Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2017 Dec 1.
Published in final edited form as: Behav Pharmacol. 2016 Dec;27(8):640–648. doi: 10.1097/FBP.0000000000000258

Oxytocin for the treatment of drug and alcohol use disorders

Mary R Lee 1, Elise M Weerts 2,*
PMCID: PMC5089904  NIHMSID: NIHMS811517  PMID: 27603752

Abstract

There is growing interest in the use of oxytocin (OT) as a potential treatment for alcohol and other substance use disorders. OT is a neuropeptide that modulates adaptive processes associated with addiction including reward, tolerance, associative learning, memory, and stress responses. OT exerts its effects via interactions with the hypothalamic–pituitary–adrenal (HPA) axis, and multiple neurotransmitter systems including the dopamine mesolimbic reward and corticotrophin-releasing factor stress systems. Oxytocin effects on stress systems are of high interest given the strong link between stress, drug use and relapse, and known dysregulation of HPA-axis activity associated with substance use disorders. At the same time, the oxytocin system is itself altered by acute or chronic drug exposure. This review summarizes the preclinical and clinical literature on the oxytocin system, and its relevance to drug and alcohol addiction. In addition, findings from recent clinical trials conducted in participants with cocaine, cannabis or alcohol use disorder are included and evidence that oxytocin may help to normalize blunted stress responses, and attenuate withdrawal associated hypercortisolism, negative mood and withdrawal symptoms are summarized.

Keywords: Oxytocin, addiction, dependence, substance use disorder, alcoholism, treatment

Introduction

Oxytocin (OT) is a 9 amino acid polypeptide hormone that acts via a specific receptor and is widely distributed in the central nervous system (CNS) and peripheral tissues (Gimpl and Fahrenholz 2001). OT is involved in the regulation and release of adenohypophyseal hormones including prolactin, adrenocorticotropin (ACTH), gonadotropins, and corticotrophin-releasing factor (CRF). Initially, OT was thought to be primarily involved in sexual behaviors, female parturition and lactation. Subsequent research has determined that OT is also involved in emotional regulation, pain and stress, and modulates response to rewarding behaviors promoted by food, sex and drugs (Meyer-Lindenberg et al. 2011; Onaka et al. 2012). The co-modulation of both stress and motivational processes is believed to be due to the important role of OT to shift salience to social, affiliative processes, both by increasing the salience itself of rewarding stimuli and/or by reducing stress, allowing for attention to social bonding (Baskerville and Douglas 2010). This is obviously relevant to addiction, where salience of drug stimuli overshadows motivation for social affiliation, and where stress may trigger drug seeking and relapse (Sinha 2008). In the current review, we will focus on the role of the oxytocin system in drug and alcohol addiction and highlight key findings to date on the use of intranasal OT to treat substance use disorders.

Oxytocin and stress

The influence of OT to dampen stress responses is important. Neuroendocrine pathways that modulate the response to stress include three interconnecting circuits, the HPA axis, the adrenomedullary system, and the extra-hypothalamic CRF system. The HPA axis releases CRF from paraventricular neurons within the hypothalamus, stimulating the synthesis and release of adrenocorticotropin (ACTH) by the anterior pituitary, which in turn stimulates the synthesis and release of corticosteroids (CORT) (cortisol in human and nonhuman primates and corticosterone in rodents) via the adrenal cortex. The sympathetic adrenomedullary system, which releases norepinephrine and epinephrine, and CRF expression in the extra-hypothalamic brain regions including limbic regions, are key substrates involved in anxiety and other stress-related behaviors. Stress, defined as any stimulus that disrupts physiological homeostasis, triggers a cascade of adaptive responses involving any or all of these pathways to return the organism to homeostasis.

There is strong evidence from the preclinical literature that stress exposure is an important contributor to relapse. In rats and monkeys, acute stress enhances alcohol preference and reward (Funk et al. 2004), and increased alcohol intake is correlated with stress-induced increases in CORT levels (Fahlke et al. 2000; Fish et al. 2008). In addition, following repeated social stress exposure (e.g., defeat, low social rank, and maternal separation), rats and monkeys subsequently show greater alcohol intake when compared to non-stressed cohorts (Fahlke et al. 2000; Cruz et al. 2008; Fish et al. 2008). Current theories suggest that CORT release induced by stress augments drug reinforcement. Indeed, in rodents, CORT increases drug reward by increasing mesolimbic dopamine transmission (Piazza and Le Moal 1996), rats self-administer CORT itself at levels similar to those elicited by stress, and intracerebroventricular infusions of CORT enhance the reinforcing effects of alcohol (Fahlke et al. 1996).

Studies in laboratory animals have demonstrated that OT has marked anti-stress effects. When administered centrally, OT decreases stress-induced increases in CORT levels (Lang et al. 1983; Windle et al. 1997; Neumann et al. 2000) and reduces stress-induced behaviors in rodent models of anxiety and depression (Arletti and Bertolini 1987; Insel and Winslow 1991; Windle, et al. 1997; Neumann et al. 2000). At the same time, the endogenous OT system appears to be sensitive to stressors. In rats, exposure to acute stress increased OT levels in blood and in hypothalamic and extra-hypothalamic brain regions (Lang et al. 1983; Neumann et al. 1998; Ebner et al. 2000; Ondrejcakova et al. 2010) and increased OT mRNA levels (Jezova et al. 1995). Thus, OT appears to play a protective role in homeostatic regulation of stress responses, and OT administration may attenuate the effects of stress on drinking/drug use and relapse (Uhart and Wand 2009; Koob et al. 2014).

Investigations in human subjects are in line with the preclinical literature. When administered via the intranasal route, OT produces changes in measures of autonomic arousal and mood (MacDonald et al. 2011), increased positive communication during couples’ conflict discussions (Ditzen et al. 2009) and improved recognition and processing of positive facial expressions (Di Simplicio et al. 2009; Marsh et al. 2010; Lischke et al. 2012). The anti-stress effects of OT have been also been investigated using the Trier Social Stress Test, a well-validated laboratory procedure for induction of stress responses in human subjects (Foley and Kirschbaum 2010). This test, which includes components of public speaking component and oral mental arithmetic, produces a robust increase in CORT and self-reported psychological stress and these effects are attenuated by OT (Heinrichs et al. 2003; Quirin et al. 2011; Simeon et al. 2011; de Oliveira et al. 2012; Kubzansky et al. 2012). Consistent with an OT anti-stress hypothesis, a recent study that measured both OT and CORT after the TSST found that salivary OT levels increased immediately following social stress exposure, prior to increases in salivary CORT (Jong et al. 2015). Taken together, these data suggest that OT treatment may be useful to normalize the HPA-axis and reduce stress-related physiological and subjective responses (e.g., anxiety, craving) that increase drug and alcohol use and trigger relapse.

The endogenous OT system and chronic drug use

OT, the related peptide, vasopressin, and their respective receptors are highly conserved in evolution (Beets et al. 2012). The OT receptor belongs to the rhodopsin-type (class1) G protein-coupled receptor family and was cloned in 1992 (Kimura et al. 1992). Studies in laboratory animals have demonstrated that OT is synthesized in the magnocellular neurons of the paraventricular (PVN), supraoptic (SON) and accessory magnocellular (AN) nuclei of the hypothalamus and released into the bloodstream from axon terminals of these neurons, which are located in the posterior pituitary. Central release of OT occurs by two mechanisms (Ross and Young 2009). 1) There is dendritic release from the PVN and SON in the hypothalamus (Ludwig and Leng 2006) with passive diffusion to OT receptors, which are located throughout the brain (Gimpl and Fahrenholz 2001). 2) More targeted direct release of OT occurs from nerve terminals of parvocellular neurons of the PVN, which project centrally to diverse regions including, among others, the olfactory bulb, tubercule, medial and central amygdala, lateral septum, hippocampus(HC), brainstem and spinal cord (Stoop 2014).

There is evidence that chronic exposure to drugs of abuse produces compensatory neuroadaptive changes in the endogenous OT system in specific brain regions involved in addiction processes. The direction of change is region-specific. For example, OT receptor density in the rodent amygdala and hypothalamus increases following chronic administration of methamphetamine, cocaine and morphine (Zanos et al. 2014a,b, Georgiou et al. 2015), while in the hippocampus (HPC), decreased levels of OT and/or OT receptor density were reported following chronic administration of cocaine (Sarnyai and Kovacs 1994) or morphine (Zanos et al. 2014a). Chronic self-administration of methamphetamine also results in decreased OT receptor immunoreactive fibers in the nucleus accumbers (NAc) Core compared to yoked controls (Baracz et al. 2015b). Chronic exposure to Δ9 –tetrahydrocannabinol in rats downregulates OT mRNA expression in the NAc and ventral tegmental area (VTA) (Butovsky et al. 2006). Chronic administration of morphine to rats alters brain OT expression differentially with a decrease in OT mRNA in the SON and NAc and an increase in the VTA and locus coeruleus (You, Li et al. 2000).

