Next Article in Journal
Potential Biomarkers and Endometrial Immune Microenvironment in Recurrent Implantation Failure
Next Article in Special Issue
Xanthine–Dopamine Hybrid Molecules as Multitarget Drugs with Potential for the Treatment of Neurodegenerative Diseases
Previous Article in Journal
Linker Length Drives Heterogeneity of Multivalent Complexes of Hub Protein LC8 and Transcription Factor ASCIZ
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Role of Oxytocin and Vasopressin in Drug-Induced Reward—Implications for Social and Non-Social Factors

by
Olga Wronikowska-Denysiuk
*,
Weronika Mrozek
and
Barbara Budzyńska
Independent Laboratory of Behavioral Studies, Chair of Biomedical Sciences, Medical University of Lublin, Chodzki 4a Street, 20-093 Lublin, Poland
*
Author to whom correspondence should be addressed.
Biomolecules 2023, 13(3), 405; https://doi.org/10.3390/biom13030405
Submission received: 14 January 2023 / Revised: 17 February 2023 / Accepted: 19 February 2023 / Published: 21 February 2023

Abstract

:
Drug abuse is a worldwide problem that leads to negative physical, mental, and economic consequences. Although pharmacological strategies for drug addiction management have been widely studied, therapeutic options with high efficacy and a low side-effects profile are still limited. Recently, there has been a growing interest in oxytocin (OT) and vasopressin (AVP) systems as potential therapeutic targets for the treatment of drug abuse. OT and AVP are hypothalamic neuropeptides involved in numerous physiological processes. Additionally, studies show that these neurohormones are highly implicated in the modulation of a wide range of behaviors. Interestingly, ample evidence has shown that both, OT and AVP are able to decrease the consumption of different drugs of abuse, as well as to ameliorate their rewarding and reinforcing effects. Furthermore, OT and AVP have been strongly involved in prosocial effects and social reward. In particular, OT has been shown to be able to shift drug-induced reward into social-induced reward, mainly due to its interaction with the dopaminergic system. This phenomenon is also reflected in the results of clinical trials where intranasal OT shows promising efficacy in managing substance use disorder. Therefore, the aim of this review is to comprehensively characterize the involvement of OT and AVP in the rewarding and other behavioral effects of drugs of abuse in animal models, with a particular highlight on the impact of social factors on the observed effects. Understanding this relationship may contribute to higher drug development success rates, as a result of a more profound and deliberate studies design.

1. Introduction

According to the latest European Monitoring Centre for Drugs and Drug Addiction (EMCDDA) report, around 83 million or 28.9% of adults (aged 15–64) in the European Union are estimated to have used illicit drugs at least once in their lifetime. The most prominent emerging challenges presented by the European drug market consist of the widespread availability of a diverse range of drugs, their increasingly high purity or potency, and importantly, the ever more frequently observed transition from experimental to habitual and dependent consumption [1]. In the light of such circumstances, the definition of vulnerability factors that may influence the liability to drug abuse is a vastly important issue.
One of the factors that can impact the intensity of the rewarding experience derived from the intake of illicit drugs is social context during their consumption. Numerous studies in animal models [2,3,4,5], including our own study [6], have shown that drug-induced reward can be highly dependent on the social context, e.g., the company of the conspecific during drug intake. A similar pattern has been reported in humans where the social environment in which the drug is taken has influenced the liability to its abuse [7,8]. Furthermore, other social-dependent factors such as paternal behavior or friendships with peers have been shown to influence drug abuse and the development of drug addiction [9,10,11,12].
Many overlapping mechanisms can be responsible for the social-dependent and social-independent changes in drugs-triggered effects. Nevertheless, oxytocin (OT) and arginine vasopressin (AVP) are two neurohormones considered as important players in drug-related reward [3,13,14]. OT and AVP are the hypothalamic neurohormones, released into the bloodstream by the posterior pituitary [15]. The primary role of OT consists in the regulation of reproductive processes, and stimulation of labor and breast milk production [15,16], whereas the primary AVP function involves regulation of blood pressure by kidney- and vessel-associated mechanisms [15,17]. Nevertheless, these neurohormones additionally trigger complex effects in the human body, and thus, they have been appreciated in addition as neuromodulators capable of regulating social cognition and affecting a wide range of behaviors, such as social attachment, social exploration, recognition and aggression, pair bonding, as well as maternal, affiliative and sexual behaviors [18,19]. Furthermore, recent studies in humans have also suggested that central OT modulates social cognition, including an increase in interpersonal trust, eye gaze, face recognition, and the ability to infer the emotions of others based on facial cues [20].
Recently, there has been a growing interest in OT and AVP systems as potential therapeutic targets for the treatment of drug abuse. Therefore, the aim of this review is to comprehensively characterize the involvement of OT and AVP in the rewarding and other behavioral effects of drugs of abuse in animal models, with a special highlight on the impact of social factors on the observed effects. Specifically, the relationship between OT and AVP neurotransmission and the behavioral/rewarding effects of 3,4-methylenedioxymethamphetamine (MDMA), cocaine, ethanol, amphetamine, methamphetamine (METH), morphine, heroin and nicotine was reviewed in this paper. Because several clinical trials on OT use in drug addiction have already been conducted (see Section 6), a thoughtful insight into ligands acting via receptors for OT and AVP may result in identifying novel therapeutic targets for the treatment of substance use disorder.

2. Neuromodulation of OT and AVP

OT and AVP are neuropeptides consisting of nine aminoacids (nonapeptides) and differing only in two of them, at position 3 and 8 (Figure 1).
There are two major hypothalamic cells responsible for OT and AVP synthesis: magnocellular neurons and parvocellular neurons [21]. Previous theories assumed that these neurons were responsible for transmitting different signals. Precisely, it was believed that magnocellular neurons project to the posterior pituitary gland from where OT and AVP are released into the bloodstream, whereas parvocellular neurons project to other parts of the brain regulating different types of behavior. Nevertheless, the newest data, extensively reviewed by Grinevich and Ludwig (2021) [22], suggest that magnocellular OT and AVP neurons may additionally send axonal collaterals to forebrain regions and release these neuropeptides in micro volumes non-synaptically to modulate and control various behavioral responses (additionally by producing releasing factors, as it is reported for classical hypothalamic neurons) [23,24,25]. Importantly, the magnocellular OT neurons are controlled by parvocellular OT neurons in order to maintain homeostasis and regulate behavioral response. Nevertheless, a similar pattern has not been observed for parvocellular AVP neurons, and, at this stage of knowledge, they cannot be considered as independent cells [22].
The location of OT neurons and AVP neurons in rodents’ brains is similar; however, some differences have been identified. OT neurons are expressed mainly in the hypothalamic paraventricular nucleus (PVN) and supraoptic nucleus (SON) with an additional minor scatter in the bed nucleus of stria terminalis (BNST) [19,22,26]. AVP neurons are also expressed in PVN and SON but have been found additionally in the suprachiasmatic nucleus (SCN), BNST, medial nucleus of the amygdala and entorhinal cortex [27,28]. Interestingly, OT and AVP neurons can project to distinct locations through the brain, including the obvious, the pituitary, but also other regions and structures such as the amygdala, hippocampus, striatum, brainstem and spinal cord [19,22,29].
The effects of OT and AVP are exerted via modulation of their receptors (OT receptors, OTRs and AVP receptors AVPRs, respectively). These are G-protein coupled receptors (GPCR) and their activation leads to a conformational change in the receptor structure which leads to the activation of G proteins and subsequent Ca2+ release from intracellular stores [30,31]. One receptor for OT (OTR) and three types of AVPRs (AVPV1ARs, AVPV1BRs and AVPV2Rs) have been identified [32,33,34]. Extensive studies on rodents have identified the sites of expression of OTRs and AVPRs, both centrally and peripherally. In the brain, OTRs are expressed, among others, in the amygdala, nucleus accumbens (NAc), BNST, PVN, medial preoptic area, ventromedial nucleus of the hypothalamus, hippocampus, ventral pallidum, periaqueductal gray, striatum, lateral septum, ventral tegmental area, and olfactory bulb. OTRs are additionally expressed peripherally, mainly in the uterus, placenta and cardiovascular system (for a detailed list of central and peripheral OTRs locations see [29,33]). AVPV1ARs are expressed both peripherally, largely on the vascular smooth muscle but also in the testis, uterus, liver, blood vessels, and renal medulla, as well as centrally, in different brain areas, such as the lateral septal nucleus, thalamic nuclei, hippocampus, parts of the basal ganglia, and different brainstem nuclei [35,36]. AVPV1BRs are detectable mainly in the anterior pituitary, whereas AVPV2Rs are found mainly in the kidneys [29].
OT is released in response to sexual stimulation, stretching of the cervix and uterine dilatation during labor and with stimulation of the nipples during breastfeeding [15]. AVP displays a binary effect and is released into the circulation in response to extracellular fluid hyperosmolarity with a subsequent (1) increase in the reabsorption of solute-free water to blood from kidneys’ filtrate and an (2) increase in peripheral vascular resistance triggered by vessels constriction [15,17]. Apart from OT and AVP systemic activity, ample evidence has proven their involvement in regulating social and aggressive behaviors, stress adaptation, social memory, as well as maternal and sexual behaviors [37].

3. The Impact of OT/AVP and OTRs/AVPRs Ligands on Behavioral and Rewarding Effects of Drugs of Abuse in Animal Models

Extensive studies in animal models have proven the involvement of OT and AVP neurotransmission in behavioral and rewarding effects of different drugs of abuse. Specifically, the effects of OT, OTRs ligands, AVP and AVPRs ligands have been summarized in this review. The selection of studies included in these tables has been based on the following criteria: (1) systemic or central administration of OT/AVP or OTRs/AVPRs ligands combined with the systemic or central administration of any drug of abuse from MDMA, cocaine, ethanol, morphine, heroin, amphetamine, METH or nicotine; (2) presence of the behavioral impact of OT/AVP or OTRs/AVPRs ligands on the rewarding or other effects of the drug of abuse; (3) study performed in one of the species from rats or mice (any strain, including strains with genetic modifications), prairie wolves or zebrafish; (4) publication dates from 2000 (Figure 2). The choice of the animal species included in this review has been based on (a) their sociability and willingness to live in groups and (b) the presence of data on the chosen drugs of abuse and correlated addictive-like behaviors in those species. The abundance of presented data certainly proves the involvement of OT/AVP in the modulation of different effects of drugs of abuse; nevertheless, to comprehensively summarize collected data, several aspects require underlining, clarification and comment.

3.1. The Involvement of OT Transmission in Drug-Induced Reward

Findings reported in Table 1 and Table 2 suggest a general conclusion that the administration of (1) OT itself; (2) OT analog (Thr4,Gly7-OT) or (3) OTRs agonists (PF-06655075, WAY-267464 or carbetocin) and subsequent activation of OT transmission lead to attenuation of the rewarding effects of different drugs of abuse.
Such effects have been observed in different behavioral paradigms where OT has been shown to decrease the rewarding effects of METH [38,39] and morphine [40], measured in CPP. Furthermore, OT has been proven to reduce the intake of cocaine [41,42], METH [43,44,45] and ethanol [46,47,48,49,50,51,52,53,54]. Additionally, OT prevented the drug-, stress- and cue-induced reinstatement of cocaine [41,55,56,57,58,59,60], METH [39,44,45,61,62,63,64,65,66,67,68,69,70,71,72] and ethanol [73,74]. Finally, OT has been shown to prevent seeking during extinction and to facilitate the extinction of cocaine- [58,59] and METH-addictive effects [39].
Similar effects were observed for agonists of OTRs which were also shown to ameliorate drug reward. For example, carbetocin was able to prevent stress- and morphine-induced reinstatement to morphine-seeking [75,76] and attenuate the acquisition, facilitate extinction and block reinstatement of ethanol-induced CPP [77]. OT analog, Thr4,Gly7-oxytocin was able to block the cue-induced reinstatement of cocaine seeking [60] and the OTRs agonist while PF-06655075 was shown to decrease ethanol intake in dependent rats [52].
Interestingly, one study performed in adolescent rats reported that peripheral OT administration led to an increase in nicotine intake measured in two bottle free-choice paradigms [78], which could suggest that OT effects on drug-induced rewards may be age-dependent. Nevertheless, another study that took age into account showed that adolescent pretreatment with OT reduced METH-self administration and reinstatement to METH abuse in adult rats [44]. Therefore, the interaction between OT and the age-dependent effects of a drugs-induced reward needs further clarification. Importantly, the attenuation of drug reward triggered by OT activation can be prevented by the administration of OTRs antagonists (L,368,899 and atosiban) [39,47,72,79], which proves the involvement of OTRs in the observed effects. The effects of OT and OTRs ligands on behavioral/rewarding effects of different drugs of abuse has been summarized in Table 1 and Table 2, respectively.
Table 1. The effects of OT on behavioral/rewarding effects of different drugs of abuse.
Table 1. The effects of OT on behavioral/rewarding effects of different drugs of abuse.
Species and SexDrug of Abuse and DoseOT Effective DoseBehavioral TestOT-Induced EffectRef.
Long–Evans rats ♂MDMA
2.5 mg/kg, ip
0.25 mg/kg, ipSI↑ in adjacent lying[80]
Sprague Dawley rats ♂, ♀cocaine
(♂) 0.2 mg/50 μL/bolus, iv
(♀) 0.15 mg/50 μL/bolus, iv
0.1, 0.3, 1, 3 mg/kg, ipSA↓ cocaine intake (in ♀)[41]
1.0 mg/kg, ipSA↓ cue-induced cocaine seeking following extinction (in ♀)
Sprague Dawley rats ♂, ♀cocaine
0.5 mg/kg/infusion, iv
0.6 nmol/0.25 μL/side into NAcSA↓ cue-induced reinstatement of cocaine seeking[56]
0.6 nmol/0.25 μL/side into PFCSA↑ reinstatement to cocaine-associated cues
Sprague Dawley rats ♂cocaine
0.2 mg/50 μL/infusion, iv
10 mg/kg, ip, for cocaine- priming
0.3, 1, 3 mg/kg, ip during SASA↓ cocaine intake[42]
0.3, 1 mg/kg, ip during reinstatementSA↓ cocaine prime-induced (0.3 and 1 mg/kg) and cue-induced (1 mg/kg) reinstatement to cocaine seeking
Sprague Dawley rats ♂, ♀cocaine
(♂) 0.2 mg/50 μL/infusion, iv
(♀) 0.16 mg/50 μL/infusion, iv
0.3, 1 mg/kg, ipSA↓ cocaine-seeking during extinction and cue-induced reinstatement of cocaine-seeking[58]
Sprague Dawley rats ♂, ♀cocaine
(♂) 0.2 mg/50 μL/bolus, iv
(♀) 0.15 mg /50 μL/bolus, iv
1 mg/kg, ipSA↓ cue-induced cocaine reinstatement[55]
3 μg/0.5 μL/side, icvSA
OF1 mice ♂cocaine
1, 10 mg/kg, ip
1 mg/kg, ipCPP↓ social defeat-induced increase of cocaine (1 mg/kg) rewarding effects; facilitation of the extinction of cocaine (10 mg/kg)-CPP;[59]
cocaine
0.5 mg/kg/infusion, iv
10 mg/kg, ip for cocaine-priming
SA
facilitation of the extinction of cocaine-seeking behavior;
↓ of the cocaine-primed reinstatement of social defeat-induced cocaine-seeking
Sprague Dawley rats ♂cocaine
0.25 mg/0.1 mL.infusion, iv
10 ng/10 μL, icvSA↓ reinstatement of cue-induced cocaine seeking behavior[60]
EPM↓ anxiety triggered by cue-induced reinstatement conditions and cocaine-paired conditioned locomotion
Sprague Dawley rats ♂, ♀METH
0.1 mg/kg/50 μL/infusion, iv
0.3, 1 mg/kg, ipSA↓ cue-induced reinstatement (more in STs than in GTs)[69]
Swiss mice ♂METH
2 mg/kg, ip
0.5, 2.5 μg/μL, icvlocomotor activity test↓ METH-induced hyperactivity[79]
Swiss mice ♂METH
2 mg/kg, ip
0.5, 2.5 μg/μL into mPFC
2.5 μg/μL into DHC
CPP↓ stress-reinstained METH-induced CPP[72]
Long–Evans rats ♀METH
0.06 mg/kg/infusion, iv; PR
0.3 mg/kg, ipSA↓ BP in individually- and socially-housed rats[81]
Sprague Dawley rats ♂METH
2 mg/kg, ip
2 mg/kg, iplocomotor activity test↓ METH-induced hyperactivity[82]
Sprague Dawley rats ♂METH
0.1 mg/kg/infusion, iv
1 mg/kg, ip for METH-priming
1.5, 4.5 pmol
(500 nL/side) into NAc core
SA↓ METH-primed reinstatement[62]
Sprague Dawley rats ♂METH
0.1 mg/kg/infusion, iv
1 mg/kg, ip for METH-priming
3.6 pmol
(200 nL/side) into the STh
SA↓ METH-primed reinstatement[61]
Sprague Dawley rats ♀METH
0.01, 0.03, 0.1, 0.3, 1  mg/kg, iv
1 mg/kg, ip for METH-priming
1 mg/kg, ip
(during adolescence)
SA↓ METH (0.03 mg/kg) self-administration (in PR and not FR);
↓ METH (1 mg/kg)-primed reinstatement
[44]
Sprague Dawley rats ♂METH
0.1 mg/kg/50 μL/infusion, iv
1 mg/kg, ip for METH-priming
0.1 μg/side into the PrLSA↓ cue-induced METH reinstatement[68]
1.0, 3.0 μg/side into the PrLSA↓ METH-primed reinstatement
Sprague Dawley rats ♂METH
1 mg/kg, ip
0.6 mg, ipCPP↓ METH-induced CPP[38]
0.6 ng into the NAc core (0.5 μL/side) or into the STh (0.3 μL/side)CPP
Sprague Dawley rats ♂, ♀(with SDV)METH
0.1 mg/kg/50 μL/infusion, iv
1 mg/kg, ip for METH-priming
0.3, 1.0 mg/kg, ipSA↓ METH intake in ♂ and ♀ (with SDV-prevention of this OT-induced suppressant effect);
↓ cue- and METH-primed reinstatement (with SDV-prevention of this OT-induced suppressant effect; only in ♂)
[45]
Sprague Dawley rats ♂METH
0.1 mg/kg, PR, iv
1 mg/kg, ip for METH-priming
0.001, 0.01, 0.1, 0.3 and 1 mg/kg, ip, ascending (prior to self administration) or 1 mg/kg, ip (for reinstatement)SA↓ METH intake;
↓ METH-induced hyperactivity;
↓ relapse to METH-seeking behavior
[43]
Sprague Dawley rats ♂, ♀METH
(♀) 17.5 μg/50 μL/infusion, iv
(♂) 20 μg/50 μL/infusion, iv
1 mg/kg, ipSA↓ cue-induced METH seeking in ♂ and ♀[63]
0.6 nmol/0.25 μL/side into NaC coreSA
C57BL/6 mice ♂, ♀METH
2 mg/kg, ip
1.25 or 2.5 μg into hippocampusCPP↓ context- and restraint stress-induced reinstatement of METH-CPP[64]
Sprague Dawley rats ♂, ♀METH
(♀) 17.5 μg/50 μL/infusion, iv
(♂) 20 μg/50 μL/infusion, iv
1 mg/kg ipSA
(with BE procedure)
↓ METH-demand and ↓ reinstatement to METH-seeking in ♂ and ♀[66]
0.6 μg/μL into NAc coreSA
(with BE procedure)
↓ METH-seeking
C57BL/6 mice ♂METH
2.0 mg/kg, ip
2.5 μg, icvMWM;
NOR
↓ METH-induced spatial memory enhancement
↓ METH-induced cognitive memory deficits
[83]
Sprague Dawley rats ♂, ♀METH
0.1 mg/kg/50 μL/infusion, iv (followed by ShA or LgA sessions)
1 mg/kg, ip for METH-priming
1 mg/kg, ip
(during METH abstinence)
SA
EPM
↓ incubation and METH-primed reinstatement in ♂ and ♀
↓ of LgA-induced heightened anxiety phenotype effects
[70]
Sprague Dawley rats ♂METH
0.02 ug/50 μL/infusion, iv
1 mg/kg, ip, before reinstatementSA in rats pre-exposed to a predator odor threat (TMT)↓ METH-seeking in both saline- and TMT pre-exposed rats[71]
1 mg/kg, ip, prior to METH self-administration↓ stress-induced exacerbation of drug-seeking in TMT pre-exposed rats
Long–Evans rats ♂ and/or ♀METH
(♀) 17.5 μg/50 μL/infusion, iv
(♂) 20 μg/50 μL/infusion, iv
1 mg/kg, ip for METH-priming
1 mg/kg, ipSA↓ METH seeking and ↓ PR responding for METH in ♀;
↓ cue-induced METH-reinstatement in ♀;
↓ METH-primed induced METH-seeking in ♂ and ♀
[65]
Sprague Dawley rats ♂METH
0.1 mg /kg/50 μL/infusion, iv
1 mg/kg, ip for METH-priming
1 mg/kg, ipSA↓ METH-primed reinstatement[67]
3 pmol (500 nL/side) into the NAccSA
Swiss mice ♂METH
2 mg/kg, ip
0.1, 0.5, 2.5 μg/μL, icvCPP↓ acquisition METH-CPP;
facilitation of the extinction of METH-CPP;
↓ restraint stress-induced reinstatement to METH-CPP
[39]
Sprague Dawley rats ♂ (adolescent)nicotine
25 μg/mL per bottle
0.01 mg/kg, sctwo bottle free-choice paradigm↓ nicotine aversion after acclimation to nicotine solution[78]
1 mg/kg, sctwo bottle free-choice paradigm↑ nicotine intake
Wistar rats ♂nicotine
3.2 mg/kg/day, sc, in osmotic minipump
0.06, 0.125, 0.25, 0.50, 0.75, or 1.0 mg/kg, ipICSS and somatic signs evaluation↓ withdrawal-induced elevations in somatic signs in nicotine-dependent rats with no effect on nicotine withdrawal-induced elevations in ICSS thresholds[84]
Wistar rats ♂morphine
5 mg/kg, sc
0.2 μg, icvCPP↑ expression, but not acquisition, of morphine-induced CPP[40]
Wistar rats ♂morphine
5 and 10 μg/site into the mPFC
5 and 10 ng/site into the mPFCMWM↓ morphine-induced decrease in memory related activities[85]
Sprague Dawley rats ♂ethanol
10% and 15%
0.1, 0.3, and 0.5mg/kg, ipthree-bottle choice (modified DID model)↓ ethanol consumption[48]
ethanol
10% with gelatin
0.3 mg/kg, ipSA (oral)
C57BL/6J mice ♂, ♀ethanol
12%, 20 μL into the well
(a) 0.5, 1 mg/kg, ip (♀) and 1 mg/kg, ip (♂)
(b) 1 mg/kg, ip (♂ and ♀)
SA (oral)(a) ↓ of TMT-induced reinstatement of ethanol-seeking behavior;
(b) ↓ of yohimbine-induced reinstatement of ethanol-seeking behavior
[74]
Wistar rats ♂ethanol
20%
1 µg/5 µL, icvtwo-bottle free-choice paradigm↓ ethanol consumption[50]
C57BL/6J mice♂, ♀ethanol
3 and 6%
3 mg/kg, iptwo-bottle free-choice paradigm with RFIDs↓ ethanol consumption on 3 out 4 treatment days[51]
Wistar rats ♂ethanol
10%, 0.1 mL followed by exposition to ethanol vapor
0.25, 0.5, and 1 mg/kg for FR; 0.125, and 0.25 mg/kg for PRSA (oral) and alcohol vapor exposure↓ escalation of ethanol drinking (FR)
↓ enhanced motivation for ethanol (PR)
[52]
0.25, 0.5 and 1 mg/kg/20 μL; intranasal for FR and 1 mg/kg/20 μL intranasal for PRSA (oral) and ethanol vapor exposure
3, 10 and 30 μg, icvSA (oral) and ethanolapor exposure↓ ethanol consumption in dependent rats
Sprague Dawley rats ♂ethanol
20%
1 mg/kg, ipSA (oral)↓ of yohimbine-induced reinstatement of ethanol-seeking behavior[73]
0.5 μg intra-CeASA (oral)
Prairie voles ♂, ♀ethanol
15%
(a) 1, 3 and 10 mg/kg, ip
(b) 3 mg/kg, ip
two-bottle free-choiceparadigm(a) ↓ ethanol consumption (with restricted access to 15% ethanol)
(b) ↓ ethanol consumption (with continuous access to 15% ethanol), depending on time of testing
[54]
Wistar rats ♂ethanol
15%, 1.5 g/kg, ip
1 µg /5 μL, icvOF; wire-hanging test;righting-reflex test↓ ethanol-induced motor impairment (sedation and ataxia)[86]
C57BL/6N mice ♂ethanol2, 4, 6 and 8% (escalating)10 mg/kg, iptwo-bottle free-choice paradigm↓ ethanol consumption in control but not in CSC-triggered stressed mice[49]
C57BL/6N mice ♂ethanol
20%
1, 3, or 10 mg/kg, ipbinge-like DID↓ ethanol consumption[46]
1 mg/kg, iptwo-bottle free-choice paradigm↓ ethanol consumption
ethanol
12%, 20 μL to the well
0.1, 0.3, or 1 mg/kg, ip (FR) and 0.3 mg/kg, ip (PR)SA (oral)↓ ethanol consumption (FR) and ↓ motivation to seek ethanol reinforcement (PR)
Oxt-IRES-Cre mice without viral infusion ♂ethanol
20%
1 mg/kg, ipbinge-like drinking (DID)↓ ethanol consumption[47]
OF1 mice ♂ethanol
7.6%, 36 μLper nose poke
1 mg/kg, ipSA (oral)↓ (social-defeat)-induced increase in ethanol consumption[53]
BE—behavioral-economic; BP—breaking point (highest number of responses after 4 h or if 1 h elapsed without an infusion); CeA—central amygdala; CPP—conditioned place preference; CSC—subordinate colony housing (for stress induction); DHC—dorsal hippocampus; DID—drinking-in-the-dark model; EPM—elevated plus maze; FR—fixed ratio; GTs—“goal trackers”—rats for whom food reward-paired cue do not become imbued with incentive value, and do not motivate approach behavior; ICSS—intracranial self-stimulation; icv—intracerebroventricular injection; ip—intraperitoneal injection; LgA—long-access (6 h/day); METH—methamphetamine; mPFC—medial prefrontal cortex; MWM—Morris water maze; NAc—nucleus accumbens; NOR—novel object recognition; OF—open field test; PR—progressive ratio; RFIDs—radiofrequency identification tags (for monitoring of individual consumption); SA—self-administration; sc—subcutaneous injection; SDV—subdiaphragmatic vagotomy; ShA—short-access (2 h/day); SI—social interactions paradigm; STh—subthalamic nucleus; STs—“sign trackers”—rats for whom food reward-paired cue acquires control over motivated behaviors; TMT—2,5-dihydro-2,4,5-trimethylthiazoline (predator odor for stress induction); —decrease; —increase; ♀—female; ♂—male.
Table 2. The effects of OTRs ligands on behavioral/rewarding effects of different drugs of abuse.
Table 2. The effects of OTRs ligands on behavioral/rewarding effects of different drugs of abuse.
Species and SexDrug of Abuse and DoseOTR Ligand and DoseBehavioral TestOTR Ligand-Induced EffectRef.
Sprague Dawley rats ♂cocaine
0.25 mg/0.1 mL/infusion, iv
Thr4,Gly7-oxytocin
10 nmol/10 μL, icv
SA↓ reinstatement of cue-induced cocaine seeking behavior[60]
Wistar rats ♂ethanol
10%, 0.1 mL followed by exposition to ethanol vapor
PF-06655075
30 μg, icv
SA (oral) and ethanol vapor exposure↓ ethanol drinking in dependent rats[52]
Sprague Dawley rats
♂, ♀
ethanol
adolescent intermittent exposure—4 g/kg every 48 h for a total of 11 exposure
WAY-267464
5 mg/kg, ip
SIreversal of ethanol-induced social anxiety in ♂[87]
C57BL/6J mice ♂morphine
20–100 mg/kg/day—escalating, ip
carbetocin
6.4 mg/kg, ip
EPM, FST,
sociability and social novelty test
↓ withdrawal-induced negative emotional consequences (↓ of anxiety- and depressive-like and restoration of sociability behaviors)[75]
morphine
10 mg/kg, sc
carbetocin
6.4 mg/kg, ip
CPPprevention of stress-induced reinstatement to morphine-seeking
C57BL/6J mice ♂morphine
10 mg/kg, sc for conditioning;
2 mg/kg, ip for morphine-priming
carbetocin
6.4 mg/kg, ip
CPPprevention of morphine priming-induced reinstatement to morphine CPP[76]
C57BL/6 mice ♂ethanol
10%, 2 g/kg, ip
carbetocin
6.4 mg/kg, ip
CPP↓ acquisition of ethanol CPP;
facilitation of extinction of ethanol-CPP;
↓ reinstatement induced by ethanol priming
[77]
Sprague Dawley rats♂, ♀MDMA
1.5 mg/kg, ip
amphetamine
1 mg/kg, ip
carbetocin
2 and 20 mg/kg, ip
three-lever drug discrimination paradigm (MDMA/
amphetamine/saline)
↑ MDMA lever presses—carbetocin generalized to the MDMA training cue;
no effect on amphetamine lever selection
[88]
Swiss mice ♂ethanol
20%, 2 g/kg, ip
carbetocin
6.4 mg/kg, ip
CPPmimicking of behavioral effects of EE on ethanol-CPP
↑ of ethanol-CPP
[89]
L,368,899
5 mg/kg, ip
(during EE exposure but not during acquisition of ethanol CPP)
CPP↓ EE-induced ethanol CPP
C57BL/6J mice ♂MDMA
3 mg/kg, ip
L,368,899
10 mg/kg, ip
sociability test↓ prosocial effects of MDMA in highly sociable mice;
no effect in low sociable mice
[90]
Oxt-IRES-Cre mice with
intra-PVN infusion of active virus * ♂
ethanol
20%
L,368,899
10 mg/kg, ip
binge-like drinking (DID)reversal of (chemogenetic activation of PVN OT neurons)-induced ↓ of binge-like ethanol drinking[47]
C57BL/6J mice ♂ethanol
20%
L,368,899
10 mg/kg, ip
binge-like drinking (DID)↓ of OT-induced reduction in binge-like ethanol consumption[46]
Swiss mice ♂METH
2 mg/kg, ip
atosiban
2 μg/μL, icv
locomotor activity test↓ inhibitory effect of OT (0.5, 2.5 μg) on METH-induced hyperactivity in mice[79]
Swiss mice ♂METH
2 mg/kg, ip
atosiban
10 μg/μL into mPFC
CPP↓ OT (2.5μg/μL into mPFC)-induced inhibition of stress-reinstained METH-induced CPP[72]
Swiss mice ♂METH
2 mg/kg, ip
atosiban
2.0 μg/μL, icv
CPP↓ OT-induced effects (see Table 1)[39]
OF1 mice ♂cocaine
1 mg/kg, ip
atosiban
1 mg/kg, ip
CPPreversal of (positive social housing)-protective effect against increased cocaine reward[91]
Sprague Dawley rats
♂, ♀
MDMA
1.5 mg/kg, ip amphetamine
1 mg/kg, ip
atosiban
10 mg/kg, ip
three-lever drug discrimination paradigm (MDMA/
amphetamine/
saline)
disruption of MDMA- (but not AMP-) appropriate responding[88]
Wistar rats ♂MDMA
5 mg/kg, ip
tocinoic acid
20 μg/μL, icv
SI↓ the facilitation of MDMA-induced social interactions[92]
* with chemogenetic activation of OT-containing neurons; CPP—conditioned place preference; DID—drinking-in-the-dark model; EE—model of “eustress”—animals were exposed to different stimuli, such as toys, tubes, ladders, houses and running wheels (objects were changed/moved three times a week); EPM—elevated plus maze; FST—forced swim test; HPA—hypothalamic-pituitary-adrenal; icv—intracerebroventricular injection; ip—intraperitoneal injection; METH—methamphetamine; OT analog—Thr4,Gly7-oxytocin; OTRs agonists—PF-06655075, WAY-267464, carbetocin; OTRs antagonists—L,368,899, atosiban, tocinoic acid; SA—self-administration; sc—subcutaneous injection; SI—social interactions paradigm; ↓—decrease; ↑—increase; ♀—female; ♂—male.

