Comprehensive Physiology Wiley Online Library

Neuropeptide Regulation of Social Attachment: The Prairie Vole Model

Full Article on Wiley Online Library



ABSTRACT

Social attachments are ubiquitous among humans and integral to human health. Although great efforts have been made to elucidate the neural underpinnings regulating social attachments, we still know relatively little about the neuronal and neurochemical regulation of social attachments. As a laboratory animal research model, the socially monogamous prairie vole (Microtus ochrogaster) displays behaviors paralleling human social attachments and thus has provided unique insights into the neural regulation of social behaviors. Research in prairie voles has particularly highlighted the significance of neuropeptidergic regulation of social behaviors, especially of the roles of oxytocin (OT) and vasopressin (AVP). This article aims to review these findings. We begin by discussing the role of the OT and AVP systems in regulating social behaviors relevant to social attachments, and thereafter restrict our discussion to studies in prairie voles. Specifically, we discuss the role of OT and AVP in adult mate attachments, biparental care, social isolation, and social buffering as informed by studies utilizing the prairie vole model. Not only do these studies offer insight into social attachments in humans, but they also point to dysregulated mechanisms in several mental disorders. We conclude by discussing these implications for human health. © 2017 American Physiological Society. Compr Physiol 7:81‐104, 2017.

Comprehensive Physiology offers downloadable PowerPoint presentations of figures for non-profit, educational use, provided the content is not modified and full credit is given to the author and publication.

Download a PowerPoint presentation of all images


Figure 1. Figure 1. Schematic drawings of sagittal brain sections illustrating OT (right) and AVP (left) neurons and their projections to selected brain regions important in social behaviors. Colored brain regions also indicate the distribution and regional density of OT receptors (red) and AVP receptors (blue) in the brain. OT and AVP are released from the pituitary gland into the blood circulation to regulate peripheral functions such as the milk letdown reflex and uterine contractions in females as well as vasoconstriction and water retention in both males and females. In addition, OT and AVP are released throughout the brain to regulate a variety of complex social behaviors including social recognition, mating, bonding, parenting, and social buffering. AH, anterior hypothalamus; BNST, bed nucleus of the stria terminalis; HP, hippocampus; LS, lateral septum; MeA medial amygdala. MPOA, medial preoptic area of the hypothalamus; NAcc, nucleus accumbens; OB, olfactory bulb; PFC, prefrontal cortex; Pit, pituitary gland; SON, supraoptic nucleus; VMH, ventromedial hypothalamus; VP, ventral pallidum; VTA, ventral tegmental area.
Figure 2. Figure 2. Socially monogamous prairie voles display several types of social behaviors that have been studied in laboratory conditions. (A) Photograph of a pair of male and female prairie voles with their pups in the nest. (B) Pair bonding behavior is measured using a 3‐h partner preference test. The testing apparatus consists of three chambers connected by hollow tubes. At the beginning of the test, the subject is placed in the center cage and allowed to freely explore the other two cages containing either the partner or a conspecific stranger. (C) In both male and female prairie voles, 24‐h cohabitation with mating reliably induces an increase in side‐by‐side contact with the partner versus a stranger, and this partner preference is not observed following 6‐h cohabitation. (D) Selective aggression is another indicator of pair bonding. While sexually naïve males are not aggressive, pair‐bonded males display aggression selectively toward stranger males and females, but not toward their partners. (E) Upon litter birth, both male and female prairie voles share the natal nest and engage in parental care. Data are shown as mean ± SEM. *P < 0.05. Alphabetic letters indicate the results from a post‐hoc test following an ANOVA. Bars labeled with different letters differ significantly from each other. Data adapted, with permission, from (11,102,274,283).
Figure 3. Figure 3. Autoradiograms showing the distribution of the OTR and vasopressin 1a receptor (V1aR) in the brain of the monogamous prairie voles and nonmonogamous montane voles. The densities of OTR binding in the nucleus accumbens (NAcc) and medial prefrontal cortex (mPFC) are higher in the prairie vole (A) than in the montane vole (B). Additionally, the density of V1aR binding is higher in the BNST and lower in the lateral septum (LS) in the prairie vole (C) compared to the montane vole (D). Data adapted, with permission, from (137,138,279).
Figure 4. Figure 4. The effects of OT and AVP on partner preference behavior in prairie voles. (A) Control females that receive ICV injections of CSF do not display partner preferences after 6 h of cohabitation with a male, whereas females receiving OT injections into the ventricle (OT ICV), nucleus accumbens (OT NAcc), or viral vector injections (AAV‐OTR) for OTR over expression in the NAcc, do display partner preferences. (B) 24 h of mating and cohabitation with a male reliably induces partner preferences in control females (CSF), but this behavior is prevented by ICV or intra‐NAcc injections of an OTR antagonist (OTRA) or downregulation of OXTR by injections of interfering short hairpin RNA (OTR‐shRNA). (C) In male prairie voles, brief cohabitation does not induce partner preferences. However, AVP injections into the ventricle (ICV) and lateral septum (LS) as well as upregulation of the V1aR in the ventral pallidum (VP) via viral vector mediated gene transfer (AAV‐V1aR) facilitate partner preference formation. (D) 24‐h mating and cohabitation with a female induces partner preference in male prairie voles but this behavior is blocked by injections of the V1aR antagonist (V1aRA) into the ventricle (ICV), LS, or VP. Although OT and AVP effects are illustrated here by data from females and males, respectively, both neuropeptides have been shown to affect partner preference behavior in male and female prairie voles. Data are shown as mean ± SEM. *P < 0.05, **P < 0.01. Data adapted, with permission, from (135,171,172,215,231,282,283).
Figure 5. Figure 5. The role of brain OT and AVP in the regulation of parental behaviors in prairie voles. (A) Oxytocin receptor (OTR) binding in the nucleus accumbens (NAcc) is higher in spontaneously maternal than in nonmaternal females. (B) Intra‐NAcc injections of an OTR antagonist (OTR‐NA) result in more female voles that do not display spontaneous maternal behavior compared to controls (CSF‐NA). (C) Downregulating OTR expression in the NAcc via injections of a short hairpin OTR interfering RNA (shRNA‐OTR) decreases the number of juvenile female voles displaying alloparental behavior, compared to control females (control) injected with a scrambled sequence. (D) Female voles receiving shRNA‐OTR injections into the NAcc spend less time licking and grooming pups, compared to control females. (E) Male prairie voles receiving ICV injections of AVP display a higher level of spontaneous paternal responsiveness to pups compared to males receiving control injections (Saline) or injections of AVP with an AVP receptor antagonist (antagonist/AVP). Data are shown as mean ± SEM. *P < 0.05. Data adapted, with permission, from (24,198,272).
Figure 6. Figure 6. The effects of social isolation on behaviors, brain OT, and immune responses in sexually naive female prairie voles. Compared to the pair‐housed controls (paired), females that are socially isolated from cage mates for 4 weeks (isolated) spend less time in the open arms during an elevated plus maze test (EPM) (A) and more time immobile during a forced swim test (FST) (B). Isolation experience also decreases CH50, which measures the activity of the immune system's classical complement pathway (C). In addition, Isolated females show a decrease in OTR mRNA expression in the hypothalamus (D), an increase in the percentage of c‐Fos labeled OT‐immunoreactive neurons in the PVN following a 5‐min RIT (E), and an elevation in circulating OT levels (F), compared to the Paired controls. Such social isolation‐induced increases in heart rate (G) and immobile duration during the FST (H) as well as the decrease in sucrose intake (I) are prevented by daily OT administration. Data are shown as mean ± SEM. *P < 0.05. Alphabetic letters indicate the results from a post‐hoc test following an ANOVA. Bars labeled with different letters differ significantly from each other. Data adapted, with permission, from (109,110,114,216,236).
Figure 7. Figure 7. The effects of partner separation on the physiology, behavior, and neurochemical staining in the brain of pair‐bonded prairie voles. Compared to the paired controls (paired), 5 days of separation from the mating partner (separated) resulted in increases in heat rate (A), immobile time during a forced swim test (FST) (B), and circulating levels of CORT following the FST (C) in both male and female prairie voles. In male prairie voles, 2 weeks of separation from their female partners led to a decreased entry to the open arms during an elevated plus maze test (EPM) (D) and an increased duration in the dark box during a light‐dark box test (E), in comparison to the Paired controls. Furthermore, these Separated males also showed an increase in social affiliation and a decrease in aggression during an RIT (F) as well as increases in the number of neurons stained for OT and AVP in the paraventricular nucleus of the hippocampus (PVN) (G, H, and I), compared to Paired controls. Data are shown as mean ± SEM. *P < 0.05. Scale bar = 100 μm. Data adapted, with permission, from (179,255).
Figure 8. Figure 8. The effects of IMO stress and social buffering on anxiety‐like behaviors and brain OT activity in female prairie voles. Subjects experience 1‐h IMO stress, recover alone (alone) or with the male partner (partner) for 30 min, and receive a 5‐min EPM test. (A) Females that recover alone (alone) enter the open arms less frequently and spend less time there in the EPM test, compared to handled females (control) and females recovering with a partner (partner). (B) Data from in vivo brain microdialysis show that OT release in the PVN increases during IMO stress, and this increased OT release is sustained during the recovery if the subject recovers with a partner but not alone. (C and D) Intra‐PVN injections of an OTRA in the subjects recovering with the partner block social buffering effects on anxiety‐like behaviors and plasma CORT. Conversely, OT injections into the PVN of the subjects recovering alone mimic the effects of a partner by reducing anxiety like behaviors and circulating levels of CORT (D). Data are shown as mean ± SEM. *P < 0.05. Alphabetic letters indicate the results from a post‐hoc test following an ANOVA. Bars labeled with different letters differ significantly from each other. Data adapted, with permission, from (248).
Figure 9. Figure 9. The effects of OT treatment in the PVN on stress induced changes in circulating CORT, anxiety‐like behaviors, and neural activity in female prairie voles. Intra‐PVN injections of OT before an EPS inhibit the stress‐induced rise in plasma CORT (A) and anxiety‐like behavior during the EPM (B). Although such OT injection does not alter the percentage of OT (C) or AVP (D) neurons in the PVN that are double‐labeled with c‐Fos, it decreases the number of CRH/Fos neurons (E) and increases the number of GABA/Fos (F) neurons in the PVN. Blue arrows point to double labeled cells. Furthermore, intra‐PVN injections of the GABAA receptor antagonist, bicuculline (Bic), prevent OT effects in reducing anxiety‐like behavior following the EPS stress (G). (H) A hypothetical model suggesting that OT release in the PVN may activate GABAergic interneurons in the PVN and GABAergic projecting neurons to the PVN, which, in turn, can inhibit CRH neurons and potentially decrease activity of the hypothalamic‐pituitary‐adrenal axis. Data are shown as mean ± SEM. *P < 0.05. Data adapted, with permission, from (246).


Figure 1. Schematic drawings of sagittal brain sections illustrating OT (right) and AVP (left) neurons and their projections to selected brain regions important in social behaviors. Colored brain regions also indicate the distribution and regional density of OT receptors (red) and AVP receptors (blue) in the brain. OT and AVP are released from the pituitary gland into the blood circulation to regulate peripheral functions such as the milk letdown reflex and uterine contractions in females as well as vasoconstriction and water retention in both males and females. In addition, OT and AVP are released throughout the brain to regulate a variety of complex social behaviors including social recognition, mating, bonding, parenting, and social buffering. AH, anterior hypothalamus; BNST, bed nucleus of the stria terminalis; HP, hippocampus; LS, lateral septum; MeA medial amygdala. MPOA, medial preoptic area of the hypothalamus; NAcc, nucleus accumbens; OB, olfactory bulb; PFC, prefrontal cortex; Pit, pituitary gland; SON, supraoptic nucleus; VMH, ventromedial hypothalamus; VP, ventral pallidum; VTA, ventral tegmental area.


Figure 2. Socially monogamous prairie voles display several types of social behaviors that have been studied in laboratory conditions. (A) Photograph of a pair of male and female prairie voles with their pups in the nest. (B) Pair bonding behavior is measured using a 3‐h partner preference test. The testing apparatus consists of three chambers connected by hollow tubes. At the beginning of the test, the subject is placed in the center cage and allowed to freely explore the other two cages containing either the partner or a conspecific stranger. (C) In both male and female prairie voles, 24‐h cohabitation with mating reliably induces an increase in side‐by‐side contact with the partner versus a stranger, and this partner preference is not observed following 6‐h cohabitation. (D) Selective aggression is another indicator of pair bonding. While sexually naïve males are not aggressive, pair‐bonded males display aggression selectively toward stranger males and females, but not toward their partners. (E) Upon litter birth, both male and female prairie voles share the natal nest and engage in parental care. Data are shown as mean ± SEM. *P < 0.05. Alphabetic letters indicate the results from a post‐hoc test following an ANOVA. Bars labeled with different letters differ significantly from each other. Data adapted, with permission, from (11,102,274,283).


Figure 3. Autoradiograms showing the distribution of the OTR and vasopressin 1a receptor (V1aR) in the brain of the monogamous prairie voles and nonmonogamous montane voles. The densities of OTR binding in the nucleus accumbens (NAcc) and medial prefrontal cortex (mPFC) are higher in the prairie vole (A) than in the montane vole (B). Additionally, the density of V1aR binding is higher in the BNST and lower in the lateral septum (LS) in the prairie vole (C) compared to the montane vole (D). Data adapted, with permission, from (137,138,279).


Figure 4. The effects of OT and AVP on partner preference behavior in prairie voles. (A) Control females that receive ICV injections of CSF do not display partner preferences after 6 h of cohabitation with a male, whereas females receiving OT injections into the ventricle (OT ICV), nucleus accumbens (OT NAcc), or viral vector injections (AAV‐OTR) for OTR over expression in the NAcc, do display partner preferences. (B) 24 h of mating and cohabitation with a male reliably induces partner preferences in control females (CSF), but this behavior is prevented by ICV or intra‐NAcc injections of an OTR antagonist (OTRA) or downregulation of OXTR by injections of interfering short hairpin RNA (OTR‐shRNA). (C) In male prairie voles, brief cohabitation does not induce partner preferences. However, AVP injections into the ventricle (ICV) and lateral septum (LS) as well as upregulation of the V1aR in the ventral pallidum (VP) via viral vector mediated gene transfer (AAV‐V1aR) facilitate partner preference formation. (D) 24‐h mating and cohabitation with a female induces partner preference in male prairie voles but this behavior is blocked by injections of the V1aR antagonist (V1aRA) into the ventricle (ICV), LS, or VP. Although OT and AVP effects are illustrated here by data from females and males, respectively, both neuropeptides have been shown to affect partner preference behavior in male and female prairie voles. Data are shown as mean ± SEM. *P < 0.05, **P < 0.01. Data adapted, with permission, from (135,171,172,215,231,282,283).


Figure 5. The role of brain OT and AVP in the regulation of parental behaviors in prairie voles. (A) Oxytocin receptor (OTR) binding in the nucleus accumbens (NAcc) is higher in spontaneously maternal than in nonmaternal females. (B) Intra‐NAcc injections of an OTR antagonist (OTR‐NA) result in more female voles that do not display spontaneous maternal behavior compared to controls (CSF‐NA). (C) Downregulating OTR expression in the NAcc via injections of a short hairpin OTR interfering RNA (shRNA‐OTR) decreases the number of juvenile female voles displaying alloparental behavior, compared to control females (control) injected with a scrambled sequence. (D) Female voles receiving shRNA‐OTR injections into the NAcc spend less time licking and grooming pups, compared to control females. (E) Male prairie voles receiving ICV injections of AVP display a higher level of spontaneous paternal responsiveness to pups compared to males receiving control injections (Saline) or injections of AVP with an AVP receptor antagonist (antagonist/AVP). Data are shown as mean ± SEM. *P < 0.05. Data adapted, with permission, from (24,198,272).


Figure 6. The effects of social isolation on behaviors, brain OT, and immune responses in sexually naive female prairie voles. Compared to the pair‐housed controls (paired), females that are socially isolated from cage mates for 4 weeks (isolated) spend less time in the open arms during an elevated plus maze test (EPM) (A) and more time immobile during a forced swim test (FST) (B). Isolation experience also decreases CH50, which measures the activity of the immune system's classical complement pathway (C). In addition, Isolated females show a decrease in OTR mRNA expression in the hypothalamus (D), an increase in the percentage of c‐Fos labeled OT‐immunoreactive neurons in the PVN following a 5‐min RIT (E), and an elevation in circulating OT levels (F), compared to the Paired controls. Such social isolation‐induced increases in heart rate (G) and immobile duration during the FST (H) as well as the decrease in sucrose intake (I) are prevented by daily OT administration. Data are shown as mean ± SEM. *P < 0.05. Alphabetic letters indicate the results from a post‐hoc test following an ANOVA. Bars labeled with different letters differ significantly from each other. Data adapted, with permission, from (109,110,114,216,236).


Figure 7. The effects of partner separation on the physiology, behavior, and neurochemical staining in the brain of pair‐bonded prairie voles. Compared to the paired controls (paired), 5 days of separation from the mating partner (separated) resulted in increases in heat rate (A), immobile time during a forced swim test (FST) (B), and circulating levels of CORT following the FST (C) in both male and female prairie voles. In male prairie voles, 2 weeks of separation from their female partners led to a decreased entry to the open arms during an elevated plus maze test (EPM) (D) and an increased duration in the dark box during a light‐dark box test (E), in comparison to the Paired controls. Furthermore, these Separated males also showed an increase in social affiliation and a decrease in aggression during an RIT (F) as well as increases in the number of neurons stained for OT and AVP in the paraventricular nucleus of the hippocampus (PVN) (G, H, and I), compared to Paired controls. Data are shown as mean ± SEM. *P < 0.05. Scale bar = 100 μm. Data adapted, with permission, from (179,255).


Figure 8. The effects of IMO stress and social buffering on anxiety‐like behaviors and brain OT activity in female prairie voles. Subjects experience 1‐h IMO stress, recover alone (alone) or with the male partner (partner) for 30 min, and receive a 5‐min EPM test. (A) Females that recover alone (alone) enter the open arms less frequently and spend less time there in the EPM test, compared to handled females (control) and females recovering with a partner (partner). (B) Data from in vivo brain microdialysis show that OT release in the PVN increases during IMO stress, and this increased OT release is sustained during the recovery if the subject recovers with a partner but not alone. (C and D) Intra‐PVN injections of an OTRA in the subjects recovering with the partner block social buffering effects on anxiety‐like behaviors and plasma CORT. Conversely, OT injections into the PVN of the subjects recovering alone mimic the effects of a partner by reducing anxiety like behaviors and circulating levels of CORT (D). Data are shown as mean ± SEM. *P < 0.05. Alphabetic letters indicate the results from a post‐hoc test following an ANOVA. Bars labeled with different letters differ significantly from each other. Data adapted, with permission, from (248).


Figure 9. The effects of OT treatment in the PVN on stress induced changes in circulating CORT, anxiety‐like behaviors, and neural activity in female prairie voles. Intra‐PVN injections of OT before an EPS inhibit the stress‐induced rise in plasma CORT (A) and anxiety‐like behavior during the EPM (B). Although such OT injection does not alter the percentage of OT (C) or AVP (D) neurons in the PVN that are double‐labeled with c‐Fos, it decreases the number of CRH/Fos neurons (E) and increases the number of GABA/Fos (F) neurons in the PVN. Blue arrows point to double labeled cells. Furthermore, intra‐PVN injections of the GABAA receptor antagonist, bicuculline (Bic), prevent OT effects in reducing anxiety‐like behavior following the EPS stress (G). (H) A hypothetical model suggesting that OT release in the PVN may activate GABAergic interneurons in the PVN and GABAergic projecting neurons to the PVN, which, in turn, can inhibit CRH neurons and potentially decrease activity of the hypothalamic‐pituitary‐adrenal axis. Data are shown as mean ± SEM. *P < 0.05. Data adapted, with permission, from (246).
References
 1.Acher R, Chauvet J, Chauvet MT. Man and the chimaera. Selective versus neutral oxytocin evolution. Adv Exp Med Biol 395: 615‐627, 1995.
 2.Ahern TH, Hammock EAD, Young LJ. Parental division of labor, coordination, and the effects of family structure on parenting in monogamous prairie voles (Microtus ochrogaster). Dev Psychobiol 53(2): 118‐131, 2011.
 3.Ahern TH, Young LJ. The impact of early life family structure on adult social attachment, alloparental behavior, and the neuropeptide systems regulating affiliative behaviors in the monogamous prairie vole (Microtus ochrogaster). Front Behav Neurosci 3: 17, 2009.
 4.Ainsworth MD. Object relations, dependency, and attachment: A theoretical review of the infant‐mother relationship. Child Dev 40(4): 969‐1025, 1969.
 5.Akther S, Korshnova N, Zhong J, Liang M, Cherepanov SM, Lopatina O, Komleva YK, Salmina AB, Nishimura T, Fakhrul AA, Hirai H, Kato I, Yamamoto Y, Takasawa S, Okamoto H, Higashida H. Cd38 in the nucleus accumbens and oxytocin are related to paternal behavior in mice. Mol Brain 6: 41, 2013.
 6.Albers HE. Species, sex and individual differences in the vasotocin/vasopressin system: Relationship to neurochemical signaling in the social behavior neural network. Front Neuroendocrinol 36:49‐71, 2015.
 7.Anacker AMJ, Ahern TH, Hostetler CM, Dufour BD, Smith ML, Cocking DL, Li J, Young LJ, Loftis JM, Ryabinin AE. Drinking alcohol has sex‐dependent effects on pair bond formation in prairie voles. Proc Natl Acad Sci U S A 111(16): 6052‐6057, 2014.
 8.Anacker AMJ, Loftis JM, Kaur S, Ryabinin AE. Prairie voles as a novel model of socially facilitated excessive drinking. Addict Biol 16(1): 92‐107, 2011.
 9.Anagnostou E, Soorya L, Brian J, Dupuis A, Mankad D, Smile S, Jacob S. Intranasal oxytocin in the treatment of autism spectrum disorders: A review of literature and early safety and efficacy data in youth. Brain Res 1580: 188‐198, 2014.
 10.Aragona BJ, Liu Y, Yu YJ, Curtis JT, Detwiler JM, Insel TR, Wang Z. Nucleus accumbens dopamine differentially mediates the formation and maintenance of monogamous pair bonds. Nat Neurosci 9(1): 133‐139, 2006.
 11.Aragona BJ, Wang Z. Dopamine regulation of social choice in a monogamous rodent species. Front Behav Neurosci 3: 15, 2009.
 12.Argiolas A, Melis MR. Neuropeptides and central control of sexual behaviour from the past to the present: A review. Prog Neurobiol 108: 80‐107, 2013.
 13.Armstrong WE, Warach S, Hatton GI, McNeill TH. Subnuclei in the rat hypothalamic paraventricular nucleus: A cytoarchitectural, horseradish peroxidase and immunocytochemical analysis. Neuroscience 5(11): 1931‐1958, 1980.
 14.Auyeung B, Lombardo MV, Heinrichs M, Chakrabarti B, Sule A, Deakin JB, Bethlehem RAI, Dickens L, Mooney N, Sipple JAN, Thiemann P, Baron‐Cohen S. Oxytocin increases eye contact during a real‐time, naturalistic social interaction in males with and without autism. Transl Psychiatry 5: e507, 2015.
 15.Badura B. Social support and community health care. Soz‐ Präventivmedizin 33(2): 79‐85, 1988.
 16.Bales KL, Carter CS. Sex differences and developmental effects of oxytocin on aggression and social behavior in prairie voles (Microtus ochrogaster). Horm Behav 44(3): 178‐184, 2003.
 17.Bales KL, Kim AJ, Lewis‐Reese AD, Sue Carter C. Both oxytocin and vasopressin may influence alloparental behavior in male prairie voles. Horm Behav 45(5): 354‐361, 2004.
 18.Bales KL, Perkeybile AM, Conley OG, Lee MH, Guoynes CD, Downing GM, Yun CR, Solomon M, Jacob S, Mendoza SP. Chronic intranasal oxytocin causes long‐term impairments in partner preference formation in male prairie voles. Biol Psychiatry 74(3): 180‐188, 2013.
 19.Bales KL, Pfeifer LA, Carter CS. Sex differences and developmental effects of manipulations of oxytocin on alloparenting and anxiety in prairie voles. Dev Psychobiol 44(2): 123‐131, 2004.
 20.Bales KL, Plotsky PM, Young LJ, Lim MM, Grotte N, Ferrer E, Carter CS. Neonatal oxytocin manipulations have long‐lasting, sexually dimorphic effects on vasopressin receptors. Neuroscience 144(1): 38‐45, 2007.
 21.Bamshad M, Novak MA, De Vries GJ. Sex and species differences in the vasopressin innervation of sexually naive and parental prairie voles, Microtus ochrogaster and meadow voles, Microtus pennsylvanicus. J Neuroendocrinol 5(3): 247‐255, 1993.
 22.Bamshad M, Novak MA, de Vries GJ. Cohabitation alters vasopressin innervation and paternal behavior in prairie voles (Microtus ochrogaster). Physiol Behav 56(4): 751‐758, 1994.
 23.Barrett CE, Keebaugh AC, Ahern TH, Bass CE, Terwilliger EF, Young LJ. Variation in vasopressin receptor (AVPR1a) expression creates diversity in behaviors related to monogamy in prairie voles. Horm Behav 63(3): 518‐526, 2013.
 24.Barrett CE, Keebaugh AC, Laprairie JL, Jenkins JJ, Young LJ. RNAi knockdown of oxytocin receptor in the nucleus accumbens inhibits social attachment and parental care in monogamous female prairie voles. Soc Neurosci, 10: 561‐570, 2015.
 25.Bartz J, Simeon D, Hamilton H, Kim S, Crystal S, Braun A, Vicens V, Hollander E. Oxytocin can hinder trust and cooperation in borderline personality disorder. Soc Cogn Affect Neurosci 6(5): 556‐563, 2011.
 26.Bealer SL, Lipschitz DL, Ramoz G, Crowley WR. Oxytocin receptor binding in the hypothalamus during gestation in rats. Am J Physiol Regul Integr Comp Physiol 291(1): R53‐R58, 2006.
 27.Beery AK, Routman DM, Zucker I. Same‐sex social behavior in meadow voles: Multiple and rapid formation of attachments. Physiol Behav 97(1): 52‐57, 2009.
 28.Bielsky IF, Hu S‐B, Ren X, Terwilliger EF, Young LJ. The V1a vasopressin receptor is necessary and sufficient for normal social recognition: A gene replacement study. Neuron 47(4): 503‐513, 2005.
 29.Bluthe RM, Schoenen J, Dantzer R. Androgen‐dependent vasopressinergic neurons are involved in social recognition in rats. Brain Res 519(1‐2): 150‐157, 1990.
 30.Bosch OJ. Maternal aggression in rodents: Brain oxytocin and vasopressin mediate pup defence. Philos Trans R Soc Lond B Biol Sci 368(1631): 20130085, 2013.
 31.Bosch OJ, Dabrowska J, Modi ME, Johnson ZV, Keebaugh AC, Barrett CE, Ahern TH, Guo J, Grinevich V, Rainnie DG, Neumann ID, Young LJ. Oxytocin in the nucleus accumbens shell reverses CRFR2‐evoked passive stress‐coping after partner loss in monogamous male prairie voles. Psychoneuroendocrinology 64: 66‐78, 2016.
 32.Bosch OJ, Nair HP, Ahern TH, Neumann ID, Young LJ. The CRF system mediates increased passive stress‐coping behavior following the loss of a bonded partner in a monogamous rodent. Neuropsychopharmacol 34(6): 1406‐1415, 2009.
 33.Bosch OJ, Neumann ID. Brain vasopressin is an important regulator of maternal behavior independent of dams' trait anxiety. Proc Natl Acad Sci U S A 105(44): 17139‐17144, 2008.
 34.Bosch OJ, Neumann ID. Vasopressin released within the central amygdala promotes maternal aggression. Eur J Neurosci 31(5): 883‐891, 2010.
 35.Bosch OJ, Neumann ID. Both oxytocin and vasopressin are mediators of maternal care and aggression in rodents: From central release to sites of action. Horm Behav 61(3): 293‐303, 2012.
 36.Bosch OJ, Pförtsch J, Beiderbeck DI, Landgraf R, Neumann ID. Maternal behaviour is associated with vasopressin release in the medial preoptic area and bed nucleus of the stria terminalis in the rat. J Neuroendocrinol 22(5): 420‐429, 2010.
 37.Bowlby J. Attachment and Loss. New York: Basic Books, 1969.
 38.Burkett JP, Andari E, Johnson ZV, Curry DC, de Waal FBM, Young LJ. Oxytocin‐dependent consolation behavior in rodents. Science 351(6271): 375‐378, 2016.
 39.Burkett JP, Young LJ. The behavioral, anatomical and pharmacological parallels between social attachment, love and addiction. Psychopharmacology (Berl) 224(1): 1‐26, 2012.
 40.Cacioppo JT, Cacioppo S. The phenotype of loneliness. Eur J Dev Psychol 9(4): 446‐452, 2012.
 41.Cacioppo JT, Cacioppo S, Capitanio JP, Cole SW. The neuroendocrinology of social isolation. Annu Rev Psychol 66: 733‐767, 2015.
 42.Cacioppo JT, Cacioppo S, Cole SW, Capitanio JP, Goossens L, Boomsma DI. Loneliness across phylogeny and a call for comparative studies and animal models. Perspect Psychol Sci 10(2): 202‐212, 2015.
 43.Cacioppo JT, Hawkley LC, Thisted RA. Perceived social isolation makes me sad: 5‐year cross‐lagged analyses of loneliness and depressive symptomatology in the chicago health, aging, and social relations study. Psychol Aging 25(2): 453‐463, 2010.
 44.Cacioppo S, Capitanio JP, Cacioppo JT. Toward a neurology of loneliness. Psychol Bull 140(6): 1464‐1504, 2014.
 45.Cacioppo S, Grippo AJ, London S, Goossens L, Cacioppo JT. Loneliness: Clinical import and interventions. Perspect Psychol Sci 10(2): 238‐249, 2015.
 46.Carter CS. Attachment and Bonding: A New Synthesis. Cambridge, Mass: MIT Press. p. 509, 2005.
 47.Carter CS, Boone EM, Pournajafi‐Nazarloo H, Bales KL. Consequences of early experiences and exposure to oxytocin and vasopressin are sexually dimorphic. Dev Neurosci 31(4): 332‐341, 2009.
 48.Carter CS, DeVries AC, Getz LL. Physiological substrates of mammalian monogamy: The prairie vole model. Neurosci Biobehav Rev 19(2): 303‐314, 1995.
 49.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. Proc Natl Acad Sci U S A 98(22): 12736‐12741, 2001.
 50.Champagne FA, Weaver ICG, Diorio J, Sharma S, Meaney MJ. Natural variations in maternal care are associated with estrogen receptor alpha expression and estrogen sensitivity in the medial preoptic area. Endocrinology 144(11): 4720‐4724, 2003.
 51.Chen FS, Barth ME, Johnson SL, Gotlib IH, Johnson SC. Oxytocin receptor (OXTR) polymorphisms and attachment in human infants. Front Psychol 2: 200, 2011.
 52.Cho MM, DeVries AC, Williams JR, Carter CS. The effects of oxytocin and vasopressin on partner preferences in male and female prairie voles (Microtus ochrogaster). Behav Neurosci 113(5): 1071‐1079, 1999.
 53.Churchland PS, Winkielman P. Modulating social behavior with oxytocin: How does it work? What does it mean? Horm Behav 61(3): 392‐399, 2012.
 54.Clark‐Elford R, Nathan PJ, Auyeung B, Mogg K, Bradley BP, Sule A, Müller U, Dudas RB, Sahakian BJ, Baron‐Cohen S. Effects of oxytocin on attention to emotional faces in healthy volunteers and highly socially anxious males. Int J Neuropsychopharmacol 18(2), n/a. doi: 10.1093/ijnp/pyu012, 2015.
 55.Cohen S, Wills TA. Stress, social support, and the buffering hypothesis. Psychol Bull 98(2): 310‐357, 1985.
 56.Cole SW, Capitanio JP, Chun K, Arevalo JMG, Ma J, Cacioppo JT. Myeloid differentiation architecture of leukocyte transcriptome dynamics in perceived social isolation. Proc Natl Acad Sci U S A 112: 15142‐15147, 2015.
 57.Cullinan WE. GABA(a) receptor subunit expression within hypophysiotropic CRH neurons: A dual hybridization histochemical study. J Comp Neurol 419(3): 344‐351, 2000.
 58.Cushing BS. Neonatal Effects of Oxytocin: Development and Organizational. Hauppauge: Nova Science Publishers, Inc., 2009.
 59.Cushing BS, Okorie U, Young LJ. The effects of neonatal castration on the subsequent behavioural response to centrally administered arginine vasopressin and the expression of V1a receptors in adult male prairie voles. J Neuroendocrinol 15(11): 1021‐1026, 2003.
 60.Cushing BS, Razzoli M, Murphy AZ, Epperson PM, Le W‐W, Hoffman GE. Intraspecific variation in estrogen receptor alpha and the expression of male sociosexual behavior in two populations of prairie voles. Brain Res 1016(2): 247‐254, 2004.
 61.Dadds MR, Moul C, Cauchi A, Dobson‐Stone C, Hawes DJ, Brennan J, Urwin R, Ebstein RE. Polymorphisms in the oxytocin receptor gene are associated with the development of psychopathy. Dev Psychopathol 26(1): 21‐31, 2014.
 62.Dantzer R, Bluthe RM, Koob GF, Le Moal M. Modulation of social memory in male rats by neurohypophyseal peptides. Psychopharmacology (Berl) 91(3): 363‐368, 1987.
 63.Dantzer R, Koob GF, Bluthé RM, Le Moal M. Septal vasopressin modulates social memory in male rats. Brain Res 457(1): 143‐147, 1988.
 64.De Dreu CKW, Kret ME. Oxytocin conditions intergroup relations through upregulated in‐group empathy, cooperation, conformity, and defense. Biol Psychiatry 79(3): 165‐173, 2016.
 65.De Vries GJ, Best W, Sluiter AA. The influence of androgens on the development of a sex difference in the vasopressinergic innervation of the rat lateral septum. Brain Res 284(2‐3): 377‐380, 1983.
 66.De Vries GJ, Panzica GC. Sexual differentiation of central vasopressin and vasotocin systems in vertebrates: Different mechanisms, similar endpoints. Neuroscience 138(3): 947‐955, 2006.
 67.Decavel C, Van den Pol AN. GABA: A dominant neurotransmitter in the hypothalamus. J Comp Neurol 302(4): 1019‐1037, 1990.
 68.DeVries AC. Interaction among social environment, the hypothalamic‐pituitary‐adrenal axis, and behavior. Horm Behav 41(4): 405‐413, 2002.
 69.DeVries AC, DeVries MB, Taymans S, Carter CS. Modulation of pair bonding in female prairie voles (Microtus ochrogaster) by corticosterone. Proc Natl Acad Sci U S A 92(17): 7744‐7748, 1995.
 70.DeVries AC, DeVries MB, Taymans SE, Carter CS. The effects of stress on social preferences are sexually dimorphic in prairie voles. Proc Natl Acad Sci U S A 93(21): 11980‐11984, 1996.
 71.DeVries AC, Guptaa T, Cardillo S, Cho M, Carter CS. Corticotropin‐releasing factor induces social preferences in male prairie voles. Psychoneuroendocrinology 27(6): 705‐714, 2002.
 72.DeVries AC, Taymans SE, Carter CS. Social modulation of corticosteroid responses in male prairie voles. Ann N Y Acad Sci 807: 494‐497, 1997.
 73.DeVries GJ, Buijs RM, Van Leeuwen FW, Caffé AR, Swaab DF. The vasopressinergic innervation of the brain in normal and castrated rats. J Comp Neurol 233(2): 236‐254, 1985.
 74.Ditzen B, Schaer M, Gabriel B, Bodenmann G, Ehlert U, Heinrichs M. Intranasal oxytocin increases positive communication and reduces cortisol levels during couple conflict. Biol Psychiatry 65(9): 728‐731, 2009.
 75.Dluzen DE, Muraoka S, Engelmann M, Landgraf R. The effects of infusion of arginine vasopressin, oxytocin, or their antagonists into the olfactory bulb upon social recognition responses in male rats. Peptides 19(6): 999‐1005, 1998.
 76.Dodhia S, Hosanagar A, Fitzgerald DA, Labuschagne I, Wood AG, Nathan PJ, Phan KL. Modulation of resting‐state amygdala‐frontal functional connectivity by oxytocin in generalized social anxiety disorder. Neuropsychopharmacol 39(9): 2061‐2069, 2014.
 77.Domes G, Kumbier E, Heinrichs M, Herpertz SC. Oxytocin promotes facial emotion recognition and amygdala reactivity in adults with asperger syndrome. Neuropsychopharmacol 39(3): 698‐706, 2014.
 78.Donaldson ZR, Spiegel L, Young LJ. Central vasopressin V1a receptor activation is independently necessary for both partner preference formation and expression in socially monogamous male prairie voles. Behav Neurosci 124(1): 159‐163, 2010.
 79.Dumais KM, Veenema AH. Vasopressin and oxytocin receptor systems in the brain: Sex differences and sex‐specific regulation of social behavior. Front Neuroendocrinol 40: 1‐23, 2015.
 80.Ebstein RP, Knafo A, Mankuta D, Chew SH, Lai PS. The contributions of oxytocin and vasopressin pathway genes to human behavior. Horm Behav 61(3): 359‐379, 2012.
 81.Eckstein M, Scheele D, Weber K, Stoffel‐Wagner B, Maier W, Hurlemann R. Oxytocin facilitates the sensation of social stress. Hum Brain Mapp 35(9): 4741‐4750, 2014.
 82.Eliava M, Melchior M, Knobloch‐Bollmann HS, Wahis J, da Silva Gouveia M, Tang Y, Ciobanu AC, Triana Del Rio R, Roth LC, Althammer F, Chavant V, Goumon Y, Gruber T, Petit‐Demoulière N, Busnelli M, Chini B, Tan LL, Mitre M, Froemke RC, Chao MV, Giese G, Sprengel R, Kuner R, Poisbeau P, Seeburg PH, Stoop R, Charlet A, Grinevich V. A new population of parvocellular oxytocin neurons controlling magnocellular neuron activity and inflammatory pain processing. Neuron 89(6): 1291‐1304, 2016.
 83.Evans S. The pair‐bond of the common marmoset, Callithrix jacchus jacchus: An experimental investigation. Anim Behav 31(3): 651‐658, 1983.
 84.Feldman R, Monakhov M, Pratt M, Ebstein RP. Oxytocin pathway genes: Evolutionary ancient system impacting on human affiliation, sociality, and psychopathology. Biol Psychiatry 79(3): 174‐184, 2016.
 85.Ferguson JN, Aldag JM, Insel TR, Young LJ. Oxytocin in the medial amygdala is essential for social recognition in the mouse. J Neurosci 21(20): 8278‐8285, 2001.
 86.Ferris CF, Yee JR, Kenkel WM, Dumais KM, Moore K, Veenema AH, Kulkarni P, Perkybile AM, Carter CS. Distinct bold activation profiles following central and peripheral oxytocin administration in awake rats. Front Behav Neurosci 9: 245, 2015.
 87.Flanagan LM, Pfaus JG, Pfaff DW, McEwen BS. Induction of FOS immunoreactivity in oxytocin neurons after sexual activity in female rats. Neuroendocrinology 58(3): 352‐358, 1993.
 88.Fliers E, Guldenaar SE, van de Wal N, Swaab DF. Extrahypothalamic vasopressin and oxytocin in the human brain; presence of vasopressin cells in the bed nucleus of the stria terminalis. Brain Res 375(2): 363‐367, 1986.
 89.Freeman SM, Inoue K, Smith AL, Goodman MM, Young LJ. The neuroanatomical distribution of oxytocin receptor binding and mRNA in the male rhesus macaque (Macaca mulatta). Psychoneuroendocrinology 45: 128‐141, 2014.
 90.Freeman SM, Walum H, Inoue K, Smith AL, Goodman MM, Bales KL, Young LJ. Neuroanatomical distribution of oxytocin and vasopressin 1a receptors in the socially monogamous coppery titi monkey (Callicebus cupreus). Neuroscience 273: 12‐23, 2014.
 91.Freeman SM, Young LJ. Comparative perspectives on oxytocin and vasopressin receptor research in rodents and primates: Translational implications. J Neuroendocrinol 28(4), n/a. doi:10.1111/jne.12382, 2016.
 92.Fujisawa TX, Nishitani S, Obara T, Shinohara K. Loneliness depends on salivary estradiol levels in adolescent females. Neuro Endocrinol Lett 33(5): 525‐529, 2012.
 93.Gavish L, Carter CS, Getz LL. Further evidences for monogamy in the prairie vole. Anim Behav 29(3): 955‐957, 1981.
 94.Getz LL. Aggressive behavior of the meadow and prairie voles. J Mammal 43(3): 351‐358, 1962.
 95.Getz LL, Carter CS. Prairie‐vole partnerships. Am Sci 84(1): 56‐62, 1996.
 96.Getz LL, Carter CS, Gavish L. The mating system of the prairie vole, Microtus ochrogaster: Field and laboratory evidence for pair‐bonding. Behav Ecol Sociobiol 8(3): 189‐194, 1981.
 97.Getz LL, McGuire B, Pizzuto T, Hofmann JE, Frase B. Social organization of the prairie vole (Microtus ochrogaster). J Mammal 74(1): 44‐58, 1993.
 98.Gimpl G, Fahrenholz F. The oxytocin receptor system: Structure, function, and regulation. Physiol Rev 81(2): 629‐683, 2001.
 99.Gobrogge K, Wang Z. Neuropeptidergic regulation of pair‐bonding and stress buffering: Lessons from voles. Horm Behav 76: 91‐105, 2015.
 100.Gobrogge KL. Sex, drugs, and violence: Neuromodulation of attachment and conflict in voles. Curr Top Behav Neurosci 17: 229‐264, 2014.
 101.Gobrogge KL, Jia X, Liu Y, Wang Z. Neurochemical mediation of affiliation and aggression associated with pair‐bonding. Biol Psychiatry, 2016. n/a (in press) doi: 10.1016/j.biopsych.2016.02.013.
 102.Gobrogge KL, Liu Y, Jia X, Wang Z. Anterior hypothalamic neural activation and neurochemical associations with aggression in pair‐bonded male prairie voles. J Comp Neurol 502(6): 1109‐1122, 2007.
 103.Gobrogge KL, Liu Y, Young LJ, Wang Z. Anterior hypothalamic vasopressin regulates pair‐bonding and drug‐induced aggression in a monogamous rodent. Proc Natl Acad Sci USA 106(45): 19144‐19149, 2009.
 104.Goodson JL, Kabelik D. Dynamic limbic networks and social diversity in vertebrates: From neural context to neuromodulatory patterning. Front Neuroendocrinol 30(4): 429‐441, 2009.
 105.Gray GD, Dewsbury DA. A quantitative description of copulatory behavior in prairie voles (Microtus ochrogaster). Brain Behav Evol 8(6): 426‐452, 1973.
 106.Grinevich V, Desarménien MG, Chini B, Tauber M, Muscatelli F. Ontogenesis of oxytocin pathways in the mammalian brain: Late maturation and psychosocial disorders. Front Neuroanat 8: 164, 2014.
 107.Grinevich V, Knobloch‐Bollmann HS, Eliava M, Busnelli M, Chini B. Assembling the puzzle: Pathways of oxytocin signaling in the brain. Biol Psychiatry 79(3): 155‐164, 2016.
 108.Grippo AJ, Carter CS, McNeal N, Chandler DL, Larocca MA, Bates SL, Porges SW. 24‐hour autonomic dysfunction and depressive behaviors in an animal model of social isolation: Implications for the study of depression and cardiovascular disease. Psychosom Med 73(1): 59‐66, 2011.
 109.Grippo AJ, Gerena D, Huang J, Kumar N, Shah M, Ughreja R, Carter CS. Social isolation induces behavioral and neuroendocrine disturbances relevant to depression in female and male prairie voles. Psychoneuroendocrinology 32(8‐10): 966‐980, 2007.
 110.Grippo AJ, Ihm E, Wardwell J, McNeal N, Scotti M‐AL, Moenk DA, Chandler DL, LaRocca MA, Preihs K. The effects of environmental enrichment on depressive and anxiety‐relevant behaviors in socially isolated prairie voles. Psychosom Med 76(4): 277‐284, 2014.
 111.Grippo AJ, Lamb DG, Carter CS, Porges SW. Social isolation disrupts autonomic regulation of the heart and influences negative affective behaviors. Biol Psychiatry 62(10): 1162‐1170, 2007.
 112.Grippo AJ, Moffitt JA, Sgoifo A, Jepson AJ, Bates SL, Chandler DL, McNeal N, Preihs K. The integration of depressive behaviors and cardiac dysfunction during an operational measure of depression: Investigating the role of negative social experiences in an animal model. Psychosom Med 74(6): 612‐619, 2012.
 113.Grippo AJ, Pournajafi‐Nazarloo H, Sanzenbacher L, Trahanas DM, McNeal N, Clarke DA, Porges SW, Sue Carter C. Peripheral oxytocin administration buffers autonomic but not behavioral responses to environmental stressors in isolated prairie voles. Stress Amst Neth 15(2): 149‐161, 2012.
 114.Grippo AJ, Trahanas DM, Zimmerman RR, Porges SW, Carter CS. Oxytocin protects against negative behavioral and autonomic consequences of long‐term social isolation. Psychoneuroendocrinology 34(10): 1542‐1553, 2009.
 115.Grippo AJ, Wu KD, Hassan I, Carter CS. Social isolation in prairie voles induces behaviors relevant to negative affect: Toward the development of a rodent model focused on co‐occurring depression and anxiety. Depress Anxiety 25(6): E17‐E26, 2008.
 116.Groppe SE, Gossen A, Rademacher L, Hahn A, Westphal L, Gründer G, Spreckelmeyer KN. Oxytocin influences processing of socially relevant cues in the ventral tegmental area of the human brain. Biol Psychiatry 74(3): 172‐179, 2013.
 117.Gruder‐Adams S, Getz LL. Comparison of the mating system and paternal behavior in Microtus ochrogaster and M pennsylvanicus. J Mammal 66(1): 165‐167, 1985.
 118.Guastella AJ, Kenyon AR, Alvares GA, Carson DS, Hickie IB. Intranasal arginine vasopressin enhances the encoding of happy and angry faces in humans. Biol Psychiatry 67(12): 1220‐1222, 2010.
 119.Guastella AJ, Mitchell PB, Dadds MR. Oxytocin increases gaze to the eye region of human faces. Biol Psychiatry 63(1): 3‐5, 2008.
 120.Hammock EAD, Lim MM, Nair HP, Young LJ. Association of vasopressin 1a receptor levels with a regulatory microsatellite and behavior. Genes Brain Behav 4(5): 289‐301, 2005.
 121.Hammock EAD. Developmental perspectives on oxytocin and vasopressin. Neuropsychopharmacol 40(1): 24‐42, 2015.
 122.Hammock EAD, Levitt P. Oxytocin receptor ligand binding in embryonic tissue and postnatal brain development of the c57bl/6j mouse. Front Behav Neurosci 7: 195, 2013.
 123.Hammock EAD, Young LJ. Variation in the vasopressin V1a receptor promoter and expression: implications for inter‐ and intraspecific variation in social behaviour. Eur J Neurosci 16(3): 399‐402, 2002.
 124.Hammock EAD, Young LJ. Functional microsatellite polymorphism associated with divergent social structure in vole species. Mol Biol Evol 21(6): 1057‐1063, 2004.
 125.Hawkley LC, Browne MW, Cacioppo JT. How can i connect with thee? let me count the ways. Psychol Sci 16(10): 798‐804, 2005.
 126.Hawkley LC, Cacioppo JT. Loneliness matters: A theoretical and empirical review of consequences and mechanisms. Ann Behav Med 40(2): 218‐227, 2010.
 127.Heinrich LM, Gullone E. The clinical significance of loneliness: A literature review. Clin Psychol Rev 26(6): 695‐718, 2006.
 128.Heinrichs M, Baumgartner T, Kirschbaum C, Ehlert U. Social support and oxytocin interact to suppress cortisol and subjective responses to psychosocial stress. Biol Psychiatry 54(12): 1389‐1398, 2003.
 129.Herman JP, Tasker JG, Ziegler DR, Cullinan WE. Local circuit regulation of paraventricular nucleus stress integration: Glutamate‐GABA connections. Pharmacol Biochem Behav 71(3): 457‐468, 2002.
 130.Hofmann JE, Getz LL, Gavish L. Home range overlap and nest cohabitation of male and female prairie voles. Am Midl Nat 112(2): 314‐319, 1984.
 131.Holt‐Lunstad J, Smith TB, Baker M, Harris T, Stephenson D. Loneliness and social isolation as risk factors for mortality: A meta‐analytic review. Perspect Psychol Sci 10(2): 227‐237, 2015.
 132.Hostinar CE, Sullivan RM, Gunnar MR. Psychobiological mechanisms underlying the social buffering of the hypothalamic‐pituitary‐adrenocortical axis: a review of animal models and human studies across development. Psychol Bull 140(1): 256‐282, 2014.
 133.House JS, Landis KR, Umberson D. Social relationships and health. Science 241(4865): 540‐545, 1988.
 134.Hurlemann R, Scheele D. Dissecting the role of oxytocin in the formation and loss of social relationships. Biol Psychiatry 79(3): 185‐193, 2016.
 135.Insel TR, Hulihan TJ. A gender‐specific mechanism for pair bonding: Oxytocin and partner preference formation in monogamous voles. Behav Neurosci 109(4): 782‐789, 1995.
 136.Insel TR, Preston S, Winslow JT. Mating in the monogamous male: Behavioral consequences. Physiol Behav 57(4): 615‐627, 1995.
 137.Insel TR, Shapiro LE. Oxytocin receptor distribution reflects social organization in monogamous and polygamous voles. Proc Natl Acad Sci 89(13): 5981‐5985, 1992.
 138.Insel TR, Wang ZX, Ferris CF. Patterns of brain vasopressin receptor distribution associated with social organization in microtine rodents. J Neurosci 14(9): 5381‐5392, 1994.
 139.Insel TR, Winslow JT, Williams JR, Hastings N, Shapiro LE, Carter CS. The role of neurohypophyseal peptides in the central mediation of complex social processes–evidence from comparative studies. Regul Pept 45(1‐2): 127‐131, 1993.
 140.Insel TR, Young L, Wang Z. Molecular aspects of monogamy. Ann N Y Acad Sci 807(1): 302‐316, 1997.
 141.Israel S, Lerer E, Shalev I, Uzefovsky F, Reibold M, Bachner‐Melman R, Granot R, Bornstein G, Knafo A, Yirmiya N, Ebstein RP. Molecular genetic studies of the arginine vasopressin 1a receptor (AVPR1a) and the oxytocin receptor (OXTR) in human behaviour: From autism to altruism with some notes in between. Prog Brain Res 170:435‐449, 2008.
 142.Jean‐Baptiste N, Terleph TA, Bamshad M. Changes in paternal responsiveness of prairie voles (Microtus ochrogaster) in response to olfactory cues and continuous physical contact with a female. Ethology 114(12): 1239‐1246, 2008.
 143.Jensen Peña C, Champagne FA. Implications of temporal variation in maternal care for the prediction of neurobiological and behavioral outcomes in offspring. Behav Neurosci 127(1): 33‐46, 2013.
 144.Johnson ZV, Walum H, Jamal YA, Xiao Y, Keebaugh AC, Inoue K, Young LJ. Central oxytocin receptors mediate mating‐induced partner preferences and enhance correlated activation across forebrain nuclei in male prairie voles. Horm Behav 79: 8‐17, 2016.
 145.Kang HJ, Kawasawa YI, Cheng F, Zhu Y, Xu X, Li M, Sousa AMM, Pletikos M, Meyer KA, Sedmak G, Guennel T, Shin Y, Johnson MB, Krsnik Z, Mayer S, Fertuzinhos S, Umlauf S, Lisgo SN, Vortmeyer A, Weinberger DR, Mane S, Hyde TM, Huttner A, Reimers M, Kleinman JE, Sestan N. Spatio‐temporal transcriptome of the human brain. Nature 478(7370): 483‐489, 2011.
 146.Keebaugh AC, Barrett CE, Laprairie JL, Jenkins JJ, Young LJ. RNAi knockdown of oxytocin receptor in the nucleus accumbens inhibits social attachment and parental care in monogamous female prairie voles. Soc Neurosci 10(5): 561‐570, 2015.
 147.Keebaugh AC, Young LJ. Increasing oxytocin receptor expression in the nucleus accumbens of pre‐pubertal female prairie voles enhances alloparental responsiveness and partner preference formation as adults. Horm Behav 60(5): 498‐504, 2011.
 148.Kenkel WM, Paredes J, Yee JR, Pournajafi‐Nazarloo H, Bales KL, Carter CS. Neuroendocrine and behavioural responses to exposure to an infant in male prairie voles. J Neuroendocrinol 24(6): 874‐886, 2012.
 149.Kenkel WM, Suboc G, Carter CS. Autonomic, behavioral and neuroendocrine correlates of paternal behavior in male prairie voles. Physiol Behav 128: 252‐259, 2014.
 150.Kiecolt‐Glaser JK, Newton TL. Marriage and health: his and hers. Psychol Bull 127(4): 472‐503, 2001.
 151.Kim P, Rigo P, Mayes LC, Feldman R, Leckman JF, Swain JE. Neural plasticity in fathers of human infants. Soc Neurosci 9(5): 522‐535, 2014.
 152.Kirkpatrick B, Carter CS, Newman SW, Insel TR. Axon‐sparing lesions of the medial nucleus of the amygdala decrease affiliative behaviors in the prairie vole (Microtus ochrogaster): Behavioral and anatomical specificity. Behav Neurosci 108(3): 501‐513, 1994.
 153.Kirkpatrick B, Kim JW, Insel TR. Limbic system fos expression associated with paternal behavior. Brain Res 658(1‐2): 112‐118, 1994.
 154.Kirkpatrick B, Williams JR, Slotnick BM, Carter CS. Olfactory bulbectomy decreases social behavior in male prairie voles (M. ochrogaster). Physiol Behav 55(5): 885‐889, 1994.
 155.Kleiman DG. Monogamy in mammals. Q Rev Biol 52(1): 39‐69, 1977.
 156.Knobloch HS, Charlet A, Hoffmann LC, Eliava M, Khrulev S, Cetin AH, Osten P, Schwarz MK, Seeburg PH, Stoop R, Grinevich V. Evoked axonal oxytocin release in the central amygdala attenuates fear response. Neuron 73(3): 553‐566, 2012.
 157.Kosfeld M, Heinrichs M, Zak PJ, Fischbacher U, Fehr E. Oxytocin increases trust in humans. Nature 435(7042): 673‐676, 2005.
 158.Kovács GL, Buijs RM, Bohus B, van Wimersma Greidanus TB. Microinjection of arginine8‐vasopressin antiserum into the dorsal hippocampus attenuates passive avoidance behavior in rats. Physiol Behav 28(1): 45‐48, 1982.
 159.Kramer KM, Yoshida S, Papademetriou E, Cushing BS. The organizational effects of oxytocin on the central expression of estrogen receptor alpha and oxytocin in adulthood. BMC Neurosci 8: 71, 2007.
 160.Kuroda KO, Tachikawa K, Yoshida S, Tsuneoka Y, Numan M. Neuromolecular basis of parental behavior in laboratory mice and rats: With special emphasis on technical issues of using mouse genetics. Prog Neuropsychopharmacol Biol Psychiatry 35(5): 1205‐1231, 2011.
 161.Landgraf R, Neumann ID. Vasopressin and oxytocin release within the brain: A dynamic concept of multiple and variable modes of neuropeptide communication. Front Neuroendocrinol 25(3‐4): 150‐176, 2004.
 162.Landgraf R, Neumann I, Pittman QJ. Septal and hippocampal release of vasopressin and oxytocin during late pregnancy and parturition in the rat. Neuroendocrinology 54(4): 378‐383, 1991.
 163.Lei K, Cushing BS, Musatov S, Ogawa S, Kramer KM. Estrogen receptor‐alpha in the bed nucleus of the stria terminalis regulates social affiliation in male prairie voles (Microtus ochrogaster). Plos One 5(1): e8931, 2010.
 164.Leknes S, Wessberg J, Ellingsen D‐M, Chelnokova O, Olausson H, Laeng B. Oxytocin enhances pupil dilation and sensitivity to “hidden” emotional expressions. Soc Cogn Affect Neurosci 8(7): 741‐749, 2013.
 165.Leng G, Ludwig M. Intranasal oxytocin: Myths and delusions. Biol Psychiatry 79(3): 243‐250, 2016.
 166.Levin R, Heresco‐Levy U, Bachner‐Melman R, Israel S, Shalev I, Ebstein RP. Association between arginine vasopressin 1a receptor (AVPR1a) promoter region polymorphisms and prepulse inhibition. Psychoneuroendocrinology 34(6): 901‐908, 2009.
 167.Licht G, Meredith M. Convergence of main and accessory olfactory pathways onto single neurons in the hamster amygdala. Exp Brain Res 69(1): 7‐18, 1987.
 168.Lieberwirth C, Liu Y, Jia X, Wang Z. Social isolation impairs adult neurogenesis in the limbic system and alters behaviors in female prairie voles. Horm Behav 62(4): 357‐366, 2012.
 169.Lim MM, Wang Z, Olazábal DE, Ren X, Terwilliger EF, Young LJ. Enhanced partner preference in a promiscuous species by manipulating the expression of a single gene. Nature. 429(6993): 754‐757, 2004.
 170.Lim MM, Young LJ. Vasopressin‐dependent neural circuits underlying pair bond formation in the monogamous prairie vole. Neuroscience 125(1): 35‐45, 2004.
 171.Liu Y, Curtis JT, Wang Z. Vasopressin in the lateral septum regulates pair bond formation in male prairie voles (Microtus ochrogaster). Behav Neurosci 115(4): 910‐919, 2001.
 172.Liu Y, Wang ZX. Nucleus accumbens oxytocin and dopamine interact to regulate pair bond formation in female prairie voles. Neuroscience 121(3): 537‐544, 2003.
 173.Lonstein JS, De Vries GJ. Comparison of the parental behavior of pair‐bonded female and male prairie voles (Microtus ochrogaster). Physiol Behav 66(1): 33‐40, 1999.
 174.Lorenz FO, Wickrama KAS, Conger RD, Elder GH. The short‐term and decade‐long effects of divorce on women's midlife health. J Health Soc Behav 47(2): 111‐125, 2006.
 175.Luppino D, Moul C, Hawes DJ, Brennan J, Dadds MR. Association between a polymorphism of the vasopressin 1b receptor gene and aggression in children. Psychiatr Genet 24(5): 185‐190, 2014.
 176.Mason WA, Mendoza SP. Generic aspects of primate attachments: Parents, offspring and mates. Psychoneuroendocrinology 23(8): 765‐778, 1998.
 177.McBride WJ, Murphy JM, Ikemoto S. Localization of brain reinforcement mechanisms: Intracranial self‐administration and intracranial place‐conditioning studies. Behav Brain Res 101(2): 129‐152, 1999.
 178.McGuire B, Novak M. A comparison of maternal behaviour in the meadow vole (Microtus pennsylvanicus), prairie vole (M. ochrogaster) and pine vole (M. pinetorum). Anim Behav 32(4): 1132‐1141, 1984.
 179.McNeal N, Scotti M‐AL, Wardwell J, Chandler DL, Bates SL, Larocca M, Trahanas DM, Grippo AJ. Disruption of social bonds induces behavioral and physiological dysregulation in male and female prairie voles. Auton Neurosci Basic Clin 180: 9‐16, 2014.
 180.Melchers M, Montag C, Markett S, Reuter M. Relationship between oxytocin receptor genotype and recognition of facial emotion. Behav Neurosci 127(5): 780‐787, 2013.
 181.Melis MR, Argiolas A. Central control of penile erection: A re‐visitation of the role of oxytocin and its interaction with dopamine and glutamic acid in male rats. Neurosci Biobehav Rev 35(3): 939‐955, 2011.
 182.Miller MA, Urban JH, Dorsa DM. Steroid dependency of vasopressin neurons in the bed nucleus of the stria terminalis by in situ hybridization. Endocrinology 125(5): 2335‐2340, 1989.
 183.Mohedano‐Moriano A, de la Rosa‐Prieto C, Saiz‐Sanchez D, Ubeda‐Bañon I, Pro‐Sistiaga P, de Moya‐Pinilla M, Martinez‐Marcos A. Centrifugal telencephalic afferent connections to the main and accessory olfactory bulbs. Front Neuroanat 6: 19, 2012.
 184.Mohr E, Meyerhof W, Richter D. Vasopressin and oxytocin: Molecular biology and evolution of the peptide hormones and their receptors. Vitam Horm 51: 235‐266, 1995.
 185.Moll J, Bado P, de Oliveira‐Souza R, Bramati IE, Lima DO, Paiva FF, Sato JR, Tovar‐Moll F, Zahn R. A neural signature of affiliative emotion in the human septohypothalamic area. J Neurosci 32(36): 12499‐12505, 2012.
 186.Neumann I, Russell JA, Landgraf R. Oxytocin and vasopressin release within the supraoptic and paraventricular nuclei of pregnant, parturient and lactating rats: A microdialysis study. Neuroscience 53(1): 65‐75, 1993.
 187.Neumann ID, Slattery DA. Oxytocin in general anxiety and social fear: A translational approach. Biol Psychiatry 79(3): 213‐221, 2016.
 188.Neumann ID, Wigger A, Torner L, Holsboer F, Landgraf R. Brain oxytocin inhibits basal and stress‐induced activity of the hypothalamo‐pituitary‐adrenal axis in male and female rats: Partial action within the paraventricular nucleus. J Neuroendocrinol 12(3): 235‐243, 2000.
 189.Ni R‐J, Shu Y‐M, Wang J, Yin J‐C, Xu L, Zhou J‐N. Distribution of vasopressin, oxytocin and vasoactive intestinal polypeptide in the hypothalamus and extrahypothalamic regions of tree shrews. Neuroscience 265: 124‐136, 2014.
 190.Numan M, Numan MJ, English JB. Excitotoxic amino acid injections into the medial amygdala facilitate maternal behavior in virgin female rats. Horm Behav 27(1): 56‐81, 1993.
 191.Numan M, Stolzenberg DS. Medial preoptic area interactions with dopamine neural systems in the control of the onset and maintenance of maternal behavior in rats. Front Neuroendocrinol 30(1): 46‐64, 2009.
 192.Numan M, Young LJ. Neural mechanisms of mother‐infant bonding and pair bonding: Similarities, differences, and broader implications. Horm Behav 77: 98‐112, 2016.
 193.Nyuyki KD, Waldherr M, Baeuml S, Neumann ID. Yes, i am ready now: Differential effects of paced versus unpaced mating on anxiety and central oxytocin release in female rats. PloS One 6(8): e23599, 2011.
 194.Okhovat M, Berrio A, Wallace G, Ophir AG, Phelps SM. Sexual fidelity trade‐offs promote regulatory variation in the prairie vole brain. Science 350(6266): 1371‐1374, 2015.
 195.Olazábal DE. Comparative analysis of oxytocin receptor density in the nucleus accumbens: An adaptation for female and male alloparental care? J Physiol Paris 108(2‐3): 213‐220, 2014.
 196.Olazábal DE, Pereira M, Agrati D, Ferreira A, Fleming AS, González‐Mariscal G, Lévy F, Lucion AB, Morrell JI, Numan M, Uriarte N. Flexibility and adaptation of the neural substrate that supports maternal behavior in mammals. Neurosci Biobehav Rev 37(8): 1875‐1892, 2013.
 197.Olazábal DE, Young LJ. Species and individual differences in juvenile female alloparental care are associated with oxytocin receptor density in the striatum and the lateral septum. Horm Behav 49(5): 681‐687, 2006.
 198.Olazábal DE, Young LJ. Oxytocin receptors in the nucleus accumbens facilitate “spontaneous” maternal behavior in adult female prairie voles. Neuroscience 141(2): 559‐568, 2006.
 199.Oliveras D, Novak M. A comparison of paternal behaviour in the meadow vole Microtus pennsylvanicus, the pine vole m. pinetorum and the prairie vole m. cchrogaster. Anim Behav 34(2): 519‐526, 1986.
 200.Ophir AG, Sorochman G, Evans BL, Prounis GS. Stability and dynamics of forebrain vasopressin receptor and oxytocin receptor during pregnancy in prairie voles. J Neuroendocrinol 25(8): 719‐728, 2013.
 201.Pan Y, Liu Y, Young KA, Zhang Z, Wang Z. Post‐weaning social isolation alters anxiety‐related behavior and neurochemical gene expression in the brain of male prairie voles. Neurosci Lett 454(1): 67‐71, 2009.
 202.Parker KJ, Lee TM. Interaction of photoperiod and testes development is associated with paternal care in Microtus pennsylvanicus (meadow voles). Physiol Behav 75(1‐2): 91‐95, 2002.
 203.Parker KJ, Lee TM. Female meadow voles (Microtus pennsylvanicus) demonstrate same‐sex partner preferences. J Comp Psychol 117(3): 283‐289, 2003.
 204.Pedersen CA. Oxytocin control of maternal behavior regulation by sex steroids and offspring stimuli. Ann N Y Acad Sci 807: 126‐145, 1997.
 205.Pedersen CA, Ascher JA, Monroe YL, Prange AJ. Oxytocin induces maternal behavior in virgin female rats. Science 216(4546): 648‐650, 1982.
 206.Pedersen CA, Boccia ML. Oxytocin antagonism alters rat dams' oral grooming and upright posturing over pups. Physiol Behav 80(2‐3): 233‐241, 2003.
 207.Pedersen CA, Boccia ML. Vasopressin interactions with oxytocin in the control of female sexual behavior. Neuroscience 139(3): 843‐851, 2006.
 208.Pedersen CA, Caldwell JD, Walker C, Ayers G, Mason GA. Oxytocin activates the postpartum onset of rat maternal behavior in the ventral tegmental and medial preoptic areas. Behav Neurosci 108(6): 1163‐1171, 1994.
 209.Perkeybile AM, Griffin LL, Bales KL. Natural variation in early parental care correlates with social behaviors in adolescent prairie voles (Microtus ochrogaster). Front Behav Neurosci 7: 21, 2013.
 210.Petitte T, Mallow J, Barnes E, Petrone A, Barr T, Theeke L. A systematic review of loneliness and common chronic physical conditions in adults. Open Psychol J 8(Suppl 2): 113‐132, 2015.
 211.Petrulis A. Chemosignals, hormones and mammalian reproduction. Horm Behav 63(5): 723‐741, 2013.
 212.Peuler JD, Scotti M‐AL, Phelps LE, McNeal N, Grippo AJ. Chronic social isolation in the prairie vole induces endothelial dysfunction: Implications for depression and cardiovascular disease. Physiol Behav 106(4): 476‐484, 2012.
 213.Phares V, Compas BE. The role of fathers in child and adolescent psychopathology: Make room for daddy. Psychol Bull 111(3): 387‐412, 1992.
 214.Phelps SM, Young LJ. Extraordinary diversity in vasopressin (V1a) receptor distributions among wild prairie voles (Microtus ochrogaster): Patterns of variation and covariation. J Comp Neurol 466(4): 564‐576, 2003.
 215.Pitkow LJ, Sharer CA, Ren X, Insel TR, Terwilliger EF, Young LJ. Facilitation of affiliation and pair‐bond formation by vasopressin receptor gene transfer into the ventral forebrain of a monogamous vole. J Neurosci 21(18): 7392‐7396, 2001.
 216.Pournajafi‐Nazarloo H, Kenkel W, Mohsenpour SR, Sanzenbacher L, Saadat H, Partoo L, Yee J, Azizi F, Carter CS. Exposure to chronic isolation modulates receptors mRNAs for oxytocin and vasopressin in the hypothalamus and heart. Peptides 43: 20‐26, 2013.
 217.Pow DV, Morris JF. Dendrites of hypothalamic magnocellular neurons release neurohypophysial peptides by exocytosis. Neuroscience 32(2): 435‐439, 1989.
 218.Quattrocki E, Friston K. Autism, oxytocin and interoception. Neurosci Biobehav Rev 47: 410‐430, 2014.
 219.Remage‐Healey L, Adkins‐Regan E, Romero LM. Behavioral and adrenocortical responses to mate separation and reunion in the zebra finch. Horm Behav 43(1): 108‐114, 2003.
 220.Resendez SL, Aragona BJ. Aversive motivation and the maintenance of monogamous pair bonding. Rev Neurosci 24(1): 51‐60, 2013.
 221.Resendez SL, Dome M, Gormley G, Franco D, Nevárez N, Hamid AA, Aragona BJ. M‐opioid receptors within subregions of the striatum mediate pair bond formation through parallel yet distinct reward mechanisms. J Neurosci 33(21): 9140‐9149, 2013.
 222.Resendez SL, Kuhnmuench M, Krzywosinski T, Aragona BJ. K‐opioid receptors within the nucleus accumbens shell mediate pair bond maintenance. J Neurosci 32(20): 6771‐6784, 2012.
 223.Richard S, Zingg HH. The human oxytocin gene promoter is regulated by estrogens. J Biol Chem 265(11): 6098‐6103, 1990.
 224.Rilling JK, Demarco AC, Hackett PD, Chen X, Gautam P, Stair S, Haroon E, Thompson R, Ditzen B, Patel R, Pagnoni G. Sex differences in the neural and behavioral response to intranasal oxytocin and vasopressin during human social interaction. Psychoneuroendocrinology 39: 237‐248, 2014.
 225.Rilling JK, DeMarco AC, Hackett PD, Thompson R, Ditzen B, Patel R, Pagnoni G. Effects of intranasal oxytocin and vasopressin on cooperative behavior and associated brain activity in men. Psychoneuroendocrinology 37(4): 447‐461, 2012.
 226.Rilling JK, Young LJ. The biology of mammalian parenting and its effect on offspring social development. Science 345(6198): 771‐776, 2014.
 227.Roberts RL, Miller AK, Taymans SE, Carter CS. Role of social and endocrine factors in alloparental behavior of prairie voles (Microtus ochrogaster). Can J Zool 76(10): 1862‐1868, 1998.
 228.Rodrigues SM, Saslow LR, Garcia N, John OP, Keltner D. Oxytocin receptor genetic variation relates to empathy and stress reactivity in humans. Proc Natl Acad Sci 106(50): 21437‐21441, 2009.
 229.Ross HE, Cole CD, Smith Y, Neumann ID, Landgraf R, Murphy AZ, Young LJ. Characterization of the oxytocin system regulating affiliative behavior in female prairie voles. Neuroscience 162(4): 892‐903, 2009.
 230.Ross HE, Freeman SM, Spiegel LL, Ren X, Terwilliger EF, Young LJ. Variation in oxytocin receptor density in the nucleus accumbens has differential effects on affiliative behaviors in monogamous and polygamous voles. J Neurosci 29(5): 1312‐1318, 2009.
 231.Ross HE, Young LJ. Oxytocin and the neural mechanisms regulating social cognition and affiliative behavior. Front Neuroendocrinol 30(4): 534‐547, 2009.
 232.Ruscio MG, Sweeny T, Hazelton J, Suppatkul P, Sue Carter C. Social environment regulates corticotropin releasing factor, corticosterone and vasopressin in juvenile prairie voles. Horm Behav 51(1): 54‐61, 2007.
 233.Sanna F, Argiolas A, Melis MR. Oxytocin‐induced yawning: Sites of action in the brain and interaction with mesolimbic/mesocortical and incertohypothalamic dopaminergic neurons in male rats. Horm Behav 62(4): 505‐514, 2012.
 234.Sarkadi A, Kristiansson R, Oberklaid F, Bremberg S. Fathers' involvement and children's developmental outcomes: A systematic review of longitudinal studies. Acta Pædiatrica 97(2): 153‐158, 2008.
 235.Scheele D, Wille A, Kendrick KM, Stoffel‐Wagner B, Becker B, Güntürkün O, Maier W, Hurlemann R. Oxytocin enhances brain reward system responses in men viewing the face of their female partner. Proc Natl Acad Sci U S A 110(50): 20308‐20313, 2013.
 236.Scotti M‐AL, Carlton ED, Demas GE, Grippo AJ. Social isolation disrupts innate immune responses in both male and female prairie voles and enhances agonistic behavior in female prairie voles (Microtus ochrogaster). Horm Behav 70: 7‐13, 2015.
 237.Shahrestani S, Kemp AH, Guastella AJ. The impact of a single administration of intranasal oxytocin on the recognition of basic emotions in humans: a meta‐analysis. Neuropsychopharmacol 38(10): 1929‐1936, 2013.
 238.Shahrokh DK, Zhang T‐Y, Diorio J, Gratton A, Meaney MJ. Oxytocin‐dopamine interactions mediate variations in maternal behavior in the rat. Endocrinology 151(5): 2276‐2286, 2010.
 239.Shamay‐Tsoory SG, Abu‐Akel A. The social salience hypothesis of oxytocin. Biol Psychiatry 79(3): 194‐202, 2016.
 240.Shamay‐Tsoory SG, Fischer M, Dvash J, Harari H, Perach‐Bloom N, Levkovitz Y. Intranasal administration of oxytocin increases envy and schadenfreude (gloating). Biol Psychiatry 66(9): 864‐870, 2009.
 241.Shapiro LE, Dewsbury DA. Differences in affiliative behavior, pair bonding, and vaginal cytology in two species of vole (Microtus ochrogaster and M. montanus). J Comp Psychol 104(3): 268‐274, 1990.
 242.Shear MK. Complicated grief. N Engl J Med 372(2): 153‐160, 2015.
 243.Skuse DH, Lori A, Cubells JF, Lee I, Conneely KN, Puura K, Lehtimäki T, Binder EB, Young LJ. Common polymorphism in the oxytocin receptor gene (OXTR) is associated with human social recognition skills. Proc Natl Acad Sci U S A 111(5): 1987‐1992, 2014.
 244.Smeltzer MD, Curtis JT, Aragona BJ, Wang Z. Dopamine, oxytocin, and vasopressin receptor binding in the medial prefrontal cortex of monogamous and promiscuous voles. Neurosci Lett 394(2): 146‐151, 2006.
 245.Smith AS, Lieberwirth C, Wang Z. Behavioral and physiological responses of female prairie voles (Microtus ochrogaster) to various stressful conditions. Stress Amst Neth 16(5): 531‐539, 2013.
 246.Smith AS, Tabbaa M, Lei K, Eastham P, Butler MJ, Linton L, Altshuler R, Liu Y, Wang Z. Local oxytocin tempers anxiety by activating GABAa receptors in the hypothalamic paraventricular nucleus. Psychoneuroendocrinology 63: 50‐58, 2015.
 247.Smith AS, Wang Z. Salubrious effects of oxytocin on social stress‐induced deficits. Horm Behav 61(3): 320‐330, 2012.
 248.Smith AS, Wang Z. Hypothalamic oxytocin mediates social buffering of the stress response. Biol Psychiatry 76(4): 281‐288, 2014.
 249.Song Z, McCann KE, McNeill JK, Larkin TE, Huhman KL, Albers HE. Oxytocin induces social communication by activating arginine‐vasopressin V1a receptors and not oxytocin receptors. Psychoneuroendocrinology 50: 14‐19, 2014.
 250.Steptoe A, Owen N, Kunz‐Ebrecht SR, Brydon L. Loneliness and neuroendocrine, cardiovascular, and inflammatory stress responses in middle‐aged men and women. Psychoneuroendocrinology 29(5): 593‐611, 2004.
 251.Stolzenberg DS, Champagne FA. Hormonal and non‐hormonal bases of maternal behavior: The role of experience and epigenetic mechanisms. Horm Behav 77: 204‐210, 2015.
 252.Stoop R. Neuromodulation by oxytocin and vasopressin in the central nervous system as a basis for their rapid behavioral effects. Curr Opin Neurobiol 29: 187‐193, 2014.
 253.Striepens N, Matusch A, Kendrick KM, Mihov Y, Elmenhorst D, Becker B, Lang M, Coenen HH, Maier W, Hurlemann R, Bauer A. Oxytocin enhances attractiveness of unfamiliar female faces independent of the dopamine reward system. Psychoneuroendocrinology 39: 74‐87, 2014.
 254.Succu S, Sanna F, Cocco C, Melis T, Boi A, Ferri G‐L, Argiolas A, Melis MR. Oxytocin induces penile erection when injected into the ventral tegmental area of male rats: Role of nitric oxide and cyclic gmp. Eur J Neurosci 28(4): 813‐821, 2008.
 255.Sun P, Smith AS, Lei K, Liu Y, Wang Z. Breaking bonds in male prairie vole: Long‐term effects on emotional and social behavior, physiology, and neurochemistry. Behav Brain Res 265: 22‐31, 2014.
 256.Swain JE, Kim P, Spicer J, Ho SS, Dayton CJ, Elmadih A, Abel KM. Approaching the biology of human parental attachment: Brain imaging, oxytocin and coordinated assessments of mothers and fathers. Brain Res 1580: 78‐101, 2014.
 257.Swanson LW, Sawchenko PE. Hypothalamic integration: Organization of the paraventricular and supraoptic nuclei. Annu Rev Neurosci 6: 269‐324, 1983.
 258.Tabak BA, Meyer ML, Castle E, Dutcher JM, Irwin MR, Han JH, Lieberman MD, Eisenberger NI. Vasopressin, but not oxytocin, increases empathic concern among individuals who received higher levels of paternal warmth: A randomized controlled trial. Psychoneuroendocrinology 51: 253‐261, 2015.
 259.Thomas JA, Birney EC. Parental care and mating system of the prairie vole. Microtus ochrogaster Behav Ecol Sociobiol 5(2): 171‐186, 1979.
 260.Tobin VA, Hashimoto H, Wacker DW, Takayanagi Y, Langnaese K, Caquineau C, Noack J, Landgraf R, Onaka T, Leng G, Meddle SL, Engelmann M, Ludwig M. An intrinsic vasopressin system in the olfactory bulb is involved in social recognition. Nature 464(7287): 413‐417, 2010.
 261.Tops M, van Peer JM, Korf J, Wijers AA, Tucker DM. Anxiety, cortisol, and attachment predict plasma oxytocin. Psychophysiology 44(3): 444‐449, 2007.
 262.van den Pol AN. The magnocellular and parvocellular paraventricular nucleus of rat: Intrinsic organization. J Comp Neurol 206(4): 317‐345, 1982.
 263.van Wimersma Greidanus TB, Maigret C. The role of limbic vasopressin and oxytocin in social recognition. Brain Res 713(1‐2): 153‐159, 1996.
 264.Veenema AH, Beiderbeck DI, Lukas M, Neumann ID. Distinct correlations of vasopressin release within the lateral septum and the bed nucleus of the stria terminalis with the display of intermale aggression. Horm Behav 58(2): 273‐281, 2010.
 265.Waldherr M, Neumann ID. Centrally released oxytocin mediates mating‐induced anxiolysis in male rats. Proc Natl Acad Sci U S A 104(42): 16681‐16684, 2007.
 266.Waltz M, Badura B, Pfaff H, Schott T. Marriage and the psychological consequences of a heart attack: A longitudinal study of adaptation to chronic illness after 3 years. Soc Sci Med 27(2): 149‐158, 1988.
 267.Walum H, Waldman ID, Young LJ. Statistical and methodological considerations for the interpretation of intranasal oxytocin studies. Biol Psychiatry 79(3): 251‐257, 2016.
 268.Wang B, Li Y, Wu R, Zhang S, Tai F. Behavioral responses to pups in males with different reproductive experiences are associated with changes in central ot, th and otr, d1r, d2r mRNA expression in mandarin voles. Horm Behav 67: 73‐82, 2015.
 269.Wang H, Duclot F, Liu Y, Wang Z, Kabbaj M. Histone deacetylase inhibitors facilitate partner preference formation in female prairie voles. Nat Neurosci 16(7): 919‐924, 2013.
 270.Wang Z. Species differences in the vasopressin‐immunoreactive pathways in the bed nucleus of the stria terminalis and medial amygdaloid nucleus in prairie voles (Microtus ochrogaster) and meadow voles (Microtus pennsylvanicus). Behav Neurosci 109(2): 305‐311, 1995.
 271.Wang Z, Aragona BJ. Neurochemical regulation of pair bonding in male prairie voles. Physiol Behav 83(2): 319‐328, 2004.
 272.Wang Z, Ferris CF, De Vries GJ. Role of septal vasopressin innervation in paternal behavior in prairie voles (Microtus ochrogaster). Proc Natl Acad Sci U S A 91(1): 400‐404, 1994.
 273.Wang Z, Hulihan TJ, Insel TR. Sexual and social experience is associated with different patterns of behavior and neural activation in male prairie voles. Brain Res 767(2): 321‐332, 1997.
 274.Wang Z, Insel T. Parental Care: Evolution, Mechanisms, And Adaptive Significance. San Diego, CA: Academic Press. p. 737, 1996.
 275.Wang Z, Novak MA. Influence of the social environment on parental behavior and pup development of meadow voles (Microtus pennsylvanicus) and prairie voles (M. ochrogaster). J Comp Psychol 106(2): 163‐171, 1992.
 276.Wang Z, Novak MA. Alloparental care and the influence of father presence on juvenile prairie voles, Microtus ochrogaster. Anim Behav 47(2): 281‐288, 1994.
 277.Wang Z, Smith W, Major DE, De Vries GJ. Sex and species differences in the effects of cohabitation on vasopressin messenger RNA expression in the bed nucleus of the stria terminalis in prairie voles (Microtus ochrogaster) and meadow voles (Microtus pennsylvanicus). Brain Res 650(2): 212‐218, 1994.
 278.Wang Z, Young LJ, Liu Y, Insel TR. Species differences in vasopressin receptor binding are evident early in development: Comparative anatomic studies in prairie and montane voles. J Comp Neurol 378(4): 535‐546, 1997.
 279.Wang Z, Zhou L, Hulihan TJ, Insel TR. Immunoreactivity of central vasopressin and oxytocin pathways in microtine rodents: A quantitative comparative study. J Comp Neurol 366(4): 726‐737, 1996.
 280.Wang ZX, Liu Y, Young LJ, Insel TR. Hypothalamic vasopressin gene expression increases in both males and females postpartum in a biparental rodent. J Neuroendocrinol 12(2): 111‐120, 2000.
 281.Williams JR, Catania KC, Carter CS. Development of partner preferences in female prairie voles (Microtus ochrogaster): The role of social and sexual experience. Horm Behav 26(3): 339‐349, 1992.
 282.Williams JR, Insel TR, Harbaugh CR, Carter CS. Oxytocin administered centrally facilitates formation of a partner preference in female prairie voles (Microtus ochrogaster). J Neuroendocrinol 6(3): 247‐250, 1994.
 283.Winslow JT, Hastings N, Carter CS, Harbaugh CR, Insel TR. A role for central vasopressin in pair bonding in monogamous prairie voles. Nature 365(6446): 545‐548, 1993.
 284.Wu N, Li Z, Su Y. The association between oxytocin receptor gene polymorphism (OXTR) and trait empathy. J Affect Disord 138(3): 468‐472, 2012.
 285.Wu N, Shang S, Su Y. The arginine vasopressin v1b receptor gene and prosociality: Mediation role of emotional empathy. PsyCh J 4(3): 160‐165, 2015.
 286.Yamamoto Y, Carter CS, Cushing BS. Neonatal manipulation of oxytocin affects expression of estrogen receptor alpha. Neuroscience 137(1): 157‐164, 2006.
 287.Yamamoto Y, Cushing BS, Kramer KM, Epperson PD, Hoffman GE, Carter CS. Neonatal manipulations of oxytocin alter expression of oxytocin and vasopressin immunoreactive cells in the paraventricular nucleus of the hypothalamus in a gender‐specific manner. Neuroscience 125(4): 947‐955, 2004.
 288.Young KA, Gobrogge KL, Liu Y, Wang Z. The neurobiology of pair bonding: Insights from a socially monogamous rodent. Front Neuroendocrinol 32(1): 53‐69, 2011.
 289.Young KA, Liu Y, Gobrogge KL, Wang H, Wang Z. Oxytocin reverses amphetamine‐induced deficits in social bonding: Evidence for an interaction with nucleus accumbens dopamine. J Neurosci 34(25): 8499‐8506, 2014.
 290.Young LJ. Oxytocin and vasopressin receptors and species‐typical social behaviors. Horm Behav 36(3): 212‐221, 1999.
 291.Young LJ, Barrett CE. Can oxytocin treat autism? Science 347(6224): 825‐826, 2015.
 292.Young LJ, Huot B, Nilsen R, Wang Z, Insel TR. Species differences in central oxytocin receptor gene expression: Comparative analysis of promoter sequences. J Neuroendocrinol 8(10): 777‐783, 1996.
 293.Young LJ, Lim MM, Gingrich B, Insel TR. Cellular mechanisms of social attachment. Horm Behav 40(2): 133‐138, 2001.
 294.Young LJ, Murphy Young AZ, Hammock EAD. Anatomy and neurochemistry of the pair bond. J Comp Neurol 493(1): 51‐57, 2005.
 295.Young LJ, Nilsen R, Waymire KG, MacGregor GR, Insel TR. Increased affiliative response to vasopressin in mice expressing the V1a receptor from a monogamous vole. Nature 400(6746): 766‐768, 1999.
 296.Young LJ, Wang Z. The neurobiology of pair bonding. Nat Neurosci 7(10): 1048‐1054, 2004.
 297.Young LJ, Wang Z, Insel TR. Neuroendocrine bases of monogamy. Trends Neurosci 21(2): 71‐75, 1998.
 298.Young LJ, Winslow JT, Nilsen R, Insel TR. Species differences in V1a receptor gene expression in monogamous and nonmonogamous voles: Behavioral consequences. Behav Neurosci 111(3): 599‐605, 1997.
 299.Zahed SR, Kurian AV, Snowdon CT. Social dynamics and individual plasticity of infant care behavior in cooperatively breeding cotton‐top tamarins. Am J Primatol 72(4): 296‐306, 2010.
 300.Zheng J‐J, Li S‐J, Zhang X‐D, Miao W‐Y, Zhang D, Yao H, Yu X. Oxytocin mediates early experience‐dependent cross‐modal plasticity in the sensory cortices. Nat Neurosci 17(3): 391‐399, 2014.

Contact Editor

Submit a note to the editor about this article by filling in the form below.

* Required Field

How to Cite

Manal Tabbaa, Brennan Paedae, Yan Liu, Zuoxin Wang. Neuropeptide Regulation of Social Attachment: The Prairie Vole Model. Compr Physiol 2016, 7: 81-104. doi: 10.1002/cphy.c150055