych.2007.11.015
    1. Zaninetti M., Raggenbass M. (2000). Oxytocin receptor agonists enhance inhibitory synaptic transmission in the rat hippocampus by activating interneurons in stratum pyramidale. Eur. J. Neurosci. 12, 3975–3984. 10.1046/j.1460-9568.2000.00290.x
    1. Zhang G., Bai H., Zhang H., Dean C., Wu Q., Li J., et al. . (2011). Neuropeptide exocytosis involving synaptotagmin-4 and oxytocin in hypothalamic programming of body weight and energy balance. Neuron 69, 523–535. 10.1016/j.neuron.2010.12.036
    1. Zhang G., Cai D. (2011). Circadian intervention of obesity development via resting-stage feeding manipulation or oxytocin treatment. Am. J. Physiol. Endocrinol. Metab. 301, E1004–E1012. 10.1152/ajpendo.00196.2011
    1. Zimmerman E. A., Nilaver G., Hou-Yu A., Silverman A. J. (1984). Vasopressinergic and oxytocinergic pathways in the central nervous system. Fed. Proc. 43, 91–96.

Source: PubMed

3
Tilaa