3.2. The Involvement of AVP Transmission in Drug-Induced Reward

Research suggests that the administration of AVP (similarly to the administration of OT) can lead to the amelioration of drug-induced reward; however, the amount of available data is less abundant than reported for OT (Table 3). Specifically, it has been shown that central administration of AVP into the lateral septum (LS) blocked the expression of amphetamine-induced CPP [93]. Interestingly, the effects of the AVPV1ARs antagonist (SR49059) and the AVPV1BRs antagonist (SSR149415) on the drug-induced reward in rodents seem to be opposite (Table 4). Precisely, the effects of the AVPV1ARs blockage by its antagonist, SR49059, prevented OT-induced attenuation of METH-primed reinstatement in rats [67]. Conversely, the effects of the AVPV1BRs blockage by its antagonist, SSR149415, seem to be opposite to the effects of the AVPV1ARs blockage. Specifically, the administration of SSR149415 resulted in a decrease in ethanol intake [57,94,95], blockage of the acquisition of morphine-CPP in rats housed with identically treated conspecifics [14], attenuation of foot shock- and heroin priming-induced reinstatement to heroin self-administration [96] and prevention of nicotine withdrawal-induced dysphoria in intracranial self-stimulation (ICSS) [97]. The above-mentioned AVPV1ARs and AVPV1BRs distinctions suggest that a drug-induced reward in rodents may be attenuated by the blockage of AVPV1BRs, whereas the blockage of AVPV1ARs prevents the amelioration of a drug reward. Interestingly, the SR49059-induced AVPV1ARs blockage prevented the acquisition of MDMA-CPP in adult zebrafish [98], indicating a possible species-dependent effect; however, available data are insufficient to fully confirm this hypothesis. The effects of AVP and AVPRs ligands on behavioral/rewarding effects of different drugs of abuse has been summarized in Table 3 and Table 4, respectively.
Table 3. The effects of AVP on behavioral/rewarding effects of different drugs of abuse.
Table 3. The effects of AVP on behavioral/rewarding effects of different drugs of abuse.
Species and SexDrug of Abuse and DoseAVP Effective DoseTestAVP-Induced EffectRef.
Long–Evans rats ♂MDMA0.0025 mg/kg, ipSI↑ adjacent lying[80]
2.5 mg/kg, ip
Wistar or Sprague Dawley rats ♂ethanol
4.5%
(or CIE or modified CIE)
4 μg or 0.4 μg/0.5 μL * into CEASI↑ ethanol withdrawal-induced social anxiety[99]
Rats **amphetamine50 ng, icvnormal/abnormal
behavior
assessment ***
cross-sensitization of rats to amphetamine hyperlocomotion[100]
Sprague Dawley rats ♂amphetamine0.2 ng/side into LSCPP↓ expression of amphetamine-induced CPP[93]
* 4 μg/0.5 μL in both strains and 0.4 μg/0.5 μL only in Wistar rats; ** only information provided—Charles River strain; *** abnormal behaviors associated with AVP treatment: severe motor disturbances, sprawled-out posture with hind-limb extension and motor difficulty, and excessive scratching; abnormal behaviors associated with amphetamine administration: excessive locomotion and motor stereotypy; CEA—central nucleus of the amygdala; CIE—chronic intermittent ethanol exposure; CPP—conditioned place preference; icv—intracerebroventricular injection; ip—intraperitoneal injection; LS—lateral septum; SI—social interactions paradigm; ↓—decrease; ↑—increase; ♂—male.
Table 4. The effects of AVPRs ligands on behavioral/rewarding effects of different drugs of abuse.
Table 4. The effects of AVPRs ligands on behavioral/rewarding effects of different drugs of abuse.
Species and SexDrug of Abuse and DoseAVPR Ligand and DoseTestAVPR Ligand-Induced EffectRef.
adult zebrafishMDMA
5 mg/kg im
SR49059
0.01, and 0.1 ng/kg, im
CPP↓ MDMA-induced CPP[98]
SR49059
0.01, and 0.1 ng/kg, im
social preference test↓ MDMA-induced social preference
MDMA
10 mg/kg im
SR49059
0.1 and 1 ng/kg, im
novel tank diving test↓ MDMA-induced anxiolytic effect
MDMA
2.5 mg/kg im
SR49059
0.01 and 0.1 ng/kg, im
light-dark test↓ MDMA-induced anxiolytic effect
Sprague Dawley rats ♂METH
0.1 mg/kg/50 μL/ infusion, iv;
1 mg/kg, ip for METH-primed reinstatement
SR49059
1 mg/kg, ip
SA↓ OT-induced prevention of METH-primed reinstatement[67]
Long–Evans rats ♂MDMA
5 mg/kg, ip
SR49059
1 mg/kg, ip
SI↓ MDMA-induced adjacent lying[80]
Sprague Dawley rats ♂, ♀ethanol
adolescent intermittent exposure—4 g/kg every 48 h for a total of 11 exposures
SSR149415
5, 10, 20 mg/kg, ip
SI↓ ethanol-induced social anxiety[87]
Wistar rats ♂nicotine
0.4 mg/kg, sc
SSR149415
30 mg/kg, ip
locomotor activity test↓ expression of nicotine-induced sensitization[101]
C57BL/6J mice ♂, ♀ethanol
15%
SSR149415
10 and 30 mg/kg ip
two-bottle choice paradigm with IA↓ ethanol intake and preference[57]
SSR149415
1 and 3 mg/kg + naltrexone 1 mg/kg
sP and sNP rats ♂ethanol
10%
SSR149415
30 mg/kg, ip
two-bottle choice paradigm↓ ethanol intake in sP rats[95]
Wistar rats ♂nicotine
3.16 mg/kg/day in osmotic sc minipumps
SSR149415
0.1, 0.5, and 2 μg, icv (acute);
0.5 μg/day for 6 days, icv (chronic)
ICSS
(mecamylamine-precipitated nicotine withdrawal)
chronic treatment: complete prevention of the elevations in brain reward thresholds linked with nicotine withdrawal (prevention of the nicotine withdrawal-caused dysphoria);
acute treatment: partial prevention of nicotine withdrawal
[97]
C57BL/6N mice ♂morphine
10, 20 or 40 mg, sc
(6 day progressive ratio)
SSR149415
10 mg/kg, ip
CPP↓ acquisition of morphine-CPP in the morphine only mice * (no effect on the acquisition of morphine CPP in the morphine cage-mate mice **)[14]
Wistar rats ♂ethanol
CIEV adjusted by controlling BALs
SSR149415
30 mg/kg, ip
SA↓ excessive levels of ethanol SA observed in dependent animals without affecting ethanol drinking in non-dependent animals[94]
Fischer rats ♂heroin
0.05 mg/kg/infusion, iv
0.25 mg/kg, sc, for priming
SSR149415
30 mg/kg, ip
SA↓ foot shock-induced heroin reinstatement
↓ heroin-primed heroin reinstatement
[96]
Fischer rats ♂cocaine
45–90 mg/kg/day, ip
(chronic binge pattern with EDC)
SSR149415
5 mg/kg, ip
chronic EDC binge cocaine with acute withdrawal paradigm↓ acute withdrawal-induced HPA axis activation (ACTH increase) after EDC[102]
Wistar rats ♂ethanol
4.5% modified CIE or CIE
SSR149415
5 μg in 0.5 μL into CEA
SIbinary effect: ↓ of social interactions in control animals but reversal of ethanol withdrawal-induced decrease in social interactions[99]
* morphine-treated animals housed with only morphine-treated animals; ** morphine-treated animals housed with drug-naïve anim; BALs—blood alcohol levels; CEA—central nucleus of the amygdala; CIE—chronic intermittent ethanol exposure; CIEV—chronic, intermittent, exposure to ethanol vapors; CPP—conditioned place preference; EDC—escalating-dose cocaine; IA—chronic intermittent access; icv—intracerebroventricular injection; ip—intraperitoneal injection; SA—self-administration; sc—subcutaneous injection; SI—social interactions paradigm; sNP—Sardinian alcohol-nonpreferring rats; sP—Sardinian alcohol-preferring rats; SR49059—AVPV1ARs antagonist; SSR149415—AVPV1BRs antagonist ↓—decrease; ♀—female; ♂—male.

3.3. The Relationship between Social Factor and OT/AVP Impact on the Effects of Drugs of Abuse

Data summarized in the previous sections (Section 3.1 and Section 3.2) undeniably show that alternations in OT and AVP transmission may influence the drug-induced reward. Nevertheless, those effects can be additionally strongly modified by a social factor.
In rats, it has been shown that MDMA, OT and AVP do not induce classic-CPP but, interestingly, MDMA and OT are able to produce social-CPP (conducted with the presence of another conspecific during conditioning sessions) [3]. In the social interactions test MDMA, OT and AVP increased the time of the adjacent lying of rats (a prosocial effect) and this effect was attenuated by SR49059 (AVPV1ARs antagonist), suggesting an involvement of these receptors in the observed phenomenon [80]. A similar pattern has been reported in zebrafish, where SR49059 was able to block MDMA-induced CPP and decrease MDMA-induced social preference [98]. In terms of OTRs, it has been shown that the OTR agonist, tocinoid acid, was able to attenuate the MDMA-induced facilitation of social interactions in rats [92] and that the OTR antagonist, L-368,899 was able to abolish the prosocial effects of MDMA in highly sociable mice with no effect on low-sociable mice [90]. Conversely, another OTRs antagonist, compound 25 (5-{3-[3-(2-chloro-4-fluorophenoxy)azetidin-1-yl]-5-(methoxymethyl)-4H-1,2,4-triazol-4-yl}-2-methoxypyridine), failed to attenuate an MDMA-, OT- and AVP-induced increase in adjacent lying [80], pointing to the need for further clarification. What needs to be highlighted is that MDMA is an empathogenic/entactogenic drug with strong prosocial effects in humans [103,104]. This is a distinctive feature that differentiates this drug from other psychoactive drugs reviewed in this paper. Nevertheless, the social factor has also been shown to alter OT-/AVP-dependent rewarding effects of other drugs of abuse.
It has been reported that OT can attenuate the social defeat-induced increase in cocaine reward and attenuate the cocaine-primed reinstatement of social defeat-induced cocaine seeking [59]. OT also decreased the social defeat-induced increase in ethanol consumption [53] and the OTR agonist (WAY-267464) has been shown to reverse ethanol-induced social anxiety [87]. Treatment with the AVPV1BRs antagonist (SSR149415) also led to attenuation of ethanol-induced social anxiety [87]. SSR149415 also produced an interesting effect in the morphine-CPP experiment where it was able to block the acquisition of morphine-CPP in morphine-treated animals housed with only morphine-treated animals, but had no effect on morphine-CPP in morphine-treated animals housed with drug-naïve animals [14]. The graphical summary of the OT/AVP-dependent social effects is presented in Figure 3.
All of the above-mentioned examples show that the social factor is a highly important component of OT/AVP-dependent drug reward. Unfortunately, because the vast majority of currently available studies did not take this variable into consideration, full understanding of this relationship is yet to be discovered. Nevertheless, some hypotheses can be proposed.
Firstly, most studies in this area have been performed on rodents which are naturally social animals [15] in which negative social encounters can facilitate the development of drug addiction [105] and in which drug withdrawal can lead to social impairment [106]. Conversely, positive social interactions and social experience have the potential to manage or overcome drug addiction, most likely due to the backward shift of the drug reward onto social reward. In the light of this hypothesis (excellently reviewed in [93]), OT is able to: (a) reduce drug reward by decreasing the DA release and DA transmission in brain areas involved in drug addiction [38,39,50,66,79,107]; (b) enhance social reward by increasing positive prosocial behaviors [80]) and by restoring drugs-induced social deficits [75,108,109]; (c) regulate emotional states by decreasing drugs- and cue-induced anxiety [60,70,75] and drugs-induced depressive-like behaviors [75].
Secondly, following a closer look into the OT/AVP-related effects in different drugs of abuse (Table 1, Table 2, Table 3 and Table 4), a specific pattern can be observed, suggesting that these OT/AVP effects may be dependent on the type of the studied drug (empathogens vs. other psychoactive drugs). As mentioned above, MDMA is a classic empathogenic drug that in humans increases empathy, willingness to socialize and being close to others, talkativeness, amicability, and gregariousness [103]. In animal models, MDMA, OT and AVP triggered similar prosocial effects (increase in adjacent lying, induction of social- but not classic-CPP) [3,80]. Additionally, MDMA administration has been reported to increase plasma OT level in animals [92] and humans [110,111]. Conversely, numerous available studies on various psychoactive drugs that are not classified as empathogens (e.i. morphine, cocaine, ethanol) showed that chronic exposure to these drugs leads to a reduction in the plasma and central OT level [75,76,112,113]. Interestingly, in rats trained to discriminate MDMA, amphetamine and saline in a three-lever drug discrimination paradigm, carbetocin (an OTRs agonist) substitution led to an increase in MDMA-lever presses when compared to saline-lever presses while atosiban (an OTRs antagonist) was able to selectively disrupt MDMA-, but not amphetamine-, lever responding [88]. This distinction between MDMA and other mentioned drugs may indicate that OT/AVP-related effects are different in empathogens and classic psychostimulants. This observation needs to be taken into consideration in future studies on new empathogenic drugs.
Thirdly, an important aspect of an unaware social component in the experiment design needs to be discussed. Although many studies on OT/AVP involvement in drug reward did not take into consideration the impact of social factors on their results, an unintended and unplanned effect of the social variables, such as (a) housing; (b) same/different treatment regimen with/without cage mates and (c) waiting time between drug administration and testing with/without other conspecifics in home cages/transfer cages; are additionally important factors that can influence the experience derived from drug administration. This is specifically important for experiment designs that involve treatment with OT/AVP or their ligands. As mentioned above, it has been already proven that housing conditions (housing with drug- or vehicle-treated cage mates) can affect the acquisition of morphine-CPP [14]. Therefore, based on the above-mentioned reasons, a strong conclusion should be stated that while studying drug- and social-reward, as well as the OT/AVP-dependent effects of addictive substances, a social factor should be considered as an important variable and should not be neglected in the experiment design and analysis.

3.4. The Impact of OT/AVP and OTRs/AVPRs Ligands on Other Behavioral Effects of Drugs of Abuse

Although the main objective of this review is to elaborate on the interaction between OT/AVP and drug-reward, the impact of these neuropeptides on other drug-induced behavioral effects cannot remain unremarked on. Specifically, the OT/AVP impact on anxiety- or depressive-like behaviors (as indicators of emotional states), memory performance and locomotor effects needs to be highlighted.
Available research reported that OT may decrease anxiety (measured in an elevated plus maze (EPM) test) triggered by long access (LgA) to METH [70] and by the reinstatement induced by cocaine and cues [60]. A similar effect has been observed for the OTRs agonist, carbetocin, which was able to reduce morphine withdrawal-induced anxiety and depressive-like behaviors evaluated in the EPM and forced swim test (FST), respectively [75]. In terms of memory performance, OT was able to decrease METH-induced cognitive memory deficits in the novel object recognition (NOR) test [83] and to decrease morphine-induced spatial memory deterioration in the Morris water maze (MWM) test [85]. However, OT also decreased METH-induced spatial memory enhancement in the MWM [83], suggesting drug- and memory-type-dependent effects. OT also reduced METH-induced hyperlocomotion [79,82] and the OTRs antagonist, atosiban, was able to block this OT-induced effect [79]. Furthermore, OT prevented ethanol-induced motor impairment assessed in an open field (OF), wire-hanging and righting-reflex test [86]. Finally, OT was able to reduce withdrawal somatic signs in nicotine-dependent animals [84].
The effects of AVP treatment on anxiety levels are difficult to assess due to the scarcity of available data. Nevertheless, it has been reported that AVP microinjected into the central nucleus of the amygdala (CEA) can induce social anxiety-like behavior in rats exposed to chronic ethanol and that SSR149415 (AVPV1BRs antagonist) was able to reduce this ethanol-induced social anxiety [99]. Interestingly, administration of SR49059 (AVPV1ARs antagonist) in zebrafish led to a decrease in the MDMA-induced anxiolytic effect [98], which further supports the hypothesis of the bidirectional effects of AVPV1ARs and AVPV1BRs antagonists. In terms of AVP effects on animals’ locomotion, it has been shown that AVP treatment leads to cross-sensitization to amphetamine-induced hyperlocomotion [100] and that SSR149415 is able to decrease the expression of nicotine-induced locomotor sensitization [101]. The impact of AVP and AVPRs ligands on depressive-like behavior and memory performance is yet to be discovered.

4. Interactions between OT/AVP Transmission, DA Release and Drug Reward System

Ample evidence has proven the involvement of OT and AVP in drugs of abuse-related effects, showing that OT/AVP treatment contributes to the attenuation of drug consumption and reward (Table 1, Table 2, Table 3 and Table 4). Therefore, recognizing the interactions between OT/AVP, DA and the drug reward system is crucial to fully understand the mechanisms underlying the observed phenomenon. Although it is a complex and extensive issue, several aspects must be highlighted.
Firstly, OT has been shown to directly interact with dopaminergic brain reward regions. For example, OTRs have been found on the VTA dopaminergic neurons that project from the PVN into the NAc and medial prefrontal cortex (mPFC) [114,115]. The location of OTRs in the mesocorticolimbic structures triggers direct OT and DA interactions. It has been reported that subchronic OT treatment decreased DA release in the NAc and amygdala in drug non-exposed animals [107]. Furthermore, central OT administration blocked DA release in the NAc in ethanol-treated rats [50], inhibited METH-induced DA turnover in the NAc [79] and decreased DA release [38,66] in the NAc. Importantly, chronic treatment with various drugs of abuse has been shown to decrease plasma and the central OT level in animal models [75,76,112,113,116], which suggests drug-induced hypofunction of the OT system and may contribute to the dysfunction of social behavior and further development of addiction [117]. These findings were consistent with effects observed in humans where chronic cocaine use during pregnancy led to a decrease in the plasma OT level and mood disruption in mothers [118].
Another aspect that needs highlighting is a differentiation in the role of dopaminergic D1 and D2 receptors (D1Rs and D2Rs, respectively) in addiction. Available reports suggest that D1Rs are mainly involved in drug reward whereas D2Rs are mostly involved in social reward [106,119]. Drug administration leads to an increased level in DA which can interact with D1Rs, as well as D2Rs, creating both drug- and social-related rewarding effects, respectively [120]. Nevertheless, chronic drug consumption has been linked with decreased DA levels in the basal striatum and downregulation of D2Rs [105,106,120]. This may contribute to higher D1Rs versus D2Rs activation and further reinforce drug-reward and weaken social-reward [106]. Interestingly, recent studies have reported that OTRs can form heterodimer complexes with D2Rs (OTRs/D2Rs) in NAc, amygdala and the dorsal striatum where the activation of OTRs led to increased D2Rs signaling (by increased affinity and density of D2Rs) [121,122,123,124,125]. Therefore, OT may be able to reverse the above-mentioned imbalance in D1Rs/D2Rs activation, increase D2Rs activation and thus, shift the drug-reward into social-reward [106]. Taking into account the fact that drug abuse can lead to social impairment [126], the OT regulating hypothesis is strongly relevant in terms of OT use in the treatment of drug addiction and drug-induced social disruptions.
Regrettably, current knowledge on AVP and DA interactions is scarce. Hitherto, it has been shown that amphetamine treatment leads to a decrease in AVP levels in LS [93,127] and that AVP microinjected into LS decreases DA release in the NAc 930. Furthermore, an increase in striatal expression of D1Rs (but not D2Rs) and an increase in the striatal AVP expression level was observed in morphine-treated animals when housed with identically treated conspecifics but not when housed with drug-naive animals [14], which highlights the significance of social factors in the development of drug addiction. Interestingly, AVP gene expression analysis showed its down-regulation during early- and mid-acquisition and up-regulation during the late-acquisition and expression of environment-elicited cocaine conditioning [128], which suggests addiction stage-dependent changes. Nevertheless, further studies are needed to fully elucidate the interaction between the AVP and DA mesocorticolimbic system in the context of drug reward.

5. Interactions between OT/AVP and Serotonergic Transmission

One of the most important findings in the area of interest has been presented by Dölen et al. (2013) [129], who showed that social reward is dependent on the combined action of OT and serotonin (5-HT) in the NAc core. The activation of OTRs within the NAc of mice (which are colocalized with presynaptic terminals of serotonergic inputs from the dorsal raphe nucleus) leads to the release of 5-HT and is required for the social reinforcement measured in social-CPP. Interestingly, this effect is abolished by the presence of the 5-HT1B antagonist [129]. This finding may be specifically important in terms of elucidating the difference between the social reward of empathogens (such as MDMA) and other psychoactive drugs. The most prominent difference between these drugs is the intensity of interaction with the serotonergic transmission. MDMA (apart from classic dopaminergic mechanisms) can strongly interact with the 5-HT transporter (SERT) and stimulate the release of 5-HT which is (along with OT mechanisms) responsible for the prosocial effects [130]. This combined strong interactions with OT/AVP [3,80,92] and serotonergic transmission [130] seems to be the key feature of MDMA empathogenic and prosocial activity. This may be also connected with the fact that OT infusion is able to promote 5-HT release within the median raphe nucleus which has been linked with subsequent anxiolytic activity [131].
Interestingly, the OT/5-HT interactions have additionally been shown to take part in important aspects of motherhood, such as postpartum nursing, anxiety, aggression and stress management [132]. Several studies have also demonstrated the significance of OT/5-HT interactions in the autistic spectrum disorder (ASD), a condition that is characterized, among other features, by disruptions in social behavior [133,134]. Specifically, OT/5-HT interactions via 5-HT1A receptors have been shown to play an important role in the development of social behavior showing a possible novel strategy for the treatment of ASD [135].

6. Clinical Trials of Intranasal OT in Drug Abuse

The importance of conducting further preclinical studies on OT/AVP involvement in drug addiction is supported by promising results of available clinical trials on intranasal OT in the treatment of drug abuse. This route of administration allows us to increase central oxytocin levels through a direct nose-to-brain delivery [136]. Specifically, intranasal OT has been shown to reduce [137] heroin cravings. Additionally, intranasal OT has been reported to improve social perception and decrease the appetitive approach [138], as well as to reduce ethanol cravings and withdrawal symptoms in humans with ethanol abuse disorder [138,139]. Furthermore, intranasal OT has successfully decreased cocaine [140], nicotine [141] and cannabis [142] craving or use, as well as reducing cannabis-induced anxiety [142]. The above-mentioned examples certainly prove the possibility of the utility of intranasal OT in the treatment of drug addiction. However, these positive and promising effects need to be counterbalanced with other reports indicating that intranasal OT does not influence ethanol withdrawal scores [143], may have a negative effect on ethanol-dependent patients with anxious emotional states [138] and increase the craving for cocaine use [144]. Finally, it needs to be mentioned that all of the above-mentioned effects may be dependent on: (a) the administered dose—40 IU [137,138,140,141,142] vs. 24 IU [139,143,144] and (b) dosage regimen, namely single [137,138,141,142] vs. repeated administration [139,140,143]. Therefore, creating an appropriate dosage model may be the key to going successfully through all stages of the clinical trials and drug registration process.

7. Conclusions and Future Research Directions

All of the findings reviewed in this paper certainly prove the importance of OT and AVP systems as promising targets for the management of drug addiction. One of the most important observations is that OT has been shown to be able to reverse drug-induced social disruptions and to shift drug-induced reward into social-induced reward. These complementing effects of decreasing drug reward on the one hand, and reducing drug-related social impairment on the other, seem to be a unique feature in currently available pharmacological treatment options for drug abuse. Further, ever more studies in animal models indicate the potential utility of the AVPRs ligands in the management of addiction. However, since the quantity of data is still scarce, future studies should: (a) focus on advancing knowledge about AVPRs ligands, aim to select the most promising substance for possible clinical trials; (b) clarify the bidirectional effects of AVPV1ARs and AVPV1BRs in the mitigation of drug-reward; (c) take social factors into consideration while designing the drug experiment and incorporate it in results analysis; (d) further evaluate binary OT/AVP-dependent effects in empathogens vs. other psychoactive substances—preferably in newly synthesized MDMA-like empathogens. Explanation of these above-mentioned unknown phenomena will undeniably broaden our understanding of the role of OT and AVP in drug abuse and contribute to the development of effective interventions for treating substance use disorders.

Author Contributions

O.W.-D.—writing—original draft; W.M.—writing—original draft; B.B.—writing—review and editing. All authors have read and agreed to the published version of the manuscript.

Funding

This work is a part of research projects supported by (1) the Grant No NCN 2020/37/N/NZ7/01564 from the National Science Centre (Poland) and (2) intramural funding for young scientists of the Medical University of Lublin, Poland (Project No. PBmb 211).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

We would like to thank our team member, Agnieszka Michalak, for help in the preparation of the Figures, all of which were created with https://biorender.com/ (accessed on 10 January 2023).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. European Monitoring Centre for Drugs and Drug Addiction (EMCDDA). European Drug Report 2021: Trends and Developments; Publications Office of the European Union: Luxembourg, 2021. [Google Scholar]
  2. Cole, S.L.; Hofford, R.S.; Evert, D.J.; Wellman, P.J.; Eitan, S. Social influences on morphine conditioned place preference in adolescent mice. Addict. Biol. 2013, 18, 274–285. [Google Scholar] [CrossRef] [PubMed]
  3. Ramos, L.; Hicks, C.; Caminer, A.; Goodwin, J.; McGregor, I.S. Oxytocin and MDMA (‘Ecstasy’) enhance social reward in rats. Psychopharmacology 2015, 232, 2631–2641. [Google Scholar] [CrossRef]
  4. Thiel, K.J.; Sanabria, F.; Neisewander, J.L. Synergistic interaction between nicotine and social rewards in adolescent male rats. Psychopharmacology 2009, 204, 391–402. [Google Scholar] [CrossRef] [Green Version]
  5. Thiel, K.J.; Okun, A.C.; Neisewander, J.L. Social reward-conditioned place preference: A model revealing an interaction between cocaine and social context rewards in rats. Drug Alcohol Depend. 2008, 96, 202–212. [Google Scholar] [CrossRef] [Green Version]
  6. Wronikowska, O.; Zykubek, M.; Kurach, Ł.; Michalak, A.; Boguszewska-Czubara, A.; Budzyńska, B. Vulnerability factors for mephedrone-induced conditioned place preference in rats-the impact of sex differences, social-conditioning and stress. Psychopharmacology 2021, 238, 2947–2961. [Google Scholar] [CrossRef]
  7. Bahr, S.J.; Hoffmann, J.P.; Yang, X. Parental and peer influences on the risk of adolescent drug use. J. Prim. Prev. 2005, 26, 529–551. [Google Scholar] [CrossRef]
  8. Simons-Morton, B.; Chen, R.S. Over time relationships between early adolescent and peer substance use. Addict. Behav. 2006, 31, 1211–1223. [Google Scholar] [CrossRef]
  9. Andrews, J.A.; Tildesley, E.; Hops, H.; Li, F. The influence of peers on young adult substance use. Health Psychol. 2002, 21, 349–357. [Google Scholar] [CrossRef] [PubMed]
  10. Beloate, L.N.; Coolen, L.M. Influences of social reward experience on behavioral responses to drugs of abuse: Review of shared and divergent neural plasticity mechanisms for sexual reward and drugs of abuse. Neurosci. Biobehav. Rev. 2017, 83, 356–372. [Google Scholar] [CrossRef]
  11. Hosseinbor, M.; Yassini Ardekani, S.M.; Bakhshani, S.; Bakhshani, S. Emotional and social loneliness in individuals with and without substance dependence disorder. Int. J. High Risk Behav. Addict. 2014, 3, e22688. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Zeinali, A.; Sharifi, H.; Enayati, M.; Asgari, P.; Pasha, G. The mediational pathway among parenting styles, attachment styles and self-regulation with addiction susceptibility of adolescents. J. Res. Med. Sci. 2011, 16, 1105–1121. [Google Scholar] [PubMed]
  13. Bachner-Melman, R.; Ebstein, R.P. The role of oxytocin and vasopressin in emotional and social behaviors. Handb. Clin. Neurol. 2014, 124, 53–68. [Google Scholar] [PubMed]
  14. Bates, M.L.S.; Hofford, R.S.; Emery, M.A.; Wellman, P.J.; Eitan, S. The role of the vasopressin system and dopamine D1 receptors in the effects of social housing condition on morphine reward. Drug Alcohol Depend. 2018, 188, 113–118. [Google Scholar] [CrossRef] [PubMed]
  15. Insel, T.R. The challenge of translation in social neuroscience: A review of oxytocin, vasopressin, and affiliative behavior. Neuron 2010, 65, 768–779. [Google Scholar] [CrossRef] [Green Version]
  16. Landgraf, R.; Neumann, I.D. Vasopressin and oxytocin release within the brain: A dynamic concept of multiple and variable modes of neuropeptide communication. Front. Neuroendocrinol. 2004, 25, 150–176. [Google Scholar] [CrossRef]
  17. Lozić, M.; Šarenac, O.; Murphy, D.; Japundžić-Žigon, N. Vasopressin, Central Autonomic Control and Blood Pressure Regulation. Curr. Hypertens. Rep. 2018, 20, 11. [Google Scholar] [CrossRef]
  18. Love, T.M. Oxytocin, motivation and the role of dopamine. Pharmacol. Biochem. Behav. 2014, 119, 49–60. [Google Scholar] [CrossRef] [Green Version]
  19. Meyer-Lindenberg, A.; Domes, G.; Kirsch, P.; Heinrichs, M. Oxytocin and vasopressin in the human brain: Social neuropeptides for translational medicine. Nat. Rev. Neurosci. 2011, 12, 524–538. [Google Scholar] [CrossRef]
  20. Ross, H.E.; Young, L.J. Oxytocin and the neural mechanisms regulating social cognition and affiliative behavior. Front. Neuroendocrinol. 2009, 30, 534–547. [Google Scholar] [CrossRef] [Green Version]
  21. Althammer, F.; Grinevich, V. Diversity of oxytocin neurons: Beyond magno- and parvocellular cell types? J. Neuroendocrinol. 2017, 30, e12549. [Google Scholar] [CrossRef]
  22. Grinevich, V.; Ludwig, M. The multiple faces of the oxytocin and vasopressin systems in the brain. J. Neuroendocrinol. 2021, 33, e13004. [Google Scholar] [CrossRef] [PubMed]
  23. Knobloch, H.S.; Charlet, A.; Hoffmann, L.C.; Eliava, M.; Khrulev, S.; Cetin, A.H.; Osten, P.; Schwarz, M.K.; Seeburg, P.H.; Stoop, R.; et al. Evoked axonal oxytocin release in the central amygdala attenuates fear response. Neuron 2012, 73, 553–566. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Mitre, M.; Marlin, B.J.; Schiavo, J.K.; Morina, E.; Norden, S.E.; Hackett, T.A.; Aoki, C.J.; Chao, M.V.; Froemke, R.C. A distributed network for social cognition enriched for oxytocin receptors. J. Neurosci. 2016, 36, 2517–2535. [Google Scholar] [CrossRef] [Green Version]
  25. Zhang, B.; Qiu, L.; Xiao, W.; Ni, H.; Chen, L.; Wang, F.; Mai, W.; Wu, J.; Bao, A.; Hu, H.; et al. Reconstruction of the hypothalamoneurohypophysial system and functional dissection of magnocellular oxytocin neurons in the brain. Neuron 2021, 109, 331–346.e7. [Google Scholar] [CrossRef] [PubMed]
  26. Duque-Wilckens, N.; Torres, L.Y.; Yokoyama, S.; Minie, V.A.; Tran, A.M.; Petkova, S.P.; Hao, R.; Ramos-Maciel, S.; Rios, R.A.; Jackson, K.; et al. Extrahypothalamic oxytocin neurons drive stress-induced social vigilance and avoidance. Proc. Natl. Acad. Sci. USA 2020, 117, 26406–26413. [Google Scholar] [CrossRef]
  27. De Vries, G.J.; Buijs, R.M.; Van Leeuwen, F.W. Sex differences in vasopressin and other neurotransmitter systems in the brain. Prog. Brain Res. 1984, 61, 185–203. [Google Scholar]
  28. Zhang, L.; Hernandez, V.S.; Zetter, M.A.; Eiden, L.E. VGLUT-VGAT expression delineates functionally specialised populations of vasopressin-containing neurones including a glutamatergic perforant path-projecting cell group to the hippocampus in rat and mouse brain. J. Neuroendocrinol. 2020, 32, e12831. [Google Scholar] [CrossRef]
  29. Baribeau, D.A.; Anagnostou, E. Oxytocin and vasopressin: Linking pituitary neuropeptides and their receptors to social neurocircuits. Front. Neurosci. 2015, 9, 335. [Google Scholar] [CrossRef] [Green Version]
  30. Kimura, T.; Tanizawa, O.; Mori, K.; Brownstein, M.J.; Okayama, H. Structure and expression of a human oxytocin receptor. Nature 1992, 356, 526–529. [Google Scholar] [CrossRef]
  31. Zingg, H.H.; Laporte, S.A. The oxytocin receptor. Trends Endocrinol. Metab. 2003, 14, 222–227. [Google Scholar] [CrossRef]
  32. De Keyzer, Y.; Auzan, C.; Lenne, F.; Beldjord, C.; Thibonnier, M.; Bertagna, X.; Clauser, E. Cloning and characterization of the human V3 pituitary vasopressin receptor. FEBS Lett. 1994, 356, 215–220. [Google Scholar] [CrossRef] [Green Version]
  33. Jurek, B.; Neumann, I.D. The Oxytocin Receptor: From Intracellular Signaling to Behavior. Physiol. Rev. 2018, 98, 1805–1908. [Google Scholar] [CrossRef] [PubMed]
  34. Thibonnier, M.; Coles, P.; Thibonnier, A.; Shoham, M. Molecular pharmacology and modeling of vasopressin receptors. Prog. Brain Res. 2002, 139, 179–196. [Google Scholar] [PubMed]
  35. Loup, F.; Tribollet, E.; Dubois-Dauphin, M.; Dreifuss, J.J. Localization of high-affinity binding sites for oxytocin and vasopressin in the human brain. An autoradiographic study. Brain Res. 1991, 555, 220–232. [Google Scholar] [CrossRef] [PubMed]
  36. Holmes, C.L.; Landry, D.W.; Granton, J.T. Science review: Vasopressin and the cardiovascular system part 1—Receptor physiology. Crit. Care 2003, 7, 427–434. [Google Scholar] [CrossRef] [Green Version]
  37. Caldwell, H.K. Oxytocin and Vasopressin: Powerful Regulators of Social Behavior. Neuroscientist 2017, 23, 517–528. [Google Scholar] [CrossRef]
  38. Baracz, S.J.; Rourke, P.I.; Pardey, M.C.; Hunt, G.E.; McGregor, I.S.; Cornish, J.L. Oxytocin directly administered into the nucleus accumbens core or subthalamic nucleus attenuates methamphetamine-induced conditioned place preference. Behav. Brain Res. 2012, 228, 185–193. [Google Scholar] [CrossRef] [PubMed]
  39. Qi, J.; Yang, J.Y.; Wang, F.; Zhao, Y.N.; Song, M.; Wu, C.F. Effects of oxytocin on methamphetamine-induced conditioned place preference and the possible role of glutamatergic neurotransmission in the medial prefrontal cortex of mice in reinstatement. Neuropharmacology 2009, 56, 856–865. [Google Scholar] [CrossRef] [PubMed]
  40. Moaddab, M.; Hyland, B.I.; Brown, C.H. Oxytocin enhances the expression of morphine-induced conditioned place preference in rats. Psychoneuroendocrinology 2015, 53, 159–169. [Google Scholar] [CrossRef]
  41. Leong, K.C.; Zhou, L.; Ghee, S.M.; See, R.E.; Reichel, C.M. Oxytocin decreases cocaine taking, cocaine seeking, and locomotor activity in female rats. Exp. Clin. Psychopharmacol. 2016, 24, 55–64. [Google Scholar] [CrossRef]
  42. Zhou, L.; Sun, W.L.; Young, A.B.; Lee, K.; McGinty, J.F.; See, R.E. Oxytocin reduces cocaine seeking and reverses chronic cocaine-induced changes in glutamate receptor function. Int. J. Neuropsychopharmacol. 2014, 18, pyu009. [Google Scholar] [CrossRef] [Green Version]
  43. Carson, D.S.; Cornish, J.L.; Guastella, A.J.; Hunt, G.E.; McGregor, I.S. Oxytocin decreases methamphetamine self-administration, methamphetamine hyperactivity, and relapse to methamphetamine-seeking behaviour in rats. Neuropharmacology 2010, 58, 38–43. [Google Scholar] [CrossRef] [PubMed]
  44. Hicks, C.; Cornish, J.L.; Baracz, S.J.; Suraev, A.; McGregor, I.S. Adolescent pre-treatment with oxytocin protects against adult methamphetamine-seeking behavior in female rats. Addict. Biol. 2016, 21, 304–315. [Google Scholar] [CrossRef]
  45. Everett, N.A.; Turner, A.J.; Costa, P.A.; Baracz, S.J.; Cornish, J.L. The vagus nerve mediates the suppressing effects of peripherally administered oxytocin on methamphetamine self-administration and seeking in rats. Neuropsychopharmacology 2021, 46, 297–304. [Google Scholar] [CrossRef] [PubMed]
  46. King, C.E.; Griffin, W.C.; Luderman, L.N.; Kates, M.M.; McGinty, J.F.; Becker, H.C. Oxytocin Reduces Ethanol Self-Administration in Mice. Alcohol. Clin. Exp. Res. 2017, 41, 955–964. [Google Scholar] [CrossRef] [Green Version]
  47. King, C.E.; Griffin, W.C.; Lopez, M.F.; Becker, H.C. Activation of hypothalamic oxytocin neurons reduces binge-like alcohol drinking through signaling at central oxytocin receptors. Neuropsychopharmacology 2021, 46, 1950–1957. [Google Scholar] [CrossRef]
  48. MacFadyen, K.; Loveless, R.; DeLucca, B.; Wardley, K.; Deogan, S.; Thomas, C.; Peris, J. Peripheral oxytocin administration reduces ethanol consumption in rats. Pharm. Biochem. Behav. 2016, 140, 27–32. [Google Scholar] [CrossRef] [Green Version]
  49. Peters, S.; Slattery, D.A.; Flor, P.J.; Neumann, I.D.; Reber, S.O. Differential effects of baclofen and oxytocin on the increased ethanol consumption following chronic psychosocial stress in mice. Addict. Biol. 2013, 18, 66–67. [Google Scholar] [CrossRef] [PubMed]
  50. Peters, S.T.; Bowen, M.T.; Bohrer, K.; McGregor, I.S.; Neumann, I.D. Oxytocin inhibits ethanol consumption and ethanol-induced dopamine release in the nucleus accumbens. Addict. Biol. 2017, 22, 702–711. [Google Scholar] [CrossRef]
  51. Caruso, M.A.; Robins, M.T.; Fulenwider, H.D.; Ryabinin, A.E. Temporal analysis of individual ethanol consumption in socially housed mice and the effects of oxytocin. Psychopharmacology 2021, 238, 899–911. [Google Scholar] [CrossRef]
  52. Tunstall, B.J.; Kirson, D.; Zallar, L.J.; McConnell, S.A.; Vendruscolo, J.C.M.; Ho, C.P.; Oleata, C.S.; Khom, S.; Manning, M.; Lee, M.R.; et al. Oxytocin blocks enhanced motivation for alcohol in alcohol dependence and blocks alcohol effects on GABAergic transmission in the central amygdala. PLoS Biol. 2019, 17, e2006421. [Google Scholar] [CrossRef] [PubMed]
  53. Reguilón, M.D.; Ferrer-Pérez, C.; Miñarro, J.; Rodríguez-Arias, M. Oxytocin reverses ethanol consumption and neuroinflammation induced by social defeat in male mice. Horm. Behav. 2021, 127, 104875. [Google Scholar] [CrossRef] [PubMed]
  54. Stevenson, J.R.; Wenner, S.M.; Freestone, D.M.; Romaine, C.C.; Parian, M.C.; Christian, S.M.; Bohidar, A.E.; Ndem, J.R.; Vogel, I.R.; O’Kane, C.M. Oxytocin reduces alcohol consumption in prairie voles. Physiol. Behav. 2017, 179, 411–421. [Google Scholar] [CrossRef] [PubMed]
  55. Leong, K.C.; Freeman, L.R.; Berini, C.R.; Ghee, S.M.; See, R.E.; Reichel, C.M. Oxytocin Reduces Cocaine Cued Fos Activation in a Regionally Specific Manner. Int. J. Neuropsychopharmacol. 2017, 20, 844–854. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Weber, R.A.; Logan, C.N.; Leong, K.C.; Peris, J.; Knackstedt, L.; Reichel, C.M. Regionally Specific Effects of Oxytocin on Reinstatement of Cocaine Seeking in Male and Female Rats. Int. J. Neuropsychopharmacol. 2018, 21, 677–686. [Google Scholar] [CrossRef] [PubMed]
  57. Zhou, Y.; Rubinstein, M.; Low, M.J.; Kreek, M.J. V1b Receptor Antagonist SSR149415 and Naltrexone Synergistically Decrease Excessive Alcohol Drinking in Male and Female Mice. Alcohol. Clin. Exp. Res. 2018, 42, 195–205. [Google Scholar] [CrossRef]
  58. Kohtz, A.S.; Lin, B.; Smith, M.E.; Aston-Jones, G. Attenuated cocaine-seeking after oxytocin administration in male and female rats. Psychopharmacology 2018, 235, 2051–2063. [Google Scholar] [CrossRef]
  59. Ferrer-Pérez, C.; Castro-Zavala, A.; Luján, M.Á.; Filarowska, J.; Ballestín, R.; Miñarro, J.; Valverde, O.; Rodríguez-Arias, M. Oxytocin prevents the increase of cocaine-related responses produced by social defeat. Neuropharmacology 2019, 146, 50–64. [Google Scholar] [CrossRef]
  60. Morales-Rivera, A.; Hernández-Burgos, M.M.; Martínez-Rivera, A.; Pérez-Colón, J.; Rivera, R.; Montalvo, J.; Rodríguez-Borrero, E.; Maldonado-Vlaar, C.S. Anxiolytic effects of oxytocin in cue-induced cocaine seeking behavior in rats. Psychopharmacology 2014, 231, 4145–4155. [Google Scholar] [CrossRef]
  61. Baracz, S.J.; Everett, N.A.; Cornish, J.L. The Involvement of Oxytocin in the Subthalamic Nucleus on Relapse to Methamphetamine-Seeking Behaviour. PLoS ONE 2015, 10, e0136132. [Google Scholar] [CrossRef] [Green Version]
  62. Baracz, S.J.; Everett, N.A.; McGregor, I.S.; Cornish, J.L. Oxytocin in the nucleus accumbens core reduces reinstatement of methamphetamine-seeking behaviour in rats. Addict. Biol. 2016, 21, 316–325. [Google Scholar] [CrossRef] [PubMed]
  63. Bernheim, A.; Leong, K.C.; Berini, C.; Reichel, C.M. Antagonism of mGlu2/3 receptors in the nucleus accumbens prevents oxytocin from reducing cued methamphetamine seeking in male and female rats. Pharmacol. Biochem. Behav. 2017, 161, 13–21. [Google Scholar] [CrossRef] [PubMed]
  64. Cai, J.; Che, X.; Xu, T.; Luo, Y.; Yin, M.; Lu, X.; Wu, C.; Yang, J. Repeated oxytocin treatment during abstinence inhibited context- or restraint stress-induced reinstatement of methamphetamine-conditioned place preference and promoted adult hippocampal neurogenesis in mice. Exp. Neurol. 2022, 347, 113907. [Google Scholar] [CrossRef]
  65. Cox, B.M.; Young, A.B.; See, R.E.; Reichel, C.M. Sex differences in methamphetamine seeking in rats: Impact of oxytocin. Psychoneuroendocrinology 2013, 38, 2343–2353. [Google Scholar] [CrossRef] [Green Version]
  66. Cox, B.M.; Bentzley, B.S.; Regen-Tuero, H.; See, R.E.; Reichel, C.M.; Aston-Jones, G. Oxytocin Acts in Nucleus Accumbens to Attenuate Methamphetamine Seeking and Demand. Biol. Psychiatry 2017, 81, 949–958. [Google Scholar] [CrossRef] [PubMed]
  67. Everett, N.A.; McGregor, I.S.; Baracz, S.J.; Cornish, J.L. The role of the vasopressin V1A receptor in oxytocin modulation of methamphetamine primed reinstatement. Neuropharmacology 2018, 133, 1–11. [Google Scholar] [CrossRef]
  68. Everett, N.; Baracz, S.; Cornish, J. Oxytocin treatment in the prelimbic cortex reduces relapse to methamphetamine-seeking and is associated with reduced activity in the rostral nucleus accumbens core. Pharmacol. Biochem. Behav. 2019, 183, 64–71. [Google Scholar] [CrossRef]
  69. Everett, N.A.; Carey, H.A.; Cornish, J.L.; Baracz, S.J. Sign tracking predicts cue-induced but not drug-primed reinstatement to methamphetamine seeking in rats: Effects of oxytocin treatment. J. Psychopharmacol. 2020, 34, 1271–1279. [Google Scholar] [CrossRef]
  70. Everett, N.A.; Baracz, S.J.; Cornish, J.L. The effect of chronic oxytocin treatment during abstinence from methamphetamine self-administration on incubation of craving, reinstatement, and anxiety. Neuropsychopharmacology 2020, 45, 597–605. [Google Scholar] [CrossRef]
  71. Ferland, C.L.; Reichel, C.M.; McGinty, J.F. Effects of oxytocin on methamphetamine-seeking exacerbated by predator odor pre-exposure in rats. Psychopharmacology 2016, 233, 1015–1024. [Google Scholar] [CrossRef] [Green Version]
  72. Han, W.Y.; Du, P.; Fu, S.Y.; Wang, F.; Song, M.; Wu, C.F.; Yang, J.Y. Oxytocin via its receptor affects restraint stress-induced methamphetamine CPP reinstatement in mice: Involvement of the medial prefrontal cortex and dorsal hippocampus glutamatergic system. Pharm. Biochem. Behav. 2014, 119, 80–87. [Google Scholar] [CrossRef]
  73. Ballas, H.S.; Wilfur, S.M.; Freker, N.A.; Leong, K.C. Oxytocin Attenuates the Stress-Induced Reinstatement of Alcohol-Seeking in Male Rats: Role of the Central Amygdala. Biomedicines 2021, 9, 1919. [Google Scholar] [CrossRef]
  74. King, C.E.; Becker, H.C. Oxytocin attenuates stress-induced reinstatement of alcohol seeking behavior in male and female mice. Psychopharmacology 2019, 236, 2613–2622. [Google Scholar] [CrossRef]
  75. Zanos, P.; Georgiou, P.; Wright, S.R.; Hourani, S.M.; Kitchen, I.; Winsky-Sommerer, R.; Bailey, A. The oxytocin analogue carbetocin prevents emotional impairment and stress-induced reinstatement of opioid-seeking in morphine-abstinent mice. Neuropsychopharmacology 2014, 39, 855–865. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Georgiou, P.; Zanos, P.; Garcia-Carmona, J.A.; Hourani, S.; Kitchen, I.; Kieffer, B.L.; Laorden, M.L.; Bailey, A. The oxytocin analogue carbetocin prevents priming-induced reinstatement of morphine-seeking: Involvement of dopaminergic, noradrenergic and MOPr systems. Eur. Neuropsychopharmacol. 2015, 25, 2459–2464. [Google Scholar] [CrossRef] [PubMed]
  77. Bahi, A. The oxytocin receptor impairs ethanol reward in mice. Physiol. Behav. 2015, 139, 321–327. [Google Scholar] [CrossRef]
  78. Lee, H.; Jang, M.; Noh, J. Oxytocin attenuates aversive response to nicotine and anxiety-like behavior in adolescent rats. Neurosci. Res. 2017, 115, 29–36. [Google Scholar] [CrossRef]
  79. Qi, J.; Yang, J.Y.; Song, M.; Li, Y.; Wang, F.; Wu, C.F. Inhibition by oxytocin of methamphetamine-induced hyperactivity related to dopamine turnover in the mesolimbic region in mice. Naunyn. Schmiedebergs Arch. Pharmacol. 2008, 376, 441–448. [Google Scholar] [CrossRef]
  80. Ramos, L.; Hicks, C.; Kevin, R.; Caminer, A.; Narlawar, R.; Kassiou, M.; McGregor, I.S. Acute prosocial effects of oxytocin and vasopressin when given alone or in combination with 3,4-methylenedioxymethamphetamine in rats: Involvement of the V1A receptor. Neuropsychopharmacology 2013, 38, 2249–2259. [Google Scholar] [CrossRef]
  81. Westenbroek, C.; Perry, A.N.; Jagannathan, L.; Becker, J.B. Effect of social housing and oxytocin on the motivation to self-administer methamphetamine in female rats. Physiol. Behav. 2019, 203, 10–17. [Google Scholar] [CrossRef] [PubMed]
  82. Carson, D.S.; Hunt, G.E.; Guastella, A.J.; Barber, L.; Cornish, J.L.; Arnold, J.C.; Boucher, A.A.; McGregor, I.S. Systemically administered oxytocin decreases methamphetamine activation of the subthalamic nucleus and accumbens core and stimulates oxytocinergic neurons in the hypothalamus. Addict. Biol. 2010, 15, 448–463. [Google Scholar] [CrossRef] [PubMed]
  83. Fan, X.Y.; Yang, J.Y.; Dong, Y.X.; Hou, Y.; Liu, S.; Wu, C.F. Oxytocin inhibits methamphetamine-associated learning and memory alterations by regulating DNA methylation at the Synaptophysin promoter. Addict. Biol. 2020, 25, e12697. [Google Scholar] [CrossRef]
  84. Manbeck, K.E.; Shelley, D.; Schmidt, C.E.; Harris, A.C. Effects of oxytocin on nicotine withdrawal in rats. Pharmacol. Biochem. Behav. 2014, 116, 84–89. [Google Scholar] [CrossRef]
  85. Salighedar, R.; Erfanparast, A.; Tamaddonfard, E.; Soltanalinejad, F. Medial prefrontal cortex oxytocin-opioid receptors interaction in spatial memory processing in rats. Physiol. Behav. 2019, 209, 112599. [Google Scholar] [CrossRef]
  86. Bowen, M.T.; Peters, S.T.; Absalom, N.; Chebib, M.; Neumann, I.D.; McGregor, I.S. Oxytocin prevents ethanol actions at δ subunit-containing GABAA receptors and attenuates ethanol-induced motor impairment in rats. Proc. Natl. Acad. Sci. USA 2015, 112, 3104–3109. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Dannenhoffer, C.A.; Kim, E.U.; Saalfield, J.; Werner, D.F.; Varlinskaya, E.I.; Spear, L.P. Oxytocin and vasopressin modulation of social anxiety following adolescent intermittent ethanol exposure. Psychopharmacology (Berl) 2018, 235, 3065–3077. [Google Scholar] [CrossRef]
  88. Broadbear, J.H.; Tunstall, B.; Beringer, K. 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. 2011, 16, 202–214. [Google Scholar] [CrossRef]
  89. Rae, M.; Zanos, P.; Georgiou, P.; Chivers, P.; Bailey, A.; Camarini, R. Environmental enrichment enhances conditioned place preference to ethanol via an oxytocinergic-dependent mechanism in male mice. Neuropharmacology 2018, 138, 267–274. [Google Scholar] [CrossRef] [PubMed]
  90. Kuteykin-Teplyakov, K.; Maldonado, R. Looking for prosocial genes: ITRAQ analysis of proteins involved in MDMA-induced sociability in mice. Eur. Neuropsychopharmacol. 2014, 24, 1773–1783. [Google Scholar] [CrossRef]
  91. Ferrer-Pérez, C.; Reguilón, M.D.; Miñarro, J.; Rodríguez-Arias, M. Endogenous oxytocin is essential for the buffering effects of pair housing against the increase in cocaine reward induced by social stress. Physiol. Behav. 2020, 221, 112913. [Google Scholar] [CrossRef]
  92. Thompson, M.R.; Callaghan, P.D.; Hunt, G.E.; Cornish, J.L.; McGregor, I.S. A role for oxytocin and 5-HT(1A) receptors in the prosocial effects of 3,4 methylenedioxymethamphetamine (“ecstasy”). Neuroscience 2007, 146, 509–514. [Google Scholar] [CrossRef] [PubMed]
  93. Gárate-Pérez, M.F.; Méndez, A.; Bahamondes, C.; Sanhueza, C.; Guzmán, F.; Reyes-Parada, M.; Sotomayor-Zárate, R.; Renard, G.M. Vasopressin in the lateral septum decreases conditioned place preference to amphetamine and nucleus accumbens dopamine release. Addict. Biol. 2021, 26, e12851. [Google Scholar] [CrossRef]
  94. Edwards, S.; Guerrero, M.; Ghoneim, O.M.; Roberts, E.; Koob, G.F. Evidence that vasopressin V1b receptors mediate the transition to excessive drinking in ethanol-dependent rats. Addict. Biol. 2012, 17, 76–85. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Zhou, Y.; Litvin, Y.; Piras, A.P.; Pfaff, D.W.; Kreek, M.J. Persistent increase in hypothalamic arginine vasopressin gene expression during protracted withdrawal from chronic escalating-dose cocaine in rodents. Neuropsychopharmacology 2011, 36, 2062–2075. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Zhou, Y.; Leri, F.; Cummins, E.; Hoeschele, M.; Kreek, M.J. Involvement of arginine vasopressin and V1b receptor in heroin withdrawal and heroin seeking precipitated by stress and by heroin. Neuropsychopharmacology 2008, 33, 226–236. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Qi, X.; Guzhva, L.; Ji, Y.; Bruijnzeel, A.W. Chronic treatment with the vasopressin 1b receptor antagonist SSR149415 prevents the dysphoria associated with nicotine withdrawal in rats. Behav. Brain Res. 2015, 292, 259–265. [Google Scholar] [CrossRef] [Green Version]
  98. Ponzoni, L.; Braida, D.; Bondiolotti, G.; Sala, M. The Non-Peptide Arginine-Vasopressin v1a Selective Receptor Antagonist, SR49059, Blocks the Rewarding, Prosocial, and Anxiolytic Effects of 3,4-Methylenedioxymethamphetamine and Its Derivatives in Zebrafish. Front. Psychiatry 2017, 8, 146. [Google Scholar] [CrossRef] [Green Version]
  99. Harper, K.M.; Knapp, D.J.; Butler, R.K.; Cook, C.A.; Criswell, H.E.; Stuber, G.D.; Breese, G.R. Amygdala Arginine Vasopressin Modulates Chronic Ethanol Withdrawal Anxiety-Like Behavior in the Social Interaction Task. Alcohol. Clin. Exp. Res. 2019, 43, 2134–2143. [Google Scholar] [CrossRef]
  100. McBride, S.M.; Flynn, F.W. Centrally administered vasopressin cross-sensitizes rats to amphetamine and drinking hypertonic NaCl. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2007, 293, R1452-8. [Google Scholar] [CrossRef]
  101. Goutier, W.; Kloeze, M.; McCreary, A.C. Nicotine-induced locomotor sensitization: Pharmacological analyses with candidate smoking cessation aids. Addict. Biol. 2016, 21, 234–241. [Google Scholar] [CrossRef]
  102. Zhou, Y.; Colombo, G.; Carai, M.A.; Ho, A.; Gessa, G.L.; Kreek, M.J. Involvement of arginine vasopressin and V1b receptor in alcohol drinking in Sardinian alcohol-preferring rats. Alcohol. Clin. Exp. Res. 2011, 35, 1876–1883. [Google Scholar] [CrossRef]
  103. Bedi, G.; Hyman, D.; de Wit, H. Is ecstasy an “empathogen”? Effects of ±3,4-methylenedioxymethamphetamine on prosocial feelings and identification of emotional states in others. Biol. Psychiatry 2010, 68, 1134–1140. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Parrott, A.C. Human psychobiology of MDMA or ‘Ecstasy’: An overview of 25 years of empirical research. Hum. Psychopharmacol. 2013, 28, 289–307. [Google Scholar] [CrossRef]
  105. Pelloux, Y.; Giorla, E.; Montanari, C.; Baunez, C. Social modulation of drug use and drug addiction. Neuropharmacology 2019, 159, 107545. [Google Scholar] [CrossRef]
  106. Che, X.; Cai, J.; Liu, Y.; Xu, T.; Yang, J.; Wu, C. Oxytocin signaling in the treatment of drug addiction: Therapeutic opportunities and challenges. Pharmacol. Ther. 2021, 223, 107820. [Google Scholar] [CrossRef] [PubMed]
  107. Estes, M.K.; Freels, T.G.; Prater, W.T.; Lester, D.B. Systemic oxytocin administration alters mesolimbic dopamine release in mice. Neuroscience 2019, 408, 226–238. [Google Scholar] [CrossRef]
  108. Young, K.A.; Liu, Y.; Gobrogge, K.L.; Wang, H.; Wang, Z. Oxytocin reverses amphetamine-induced deficits in social bonding: Evidence for an interaction with nucleus accumbens dopamine. J. Neurosci. Off. J. Soc. Neurosci. 2014, 34, 8499–8506. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  109. Ferguson, J.N.; Aldag, J.M.; Insel, T.R.; Young, L.J. Oxytocin in the medial amygdala is essential for social recognition in the mouse. J. Neurosci. 2001, 21, 8278–8285. [Google Scholar] [CrossRef]
  110. Hysek, C.M.; Schmid, Y.; Simmler, L.D.; Domes, G.; Heinrichs, M.; Eisenegger, C.; Preller, K.H.; Quednow, B.B.; Liechti, M.E. MDMA enhances emotional empathy and prosocial behavior. Soc. Cogn. Affect. Neurosci. 2014, 9, 1645–1652. [Google Scholar] [CrossRef] [Green Version]
  111. Kirkpatrick, M.G.; Francis, S.M.; Lee, R.; de Wit, H.; Jacob, S. Plasma oxytocin concentrations following MDMA or intranasal oxytocin in humans. Psychoneuroendocrinology 2014, 46, 23–31. [Google Scholar] [CrossRef] [Green Version]
  112. Johns, J.M.; McMurray, M.S.; Joyner, P.W.; Jarrett, T.M.; Williams, S.K.; Cox, E.T.; Black, M.A.; Middleton, C.L.; Walker, C.H. Effects of chronic and intermittent cocaine treatment on dominance, aggression, and oxytocin levels in post-lactational rats. Psychopharmacology 2010, 211, 175–185. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. McMurray, M.S.; Joyner, P.W.; Middleton, C.W.; Jarrett, T.M.; Elliott, D.L.; Black, M.A.; Hofler, V.E.; Walker, C.H.; Johns, J.M. Intergenerational effects of cocaine on maternal aggressive behavior and brain oxytocin in rat dams. Stress 2008, 11, 398–410. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Knobloch, H.S.; Grinevich, V. Evolution of oxytocin pathways in the brain of vertebrates. Front. Behav. Neurosci. 2014, 8, 31. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Peris, J.; MacFadyen, K.; Smith, J.A.; de Kloet, A.D.; Wang, L.; Krause, E.G. Oxytocin receptors are expressed on dopamine and glutamate neurons in the mouse ventral tegmental area that project to nucleus accumbens and other mesolimbic targets. J. Comp. Neurol. 2017, 525, 1094–1108. [Google Scholar] [CrossRef]
  116. Li, J.; You, Z.; Chen, Z.; Song, C.; Lu, C. Chronic morphine treatment inhibits oxytocin release from the supraoptic nucleus slices of rats. Neurosci. Lett. 2001, 300, 54–58. [Google Scholar] [CrossRef]
  117. Bowen, M.T.; Neumann, I.D. Rebalancing the Addicted Brain: Oxytocin Interference with the Neural Substrates of Addiction. Trends Neurosci. 2017, 40, 691–708. [Google Scholar] [CrossRef]
  118. Light, K.C.; Grewen, K.M.; Amico, J.A.; Boccia, M.; Brownley, K.A.; Johns, J.M. Deficits in plasma oxytocin responses and increased negative affect, stress, and blood pressure in mothers with cocaine exposure during pregnancy. Addict. Behav. 2004, 29, 1541–1564. [Google Scholar] [CrossRef] [Green Version]
  119. Koob, G.F.; Volkow, N.D. Neurobiology of addiction: A neurocircuitry analysis. Lancet Psychiatry 2016, 3, 760–773. [Google Scholar] [CrossRef]
  120. Volkow, N.D.; Michaelides, M.; Baler, R. The Neuroscience of Drug Reward and Addiction. Physiol. Rev. 2019, 99, 2115–2140. [Google Scholar] [CrossRef]
  121. Baskerville, T.A.; Allard, J.; Wayman, C.; Douglas, A.J. Dopamine-oxytocin interactions in penile erection. Eur. J. Neurosci. 2009, 30, 2151–2164. [Google Scholar] [CrossRef]
  122. Baskerville, T.A.; Douglas, A.J. Dopamine and oxytocin interactions underlying behaviors: Potential contributions to behavioral disorders. CNS Neurosci. Ther. 2010, 16, e92–e123. [Google Scholar] [CrossRef]
  123. Borroto-Escuela, D.O.; Carlsson, J.; Ambrogini, P.; Narvaez, M.; Wydra, K.; Tarakanov, A.O.; Li, X.; Millón, C.; Ferraro, L.; Cuppini, R.; et al. Understanding the role of GPCR heteroreceptor complexes inmodulating the brain networks in health and disease. Front. Cell. Neurosci. 2017, 11, 37. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. de la Mora, M.P.; Pérez-Carrera, D.; Crespo-Ramírez, M.; Tarakanov, A.; Fuxe, K.; Borroto-Escuela, D.O. Signaling in dopamine D2 receptor-oxytocin receptor heterocomplexes and its relevance for the anxiolytic effects of dopamine and oxytocin interactions in the amygdala of the rat. Biochim. Biophys. Acta 2016, 1862, 2075–2085. [Google Scholar] [CrossRef]
  125. Romero-Fernandez, W.; Borroto-Escuela, D.O.; Agnati, L.F.; Fuxe, K. Evidence for the existence of dopamine D2-oxytocin receptor heteromers in the ventral and dorsal striatum with facilitatory receptor-receptor interactions. Mol. Psychiatry 2013, 18, 849–850. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Zou, Z.; Wang, H.; d’Oleire Uquillas, F.; Wang, X.; Ding, J.; Chen, H. Definition of Substance and Non-substance Addiction. Adv. Exp. Med. Biol. 2017, 1010, 21–41. [Google Scholar] [PubMed]
  127. Ahumada, C.; Bahamondes, C.; Cerda, C.A.; Silva, R.A.; Cruz, G.; Moya, P.R.; Sotomayor-Zárate, R.; Renard, G.M. Amphetamine treatment affects the extra-hypothalamic vasopressinergic system in a sex- and nucleus-dependent manner. J. Neuroendocrinol. 2017, 29. [Google Scholar] [CrossRef] [PubMed]
  128. Rodríguez-Borrero, E.; Rivera-Escalera, F.; Candelas, F.; Montalvo, J.; Muñoz-Miranda, W.J.; Walker, J.R.; Maldonado-Vlaar, C.S. Arginine vasopressin gene expression changes within the nucleus accumbens during environment elicited cocaine-conditioned response in rats. Neuropharmacology 2010, 58, 88–101. [Google Scholar] [CrossRef] [Green Version]
  129. Dölen, G.; Darvishzadeh, A.; Huang, K.W.; Malenka, R.C. Social reward requires coordinated activity of nucleus accumbens oxytocin and serotonin. Nature 2013, 501, 179–184. [Google Scholar] [CrossRef] [Green Version]
  130. Costa, G.; Gołembiowska, K. Neurotoxicity of MDMA: Main effects and mechanisms. Exp. Neurol. 2022, 347, 113894. [Google Scholar] [CrossRef]
  131. Yoshida, M.; Takayanagi, Y.; Inoue, K.; Kimura, T.; Young, L.J.; Onaka, T.; Nishimori, K. Evidence that oxytocin exerts anxiolytic effects via oxytocin receptor expressed in serotonergic neurons in mice. J. Neurosci. 2009, 29, 2259–2271. [Google Scholar] [CrossRef] [Green Version]
  132. Grieb, Z.A.; Lonstein, J.S. Oxytocin interactions with central dopamine and serotonin systems regulate different components of motherhood. Phil. Trans. R. Soc. B 2022, 377. [Google Scholar] [CrossRef] [PubMed]
  133. Dölen, G. Autism: Oxytocin, serotonin, and social reward. Soc. Neurosci. 2015, 10, 450–465. [Google Scholar] [CrossRef] [PubMed]
  134. Zhao, F.; Zhang, H.; Wang, P.; Cui, W.; Xu, K.; Chen, D.; Hu, M.; Li, Z.; Geng, X.; Wei, S. Oxytocin and serotonin in the modulation of neural function: Neurobiological underpinnings of autism-related behavior. Front. Neurosci. 2022, 16, 919890. [Google Scholar] [CrossRef]
  135. Nagano, M.; Takumi, T.; Suzuki, H. Critical roles of serotonin-oxytocin interaction during the neonatal period in social behavior in 15q dup mice with autistic traits. Sci. Rep. 2018, 8, 13675. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Quintana, D.S.; Lischke, A.; Grace, S.; Scheele, D.; Ma, Y.; Becker, B. Advances in the field of intranasal oxytocin research: Lessons learned and future directions for clinical research. Mol. Psychiatry 2021, 26, 80–91. [Google Scholar] [CrossRef]
  137. Moeini, M.; Omidi, A.; Sehat, M.; Banafshe, H.R. The effects of oxytocin on withdrawal, craving and stress response in heroin-dependent patients: A randomized, double-blind clinical trial. Eur. Addict. Res. 2019, 25, 41–47. [Google Scholar] [CrossRef]
  138. Mitchell, J.M.; Arcuni, P.A.; Weinstein, D.; Woolley, J.D. Intranasal Oxytocin Selectively Modulates Social Perception, Craving, and Approach Behavior in Subjects With Alcohol Use Disorder. J. Addict. Med. 2016, 10, 182–189. [Google Scholar] [CrossRef]
  139. Pedersen, C.A.; Smedley, K.L.; Leserman, J.; Jarskog, L.F.; Rau, S.W.; Kampov-Polevoi, A.; Garbutt, J.C. Intranasal oxytocin blocks alcohol withdrawal in human subjects. Alcohol. Clin. Exp. Res. 2013, 37, 484–489. [Google Scholar] [CrossRef] [Green Version]
  140. Stauffer, C.S.; Musinipally, V.; Suen, A.; Lynch, K.L.; Shapiro, B.; Woolley, J.D. A two-week pilot study of intranasal oxytocin for cocaine-dependent individuals receiving methadone maintenance treatment for opioid use disorder. Addict. Res. Theory 2016, 24, 490–498. [Google Scholar] [CrossRef] [Green Version]
  141. Van Hedger, K.; Kushner, M.J.; Lee, R.; de Wit, H. Oxytocin reduces cigarette consumption in daily smokers. Nicotine Tob. Res. 2019, 21, 799–804. [Google Scholar] [CrossRef]
  142. McRae-Clark, A.L.; Baker, N.L.; Maria, M.M.; Brady, K.T. Effect of oxytocin on craving and stress response in marijuana-dependent individuals: A pilot study. Psychopharmacology 2013, 228, 623–631. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Melby, K.; Grawe, R.W.; Aamo, T.O.; Salvesen, O.; Spigset, O. Effect of intranasal oxytocin on alcohol withdrawal syndrome: A randomized placebo-controlled double-blind clinical trial. Drug Alcohol Depend. 2019, 197, 95–101. [Google Scholar] [CrossRef] [PubMed]
  144. Lee, M.R.; Glassman, M.; King-Casas, B.; Kelly, D.L.; Stein, E.A.; Schroeder, J.; Salmeron, B.J. Complexity of oxytocins effects in a chronic cocaine dependent population. Eur. Neuropsychopharmacol. 2014, 24, 1483–1491. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. The structure and amino acids sequence of (A) OT and (B) AVP. The lines indicate cysteine residues that form a sulfur bridge by disulfide bonds. OT and AVP structures differ in the first and eighth place in the aminoacid sequence (dark green). This figure was created with https://biorender.com/ (accessed on 10 January 2023).
Figure 1. The structure and amino acids sequence of (A) OT and (B) AVP. The lines indicate cysteine residues that form a sulfur bridge by disulfide bonds. OT and AVP structures differ in the first and eighth place in the aminoacid sequence (dark green). This figure was created with https://biorender.com/ (accessed on 10 January 2023).
Biomolecules 13 00405 g001
Figure 2. Including criteria for the selection of studies chosen for the Table 1, Table 2, Table 3 and Table 4. This figure was created with https://biorender.com/ (accessed on 10 January 2023).
Figure 2. Including criteria for the selection of studies chosen for the Table 1, Table 2, Table 3 and Table 4. This figure was created with https://biorender.com/ (accessed on 10 January 2023).
Biomolecules 13 00405 g002
Figure 3. The summary of available data on OT/AVP effects on drug reward and social effects. Detailed descriptions are presented in Section 3.1, Section 3.2 and Section 3.3. This figure was created with https://biorender.com/ (accessed on 10 January 2023).
Figure 3. The summary of available data on OT/AVP effects on drug reward and social effects. Detailed descriptions are presented in Section 3.1, Section 3.2 and Section 3.3. This figure was created with https://biorender.com/ (accessed on 10 January 2023).
Biomolecules 13 00405 g003
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Wronikowska-Denysiuk, O.; Mrozek, W.; Budzyńska, B. The Role of Oxytocin and Vasopressin in Drug-Induced Reward—Implications for Social and Non-Social Factors. Biomolecules 2023, 13, 405. https://doi.org/10.3390/biom13030405

AMA Style

Wronikowska-Denysiuk O, Mrozek W, Budzyńska B. The Role of Oxytocin and Vasopressin in Drug-Induced Reward—Implications for Social and Non-Social Factors. Biomolecules. 2023; 13(3):405. https://doi.org/10.3390/biom13030405

Chicago/Turabian Style

Wronikowska-Denysiuk, Olga, Weronika Mrozek, and Barbara Budzyńska. 2023. "The Role of Oxytocin and Vasopressin in Drug-Induced Reward—Implications for Social and Non-Social Factors" Biomolecules 13, no. 3: 405. https://doi.org/10.3390/biom13030405

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop