1
|
Wang R, Liu G, Chen H, Hou H, Hu Q. Linking oxytocin to nicotine dependence: An experimental study of the brain, behavior, and gut microbiota in rats. Neurosci Lett 2025; 852:138198. [PMID: 40086613 DOI: 10.1016/j.neulet.2025.138198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 03/06/2025] [Accepted: 03/11/2025] [Indexed: 03/16/2025]
Abstract
Oxytocin (OXT) is a hypothalamic neuropeptide, and numerous studies have indicated that exposure to addictive substances, such as opioids, cocaine, etc., can result in decreased function of the OXT system. The study also found that OXT can reduce addictive behavior for certain drugs, including methamphetamine, alcohol, and cocaine, suggesting a close relationship between the OXT system and drug abuse. Although nicotine is the main addictive substance in tobacco, its interaction with the OXT system is unknown and requires further study. Therefore, OXT levels in plasma and brain regions associated with addiction were measured by enzyme-linked immunosorbent assay (ELISA) using chronic nicotine administration via a slow-release pump. In addition, the effects of OXT injection on nicotine self-administration behavior, motor activity, and intestinal microbiota in rats were examined by nicotine self-administration experiment, open field experiment, and 16S sequencing experiment. By depleting gut microbiota with oral antibiotics, this study aims to investigate whether gut microbiota mediates oxytocin effect on the nicotine self-administration behavior in rats. This study shows that chronic nicotine administration can reduce OXT levels in plasma and brain regions such as the paraventricular nucleus (PVN), ventral tegmental area (VTA), and caudate putamen (CPU). OXT at a dose of 1.0 mg/kg significantly reduced the number of nicotine infusions and the abundance of Lactobacillus in rats. Notably, our findings indicate that other mechanisms besides gut microbes are involved in the effect of peripheral OXT administration on the inhibition of nicotine self-administration.
Collapse
Affiliation(s)
- Ruiyan Wang
- Institute of Biomedical Engineering, College of Life Sciences, Qingdao University, Qingdao 266071, China; China National Tobacco Quality Supervision &Test Center, Zhengzhou 450000, China; Beijing Life Science Academy, Beijing 100000, China; Key Laboratory of Tobacco Biological Effects, Zhengzhou 450000, China
| | - Guanglin Liu
- China National Tobacco Quality Supervision &Test Center, Zhengzhou 450000, China; Beijing Life Science Academy, Beijing 100000, China; Key Laboratory of Tobacco Biological Effects, Zhengzhou 450000, China
| | - Huan Chen
- China National Tobacco Quality Supervision &Test Center, Zhengzhou 450000, China; Beijing Life Science Academy, Beijing 100000, China; Key Laboratory of Tobacco Biological Effects, Zhengzhou 450000, China.
| | - Hongwei Hou
- China National Tobacco Quality Supervision &Test Center, Zhengzhou 450000, China; Beijing Life Science Academy, Beijing 100000, China; Key Laboratory of Tobacco Biological Effects, Zhengzhou 450000, China.
| | - Qingyuan Hu
- China National Tobacco Quality Supervision &Test Center, Zhengzhou 450000, China; Beijing Life Science Academy, Beijing 100000, China; Key Laboratory of Tobacco Biological Effects, Zhengzhou 450000, China
| |
Collapse
|
2
|
Jeong H, Chon Y, Yoon S, Choi EK, Lee N, Oh JK, Chung YA, Song IU. Structural and functional alterations in hypothalamic subregions in male patients with alcohol use disorder. Drug Alcohol Depend 2025; 268:112554. [PMID: 39848134 DOI: 10.1016/j.drugalcdep.2025.112554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 12/29/2024] [Accepted: 01/04/2025] [Indexed: 01/25/2025]
Abstract
BACKGROUND The hypothalamus is involved in stress regulation and reward processing, with its various nuclei exhibiting unique functions and connections. However, human neuroimaging studies on the hypothalamic subregions are limited in drug addiction. This study examined the volumes and functional connectivity of hypothalamic subregions in individuals with alcohol use disorder (AUD). METHOD The study included 24 male patients with AUD who had maintained abstinence and 24 healthy male controls, all of whom underwent brain structural and resting-state functional magnetic resonance imaging. The hypothalamus was segmented into five subunits using a deep learning-based algorithm, with comparisons of volumes and functional connectivity (FC) between the two groups. The relationships between these measures and alcohol-related characteristics were examined in the AUD group. RESULTS Findings indicated lower volumes in the anterior-superior (corrected-p < 0.001) and tuberal-superior subunits (corrected-p = 0.002) and altered FC of these and the anterior-inferior subunit among AUD patients (corrected-p < 0.05). Moreover, greater disease severity and a longer history of heavy drinking correlated with lower volumes in the anterior-superior (r = -0.42, p = 0.045) and tuberal-superior subregions (r = -0.61, p = 0.013), respectively. Conversely, a longer abstinence duration was associated with larger volumes in the anterior-superior (r = 0.56, p = 0.008) and tuberal-superior subunits (r = 0.40, p = 0.048) and with higher FC between the tuberal-superior hypothalamus and the thalamus, caudate, and anterior cingulate cortex (r = 0.55, p = 0.014). CONCLUSIONS Our results suggest that specific regional alterations within the hypothalamus, particularly the superior subregions, are associated with AUD, and more importantly, that these alterations may be reversible with prolonged abstinence.
Collapse
Affiliation(s)
- Hyeonseok Jeong
- Department of Radiology, Incheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea; Department of Neurology, Incheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Younghoon Chon
- Department of Psychiatry, Incheon Chamsarang Hospital, Incheon, Republic of Korea
| | - Sujung Yoon
- Ewha Brain Institute, Ewha W. University, Seoul, Republic of Korea; Department of Brain and Cognitive Sciences, Ewha W. University, Seoul, Republic of Korea
| | - Eun Kyoung Choi
- Department of Nuclear Medicine, Incheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Narae Lee
- Department of Psychiatry, Incheon Chamsarang Hospital, Incheon, Republic of Korea
| | - Jin Kyoung Oh
- Department of Nuclear Medicine, Incheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.
| | - Yong-An Chung
- Department of Nuclear Medicine, Incheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - In-Uk Song
- Department of Neurology, Incheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| |
Collapse
|
3
|
Wilkinson CS, Modrak CG, Thompson TD, Conrad RC, Leon I, Knackstedt LA. Consumption of oxycodone prevents oxytocin from attenuating alcohol intake in rats. Alcohol 2025; 122:43-53. [PMID: 39447851 DOI: 10.1016/j.alcohol.2024.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 08/26/2024] [Accepted: 10/18/2024] [Indexed: 10/26/2024]
Abstract
Alcohol and opioid polysubstance use (PSU) is common and often accompanied by higher trait anxiety. Oxytocin decreases anxiety, alcohol- and opioid-seeking and -taking but has not been assessed in the context of PSU. Here we developed a rat model of sequential oxycodone and alcohol PSU to examine the relationship between anxiety, alcohol and oxycodone intake, and the efficacy of systemic oxytocin to attenuate alcohol intake. Male and female Sprague-Dawley rats were assessed for baseline anxiety-like behavior using acoustic startle and the elevated plus maze (EPM). Rats were then given 2-bottle choice access to oxycodone and/or water for 6-hr/day for 7 days, followed by 2-bottle choice access to alcohol (20% v/v) and/or water for five 24-hr sessions across 10 days. Next, monosubstance (oxycodone- or alcohol-alone) rats continued to have access to only one substance/day while PSU rats had access to oxycodone and water for 3-hr, followed by alcohol and water for 6-hr. After 12 days, rats were tested in the EPM 20 h after alcohol access to examine withdrawal-related anxiety. Next, oxytocin (0, 0.3 or 1.0 mg/kg IP) was administered following the oxycodone/water session, 30 min prior to alcohol access. Rats received intragastric oxycodone (2 mg/kg) or water followed by intragastric alcohol (2 g/kg) and blood was collected to determine blood alcohol levels. Elevated baseline anxiety-like behavior was accompanied by reduced alcohol intake. Consumption of oxycodone did not alter alcohol intake but resulted in less anxiety-like behavior during withdrawal and prevented oxytocin from attenuating alcohol intake. Oxytocin (1 mg/kg) reduced alcohol intake in the alcohol-only condition, an effect that persisted for days after a single oxytocin administration. Rats that received oxycodone prior to non-contingent alcohol displayed higher blood alcohol levels than those that did not. These results support the necessity for the testing of medications for substance use in rodent models of PSU.
Collapse
Affiliation(s)
- C S Wilkinson
- Psychology Department, 945 Center Drive, University of Florida, Gainesville, FL, USA; Center for Addiction Research and Education, 1395 Center Dr, Suite D2-013, University of Florida, Gainesville, FL, USA
| | - C G Modrak
- Psychology Department, 945 Center Drive, University of Florida, Gainesville, FL, USA; Center for Addiction Research and Education, 1395 Center Dr, Suite D2-013, University of Florida, Gainesville, FL, USA
| | - T D Thompson
- Psychology Department, 945 Center Drive, University of Florida, Gainesville, FL, USA
| | - R C Conrad
- Psychology Department, 945 Center Drive, University of Florida, Gainesville, FL, USA
| | - I Leon
- Psychology Department, 945 Center Drive, University of Florida, Gainesville, FL, USA
| | - L A Knackstedt
- Psychology Department, 945 Center Drive, University of Florida, Gainesville, FL, USA; Center for Addiction Research and Education, 1395 Center Dr, Suite D2-013, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
4
|
Scarboro SM, López MF, Becker HC. Role of oxytocin and vasopressin in alcohol use disorder. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2024; 178:23-57. [PMID: 39523056 DOI: 10.1016/bs.irn.2024.08.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Published works highlight the role of neuropeptides in both the development and treatment of AUD. Closely related hypothalamic neuropeptides, oxytocin (OT) and vasopressin (VP), initially recognized for their physiological hormone effects, are increasingly acknowledged for their behavioral influences. Studies consistently demonstrate that OT and VP impact alcohol consumption and related behaviors, implicating them in the neurobiology of addiction. Moreover, stress is a pivotal risk factor for alcohol use and relapse, with OT and VP playing an integral role in the body's stress response system. While previous work has explored the interaction of OT and VP with other substances of abuse, this review focuses on their roles in alcohol-associated behaviors specifically to better understand the role of OT and VP in AUD. Here we synthesize recent preclinical and clinical literature examining changes in OT and VP protein and receptor expression in response to alcohol, as well as research investigating the effects of modulating these systems on alcohol-related behaviors. This review aims to deepen the understanding of OT and VP in the context of AUD with the goal of facilitating future research and enhancing treatment outcomes.
Collapse
Affiliation(s)
- Sutton M Scarboro
- Department of Psychiatry and Behavioral Sciences, Charleston Alcohol Research Center, Medical University of South Carolina, Charleston, SC, United States; Department of Neuroscience, Charleston Alcohol Research Center, Medical University of South Carolina, Charleston, SC, United States
| | - Marcelo F López
- Department of Psychiatry and Behavioral Sciences, Charleston Alcohol Research Center, Medical University of South Carolina, Charleston, SC, United States
| | - Howard C Becker
- Department of Psychiatry and Behavioral Sciences, Charleston Alcohol Research Center, Medical University of South Carolina, Charleston, SC, United States; Department of Neuroscience, Charleston Alcohol Research Center, Medical University of South Carolina, Charleston, SC, United States; RHJ Veterans Administartion Health Care System, Charleston, SC, United States.
| |
Collapse
|
5
|
Kamrani-Sharif R, Hayes AW, Gholami M, Salehirad M, Allahverdikhani M, Motaghinejad M, Emanuele E. Oxytocin as neuro-hormone and neuro-regulator exert neuroprotective properties: A mechanistic graphical review. Neuropeptides 2023; 101:102352. [PMID: 37354708 DOI: 10.1016/j.npep.2023.102352] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 03/28/2023] [Accepted: 06/12/2023] [Indexed: 06/26/2023]
Abstract
BACKGROUND Neurodegeneration is progressive cell loss in specific neuronal populations, often resulting in clinical consequences with significant medical, societal, and economic implications. Because of its antioxidant, anti-inflammatory, and anti-apoptotic properties, oxytocin has been proposed as a potential neuroprotective and neurobehavioral therapeutic agent, including modulating mood disturbances and cognitive enchantment. METHODS Literature searches were conducted using the following databases Web of Science, PubMed, Elsevier Science Direct, Google Scholar, the Core Collection, and Cochrane from January 2000 to February 2023 for articles dealing with oxytocin neuroprotective properties in preventing or treating neurodegenerative disorders and diseases with a focus on oxidative stress, inflammation, and apoptosis/cell death. RESULTS The neuroprotective effects of oxytocin appears to be mediated by its anti-inflammatory properties, inhibition of neuro inflammation, activation of several antioxidant enzymes, inhibition of oxidative stress and free radical formation, activation of free radical scavengers, prevent of mitochondrial dysfunction, and inhibition of apoptosis. CONCLUSION Oxytocin acts as a neuroprotective agent by preventing neuro-apoptosis, neuro-inflammation, and neuronal oxidative stress, and by restoring mitochondrial function.
Collapse
Affiliation(s)
- Roya Kamrani-Sharif
- Chronic Respiratory Disease Research Center (CRDRC), National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - A Wallace Hayes
- University of South Florida College of Public Health, Tampa, FL, USA; Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, USA
| | - Mina Gholami
- Chronic Respiratory Disease Research Center (CRDRC), National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahsa Salehirad
- Cognitive and Neuroscience Research Center (CNRC), Amir-Almomenin Hospital, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Maryam Allahverdikhani
- Chronic Respiratory Disease Research Center (CRDRC), National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Majid Motaghinejad
- Chronic Respiratory Disease Research Center (CRDRC), National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | | |
Collapse
|
6
|
Smoking Is Positively Related and Alcohol Consumption Is Negatively Related to an Increased Risk of Meniere's Disease. J Clin Med 2022; 11:jcm11175007. [PMID: 36078935 PMCID: PMC9457180 DOI: 10.3390/jcm11175007] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/20/2022] [Accepted: 08/24/2022] [Indexed: 12/02/2022] Open
Abstract
A few prior researchers presumed the impacts of smoking and alcohol on the risk of Meniere’s disease (MD). This study investigated the relationship between smoking, alcohol consumption, and obesity with Meniere’s disease in an adult population. The ≥40-year-old population in the Korean National Health Insurance Service-Health Screening Cohort 2002−2019 was analyzed. A total of 15,208 patients with MD were matched with 499,658 comparison participants. The current smoking, alcohol consumption, and past medical histories were collected. Body mass index (BMI) was grouped into underweight, normal, overweight, obese I, and obese II. The odds of histories of smoking and alcohol consumption and the BMI group for MD were analyzed using conditional logistic regression analysis. These associations were further analyzed in subgroups of age, sex, smoking, alcohol consumption, and BMI. In the overall adult population, smoking and alcohol consumption did not show an association with MD. Being underweight was linked with lower odds for MD (adjusted OR [aOR] = 0.80, 95% CI = 0.68−0.93, p = 0.004). In the male group, smoking was positively associated with MD (aOR = 1.08, 95% CI = 1.00−1.17, p = 0.043), while alcohol consumption was negatively related to MD (aOR = 0.87, 95% CI = 0.81−0.94, p < 0.001). Being underweight was related to a lower risk of MD. In adult men, smoking was predicted to increase, while alcohol consumption was predicted to decrease the risk of MD.
Collapse
|
7
|
Towner TT, Papastrat KM, Spear LP, Varlinskaya EI, Werner DF. Impact of adolescent intermittent ethanol exposure in male and female rats on social drinking and neuropeptide gene expression. Alcohol Clin Exp Res 2022; 46:979-993. [PMID: 35470441 DOI: 10.1111/acer.14847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 03/11/2022] [Accepted: 04/15/2022] [Indexed: 11/27/2022]
Abstract
BACKGROUND Alcohol use during adolescence can alter maturational changes that occur in brain regions associated with social and emotional responding. Our previous studies have shown that adult male, but not female rats demonstrate social anxiety-like alterations and enhanced sensitivity to ethanol-induced social facilitation following adolescent intermittent ethanol exposure (AIE). These consequences of AIE may influence adult social drinking in a sex-specific manner. METHODS To test the effects of AIE on social drinking, male and female Sprague-Dawley rats exposed to water or ethanol (0 or 4 g/kg, intragastrically, every other day, between postnatal day [P] 25 and 45) were tested as adults (P72-83) in a social drinking paradigm (30-minute access to a 10% ethanol solution in supersac or supersac alone in groups of three same-sex littermates across two 4-day cycles separated by 4 days off). Social behavior was assessed during the last drinking session, along with assessment of oxytocin (OXT), oxytocin receptor (OXTR), vasopressin (AVP), and vasopressin receptors 1a and 1b (AVPR1a, AVPR1b) in the hypothalamus and lateral septum. RESULTS Males exposed to AIE consumed more ethanol than water-exposed controls during the second drinking cycle, whereas AIE did not affect supersac intake in males. AIE-exposed females consumed less ethanol and more supersac than water-exposed controls. Water-exposed females drinking ethanol showed more social investigation and significantly higher hypothalamic OXTR, AVP, and AVPR1b gene expression than their counterparts ingesting supersac and AIE females drinking ethanol. In males, hypothalamic AVPR1b gene expression was affected by drinking solution, with significantly higher expression evident in males drinking ethanol than those consuming supersac. CONCLUSIONS Collectively, these findings provide new evidence regarding sex-specific effects of AIE on social drinking and suggest that the hypothalamic OXT and AVP systems are implicated in the effects of ingested ethanol on social behavior in a sex- and adolescent-exposure-dependent manner.
Collapse
Affiliation(s)
- Trevor T Towner
- Neurobiology of Adolescent Drinking in Adulthood Consortium, Center for Development and Behavioral Neuroscience, Department of Psychology, Binghamton University, Binghamton, New York, USA
| | - Kimberly M Papastrat
- Neurobiology of Adolescent Drinking in Adulthood Consortium, Center for Development and Behavioral Neuroscience, Department of Psychology, Binghamton University, Binghamton, New York, USA
| | - Linda P Spear
- Neurobiology of Adolescent Drinking in Adulthood Consortium, Center for Development and Behavioral Neuroscience, Department of Psychology, Binghamton University, Binghamton, New York, USA
| | - Elena I Varlinskaya
- Neurobiology of Adolescent Drinking in Adulthood Consortium, Center for Development and Behavioral Neuroscience, Department of Psychology, Binghamton University, Binghamton, New York, USA
| | - David F Werner
- Neurobiology of Adolescent Drinking in Adulthood Consortium, Center for Development and Behavioral Neuroscience, Department of Psychology, Binghamton University, Binghamton, New York, USA
| |
Collapse
|
8
|
Peris J, Totten K, Montgomery D, Lester H, Weatherington A, Piotrowski B, Sowell S, Doyle K, Scott K, Tan Y, MacFadyen KA, Engle H, de Kloet AD, Krause EG. Conditioned social preference and reward value of activating oxytocin-receptor-expressing ventral tegmental area neurons following repeated daily binge ethanol intake. Alcohol Clin Exp Res 2022; 46:194-206. [PMID: 34964139 PMCID: PMC8858886 DOI: 10.1111/acer.14769] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 11/29/2021] [Accepted: 12/22/2021] [Indexed: 02/03/2023]
Abstract
BACKGROUND Individuals with alcohol use disorder (AUD) exhibit a disruption of social behavior and dysregulation of oxytocin signaling in the brain, possibly reflecting decreased activation of oxytocin receptors (OxTRs) in reward pathways in response to social stimuli. We hypothesize that daily binge ethanol intake causes a deficit in social reward and oxytocin signaling in the ventral tegmental area (VTA). METHODS After 9 weeks of daily binge ethanol intake (blood ethanol concentration >80 mg%), OxTR-cre mice underwent conditioned place preference for social reward. Separate groups of mice were tested for the effects of binge ethanol on voluntary social interactions, food reward, locomotion, and anxiety-like behaviors. A subset of mice underwent transfection of OxTR-expressing VTA neurons (VTAOxtr ) with a light-sensitive opsin, followed by operant training to respond to light delivered to VTA. RESULTS Ethanol-naïve male mice increased the time spent on the side previously paired with novel mice while ethanol-treated mice did not. Binge ethanol did not affect conditioned place preference for food reward in males, but this response was weakened in ethanol-treated females. Ethanol treatment also caused a sex-specific impairment of voluntary social interactions with novel mice. There were minimal differences between groups in measures of anxiety and locomotion. Ethanol-naïve mice had significantly greater operant responding for activation of VTAOxtr than sham-transfected mice but ethanol-treated mice did not. There was no difference in the number of VTAOxtr after binge ethanol. CONCLUSIONS Daily binge ethanol causes social reward deficits that cannot be explained by nonspecific effects on other behaviors, at least in males. Only ethanol-naïve mice exhibited positive reinforcement caused by activation of VTAOxtr while daily binge ethanol did not alter the number of VTAOxtr in either males or females. Thus, subtle dysregulation of VTAOxtr function may be related to the social reward deficits caused by daily binge ethanol.
Collapse
Affiliation(s)
- Joanna Peris
- University of Florida, Department of Pharmacodynamics, Gainesville FL 32610 USA
| | - Katye Totten
- University of Florida, Department of Pharmacodynamics, Gainesville FL 32610 USA
| | - Darrice Montgomery
- University of Florida, Department of Pharmacodynamics, Gainesville FL 32610 USA
| | - Hannah Lester
- University of Florida, Department of Pharmacodynamics, Gainesville FL 32610 USA
| | | | - Brian Piotrowski
- University of Florida, Department of Pharmacodynamics, Gainesville FL 32610 USA
| | - Sam Sowell
- University of Florida, Department of Pharmacodynamics, Gainesville FL 32610 USA
| | - Kristen Doyle
- University of Florida, Department of Pharmacodynamics, Gainesville FL 32610 USA
| | - Karen Scott
- University of Florida, Department of Pharmacodynamics, Gainesville FL 32610 USA
| | - Yalun Tan
- University of Florida, Department of Pharmacodynamics, Gainesville FL 32610 USA
| | - Kaley A. MacFadyen
- University of Florida, Department of Pharmacodynamics, Gainesville FL 32610 USA
| | - Hannah Engle
- University of Florida, Department of Pharmacodynamics, Gainesville FL 32610 USA
| | | | - Eric G. Krause
- University of Florida, Department of Pharmacodynamics, Gainesville FL 32610 USA
| |
Collapse
|
9
|
King CE, Griffin WC, Lopez MF, Becker HC. Activation of hypothalamic oxytocin neurons reduces binge-like alcohol drinking through signaling at central oxytocin receptors. Neuropsychopharmacology 2021; 46:1950-1957. [PMID: 34127796 PMCID: PMC8429589 DOI: 10.1038/s41386-021-01046-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 05/04/2021] [Accepted: 05/18/2021] [Indexed: 02/05/2023]
Abstract
Preclinical and clinical evidence suggests that exogenous administration of oxytocin (OT) may hold promise as a therapeutic strategy for reducing heavy alcohol drinking. However, it remains unknown whether these effects are mediated by stimulation of endogenous sources of OT and signaling at oxytocin receptors (OTR) in brain or in the periphery. To address this question, we employed a targeted chemogenetic approach to examine whether selective activation of OT-containing neurons in the paraventricular nucleus of the hypothalamus (PVN) alters alcohol consumption in a binge-like drinking ("Drinking-in-the-Dark"; DID) model. Adult male Oxt-IRES-Cre mice received bilateral infusion of a Cre-dependent virus containing an excitatory DREADD (AAV8-hSyn-DIO-hM3Dq-mCherry) or control virus (AAV8-hSyn-DIO-mCherry) into the PVN. Chemogenetic activation of PVNOT+ neurons following clozapine-N-oxide injection reduced binge-like alcohol drinking in a similar manner as systemic administration of the neuropeptide. Pretreatment with a brain-penetrant OTR antagonist (L-368,899) reversed this effect while systemic administration of a peripherally restricted OTR antagonist (Atosiban) did not alter reduced alcohol drinking following chemogenetic activation of PVNOT+ neurons. Altogether, these data are the first to demonstrate that targeted activation of hypothalamic (endogenous) OT reduces alcohol consumption, providing further evidence that this neuropeptide plays a role in regulation of alcohol self-administration behavior. Further, results indicate that the ability OT to reduce alcohol drinking is mediated by signaling at OTR in the brain.
Collapse
Affiliation(s)
- Courtney E King
- Charleston Alcohol Research Center, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - William C Griffin
- Charleston Alcohol Research Center, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Marcelo F Lopez
- Charleston Alcohol Research Center, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Howard C Becker
- Charleston Alcohol Research Center, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, USA.
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA.
- RHJ Department of Veterans Affairs Medical Center, Charleston, SC, USA.
| |
Collapse
|
10
|
Alcohol and oxytocin: Scrutinizing the relationship. Neurosci Biobehav Rev 2021; 127:852-864. [PMID: 34102150 DOI: 10.1016/j.neubiorev.2021.06.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 05/19/2021] [Accepted: 06/03/2021] [Indexed: 02/07/2023]
Abstract
The initial enthusiasm towards oxytocin (OXT) as a potential treatment for alcohol use disorder has been recently tempered by recognizing existing gaps in literature and the recent appearance of a relatively small number of clinical studies with negative outcomes. On the other hand, several new studies continue to support the OXT system's potential for such treatment. In this review, we thoroughly analyze existing literature assessing both alcohol's effects on the OXT system and OXT's effects on alcohol-related behaviors. Both rodent and clinical research is discussed. We identify areas that have been studied extensively and those that have been undeservingly understudied. OXT's potential effects on tolerance, withdrawal, craving, anxiety and social behaviors, and how these processes ultimately affect alcohol consumption, are critically explored. We conclude that while OXT can affect alcohol consumption in males and females, more comprehensive studies on OXT's effects on alcohol-related tolerance, withdrawal, craving, anxiety and social affiliations in subjects of both sexes and across several levels of analyses are needed.
Collapse
|
11
|
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. [PMID: 33600854 DOI: 10.1016/j.pharmthera.2021.107820] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/15/2021] [Indexed: 12/13/2022]
Abstract
Drug addiction is one of the leading causes of mortality worldwide. Despite great advances were achieved in understanding the neurobiology of drug addiction, the therapeutic options are severely limited, with poor effectiveness and serious side effects. The neuropeptide oxytocin (OXT) is well known for its effects on uterine contraction, sexual/maternal behaviors, social affiliation, stress and learning/memory by interacting with the OXT receptor and other neuromodulators. Emerging evidence suggests that the acute or chronic exposure to drugs can affect the OXT system. Additionally, OXT administration can ameliorate a wide range of abused drug-induced neurobehavioral changes. Overall, OXT not only suppresses drug reward in the binge stage of drug addiction, but also reduces stress responses and social impairments during the withdrawal stage and, finally, prevents drug/cue/stress-induced reinstatement. More importantly, clinical studies have also shown that OXT can exert beneficial effects on reducing substance use disorders of a series of drugs, such as heroin, cocaine, alcohol, cannabis and nicotine. Thus, the present review focuses on the role of OXT in treating drug addiction, including the preclinical and clinical therapeutic potential of OXT and its analogs on the neurobiological perspectives of drugs, to provide a better insight of the efficacy of OXT as a clinical addiction therapeutic agent.
Collapse
Affiliation(s)
- Xiaohang Che
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, PR China; Key Laboratory of New Drug Screening of Liaoning Province, Shenyang Pharmaceutical University, Shenyang, PR China; Key Laboratory of New Drug Pharmacodynamics Evaluation of Liaoning Province, Shenyang Pharmaceutical University, Shenyang, PR China
| | - Jialing Cai
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, PR China
| | - Yueyang Liu
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, PR China; Key Laboratory of New Drug Screening of Liaoning Province, Shenyang Pharmaceutical University, Shenyang, PR China; Key Laboratory of New Drug Pharmacodynamics Evaluation of Liaoning Province, Shenyang Pharmaceutical University, Shenyang, PR China
| | - Tianyu Xu
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, PR China
| | - Jingyu Yang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, PR China; Key Laboratory of New Drug Screening of Liaoning Province, Shenyang Pharmaceutical University, Shenyang, PR China; Key Laboratory of New Drug Pharmacodynamics Evaluation of Liaoning Province, Shenyang Pharmaceutical University, Shenyang, PR China.
| | - Chunfu Wu
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, PR China; Key Laboratory of New Drug Screening of Liaoning Province, Shenyang Pharmaceutical University, Shenyang, PR China; Key Laboratory of New Drug Pharmacodynamics Evaluation of Liaoning Province, Shenyang Pharmaceutical University, Shenyang, PR China.
| |
Collapse
|
12
|
Salles J, Lacassagne E, Eddiry S, Franchitto N, Salles JP, Tauber M. What can we learn from PWS and SNORD116 genes about the pathophysiology of addictive disorders? Mol Psychiatry 2021; 26:51-59. [PMID: 33082508 DOI: 10.1038/s41380-020-00917-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 09/16/2020] [Accepted: 10/05/2020] [Indexed: 02/07/2023]
Abstract
Addictive disorders have been much investigated and many studies have underlined the role of environmental factors such as social interaction in the vulnerability to and maintenance of addictive behaviors. Research on addiction pathophysiology now suggests that certain behavioral disorders are addictive, one example being food addiction. Yet, despite the growing body of knowledge on addiction, it is still unknown why only some of the individuals exposed to a drug become addicted to it. This observation has prompted the consideration of genetic heritage, neurodevelopmental trajectories, and gene-environment interactions in addiction vulnerability. Prader-Willi syndrome (PWS) is a rare neurodevelopmental disorder in which children become addicted to food and show early social impairment. PWS is caused by the deficiency of imprinted genes located on the 15q11-q13 chromosome. Among them, the SNORD116 gene was identified as the minimal gene responsible for the PWS phenotype. Several studies have also indicated the role of the Snord116 gene in animal and cellular models to explain PWS pathophysiology and phenotype (including social impairment and food addiction). We thus present here the evidence suggesting the potential involvement of the SNORD116 gene in addictive disorders.
Collapse
Affiliation(s)
- Juliette Salles
- Université de Toulouse III, F-31000, Toulouse, France.,CHU de Toulouse, Service de psychiatrie et psychologie, psychiatrie Toulouse, F-31000, Toulouse, France.,Inserm Unité 1043, CNRS 5828, Université Paul Sabatier, Toulouse III, F-31000, Toulouse, France.,CHU de Toulouse, Institut des Handicaps Neurologiques, Psychiatriques et Sensoriels, F-31000, Toulouse, France
| | - Emmanuelle Lacassagne
- Inserm Unité 1043, CNRS 5828, Université Paul Sabatier, Toulouse III, F-31000, Toulouse, France
| | - Sanaa Eddiry
- Inserm Unité 1043, CNRS 5828, Université Paul Sabatier, Toulouse III, F-31000, Toulouse, France
| | - Nicolas Franchitto
- Université de Toulouse III, F-31000, Toulouse, France.,CHU de Toulouse, Service d'addictologie clinique, urgences réanimation médecine, F-31000, Toulouse, France
| | - Jean-Pierre Salles
- Inserm Unité 1043, CNRS 5828, Université Paul Sabatier, Toulouse III, F-31000, Toulouse, France
| | - Maithé Tauber
- Université de Toulouse III, F-31000, Toulouse, France. .,Inserm Unité 1043, CNRS 5828, Université Paul Sabatier, Toulouse III, F-31000, Toulouse, France. .,CHU de Toulouse, Institut des Handicaps Neurologiques, Psychiatriques et Sensoriels, F-31000, Toulouse, France. .,CHU de Toulouse, Centre de référence du Syndrome de Prader-Willi et autres syndromes avec troubles du comportement alimentaire, Unité d'endocrinologie, obésités, maladies osseuses, génétique et gynécologie médicale, F-31000, Toulouse, France.
| |
Collapse
|
13
|
Rodriguez KM, Smith BL, Caldwell HK. Voluntary alcohol consumption is increased in female, but not male, oxytocin receptor knockout mice. Brain Behav 2020; 10:e01749. [PMID: 32666677 PMCID: PMC7507036 DOI: 10.1002/brb3.1749] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 06/17/2020] [Accepted: 06/19/2020] [Indexed: 01/30/2023] Open
Abstract
INTRODUCTION The oxytocin (Oxt) system, while typically associated with the neural regulation of social behaviors, also plays a role in an individual's vulnerability to develop alcohol use disorders (AUD). In humans, changes to the Oxt system, due to early life experience and/or genetic mutations, are associated with increased vulnerability to AUD. While a considerable amount is known about Oxt's role in AUD in males, less is known or understood, about how Oxt may affect AUD in females, likely due to many clinical and preclinical studies of AUD not directly considering sex as a biological variable. This is unfortunate given that females are more vulnerable to the effects of alcohol and have increased alcohol consumption, as compared to males. Therefore, in the current study we wanted to determine whether genetic disruption of the Oxt receptor (Oxtr), that is, Oxtr knockout (-/-) mice, affected stress-induced alcohol consumption in males and females. We hypothesized that genetic disruption of the Oxtr would result in increased stress-induced alcohol consumption in both males and females compared to wild-type (+/+) controls. Though, we predicted that these disruptions might be greater in female Oxtr -/- mice. METHODS To test this hypothesis, a two-bottle preference test was utilized along with the forced swim test (FST), and pre- and poststress alcohol consumption and preference measured within each sex (males and females were run separately). As a follow-up experiment, a taste preference test, to control for possible genotypic differences in taste, was also performed. RESULTS In males, we found no significant genotypic differences in alcohol consumption or preference. However, in females, we found that genetic disruption of the Oxtr resulted in a greater consumption of alcohol both pre- and poststress compared to controls. CONCLUSION These data suggest that in females, disruptions in Oxt signaling may contribute to increased vulnerability to alcohol-associated addiction.
Collapse
Affiliation(s)
- Karla M Rodriguez
- School of Biomedical Sciences and the Brain Health Research Institute, Kent State University, Kent, OH, USA.,Laboratory of Neuroendocrinology and Behavior, Department of Biological Sciences, Kent State University, Kent, OH, USA
| | - Brittany L Smith
- Laboratory of Neuroendocrinology and Behavior, Department of Biological Sciences, Kent State University, Kent, OH, USA.,Department of Pharmacology & Systems Physiology, University of Cincinnati, Cincinnati, OH, USA
| | - Heather K Caldwell
- School of Biomedical Sciences and the Brain Health Research Institute, Kent State University, Kent, OH, USA.,Laboratory of Neuroendocrinology and Behavior, Department of Biological Sciences, Kent State University, Kent, OH, USA
| |
Collapse
|
14
|
Vena AA, Zandy SL, Cofresí RU, Gonzales RA. Behavioral, neurobiological, and neurochemical mechanisms of ethanol self-administration: A translational review. Pharmacol Ther 2020; 212:107573. [PMID: 32437827 PMCID: PMC7580704 DOI: 10.1016/j.pharmthera.2020.107573] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/29/2020] [Indexed: 12/16/2022]
Abstract
Alcohol use disorder has multiple characteristics including excessive ethanol consumption, impaired control over drinking behaviors, craving and withdrawal symptoms, compulsive seeking behaviors, and is considered a chronic condition. Relapse is common. Determining the neurobiological targets of ethanol and the adaptations induced by chronic ethanol exposure is critical to understanding the clinical manifestation of alcohol use disorders, the mechanisms underlying the various features of the disorder, and for informing medication development. In the present review, we discuss ethanol's interactions with a variety of neurotransmitter systems, summarizing findings from preclinical and translational studies to highlight recent progress in the field. We then describe animal models of ethanol self-administration, emphasizing the value, limitations, and validity of commonly used models. Lastly, we summarize the behavioral changes induced by chronic ethanol self-administration, with an emphasis on cue-elicited behavior, the role of ethanol-related memories, and the emergence of habitual ethanol seeking behavior.
Collapse
Affiliation(s)
- Ashley A Vena
- Department of Psychiatry and Behavioral Neuroscience, University of Chicago, United States of America
| | | | - Roberto U Cofresí
- Psychological Sciences, University of Missouri, United States of America
| | - Rueben A Gonzales
- Division of Pharmacology and Toxicology, College of Pharmacy and Institute for Neuroscience, The University of Texas at Austin, United States of America.
| |
Collapse
|
15
|
King CE, Gano A, Becker HC. The role of oxytocin in alcohol and drug abuse. Brain Res 2020; 1736:146761. [PMID: 32142721 PMCID: PMC7137097 DOI: 10.1016/j.brainres.2020.146761] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 02/28/2020] [Accepted: 03/02/2020] [Indexed: 02/07/2023]
Abstract
The neuropeptide oxytocin (OXT) plays a key role in adaptive processes associated with reward, tolerance, memory and stress responses. Through interactions with brain reward and stress systems, OXT is known to play a role in several neuropsychiatric disorders, particularly those that involve altered social integration, such as alcohol and drug addiction (Heilig et al., 2016). As such, there is growing interest in the oxytocin system as a potential therapeutic target for the treatment of alcohol and substance use disorders. Accumulating preclinical evidence suggests that administration of OXT influences the development of tolerance, sensitization and withdrawal symptoms, and modulates numerous alcohol/drug-seeking and alcohol/drug-taking behaviors. Further, there is some evidence to suggest that OXT may help to reverse neuroadaptations that occur as a result of chronic alcohol or drug exposure. To date, there have been only a handful of clinical studies conducted in alcohol and drug dependent populations. This review summarizes the preclinical and clinical literature on the effects of OXT administration on alcohol- and drug-related behaviors. In addition, we discuss OXT interactions with the hypothalamic-pituitaryadrenal axis and multiple neurotransmitter systems within addiction circuitry.
Collapse
Affiliation(s)
- Courtney E King
- Charleston Alcohol Research Center, Department of Psychiatry and Neuroscience, Medical University of South Carolina & VAMC, Charleston, SC 29425, United States
| | - Anny Gano
- Charleston Alcohol Research Center, Department of Psychiatry and Neuroscience, Medical University of South Carolina & VAMC, Charleston, SC 29425, United States
| | - Howard C Becker
- Charleston Alcohol Research Center, Department of Psychiatry and Neuroscience, Medical University of South Carolina & VAMC, Charleston, SC 29425, United States.
| |
Collapse
|
16
|
Peris J, Steck MR, Krause EG. Oxytocin treatment for alcoholism: Potential neurocircuitry targets. Neuropharmacology 2020; 171:108091. [PMID: 32304701 DOI: 10.1016/j.neuropharm.2020.108091] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Revised: 03/19/2020] [Accepted: 04/03/2020] [Indexed: 12/19/2022]
Abstract
Oxytocin (OT) has gained considerable interest in recent years as a potential treatment for alcoholism and other substance use disorders. Evidence continues to mount that OT administered either centrally, peripherally or intranasally can decrease ethanol intake in both humans and animal models. The potential mechanisms for the ability of OT to decrease ethanol reward, and importantly, cue- and stress-induced ethanol relapse, are explored by reviewing the specific neuronal circuits involved in mediating these actions and their sensitivity to OT. In addition to dopamine neurons that project from ventral tegmental area (VTA) to nucleus accumbens (NAc) to signal positively reinforcing events, OT receptors (OxTR) are also expressed by dopamine neurons that project from VTA to brain regions that can convey aversive properties of a stimulus. Moreover, OxTR are expressed by non-dopaminergic neurons in the VTA, such as GABA and glutamate neurons, which can both modulate the activity of dopamine VTA neurons locally (in opposite directions) or can project to other brain regions, including the NAc, where it can alter either positive reinforcement or aversion caused by ethanol. The ability of OT to regulate limbic circuitry and the hypothalamic-pituitary-adrenal axis is discussed as a potential mechanism for the ability of OT to inhibit ethanol-induced negative reinforcement. Together, understanding the diversity and complexity of OT regulation of ethanol reward may contribute to more effective use of OT as pharmacotherapy for alcohol use disorder. This article is part of the special issue on Neuropeptides.
Collapse
Affiliation(s)
- Joanna Peris
- Department of Pharmacodynamics, University of Florida, Gainesville, FL, 32610, USA.
| | - Madeline R Steck
- Department of Pharmacodynamics, University of Florida, Gainesville, FL, 32610, USA
| | - Eric G Krause
- Department of Pharmacodynamics, University of Florida, Gainesville, FL, 32610, USA
| |
Collapse
|
17
|
Walcott AT, Ryabinin AE. Effects of Alcohol Consumption on Pair Bond Maintenance and Potential Neural Substrates in Female Prairie Voles. Alcohol Alcohol 2020; 54:353-360. [PMID: 31062856 DOI: 10.1093/alcalc/agz041] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 04/09/2019] [Accepted: 04/15/2019] [Indexed: 12/19/2022] Open
Abstract
AIMS Discordant heavy alcohol use is a risk factor for disruption of intimate partner relationships. Modeling these relationships in prairie voles indicates that biological effects of alcohol can contribute to this risk. In particular, alcohol consumption disrupted an established preference for a female partner in male prairie voles if the partner was drinking water, but not if the partner was drinking alcohol. The current study investigated the effects of alcohol consumption on pair bonds in female prairie voles. METHODS Female and male prairie voles established pair bonds during 1 week of cohabitation. Following cohabitation, females and their partners were put into mesh-divided cages where they were given access to 10% ethanol and water or only water for 1 week. Pair bonds in female prairie voles were tested using the partner preference test (PPT). Following the PPT, we examined oxytocin, vasopressin and FosB immunoreactivity across several brain regions. RESULTS Female prairie voles consumed more alcohol if their male partner was also drinking alcohol, but not if their partner was drinking water. During PPT, females preferred their partner over a stranger, regardless of their partner's drinking status. Alcohol consumption decreased oxytocin immunoreactivity in the paraventricular nucleus of the hypothalamus and increased FosB immunoreactivity in the centrally projecting Edinger-Westphal nucleus. CONCLUSIONS Established partner preference in female prairie voles is resistant to alcohol consumption. This finding suggests that the risk for disruption of intimate partner relationships in females is not mediated by a decreased motivation to be with their partners.
Collapse
Affiliation(s)
- Andre T Walcott
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, USA
| | - Andrey E Ryabinin
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
18
|
Stauffer CS, Meinzer NK, Morrison T, Wen JH, Radanovich L, Leung D, Niles A, O'Donovan A, Batki SL, Woolley JD. Effects of Oxytocin Administration on Cue-Induced Craving in Co-occurring Alcohol Use Disorder and PTSD: A Within-Participant Randomized Clinical Trial. Alcohol Clin Exp Res 2019; 43:2627-2636. [PMID: 31610033 PMCID: PMC7450809 DOI: 10.1111/acer.14217] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 10/07/2019] [Indexed: 12/30/2022]
Abstract
BACKGROUND Individuals with alcohol use disorder (AUD) are much more likely to meet criteria for posttraumatic stress disorder (PTSD) than the general population. Compared to AUD alone, those with comorbid AUD-PTSD experience worse outcomes. Prior literature suggests that oxytocin, a hypothalamic neuropeptide, may be effective in the treatment of both AUD and PTSD when administered intranasally, although specific mechanisms remain elusive. METHODS Forty-seven male patients with comorbid AUD-PTSD were administered intranasal oxytocin in a randomized, double-blind, dose-ranging (20 IU, 40 IU, and matched placebo), within-participant design with study visits at least 1 week apart. A cue-induced craving paradigm was conducted using each participant's preferred alcoholic beverage versus a neutral water cue. Self-reported alcohol craving and heart rate (HR) were recorded and analyzed using linear mixed-effect models. RESULTS While alcohol cues significantly induced self-reported craving and increased HR compared to neutral water cues, neither dosage of oxytocin compared to placebo reduced self-reported cue-induced alcohol craving nor cue-induced changes in HR in patients with PTSD-AUD. CONCLUSIONS These preliminary findings suggest that oxytocin does not affect cue-induced craving. Our results contribute to an ever-growing field of research investigating the effects of intranasal oxytocin on the symptoms of substance use disorders and will help further refine methodology and streamline future inquiries in this area.
Collapse
Affiliation(s)
- Christopher S Stauffer
- From the, San Francisco Veterans Affairs Medical Center, University of California, San Francisco, San Francisco, California
| | | | - Tyler Morrison
- University of California, San Francisco, San Francisco, California
| | - Jin-Hui Wen
- University of British Columbia, Vancouver, British Columbia
| | - Lily Radanovich
- San Francisco Veterans Affairs Medical Center, San Francisco, California
| | - David Leung
- San Francisco Veterans Affairs Medical Center, San Francisco, California
| | | | - Aoife O'Donovan
- San Francisco Veterans Affairs Medical Center, San Francisco, California
| | - Steven L Batki
- From the, San Francisco Veterans Affairs Medical Center, University of California, San Francisco, San Francisco, California
| | - Joshua D Woolley
- From the, San Francisco Veterans Affairs Medical Center, University of California, San Francisco, San Francisco, California
| |
Collapse
|
19
|
Zhou Y, Liang Y, Low MJ, Kreek MJ. Nuclear transcriptional changes in hypothalamus of Pomc enhancer knockout mice after excessive alcohol drinking. GENES BRAIN AND BEHAVIOR 2019; 18:e12600. [PMID: 31339663 DOI: 10.1111/gbb.12600] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 07/13/2019] [Accepted: 07/19/2019] [Indexed: 12/13/2022]
Abstract
Persistent alterations of proopiomelanocortin (Pomc) and mu-opioid receptor (Oprm1) activity and stress responses after alcohol are critically involved in vulnerability to alcohol dependency. Gene transcriptional regulation altered by alcohol may play important roles. Mice with genome-wide deletion of neuronal Pomc enhancer1 (nPE1-/- ), had hypothalamic-specific partial reductions of beta-endorphin and displayed lower alcohol consumption, compared to wildtype littermates (nPE1+/+ ). We used RNA-Seq to measure steady-state nuclear mRNA transcripts of opioid and stress genes in hypothalamus of nPE1+/+ and nPE1-/- mice after 1-day acute withdrawal from chronic excessive alcohol drinking or after water. nPE1-/- had lower basal Pomc and Pdyn (prodynorphin) levels compared to nPE1+/+ , coupled with increased basal Oprm1 and Oprk1 (kappa-opioid receptor) levels, and low alcohol drinking increased Pomc and Pdyn to the basal levels of nPE1+/+ in the water group, without significant effects on Oprm1 and Oprk1. In nPE1+/+ , excessive alcohol intake increased Pomc and Oprm1, with no effect on Pdyn or Oprk1. For stress genes, nPE1-/- had lowered basal Oxt (oxytocin) and Avp (arginine vasopressin) that were restored by low alcohol intake to basal levels of nPE1+/+ . In nPE1+/+ , excessive alcohol intake decreased Oxt and Avpi1 (AVP-induced protein1). Functionally examining the effect of pharmacological blockade of mu-opioid receptor, we found that naltrexone reduced excessive alcohol intake in nPE1+/+ , but not nPE1-/- . Our results provide evidence relevant to the transcriptional profiling of the critical genes in mouse hypothalamus: enhanced opioid and reduced stress gene transcripts after acute withdrawal from excessive alcohol may contribute to altered reward and stress responses.
Collapse
Affiliation(s)
- Yan Zhou
- Laboratory of the Biology of Addictive Diseases, The Rockefeller University, New York, New York
| | - Yupu Liang
- Research Bioinformatics, CCTS, The Rockefeller University, New York, New York
| | - Malcolm J Low
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Mary J Kreek
- Laboratory of the Biology of Addictive Diseases, The Rockefeller University, New York, New York
| |
Collapse
|
20
|
Abstract
The neuropeptide Oxytocin (ΟΤ) is involved as a neurohormone, a neurotransmitter, or a neuromodulator in an extensive range of central and peripheral effects, complex emotional and social human behaviors, memory and learning processes. It is implicated in homeostatic, neuroadaptive processes associated with stress responses and substance use via interactions with the hypothalamic-pituitary-adrenal (HPA) axis and the dopamine mesolimbic reward stress system. This chapter reviews the preclinical and clinical literature on the complicated relationships between endogenous and exogenous opioids and ΟΤ systems and attempts to highlight key findings to date on the effectiveness of intranasal OT administration to treat opioid use disorders. OΤ seems to attenuate, even inhibit, the development of opioid use disorders in preclinical models but is still under clinical research as a promising pharmacological agent in the treatment of opioid use related behaviors. Evidence suggests a role for OT as an adjunctive or stand-alone treatment of behavioral, cognitive and emotional deficits associated with substance use, which may be responsible for seeking behavior and relapse. The mechanisms by which oxytocin acts to reverse the neural substrates of these deficits, partially due to substance induced alterations of the endogenous OT system, and thus modify the behavioral response to substance use are discussed. Other clinically relevant issues are also discussed.
Collapse
|
21
|
Skinner JA, Campbell EJ, Dayas CV, Garg ML, Burrows TL. The relationship between oxytocin, dietary intake and feeding: A systematic review and meta-analysis of studies in mice and rats. Front Neuroendocrinol 2019; 52:65-78. [PMID: 30315826 DOI: 10.1016/j.yfrne.2018.09.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 09/13/2018] [Accepted: 09/28/2018] [Indexed: 01/11/2023]
Abstract
The neuropeptide oxytocin has been associated with food intake and feeding behaviour. This systematic review aimed to investigate the impact of oxytocin on dietary intake and feeding behaviour in rodent studies. Six electronic databases were searched to identify published studies to April 2018. Preclinical studies in mice and rats were included if they reported: (1) a dietary measure (i.e. food or nutrient and/or behaviour (2) an oxytocin measure, and (3) relationship between the two measures. A total of 75 articles (n = 246 experiments) were included, and study quality appraised. The majority of studies were carried out in males (87%). The top three oxytocin outcomes assessed were: exogenous oxytocin administration (n = 126), oxytocin-receptor antagonist administration (n = 46) and oxytocin gene deletion (n = 29). Meta-analysis of exogenous studies in mice (3 studies, n = 43 comparisons) and rats (n = 8 studies, n = 82 comparisons) showed an overall decrease in food intake with maximum effect shown at 2 h post-administration.
Collapse
Affiliation(s)
- Janelle A Skinner
- Nutrition and Dietetics, School of Health Sciences, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW 2308, Australia; Priority Research Centre for Physical Activity and Nutrition, University of Newcastle, Callaghan, NSW 2308, Australia.
| | - Erin J Campbell
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria 3052, Australia; Florey Department of Neuroscience and Mental Health, University of Melbourne, Victoria 3010, Australia.
| | - Christopher V Dayas
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW 2308, Australia.
| | - Manohar L Garg
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW 2308, Australia.
| | - Tracy L Burrows
- Nutrition and Dietetics, School of Health Sciences, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW 2308, Australia; Priority Research Centre for Physical Activity and Nutrition, University of Newcastle, Callaghan, NSW 2308, Australia.
| |
Collapse
|
22
|
Zanos P, Keyworth H, Georgiou P, Hambsch B, Otte DM, Kitchen I, Zimmer A, Bailey A. Chronic nicotine administration restores brain region specific upregulation of oxytocin receptor binding levels in a G72 mouse model of schizophrenia. Eur J Neurosci 2018; 50:2255-2263. [DOI: 10.1111/ejn.14155] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Revised: 08/07/2018] [Accepted: 08/26/2018] [Indexed: 12/13/2022]
Affiliation(s)
- Panos Zanos
- Department of Biochemistry and Physiology Faculty of Health and Medical Sciences University of Surrey Guildford Surrey UK
- Department of Psychiatry School of Medicine University of Maryland Baltimore Maryland USA
| | - Helen Keyworth
- Department of Biochemistry and Physiology Faculty of Health and Medical Sciences University of Surrey Guildford Surrey UK
| | - Polymnia Georgiou
- Department of Biochemistry and Physiology Faculty of Health and Medical Sciences University of Surrey Guildford Surrey UK
- Department of Psychiatry School of Medicine University of Maryland Baltimore Maryland USA
| | - Boris Hambsch
- GKM Gesellschaft für Therapieforschung mbH Lessingstraße München Germany
| | - David M. Otte
- Institute for Molecular Psychiatry Medical Faculty University of Bonn Bonn Germany
| | - Ian Kitchen
- Department of Biochemistry and Physiology Faculty of Health and Medical Sciences University of Surrey Guildford Surrey UK
| | - Andreas Zimmer
- Institute for Molecular Psychiatry Medical Faculty University of Bonn Bonn Germany
| | - Alexis Bailey
- Department of Biochemistry and Physiology Faculty of Health and Medical Sciences University of Surrey Guildford Surrey UK
- Institute of Medical and Biomedical Education St George's University of London London SW17 0RE UK
| |
Collapse
|
23
|
Zhou Y, Kreek MJ. Involvement of Activated Brain Stress Responsive Systems in Excessive and "Relapse" Alcohol Drinking in Rodent Models: Implications for Therapeutics. J Pharmacol Exp Ther 2018; 366:9-20. [PMID: 29669731 PMCID: PMC5988024 DOI: 10.1124/jpet.117.245621] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 04/16/2018] [Indexed: 02/06/2023] Open
Abstract
Addictive diseases, including addiction to alcohol, pose massive public health costs. Addiction is a chronic relapsing disease caused by both the direct effects induced by drugs and persistent neuroadaptations at the molecular, cellular, and behavioral levels. These drug-type specific neuroadaptations are brought on largely by the reinforcing effects of drugs on the central nervous system and environmental stressors. Results from animal experiments have demonstrated important interactions between alcohol and stress-responsive systems. Addiction to specific drugs such as alcohol, psychostimulants, and opioids shares some common direct or downstream effects on the brain's stress-responsive systems, including arginine vasopressin and its V1b receptors, dynorphin and the κ-opioid receptors, pro-opiomelanocortin/β-endorphin and the μ-opioid receptors, and the endocannabinoids. Further study of these systems through laboratory-based and translational research could lead to the discovery of novel treatment targets and the early optimization of interventions (for example, combination) for the pharmacologic therapy of alcoholism.
Collapse
Affiliation(s)
- Yan Zhou
- Laboratory of Biology of Addictive Diseases, Rockefeller University, New York, New York
| | - Mary Jeanne Kreek
- Laboratory of Biology of Addictive Diseases, Rockefeller University, New York, New York
| |
Collapse
|
24
|
Silva CP, Horton WJ, Caruso MJ, Sebastian A, Klein LC, Albert I, Kamens HM. The influence of adolescent nicotine exposure on ethanol intake and brain gene expression. PLoS One 2018; 13:e0198935. [PMID: 29912970 PMCID: PMC6005571 DOI: 10.1371/journal.pone.0198935] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 05/29/2018] [Indexed: 01/14/2023] Open
Abstract
Nicotine and alcohol are often co-abused. Adolescence is a vulnerable period for the initiation of both nicotine and alcohol use, which can lead to subsequent neurodevelopmental and behavioral alterations. It is possible that during this vulnerable period, use of one drug leads to neurobiological alterations that affect subsequent consumption of the other drug. The aim of the present study was to determine the effect of nicotine exposure during adolescence on ethanol intake, and the effect of these substances on brain gene expression. Forty-three adolescent female C57BL/6J mice were assigned to four groups. In the first phase of the experiment, adolescent mice (PND 36-41 days) were exposed to three bottles filled with water or nicotine (200 μg/ml) for 22 h a day and a single bottle of water 2 h a day for six days. In the second phase (PND 42-45 days), the 4-day Drinking-in-the-Dark paradigm consisting of access to 20% v/v ethanol or water for 2h or 4h (the last day) was overlaid during the time when the mice did not have nicotine available. Ethanol consumption (g/kg) and blood ethanol concentrations (BEC, mg %) were measured on the final day and whole brains including the cerebellum, were dissected for RNA sequencing. Differentially expressed genes (DEG) were detected with CuffDiff and gene networks were built using WGCNA. Prior nicotine exposure increased ethanol consumption and resulting BEC. Significant DEG and biological pathways found in the group exposed to both nicotine and ethanol included genes important in stress-related neuropeptide signaling, hypothalamic-pituitary-adrenal (HPA) axis activity, glutamate release, GABA signaling, and dopamine release. These results replicate our earlier findings that nicotine exposure during adolescence increases ethanol consumption and extends this work by examining gene expression differences which could mediate these behavioral effects.
Collapse
Affiliation(s)
- Constanza P. Silva
- Biobehavioral Health Department, Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - William J. Horton
- Department of Animal Science, Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Michael J. Caruso
- Biobehavioral Health Department, Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Aswathy Sebastian
- Biochemistry and Molecular Biology, Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Laura C. Klein
- Biobehavioral Health Department, Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Istvan Albert
- Biochemistry and Molecular Biology, Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Helen M. Kamens
- Biobehavioral Health Department, Pennsylvania State University, University Park, Pennsylvania, United States of America
| |
Collapse
|
25
|
Sánchez-Sellero I, San-Román-Rodríguez E, Santos-Pérez S, Rossi-Izquierdo M, Soto-Varela A. Alcohol consumption in Menière’s disease patients. Nutr Neurosci 2018; 23:68-74. [DOI: 10.1080/1028415x.2018.1470372] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Affiliation(s)
- Inés Sánchez-Sellero
- Division of Toxicology, Department of Forensic Sciences, Pathology, Gynecology and Obstetrics, and Pediatrics, Veterinary School, Universidade de Santiago de Compostela, Lugo, Spain
| | | | - Sofía Santos-Pérez
- Division of Neurotology, Department of Otorhinolaryngology, Complexo Hospitalario Universitario de Santiago de Compostela, Department of Surgery and Medical-Surgical Specialities, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Marcos Rossi-Izquierdo
- Department of Otorhinolaryngology, Complexo Hospitalario Universitario Lucus Augusti, Lugo, Spain
| | - Andrés Soto-Varela
- Division of Neurotology, Department of Otorhinolaryngology, Complexo Hospitalario Universitario de Santiago de Compostela, Department of Surgery and Medical-Surgical Specialities, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| |
Collapse
|
26
|
Oxytocin Reduces Alcohol Cue-Reactivity in Alcohol-Dependent Rats and Humans. Neuropsychopharmacology 2018; 43:1235-1246. [PMID: 29090683 PMCID: PMC5916348 DOI: 10.1038/npp.2017.257] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 10/08/2017] [Accepted: 10/24/2017] [Indexed: 12/25/2022]
Abstract
Approved pharmacological treatments for alcohol use disorder are limited in their effectiveness, and new drugs that can easily be translated into the clinic are warranted. One of those candidates is oxytocin because of its interaction with several alcohol-induced effects. Alcohol-dependent rats as well as post-mortem brains of human alcoholics and controls were analyzed for the expression of the oxytocin system by qRT-PCR, in situ hybridization, receptor autoradiography ([125I]OVTA binding), and immunohistochemistry. Alcohol self-administration and cue-induced reinstatement behavior was measured after intracerebroventricular injection of 10 nM oxytocin in dependent rats. Here we show a pronounced upregulation of oxytocin receptors in brain tissues of alcohol-dependent rats and deceased alcoholics, primarily in frontal and striatal areas. This upregulation stems most likely from reduced oxytocin expression in hypothalamic nuclei. Pharmacological validation showed that oxytocin reduced cue-induced reinstatement response in dependent rats-an effect that was not observed in non-dependent rats. Finally, a clinical pilot study (German clinical trial number DRKS00009253) using functional magnetic resonance imaging in heavy social male drinkers showed that intranasal oxytocin (24 IU) decreased neural cue-reactivity in brain networks similar to those detected in dependent rats and humans with increased oxytocin receptor expression. These studies suggest that oxytocin might be used as an anticraving medication and thus may positively affect treatment outcomes in alcoholics.
Collapse
|
27
|
Zhou Y, Rubinstein M, Low MJ, Kreek MJ. V1b Receptor Antagonist SSR149415 and Naltrexone Synergistically Decrease Excessive Alcohol Drinking in Male and Female Mice. Alcohol Clin Exp Res 2018; 42:195-205. [PMID: 29105118 PMCID: PMC5750120 DOI: 10.1111/acer.13544] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 10/26/2017] [Indexed: 11/30/2022]
Abstract
BACKGROUND A recent clinical trial found that pharmacological blockade of V1b receptors reduces alcohol relapse in alcohol-dependent patients. SSR149415 is a selective V1b receptor antagonist that has potential for development as an alcohol dependency treatment. In this study, we investigated whether SSR149415 alone or in combination with the mu-opioid receptor (MOP-r) antagonist naltrexone (NTN) would alter excessive alcohol drinking in mice. METHODS Both sexes of C57BL/6J (B6) mice were subjected to a chronic intermittent access (IA) drinking paradigm (2-bottle choice, 24-hour access every other day) for 3 weeks. Sucrose and saccharin drinking were used as controls for alcohol-specific drug effects. Neuronal proopiomelanocortin (POMC) enhancer (nPE) knockout mice with hypothalamic-specific loss of POMC (including beta-endorphin, the main endogenous ligand of MOP-r) were used as a genetic control for the effects of NTN. RESULTS Acute administration of SSR149415 (1 to 30 mg/kg) reduced alcohol intake and preference in a dose-dependent manner in both male and female B6 mice after IA. To investigate potential synergistic effects between NTN and SSR149415, we tested 6 different combination doses of SSR149415 and NTN, and found that a combination of SSR149415 (3 mg/kg) and NTN (1 mg/kg) reduced alcohol intake profoundly at doses lower than the individual effective doses in both sexes of B6 mice. We confirmed the effect of SSR149415 on reducing alcohol intake in nPE-/- male mice, consistent with independent mechanisms by which SSR149415 and NTN decrease alcohol drinking. CONCLUSIONS The combination of V1b antagonist SSR149415 with NTN at individual subthreshold doses shows potential in alcoholism treatment, possibly with less adverse effects.
Collapse
Affiliation(s)
- Yan Zhou
- Laboratory of the Biology of Addictive Diseases, The Rockefeller University, NY
| | | | - Malcolm J. Low
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, MI
| | - Mary Jeanne Kreek
- Laboratory of the Biology of Addictive Diseases, The Rockefeller University, NY
| |
Collapse
|
28
|
Bowen MT, Neumann ID. Rebalancing the Addicted Brain: Oxytocin Interference with the Neural Substrates of Addiction. Trends Neurosci 2017; 40:691-708. [PMID: 29128108 DOI: 10.1016/j.tins.2017.10.003] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 10/05/2017] [Accepted: 10/06/2017] [Indexed: 12/21/2022]
Abstract
Drugs that act on the brain oxytocin (OXT) system may provide a much-needed treatment breakthrough for substance-use disorders. Targeting the brain OXT system has the potential to treat addiction to all major classes of addictive substance and to intervene across all stages of the addiction cycle. Emerging evidence suggests that OXT is able to interfere with such a wide range of addictive behaviours for such a wide range of addictive substances by rebalancing core neural systems that become dysregulated over the course of addiction. By improving our understanding of these interactions between OXT and the neural substrates of addiction, we will not only improve our understanding of addiction, but also our ability to effectively treat these devastating disorders.
Collapse
Affiliation(s)
- Michael T Bowen
- The University of Sydney, Faculty of Science, School of Psychology, Sydney, NSW, Australia; The University of Sydney, Brain and Mind Centre, Sydney, NSW, Australia
| | - Inga D Neumann
- Regensburg Center of Neuroscience, Department of Behavioural and Molecular Neurobiology, University of Regensburg, Regensburg, Germany.
| |
Collapse
|
29
|
Lee MR, Schwandt ML, Sankar V, Suchankova P, Sun H, Leggio L. Effect of alcohol use disorder on oxytocin peptide and receptor mRNA expression in human brain: A post-mortem case-control study. Psychoneuroendocrinology 2017; 85:14-19. [PMID: 28787642 DOI: 10.1016/j.psyneuen.2017.07.481] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Revised: 07/05/2017] [Accepted: 07/11/2017] [Indexed: 11/28/2022]
Abstract
Animal and human evidence supports a role for oxytocin in alcohol-seeking behaviors. There is interest, therefore, in targeting the oxytocin pathway as a new pharmacologic approach to treat alcohol use disorder. To this end, it is important to understand the effect of alcohol use disorder on endogenous oxytocin in brain regions that are relevant for the initiation and maintenance of alcohol use disorder. We examined human post-mortem brain tissue from males with alcohol use disorder (n=11) compared to nonalcohol dependent male controls (n=16). We a priori targeted five brain regions that in rodent studies, are projection areas for oxytocin neurons: nucleus accumbens, amygdala, hippocampus, ventral tegmental area and prefrontal cortex. Fold change in mRNA levels of oxytocin peptide and receptor were measured in each of the brain regions studied. Fold change for oxytocin peptide mRNA was significantly elevated in the prefrontal cortex of subjects with alcohol use disorder compared to controls (uncorrected p=0.0001; FDR-corrected p=0.001). For the entire sample of 27 subjects, there was a significant positive correlation between the fold change in oxytocin peptide mRNA in the prefrontal cortex and both daily alcohol intake (r2=0.38; p=0.002) and drinks per week (r2=0.24; p=0.02). Results are discussed in light of the previous animal and human literature on changes in the endogenous oxytocin system as an effect of chronic alcohol exposure.
Collapse
Affiliation(s)
- Mary R Lee
- Section on Clinical Psychoneuroendocrinology and Neuropsychopharmacology, National Institute on Alcohol Abuse and Alcoholism and National Institute on Drug Abuse, National Institutes of Health, Bethesda, MD, USA.
| | - Melanie L Schwandt
- Office of the Clinical Director, National Institute on Alcohol Abuse and Alcoholism and National Institute on Drug Abuse, National Institutes of Health, Bethesda, MD, USA
| | - Vignesh Sankar
- Section on Clinical Psychoneuroendocrinology and Neuropsychopharmacology, National Institute on Alcohol Abuse and Alcoholism and National Institute on Drug Abuse, National Institutes of Health, Bethesda, MD, USA
| | - Petra Suchankova
- Section on Clinical Psychoneuroendocrinology and Neuropsychopharmacology, National Institute on Alcohol Abuse and Alcoholism and National Institute on Drug Abuse, National Institutes of Health, Bethesda, MD, USA; Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Hui Sun
- Office of the Clinical Director, National Institute on Alcohol Abuse and Alcoholism and National Institute on Drug Abuse, National Institutes of Health, Bethesda, MD, USA
| | - Lorenzo Leggio
- Section on Clinical Psychoneuroendocrinology and Neuropsychopharmacology, National Institute on Alcohol Abuse and Alcoholism and National Institute on Drug Abuse, National Institutes of Health, Bethesda, MD, USA; Center for Alcohol and Addiction Studies, Department of Behavioral and Social Sciences, Brown University, Providence, RI, USA
| |
Collapse
|
30
|
Stevenson JR, Young KA, Bohidar AE, Francomacaro LM, Fasold TR, Buirkle JM, Ndem JR, Christian SC. Alcohol Consumption Decreases Oxytocin Neurons in the Anterior Paraventricular Nucleus of the Hypothalamus in Prairie Voles. Alcohol Clin Exp Res 2017; 41:1444-1451. [PMID: 28617958 DOI: 10.1111/acer.13430] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Accepted: 06/05/2017] [Indexed: 12/22/2022]
Abstract
BACKGROUND Alcohol use disorders are associated with dysfunctional social relationships and stress responses. The neuropeptides oxytocin (OT) and vasopressin (AVP) are known to orchestrate or mediate many aspects of social behavior, stress responses, and ingestive behaviors. Because of the overlap between the effects of alcohol and the roles of OT and AVP, we sought to determine whether alcohol consumption altered expression of OT and AVP in the paraventricular nucleus (PVN) of the hypothalamus, one of the key sites for OT and AVP synthesis. METHODS Pair-housed adult male prairie voles were allowed to consume 15% ethanol versus water in the home cage continuously (Continuous-Access [CA] group) or every other day for 4 hours (Intermittent-Access [IA] group). Control animals never had access to alcohol. After 7 weeks, animals were sacrificed and their brains were removed and immunohistochemical analysis of OT- and AVP-immunopositive neurons was performed. RESULTS OT-immunopositive neurons were significantly decreased in the anterior PVN in the CA but not IA group, relative to Control animals, suggesting that continuous alcohol consumption decreases the number of OT neurons. There was no effect of alcohol consumption on posterior PVN OT neurons, and no effect on PVN AVP neurons. CONCLUSIONS These data show that continuous-access voluntary alcohol consumption is associated with decreased OT neurons in the anterior PVN, suggesting that alcohol-induced alterations in the OT system should be investigated as a mechanism for alcohol-related changes in social behavior, stress responses, and exacerbation of alcohol use disorders.
Collapse
Affiliation(s)
- Jennie R Stevenson
- Department of Psychology, Bucknell University, Lewisburg, Pennsylvania.,Program in Neuroscience, Bucknell University, Lewisburg, Pennsylvania.,Program in Animal Behavior, Bucknell University, Lewisburg, Pennsylvania
| | - Katelyn A Young
- Department of Psychology, Bucknell University, Lewisburg, Pennsylvania.,Program in Neuroscience, Bucknell University, Lewisburg, Pennsylvania.,Program in Animal Behavior, Bucknell University, Lewisburg, Pennsylvania
| | - Amelia E Bohidar
- Department of Psychology, Bucknell University, Lewisburg, Pennsylvania.,Program in Neuroscience, Bucknell University, Lewisburg, Pennsylvania.,Program in Animal Behavior, Bucknell University, Lewisburg, Pennsylvania
| | - Lisa M Francomacaro
- Department of Psychology, Bucknell University, Lewisburg, Pennsylvania.,Program in Neuroscience, Bucknell University, Lewisburg, Pennsylvania.,Program in Animal Behavior, Bucknell University, Lewisburg, Pennsylvania
| | - Terra R Fasold
- Department of Psychology, Bucknell University, Lewisburg, Pennsylvania.,Program in Neuroscience, Bucknell University, Lewisburg, Pennsylvania.,Program in Animal Behavior, Bucknell University, Lewisburg, Pennsylvania
| | - Julia M Buirkle
- Department of Psychology, Bucknell University, Lewisburg, Pennsylvania.,Program in Neuroscience, Bucknell University, Lewisburg, Pennsylvania.,Program in Animal Behavior, Bucknell University, Lewisburg, Pennsylvania
| | - Jackie R Ndem
- Department of Psychology, Bucknell University, Lewisburg, Pennsylvania.,Program in Neuroscience, Bucknell University, Lewisburg, Pennsylvania.,Program in Animal Behavior, Bucknell University, Lewisburg, Pennsylvania
| | - Sara C Christian
- Department of Psychology, Bucknell University, Lewisburg, Pennsylvania.,Program in Neuroscience, Bucknell University, Lewisburg, Pennsylvania.,Program in Animal Behavior, Bucknell University, Lewisburg, Pennsylvania
| |
Collapse
|
31
|
Peters ST, Bowen MT, Bohrer K, McGregor IS, Neumann ID. Oxytocin inhibits ethanol consumption and ethanol-induced dopamine release in the nucleus accumbens. Addict Biol 2017; 22:702-711. [PMID: 26810371 DOI: 10.1111/adb.12362] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Revised: 12/04/2015] [Accepted: 12/07/2015] [Indexed: 11/29/2022]
Abstract
Alcohol (EtOH) is one of the most widely abused recreational drugs and is arguably the most harmful. However, current treatment options for alcohol-use disorders generally have limited efficacy and poor uptake in the community. In this context, the neuropeptide oxytocin (OXT) has emerged as a promising potential treatment option for a number of substance-use disorders, including alcoholism. The utility of OXT in reducing consumption of and craving for a wide range of substances may lie in its ability to modulate drug-induced neurochemical effects within the mesolimbic dopamine pathway. However, the impact of OXT on EtOH actions in this pathway has yet to be explored. Here, we reveal that an acute intracerebroventricular (icv) infusion of OXT (1 µg/5 µl) attenuated voluntary EtOH (20 percent) self-administration after chronic intermittent access to EtOH for 59 days (28 drinking sessions) in male Wistar rats. Next, we demonstrated that an acute intraperitoneal (ip) injection of EtOH (1.5 g/kg, 15 percent w/v) increased dopamine release within the nucleus accumbens in both EtOH-naive rats and rats that had received 10 daily ip injections of EtOH. Icv OXT completely blocked the EtOH-induced dopamine release in both EtOH-naive and chronically treated rats. The attenuation of EtOH-induced dopamine release by OXT may help to explain the reduced EtOH self-administration observed following icv OXT infusion.
Collapse
Affiliation(s)
- Sebastian T. Peters
- Department of Behavioral and Molecular Neurobiology; University of Regensburg; Germany
- Current address: Department of Neurology; University Clinic Regensburg; Germany
| | | | - Kathrin Bohrer
- Department of Behavioral and Molecular Neurobiology; University of Regensburg; Germany
| | | | - Inga D. Neumann
- Department of Behavioral and Molecular Neurobiology; University of Regensburg; Germany
| |
Collapse
|
32
|
Nikolaou K, Kapoukranidou D, Ndungu S, Floros G, Kovatsi L. Severity of Withdrawal Symptoms, Plasma Oxytocin Levels, and Treatment Outcome in Heroin Users Undergoing Acute Withdrawal. J Psychoactive Drugs 2017; 49:233-241. [PMID: 28443705 DOI: 10.1080/02791072.2017.1312644] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Pre-clinical studies show that, following chronic opioid exposure, oxytocin neurons exhibit over-excitation upon withdrawal, causing an increase in oxytocin brain and plasma levels. Relevant clinical data on humans are scarce. This study investigates the opioid withdrawal stress effect on oxytocin plasma levels in humans. We evaluated 57 male chronic heroin users in a residential detoxification program. We determined plasma oxytocin levels by ELISA and measured the stress effects of withdrawal using the COWS scale for opioid withdrawal, the VAS scale for craving, and the Hamilton scales for anxiety and depression on the second day of admission. Out of the 57 patients enrolled in the study, 27 completed the 21-day program, while the remaining 30 dropped out prior to completion. Plasma oxytocin levels were significantly higher in those individuals who dropped out than in those who completed the program. Participants who dropped out at some stage scored higher in the COWS, VAS-Craving, and Hamilton-anxiety scales, indicating a higher stress and explaining the higher oxytocin levels. In addition, plasma oxytocin levels correlated positively with the scores achieved in the COWS and Hamilton-anxiety scales. Higher withdrawal stress levels are associated with higher plasma oxytocin levels and early treatment discharge.
Collapse
Affiliation(s)
- Kakia Nikolaou
- a Consultant Psychiatrist, Head of the Addictions Department IANOS , Papanikolaou General Hospital of Thessaloniki-Psychiatric Hospital of Thessaloniki , Thessaloniki , Greece
| | - Dorothea Kapoukranidou
- b Associate Professor, Department of Physiology, School of Medicine , Aristotle University of Thessaloniki , Thessaloniki , Greece
| | - Samuel Ndungu
- c Emeritus Professor, Laboratory of Forensic Medicine and Toxicology, School of Medicine , Aristotle University of Thessaloniki , Thessaloniki , Greece
| | - Georgios Floros
- d Scientific Associate, Second Department of Psychiatry, School of Medicine , Aristotle University of Thessaloniki , Thessaloniki , Greece
| | - Leda Kovatsi
- e Assistant Professor, Laboratory of Forensic Medicine and Toxicology, School of Medicine , Aristotle University of Thessaloniki , Thessaloniki , Greece
| |
Collapse
|
33
|
Chronic ethanol intake induces partial microglial activation that is not reversed by long-term ethanol withdrawal in the rat hippocampal formation. Neurotoxicology 2017; 60:107-115. [PMID: 28408342 DOI: 10.1016/j.neuro.2017.04.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Revised: 04/06/2017] [Accepted: 04/07/2017] [Indexed: 12/12/2022]
Abstract
Neuroinflammation has been implicated in the pathogenesis of several disorders. Activation of microglia leads to the release of pro-inflammatory mediators and microglial-mediated neuroinflammation has been proposed as one of the alcohol-induced neuropathological mechanisms. The present study aimed to examine the effect of chronic ethanol exposure and long-term withdrawal on microglial activation and neuroinflammation in the hippocampal formation. Male rats were submitted to 6 months of ethanol treatment followed by a 2-month withdrawal period. Stereological methods were applied to estimate the total number of microglia and activated microglia detected by CD11b immunohistochemistry in the hippocampal formation. The expression levels of the pro-inflammatory cytokines TNF-α, COX-2 and IL-15 were measured by qRT-PCR. Alcohol consumption was associated with an increase in the total number of activated microglia but morphological assessment indicated that microglia did not exhibit a full activation phenotype. These data were supported by functional evidence since chronic alcohol consumption produced no changes in the expression of TNF-α or COX-2. The levels of IL-15 a cytokine whose expression is increased upon activation of both astrocytes and microglia, was induced by chronic alcohol treatment. Importantly, the partial activation of microglia induced by ethanol was not reversed by long-term withdrawal. This study suggests that chronic alcohol exposure induces a microglial phenotype consistent with partial activation without significant increase in classical cytokine markers of neuroinflammation in the hippocampal formation. Furthermore, long-term cessation of alcohol intake is not sufficient to alter the microglial partial activation phenotype induced by ethanol.
Collapse
|
34
|
Dolder PC, Holze F, Liakoni E, Harder S, Schmid Y, Liechti ME. Alcohol acutely enhances decoding of positive emotions and emotional concern for positive stimuli and facilitates the viewing of sexual images. Psychopharmacology (Berl) 2017; 234:41-51. [PMID: 27640999 DOI: 10.1007/s00213-016-4431-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Accepted: 09/05/2016] [Indexed: 12/23/2022]
Abstract
RATIONALE Social cognition influences social interactions. Alcohol reportedly facilitates social interactions. However, the acute effects of alcohol on social cognition are relatively poorly studied. METHODS We investigated the effects of alcoholic or non-alcoholic beer on emotion recognition, empathy, and sexual arousal using the dynamic face emotion recognition task (FERT), Multifaceted Empathy Test (MET), and Sexual Arousal Task (SAT) in a double-blind, random-order, cross-over study in 60 healthy social drinkers. We also assessed subjective effects using visual analog scales (VASs), blood alcohol concentrations, and plasma oxytocin levels. RESULTS Alcohol increased VAS ratings of stimulated, happy, talkative, open, and want to be with others. The subjective effects of alcohol were greater in participants with higher trait inhibitedness. Alcohol facilitated the recognition of happy faces on the FERT and enhanced emotional empathy for positive stimuli on the MET, particularly in participants with low trait empathy. Pictures of explicit sexual content were rated as less pleasant than neutral pictures after non-alcoholic beer but not after alcoholic beer. Explicit sexual pictures were rated as more pleasant after alcoholic beer compared with non-alcoholic beer, particularly in women. Alcohol did not alter the levels of circulating oxytocin. CONCLUSIONS Alcohol biased emotion recognition toward better decoding of positive emotions and increased emotional concern for positive stimuli. No support was found for a modulatory role of oxytocin. Alcohol also facilitated the viewing of sexual images, consistent with disinhibition, but it did not actually enhance sexual arousal. These effects of alcohol on social cognition likely enhance sociability. TRIAL REGISTRATION www.clinicaltrials.gov/ct2/show/NCT02318823.
Collapse
Affiliation(s)
- Patrick C Dolder
- Division of Clinical Pharmacology and Toxicology, Department of Biomedicine and Department of Clinical Research, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Friederike Holze
- Division of Clinical Pharmacology and Toxicology, Department of Biomedicine and Department of Clinical Research, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Evangelia Liakoni
- Division of Clinical Pharmacology and Toxicology, Department of Biomedicine and Department of Clinical Research, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Samuel Harder
- Division of Clinical Pharmacology and Toxicology, Department of Biomedicine and Department of Clinical Research, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Yasmin Schmid
- Division of Clinical Pharmacology and Toxicology, Department of Biomedicine and Department of Clinical Research, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Matthias E Liechti
- Division of Clinical Pharmacology and Toxicology, University Hospital Basel, Hebelstrasse 2, 4031, Basel, CH, Switzerland.
| |
Collapse
|
35
|
Walcott AT, Ryabinin AE. Alcohol's Effects on Pair-Bond Maintenance in Male Prairie Voles. Front Psychiatry 2017; 8:226. [PMID: 29204125 PMCID: PMC5698799 DOI: 10.3389/fpsyt.2017.00226] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 10/24/2017] [Indexed: 12/18/2022] Open
Abstract
Alcohol abuse can have devastating effects on social relationships. In particular, discrepant patterns of heavy alcohol consumption are associated with increased rates of separation and divorce. Previous studies have attempted to model these effects of alcohol using socially monogamous prairie voles. These studies showed that alcohol consumption can inhibit the formation of pair bonds in this species. While these findings indicated that alcohol's effects on social attachments can involve biological mechanisms, the formation of pair bonds does not properly model long-term human attachments. To overcome this caveat, this study explored whether discordant or concordant alcohol consumption between individuals within established pairs affects maintenance of pair bonds in male prairie voles. Male and female prairie voles were allowed to form a pair bond for 1 week. Following this 1-week cohabitation period, males received access to 10% continuous ethanol; meanwhile, their female partners had access to either alcohol and water or just water. When there was a discrepancy in alcohol consumption, male prairie voles showed a decrease in partner preference (PP). Conversely, when concordant drinking occurred, males showed no inhibition in PP. Further analysis revealed a decrease in oxytocin immunoreactivity in the paraventricular nucleus of alcohol-exposed males that was independent of the drinking status of their female partners. On the other hand, only discordant alcohol consumption resulted in an increase of FosB immunoreactivity in the periaqueductal gray of male voles, a finding suggesting a potential involvement of this brain region in the effects of alcohol on maintenance of pair bonds. Our studies provide the first evidence that alcohol has effects on established pair bonds and that partner drinking status plays a large role in these effects.
Collapse
Affiliation(s)
- Andre T Walcott
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, OR, United States
| | - Andrey E Ryabinin
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, OR, United States
| |
Collapse
|
36
|
Kim S, Kwok S, Mayes LC, Potenza MN, Rutherford HJV, Strathearn L. Early adverse experience and substance addiction: dopamine, oxytocin, and glucocorticoid pathways. Ann N Y Acad Sci 2016; 1394:74-91. [PMID: 27508337 DOI: 10.1111/nyas.13140] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Revised: 05/12/2016] [Accepted: 05/20/2016] [Indexed: 12/17/2022]
Abstract
Substance addiction may follow a chronic, relapsing course and critically undermine the physical and psychological well-being of the affected individual and the social units of which the individual is a member. Despite the public health burden associated with substance addiction, treatment options remain suboptimal, with relapses often seen. The present review synthesizes growing insights from animal and human research to shed light upon developmental and neurobiological pathways that may increase susceptibility to addiction. We examine the dopamine system, the oxytocin system, and the glucocorticoid system, as they are particularly relevant to substance addiction. Our aim is to delineate how early adverse experience may induce long-lasting alterations in each of these systems at molecular, neuroendocrine, and behavioral levels and ultimately lead to heightened vulnerability to substance addiction. We further discuss how substance addiction in adulthood may increase the risk of suboptimal caregiving for the next generation, perpetuating the intergenerational cycle of early adverse experiences and addiction.
Collapse
Affiliation(s)
- Sohye Kim
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, Texas.,Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, Texas.,Department of Pediatrics, Baylor College of Medicine, Houston, Texas.,Attachment and Neurodevelopment Laboratory, Children's Nutrition Research Center, Baylor College of Medicine, Houston, Texas
| | - Stephanie Kwok
- Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, Texas
| | - Linda C Mayes
- Yale Child Study Center, Yale University School of Medicine, New Haven, Connecticut
| | - Marc N Potenza
- Yale Child Study Center, Yale University School of Medicine, New Haven, Connecticut.,Departments of Psychiatry and Neuroscience and the National Center on Addiction and Substance Abuse (CASAColumbia), Yale University School of Medicine, New Haven, Connecticut.,Connecticut Mental Health Center, New Haven, Connecticut
| | | | - Lane Strathearn
- Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, Texas.,Department of Pediatrics, Baylor College of Medicine, Houston, Texas.,Attachment and Neurodevelopment Laboratory, Children's Nutrition Research Center, Baylor College of Medicine, Houston, Texas.,Stead Family Department of Pediatrics, University of Iowa Carver College of Medicine, Iowa City, Iowa
| |
Collapse
|
37
|
Rutigliano G, Rocchetti M, Paloyelis Y, Gilleen J, Sardella A, Cappucciati M, Palombini E, Dell'Osso L, Caverzasi E, Politi P, McGuire P, Fusar-Poli P. Peripheral oxytocin and vasopressin: Biomarkers of psychiatric disorders? A comprehensive systematic review and preliminary meta-analysis. Psychiatry Res 2016; 241:207-20. [PMID: 27183106 DOI: 10.1016/j.psychres.2016.04.117] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 04/28/2016] [Accepted: 04/29/2016] [Indexed: 11/29/2022]
Abstract
A large array of studies have investigated peripheral oxytocin (OT) and vasopressin (ADH) as potential biomarkers of psychiatric disorders, with highly conflicting and heterogenous findings. We searched Web of KnowledgeSM and Scopus® for English original articles investigating OT and/or ADH levels in different biological fluids (plasma/serum, saliva, urine and cerebrospinal fluid) across several psychiatric disorders. Sixty-four studies were included. We conducted 19 preliminary meta-analyses addressing OT alterations in plasma/serum, saliva, urine and cerebrospinal fluid of 7 psychiatric disorders and ADH alterations in plasma/serum, saliva, urine and cerebrospinal fluid of 6 psychiatric disorders compared to controls. Hedge's g was used as effect size measure, together with heterogeneity analyses, test of publication biases and quality control. None of them (except serum OT in anorexia nervosa) revealed significant differences. There is no convincing evidence that peripheral ADH or OT might be reliable biomarkers in psychiatric disorders. However, the lack of significant results was associated with high methodological heterogeneity, low quality of the studies, small sample size, and scarce reliability of the methods used in previous studies, which need to be validated and standardized.
Collapse
Affiliation(s)
- Grazia Rutigliano
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, PO63, De Crespigny Park, SE58AF London, UK; Department of Clinical and Experimental Medicine, University of Pisa, Via Roma, 67-56126 Pisa, Italy
| | - Matteo Rocchetti
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, PO63, De Crespigny Park, SE58AF London, UK; Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Yannis Paloyelis
- Neuroimaging Department, Institute of Psychiatry, Psychology and Neuroscience, King's College London, PO89, De Crespigny Park, SE58AF London, UK
| | - James Gilleen
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, PO63, De Crespigny Park, SE58AF London, UK; Department of Psychology, University of Roehampton, UK
| | - Alberto Sardella
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, PO63, De Crespigny Park, SE58AF London, UK
| | - Marco Cappucciati
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, PO63, De Crespigny Park, SE58AF London, UK; Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Erika Palombini
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, PO63, De Crespigny Park, SE58AF London, UK; Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Liliana Dell'Osso
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma, 67-56126 Pisa, Italy
| | - Edgardo Caverzasi
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Pierluigi Politi
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Philip McGuire
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, PO63, De Crespigny Park, SE58AF London, UK
| | - Paolo Fusar-Poli
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, PO63, De Crespigny Park, SE58AF London, UK; OASIS clinic, SLaM NHS Foundation Trust, 190 Kennington Lane, SE11 5DL London, UK.
| |
Collapse
|
38
|
Nucleus accumbens lentiviral-mediated gain of function of the oxytocin receptor regulates anxiety- and ethanol-related behaviors in adult mice. Physiol Behav 2016; 164:249-58. [PMID: 27306084 DOI: 10.1016/j.physbeh.2016.06.009] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Revised: 06/10/2016] [Accepted: 06/11/2016] [Indexed: 12/23/2022]
Abstract
Anxiety is believed to influence ethanol use human in alcoholics. Studies using laboratory animals suggested an interaction between oxytocin and the behavioral effects of ethanol. Our previous study implicated a potential role for the oxytocin receptor (OxtR) in regulating ethanol-conditioned place preference. Here, we examined anxiety and the behavioral responses to ethanol in C57BL/6 mice stereotaxically injected in the nucleus accumbens (NAcc) with lentiviral vectors expressing an empty vector (Mock) or the OxtR cDNA. For anxiety we used the elevated-plus maze, the open-field and the marble-burying tests and for ethanol we used the two-bottle choice paradigm, the wire-hanging and ethanol-induced loss-of-righting-reflex tests. We found that, compared to Mock, OxtR overexpression led to anxiolytic-like behavior without altering spontaneous locomotor activity. Most importantly, we found that, relative to Mock controls, increased expression of the OxtR in the NAcc led to decreased ethanol consumption and preference in the two-bottle choice protocol and increased resistance to ethanol-induced sedation. We also compared the consequence of OxtR modulation on the consumption and preference of saccharin and quinine and found that the two experimental groups did not differ for any tastant. These results provide further evidence that the oxytocin system contributes to the regulation of ethanol drinking and sensitivity and position OxtR as a central molecular mediator of ethanol's effects within the mesolimbic system. Taken together, the current findings suggest that OxtR manipulation may be a relevant strategy to address ethanol use disorders.
Collapse
|
39
|
Rebouças ECC, Leal S, Silva SM, Sá SI. Changes in the female arcuate nucleus morphology and neurochemistry after chronic ethanol consumption and long-term withdrawal. J Chem Neuroanat 2016; 77:30-40. [PMID: 27154870 DOI: 10.1016/j.jchemneu.2016.05.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Revised: 05/02/2016] [Accepted: 05/02/2016] [Indexed: 12/26/2022]
Abstract
Ethanol is a macronutrient whose intake is a form of ingestive behavior, sharing physiological mechanisms with food intake. Chronic ethanol consumption is detrimental to the brain, inducing gender-dependent neuronal damage. The hypothalamic arcuate nucleus (ARN) is a modulator of food intake that expresses feeding-regulatory neuropeptides, such as alpha melanocyte-stimulating hormone (α-MSH) and neuropeptide Y (NPY). Despite its involvement in pathways associated with eating disorders and ethanol abuse, the impact of ethanol consumption and withdrawal in the ARN structure and neurochemistry in females is unknown. We used female rat models of 20% ethanol consumption for six months and of subsequent ethanol withdrawal for two months. Food intake and body weights were measured. ARN morphology was stereologically analyzed to estimate its volume, total number of neurons and total number of neurons expressing NPY, α-MSH, tyrosine hydroxylase (TH) and estrogen receptor alpha (ERα). Ethanol decreased energy intake and body weights. However, it did not change the ARN morphology or the expression of NPY, α-MSH and TH, while increasing ERα expression. Withdrawal induced a significant volume and neuron loss that was accompanied by an increase in NPY expression without affecting α-MSH and TH expression. These findings indicate that the female ARN is more vulnerable to withdrawal than to excess alcohol. The data also support the hypothesis that the same pathways that regulate the expression of NPY and α-MSH in long-term ethanol intake may regulate food intake. The present model of long-term ethanol intake and withdrawal induces new physiological conditions with adaptive responses.
Collapse
Affiliation(s)
- Elce C C Rebouças
- Department of Natural Sciences, State University of Southwestern Bahia, Praça Primavera, 40-Bairro Primavera, Itapetinga, BA 45700-000, Brazil; Department of Anatomy, Faculty of Medicine, University of Porto, Al. Prof. Hernâni Monteiro, 4200-319 Porto, Portugal; Center for Health Technology and Services Research (CINTESIS), Faculty of Medicine, University of Porto, Rua Dr. Plácido da Costa, 4200-450 Porto, Portugal.
| | - Sandra Leal
- Department of Anatomy, Faculty of Medicine, University of Porto, Al. Prof. Hernâni Monteiro, 4200-319 Porto, Portugal; Center for Health Technology and Services Research (CINTESIS), Faculty of Medicine, University of Porto, Rua Dr. Plácido da Costa, 4200-450 Porto, Portugal; Institute of Research and Advanced Training in Health Sciences and Technologies (IINFACTS), Department of Sciences, Instituto Universitário de Ciências da Saúde (IUCS), CESPU, CRL, R. Central da Gandra 1317, 4585-116 Gandra, Portugal.
| | - Susana M Silva
- Department of Anatomy, Faculty of Medicine, University of Porto, Al. Prof. Hernâni Monteiro, 4200-319 Porto, Portugal; Center for Health Technology and Services Research (CINTESIS), Faculty of Medicine, University of Porto, Rua Dr. Plácido da Costa, 4200-450 Porto, Portugal.
| | - Susana I Sá
- Department of Anatomy, Faculty of Medicine, University of Porto, Al. Prof. Hernâni Monteiro, 4200-319 Porto, Portugal; Center for Health Technology and Services Research (CINTESIS), Faculty of Medicine, University of Porto, Rua Dr. Plácido da Costa, 4200-450 Porto, Portugal.
| |
Collapse
|
40
|
Todkar A, Granholm L, Aljumah M, Nilsson KW, Comasco E, Nylander I. HPA Axis Gene Expression and DNA Methylation Profiles in Rats Exposed to Early Life Stress, Adult Voluntary Ethanol Drinking and Single Housing. Front Mol Neurosci 2016; 8:90. [PMID: 26858597 PMCID: PMC4726785 DOI: 10.3389/fnmol.2015.00090] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 12/24/2015] [Indexed: 11/24/2022] Open
Abstract
The neurobiological basis of early life stress (ELS) impact on vulnerability to alcohol use disorder is not fully understood. The effect of ELS, adult ethanol consumption and single housing, on expression of stress and DNA methylation regulatory genes as well as blood corticosterone levels was investigated in the hypothalamus and pituitary of adult out-bred Wistar rats subjected to different rearing conditions. A prolonged maternal separation (MS) of 360 min (MS360) was used to study the effect of ELS, and a short MS of 15 min (MS15) was used as a control. Voluntary ethanol drinking was assessed using a two-bottle free choice paradigm to simulate human episodic drinking. The effects of single housing and ethanol were assessed in conventional animal facility rearing (AFR) conditions. Single housing in adulthood was associated with lower Crhr1 and higher Pomc expression in the pituitary, whereas ethanol drinking was associated with higher expression of Crh in the hypothalamus and Crhr1 in the pituitary, accompanied by lower corticosterone levels. As compared to controls with similar early life handling, rats exposed to ELS displayed lower expression of Pomc in the hypothalamus, and higher Dnmt1 expression in the pituitary. Voluntary ethanol drinking resulted in lower Fkbp5 expression in the pituitary and higher Crh expression in the hypothalamus, independently of rearing conditions. In rats exposed to ELS, water and ethanol drinking was associated with higher and lower corticosterone levels, respectively. The use of conventionally reared rats as control group yielded more significant results than the use of rats exposed to short MS. Positive correlations, restricted to the hypothalamus and ELS group, were observed between the expression of the hypothalamus-pituitary-adrenal receptor and the methylation-related genes. Promoter DNA methylation and expression of respective genes did not correlate suggesting that other loci are involved in transcriptional regulation. Concluding, single housing is a confounding factor to be considered in voluntary ethanol drinking paradigms. ELS and ethanol drinking in adulthood exert independent effects on hypothalamic and pituitary related genes, however, in a manner dependent on the control group used.
Collapse
Affiliation(s)
| | - Linnea Granholm
- Department of Pharmaceutical Bioscience, Uppsala University Uppsala, Sweden
| | - Mujtaba Aljumah
- Department of Neuroscience, Uppsala University Uppsala, Sweden
| | - Kent W Nilsson
- Centre for Clinical Research, Västerås Central Hospital, Uppsala University Uppsala, Sweden
| | - Erika Comasco
- Department of Neuroscience, Uppsala University Uppsala, Sweden
| | - Ingrid Nylander
- Department of Pharmaceutical Bioscience, Uppsala University Uppsala, Sweden
| |
Collapse
|
41
|
Blum K, Thompson B, Demotrovics Z, Femino J, Giordano J, Oscar-Berman M, Teitelbaum S, Smith DE, Roy AK, Agan G, Fratantonio J, Badgaiyan RD, Gold MS. The Molecular Neurobiology of Twelve Steps Program & Fellowship: Connecting the Dots for Recovery. JOURNAL OF REWARD DEFICIENCY SYNDROME 2015; 1:46-64. [PMID: 26306329 PMCID: PMC4545669 DOI: 10.17756/jrds.2015-008] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
There are some who suggest that alcoholism and drug abuse are not diseases at all and that they are not consequences of a brain disorder as espoused recently by the American Society of Addiction Medicine (ASAM). Some would argue that addicts can quit on their own and moderate their alcohol and drug intake. When they present to a treatment program or enter the 12 Step Program & Fellowship, many addicts finally achieve complete abstinence. However, when controlled drinking fails, there may be successful alternatives that fit particular groups of individuals. In this expert opinion, we attempt to identify personal differences in recovery, by clarifying the molecular neurobiological basis of each step of the 12 Step Program. We explore the impact that the molecular neurobiological basis of the 12 steps can have on Reward Deficiency Syndrome (RDS) despite addiction risk gene polymorphisms. This exploration has already been accomplished in part by Blum and others in a 2013 Springer Neuroscience Brief. The purpose of this expert opinion is to briefly, outline the molecular neurobiological and genetic links, especially as they relate to the role of epigenetic changes that are possible in individuals who regularly attend AA meetings. It begs the question as to whether "12 steps programs and fellowship" does induce neuroplasticity and continued dopamine D2 receptor proliferation despite carrying hypodopaminergic type polymorphisms such as DRD2 A1 allele. "Like-minded" doctors of ASAM are cognizant that patients in treatment without the "psycho-social-spiritual trio," may not be obtaining the important benefits afforded by adopting 12-step doctrines. Are we better off with coupling medical assisted treatment (MAT) that favors combining dopamine agonist modalities (DAM) as possible histone-deacetylase activators with the 12 steps followed by a program that embraces either one or the other? While there are many unanswered questions, at least we have reached a time when "science meets recovery," and in doing so, can further redeem joy in recovery.
Collapse
Affiliation(s)
- Kenneth Blum
- Department of Psychiatry, School of Medicine and McKnight Brain Institute, University of Florida, Gainesville, FL, USA
- Department of Addiction Research and Therapy, Malibu Beach Recovery Center, Malibu Beach, CA, USA
- Dominion Diagnostics, Inc., North Kingstown, RI, USA
- IGENE, LLC., Austin, TX, USA
- RDSolutions, Del Mar, CA, USA
- National Institute for Holistic Medicine, North Miami Beach, FL, USA
| | - Benjamin Thompson
- Behavioral Neuroscience Program, Boston University School of Medicine, and Boston VA Healthcare System, Boston, MA, USA
| | - Zsolt Demotrovics
- Eötvös Loránd University, Institute of Psychology, Budapest, Hungary
| | - John Femino
- Dominion Diagnostics, Inc., North Kingstown, RI, USA
- Meadows Edge Recovery Center, North Kingstown, RI, USA
| | - John Giordano
- National Institute for Holistic Medicine, North Miami Beach, FL, USA
| | - Marlene Oscar-Berman
- Departments of Psychiatry, Neurology, and Anatomy & Neurobiology, Boston University School of Medicine, and Boston VA Healthcare System, Boston, MA, USA
| | - Scott Teitelbaum
- Department of Psychiatry, School of Medicine and McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - David E. Smith
- Dominion Diagnostics, Inc., North Kingstown, RI, USA
- Institute of Health & Aging, University of California at San Francisco, San Francisco, CA, USA
| | | | - Gozde Agan
- Dominion Diagnostics, Inc., North Kingstown, RI, USA
| | | | - Rajendra D. Badgaiyan
- Department of Psychiatry, University of Minnesota College of Medicine, Minneapolis, MN, USA
| | - Mark S. Gold
- Director of Research, Drug Enforcement Administration (DEA) Educational Foundation, Washington, D.C, USA
- Departments of Psychiatry & Behavioral Sciences at the Keck, University of Southern California, School of Medicine, CA, USA
| |
Collapse
|
42
|
Abstract
Many drugs, including alcohol and stimulants, demonstrably increase sociability and verbal interaction and are recreationally consumed in social settings. One drug, 3,4-methylenedioxymethamphetamine (MDMA, ecstasy), seems to produce its prosocial effects by increasing plasma oxytocin levels, and the oxytocin system has been implicated in responses to several other drugs of abuse. Here, we sought to investigate the effects of 2 other "social" drugs on plasma oxytocin levels--methamphetamine and alcohol. Based on their shared capacity to enhance sociability, we hypothesized that both methamphetamine and alcohol would increase plasma oxytocin levels. In study 1, 11 healthy adult volunteers attended 3 sessions during which they received methamphetamine (10 mg or 20 mg) or placebo under double-blind conditions. Subjective drug effects, cardiovascular effects, and plasma oxytocin levels were measured at regular intervals throughout the sessions. In study 2, 8 healthy adult volunteers attended a single session during which they received 1 beverage containing placebo, and then a beverage containing alcohol (0.8 g/kg). Subjective effects, breath alcohol levels, and plasma oxytocin levels were measured at regular intervals. Both methamphetamine and alcohol produced their expected physiological and subjective effects, but neither of these drugs increased plasma oxytocin levels. The neurobiological mechanisms mediating the prosocial effects of drugs such as alcohol and methamphetamine remain to be identified.
Collapse
|
43
|
Zanos P, Georgiou P, Metaxas A, Kitchen I, Winsky-Sommerer R, Bailey A. Region-specific up-regulation of oxytocin receptor binding in the brain of mice following chronic nicotine administration. Neurosci Lett 2015; 600:33-7. [PMID: 26037668 DOI: 10.1016/j.neulet.2015.05.054] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Revised: 05/18/2015] [Accepted: 05/24/2015] [Indexed: 12/25/2022]
Abstract
Nicotine addiction is considered to be the main preventable cause of death worldwide. While growing evidence indicates that the neurohypophysial peptide oxytocin can modulate the addictive properties of several abused drugs, the regulation of the oxytocinergic system following nicotine administration has so far received little attention. Here, we examined the effects of long-term nicotine or saline administration on the central oxytocinergic system using [(125)I]OVTA autoradiographic binding in mouse brain. Male, 7-week old C57BL6J mice were treated with either nicotine (7.8 mg/kg daily; rate of 0.5 μl per hour) or saline for a period of 14-days via osmotic minipumps. Chronic nicotine administration induced a marked region-specific upregulation of the oxytocin receptor binding in the amygdala, a brain region involved in stress and emotional regulation. These results provide direct evidence for nicotine-induced neuroadaptations in the oxytocinergic system, which may be involved in the modulation of nicotine-seeking as well as emotional consequence of chronic drug use.
Collapse
Affiliation(s)
- Panos Zanos
- Sleep, Chronobiology & Addiction Group, School of Biosciences and Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7XH Surrey, UK
| | - Polymnia Georgiou
- Sleep, Chronobiology & Addiction Group, School of Biosciences and Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7XH Surrey, UK
| | - Athanasios Metaxas
- Sleep, Chronobiology & Addiction Group, School of Biosciences and Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7XH Surrey, UK
| | - Ian Kitchen
- Sleep, Chronobiology & Addiction Group, School of Biosciences and Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7XH Surrey, UK
| | - Raphaelle Winsky-Sommerer
- Sleep, Chronobiology & Addiction Group, School of Biosciences and Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7XH Surrey, UK
| | - Alexis Bailey
- Sleep, Chronobiology & Addiction Group, School of Biosciences and Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7XH Surrey, UK.
| |
Collapse
|
44
|
Acetaldehyde self-administration by a two-bottle choice paradigm: consequences on emotional reactivity, spatial learning, and memory. Alcohol 2015; 49:139-48. [PMID: 25636827 DOI: 10.1016/j.alcohol.2015.01.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Acetaldehyde, the first alcohol metabolite, is responsible for many pharmacological effects that are not clearly distinguishable from those exerted by its parent compound. It alters motor performance, induces reinforced learning and motivated behavior, and produces different reactions according to the route of administration and the relative accumulation in the brain or in the periphery. The effective activity of oral acetaldehyde represents an unresolved field of inquiry that deserves further investigation. Thus, this study explores the acquisition and maintenance of acetaldehyde drinking behavior in adult male rats, employing a two-bottle choice paradigm for water and acetaldehyde solution (from 0.9% to 3.2% v/v), over 8 weeks. The behavioral consequences exerted by chronic acetaldehyde intake are assessed by a set of different tests: trials in an open-field arena and elevated-plus maze provided information on both general motor and explorative activity, and anxiety-driven behavioral responses. The Morris water maze allowed the exploration of cognitive processes such as spatial learning and memory. Determination of acetaldehyde levels in the brain was carried out at the end of the drinking paradigm. Our results indicate that rats exposed for the first time to acetaldehyde at 0.9% displayed a regular and stable daily drinking pattern that reached higher values and a "peaks and drops" shaped-trend when acetaldehyde concentration was increased to 3.2%. Accordingly, an increase in acetaldehyde levels in the brain was determined compared to non-acetaldehyde drinking rats. Acetaldehyde intake during the free-choice paradigm exerted an anxiogenic response in the open-field arena and elevated-plus maze, which in turn correlates with an enhancement in cognitive flexibility and spatial orientation skills, when an adaptive response to a stressful environmental challenge was required. These findings further support the idea that acetaldehyde is indeed a centrally active and behaviorally relevant metabolite of alcohol.
Collapse
|
45
|
Cavanaugh SE. A Transition in Fetal Alcohol Syndrome Research: The Shift from Animal Modeling to Human Intervention. Alcohol Alcohol 2015; 50:251-5. [DOI: 10.1093/alcalc/agu108] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
46
|
Bahi A. The oxytocin receptor impairs ethanol reward in mice. Physiol Behav 2014; 139:321-7. [PMID: 25449413 DOI: 10.1016/j.physbeh.2014.11.046] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Revised: 11/13/2014] [Accepted: 11/14/2014] [Indexed: 01/24/2023]
Abstract
It is well established that oxytocin, and its receptor (OxtR), play a crucial role in addiction and that the stimulation of oxytocin neurotransmission reduces addictive behaviors to ethanol in laboratory animals. However, the impact of OxtR modulation on acquisition, extinction and reinstatement of drug-elicited ethanol-conditioned place preference (EtOH-CPP) has not yet been investigated. In this study, we evaluated the effects of OxtR pharmacological modulation, using the oxytocin analog Carbetocin, and genetic overexpression in the nucleus accumbens (NAcc), using lentiviral-mediated gene transfer technology, of the OxtR on acquisition, extinction and reinstatement of drug-elicited EtOH-CPP in mice. In the first experiment, results showed that Carbetocin administration and NAcc OxtR-overexpression (LV-OxtR) reduced EtOH-CPP establishment. In the second experiment, systemic Carbetocin treatment and OxtR overexpression resulted in decreased time spent in the ethanol-paired compartment following completion of a 7-day extinction protocol. Finally, the third experiment showed that Carbetocin and LV-OxtR suppressed primed reinstatement of EtOH-CPP. It is concluded that pharmacological and genetic modulation of the OxtR can modulate the acquisition, extinction, and reinstatement of conditioned reinforcing effects of ethanol. Taken together, the current findings add to the growing literature on oxytocin neurotransmission modulation in the pharmacotherapy of ethanol addiction and alcoholism.
Collapse
Affiliation(s)
- Amine Bahi
- Department of Anatomy, Tawam Medical Campus, College of Medicine & Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates.
| |
Collapse
|
47
|
Ryan PJ, Krstew EV, Sarwar M, Gundlach AL, Lawrence AJ. Relaxin-3 mRNA levels in nucleus incertus correlate with alcohol and sucrose intake in rats. Drug Alcohol Depend 2014; 140:8-16. [PMID: 24837581 DOI: 10.1016/j.drugalcdep.2014.04.017] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Revised: 04/02/2014] [Accepted: 04/09/2014] [Indexed: 11/17/2022]
Abstract
BACKGROUND Chronic alcohol intake produces multiple neuroadaptive changes, including up- and down-regulation of neuropeptides and receptors. There are widespread projections of relaxin-3 containing neurons to, and abundant relaxin family peptide 3 receptor (RXFP3) expression within, brain regions involved in modulating alcohol intake. Recently we demonstrated the involvement of relaxin-3/RXFP3 signalling in alcohol-seeking in rats; therefore in this study we examined whether relaxin-3 and/or RXFP3 expression were altered by chronic alcohol intake in alcohol-preferring iP rats. METHODS Expression of relaxin-3 mRNA in the hindbrain nucleus incertus and RXFP3 radioligand binding levels in discrete forebrain regions were investigated following voluntary intake of alcohol or sucrose for 12 weeks, with a 2 day washout, using quantitative in situ hybridisation histochemistry and in vitro receptor autoradiography, respectively, in cohorts of adult, male iP rats. RESULTS Levels of relaxin-3 mRNA in the hindbrain nucleus incertus were positively correlated with the level of intake of both alcohol (r(12)=0.59, p=0.03) and sucrose (r(7)=0.70, p=0.04) in iP rats. Dense binding of the RXFP3-selective radioligand, [(125)]-R3/I5, was detected in hypothalamic and extrahypothalamic sites, but no significant changes in the density of RXFP3 were observed in the brain regions quantified following chronic sucrose or ethanol intake. CONCLUSIONS Our findings suggest high endogenous relaxin-3 expression may be associated with higher intake of rewarding substances, rather than its expression being regulated in response to their intake, consistent with an active role for the relaxin-3/RXFP3 system in modulating ingestive and alcohol-related behaviours.
Collapse
Affiliation(s)
- P J Ryan
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia; Florey Department of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - E V Krstew
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia
| | - M Sarwar
- Drug Discovery Biology, Monash Institute of Pharmaceutical Science, Parkville, Victoria, Australia; Department of Pharmacology, Monash University, Clayton, Victoria, Australia
| | - A L Gundlach
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia; Florey Department of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia; Department of Anatomy and Neuroscience, The University of Melbourne, Parkville, Victoria, Australia
| | - A J Lawrence
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia; Florey Department of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia.
| |
Collapse
|
48
|
Zhou Y, Kreek MJ. Alcohol: a stimulant activating brain stress responsive systems with persistent neuroadaptation. Neuropharmacology 2014; 87:51-8. [PMID: 24929109 DOI: 10.1016/j.neuropharm.2014.05.044] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Revised: 05/08/2014] [Accepted: 05/19/2014] [Indexed: 01/08/2023]
Abstract
Addictive diseases, including addiction to alcohol, opiates or cocaine, pose massive public health costs. Addictions are chronic relapsing brain diseases, caused by drug-induced direct effects and persistent neuroadaptations at the molecular, cellular and behavioral levels. These drug-type specific neuroadapations are mainly contributed by three factors: environment, including stress, the direct reinforcing effects of the drug on the CNS, and genetics. Results from animal models and basic clinical research (including human genetic study) have shown important interactions between the stress responsive systems and alcohol abuse. In this review we will discuss the involvement of the dysregulation of the stress responsive hypothalamic-pituitary-adrenal (HPA) axis in alcohol addiction (Section I). Addictions to specific drugs such as alcohol, psychostimulants and opiates (e.g., heroin) have some common direct or downstream effects on several brain stress-responsive systems, including vasopressin and its receptor system (Section II), POMC and mu opioid receptor system (Section III) and dynorphin and kappa opioid receptor systems (Section IV). Further understanding of these systems, through laboratory-based and translational studies, have the potential to optimize early interventions and to discover new treatment targets for the therapy of alcoholism. This article is part of the Special Issue entitled 'CNS Stimulants'.
Collapse
Affiliation(s)
- Yan Zhou
- Laboratory of the Biology of Addictive Diseases, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Mary Jeanne Kreek
- Laboratory of the Biology of Addictive Diseases, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA.
| |
Collapse
|
49
|
Drinking alcohol has sex-dependent effects on pair bond formation in prairie voles. Proc Natl Acad Sci U S A 2014; 111:6052-7. [PMID: 24711424 DOI: 10.1073/pnas.1320879111] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Alcohol use and abuse profoundly influences a variety of behaviors, including social interactions. In some cases, it erodes social relationships; in others, it facilitates sociality. Here, we show that voluntary alcohol consumption can inhibit male partner preference (PP) formation (a laboratory proxy for pair bonding) in socially monogamous prairie voles (Microtus ochrogaster). Conversely, female PP is not inhibited, and may be facilitated by alcohol. Behavior and neurochemical analysis suggests that the effects of alcohol on social bonding are mediated by neural mechanisms regulating pair bond formation and not alcohol's effects on mating, locomotor, or aggressive behaviors. Several neuropeptide systems involved in the regulation of social behavior (especially neuropeptide Y and corticotropin-releasing factor) are modulated by alcohol drinking during cohabitation. These findings provide the first evidence to our knowledge that alcohol has a direct impact on the neural systems involved in social bonding in a sex-specific manner, providing an opportunity to explore the mechanisms by which alcohol affects social relationships.
Collapse
|
50
|
Yuen KW, Garner JP, Carson DS, Keller J, Lembke A, Hyde SA, Kenna HA, Tennakoon L, Schatzberg AF, Parker KJ. Plasma oxytocin concentrations are lower in depressed vs. healthy control women and are independent of cortisol. J Psychiatr Res 2014; 51:30-6. [PMID: 24405552 PMCID: PMC6714974 DOI: 10.1016/j.jpsychires.2013.12.012] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2013] [Revised: 12/05/2013] [Accepted: 12/17/2013] [Indexed: 01/01/2023]
Abstract
The neuropeptide oxytocin (OT) promotes social behavior and attenuates stress responsivity in mammals. Recent clinical evidence suggests OT concentrations may be dysregulated in major depression. This study extends previous research by testing whether: 1) OT concentrations vary systematically in depressive disorders with and without hypercortisolemia, 2) gender differences in OT concentrations are observed in depressed vs. healthy control participants, and 3) OT concentrations are predictive of clinical phenotypes. Plasma OT concentrations of psychotic major depressive (PMD; n = 14: 10 female, 4 male), non-psychotic major depressive (NPMD; n = 17: 12 female, 5 male), and non-depressed, healthy control (n = 19: 11 female, 8 male) participants were assayed at 2000, 2400, 0400, and 0800 h. Plasma cortisol concentrations were quantified at 2300 h, and clinical phenotypes were determined. As expected, PMD participants, compared to NPMD and healthy control participants, showed higher plasma cortisol concentrations. Although both depressed groups showed similar OT concentrations, a significant interaction effect between group and gender was observed. Specifically, depressed females exhibited lower mean OT concentrations than depressed males. Further, depressed vs. healthy control female participants exhibited lower mean OT concentrations, whereas depressed vs. healthy control male participants showed a trend in the opposite direction. OT concentrations were also predictive of desirability, drug dependence, and compulsivity scores as measured by the Million Clinical Multiaxial Inventory-III. All findings were independent of cortisol. These data suggest that OT signaling may provide a mechanism by which to better understand female-biased risk to develop depressive disorders and that plasma OT concentrations may be a useful biomarker of certain clinical phenotypes.
Collapse
Affiliation(s)
- Kaeli W. Yuen
- Department of Psychiatry and Behavioral Sciences, School of Medicine, Stanford University, 1201 Welch Rd. MSLS Room P104, Stanford, CA 94305-5485, USA
| | - Joseph P. Garner
- Department of Psychiatry and Behavioral Sciences, School of Medicine, Stanford University, 1201 Welch Rd. MSLS Room P104, Stanford, CA 94305-5485, USA,Department of Comparative Medicine, School of Medicine, Stanford University, 287 Campus Dr., Stanford, CA 94305-5410, USA
| | - Dean S. Carson
- Department of Psychiatry and Behavioral Sciences, School of Medicine, Stanford University, 1201 Welch Rd. MSLS Room P104, Stanford, CA 94305-5485, USA
| | - Jennifer Keller
- Department of Psychiatry and Behavioral Sciences, School of Medicine, Stanford University, 401 Quarry Rd., Stanford, CA 94305-5719, USA
| | - Anna Lembke
- Department of Psychiatry and Behavioral Sciences, School of Medicine, Stanford University, 1201 Welch Rd. MSLS Room P104, Stanford, CA 94305-5485, USA
| | - Shellie A. Hyde
- Department of Psychiatry and Behavioral Sciences, School of Medicine, Stanford University, 1201 Welch Rd. MSLS Room P104, Stanford, CA 94305-5485, USA
| | - Heather A. Kenna
- Department of Psychiatry and Behavioral Sciences, School of Medicine, Stanford University, 401 Quarry Rd., Stanford, CA 94305-5719, USA
| | - Lakshika Tennakoon
- Department of Psychiatry and Behavioral Sciences, School of Medicine, Stanford University, 401 Quarry Rd., Stanford, CA 94305-5719, USA
| | - Alan F. Schatzberg
- Department of Psychiatry and Behavioral Sciences, School of Medicine, Stanford University, 401 Quarry Rd., Stanford, CA 94305-5719, USA
| | - Karen J. Parker
- Department of Psychiatry and Behavioral Sciences, School of Medicine, Stanford University, 1201 Welch Rd. MSLS Room P104, Stanford, CA 94305-5485, USA,Corresponding author. Tel.: +1 650 736 9863; fax: +1 (650) 498 7761. (K.J. Parker)
| |
Collapse
|