To date, studies in humans are limited to post-mortem autoradiography studies and examination of circulating OT. In alcoholic individuals, plasma OT levels were decreased (Marchesi et al. 1997) and OT immunoreactivity was decreased in the hypothalamus post-mortem (Sivukhina et al. 2006). A final common pathway of drugs and alcohol is activation of stress as well as reward processing (Koob et al. 2014). It is unknown how activation of either of these pathways results in a decrease of OT synthesis, especially when in general stressors are associated with an activation of OT systems (Light et al. 2004; Hoge et al. 2008; Taylor, et al. 2010; Jong et al. 2015). Apart from the effect of the drugs of abuse on the endogenous OT system, environmental factors such as stress and traumatic social experiences all contribute to the emergence of individual differences in the endogenous OT system that may then contribute to the development of addiction (Buisman-Pijlman et al. 2014). Overall, it is also unknown if administration of exogenous OT will reverse these changes in OT synthesis created by exposure to drugs of abuse and whether this would translate to reversal of addictive behaviors. or if administration of OT can prevent the development of addiction after exposure to environmental risk factors. Lastly, the relationship between peripheral OT levels in plasma and central levels of OT is not known, but they are thought to be separately regulated (Ludwig and Leng 2006); therefore, studies that use plasma OT as a surrogate for central OT function are of limited value.

Preclinical studies examining the effect of oxytocin on drug-seeking behavior

Interest in OT as a modulator of the neurobehavioral response to alcohol and other drugs of abuse stems from work done in the 1970’s by De Wied and colleagues (Bohus et al. 1978), who demonstrated that hypophyseal hormones modulate learning and memory, with OT generally having an inhibitory effect. Since current theories conceptualize addiction as a form of maladaptive learning, a large number of preclinical studies in rodents have investigated the effect of OT on various drug-related behaviors, to determine whether OT could reverse the neuroadaptations occurring with repeated drug and alcohol use (Sarnyai and Kovacs 1994).

Since the majority of the published literature has focused on the effects of OT on behavioral and neurochemical responses to opiates, psychostimulants and alcohol we summarize those studies below.

Opiates

Early preclinical studies focused on the effect of OT to modulate the development of acute and chronic tolerance to the analgesic effect of morphine and heroin is reduced with a single dose of OT, while higher doses of OT are required to reduce established chronic tolerance (Kovacs et al. 1984, 1985b, 1987). Further, administration of an OT receptor antagonist inhibits this effect on tolerance, suggesting that the endogenous OT system opposes the development of acute tolerance (Kovacs et al. 1987). Moreover this effect appears to be centrally mediated as intracerebroventricular (ICV) injections of OT are more potent in their action to inhibit acute and chronic tolerance, as are localized injection into the HPC or NAc or administration of dipeptides derived from the C-terminal OT portion (Kovacs et al. 1985). OT also delays naloxone-induced withdrawal symptoms in a dose dependent manner (Kovacs et al. 1984, 1985b).

With respect to self-administration behavior, OT decreases both the acquisition and maintenance of heroin self-administration (Kovacs et al. 1985b). This effect is reproduced with injection of OT into the NAc or HPC (Ibragimov et al. 1987). However, the effect of OT on heroin self-administration was tested with only one dose of heroin (Kovacs et al. 1985a); therefore, without dose –response information, it is not possible to determine whether OT was inhibiting or potentiating the effects of heroin. OT given ICV increased the expression of morphine-induced conditioned place preference(Moaddab, Hyland et al. 2015). This effect may be explained in part by a well described effect of acute morphine to inhibit OT cell firing in the hypothalamus, with tolerance developing to this effect with repeated administration of morphine (Bicknell et al. 1988). During morphine withdrawal there is a rebound hyper-excitation of these cells with increased release of OT (Bicknell et al. 1988). |In light of this effect, administration of OT after conditioning with repeated injections of morphine may signal a withdrawal state, which then drives the increased conditioned place preference observed in this study (Moaddab et al. 2015). Further evidence that OT is involved in withdrawal from morphine, is that peripheral administration of carbetocin, an OT analogue, inhibits the development of anxiety and depressive behaviors and improves social behaviors during morphine withdrawal, as well as preventing stress-induced reinstatement of conditioned place preference for morphine (Zanos et al. 2014). . These results are consistent with those from Qi et al. (2009) who reported an inhibitory effect of OT on stress-primed reinstatement of conditioned place preference for methamphetamine.

In summary, there is evidence that OT inhibits tolerance to opiates, and reduces opiate self-administration, and stress-induced reinstatement of extinguished opiate-taking behaviors. At the same time, it is not clear if changes in OT levels during early drug withdrawal have a positive or negative impact on withdrawal severity and drug-taking behaviors. The increase in endogenous OT release during withdrawal may drive opiate-seeking behavior in the withdrawal state, but may also ameliorate behavioral disturbances that occur during opiate withdrawal.

Psychostimulants

Early preclinical work examined the effects of OT on psychostimulant-related behaviors. For example, OT reduced locomotor hyperactivity and stereotyped behaviors induced by cocaine (Kovacsi et al. 1990; Sarnyai et al. 1990, 1991), but not amphetamine (Kovacs et al. 1985b; Sarnyai and Kovacs 1994).

More recent preclinical work on psychostimulants has focused on methamphetamine, probing the mechanism underlying the largely inhibitory effect of OT on locomotor hyperactivity, self-administration and conditioned place preference (Qi et al. 2008, 2009; Carson et al. 2010a,b;Baracz et al. 2012; Baracz and Cornish 2013; Han et al. 2014; Bahi 2015). The recent studies examining the mechanism of drug-, stress- or cue-induced reinstatement of conditioned place preference or drug-seeking behavior on balance show that OT has an effect on all of these animal models, with some evidence that the mesocorticolimbic system is involved in modulating these effects. It should be noted, however, that there are inconsistent results with respect to the direct involvement of the OT receptor in the reported inhibitory effects; these studies are reviewed below.

One preclinical paradigm, conditioned place preference (or aversion) involves pairing repeated drug administration with a specific environment, and association of a different environment with the absence of the drug, to measure the rewarding effects of the test drug. , OT has been reported to reduce the acquisition, but not the expression, of methamphetamine-induced conditioned place preference (Qi et al. 2009). This suggests that OT reduces the rewarding effect of methamphetamine and perhaps does less to inhibit memory retrieval processes. The potential application of OT as a preventative treatment for the development of addictive behaviors was examined by Hicks and colleagues who treated female rats with daily systemic OT during adolescence and found a reduction in responding for methamphetamine under a progressive ratio schedule and reduced reinstatement to a methamphetamine prime, as well as higher plasma OT levels, compared to untreated rats (Hicks et al. 2016).

Reinstatement models of drug relapse include both operant self-administration models and place conditioning models. OT studies report mixed results depending on the reinstatement paradigm used (conditioned place preference vs. self-administration). OT decreases both stress- and cue-, (but not drug-) primed reinstatement of methamphetamine-conditioned place preference (Qi et al. 2009; Han et al. 2014; Morales-Rivera et al. 2014) . The effect of OT to reduce stress- (but not drug-) primed reinstatement of conditioned place preference for methamphetamine was accompanied by a reduction in medial prefrontal cortical (mPFC) glutamate levels as measured by microdialysis (Qi et al. 2009). This effect was reversed by an OT antagonist. The effect of OT on stress priming of the reinstatement of conditioned place preference was further explored by microinjecting OT into the dorsal HPC and mPFC (Han et al. 2014). Stress-induced reinstatement was inhibited by OT injected into the mPFC and reversed with an OT receptor antagonist. It was also inhibited when injected into the dorsal HPC, albeit at a higher dose and this effect was not reversed by an OT receptor antagonist.

Local administration of OT into the subthalamic nucleus or the NAc core decreased methamphetamine-induced reinstatement of self-administration (Baracz et al. 2015a, 2016). These effects were not reversed with co- administration of an OT antagonist suggesting that OT does not directly modulate methamphetamine relapse behavior, and that other receptor systems are likely involved.

Zhou and colleagues examined the effect of OT on cocaine self-administration under different reinforcement schedules and conditions of drug availability, as well as cue- and drug-primed reinstatement of cocaine- and sucrose-seeking behavior (Zhou et al. 2015). OT dose-dependently reduced cocaine self-administration under conditions of increasing motivational demands (i.e., progressive ratio schedules) and reduced the maximal amount of work (breakpoint) for a cocaine injection. OT also reduced the effects of cocaine- or cue-induced reinstatement of extinguished cocaine self-administration behavior (i.e., cocaine seeking) and reversed cocaine-induced changes in glutamate receptor function (Zhou et al. 2015)

Alcohol

Recent preclinical data show that administration of OT may disrupt biobehavioral adaptions associated with long-term alcohol exposure (Sarnyai and Kovacs 1994; McGregor and Bowen 2011). Early studies with OT focused on the ability of the peptide to reduce or reverse tolerance. A single dose of OT had no effect on tolerance; however, repeated dosing prior to alcohol decreased tolerance to the hypothermic response to alcohol (Szabo et al. 1985, 1989). Interestingly, once tolerance had developed, administration of OT had no modulatory effect on tolerance (Szabo et al. 1985). In alcohol-naïve or alcohol-dependent mice, a single dose of OT had no effect on the severity of picrotoxin-induced seizures (Szabo et al. 1987). In contrast, OT, administered before alcohol daily, resulted in a milder intensity of alcohol withdrawal seizures precipitated by picrotoxin (Szabo et al. 1987), suggesting rapid reversal of tolerance. In addition, OT reduces dthe evelopment of rapid tolerance to the hypnotic, myorelaxant effects of alcohol (Rigter et al. 1980; Pucilowski et al. 1985; Szabo et al. 1985, 1989), and produces a prolonged attenuation of alcohol withdrawal symptoms (Szabo et al. 1987; Kovacs et al. 1998; McGregor and Bowen 2011).

For self-administration, large doses of OT administered either peripherally or centrally have no effect on alcohol drinking using a two-bottle free-choice paradigm (Peters, Slattery et al. 2013). However, McGregor and Bowen (2012) found that a single dose of OT, 1 mg/kg, reduced preference for an alcoholic beverage as compared to a non-alcoholic sweet solution, and this effect lasted for up to six weeks. Further, OT treatment for 2 weeks before the introduction of a two-bottle choice paradigm also resulted in a significantly lower alcohol preference in OT-treated compared to control rats. MacFadyen and colleagues recently reported that OT at lower doses (0.1-0.5 mg/kg i.p.) administered systemically reduced alcohol self-administration (MacFadyen et al. 2016). In a chronic intermittent access model, OT administered ICV acutely similarly reduced ethanol self-administration and also blocked ethanol-induced dopamine release in the NAc, in both ethanol-naïve and chronically exposed rats (Peters et al. 2016), highlighting a possible mechanism for the behavioral effect observed.

Consistent with these results, administration of the OT receptor agonist carbetocin or genetic over-expression of OT receptors in the NAc via a lentiviral vector decreased acquisition and alcohol-primed reinstatement of conditioned place preference and increased rates of extinction (Bahi 2015). However, while the OT receptor seems to be involved in the rewarding properties of ethanol and this appears to involve the ventral striatum, the sedative and ataxic effects of alcohol do not appear to be mediated by the OT receptor (Bowen et al. 2015).

Overall, there is evidence that OT, administered systemically or centrally, reduces self-administration of opiates, cocaine and alcohol and reinstatement of responding induced by exposure to drug cues and stress. The mechanisms seem to involve the midbrain dopaminergic as well as medial prefrontal glutamatergic pathways.

Human Studies of OT in Alcohol and Drug Dependence

There are relatively few human studies that have examined the effects of OT in subjects with alcohol and drug dependence. Heavy alcohol drinking is associated with dysregulation of HPA-axis activity, as shown by episodes of hypercortisolism between drinking bouts and a blunted CORT response to stress during early abstinence (Kemper et al. 1990; Wand and Dobs 1991; Errico et al. 1993; Vescovi et al. 1997; Boschloo et al. 2011). A blunted CORT response has been associated with increased anxiety and craving during acute abstinence and subsequent relapse to heavy drinking (Walter et al. 2006; Higley et al. 2011; Sinha et al. 2011).

The potential of OT as a treatment for alcohol use disorder is highlighted by a recent small clinical trial (n=11) in alcohol-dependent subjects. All subjects underwent alcohol detoxification with PRN administration of the benzodiazepine lorazepam and were concurrently randomized to intranasal OT (24 IU twice daily for 3 days) or placebo during the withdrawal period. Subjects randomized to OT had significantly fewer alcohol withdrawal symptoms and required less symptom-triggered lorazepam for withdrawal symptoms compared to placebo (Pedersen et al. 2013). Relating this finding to the early preclinical literature, the authors posit that the mechanism of this effect might relate to rapid reversal of tolerance by OT.

In the context of stress provocation using the Trier-Social Stress Test, pretreatment with a single dose of OT (40 IU) reduced stress-induced craving and anxiety in cannabis-dependent individuals (McRae-Clark- et al. 2013). This was a between-subjects double-blind placebo-controlled study (N=16) in which OT reduced marijuana craving and stress scores post-session, but did not change anxiety scores. Also using the Trier Social Stress Test, Flanagan and colleagues examined the effect of OT on the cortisol response in 31 cocaine-dependent subjects. Subjects were randomized to receive a single dose of OT (40IU) or placebo prior to the Trier Social Stress Test. There was a significant relationship between the cortisol response after social stress and the degree of adverse childhood experiences for those on placebo, which was absent in the OT group (Flanagan et al. 2015). Our group reported analogous results in cocaine-dependent in-patients, where the significant positive relationship between state anger and cue-induced craving that was observed in the placebo condition was absent in the OT condition. This was a single OT dose (24IU) crossover study in cocaine-dependent in-patients (N=23) assessing the effect of OT on desire to use, cue-induced craving, and monetary reward tasks. OT, compared to placebo, increased subjects’ desire to use but had no effect on cue-induced cocaine craving (Lee, Glassman et al. 2014). .

These studies require replication as they are small and largely exploratory, with different OT dosing and patients who were all drug- or alcohol-dependent but were in different stages of treatment. Specifically, the alcoholic patients were in withdrawal, the marijuana patients were actively using, and the cocaine-dependent patients in one of the studies were abstinent and in a controlled environment. There also are no controlled studies examining the effects of OT on human drug self-administration or subjective effects in the laboratory. Clearly, more human research is needed to determine if the positive signals observed in animal models are replicated in humans. In addition, very little is known about the pharmacokinetics of OT, the effect of repeated OT dosing on behavioral outcomes over time, or the optimal dosing interval.

Brain penetrance of exogenously administered oxytocin and dosing

A key issue is whether systemically administered OT gains access to the CNS directly or whether it exerts its effects via an intermediate pathway. There is considerable controversy on the subject (Leng and Ludwig 2016). That notwithstanding, peptides have been shown to cross the blood-brain barrier, in small amounts either by extracellular active transport or by transcellular diffusion (Banks 2015). In humans, the time course of cerebrospinal fluid (CSF) levels of oxytocin and vasopressin, following their intranasal delivery, is consistent with extracellular transport, as elevated CSF levels are detected 20 −80 minutes after delivery (Born et al. 2002; Striepens et al. 2013). In similar studies, OT reached peak levels in plasma 30-40 min after intranasal administration (Lundin et al. 1986; Burri et al. 2008; Gossen et al. 2012). The relevance of circulating blood levels is unclear, given the considerable individual differences in OT peak and AUD (area under the curve) levels in plasma (Gossenet al. 2012) and the lack of correlation between plasma and CSF levels (Striepenset al. 2013; Freeman et al. 2016) . It is clear, however, that intranasal administration of OT results in elevated CSF levels of OT in humans (Born et al. 2002; Striepens et al. 2013), nonhuman primates(Chan et al. 2012; Dal Monteet al. 2014; Modi et al. 2014; Freeman et al. 2016) and rodents (Neumann et al. 2013). It is not known whether the elevation in CSF OT after systemic administration is due to the administered peptide gaining access to the CSF or whether there is a feed-forward effect stimulated by either peripheral or centrally mediated mechanisms (Ermisch et al. 1985; Carson et al. 2010). Further, it is also not clear if the delivered peptide reaches the CNS and whether it does so as an intact peptide.

The utility and feasibility of administration of intranasal OT as a medication is supported by its success in early clinical trials for the treatment of behavioral deficits associated with other neuropsychiatric disorders such as schizophrenia (Pedersen et al. 2011), social anxiety (Labuschagne et al. 2010), autism (Hollander et al. 2003), and Prader-Willi syndrome (Tauber et al. 2011). The dose of OT used has varied across studies and clinical trials. Most studies have used 24 IU OT (MacDonald et al. 2011), although 40IU OT was effective in cognition and stress studies (Ditzen et al. 2009; Simeon et al. 2011; Krueger et al. 2012; Leknes et al. 2012). Side effects of OT generally appear to be minimal. For example, a recent meta-analysis of the safety and side effects of intranasal OT, drawn from 38 randomized trials, concluded that OT produces no reliable side effects, and is not associated with adverse outcomes when delivered at 18-40 IU/dose for short-term use in controlled research (MacDonalds et al. 2011). In addition, doses as high as 320 IU/day OT have been safely administered to human subjects, without reports of adverse events (Epperson et al. 1996; Ohlsson et al. 2005).

Conclusions and Future Directions

Clinically, OT is an attractive candidate for the treatment of substance use disorders. OT exerts effects via interactions with multiple neurotransmitter systems including, the dopamine mesolimbic reward, the HPA axis, and CRF stress systems. It also has low abuse liability, and an excellent safety profile, as well as no detectable psychoactive actions. In most human studies, subjects could not discriminate between placebo and active drug (OT) (MacDonald et al. 2011). The possibility of OT remediating social deficits in drug- and alcohol-addicted patients has been proposed given the results of studies in social-cognitive neuroscience using OT (Meyer-Lindenberg et al. 2011). Indeed, there are now multiple clinical trials underway further exploring the efficacy of OT for treatment of SUD for marijuana (NCT01827332), cocaine (NCT01573273), alcohol (NCT02407340; NCT02251912; NCT02058251), opioids (NCT02548728), and tobacco (NCT02595749). While some findings appear positive, caution should be exercised given the context- and person-dependent effect of OT (Bartz et al. 2011). Current thinking is that, in addition to its anti-stress effects, OT may facilitate the effects of dopamine (Insel, 2003), which is involved in the reinforcing properties of drugs as well as conditioned responses to drugs (Volkow et al. 2006; Lee et al. 2014). Because of this effect, exogenous OT may exacerbate some drug-related behaviors in individuals with SUD. One recent study did report that OT worsened some drug-related behaviors (Lee et al. 2014). At the same time, the growing literature over the last 30 years investigating the effect of OT on neurobehavioral responses to drugs of abuse and alcohol remains promising.

To move the field forward, several important issues must be addressed in future research. First, the mechanism of action underlying the observation of reduced drug and ethanol self-administration and reinstatement in various contexts is still unknown. Additional studies in both laboratory animal and human subjects are needed. Second, OT itself has a short half-life (Kovacs et al. 1990;, McGregor and Bowen 2012), which raises concerns of actual clinical utility. Future investigations should examine the pharmacodynamic effects of OT, which are surprisingly long in some studies. Third, given the gender differences in the OT system and in addiction-related behaviors, and the growing literature on sex-specific drug effects, it is critical that OT studies include both sexes to determine whether there are sex-specific effects. Fourth, the brain penetrance of the exogenously delivered peptide remains an unanswered question, and should be explored fully. Finally, investigations with nonpeptide OT receptor agonists are also needed as this may lead to the development of medications with longer half-life and less frequent administrations, therefore maximizing patients’ treatment compliance.

Acknowledgments

Financial Support: This work was supported by a Bench-to-Bedside (B2B) Grant (PI: Lee) funded by the NIH Office of Behavioral and Social Sciences Research (OBSSR) ,and by the National Institute on Alcohol Abuse and Alcoholism (NIAAA) R21 AA022679 (PI: Weerts).

Footnotes

The authors declare that they have no conflict of interest.

REFERENCES

***PLEASE REMOVE JOURNAL ISSUE NUMBERS

  1. Arletti R, Bertolini A. Oxytocin acts as an antidepressant in two animal models of depression. Life Sci. 1987;41(14):1725–1730. doi: 10.1016/0024-3205(87)90600-x. [DOI] [PubMed] [Google Scholar]
  2. Bahi A. The oxytocin receptor impairs ethanol reward in mice. Physiol Behav. 2015;139:321–327. doi: 10.1016/j.physbeh.2014.11.046. [DOI] [PubMed] [Google Scholar]
  3. Banks WA. Peptides and the blood-brain barrier. Peptides. 2015 doi: 10.1016/j.peptides.2015.03.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Baracz SJ, Cornish JL. Oxytocin modulates dopamine-mediated reward in the rat subthalamic nucleus. Horm Behav. 2013;63(2):370–375. doi: 10.1016/j.yhbeh.2012.12.003. [DOI] [PubMed] [Google Scholar]
  5. Baracz SJ, Rourke PI, Pardey MC, Hunt GE, McGregor IS, Cornish JL. Oxytocin directly administered into the nucleus accumbens core or subthalamic nucleus attenuates methamphetamine-induced conditioned place preference. Behav Brain Res. 2012;228(1):185–193. doi: 10.1016/j.bbr.2011.11.038. [DOI] [PubMed] [Google Scholar]
  6. Baracz SJ, Everett NA, Cornish JL. The Involvement of Oxytocin in the Subthalamic Nucleus on Relapse to Methamphetamine-Seeking Behaviour. PLoS One. 2015a;10(8):e0136132. doi: 10.1371/journal.pone.0136132. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Baracz SJ, Parker LM, Suraev AS, Everett NA, Goodchild AK, McGregor IS, Cornish JL. Chronic methamphetamine self-administration dysregulates oxytocin plasma levels and oxytocin receptor fibre density in the nucleus accumbens core and subthalamic nucleus of the rat. J Neuroendocrinol. 2016a Apr;28(4) doi: 10.1111/jne.12337. AQ6. 2016. 28. doi: 10.1111/jne.12337. [DOI] [PubMed] [Google Scholar]
  8. Baracz SJ, Everett NA, McGregor IS, Cornish JL. Oxytocin in the nucleus accumbens core reduces reinstatement of methamphetamine-seeking behaviour in rats. Addict Biol. 2016;21(2):316–325. doi: 10.1111/adb.12198. [DOI] [PubMed] [Google Scholar]
  9. Bartz JA, Zaki J, Bolger N, Ochsner KN. Social effects of oxytocin in humans: context and person matter. Trends Cogn Sci. 2011;15(7):301–309. doi: 10.1016/j.tics.2011.05.002. [DOI] [PubMed] [Google Scholar]
  10. Baskerville TA, Douglas AJ. Dopamine and oxytocin interactions underlying behaviors: potential contributions to behavioral disorders. CNS Neurosci Ther. 2010;16(3):e92–123. doi: 10.1111/j.1755-5949.2010.00154.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Beets I, Janssen T, Meelkop E, Temmerman L, Suetens N, Rademakers S, Jansen G, Schoofs L. Vasopressin/oxytocin-related signaling regulates gustatory associative learning in C. elegans. Science. 2012;338(6106):543–545. doi: 10.1126/science.1226860. [DOI] [PubMed] [Google Scholar]
  12. Bicknell RJ, Leng G, Lincoln DW, Russell JA. Naloxone excites oxytocin neurones in the supraoptic nucleus of lactating rats after chronic morphine treatment. J Physiol. 1988;396:297–317. doi: 10.1113/jphysiol.1988.sp016963. [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Bohus B, Kovacs GL, de Wied D. Oxytocin, vasopressin and memory: opposite effects on consolidation and retrieval processes. Brain Res. 1978;157(2):414–417. doi: 10.1016/0006-8993(78)90052-5. [DOI] [PubMed] [Google Scholar]
  14. Born J, Lange T, Kern W, McGregor GP, Bickel U, Fehm HL. Sniffing neuropeptides: a transnasal approach to the human brain. Nat Neurosci. 2002;5(6):514–516. doi: 10.1038/nn849. [DOI] [PubMed] [Google Scholar]
  15. Boschloo L, Vogelzangs N, Licht CM, Vreeburg SA, Smit JH, van den Brink W, Veltman DJ, de Geus EJ, Beekman AT, Penninx BW. Heavy alcohol use, rather than alcohol dependence, is associated with dysregulation of the hypothalamic-pituitary-adrenal axis and the autonomic nervous system. Drug Alcohol Depend. 2011;116(1-3):170–176. doi: 10.1016/j.drugalcdep.2010.12.006. [DOI] [PubMed] [Google Scholar]
  16. Bowen MT, Peters ST, Absalom N, Chebib M, Neumann ID, McGregor IS. Oxytocin prevents ethanol actions at delta subunit-containing GABAA receptors and attenuates ethanol-induced motor impairment in rats. Proc Natl Acad Sci U S A. 2015;112(10):3104–3109. doi: 10.1073/pnas.1416900112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Buisman-Pijlman FT, Sumracki NM, Gordon JJ, Hull PR, Carter CS, Tops M. Individual differences underlying susceptibility to addiction: Role for the endogenous oxytocin system. Pharmacol Biochem Behav. 2014;119:22–38. doi: 10.1016/j.pbb.2013.09.005. [DOI] [PubMed] [Google Scholar]
  18. Burri A, Heinrichs M, Schedlowski M, Kruger TH. The acute effects of intranasal oxytocin administration on endocrine and sexual function in males. Psychoneuroendocrinology. 2008;33(5):591–600. doi: 10.1016/j.psyneuen.2008.01.014. [DOI] [PubMed] [Google Scholar]
  19. Butovsky E, Juknat A, Elbaz J, Shabat-Simon M, Eilam R, Zangen A, Altstein M, Vogel Z. Chronic exposure to Delta9-tetrahydrocannabinol downregulates oxytocin and oxytocin-associated neurophysin in specific brain areas. Mol Cell Neurosci. 2006;31(4):795–804. doi: 10.1016/j.mcn.2006.01.008. [DOI] [PubMed] [Google Scholar]
  20. Carson DS, Cornish JL, Guastella AJ, Hunt GE, McGregor IS. Oxytocin decreases methamphetamine self-administration, methamphetamine hyperactivity, and relapse to methamphetamine-seeking behaviour in rats. Neuropharmacology. 2010a;58(1):38–43. doi: 10.1016/j.neuropharm.2009.06.018. [DOI] [PubMed] [Google Scholar]
  21. Carson DS, Hunt GE, Guastella AJ, Barber L, Cornish JL, Arnold JC, Boucher AA, McGregor IS. Systemically administered oxytocin decreases methamphetamine activation of the subthalamic nucleus and accumbens core and stimulates oxytocinergic neurons in the hypothalamus. Addict Biol. 2010b;15(4):448–463. doi: 10.1111/j.1369-1600.2010.00247.x. [DOI] [PubMed] [Google Scholar]
  22. Chang SW, Barter JW, Ebitz RB, Watson KK, Platt ML. Inhaled oxytocin amplifies both vicarious reinforcement and self reinforcement in rhesus macaques (Macaca mulatta) Proc Natl Acad Sci U S A. 2012;109(3):959–964. doi: 10.1073/pnas.1114621109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Cruz FC, Quadros IM, Planeta Cda S, Miczek KA. Maternal separation stress in male mice: long-term increases in alcohol intake. Psychopharmacology (Berl) 2008;201(3):459–468. doi: 10.1007/s00213-008-1307-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Dal Monte O, Noble PL, Turchi J, Cummins A, Averbeck BB. CSF and blood oxytocin concentration changes following intranasal delivery in macaque. PLoS One. 2014;9(8):e103677. doi: 10.1371/journal.pone.0103677. [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. de Oliveira DC, Zuardi AW, Graeff FG, Queiroz RH, Crippa JA. Anxiolytic-like effect of oxytocin in the simulated public speaking test. J Psychopharmacol. 2012;26(4):497–504. doi: 10.1177/0269881111400642. [DOI] [PubMed] [Google Scholar]
  26. Di Simplicio M, Massey-Chase R, Cowen PJ, Harmer CJ. Oxytocin enhances processing of positive versus negative emotional information in healthy male volunteers. J Psychopharmacol. 2009;23(3):241–248. doi: 10.1177/0269881108095705. [DOI] [PubMed] [Google Scholar]
  27. Ditzen B, Schaer M, Gabriel B, Bodenmann G, Ehlert U, Heinrichs M. Intranasal oxytocin increases positive communication and reduces cortisol levels during couple conflict. Biol Psychiatry. 2009;65(9):728–731. doi: 10.1016/j.biopsych.2008.10.011. [DOI] [PubMed] [Google Scholar]
  28. Ebner K, Wotjak CT, Landgraf R, Engelmann M. A single social defeat experience selectively stimulates the release of oxytocin, but not vasopressin, within the septal brain area of male rats. Brain Res. 2000;872(1-2):87–92. doi: 10.1016/s0006-8993(00)02464-1. [DOI] [PubMed] [Google Scholar]
  29. Epperson CN, McDougle CJ, Price LH. Intranasal oxytocin in obsessive-compulsive disorder. Biol Psychiatry. 1996;40(6):547–549. doi: 10.1016/0006-3223(96)00120-5. [DOI] [PubMed] [Google Scholar]
  30. Ermisch A, Ruhle HJ, Landgraf R, Hess J. Blood-brain barrier and peptides. J Cereb Blood Flow Metab. 1985;5(3):350–357. doi: 10.1038/jcbfm.1985.49. [DOI] [PubMed] [Google Scholar]
  31. Errico AL, Parsons OA, King AC, Lovallo WR. Attenuated cortisol response to biobehavioral stressors in sober alcoholics. J Stud Alcohol. 1993;54(4):393–398. doi: 10.15288/jsa.1993.54.393. [DOI] [PubMed] [Google Scholar]
  32. Fahlke C, Hard E, Hansen S. Facilitation of ethanol consumption by intracerebroventricular infusions of corticosterone. Psychopharmacology (Berl) 1996;127(2):133–139. doi: 10.1007/BF02805986. [DOI] [PubMed] [Google Scholar]
  33. Fahlke C, Lorenz JG, Long J, Champoux M, Suomi SJ, Higley JD. Rearing experiences and stress-induced plasma cortisol as early risk factors for excessive alcohol consumption in nonhuman primates. Alcohol Clin Exp Res. 2000;24(5):644–650. [PubMed] [Google Scholar]
  34. Fish EW, McKenzie-Quirk SD, Bannai M, Miczek KA. 5-HT(1B) receptor inhibition of alcohol-heightened aggression in mice: comparison to drinking and running. Psychopharmacology (Berl) 2008;197(1):145–156. doi: 10.1007/s00213-007-1017-3. [DOI] [PubMed] [Google Scholar]
  35. Flanagan JC, Baker NL, McRae-Clark AL, Brady KT, Moran-Santa Maria MM. Effects of adverse childhood experiences on the association between intranasal oxytocin and social stress reactivity among individuals with cocaine dependence. Psychiatry Res. 2015;229(1-2):94–100. doi: 10.1016/j.psychres.2015.07.064. [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Foley P, Kirschbaum C. Human hypothalamus-pituitary-adrenal axis responses to acute psychosocial stress in laboratory settings. Neurosci Biobehav Rev. 2010;35(1):91–96. doi: 10.1016/j.neubiorev.2010.01.010. [DOI] [PubMed] [Google Scholar]
  37. Freeman SM, Samineni S, Allen PC, Stockinger D, Bales KL, Hwa GG, Roberts JA. Plasma and CSF oxytocin levels after intranasal and intravenous oxytocin in awake macaques. Psychoneuroendocrinology. 2016;66:185–194. doi: 10.1016/j.psyneuen.2016.01.014. [DOI] [PubMed] [Google Scholar]
  38. Funk D, Vohra S, Le AD. Influence of stressors on the rewarding effects of alcohol in Wistar rats: studies with alcohol deprivation and place conditioning. Psychopharmacology (Berl) 2004;176(1):82–87. doi: 10.1007/s00213-004-1859-x. [DOI] [PubMed] [Google Scholar]
  39. Georgiou P, Zanos P, Ehteramyan M, Hourani S, Kitchen I, Maldonado R, Bailey A. Differential regulation of mGlu5 R and MuOPr by priming- and cue-induced reinstatement of cocaine-seeking behaviour in mice. Addict Biol. 2015;20(5):902–912. doi: 10.1111/adb.12208. [DOI] [PubMed] [Google Scholar]
  40. Gimpl G, Fahrenholz F. The oxytocin receptor system: structure, function, and regulation. Physiol Rev. 2001;81(2):629–683. doi: 10.1152/physrev.2001.81.2.629. [DOI] [PubMed] [Google Scholar]
  41. Gossen A, Hahn A, Westphal L, Prinz S, Schultz RT, Grunder G, Spreckelmeyer KN. Oxytocin plasma concentrations after single intranasal oxytocin administration - a study in healthy men. Neuropeptides. 2012;46(5):211–215. doi: 10.1016/j.npep.2012.07.001. [DOI] [PubMed] [Google Scholar]
  42. Han WY, Du P, Fu SY, Wang F, Song M, Wu CF, Yang JY. Oxytocin via its receptor affects restraint stress-induced methamphetamine CPP reinstatement in mice: Involvement of the medial prefrontal cortex and dorsal hippocampus glutamatergic system. Pharmacol Biochem Behav. 2014;119:80–87. doi: 10.1016/j.pbb.2013.11.014. [DOI] [PubMed] [Google Scholar]
  43. Heinrichs M, Baumgartner T, Kirschbaum C, Ehlert U. Social support and oxytocin interact to suppress cortisol and subjective responses to psychosocial stress. Biol Psychiatry. 2003;54(12):1389–1398. doi: 10.1016/s0006-3223(03)00465-7. [DOI] [PubMed] [Google Scholar]
  44. Hicks C, Cornish JL, Baracz SJ, Suraev A, McGregor IS. Adolescent pre-treatment with oxytocin protects against adult methamphetamine-seeking behavior in female rats. Addict Biol. 2016;21(2):304–315. doi: 10.1111/adb.12197. [DOI] [PubMed] [Google Scholar]
  45. Higley AE, Crane NA, Spadoni AD, Quello SB, Goodell V, Mason BJ. Craving in response to stress induction in a human laboratory paradigm predicts treatment outcome in alcohol-dependent individuals. Psychopharmacology (Berl) 2011;218(1):121–129. doi: 10.1007/s00213-011-2355-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. Hoge EA, Pollack MH, Kaufman RE, Zak PJ, Simon NM. Oxytocin levels in social anxiety disorder. CNS Neurosci Ther. 2008;14(3):165–170. doi: 10.1111/j.1755-5949.2008.00051.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Hollander E, Novotny S, Hanratty M, Yaffe R, DeCaria CM, Aronowitz BR, Mosovich S. Oxytocin infusion reduces repetitive behaviors in adults with autistic and Asperger's disorders. Neuropsychopharmacology. 2003;28(1):193–198. doi: 10.1038/sj.npp.1300021. [DOI] [PubMed] [Google Scholar]
  48. Ibragimov R, Kovacs GL, Szabo G, Telegdy G. Microinjection of oxytocin into limbic-mesolimbic brain structures disrupts heroin self-administration behavior: a receptor-mediated event? Life Sci. 1987;41(10):1265–1271. doi: 10.1016/0024-3205(87)90205-0. [DOI] [PubMed] [Google Scholar]
  49. Insel TR. Is social attachment an addictive disorder? Physiol Behav. 2003;79(3):351–357. doi: 10.1016/s0031-9384(03)00148-3. [DOI] [PubMed] [Google Scholar]
  50. Insel TR, Winslow JT. Central administration of oxytocin modulates the infant rat's response to social isolation. Eur J Pharmacol. 1991;203(1):149–152. doi: 10.1016/0014-2999(91)90806-2. [DOI] [PubMed] [Google Scholar]
  51. Jezova D, Skultetyova I, Tokarev DI, Bakos P, Vigas M. Vasopressin and oxytocin in stress. Ann N Y Acad Sci. 1995;771:192–203. doi: 10.1111/j.1749-6632.1995.tb44681.x. [DOI] [PubMed] [Google Scholar]
  52. Jong TR, Menon R, Bludau A, Grund T, Biermeier V, Klampfl SM, Jurek B, Bosch OJ, Hellhammer J, Neumann ID. Salivary oxytocin concentrations in response to running, sexual self-stimulation, breastfeeding and the TSST: The Regensburg Oxytocin Challenge (ROC) study. Psychoneuroendocrinology. 2015;62:381–388. doi: 10.1016/j.psyneuen.2015.08.027. [DOI] [PubMed] [Google Scholar]
  53. Kemper A, Koalick F, Thiele H, Retzow A, Rathsack R, Nickel B. Cortisol and beta-endorphin response in alcoholics and alcohol abusers following a high naloxone dosage. Drug Alcohol Depend. 1990;25(3):319–326. doi: 10.1016/0376-8716(90)90158-b. [DOI] [PubMed] [Google Scholar]
  54. Kimura T, Tanizawa O, Mori K, Brownstein MJ, Okayama H. Structure and expression of a human oxytocin receptor. Nature. 1992;356(6369):526–529. doi: 10.1038/356526a0. [DOI] [PubMed] [Google Scholar]
  55. Koob GF, Buck CL, Cohen A, Edwards S, Park PE, Schlosburg JE, Schmeichel B, Vendruscolo LF, Wade CL, Whitfield TW, Jr., George O. Addiction as a stress surfeit disorder. Neuropharmacology. 2014;76:370–382. doi: 10.1016/j.neuropharm.2013.05.024. Pt B. [DOI] [PMC free article] [PubMed] [Google Scholar]
  56. Kovacs GL, Izbeki F, Horvath Z, Telegdy G. Effects of oxytocin and a derivative (Z-prolyl-D-leucine) on morphine tolerance/withdrawal are mediated by the limbic system. Behav Brain Res. 1984;14(1):1–8. doi: 10.1016/0166-4328(84)90014-7. [DOI] [PubMed] [Google Scholar]
  57. Kovacs GL, Borthaiser Z, Telegdy G. Oxytocin reduces intravenous heroin self-administration in heroin-tolerant rats. Life Sci. 1985a;37(1):17–26. doi: 10.1016/0024-3205(85)90620-4. [DOI] [PubMed] [Google Scholar]
  58. Kovacs GL, Horvath Z, Sarnyai Z, Faludi M, Telegdy G. Oxytocin and a C-terminal derivative (Z-prolyl-D-leucine) attenuate tolerance to and dependence on morphine and interact with dopaminergic neurotransmission in the mouse brain. Neuropharmacology. 1985b;24(5):413–419. doi: 10.1016/0028-3908(85)90026-7. [DOI] [PubMed] [Google Scholar]
  59. Kovacs GL, Sarnyai Z, Izbeki F, Szabo G, Telegdy G, Barth T, Jost K, Brtnik F. Effects of oxytocin-related peptides on acute morphine tolerance: opposite actions by oxytocin and its receptor antagonists. J Pharmacol Exp Ther. 1987;241(2):569–574. [PubMed] [Google Scholar]
  60. Kovacs GL, Sarnyai Z, Barbarczi E, Szabo G, Telegdy G. The role of oxytocin-dopamine interactions in cocaine-induced locomotor hyperactivity. Neuropharmacology. 1990;29(4):365–368. doi: 10.1016/0028-3908(90)90095-9. [DOI] [PubMed] [Google Scholar]
  61. Kovacs GL, Sarnyai Z, Szabo G. Oxytocin and addiction: a review. Psychoneuroendocrinology. 1998;23(8):945–962. doi: 10.1016/s0306-4530(98)00064-x. [DOI] [PubMed] [Google Scholar]
  62. Krueger F, Parasuraman R, Moody L, Twieg P, de Visser E, McCabe K, O'Hara M, Lee MR. Oxytocin selectively increases perceptions of harm for victims but not the desire to punish offenders of criminal offenses. Soc Cogn Affect Neurosci. 2012 doi: 10.1093/scan/nss026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  63. Kubzansky LD, Mendes WB, Appleton AA, Block J, Adler GK. A heartfelt response: Oxytocin effects on response to social stress in men and women. Biol Psychol. 2012;90(1):1–9. doi: 10.1016/j.biopsycho.2012.02.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Labuschagne I, Phan KL, Wood A, Angstadt M, Chua P, Heinrichs M, Stout JC, Nathan PJ. Oxytocin attenuates amygdala reactivity to fear in generalized social anxiety disorder. Neuropsychopharmacology. 2010;35(12):2403–2413. doi: 10.1038/npp.2010.123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Lang RE, Heil JW, Ganten D, Hermann K, Unger T, Rascher W. Oxytocin unlike vasopressin is a stress hormone in the rat. Neuroendocrinology. 1983;37(4):314–316. doi: 10.1159/000123566. [DOI] [PubMed] [Google Scholar]
  66. Lee MR, Glassman M, King-Casas B, Kelly DL, Stein EA, Schroeder J, Salmeron BJ. Complexity of oxytocins effects in a chronic cocaine dependent population. Eur Neuropsychopharmacol. 2014;24(9):1483–1491. doi: 10.1016/j.euroneuro.2014.06.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  67. Leknes S, Wessberg J, Ellingsen DM, Chelnokova O, Olausson H, Laeng B. Oxytocin enhances pupil dilation and sensitivity to 'hidden' emotional expressions. Soc Cogn Affect Neurosci. 2012 doi: 10.1093/scan/nss062. [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Leng G, Ludwig M. Intranasal Oxytocin: Myths and Delusions. Biol Psychiatry. 2016;79(3):243–250. doi: 10.1016/j.biopsych.2015.05.003. [DOI] [PubMed] [Google Scholar]
  69. Light KC, Grewen KM, Amico JA, Boccia M, Brownley KA, Johns JM. Deficits in plasma oxytocin responses and increased negative affect, stress, and blood pressure in mothers with cocaine exposure during pregnancy. Addict Behav. 2004;29(8):1541–1564. doi: 10.1016/j.addbeh.2004.02.062. [DOI] [PMC free article] [PubMed] [Google Scholar]
  70. Lischke A, Berger C, Prehn K, Heinrichs M, Herpertz SC, Domes G. Intranasal oxytocin enhances emotion recognition from dynamic facial expressions and leaves eye-gaze unaffected. Psychoneuroendocrinology. 2012;37(4):475–481. doi: 10.1016/j.psyneuen.2011.07.015. [DOI] [PubMed] [Google Scholar]
  71. Ludwig M, Leng G. Dendritic peptide release and peptide-dependent behaviours. Nat Rev Neurosci. 2006;7(2):126–136. doi: 10.1038/nrn1845. [DOI] [PubMed] [Google Scholar]
  72. Lundin S, Akerlund M, Fagerstrom PO, Hauksson A, Melin P. Pharmacokinetics in the human of a new synthetic vasopressin and oxytocin uterine antagonist. Acta Endocrinol (Copenh) 1986;112(4):465–472. doi: 10.1530/acta.0.1120465. [DOI] [PubMed] [Google Scholar]
  73. MacDonald E, Dadds MR, Brennan JL, Williams K, Levy F, Cauchi AJ. A review of safety, side-effects and subjective reactions to intranasal oxytocin in human research. Psychoneuroendocrinology. 2011;36(8):1114–1126. doi: 10.1016/j.psyneuen.2011.02.015. [DOI] [PubMed] [Google Scholar]
  74. MacFadyen K, Loveless R, DeLucca B, Wardley K, Deogan S, Thomas C, Peris J. Peripheral oxytocin administration reduces ethanol consumption in rats. Pharmacol Biochem Behav. 2016;140:27–32. doi: 10.1016/j.pbb.2015.10.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  75. Marchesi C, Chiodera P, Brusamonti E, Volpi R, Coiro V. Abnormal plasma oxytocin and beta-endorphin levels in alcoholics after short and long term abstinence. Prog Neuropsychopharmacol Biol Psychiatry. 1997;21(5):797–807. doi: 10.1016/s0278-5846(97)00080-8. [DOI] [PubMed] [Google Scholar]
  76. Marsh AA, Yu HH, Pine DS, Blair RJ. Oxytocin improves specific recognition of positive facial expressions. Psychopharmacology (Berl) 2010;209(3):225–232. doi: 10.1007/s00213-010-1780-4. [DOI] [PubMed] [Google Scholar]
  77. McGregor IS, Bowen MT. Breaking the loop: Oxytocin as a potential treatment for drug addiction. Horm Behav. 2011 doi: 10.1016/j.yhbeh.2011.12.001. [DOI] [PubMed] [Google Scholar]
  78. McGregor IS, Bowen MT. Breaking the loop: oxytocin as a potential treatment for drug addiction. Horm Behav. 2012;61(3):331–339. doi: 10.1016/j.yhbeh.2011.12.001. [DOI] [PubMed] [Google Scholar]
  79. McRae-Clark AL, Baker NL, Maria MM, Brady KT. Effect of oxytocin on craving and stress response in marijuana-dependent individuals: a pilot study. Psychopharmacology (Berl) 2013;228(4):623–631. doi: 10.1007/s00213-013-3062-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  80. 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(9):524–538. doi: 10.1038/nrn3044. [DOI] [PubMed] [Google Scholar]
  81. Moaddab M, Hyland BI, Brown CH. Oxytocin enhances the expression of morphine-induced conditioned place preference in rats. Psychoneuroendocrinology. 2015;53:159–169. doi: 10.1016/j.psyneuen.2015.01.003. [DOI] [PubMed] [Google Scholar]
  82. Modi ME, Connor-Stroud F, Landgraf R, Young LJ, Parr LA. Aerosolized oxytocin increases cerebrospinal fluid oxytocin in rhesus macaques. Psychoneuroendocrinology. 2014;45:49–57. doi: 10.1016/j.psyneuen.2014.02.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  83. Morales-Rivera A, Hernandez-Burgos MM, Martinez-Rivera A, Perez-Colon J, Rivera R, Montalvo J, Rodriguez-Borrero E, Maldonado-Vlaar CS. Anxiolytic effects of oxytocin in cue-induced cocaine seeking behavior in rats. Psychopharmacology (Berl) 2014;231(21):4145–4155. doi: 10.1007/s00213-014-3553-y. [DOI] [PubMed] [Google Scholar]
  84. Neumann ID, Johnstone HA, Hatzinger M, Liebsch G, Shipston M, Russell JA, Landgraf R, Douglas AJ. Attenuated neuroendocrine responses to emotional and physical stressors in pregnant rats involve adenohypophysial changes. J Physiol. 1998;508:289–300. doi: 10.1111/j.1469-7793.1998.289br.x. Pt 1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  85. Neumann ID, Torner L, Wigger A. Brain oxytocin: differential inhibition of neuroendocrine stress responses and anxiety-related behaviour in virgin, pregnant and lactating rats. Neuroscience. 2000;95(2):567–575. doi: 10.1016/s0306-4522(99)00433-9. [DOI] [PubMed] [Google Scholar]
  86. Neumann ID, Maloumby R, Beiderbeck DI, Lukas M, Landgraf R. Increased brain and plasma oxytocin after nasal and peripheral administration in rats and mice. Psychoneuroendocrinology. 2013;38(10):1985–1993. doi: 10.1016/j.psyneuen.2013.03.003. [DOI] [PubMed] [Google Scholar]
  87. Ohlsson B, Truedsson M, Bengtsson M, Torstenson R, Sjolund K, Bjornsson ES, Simren M. Effects of long-term treatment with oxytocin in chronic constipation; a double blind, placebo-controlled pilot trial. Neurogastroenterol Motil. 2005;17(5):697–704. doi: 10.1111/j.1365-2982.2005.00679.x. [DOI] [PubMed] [Google Scholar]
  88. Onaka T, Takayanagi Y, Yoshida M. Roles of oxytocin neurones in the control of stress, energy metabolism, and social behaviour. J Neuroendocrinol. 2012;24(4):587–598. doi: 10.1111/j.1365-2826.2012.02300.x. [DOI] [PubMed] [Google Scholar]
  89. Ondrejcakova M, Bakos J, Garafova A, Kovacs L, Kvetnansky R, Jezova D. Neuroendocrine and cardiovascular parameters during simulation of stress-induced rise in circulating oxytocin in the rat. Stress. 2010;13(4):314–322. doi: 10.3109/10253891003596822. [DOI] [PubMed] [Google Scholar]
  90. Pedersen CA, Gibson CM, Rau SW, Salimi K, Smedley KL, Casey RL, Leserman J, Jarskog LF, Penn DL. Intranasal oxytocin reduces psychotic symptoms and improves Theory of Mind and social perception in schizophrenia. Schizophr Res. 2011;132(1):50–53. doi: 10.1016/j.schres.2011.07.027. [DOI] [PubMed] [Google Scholar]
  91. Pedersen CA, Smedley KL, Leserman J, Jarskog LF, Rau SW, Kampov-Polevoi A, Casey RL, Fender T, Garbutt JC. Intranasal oxytocin blocks alcohol withdrawal in human subjects. Alcohol Clin Exp Res. 2013;37(3):484–489. doi: 10.1111/j.1530-0277.2012.01958.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  92. Peters S, Slattery DA, Flor PJ, Neumann ID, Reber SO. Differential effects of baclofen and oxytocin on the increased ethanol consumption following chronic psychosocial stress in mice. Addict Biol. 2013;18(1):66–77. doi: 10.1111/adb.12001. [DOI] [PubMed] [Google Scholar]
  93. Peters ST, Bowen MT, Bohrer K, McGregor IS, Neumann ID. Oxytocin inhibits ethanol consumption and ethanol-induced dopamine release in the AQ7 nucleus accumbens. Addict Biol. 2016 Jan 25; doi: 10.1111/adb.12362. 2016. doi: 10.1111/adb.12362. [Epub ahead of print] [DOI] [PubMed] [Google Scholar]
  94. Piazza PV, Le Moal ML. Pathophysiological basis of vulnerability to drug abuse: role of an interaction between stress, glucocorticoids, and dopaminergic neurons. Annu Rev Pharmacol Toxicol. 1996;36:359–378. doi: 10.1146/annurev.pa.36.040196.002043. [DOI] [PubMed] [Google Scholar]
  95. Pucilowski O, Kostowski W, Trzaskowska E. The effect of oxytocin and fragment (MIF-I) on the development of tolerance to hypothermic and hypnotic action of ethanol in the rat. Peptides. 1985;6(1):7–10. doi: 10.1016/0196-9781(85)90067-1. [DOI] [PubMed] [Google Scholar]
  96. Qi J, Yang JY, Song M, Li Y, Wang F, Wu CF. Inhibition by oxytocin of methamphetamine-induced hyperactivity related to dopamine turnover in the mesolimbic region in mice. Naunyn Schmiedebergs Arch Pharmacol. 2008;376(6):441–448. doi: 10.1007/s00210-007-0245-8. [DOI] [PubMed] [Google Scholar]
  97. Qi J, Yang JY, Wang F, Zhao YN, Song M, Wu CF. 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(5):856–865. doi: 10.1016/j.neuropharm.2009.01.010. [DOI] [PubMed] [Google Scholar]
  98. Quirin M, Kuhl J, Dusing R. Oxytocin buffers cortisol responses to stress in individuals with impaired emotion regulation abilities. Psychoneuroendocrinology. 2011;36(6):898–904. doi: 10.1016/j.psyneuen.2010.12.005. [DOI] [PubMed] [Google Scholar]
  99. Rigter H, Dortmans C, Crabbe JC., Jr. Effects of peptides related to neurohypophyseal hormones on ethanol tolerance. Pharmacol Biochem Behav. 1980;13(Suppl 1):285–290. doi: 10.1016/s0091-3057(80)80043-8. [DOI] [PubMed] [Google Scholar]
  100. Ross HE, Young LJ. Oxytocin and the neural mechanisms regulating social cognition and affiliative behavior. Front Neuroendocrinol. 2009;30(4):534–547. doi: 10.1016/j.yfrne.2009.05.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  101. Sarnyai Z, Kovacs GL. Role of oxytocin in the neuroadaptation to drugs of abuse. Psychoneuroendocrinology. 1994;19(1):85–117. doi: 10.1016/0306-4530(94)90062-0. [DOI] [PubMed] [Google Scholar]
  102. Sarnyai Z, Szabo G, Kovacs GL, Telegdy G. Oxytocin attenuates the cocaine-induced exploratory hyperactivity in mice. Neuroreport. 1990;1(3-4):200–202. doi: 10.1097/00001756-199011000-00006. [DOI] [PubMed] [Google Scholar]
  103. Sarnyai Z, Babarczy E, Krivan M, Szabo G, Kovacs GL, Barth T, Telegdy G. Selective attenuation of cocaine-induced stereotyped behaviour by oxytocin: putative role of basal forebrain target sites. Neuropeptides. 1991;19(1):51–56. doi: 10.1016/0143-4179(91)90073-r. [DOI] [PubMed] [Google Scholar]
  104. Simeon D, Bartz J, Hamilton H, Crystal S, Braun A, Ketay S, Hollander E. Oxytocin administration attenuates stress reactivity in borderline personality disorder: a pilot study. Psychoneuroendocrinology. 2011;36(9):1418–1421. doi: 10.1016/j.psyneuen.2011.03.013. [DOI] [PubMed] [Google Scholar]
  105. Sinha R. Chronic stress, drug use, and vulnerability to addiction. Ann N Y Acad Sci. 2008;1141:105–130. doi: 10.1196/annals.1441.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  106. Sinha R, Fox HC, Hong KI, Hansen J, Tuit K, Kreek MJ. Effects of adrenal sensitivity, stress- and cue-induced craving, and anxiety on subsequent alcohol relapse and treatment outcomes. Arch Gen Psychiatry. 2011;68(9):942–952. doi: 10.1001/archgenpsychiatry.2011.49. [DOI] [PMC free article] [PubMed] [Google Scholar]
  107. Sivukhina EV, Dolzhikov AA, Morozov Iu E, Jirikowski GF, Grinevich V. Effects of chronic alcoholic disease on magnocellular and parvocellular hypothalamic neurons in men. Horm Metab Res. 2006;38(6):382–390. doi: 10.1055/s-2006-944522. [DOI] [PubMed] [Google Scholar]
  108. Stoop R. Neuromodulation by oxytocin and vasopressin in the central nervous system as a basis for their rapid behavioral effects. Curr Opin Neurobiol. 2014;29:187–193. doi: 10.1016/j.conb.2014.09.012. [DOI] [PubMed] [Google Scholar]
  109. Striepens N, Kendrick KM, Hanking V, Landgraf R, Wullner U, Maier W, Hurlemann R. Elevated cerebrospinal fluid and blood concentrations of oxytocin following its intranasal administration in humans. Sci Rep. 2013;3:3440. doi: 10.1038/srep03440. [DOI] [PMC free article] [PubMed] [Google Scholar]
  110. Szabo G, Kovacs GL, Szekeli S, Telegdy G. The effects of neurohypophyseal hormones on tolerance to the hypothermic effect of ethanol. Alcohol. 1985;2(4):567–574. doi: 10.1016/0741-8329(85)90082-5. [DOI] [PubMed] [Google Scholar]
  111. Szabo G, L. Kovacs G, Telegdy G. Effects of neurohypophyseal peptide hormones on alcohol dependence and withdrawal. Alcohol Alcohol. 1987;22(1):71–74. [PubMed] [Google Scholar]
  112. Szabo G, Kovacs GL, Telegdy G. Intraventricular administration of neurohypophyseal hormones interferes with the development of tolerance to ethanol. Acta Physiol Hung. 1989;73(1):97–103. [PubMed] [Google Scholar]
  113. Tauber M, Mantoulan C, Copet P, Jauregui J, Demeer G, Diene G, Roge B, Laurier V, Ehlinger V, Arnaud C, Molinas C, Thuilleaux D. Oxytocin may be useful to increase trust in others and decrease disruptive behaviours in patients with Prader-Willi syndrome: a randomised placebo-controlled trial in 24 patients. Orphanet J Rare Dis. 2011;6:47. doi: 10.1186/1750-1172-6-47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  114. Taylor SE, Saphire-Bernstein S, Seeman TE. Are plasma oxytocin in women and plasma vasopressin in men biomarkers of distressed pair-bond relationships? Psychol Sci. 2010;21(1):3–7. doi: 10.1177/0956797609356507. [DOI] [PubMed] [Google Scholar]
  115. Uhart M, Wand GS. Stress, alcohol and drug interaction: an update of human research. Addict Biol. 2009;14(1):43–64. doi: 10.1111/j.1369-1600.2008.00131.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  116. Vescovi PP, DiGennaro C, Coiro V. Hormonal (ACTH, cortisol, beta-endorphin, and met-enkephalin) and cardiovascular responses to hyperthermic stress in chronic alcoholics. Alcohol Clin Exp Res. 1997;21(7):1195–1198. [PubMed] [Google Scholar]
  117. Volkow ND, Wang GJ, Telang F, Fowler JS, Logan J, Childress AR, Jayne M, Ma Y, Wong C. Cocaine cues and dopamine in dorsal striatum: mechanism of craving in cocaine addiction. J Neurosci. 2006;26(24):6583–6588. doi: 10.1523/JNEUROSCI.1544-06.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  118. Walter M, Gerhard U, Gerlach M, Weijers HG, Boening J, Wiesbeck GA. Cortisol concentrations, stress-coping styles after withdrawal and long-term abstinence in alcohol dependence. Addict Biol. 2006;11(2):157–162. doi: 10.1111/j.1369-1600.2006.00018.x. [DOI] [PubMed] [Google Scholar]
  119. Wand GS, Dobs AS. Alterations in the hypothalamic-pituitary-adrenal axis in actively drinking alcoholics. J Clin Endocrinol Metab. 1991;72(6):1290–1295. doi: 10.1210/jcem-72-6-1290. [DOI] [PubMed] [Google Scholar]
  120. Windle RJ, Shanks N, Lightman SL, Ingram CD. Central oxytocin administration reduces stress-induced corticosterone release and anxiety behavior in rats. Endocrinology. 1997;138(7):2829–2834. doi: 10.1210/endo.138.7.5255. [DOI] [PubMed] [Google Scholar]
  121. You ZD, Li JH, Song CY, Wang CH, Lu CL. Chronic morphine treatment inhibits oxytocin synthesis in rats. Neuroreport. 2000;11(14):3113–3116. doi: 10.1097/00001756-200009280-00015. [DOI] [PubMed] [Google Scholar]
  122. Zanos P, Georgiou P, Wright SR, Hourani SM, 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. 2014a;39(4):855–865. doi: 10.1038/npp.2013.285. [DOI] [PMC free article] [PubMed] [Google Scholar]
  123. Zanos P, Wright SR, Georgiou P, Yoo JH, Ledent C, Hourani SM, Kitchen I, Winsky-Sommerer R, Bailey A. Chronic methamphetamine treatment induces oxytocin receptor up-regulation in the amygdala and hypothalamus via an adenosine A2A receptor-independent mechanism. Pharmacol Biochem Behav. 2014b;119:72–79. doi: 10.1016/j.pbb.2013.05.009. [DOI] [PubMed] [Google Scholar]
  124. Zhou L, Sun WL, Young AB, Lee K, McGinty JF, See RE. Oxytocin reduces cocaine seeking and reverses chronic cocaine-induced changes in glutamate receptor function. Int J Neuropsychopharmacol. 2015 Jan;18(1) doi: 10.1093/ijnp/pyu009. 2015. pyu009. Published online 2014 Dec 22. doi: 10.1093/ijnp/pyu009. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES