1
|
Cornejo NE, McNeely EC, Yates TQ, Kaneko M, Leong KC. Oxytocin attenuates yohimbine-induced responding for oral oxycodone under a progressive ratio schedule in male and female rats. Behav Brain Res 2025; 488:115598. [PMID: 40268056 DOI: 10.1016/j.bbr.2025.115598] [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: 02/26/2025] [Revised: 04/07/2025] [Accepted: 04/18/2025] [Indexed: 04/25/2025]
Abstract
Opioid Use Disorder (OUD) has become an epidemic in the United States, with oxycodone (OXY) being one of the most widely misused opioids. Stress plays a key role in triggering opioid use, which can lead to addiction and relapse, underscoring the urgent need for effective therapeutic interventions. Recent studies suggest that the neuropeptide oxytocin (OXT) may reduce addiction-related behaviors and possess anxiolytic properties. The present study investigates the effect of peripheral administration of OXT on attenuating stress-induced motivation to seek oral OXY, as measured by progressive ratio (PR) responding in both male and female rats. Animals were first trained in an operant conditioning paradigm to orally self-administer a sucrose solution by pressing an active lever for access to the solution. As responding stabilized, subjects were switched to an OXY-sucrose solution, with sucrose concentration reduced overtime, until subjects were self-administering oral OXY alone. To test the effect of stress on OXY responding the pharmacological stressor, yohimbine (YOH), was administered prior to a progressive ratio test in which animals were required to produce increasingly higher responses to receive a single exposure to OXY. Through a within subjects design, when OXT was concurrently administered, this YOH-induced enhancement of OXY reward strength was attenuated in both male and female rats. These results suggest that OXT may serve as a potential therapeutic remedy to mitigate the deleterious effects of stress on OXY addiction in both sexes.
Collapse
Affiliation(s)
- Natalie E Cornejo
- Department of Psychology, Trinity University, San Antonio, TX 78212, USA
| | | | - Taylor Q Yates
- Department of Psychology, Trinity University, San Antonio, TX 78212, USA
| | - Moe Kaneko
- Department of Psychology, Trinity University, San Antonio, TX 78212, USA
| | - Kah-Chung Leong
- Department of Psychology, Trinity University, San Antonio, TX 78212, USA.
| |
Collapse
|
2
|
Zarei H, Sharafinezhad M, Hasani K. The interplay of oxytocin and dopaminergic system: Effects on food consumption in broiler chickens. Poult Sci 2025; 104:105139. [PMID: 40199125 PMCID: PMC12005875 DOI: 10.1016/j.psj.2025.105139] [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: 02/13/2025] [Revised: 04/01/2025] [Accepted: 04/03/2025] [Indexed: 04/10/2025] Open
Abstract
Oxytocin play a crucial physiological role in regulating feed consumption and maintaining energy homeostasis. Numerous studies have highlighted the influence of central dopaminergic systems on appetite control; however, there is a lack of information regarding their interaction with food intake regulation in avian species. Consequently, the present study aimed to investigate the potential interplay between the central oxytocin and dopaminergic systems in modulating food consumption in broiler chicks. In the initial experiment, various doses of oxytocin (2.5, 5, and 10 µg) and saline were administered via intracerebroventricular (ICV) injection to ascertain the effective dosage of oxytocin. The second experiment involved ICV injections of saline, SCH23390 (a D1 receptor antagonist), the identified effective dose of oxytocin, and a co-injection of SCH23390 with oxytocin. Subsequent experiments (3 to 7) followed a similar methodology, substituting SCH23390 with AMI-193 (a D2 receptor antagonist), NGB2904 (a D3 receptor antagonist), l-741,742 (a D4 receptor antagonist), 6-hydroxydopamine (a dopaminergic neurotoxin), and l-DOPA (a dopamine precursor), respectively. Cumulative food intake was measured over a period of 120 min post-injection. The findings revealed that ICV administration of oxytocin significantly reduced feed consumption in broilers compared to the control group (P < 0.05). Additionally, the co-infusion of SCH23390 and 6-hydroxydopamine alongside oxytocin significantly mitigated the hypophagic effect induced by oxytocin (P < 0.05). In contrast, AMI-193, NGB2904, l-741,742, and l-DOPA did not exhibit any significant influence on oxytocin-induced hypophagia (P ≥ 0.05). These findings suggest that the anorexigenic effect of oxytocin may be primarily mediated through D1 dopaminergic receptors in broilers.
Collapse
Affiliation(s)
- Hamed Zarei
- Department of Biology, Faculty of Basic Science, Central Tehran Branch, Islamic Azad University, Tehran, Iran.
| | - Mobina Sharafinezhad
- Department of Basic Sciences of Veterinary Medicine, Garmsar Branch, Islamic Azad University, Garmsar, Iran
| | - Keyvan Hasani
- Department of Basic Sciences of Veterinary Medicine, Garmsar Branch, Islamic Azad University, Garmsar, Iran
| |
Collapse
|
3
|
Yao S, Kendrick KM. How does oxytocin modulate human behavior? Mol Psychiatry 2025; 30:1639-1651. [PMID: 39827220 DOI: 10.1038/s41380-025-02898-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 12/19/2024] [Accepted: 01/14/2025] [Indexed: 01/22/2025]
Abstract
While the highly evolutionarily conserved hypothalamic neuropeptide, oxytocin (OT) can influence cognitive, emotional and social functions, and may have therapeutic potential in disorders with social dysfunction, it is still unclear how it acts. Here, we review the most established findings in both animal model and human studies regarding stimuli which evoke OT release, its primary functional effects and the mechanisms whereby exogenous administration influences brain and behavior. We also review progress on whether OT administration can improve social symptoms in autism spectrum disorder and schizophrenia and consider possible impediments to translational success. Importantly, we emphasize that OT acting via its extensive central or peripheral receptors primarily influences behavior indirectly through neuromodulatory interactions with classical transmitters and other peptides which themselves can independently influence behavior. We also emphasize that exogenous administration studies increasingly demonstrate peripheral effects of OT may be of greater importance than originally thought, especially involving the vagus. Overall, we propose a hierarchical model whereby OT's neuromodulatory actions influence behavior across interconnected functional domains and ultimately help to promote survival, security and sociability. Initially, OT potently facilitates attention to salient social and other important stimuli and additionally modulates cognitive, emotional and reward processing in a person- and context-dependent manner to promote interpersonal social understanding, attraction and bonds on the one hand and social group cohesion through increased conformity, altruistic punishment and moral emotions on the other. OT also increases co-operation and protection across both social domains. We hope this review and model will promote further research and help aid future translation success.
Collapse
Affiliation(s)
- Shuxia Yao
- The Center of Psychosomatic Medicine, Sichuan Provincial Center for Mental Health, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for Neuroinformation, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 611731, China.
| | - Keith M Kendrick
- The Center of Psychosomatic Medicine, Sichuan Provincial Center for Mental Health, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for Neuroinformation, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 611731, China.
| |
Collapse
|
4
|
Engeln M, Ahmed SH. Remission from addiction: erasing the wrong circuits or making new ones? Nat Rev Neurosci 2025; 26:115-130. [PMID: 39663409 DOI: 10.1038/s41583-024-00886-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/07/2024] [Indexed: 12/13/2024]
Abstract
Chronic relapse is a hallmark of substance-use disorders (SUDs), but many people with SUDs do recover and eventually enter remission. Many preclinical studies in this field aim to identify interventions that can precipitate recovery by reversing or erasing the neuronal circuit changes caused by chronic drug use. A better understanding of remission from SUDs can also come from preclinical studies that model factors known to influence recovery in humans, such as the negative consequences of drug use and positive environmental influences. In this Perspective we discuss human neuroimaging studies that have provided information about recovery from SUDs and highlight mechanisms identified in preclinical studies - such as the reconfiguration of neuronal circuits - that could contribute to remission. We also analyse how studies of memory and forgetting can provide insights into the mechanisms of remission. Overall, we propose that remission can be driven by the introduction of new neuronal changes (which outcompete those induced by drugs) as well as by the erasure of drug-induced changes.
Collapse
Affiliation(s)
- Michel Engeln
- Univ. Bordeaux, CNRS, INCIA, UMR 5287, Bordeaux, France.
| | - Serge H Ahmed
- Univ. Bordeaux, CNRS, INCIA, UMR 5287, Bordeaux, France
| |
Collapse
|
5
|
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
|
6
|
Faraji M, Viera-Resto OA, Berrios BJ, Bizon JL, Setlow B. Effects of systemic oxytocin receptor activation and blockade on risky decision making in female and male rats. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.13.593981. [PMID: 38798601 PMCID: PMC11118492 DOI: 10.1101/2024.05.13.593981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
The neuropeptide oxytocin is traditionally known for its roles in parturition, lactation, and social behavior. Other data, however, show that oxytocin can modulate behaviors outside of these contexts, including drug self-administration and some aspects of cost-benefit decision making. Here we used a pharmacological approach to investigate the contributions of oxytocin signaling to decision making under risk of explicit punishment. Female and male Long-Evans rats were trained on a risky decision-making task in which they chose between a small, "safe" food reward and a large, "risky" food reward that was accompanied by varying probabilities of mild footshock. Once stable choice behavior emerged, rats were tested in the task following acute intraperitoneal injections of oxytocin or the oxytocin receptor antagonist L-368,899. Neither drug affected task performance in males. In females, however, both oxytocin and L-368,899 caused a dose-dependent reduction in preference for large risky reward. Control experiments showed that these effects could not be accounted for by alterations in food motivation or shock sensitivity. Together, these results reveal a sex-dependent effect of oxytocin signaling on risky decision making in rats.
Collapse
Affiliation(s)
- Mojdeh Faraji
- Department of Psychiatry, University of Florida
- Center for Addiction Research and Education, University of Florida
| | | | | | - Jennifer L Bizon
- Center for Addiction Research and Education, University of Florida
- Department of Neuroscience, University of Florida
- McKnight Brain Institute, University of Florida
| | - Barry Setlow
- Department of Psychiatry, University of Florida
- Center for Addiction Research and Education, University of Florida
- McKnight Brain Institute, University of Florida
| |
Collapse
|
7
|
Girella A, Di Bartolomeo M, Dainese E, Buzzelli V, Trezza V, D'Addario C. Fatty Acid Amide Hydrolase and Cannabinoid Receptor Type 1 Genes Regulation is Modulated by Social Isolation in Rats. Neurochem Res 2024; 49:1278-1290. [PMID: 38368587 DOI: 10.1007/s11064-024-04117-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 01/08/2024] [Accepted: 01/25/2024] [Indexed: 02/19/2024]
Abstract
Social isolation is a state of lack of social connections, involving the modulation of different molecular signalling cascades and associated with high risk of mental health issues. To investigate if and how gene expression is modulated by social experience at the central level, we analyzed the effects of 5 weeks of social isolation in rats focusing on endocannabinoid system genes transcription in key brain regions involved in emotional control. We observed selective reduction in mRNA levels for fatty acid amide hydrolase (Faah) and cannabinoid receptor type 1 (Cnr1) genes in the amygdala complex and of Cnr1 in the prefrontal cortex of socially isolated rats when compared to controls, and these changes appear to be partially driven by trimethylation of Lysine 27 and acetylation of Lysine 9 at Histone 3. The alterations of Cnr1 transcriptional regulation result also directly correlated with those of oxytocin receptor gene. We here suggest that to counteract the effects of SI, it is of relevance to restore the endocannabinoid system homeostasis via the use of environmental triggers able to revert those epigenetic mechanisms accounting for the alterations observed.
Collapse
Affiliation(s)
- Antonio Girella
- Department of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Via Renato Balzarini, 1, 64100, Teramo, Italy
| | - Martina Di Bartolomeo
- Department of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Via Renato Balzarini, 1, 64100, Teramo, Italy
| | - Enrico Dainese
- Department of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Via Renato Balzarini, 1, 64100, Teramo, Italy
| | | | - Viviana Trezza
- Department of Science, Roma Tre University, Rome, Italy
- Neuroendocrinology, Metabolism and Neuropharmacology Unit, IRCSS Fondazione Santa Lucia, Rome, Italy
| | - Claudio D'Addario
- Department of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Via Renato Balzarini, 1, 64100, Teramo, Italy.
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
8
|
Fang Y, Reinl EL, Liu A, Prochaska TD, Malik M, Frolova AI, England SK, Imoukhuede PI. Quantification of surface-localized and total oxytocin receptor in myometrial smooth muscle cells. Heliyon 2024; 10:e25761. [PMID: 38384573 PMCID: PMC10878913 DOI: 10.1016/j.heliyon.2024.e25761] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 01/31/2024] [Accepted: 02/01/2024] [Indexed: 02/23/2024] Open
Abstract
Oxytocin acts through the oxytocin receptor (OXTR) to modulate uterine contractility. We previously identified OXTR genetic variants and showed that, in HEK293T cells, two of the OXTR protein variants localized to the cell surface less than wild-type OXTR. Here, we sought to measure OXTR in the more native human myometrial smooth muscle cell (HMSMC) line on both the cell-surface and across the whole cell, and used CRISPR editing to add an HA tag to the endogenous OXTR gene for anti-HA measurement. Quantitative flow cytometry revealed that these cells possessed 55,000 ± 3200 total OXTRs and 4900 ± 390 cell-surface OXTRs per cell. To identify any differential wild-type versus variant localization, we transiently transfected HMSMCs to exogenously express wild-type or variant OXTR with HA and green fluorescent protein tags. Total protein expression of wild-type OXTR and all tested variants were similar. However, the two variants with lower surface localization in HEK293T cells also presented lower surface localization in HMSMCs. Overall, we confirm the differential surface localization of variant OXTR in a more native cell type, and further demonstrate that the quantitative flow cytometry technique is adaptable to whole-cell measurements.
Collapse
Affiliation(s)
- Yingye Fang
- Department of Bioengineering, University of Washington, Seattle, WA, 98109, USA
| | - Erin L. Reinl
- Department of Obstetrics and Gynecology, Center for Reproductive Health Sciences, Washington University School of Medicine in St. Louis, St. Louis, MO, 63110, USA
| | - Audrey Liu
- Department of Obstetrics and Gynecology, Center for Reproductive Health Sciences, Washington University School of Medicine in St. Louis, St. Louis, MO, 63110, USA
| | - Trinidi D. Prochaska
- Department of Obstetrics and Gynecology, Center for Reproductive Health Sciences, Washington University School of Medicine in St. Louis, St. Louis, MO, 63110, USA
| | - Manasi Malik
- Department of Obstetrics and Gynecology, Center for Reproductive Health Sciences, Washington University School of Medicine in St. Louis, St. Louis, MO, 63110, USA
| | - Antonina I. Frolova
- Department of Obstetrics and Gynecology, Center for Reproductive Health Sciences, Washington University School of Medicine in St. Louis, St. Louis, MO, 63110, USA
| | - Sarah K. England
- Department of Obstetrics and Gynecology, Center for Reproductive Health Sciences, Washington University School of Medicine in St. Louis, St. Louis, MO, 63110, USA
| | | |
Collapse
|
9
|
Chang HM, Chen C, Lu ML, Jou S, Santos VHJ, Goh KK. The interplay of childhood trauma, oxytocin, and impulsivity in predicting the onset of methamphetamine use. CHILD ABUSE & NEGLECT 2024; 147:106579. [PMID: 38048654 DOI: 10.1016/j.chiabu.2023.106579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 11/19/2023] [Accepted: 11/23/2023] [Indexed: 12/06/2023]
Abstract
BACKGROUND Childhood trauma is associated with substance use disorders, including methamphetamine use disorder (MUD). Oxytocin, involved in social bonding, stress regulation, and reward processing, may influence addiction vulnerability and impulsivity in individuals with a history of childhood trauma. OBJECTIVE To investigate the relationships among childhood trauma, oxytocin levels, impulsivity, and the age of first methamphetamine use in individuals with MUD. PARTICIPANTS AND SETTING The study included 298 male participants (148 individuals with MUD and 150 healthy controls) from both probation offices and psychiatric clinics. METHODS Childhood trauma was assessed using the Childhood Trauma Questionnaire-Short Form (CTQ-SF), impulsivity with the Barratt Impulsiveness Scale-11 (BIS-11), and plasma oxytocin levels were obtained. RESULTS Individuals with MUD exhibited higher levels of childhood trauma, impulsivity, and lower plasma oxytocin levels compared to healthy controls. Childhood trauma was associated with a younger age of first methamphetamine use, higher impulsivity, and lower oxytocin levels among individuals with MUD. Plasma oxytocin levels partially mediated the relationship between childhood trauma and both the age of first methamphetamine use and impulsivity. Serial mediation analysis demonstrated that oxytocin levels and impulsivity sequentially mediated the relationship between childhood trauma and the age of first methamphetamine use. CONCLUSIONS The findings reveal the complex interplay among childhood trauma, oxytocin, impulsivity, and methamphetamine use, emphasizing the importance of considering these factors in prevention and intervention strategies for MUD. Future research should explore oxytocin and impulsivity-focused interventions to mitigate the effects of childhood trauma and reduce MUD development risk.
Collapse
Affiliation(s)
- Hu-Ming Chang
- Department of Addiction Sciences, Taipei City Psychiatric Center, Taipei City Hospital, Taipei, Taiwan; Psychiatric Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Chenyi Chen
- Psychiatric Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan; Graduate Institute of Injury Prevention and Control, College of Public Health, Taipei Medical University, Taipei, Taiwan; Graduate Institute of Mind, Brain and Consciousness, College of Humanities and Social Sciences, Taipei Medical University, Taipei, Taiwan; Neuroscience Research Center, Taipei Medical University Hospital, Taipei, Taiwan; Department of Physical Medicine and Rehabilitation, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan; The Innovative and Translational Research Center for Brain Consciousness, Taipei Medical University, Taipei, Taiwan
| | - Mong-Liang Lu
- Psychiatric Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan; Department of Psychiatry, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan; Department of Psychiatry, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Susyan Jou
- Psychiatric Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan; Graduate School of Criminology, National Taipei University, Taipei, Taiwan
| | - Vitor Hugo Jesus Santos
- Department of Psychiatry and Mental Health, Faculty of Health Sciences (FCS-UBI), Cova da Beira University Hospital Center, Covilhã, Portugal
| | - Kah Kheng Goh
- Psychiatric Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan; Graduate Institute of Injury Prevention and Control, College of Public Health, Taipei Medical University, Taipei, Taiwan; The Innovative and Translational Research Center for Brain Consciousness, Taipei Medical University, Taipei, Taiwan; Department of Psychiatry, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan; Department of Psychiatry, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
10
|
Penrod RD, Taniguchi M, Kearns AM, Hopkins JL, Reichel CM. Differential Roles of Oxytocin Receptors in the Prefrontal Cortex and Nucleus Accumbens on Cocaine Self-Administration and Reinstatement of Cued Cocaine Seeking in Male Rats. Int J Neuropsychopharmacol 2023; 26:817-827. [PMID: 37875346 PMCID: PMC10726405 DOI: 10.1093/ijnp/pyad059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 10/23/2023] [Indexed: 10/26/2023] Open
Abstract
BACKGROUND Little is known about the specific roles of cortical and accumbal oxytocin receptors in drug use disorders. To better understand the importance of the endogenous oxytocin system in cocaine relapse behavior, we developed an adeno-associated viral vector-expressing short hairpin (sh) RNAs to selectively degrade the rat oxytocin receptor (OxyR) mRNA in vivo. METHODS Male (Sprague-Dawley) rats received bilateral infusions of the shRNA for the oxytocin receptor (shOxyR) or an shRNA control virus into the prefrontal cortex (PFC) or the nucleus accumbens core (NAc). Rats self-administered cocaine on an escalating FR ratio for 14 days, lever responding was extinguished, and rats were tested for cued and cocaine-primed reinstatement of drug seeking. RESULTS OxyR knockdown in the PFC delayed the acquisition of lever pressing on an fixed ratio 1 schedule of reinforcement. All rats eventually acquired the same level of lever pressing and discrimination, and there were no differences in extinction. OxyR knockdown in the NAc had no effect during acquisition. In both the PFC and NAc, the shOxyR decreased cued reinstatement relative to shRNA control virus but was without effect during drug-primed reinstatement. OxyR knockdown in the PFC increased chamber activity during a social interaction task. CONCLUSIONS This study provides critical new information about how endogenous OxyRs function to affect drug seeking in response to different precipitators of relapse. The tool developed to knockdown OxyRs in rat could provide important new insights that aid development of oxytocin-based therapeutics to reduce return-to-use episodes in people with substance use disorder and other neuropsychiatric disorders.
Collapse
Affiliation(s)
- Rachel D Penrod
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Makoto Taniguchi
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Angela M Kearns
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Jordan L Hopkins
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Carmela M Reichel
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina, USA
| |
Collapse
|
11
|
Tereshchenko SY. Neurobiological risk factors for problematic social media use as a specific form of Internet addiction: A narrative review. World J Psychiatry 2023; 13:160-173. [PMID: 37303928 PMCID: PMC10251362 DOI: 10.5498/wjp.v13.i5.160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 02/13/2023] [Accepted: 04/07/2023] [Indexed: 05/19/2023] Open
Abstract
Problematic social media use (PSMU) is a behavioral addiction, a specific form of problematic Internet use associated with the uncontrolled use of social networks. It is typical mostly for modern adolescents and young adults, which are the first generations fully grown up in the era of total digitalization of society. The modern biopsychosocial model of the formation of behavioral addictions, postulating the impact of a large number of biological, psychological, and social factors on addictive behavior formation, may be quite applicable to PSMU. In this narrative review, we discussed neurobiological risk factors for Internet addiction with a focus on current evidence on the association between PSMU and structural/ functional characteristics of the brain and autonomic nervous system, neurochemical correlations, and genetic features. A review of the literature shows that the vast majority of the mentioned neurobiological studies were focused on computer games addiction and generalized Internet addiction (without taking into account the consumed content). Even though a certain number of neuroimaging studies have been conducted for PSMU, there is practically no research on neuropeptide and genetic associations for PSMU to date. This fact points to the extremely high relevance of such studies.
Collapse
Affiliation(s)
- Sergey Yu Tereshchenko
- Department of Child's Physical and Mental Health, Federal Research Center “Krasnoyarsk Science Center of the Siberian Branch of the Russian Academy of Sciences”, Research Institute of Medical Problems of the North, Krasnoyarsk 660022, Russia
| |
Collapse
|
12
|
Potretzke S, Zhang Y, Li J, Fecteau KM, Erikson DW, Hibert M, Ryabinin AE. Male-selective effects of oxytocin agonism on alcohol intake: behavioral assessment in socially housed prairie voles and involvement of RAGE. Neuropsychopharmacology 2023; 48:920-928. [PMID: 36369481 PMCID: PMC10156683 DOI: 10.1038/s41386-022-01490-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 10/20/2022] [Accepted: 10/25/2022] [Indexed: 11/13/2022]
Abstract
Targeting the oxytocin (OXT) peptide system has emerged as a promising new approach for the treatment of alcohol use disorder (AUD). However, further advancements in this development depend on properly modeling various complex social aspects of AUD and its treatment. Here we examined behavioral and molecular underpinnings of OXT receptor (OXTR) agonism in prairie voles, a rodent species with demonstrated translational validity for neurobiological mechanisms regulating social affiliations. To further improve translational validity of these studies, we examined effects of intranasal (IN) OXT administration in male and female prairie voles socially housed in the presence of untreated cagemates. IN OXT selectively inhibited alcohol drinking in male, but not female, animals. Further, we confirmed that exogenously administered OXT penetrates the prairie vole brain and showed that Receptor for Advanced Glycation End-products assists this penetration after IN, but not intraperitoneal (IP), OXT administration. Finally, we demonstrated that IP administration of LIT-001, a small-molecule OXTR agonist, inhibits alcohol intake in male, but not female, prairie voles socially housed in the presence of untreated cagemates. Taken together, results of this study support the promise of selectively targeting OXTR for individualized treatment of AUD.
Collapse
Affiliation(s)
- Sheena Potretzke
- Department of Behavioral Neuroscience, School of Medicine, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Yangmiao Zhang
- Department of Behavioral Neuroscience, School of Medicine, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Ju Li
- Department of Behavioral Neuroscience, School of Medicine, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Kristopher M Fecteau
- Endocrine Technologies Core, Oregon National Primate Research Center, Oregon Health & Science University, 505 NW 185th Avenue, Beaverton, OR, 97006, USA
| | - David W Erikson
- Endocrine Technologies Core, Oregon National Primate Research Center, Oregon Health & Science University, 505 NW 185th Avenue, Beaverton, OR, 97006, USA
| | - Marcel Hibert
- Laboratoire d'Innovation Thérapeutique, Faculté de Pharmacie, UMR7200 CNRS/Université de Strasbourg, 74 Route du Rhin, F-67412, Illkirch, France
| | - Andrey E Ryabinin
- Department of Behavioral Neuroscience, School of Medicine, Oregon Health & Science University, Portland, OR, 97239, USA.
| |
Collapse
|
13
|
Fernández-Rodríguez S, Cano-Cebrián MJ, Esposito-Zapero C, Pérez S, Guerri C, Zornoza T, Polache A. N-Acetylcysteine normalizes brain oxidative stress and neuroinflammation observed after protracted ethanol abstinence: a preclinical study in long-term ethanol-experienced male rats. Psychopharmacology (Berl) 2023; 240:725-738. [PMID: 36708386 PMCID: PMC10006045 DOI: 10.1007/s00213-023-06311-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 01/04/2023] [Indexed: 01/29/2023]
Abstract
RATIONALE Using a preclinical model based on the Alcohol Deprivation Effect (ADE), we have reported that N-Acetylcysteine (NAC) can prevent the relapse-like drinking behaviour in long-term ethanol-experienced male rats. OBJECTIVES To investigate if chronic ethanol intake and protracted abstinence affect several glutamate transporters and whether NAC, administered during the withdrawal period, could restore the ethanol-induced brain potential dysfunctions. Furthermore, the antioxidant and anti-inflammatory effects of NAC during abstinence in rats under the ADE paradigm were also explored. METHODS The expression of GLT1, GLAST and xCT in nucleus accumbens (Nacc) and dorsal striatum (DS) of male Wistar was analysed after water and chronic ethanol intake. We used the model based on the ADE within another cohort of male Wistar rats. During the fourth abstinence period, rats were treated for 9 days with vehicle or NAC (60, 100 mg/kg; s.c.). The effects of NAC treatment on (i) glutamate transporters expression in the Nacc and DS, (ii) the oxidative status in the hippocampus (Hip) and amygdala (AMG) and (iii) some neuroinflammatory markers in prefrontal cortex (PFC) were tested. RESULTS NAC chronic administration during protracted abstinence restored oxidative stress markers (GSSG and GGSH/GSH) in the Hip. Furthermore, NAC was able to normalize some neuroinflammation markers in PFC without normalizing the observed downregulation of GLT1 and GLAST in Nacc. CONCLUSIONS NAC restores brain oxidative stress and neuroinflammation that we previously observed after protracted ethanol abstinence in long-term ethanol-experienced male rats. This NAC effect could be a plausible mechanism for its anti-relapse effect. Also, brain oxidative stress and neuroinflammation could represent and identify plausible targets for searching new anti-relapse pharmacotherapies.
Collapse
Affiliation(s)
- Sandra Fernández-Rodríguez
- Department of Pharmacy and Pharmaceutical Technology and Parasitology, Faculty of Pharmacy, University of Valencia, Burjassot, 46100, Valencia, Spain
| | - María José Cano-Cebrián
- Department of Pharmacy and Pharmaceutical Technology and Parasitology, Faculty of Pharmacy, University of Valencia, Burjassot, 46100, Valencia, Spain
| | - Claudia Esposito-Zapero
- Department of Pharmacy and Pharmaceutical Technology and Parasitology, Faculty of Pharmacy, University of Valencia, Burjassot, 46100, Valencia, Spain
| | - Salvador Pérez
- Department of Physiology, Faculty of Pharmacy, University of Valencia, Burjassot, 46100, Valencia, Spain
| | - Consuelo Guerri
- Department of Molecular and Cellular Pathology of Alcohol, Príncipe Felipe Research Center, Valencia, Spain
| | - Teodoro Zornoza
- Department of Pharmacy and Pharmaceutical Technology and Parasitology, Faculty of Pharmacy, University of Valencia, Burjassot, 46100, Valencia, Spain
| | - Ana Polache
- Department of Pharmacy and Pharmaceutical Technology and Parasitology, Faculty of Pharmacy, University of Valencia, Burjassot, 46100, Valencia, Spain.
| |
Collapse
|
14
|
Asker M, Krieger JP, Liles A, Tinsley IC, Borner T, Maric I, Doebley S, Furst CD, Börchers S, Longo F, Bhat YR, De Jonghe BC, Hayes MR, Doyle RP, Skibicka KP. Peripherally restricted oxytocin is sufficient to reduce food intake and motivation, while CNS entry is required for locomotor and taste avoidance effects. Diabetes Obes Metab 2023; 25:856-877. [PMID: 36495318 PMCID: PMC9987651 DOI: 10.1111/dom.14937] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 11/21/2022] [Accepted: 12/02/2022] [Indexed: 12/14/2022]
Abstract
OBJECTIVES Oxytocin (OT) has a well-established role in reproductive behaviours; however, it recently emerged as an important regulator of energy homeostasis. In addition to central nervous system (CNS), OT is found in the plasma and OT receptors (OT-R) are found in peripheral tissues relevant to energy balance regulation. Here, we aim to determine whether peripheral OT-R activation is sufficient to alter energy intake and expenditure. METHODS AND RESULTS We first show that systemic OT potently reduced food intake and food-motivated behaviour for a high-fat reward in male and female rats. As it is plausible that peripherally, intraperitoneally (IP) injected OT crosses the blood-brain barrier (BBB) to produce some of the metabolic effects within the CNS, we screened, with a novel fluorescently labelled-OT (fAF546-OT, Roxy), for the presence of IP-injected Roxy in CNS tissue relevant to feeding control and compared such with BBB-impermeable fluorescent OT-B12 (fCy5-OT-B12; BRoxy). While Roxy did penetrate the CNS, BRoxy did not. To evaluate the behavioural and thermoregulatory impact of exclusive activation of peripheral OT-R, we generated a novel BBB-impermeable OT (OT-B12 ), with equipotent binding at OT-R in vitro. In vivo, IP-injected OT and OT-B12 were equipotent at food intake suppression in rats of both sexes, suggesting that peripheral OT acts on peripheral OT-R to reduce feeding behaviour. Importantly, OT induced a potent conditioned taste avoidance, indistinguishable from that induced by LiCl, when applied peripherally. Remarkably, and in contrast to OT, OT-B12 did not induce any conditioned taste avoidance. Limiting the CNS entry of OT also resulted in a dose-dependent reduction of emesis in male shrews. While both OT and OT-B12 proved to have similar effects on body temperature, only OT resulted in home-cage locomotor depression. CONCLUSIONS Together our data indicate that limiting systemic OT CNS penetrance preserves the anorexic effects of the peptide and reduces the clinically undesired side effects of OT: emesis, taste avoidance and locomotor depression. Thus, therapeutic targeting of peripheral OT-R may be a viable strategy to achieve appetite suppression with better patient outcomes.
Collapse
Affiliation(s)
- Mohammed Asker
- Department of Physiology/Metabolic Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Wallenberg Centre for molecular and translational medicine, University of Gothenburg, Gothenburg, Sweden
| | - Jean-Philippe Krieger
- Department of Physiology/Metabolic Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Amber Liles
- Department of Chemistry, Syracuse University, Syracuse, New York, USA
| | - Ian C Tinsley
- Department of Chemistry, Syracuse University, Syracuse, New York, USA
| | - Tito Borner
- Department of Biobehavioral Health Sciences, University of Pennsylvania, School of Nursing, Philadelphia, Pennsylvania, USA
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Ivana Maric
- Department of Physiology/Metabolic Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Department of Chemistry, Syracuse University, Syracuse, New York, USA
- Department of Nutritional Sciences, Pennsylvania State University, University Park, Pennsylvania, USA
| | - Sarah Doebley
- Department of Biobehavioral Health Sciences, University of Pennsylvania, School of Nursing, Philadelphia, Pennsylvania, USA
| | - C Daniel Furst
- Department of Biobehavioral Health Sciences, University of Pennsylvania, School of Nursing, Philadelphia, Pennsylvania, USA
| | - Stina Börchers
- Department of Physiology/Metabolic Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Wallenberg Centre for molecular and translational medicine, University of Gothenburg, Gothenburg, Sweden
| | - Francesco Longo
- Department of Physiology/Metabolic Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Yashaswini R Bhat
- Department of Nutritional Sciences, Pennsylvania State University, University Park, Pennsylvania, USA
| | - Bart C De Jonghe
- Department of Biobehavioral Health Sciences, University of Pennsylvania, School of Nursing, Philadelphia, Pennsylvania, USA
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Matthew R Hayes
- Department of Biobehavioral Health Sciences, University of Pennsylvania, School of Nursing, Philadelphia, Pennsylvania, USA
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Robert P Doyle
- Department of Chemistry, Syracuse University, Syracuse, New York, USA
- Departments of Medicine and Pharmacology, State University of New York, Upstate Medical University, Syracuse, New York, USA
| | - Karolina P Skibicka
- Department of Physiology/Metabolic Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Wallenberg Centre for molecular and translational medicine, University of Gothenburg, Gothenburg, Sweden
- Department of Nutritional Sciences, Pennsylvania State University, University Park, Pennsylvania, USA
| |
Collapse
|
15
|
Heteromerization of Dopamine D2 and Oxytocin Receptor in Adult Striatal Astrocytes. Int J Mol Sci 2023; 24:ijms24054677. [PMID: 36902106 PMCID: PMC10002782 DOI: 10.3390/ijms24054677] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 02/22/2023] [Accepted: 02/23/2023] [Indexed: 03/06/2023] Open
Abstract
The ability of oxytocin (OT) to interact with the dopaminergic system through facilitatory D2-OT receptor (OTR) receptor-receptor interaction in the limbic system is increasingly considered to play roles in social or emotional behavior, and suggested to serve as a potential therapeutic target. Although roles of astrocytes in the modulatory effects of OT and dopamine in the central nervous system are well recognized, the possibility of D2-OTR receptor-receptor interaction in astrocytes has been neglected. In purified astrocyte processes from adult rat striatum, we assessed OTR and dopamine D2 receptor expression by confocal analysis. The effects of activation of these receptors were evaluated in the processes through a neurochemical study of glutamate release evoked by 4-aminopyridine; D2-OTR heteromerization was assessed by co-immunoprecipitation and proximity ligation assay (PLA). The structure of the possible D2-OTR heterodimer was estimated by a bioinformatic approach. We found that both D2 and OTR were expressed on the same astrocyte processes and controlled the release of glutamate, showing a facilitatory receptor-receptor interaction in the D2-OTR heteromers. Biochemical and biophysical evidence confirmed D2-OTR heterodimers on striatal astrocytes. The residues in the transmembrane domains four and five of both receptors are predicted to be mainly involved in the heteromerization. In conclusion, roles for astrocytic D2-OTR in the control of glutamatergic synapse functioning through modulation of astrocytic glutamate release should be taken into consideration when considering interactions between oxytocinergic and dopaminergic systems in striatum.
Collapse
|
16
|
Carter JS, Wood SK, Kearns AM, Hopkins JL, Reichel CM. Paraventricular Nucleus of the Hypothalamus Oxytocin and Incubation of Heroin Seeking. Neuroendocrinology 2023; 113:1112-1126. [PMID: 36709749 PMCID: PMC10372195 DOI: 10.1159/000529358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 01/19/2023] [Indexed: 01/28/2023]
Abstract
INTRODUCTION There are numerous pharmacologic treatments for opioid use disorder (OUD), but none that directly target the underlying addictive effects of opioids. Oxytocin, a peptide hormone produced in the paraventricular nucleus (PVN) of the hypothalamus, has been investigated as a potential therapeutic for OUD. Promising preclinical and clinical results have been reported, but the brain region(s) and mechanism(s) by which oxytocin impacts reward processes remain undetermined. METHODS Here, we assess peripherally administered oxytocin's impacts on cued reinstatement of heroin seeking following forced abstinence and its effects on neuronal activation in the PVN and key projection regions. We also examine how designer receptors exclusively activated by designer drug (DREADD)-mediated activation or inhibition of oxytocinergic PVN neurons alters cued heroin seeking and social interaction. RESULTS As predicted, peripheral oxytocin administration successfully decreased cued heroin seeking on days 1 and 30 of abstinence. Oxytocin administration also led to increased neuronal activity within the PVN and the central amygdala (CeA). Activation of oxytocinergic PVN neurons with an excitatory (Gq) DREADD did not impact cued reinstatement or social interaction. In contrast, suppression with an inhibitory (Gi) DREADD reduced heroin seeking on abstinence day 30 and decreased time spent interacting with a novel conspecific. DISCUSSION These findings reinforce oxytocin's therapeutic potential for OUD, the basis for which may be driven in part by increased PVN-CeA circuit activity. Our results also suggest that oxytocin has distinct signaling and/or other mechanisms of action to produce these effects, as inhibition, but not activation, of oxytocinergic PVN neurons did not recapitulate the suppression in heroin seeking.
Collapse
Affiliation(s)
- Jordan S Carter
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina, USA,
| | - Samuel K Wood
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Angela M Kearns
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Jordan L Hopkins
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Carmela M Reichel
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina, USA
| |
Collapse
|
17
|
Gopinath A, Mackie PM, Phan LT, Mirabel R, Smith AR, Miller E, Franks S, Syed O, Riaz T, Law BK, Urs N, Khoshbouei H. Who Knew? Dopamine Transporter Activity Is Critical in Innate and Adaptive Immune Responses. Cells 2023; 12:cells12020269. [PMID: 36672204 PMCID: PMC9857305 DOI: 10.3390/cells12020269] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/22/2022] [Accepted: 01/04/2023] [Indexed: 01/12/2023] Open
Abstract
The dopamine transporter (DAT) regulates the dimension and duration of dopamine transmission. DAT expression, its trafficking, protein-protein interactions, and its activity are conventionally studied in the CNS and within the context of neurological diseases such as Parkinson's Diseases and neuropsychiatric diseases such as drug addiction, attention deficit hyperactivity and autism. However, DAT is also expressed at the plasma membrane of peripheral immune cells such as monocytes, macrophages, T-cells, and B-cells. DAT activity via an autocrine/paracrine signaling loop regulates macrophage responses to immune stimulation. In a recent study, we identified an immunosuppressive function for DAT, where blockade of DAT activity enhanced LPS-mediated production of IL-6, TNF-α, and mitochondrial superoxide levels, demonstrating that DAT activity regulates macrophage immune responses. In the current study, we tested the hypothesis that in the DAT knockout mice, innate and adaptive immunity are perturbed. We found that genetic deletion of DAT (DAT-/-) results in an exaggerated baseline inflammatory phenotype in peripheral circulating myeloid cells. In peritoneal macrophages obtained from DAT-/- mice, we identified increased MHC-II expression and exaggerated phagocytic response to LPS-induced immune stimulation, suppressed T-cell populations at baseline and following systemic endotoxemia and exaggerated memory B cell expansion. In DAT-/- mice, norepinephrine and dopamine levels are increased in spleen and thymus, but not in circulating serum. These findings in conjunction with spleen hypoplasia, increased splenic myeloid cells, and elevated MHC-II expression, in DAT-/- mice further support a critical role for DAT activity in peripheral immunity. While the current study is only focused on identifying the role of DAT in peripheral immunity, our data point to a much broader implication of DAT activity than previously thought. This study is dedicated to the memory of Dr. Marc Caron who has left an indelible mark in the dopamine transporter field.
Collapse
Affiliation(s)
- Adithya Gopinath
- Department of Neuroscience, University of Florida, Gainesville, FL 32611, USA
- Correspondence: (A.G.); (H.K.)
| | - Phillip M. Mackie
- Department of Neuroscience, University of Florida, Gainesville, FL 32611, USA
| | - Leah T. Phan
- Department of Neuroscience, University of Florida, Gainesville, FL 32611, USA
| | - Rosa Mirabel
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL 32611, USA
| | - Aidan R. Smith
- Department of Neuroscience, University of Florida, Gainesville, FL 32611, USA
| | - Emily Miller
- Department of Neuroscience, University of Florida, Gainesville, FL 32611, USA
| | - Stephen Franks
- Department of Neuroscience, University of Florida, Gainesville, FL 32611, USA
| | - Ohee Syed
- Department of Neuroscience, University of Florida, Gainesville, FL 32611, USA
| | - Tabish Riaz
- Department of Neuroscience, University of Florida, Gainesville, FL 32611, USA
| | - Brian K. Law
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL 32611, USA
| | - Nikhil Urs
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL 32611, USA
| | - Habibeh Khoshbouei
- Department of Neuroscience, University of Florida, Gainesville, FL 32611, USA
- Correspondence: (A.G.); (H.K.)
| |
Collapse
|
18
|
Bozkurt M. Neuroscientific Basis of Treatment for Substance Use Disorders. Noro Psikiyatr Ars 2022; 59:S75-S80. [PMID: 36578985 PMCID: PMC9767124 DOI: 10.29399/npa.28172] [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: 03/19/2022] [Accepted: 09/07/2022] [Indexed: 12/31/2022] Open
Abstract
Substance use disorder is a chronic and relapsing disease that burdens both the individual and the society. In addition to psychosocial treatment approaches, currently there are approved pharmacological treatment options for opioid, alcohol and tobacco use disorders, but only symptomatic treatment can be offered to patients with other substance use disorders. Advances in neuroscience and a better understanding of the addiction process offer an opportunity to create new treatment options. There is a wide range of studies, ranging from the use of drugs with different indications to the development of new pharmacological treatments, and from vaccine studies to neuromodulation techniques. Establishing novel treatment goals in addition to complete abstinence and individualizing treatment by focusing on endophenotypes may increase the treatment alternatives and the efficacy of these treatments for SUD.
Collapse
Affiliation(s)
- Müge Bozkurt
- İstanbul University, İstanbul Faculty of Medicine, Department of Psychiatry, İstanbul, Turkey
| |
Collapse
|
19
|
Giannotti G, Mottarlini F, Heinsbroek JA, Mandel MR, James MH, Peters J. Oxytocin and orexin systems bidirectionally regulate the ability of opioid cues to bias reward seeking. Transl Psychiatry 2022; 12:432. [PMID: 36195606 PMCID: PMC9532415 DOI: 10.1038/s41398-022-02161-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 09/06/2022] [Accepted: 09/08/2022] [Indexed: 11/17/2022] Open
Abstract
As opioid-related fatalities continue to rise, the need for novel opioid use disorder (OUD) treatments could not be more urgent. Two separate hypothalamic neuropeptide systems have shown promise in preclinical OUD models. The oxytocin system, originating in the paraventricular nucleus (PVN), may protect against OUD severity. By contrast, the orexin system, originating in the lateral hypothalamus (LH), may exacerbate OUD severity. Thus, activating the oxytocin system or inhibiting the orexin system are potential therapeutic strategies. The specific role of these systems with regard to specific OUD outcomes, however, is not fully understood. Here, we probed the therapeutic efficacy of pharmacological interventions targeting the orexin or oxytocin system on two distinct metrics of OUD severity in rats-heroin choice (versus choice for natural reward, i.e., food) and cued reward seeking. Using a preclinical model that generates approximately equal choice between heroin and food reward, we examined the impact of exogenously administered oxytocin, an oxytocin receptor antagonist (L-368,899), and a dual orexin receptor antagonist (DORA-12) on opioid choice. Whereas these agents did not alter heroin choice when rewards (heroin and food) were available, oxytocin and DORA-12 each significantly reduced heroin seeking in the presence of competing reward cues when no rewards were available. In addition, the number of LH orexin neurons and PVN oxytocin neurons correlated with specific behavioral economic variables indicative of heroin versus food motivation. These data identify a novel bidirectional role of the oxytocin and orexin systems in the ability of opioid-related cues to bias reward seeking.
Collapse
Affiliation(s)
- Giuseppe Giannotti
- Department of Anesthesiology, University of Colorado, Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Francesca Mottarlini
- Department of Anesthesiology, University of Colorado, Anschutz Medical Campus, Aurora, CO, 80045, USA
- Department of Pharmacological and Biomolecular Sciences, University of Milan, 20133, Milan, Italy
| | - Jasper A Heinsbroek
- Department of Anesthesiology, University of Colorado, Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Mitchel R Mandel
- Department of Anesthesiology, University of Colorado, Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Morgan H James
- Department of Psychiatry, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ, 08854, USA
| | - Jamie Peters
- Department of Anesthesiology, University of Colorado, Anschutz Medical Campus, Aurora, CO, 80045, USA.
- Department of Pharmacology, University of Colorado, Anschutz Medical Campus, Aurora, CO, 80045, USA.
| |
Collapse
|
20
|
The Role of Intraamygdaloid Oxytocin and D2 Dopamine Receptors in Reinforcement in the Valproate-Induced Autism Rat Model. Biomedicines 2022; 10:biomedicines10092309. [PMID: 36140411 PMCID: PMC9496370 DOI: 10.3390/biomedicines10092309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/07/2022] [Accepted: 09/12/2022] [Indexed: 11/28/2022] Open
Abstract
Background: autism spectrum disorder (ASD) is a neurodevelopmental disorder affecting around 1 out of 68 children and its incidence shows an increasing tendency. There is currently no effective treatment for ASD. In autism research, the valproate (VPA)-induced autism rodent model is widely accepted. Our previous results showed that intraamygdaloid oxytocin (OT) has anxiolytic effects on rats showing autistic signs under the VPA-induced autism model. Methods: rats were stereotaxically implanted with guide cannulae bilaterally and received intraamygdaloid microinjections. In the present study, we investigated the possible role of intraamygdaloid OT and D2 dopamine (DA) receptors on reinforcement using VPA-treated rats in a conditioned place preference test. OT and/or an OT receptor antagonist or a D2 DA antagonist were microinjected into the central nucleus of the amygdala (CeA). Results: valproate-treated rats receiving 10 ng OT spent significantly longer time in the treatment quadrant during the test session of the conditioned place preference test. Prior treatment with an OT receptor antagonist or with a D2 DA receptor antagonist blocked the positive reinforcing effects of OT. The OT receptor antagonist or D2 DA antagonist in themselves did not influence the time rats spent in the treatment quadrant. Conclusions: Our results show that OT has positive reinforcing effects under the VPA-induced autism rodent model and these effects are OT receptor-specific. Our data also suggest that the DAergic system plays a role in the positive reinforcing effects of OT because the D2 DA receptor antagonist can block these actions.
Collapse
|
21
|
Mühle C, Mazza M, Weinland C, von Zimmermann C, Bach P, Kiefer F, Grinevich V, Zoicas I, Kornhuber J, Lenz B. Elevated Oxytocin Receptor Blood Concentrations Predict Higher Risk for, More, and Earlier 24-Month Hospital Readmissions after In-Patient Detoxification in Males with Alcohol Use Disorder. Int J Mol Sci 2022; 23:ijms23179940. [PMID: 36077337 PMCID: PMC9455990 DOI: 10.3390/ijms23179940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 08/23/2022] [Accepted: 08/25/2022] [Indexed: 11/16/2022] Open
Abstract
Alcohol use disorder (AUD) is a major global mental health challenge. Knowledge concerning mechanisms underlying AUD and predictive biomarkers of AUD progression and relapse are insufficient. Recently, addiction research is focusing attention on the oxytocin system. However, to our knowledge, blood concentrations of the oxytocin receptor (OXTR) have not yet been studied in AUD. Here, in sex-separated analyses, OXTR serum concentrations were compared between early-abstinent in-patients with AUD (113 men, 87 women) and age-matched healthy controls (133 men, 107 women). The OXTR concentrations were correlated with sex hormone and oxytocin concentrations and alcohol-related hospital readmissions during a 24-month follow-up. In male patients with AUD, higher OXTR concentrations were found in those with an alcohol-related readmission than in those without (143%; p = 0.004), and they correlated with more prospective readmissions (ρ = 0.249; p = 0.008) and fewer days to the first readmission (ρ = −0.268; p = 0.004). In men and women, OXTR concentrations did not significantly differ between patients with AUD and controls. We found lower OXTR concentrations in smokers versus non-smokers in female patients (61%; p = 0.001) and controls (51%; p = 0.003). In controls, OXTR concentrations correlated with dihydrotestosterone (men, ρ = 0.189; p = 0.030) and testosterone concentrations (women, ρ = 0.281; p = 0.003). This clinical study provides novel insight into the role of serum OXTR levels in AUD. Future studies are encouraged to add to the available knowledge and investigate clinical implications of OXTR blood concentrations.
Collapse
Affiliation(s)
- Christiane Mühle
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Schwabachanlage 6, D-91054 Erlangen, Germany
- Correspondence: or ; Tel.: +49-9131-85-44738; Fax: +49-9131-85-36381
| | - Massimiliano Mazza
- Department of Addictive Behavior and Addiction Medicine, Central Institute of Mental Health (CIMH), Medical Faculty Mannheim, Heidelberg University, J 5, D-68159 Mannheim, Germany
| | - Christian Weinland
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Schwabachanlage 6, D-91054 Erlangen, Germany
| | - Claudia von Zimmermann
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Schwabachanlage 6, D-91054 Erlangen, Germany
| | - Patrick Bach
- Department of Addictive Behavior and Addiction Medicine, Central Institute of Mental Health (CIMH), Medical Faculty Mannheim, Heidelberg University, J 5, D-68159 Mannheim, Germany
| | - Falk Kiefer
- Department of Addictive Behavior and Addiction Medicine, Central Institute of Mental Health (CIMH), Medical Faculty Mannheim, Heidelberg University, J 5, D-68159 Mannheim, Germany
| | - Valery Grinevich
- Department of Neuropeptide Research in Psychiatry, Central Institute of Mental Health (CIMH), Medical Faculty Mannheim, Heidelberg University, J 5, D-68159 Mannheim, Germany
| | - Iulia Zoicas
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Schwabachanlage 6, D-91054 Erlangen, Germany
| | - Johannes Kornhuber
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Schwabachanlage 6, D-91054 Erlangen, Germany
| | - Bernd Lenz
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Schwabachanlage 6, D-91054 Erlangen, Germany
- Department of Addictive Behavior and Addiction Medicine, Central Institute of Mental Health (CIMH), Medical Faculty Mannheim, Heidelberg University, J 5, D-68159 Mannheim, Germany
| |
Collapse
|
22
|
Joseph JE, Bustos N, Crum K, Flanagan J, Baker NL, Hartwell K, Santa-Maria MM, Brady K, McRae-Clark A. Oxytocin moderates corticolimbic social stress reactivity in cocaine use disorder and healthy controls. COMPREHENSIVE PSYCHONEUROENDOCRINOLOGY 2022; 11:100150. [PMID: 35967924 PMCID: PMC9363641 DOI: 10.1016/j.cpnec.2022.100150] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 06/09/2022] [Accepted: 06/09/2022] [Indexed: 11/30/2022] Open
Abstract
Social stress can contribute to the development of substance use disorders (SUDs) and increase the likelihood of relapse. Oxytocin (OT) is a potential pharmacotherapy that may buffer the effects of social stress on arousal and reward neurocircuitry. However, more research is needed to understand how OT moderates the brain’s response to social stress in SUDs. The present study examined the effect of intransasal OT (24 IU) versus placebo (PBO) on corticolimbic functional connectivity associated with acute social stress in individuals with cocaine use disorder (CUD; n = 67) and healthy controls (HC; n = 52). Psychophysiological interaction modeling used the left and right amygdala as seed regions with the left and right orbitofrontal and anterior cingulate cortex as a priori regions of interest. Moderators of the OT response included childhood trauma history and biological sex, which were examined in independent analyses. The main finding was that OT normalized corticolimbic connectivity (left amygdala-orbitofrontal and left amygdala-anterior cingulate) as a function of childhood trauma such that connectivity was different between trauma-present and trauma-absent groups on PBO, but not between trauma groups on OT. Effects of OT on corticolimbic connectivity were not different as a function of diagnosis (CUD vs HC) or sex. However, OT reduced subjective anxiety during social stress for CUD participants who reported childhood trauma compared to PBO and normalized craving response as a function of sex in CUD. The present findings add to some prior findings of normalizing effects of OT on corticolimbic circuitry in individuals with trauma histories and provide some initial support that OT can normalize subjective anxiety and craving in CUD. Social stress-related corticolimbic connectivity was affected by childhood trauma under placebo. Under oxytocin, corticolimbic connectivity differences due to childhood trauma were absent. Oxytocin reduced subjective anxiety in cocaine users with childhood trauma. Oxytocin reduced subjective craving in male cocaine users.
Collapse
Affiliation(s)
- Jane E. Joseph
- Department of Neuroscience, Medical University of South Carolina, 135 Cannon Street, Charleston SC, 29425, USA
- Corresponding author.
| | - Nicholas Bustos
- Department of Neuroscience, Medical University of South Carolina, 135 Cannon Street, Charleston SC, 29425, USA
| | - Kathleen Crum
- Department of Neuroscience, Medical University of South Carolina, 135 Cannon Street, Charleston SC, 29425, USA
- Department of Psychiatry Indiana University School of Medicine, Indianapolis, IN, USA
| | - Julianne Flanagan
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, USA
- Ralph H. Johnson VA Medical Center, Charleston, SC, USA
| | - Nathaniel L. Baker
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Karen Hartwell
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, USA
- Ralph H. Johnson VA Medical Center, Charleston, SC, USA
| | - Megan Moran Santa-Maria
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, USA
- Boehringer Ingelheim, Athens, GA, USA
| | - Kathleen Brady
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, USA
- Ralph H. Johnson VA Medical Center, Charleston, SC, USA
| | - Aimee McRae-Clark
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, USA
- Ralph H. Johnson VA Medical Center, Charleston, SC, USA
| |
Collapse
|
23
|
Barbee BR, Gourley SL. Brain systems in cocaine abstinence-induced anxiety-like behavior in rodents: A review. ADDICTION NEUROSCIENCE 2022; 2:100012. [PMID: 37485439 PMCID: PMC10361393 DOI: 10.1016/j.addicn.2022.100012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/25/2023]
Abstract
Cocaine use disorder (CUD) is a significant public health issue that generates substantial personal, familial, and economic burdens. Still, there are no FDA-approved pharmacotherapies for CUD. Cocaine-dependent individuals report anxiety during withdrawal, and alleviation of anxiety and other negative affective states may be critical for maintaining drug abstinence. However, the neurobiological mechanisms underlying abstinence-related anxiety in humans or anxiety-like behavior in rodents are not fully understood. This review summarizes investigations regarding anxiety-like behavior in mice and rats undergoing cocaine abstinence, as assessed using four of the most common anxiety-related assays: the elevated plus (or its derivative, the elevated zero) maze, open field test, light-dark transition test, and defensive burying task. We first summarize available evidence that cocaine abstinence generates anxiety-like behavior that persists throughout protracted abstinence. Then, we examine investigations concerning neuropeptide, neurotransmitter, and neuromodulator systems in cocaine abstinence-induced anxiety-like behavior. Throughout, we discuss how differences in sex, rodent strain, cocaine dose and dosing strategy and abstinence duration interact to generate anxiety-like behavior.
Collapse
Affiliation(s)
- Britton R. Barbee
- Graduate Program in Molecular and Systems Pharmacology,
Emory University
- Department of Pediatrics, Emory University School of
Medicine; Yerkes National Primate Research Center
| | - Shannon L. Gourley
- Graduate Program in Molecular and Systems Pharmacology,
Emory University
- Department of Pediatrics, Emory University School of
Medicine; Yerkes National Primate Research Center
| |
Collapse
|
24
|
Fernández-Rodríguez S, Cano-Cebrián MJ, Rius-Pérez S, Pérez S, Guerri C, Granero L, Zornoza T, Polache A. Different brain oxidative and neuroinflammation status in rats during prolonged abstinence depending on their ethanol relapse-like drinking behavior: Effects of ethanol reintroduction. Drug Alcohol Depend 2022; 232:109284. [PMID: 35033958 DOI: 10.1016/j.drugalcdep.2022.109284] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 12/17/2021] [Accepted: 01/03/2022] [Indexed: 12/17/2022]
Abstract
RATIONALE Accumulating evidence suggests that chronic alcohol consumption is associated with excessive oxidative damage and neuroinflammatory processes and these events have been associated to early alcohol withdrawal. In the present research we wonder if brain oxidative stress and neuroinflammation remains altered during prolonged withdrawal situations and whether these alterations can be correlated with relapse behavior in alcohol consumption. The effects of alcohol reintroduction were also evaluated METHODS: We have used a model based on the alcohol deprivation effect (ADE) within a cohort of wild-type male Wistar rats. Two subpopulations were identified according to the alcohol relapse-like drinking behavior displayed (ADE and NO-ADE subpopulations). Oxidized and reduced glutathione content was determined within the hippocampus and the amygdala using a mass spectrometry method. The levels of mRNA of seven different inflammatory mediators in the prefrontal cortex of rats were quantified. All the analyses were performed in two different conditions: after 21-day alcohol deprivation (prolonged abstinence) and after 24 h of ethanol reintroduction in both subpopulations. RESULTS ADE and NO-ADE rats showed different endophenotypes. ADE rats always displayed a significant lower alcohol intake rate and ethanol preference than NO-ADE rats. The results also demonstrated the existence of altered brain redox and neuroinflammation status after prolonged abstinence exclusively in ADE rats. Moreover, when ethanol was reintroduced in the ADE subpopulation, altered oxidative stress and neuroinflammatory markers were restored. CONCLUSIONS Present findings provide new mechanisms underlying the neurobiology of relapse behavior and suggest the development of new pharmacological approaches to treat alcohol-induced relapse.
Collapse
Affiliation(s)
- S Fernández-Rodríguez
- Departament de Farmàcia i Tecnologia Farmacèutica i Parasitologia, Universitat de València, Avda Vicente Andrés Estellés, s/n 46100 Burjassot, Spain
| | - M J Cano-Cebrián
- Departament de Farmàcia i Tecnologia Farmacèutica i Parasitologia, Universitat de València, Avda Vicente Andrés Estellés, s/n 46100 Burjassot, Spain
| | - S Rius-Pérez
- Departament de Fisiologia, Universitat de València, Avda Vicente Andrés Estellés, s/n 46100 Burjassot, Spain
| | - S Pérez
- Departament de Fisiologia, Universitat de València, Avda Vicente Andrés Estellés, s/n 46100 Burjassot, Spain
| | - C Guerri
- Department of Molecular and Cellular Pathology of Alcohol, Príncipe Felipe Research Center, Carrer d'Eduardo Primo Yúfera, 3, 46012 Valencia, Spain
| | - L Granero
- Departament de Farmàcia i Tecnologia Farmacèutica i Parasitologia, Universitat de València, Avda Vicente Andrés Estellés, s/n 46100 Burjassot, Spain
| | - T Zornoza
- Departament de Farmàcia i Tecnologia Farmacèutica i Parasitologia, Universitat de València, Avda Vicente Andrés Estellés, s/n 46100 Burjassot, Spain.
| | - A Polache
- Departament de Farmàcia i Tecnologia Farmacèutica i Parasitologia, Universitat de València, Avda Vicente Andrés Estellés, s/n 46100 Burjassot, Spain
| |
Collapse
|
25
|
Ryabinin AE, Zhang Y. Barriers and Breakthroughs in Targeting the Oxytocin System to Treat Alcohol Use Disorder. Front Psychiatry 2022; 13:842609. [PMID: 35295777 PMCID: PMC8919088 DOI: 10.3389/fpsyt.2022.842609] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 02/04/2022] [Indexed: 11/13/2022] Open
Abstract
Development of better treatments for alcohol use disorder (AUD) is urgently needed. One promising opportunity for this development is the potential of targeting the oxytocin peptide system. Preclinical studies showed that administration of exogenous oxytocin or, more recently, stimulation of neurons expressing endogenous oxytocin lead to a decreased alcohol consumption across several rodent models. Initial clinical studies also showed that administration of oxytocin decreased craving for alcohol and heavy alcohol drinking. However, several more recent clinical studies were not able to replicate these effects. Thus, although targeting the oxytocin system holds promise for the treatment of AUD, more nuanced approaches toward development and application of these treatments are needed. In this mini-review we discuss potential caveats resulting in differential success of attempts to use oxytocin for modulating alcohol use disorder-related behaviors in clinical studies and evaluate three directions in which targeting the oxytocin system could be improved: (1) increasing potency of exogenously administered oxytocin, (2) developing oxytocin receptor agonists, and (3) stimulating components of the endogenous oxytocin system. Both advances and potential pitfalls of these directions are discussed.
Collapse
Affiliation(s)
- Andrey E. Ryabinin
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States
| | | |
Collapse
|
26
|
Oxytocin, a Novel Treatment for Methamphetamine Use Disorder. Neurol Int 2022; 14:186-198. [PMID: 35225885 PMCID: PMC8883935 DOI: 10.3390/neurolint14010015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 01/17/2022] [Accepted: 01/19/2022] [Indexed: 01/27/2023] Open
Abstract
The treatment of substance abuse with oxytocin is a novel approach to a challenging public health issue that continues to contribute to a growing economic cost for societies worldwide. Methamphetamine addiction is one of the leading causes of mortality worldwide, and despite advances in understanding the neurobiology of methamphetamine addiction, treatment options are limited. There are no medications that the Food and Drug Administration currently approves for stimulant use disorder. Off-label use of therapies for stimulant misuse include antidepressants, anxiolytics, and milder stimulants as replacement agents. Due to the shortcomings of these attempts to treat a complicated psychiatric disorder, recent attention to oxytocin therapy (OT) has gained momentum in clinical studies as a possible therapy in the context of social stress, social anxiety, social cognition, and psychosis. Oxytocin produces enhanced connectivity between cortical regions. The results from studies in rodents with OT suggest that central neuromodulation of oxytocin may be beneficial across transition states of stimulant dependence and may alleviate intense withdrawal symptoms. Studies of oxytocin in the context of other drugs of abuse, including cocaine, cannabis, and alcohol, also support the potential of oxytocin to treat stimulant use disorder, methamphetamine type. Methamphetamine abuse continues to be a significant cause of distress and dysfunction throughout the world. The effects of oxytocin on methamphetamine use outlined in this review should act as a catalyst for further investigation into the efficacy of treating stimulant use disorder, methamphetamine type with oxytocin in humans. More human-based research should initiate studies involving the long-term efficacy, side effects, and patient selection.
Collapse
|
27
|
Stauffer CS, Samson S, Hickok A, Hoffman WF, Batki SL. Intranasal Oxytocin for Stimulant Use Disorder Among Male Veterans Enrolled in an Opioid Treatment Program: A Randomized Controlled Trial. Front Psychiatry 2022; 12:804997. [PMID: 35111090 PMCID: PMC8801418 DOI: 10.3389/fpsyt.2021.804997] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 12/15/2021] [Indexed: 12/12/2022] Open
Abstract
The increasing prevalence of illicit stimulant use among those in opioid treatment programs poses a significant risk to public health, stimulant users have the lowest rate of retention and poorest outcomes among those in addiction treatment, and current treatment options are limited. Oxytocin administration has shown promise in reducing addiction-related behavior and enhancing salience to social cues. We conducted a randomized, double-blind, placebo-controlled clinical trial of intranasal oxytocin administered twice daily for 6 weeks to male Veterans with stimulant use disorder who were also receiving opioid agonist therapy and counseling (n = 42). There was no significant effect of oxytocin on stimulant use, stimulant craving, or therapeutic alliance over 6 weeks. However, participants receiving oxytocin (vs. placebo) attended significantly more daily opioid agonist therapy dispensing visits. This replicated previous work suggesting that oxytocin may enhance treatment engagement among individuals with stimulant and opioid use disorders, which would address a significant barrier to effective care.
Collapse
Affiliation(s)
- Christopher S. Stauffer
- Department of Mental Health, VA Portland Health Care System, Portland, OR, United States
- Social Neuroscience and Psychotherapy Lab, Department of Psychiatry, Oregon Health and Science University, Portland, OR, United States
- Department of Psychiatry and Behavioral Sciences, San Francisco School of Medicine and San Francisco VA Health Care System, University of California, San Francisco, San Francisco, CA, United States
| | - Salem Samson
- School of Nursing, Massachusetts General Hospital (MGH) Institute of Health Professions, Boston, MA, United States
| | - Alex Hickok
- Department of Mental Health, VA Portland Health Care System, Portland, OR, United States
- Social Neuroscience and Psychotherapy Lab, Department of Psychiatry, Oregon Health and Science University, Portland, OR, United States
| | - William F. Hoffman
- Department of Mental Health, VA Portland Health Care System, Portland, OR, United States
- Social Neuroscience and Psychotherapy Lab, Department of Psychiatry, Oregon Health and Science University, Portland, OR, United States
| | - Steven L. Batki
- Department of Psychiatry and Behavioral Sciences, San Francisco School of Medicine and San Francisco VA Health Care System, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
28
|
Oxytocin Attenuates the Stress-Induced Reinstatement of Alcohol-Seeking in Male Rats: Role of the Central Amygdala. Biomedicines 2021; 9:biomedicines9121919. [PMID: 34944734 PMCID: PMC8698625 DOI: 10.3390/biomedicines9121919] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 12/11/2021] [Accepted: 12/13/2021] [Indexed: 12/21/2022] Open
Abstract
Factors such as stress and anxiety often contribute to alcohol-dependent behavior and can trigger a relapse of alcohol addiction and use. Therefore, it is important to investigate potential pharmacological interventions that may alleviate the influence of stress on addiction-related behaviors. Previous studies have demonstrated that the neuropeptide oxytocin has promising anxiolytic potential in mammals and may offer a pharmacological target to diminish the emotional impact on reinstatement of alcohol-seeking. The purpose of the present study was to investigate the effect of oxytocin on stress-induced alcohol relapse and identify a neural structure mediating this effect through the use of an ethanol self-administration and yohimbine-induced reinstatement paradigm. While yohimbine administration resulted in the reinstatement of ethanol-seeking behavior, the concurrent administration of yohimbine and oxytocin attenuated this effect, suggesting that oxytocin may disrupt stress-induced ethanol-seeking behavior. The central amygdala (CeA) is a structure that drives emotional responses and robustly expresses oxytocin receptors. Intra-CeA oxytocin similarly attenuated the yohimbine-induced reinstatement of ethanol-seeking behavior. These results demonstrate that oxytocin has the potential to attenuate stress-induced relapse into ethanol-seeking behavior, and that this mechanism occurs specifically within the central amygdala.
Collapse
|
29
|
Le Roux M, Möller M, Harvey BH. Prolonged efavirenz exposure reduces peripheral oxytocin and vasopressin comparable to known drugs of addiction in male Sprague Dawley rats. IBRO Neurosci Rep 2021; 11:56-63. [PMID: 34939063 PMCID: PMC8664698 DOI: 10.1016/j.ibneur.2021.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 03/03/2021] [Accepted: 06/22/2021] [Indexed: 11/27/2022] Open
Abstract
INTRODUCTION Several drugs of abuse (DOA) are capable of modulating neurohypophysial hormones, such as oxytocin (OT) and vasopressin (VP), potentially resulting in the development of psychological abnormalities, such as cognitive dysfunction, psychoses, and affective disorders. Efavirenz (EFV), widely used in Africa and globally to treat HIV, induces diverse neuropsychiatric side effects while its abuse has become a global concern. The actions of EFV may involve neurohypophysial system (NS) disruption like that of known DOA. This study investigated whether sub-chronic EFV exposure, at a previously-determined rewarding dose, alters peripheral OT and VP levels versus that of a control, ∆9-tetrahydrocannabinol (∆9-THC), methamphetamine (MA) and cocaine. MATERIALS AND METHODS To simulate the conditions under which reward-driven behavior had previously been established for EFV, male Sprague Dawley rats (n = 16/exposure) received intraperitoneal vehicle (control) or drug administration across an alternating sixteen-day dosing protocol. Control administration (saline/olive oil; 0.2 ml) occurred on odd-numbered and drug administration (EFV: 5 mg/kg, ∆9-THC: 0.75 mg/kg, MA: 1 mg/kg, or cocaine: 20 mg/kg) on even-numbered days followed by euthanasia, trunk blood collection and plasma extraction for neuropeptide assay. Effect of drug exposure on peripheral OT and VP levels was assessed versus controls and quantified using specific ELISA kits. Statistical significance was determined by Kruskal-Wallis ANOVA, with p < 0.05. Ethics approval: NWU-00291-17-A5. RESULTS Delta-9-THC reduced OT and VP plasma levels (p < 0.0001, p = 0.0141; respectively), cocaine reduced plasma OT (p = 0.0023), while MA reduced plasma VP levels (p = 0.0001), all versus control. EFV reduced OT and VP plasma levels (p < 0.0001; OT and VP) versus control, and similar to ∆9-THC. CONCLUSION EFV markedly affects the NS in significantly reducing both plasma OT and VP equivalent to DOA. Importantly, EFV has distinct effects on peripheral OT and VP levels when assessed within the context of drug dependence. The data highlights a possible new mechanism underlying previously documented EFV-induced effects in rats, and whereby EFV may induce neuropsychiatric adverse effects clinically; also providing a deeper understanding of the suggested abuse-potential of EFV.
Collapse
Key Words
- 5-HT, 5-hydroxytryptamine (serotonin)
- ADH, antidiuretic hormone
- AEA, N-arachidonoylethanolamine (anandamide)
- ANOVA, one-way analysis of variance
- ARRIVE, animal research: reporting of in vivo experiments (guidelines)
- ARV, antiretroviral
- Ach, acetylcholine
- CB, cannabinoid
- CNS, central nervous system
- CPP, conditioned place preference
- Cocaine
- DA, dopamine
- DAT, dopamine transporter
- DOA‘s, drug(s) of abuse
- ECS, endocannabinoid system
- EFV, efavirenz
- ELISA, enzyme-linked immunosorbent assay
- Efavirenz
- GABA, gamma-aminobutyric acid
- Glu, glutamate
- HIV, human immunodeficiency virus
- HNS, hypothalamic neurohypophysial system
- HPA, hypothalamic-pituitary-adrenal (axis)
- IP, intraperitoneal
- IV, intravenous
- M, muscarinic
- MA, methamphetamine
- MAO, monoamine oxidase
- Methamphetamine
- NAc, nucleus accumbens
- NE, norepinephrine
- NO, nitric oxide
- NPAE, neuropsychiatric adverse effect
- OT, oxytocin
- OTR, oxytocin receptor
- Oxytocin
- PND, postnatal day
- PVN, paraventricular nucleus
- SC, subcutaneous
- SD, Sprague Dawley (rat)
- SEM, standard error of the mean
- SERT, serotonin transporter
- SON, supraoptic nucleus
- VMAT, vesicular monoamine transporter
- VP, vasopressin
- VPR, vasopressin receptor
- Vasopressin
- cART, combined antiretroviral therapy
- ∆9-THC, delta-9-tetrahydrocannabinol
- ∆9-tetrahydrocannabinol
Collapse
Affiliation(s)
- Mandi Le Roux
- Division of Pharmacology, School of Pharmacy, North-West University, Potchefstroom, South Africa
- Centre of Excellence for Pharmaceutical Sciences (PharmaCenTM), School of Pharmacy, North-West University, Potchefstroom, South Africa
| | - Marisa Möller
- Division of Pharmacology, School of Pharmacy, North-West University, Potchefstroom, South Africa
- Centre of Excellence for Pharmaceutical Sciences (PharmaCenTM), School of Pharmacy, North-West University, Potchefstroom, South Africa
| | - Brian H. Harvey
- Division of Pharmacology, School of Pharmacy, North-West University, Potchefstroom, South Africa
- Centre of Excellence for Pharmaceutical Sciences (PharmaCenTM), School of Pharmacy, North-West University, Potchefstroom, South Africa
| |
Collapse
|
30
|
Early life adversity promotes resilience to opioid addiction-related phenotypes in male rats and sex-specific transcriptional changes. Proc Natl Acad Sci U S A 2021; 118:2020173118. [PMID: 33593913 DOI: 10.1073/pnas.2020173118] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Experiencing some early life adversity can have an "inoculating" effect that promotes resilience in adulthood. However, the mechanisms underlying stress inoculation are unknown, and animal models are lacking. Here we used the limited bedding and nesting (LBN) model of adversity to evaluate stress inoculation of addiction-related phenotypes. In LBN, pups from postnatal days 2 to 9 and their dams were exposed to a low-resource environment. In adulthood, they were tested for addiction-like phenotypes and compared to rats raised in standard housing conditions. High levels of impulsivity are associated with substance abuse, but in males, LBN reduced impulsive choice compared to controls. LBN males also self-administered less morphine and had a lower breakpoint on a progressive ratio reinforcement schedule than controls. These effects of LBN on addiction-related behaviors were not found in females. Because the nucleus accumbens (NAc) mediates these behaviors, we tested whether LBN altered NAc physiology in drug-naïve and morphine-exposed rats. LBN reduced the frequency of spontaneous excitatory postsynaptic currents in males, but a similar effect was not observed in females. Only in males did LBN prevent a morphine-induced increase in the AMPA/NMDA ratio. RNA sequencing was performed to delineate the molecular signature in the NAc associated with LBN-derived phenotypes. LBN produced sex-specific changes in transcription, including in genes related to glutamate transmission. Collectively, these studies reveal that LBN causes a male-specific stress inoculation effect against addiction-related phenotypes. Identifying factors that promote resilience to addiction may reveal novel treatment options for patients.
Collapse
|
31
|
Houghton B, Kouimtsidis C, Duka T, Paloyelis Y, Bailey A. Can intranasal oxytocin reduce craving in automated addictive behaviours? A systematic review. Br J Pharmacol 2021; 178:4316-4334. [PMID: 34235724 DOI: 10.1111/bph.15617] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 06/21/2021] [Accepted: 06/28/2021] [Indexed: 12/24/2022] Open
Abstract
Existing pharmacotherapies for managing craving, a strong predictor of relapse to automated addictive behaviours, are limited in efficacy and characterised by increased health risks associated with their pharmacological profile. Preclinical studies have identified oxytocin as a promising pharmacotherapy with anticraving properties for addictive behaviours. Here, we provide the first systematic review of 17 human studies (n = 722; 30% female) investigating the efficacy of intranasal oxytocin to reduce craving or consumption in addictive behaviours. We identify intranasal oxytocin as a method that warrants further investigation regarding its capacity to decrease cue-induced, acute stress-induced or withdrawal-related craving and relapse related to alcohol, cannabis, opioids, cocaine or nicotine, including a potential role as ad hoc medication following exposure to drug-related cues. Future studies should investigate the role of factors such as treatment regimens and sample characteristics, including the role of the amygdala, which we propose as a distinct mechanism mediating oxytocin's anticraving properties.
Collapse
Affiliation(s)
- Ben Houghton
- Pharmacology Section, Institute of Medical and Biomedical Education, St George's University of London, London, UK
| | | | - Theodora Duka
- Behavioural and Clinical Neuroscience, School of Psychology, University of Sussex, Brighton, UK
| | - Yannis Paloyelis
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Alexis Bailey
- Pharmacology Section, Institute of Medical and Biomedical Education, St George's University of London, London, UK
| |
Collapse
|
32
|
Zhang L, Liu S, Liu X, Zhang B, An X, Ming D. Emotional Arousal and Valence Jointly Modulate the Auditory Response: A 40-Hz ASSR Study. IEEE Trans Neural Syst Rehabil Eng 2021; 29:1150-1157. [PMID: 34110997 DOI: 10.1109/tnsre.2021.3088257] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Emotion is defined as a response to external stimuli and internal mental representations. It has been characterized as a multidimensional concept, primarily comprising two dimensions: valence and arousal. Existing studies have demonstrated that emotional experience exerts a powerful impact on auditory processing in terms of valence. However, it has also been shown that while negative emotion can improve auditory perception in healthy subjects, patients with depression show deficits in auditory perception. We thus speculated that both arousal and valence jointly modulate auditory perception. To examine the emotion-driven effects on the auditory response, we induced positive, negative, and neutral emotional states in healthy subjects and collected auditory steady-state response (ASSR) evoked by a 40-Hz chirp sound. We calculated peak-to-peak amplitude (PPA) and event-related spectral perturbation (ERSP) of evoked ASSRs and observed that the positive emotions significantly enhanced brain responses to auditory stimuli (p < 0.001), but that ASSRs in a negative state were not significantly enhanced compared with the neutral state. Subsequently, regression analysis showed a significant positive multiple linear relationship between the PPA and ratings of two emotional dimensions, indicating that arousal and valence jointly regulated the auditory cortex's synchronous oscillation, rather than the valence in isolation, offering the potential to clarify the conflicting results surrounding the role of negative emotions in auditory responses. Because depression is generally characterized by low arousal and low valence in actual life, whereas the negative emotion evoked under laboratory conditions is always with low valence but high arousal.
Collapse
|
33
|
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
|
34
|
Sanna F, De Luca MA. The potential role of oxytocin in addiction: What is the target process? Curr Opin Pharmacol 2021; 58:8-20. [PMID: 33845377 DOI: 10.1016/j.coph.2021.03.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 02/26/2021] [Accepted: 03/05/2021] [Indexed: 01/27/2023]
Abstract
Oxytocin regulates a variety of centrally-mediated functions, ranging from socio-sexual behavior, maternal care, and affiliation to fear, stress, anxiety. In the past years, both clinical and preclinical studies characterized oxytocin for its modulatory role on reward-related neural substrates mainly involving the interplay with the mesolimbic and mesocortical dopaminergic pathways. This suggests a role of this nonapeptide on the neurobiology of addiction raising the possibility of its therapeutic use. Although far from a precise knowledge of the underlying mechanisms, the putative role of the bed nucleus of the stria terminalis as a key structure where oxytocin may rebalance altered neurochemical processes and neuroplasticity involved in dependence and relapse has been highlighted. This view opens new opportunities to address the health problems related to drug misuse.
Collapse
Affiliation(s)
- Fabrizio Sanna
- Department of Biomedical Sciences, University of Cagliari, Monserrato, Cagliari 09042, Italy
| | | |
Collapse
|
35
|
Abstract
Drug addiction is a chronic relapsing disorder, and a significant amount of research has been devoted to understand the factors that contribute to the development, loss of control, and persistence of compulsive addictive behaviors. In this review, we provide an overview of various theories of addiction to drugs of abuse and the neurobiology involved in elements of the addiction cycle. Specific focus is devoted to the role of the mesolimbic pathway in acute drug reinforcement and occasional drug use, the role of the mesocortical pathway and associated areas (e.g., the dorsal striatum) in escalation/dependence, and the contribution of these pathways and associated circuits to conditioned responses, drug craving, and loss of behavioral control that may underlie drug relapse. By enhancing the understanding of the neurobiological factors that mediate drug addiction, continued preclinical and clinical research will aid in the development of novel therapeutic interventions that can serve as effective long-term treatment strategies for drug-dependent individuals.
Collapse
Affiliation(s)
- Matthew W Feltenstein
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina 29425, USA
| | - Ronald E See
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina 29425, USA
- Department of Psychology, Westmont College, Santa Barbara, California 93108, USA
| | - Rita A Fuchs
- Integrative Physiology and Neuroscience, Washington State University College of Veterinary Medicine, Pullman, Washington 99164-7620, USA
| |
Collapse
|
36
|
Walcott AT, Ryabinin AE. Assessing effects of oxytocin on alcohol consumption in socially housed prairie voles using radio frequency tracking. Addict Biol 2021; 26:e12893. [PMID: 32160654 DOI: 10.1111/adb.12893] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 02/19/2020] [Accepted: 02/27/2020] [Indexed: 01/09/2023]
Abstract
Alcohol use disorder affects millions of people each year. Currently approved pharmacotherapies have limited success in treating this disorder. Evidence suggests that this lack of success is partly due to how these pharmacotherapies are tested in preclinical settings. The vast majority of preclinical studies assessing the effects of pharmacotherapies on alcohol or drug self-administration are done in individually housed animals. However, it is known that alcohol and drug intake are heavily influenced by social settings. Here, we adapted radio frequency tracking technology to determine the effects of oxytocin, a potential therapy for alcohol use disorder, on alcohol consumption in socially housed male and female prairie voles. Voluntary alcohol consumption in these animals resulted in high daily alcohol intakes, blood ethanol concentrations that are considered intoxicating, and central changes in FosB immunoreactivity, indicative of changes in neural activity. Prairie voles that received oxytocin temporarily reduced alcohol consumption but not alcohol preference, compared with control prairie voles regardless whether their cagemates received a similar treatment or not. Our results demonstrate that oxytocin can decrease consummatory behaviors in the presence of peers that are not receiving this treatment, and therefore, its potential use in clinical trials is warranted. Moreover, effectiveness of other pharmacotherapies in preclinical studies can be tested in mixed-treatment socially housed animals similarly to clinical studies in humans.
Collapse
Affiliation(s)
- Andre T. Walcott
- Department of Behavioral Neuroscience Oregon Health & Science University Portland OR 97239 USA
| | - Andrey E. Ryabinin
- Department of Behavioral Neuroscience Oregon Health & Science University Portland OR 97239 USA
| |
Collapse
|
37
|
Sundar M, Patel D, Young Z, Leong KC. Oxytocin and Addiction: Potential Glutamatergic Mechanisms. Int J Mol Sci 2021; 22:ijms22052405. [PMID: 33673694 PMCID: PMC7957657 DOI: 10.3390/ijms22052405] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 02/25/2021] [Accepted: 02/25/2021] [Indexed: 11/16/2022] Open
Abstract
Recently, oxytocin (OXT) has been investigated for its potential therapeutic role in addiction. OXT has been found to diminish various drug-seeking and drug-induced behaviors. Although its behavioral effects are well-established, there is not much consensus on how this neuropeptide exerts its effects. Previous research has given thought to how dopamine (DA) may be involved in oxytocinergic mechanisms, but there has not been as strong of a focus on the role that glutamate (Glu) has. The glutamatergic system is critical for the processing of rewards and the disruption of glutamatergic projections produces the behaviors seen in drug addicts. We introduce the idea that OXT has direct effects on Glu transmission within the reward processing pathway. Thus, OXT may reduce addictive behaviors by restoring abnormal drug-induced changes in the glutamatergic system and in its interactions with other neurotransmitters. This review offers insight into the mechanisms through which a potentially viable therapeutic target, OXT, could be used to reduce addiction-related behaviors.
Collapse
|
38
|
Manduca A, Carbone E, Schiavi S, Cacchione C, Buzzelli V, Campolongo P, Trezza V. The neurochemistry of social reward during development: What have we learned from rodent models? J Neurochem 2021; 157:1408-1435. [PMID: 33569830 DOI: 10.1111/jnc.15321] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 02/02/2021] [Accepted: 02/07/2021] [Indexed: 12/14/2022]
Abstract
Social rewards are fundamental to survival and overall health. Several studies suggest that adequate social stimuli during early life are critical for developing appropriate socioemotional and cognitive skills, whereas adverse social experiences negatively affect the proper development of brain and behavior, by increasing the susceptibility to develop neuropsychiatric conditions. Therefore, a better understanding of the neural mechanisms underlying social interactions, and their rewarding components in particular, is an important challenge of current neuroscience research. In this context, preclinical research has a crucial role: Animal models allow to investigate the neurobiological aspects of social reward in order to shed light on possible neurochemical alterations causing aberrant social reward processing in neuropsychiatric diseases, and they allow to test the validity and safety of innovative therapeutic strategies. Here, we discuss preclinical research that has investigated the rewarding properties of two forms of social interaction that occur in different phases of the lifespan of mammals, that is, mother-infant interaction and social interactions with peers, by focusing on the main neurotransmitter systems mediating their rewarding components. Together, the research performed so far helped to elucidate the mechanisms of social reward and its psychobiological components throughout development, thus increasing our understanding of the neurobiological substrates sustaining social functioning in health conditions and social dysfunction in major psychiatric disorders.
Collapse
Affiliation(s)
- Antonia Manduca
- Department of Science, Section of Biomedical Sciences and Technologies, Roma Tre University, Rome, Italy.,Neuroendocrinology, Metabolism and Neuropharmacology Unit, IRCSS Fondazione Santa Lucia, Rome, Italy
| | - Emilia Carbone
- Department of Science, Section of Biomedical Sciences and Technologies, Roma Tre University, Rome, Italy
| | - Sara Schiavi
- Department of Science, Section of Biomedical Sciences and Technologies, Roma Tre University, Rome, Italy
| | - Claudia Cacchione
- Department of Science, Section of Biomedical Sciences and Technologies, Roma Tre University, Rome, Italy
| | - Valeria Buzzelli
- Department of Science, Section of Biomedical Sciences and Technologies, Roma Tre University, Rome, Italy.,Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
| | - Patrizia Campolongo
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy.,Neurobiology of Behavior Laboratory, IRCSS Fondazione Santa Lucia, Rome, Italy
| | - Viviana Trezza
- Department of Science, Section of Biomedical Sciences and Technologies, Roma Tre University, Rome, Italy
| |
Collapse
|
39
|
Abstract
The hypothalamic peptide oxytocin has been increasingly recognized as a hormone and neurotransmitter with important effects on energy intake, metabolism, and body weight and is under investigation as a potential novel therapeutic agent for obesity. The main neurons producing oxytocin and expressing the oxytocin receptor are strategically located in brain areas known to be critically involved in homeostatic energy balance as well as hedonic and motivational aspects of eating behavior. In this chapter, we will review the central and peripheral physiology of oxytocin and the interaction of oxytocin with key hormones and neural circuitries that affect food intake and metabolism. Next, we will synthesize the available data on endogenous oxytocin levels related to caloric intake, body weight, and metabolic status. We will then review the effects of exogenous oxytocin administration on eating behavior, body weight, and metabolism in humans, including in healthy individuals as well as specific populations with suspected perturbations involving oxytocin pathways. Finally, we will address the promise and fundamental challenges of translating this line of research to clinical care.
Collapse
Affiliation(s)
- Liya Kerem
- Neuroendocrine Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States; Division of Pediatric Endocrinology, Massachusetts General Hospital for Children, Boston, MA, United States
| | - Elizabeth A Lawson
- Neuroendocrine Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States.
| |
Collapse
|
40
|
Wang JS, Liu JL, Zhang J, Tan J, Huang T, Lu CX, Peng XY, Guo Y, Zheng L. Descended Social Anxiety Disorder and Craving in Women Heroin Dependence Through Exercise Alerts Plasma Oxytocin Levels. Front Psychiatry 2021; 12:624993. [PMID: 34899404 PMCID: PMC8663167 DOI: 10.3389/fpsyt.2021.624993] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 10/25/2021] [Indexed: 11/13/2022] Open
Abstract
Purpose: This study explored the association between peripheral blood oxytocin (OT) and social anxiety disorder (SAD) and cue-induced cravings in female heroin addicts. The effect of exercise on alleviation of SAD and OT levels was also explored. Methods: A total of 72 females with heroin dependence were assigned to three groups based on SAD severity. The three groups were Non-SAD control, SAD control, and SAD exercise groups. Subjects in the SAD exercise group underwent aerobic exercise and resistance training for 8 weeks (60 min/day, 5 days/week). Enzyme-linked immunosorbent assay analysis and Liebowitz Social Anxiety Scale (LSAS) scores were used to determine plasma OT concentration and SAD, respectively. Cue-induced craving was assessed using Visual Analog Scale (VAS) and Desires for Drug Questionnaire (DDQ). Mixed-effect analysis of variance and Pearson correlation analysis were used to explore the effect and correlation between different parameters. Results: OT levels in the SAD exercise group were significantly high after exercise (p < 0.01). LSAS, VAS, and DDQ ("Desire and Intention" and "Negative reinforcement") scores in the SAD exercise group were significantly lower after exercise (p < 0.01). Plasma OT level was negatively correlated with LSAS score (r = -0.534, p < 0.001), VAS score (r = -0.609, p < 0.001), "Desire and Intention" score (r = -0.555, p < 0.001), and "Negative reinforcement" score (r = -0.332, p < 0.01) and positively correlated with the "control" score (r = 0.258, p < 0.05). LSAS was positively correlated with VAS score (r = 0.588, p < 0.001) and "Desire and Intention" score (r = 0.282, p < 0.05). Conclusions: The findings of the present study indicate that plasma OT is a potential peripheral biomarker for prediction of the severity of social anxiety in female heroin withdrawal patients. Aerobic exercise combined with resistance training plus incremental load for 8 weeks can increase plasma OT levels and significantly reduce severity of SAD and cue-induced cravings in female heroin addicts.
Collapse
Affiliation(s)
- Jing Song Wang
- Key Laboratory of Physical Fitness and Sports Rehabilitation of the Hunan Province, College of Physical Education, Hunan Normal University, Changsha, China
| | - Jing Lin Liu
- Key Laboratory of Physical Fitness and Sports Rehabilitation of the Hunan Province, College of Physical Education, Hunan Normal University, Changsha, China
| | - Jun Zhang
- Hunan Judicial Police Vocational College, Changsha, China
| | - Jun Tan
- College of Physical Education, Hunan International Economics University, Changsha, China
| | - Ting Huang
- Key Laboratory of Physical Fitness and Sports Rehabilitation of the Hunan Province, College of Physical Education, Hunan Normal University, Changsha, China
| | - Chun Xia Lu
- Key Laboratory of Physical Fitness and Sports Rehabilitation of the Hunan Province, College of Physical Education, Hunan Normal University, Changsha, China
| | - Xi Yang Peng
- Key Laboratory of Physical Fitness and Sports Rehabilitation of the Hunan Province, College of Physical Education, Hunan Normal University, Changsha, China
| | - Yin Guo
- Key Laboratory of Physical Fitness and Sports Rehabilitation of the Hunan Province, College of Physical Education, Hunan Normal University, Changsha, China
| | - Lan Zheng
- Key Laboratory of Physical Fitness and Sports Rehabilitation of the Hunan Province, College of Physical Education, Hunan Normal University, Changsha, China
| |
Collapse
|
41
|
Reguilón MD, Ferrer-Pérez C, Miñarro J, Rodríguez-Arias M. Oxytocin reverses ethanol consumption and neuroinflammation induced by social defeat in male mice. Horm Behav 2021; 127:104875. [PMID: 33069753 DOI: 10.1016/j.yhbeh.2020.104875] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 09/29/2020] [Accepted: 10/06/2020] [Indexed: 02/07/2023]
Abstract
Oxytocin (OXT) modulates social interactions, attenuates stressful responses and can decrease drug-seeking and taking behaviors. In previous studies, we observed that social defeat (SD) induced a long-lasting increase in ethanol intake and neuroinflammation in male mice. We also know that OXT blocks the increase in cocaine reward induced by SD. Therefore, in the present study we aimed to evaluate the effect of 1 mg/kg of OXT administered 30 min before each episode of SD on ethanol consumption and the neuroinflammatory response in adult male mice. Three weeks after the last SD, mice underwent oral ethanol self-administration (SA) procedure, and striatal levels of the two chemokines CX3CL1 and CXCL12 were measured after the last SD and at the end of the ethanol SA. OXT administration blocked the increase in voluntary ethanol consumption observed in defeated mice, although it did not affect motivation for ethanol. An increase in the striatal levels of CX3CL1 and CXCL12 was observed in defeated animals immediately after the last defeat and after the ethanol SA. However, defeated mice treated with OXT did not show this increase in the neuroinflammatory response. In conclusion, OXT treatment can be a powerful therapeutic target to reduce the negative effects of social stress on ethanol consumption and the neuroinflammatory process.
Collapse
Affiliation(s)
- M D Reguilón
- Department of Psychobiology, Facultad de Psicología, Universitat de Valencia, Avda. Blasco Ibáñez, 21, 46010 Valencia, Spain
| | - C Ferrer-Pérez
- Department of Psychobiology, Facultad de Psicología, Universitat de Valencia, Avda. Blasco Ibáñez, 21, 46010 Valencia, Spain
| | - J Miñarro
- Department of Psychobiology, Facultad de Psicología, Universitat de Valencia, Avda. Blasco Ibáñez, 21, 46010 Valencia, Spain
| | - M Rodríguez-Arias
- Department of Psychobiology, Facultad de Psicología, Universitat de Valencia, Avda. Blasco Ibáñez, 21, 46010 Valencia, Spain.
| |
Collapse
|
42
|
Unraveling oxytocin's peripheral vs. central mechanisms. Neuropsychopharmacology 2021; 46:273-274. [PMID: 32572151 PMCID: PMC7852891 DOI: 10.1038/s41386-020-0742-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Accepted: 06/11/2020] [Indexed: 11/08/2022]
|
43
|
Serum Oxytocin Level Among Male Patients With Opioid Dependence and Its Relation to Craving. ADDICTIVE DISORDERS & THEIR TREATMENT 2020. [DOI: 10.1097/adt.0000000000000231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
44
|
Development of Magnetic Nanobeads Modified by Artificial Fluorescent Peptides for the Highly Sensitive and Selective Analysis of Oxytocin. SENSORS 2020; 20:s20205956. [PMID: 33096804 PMCID: PMC7588987 DOI: 10.3390/s20205956] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 10/20/2020] [Indexed: 12/18/2022]
Abstract
We describe two novel fluorescent peptides (compounds 1 and 2) targeting oxytocin with a boron-dipyrromethenyl group as the fluorophore bound to an artificial peptide based on the oxytocin receptor, and their application for the analysis of oxytocin levels in human serum using nanometer-sized magnetic beads modified by fluorescent peptides (FMB-1 and FMB-2). Under the optimized experimental protocols, FMB-1 and FMB-2 emitted low levels of fluorescence but emitted much higher levels of fluorescence when associated with oxytocin. The detection limit of oxytocin by FMB-2 was 0.4 pM, which is approximately 37.5 times higher than that of conventional methods, such as ELISA. Using these fluorescent sensors, oxytocin was specifically detected over a wide linear range with high sensitivity, good reusability, stability, precision, and reproducibility. This fluorescent sensor-based detection system thus enabled the measurement of oxytocin levels in human serum, which has widespread applications for oxytocin assays across varied research fields.
Collapse
|
45
|
Cano-Cebrián MJ, Fernández-Rodríguez S, Hipólito L, Granero L, Polache A, Zornoza T. Efficacy of N-acetylcysteine in the prevention of alcohol relapse-like drinking: Study in long-term ethanol-experienced male rats. J Neurosci Res 2020; 99:638-648. [PMID: 33063355 DOI: 10.1002/jnr.24736] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 09/07/2020] [Accepted: 10/01/2020] [Indexed: 02/06/2023]
Abstract
Alcohol use disorders are chronic and highly relapsing disorders, thus alcoholic patients have a high rate of recidivism for drug use even after long periods of abstinence. The literature points to the potential usefulness of N-acetylcysteine (NAC) in the management of several substance use disorders probably due to its capacity to restore brain homeostasis of the glutamate system disrupted in addiction. However, there is little evidence in the case of alcohol. The aim of this study was to explore the potential anti-relapse efficacy of NAC using the alcohol deprivation effect (ADE) model in long-term experienced rats. Two experiments were performed in male Wistar rats to: (a) test the efficacy of NAC to prevent relapse and (b) discriminate the best administration schedule (intermittent vs. continuous) for NAC. In the first experiment, animals were implanted with mini-osmotic pumps delivering 0 or 1 mg/hr NAC during 14 days. In a second experiment, rats received 0, 60, or 100 mg/kg once daily by subcutaneous injection. The efficacy to prevent ADE was evaluated in both experiments. NAC subcutaneously administered, either by continuous infusion or by intermittent injections regimen, is able to block the ADE. The best results were obtained after using 60 mg/kg NAC dose. Our findings support the hypothesis that NAC may represent a valuable therapy in the management of alcohol relapse.
Collapse
Affiliation(s)
- María José Cano-Cebrián
- Departament de Farmàcia i Tecnologia Farmacèutica i Parasitologia, Universitat de València, Burjassot, Spain
| | - Sandra Fernández-Rodríguez
- Departament de Farmàcia i Tecnologia Farmacèutica i Parasitologia, Universitat de València, Burjassot, Spain
| | - Lucia Hipólito
- Departament de Farmàcia i Tecnologia Farmacèutica i Parasitologia, Universitat de València, Burjassot, Spain
| | - Luis Granero
- Departament de Farmàcia i Tecnologia Farmacèutica i Parasitologia, Universitat de València, Burjassot, Spain
| | - Ana Polache
- Departament de Farmàcia i Tecnologia Farmacèutica i Parasitologia, Universitat de València, Burjassot, Spain
| | - Teodoro Zornoza
- Departament de Farmàcia i Tecnologia Farmacèutica i Parasitologia, Universitat de València, Burjassot, Spain
| |
Collapse
|
46
|
László K, Péczely L, Géczi F, Kovács A, Zagoracz O, Ollmann T, Kertes E, Kállai V, László B, Berta B, Karádi Z, Lénárd L. The role of D2 dopamine receptors in oxytocin induced place preference and anxiolytic effect. Horm Behav 2020; 124:104777. [PMID: 32439347 DOI: 10.1016/j.yhbeh.2020.104777] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 04/24/2020] [Accepted: 05/15/2020] [Indexed: 12/01/2022]
Abstract
Neuropeptide oxytocin (OT) is involved in the regulation of social and non-social behaviour. The central nucleus of amygdala (CeA), part of the limbic system, plays an important role in learning, memory, anxiety and reinforcing mechanisms. CeA has been shown to be rich in OT receptors in rodents. Our previous findings indicated that OT in the rat CeA has a dose dependent rewarding and anxiolytic effect. The aim of our present study was to examine in the CeA the possible interaction of OT and D2 dopamine (DA) receptor antagonist Sulpiride on reinforcement in place preference test and on anxiety in elevated plus maze test. Wistar rats were microinjected bilaterally with 10 ng OT. In different group of animals 4 μg D2 DA receptor antagonist was applied. Other animals received D2 DA receptor antagonist 15 min before 10 ng OT treatment or vehicle solution into the CeA. Rats receiving 10 ng OT spent significantly longer time in the treatment quadrant during the test session in conditioned place preference test. Prior treatment with D2 DA receptor antagonist blocked the rewarding effects of OT. Antagonist in itself did not influence the time rats spent in the treatment quadrant. In elevated plus maze test, rats receiving 10 ng OT spent significantly longer time on the open arms. Prior treatment with D2 DA receptor antagonist blocked the effects of OT. Our results show that DA system plays a role in positive reinforcing and anxiolytic effects of OT because D2 DA receptor antagonist can block these actions.
Collapse
MESH Headings
- Amygdala/drug effects
- Amygdala/metabolism
- Animals
- Anti-Anxiety Agents/pharmacology
- Anxiety/drug therapy
- Behavior, Animal/drug effects
- Conditioning, Classical/drug effects
- Dopamine D2 Receptor Antagonists/pharmacology
- Fear/drug effects
- Freezing Reaction, Cataleptic/drug effects
- Male
- Maze Learning/drug effects
- Oxytocin/pharmacology
- Rats
- Rats, Wistar
- Receptors, Dopamine D1/metabolism
- Receptors, Dopamine D2/metabolism
- Receptors, Dopamine D2/physiology
- Receptors, Oxytocin/metabolism
- Receptors, Oxytocin/physiology
- Reinforcement, Psychology
- Reward
- Spatial Behavior/drug effects
- Sulpiride/pharmacology
Collapse
Affiliation(s)
- K László
- Institute of Physiology, University of Pécs, Medical School, Pécs, Hungary; Neuroscience Center, University of Pécs, Pécs, Hungary.
| | - L Péczely
- Institute of Physiology, University of Pécs, Medical School, Pécs, Hungary; Neuroscience Center, University of Pécs, Pécs, Hungary
| | - F Géczi
- Institute of Physiology, University of Pécs, Medical School, Pécs, Hungary; Neuroscience Center, University of Pécs, Pécs, Hungary
| | - A Kovács
- Institute of Physiology, University of Pécs, Medical School, Pécs, Hungary; Neuroscience Center, University of Pécs, Pécs, Hungary
| | - O Zagoracz
- Institute of Physiology, University of Pécs, Medical School, Pécs, Hungary; Neuroscience Center, University of Pécs, Pécs, Hungary
| | - T Ollmann
- Institute of Physiology, University of Pécs, Medical School, Pécs, Hungary; Neuroscience Center, University of Pécs, Pécs, Hungary
| | - E Kertes
- Institute of Physiology, University of Pécs, Medical School, Pécs, Hungary; Neuroscience Center, University of Pécs, Pécs, Hungary
| | - V Kállai
- Institute of Physiology, University of Pécs, Medical School, Pécs, Hungary; Neuroscience Center, University of Pécs, Pécs, Hungary
| | - B László
- Institute of Physiology, University of Pécs, Medical School, Pécs, Hungary; Neuroscience Center, University of Pécs, Pécs, Hungary
| | - B Berta
- Institute of Physiology, University of Pécs, Medical School, Pécs, Hungary; Neuroscience Center, University of Pécs, Pécs, Hungary
| | - Z Karádi
- Institute of Physiology, University of Pécs, Medical School, Pécs, Hungary; Neuroscience Center, University of Pécs, Pécs, Hungary; Molecular Endocrinology and Neurophysiology Research Group, University of Pécs, Szentágothai Center, Pécs, Hungary
| | - L Lénárd
- Institute of Physiology, University of Pécs, Medical School, Pécs, Hungary; Neuroscience Center, University of Pécs, Pécs, Hungary; Molecular Endocrinology and Neurophysiology Research Group, University of Pécs, Szentágothai Center, Pécs, Hungary
| |
Collapse
|
47
|
Sharma SR, Gonda X, Dome P, Tarazi FI. What's Love Got to do with it: Role of oxytocin in trauma, attachment and resilience. Pharmacol Ther 2020; 214:107602. [PMID: 32512017 DOI: 10.1016/j.pharmthera.2020.107602] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 05/27/2020] [Indexed: 02/06/2023]
Abstract
Oxytocin (OT) is a neurohypophysial hormone and neuropeptide produced by the hypothalamus and released by the pituitary gland. It has multiple physiological roles including stimulation of parturition and lactation, and promotion of pro-adaptive social behaviors necessary for mammalian survival. OT interacts with one receptor subtype: the OT receptor (OTR) which, upon stimulation, triggers different intracellular signal transduction cascades to mediate its physiological actions. Preclinical studies show that OT regulates social behaviors such as pair bonding, recognition and social interaction. It also coordinates the activation of the hypothalamic-pituitary-adrenal (HPA) axis and the release of corticotrophin-releasing hormone. Further evidence suggests that OT plays an important role in regulating caloric intake and metabolism, and in maintaining electrolyte and cardiovascular homeostasis. OT is also involved in attenuating the neurophysiological and neurochemical effects of trauma on the brain and body by facilitating both physical attachment such as wound healing, and psychological/social attachment, thereby increasing resilience to subsequent traumatic events. Clinical trials have reported that intranasal administration of OT provides therapeutic benefits for patients diagnosed with traumatic stress-related diseases such as major depressive disorders and post-traumatic stress disorder. OT's therapeutic benefits may result from context-dependent interactions with key neural pathways (social, cognitive, and reward), neurotransmitters (dopamine, norepinephrine, serotonin, and endogenous opioids), and biomarkers (adrenocorticotropic hormone, cortisol, and dehydroepiandrosterone sulfate), that lead to a decrease in stress -associated behaviors, and facilitate post-traumatic growth, ultimately leading to increased resilience, through improved social cohesion and attachment. OT induced-augmentation of physical and cognitive resilience may play a significant role in both the prevention of, and improved clinical outcomes for, traumatic stress-related disorders following either acute or enduring traumatic experiences.
Collapse
Affiliation(s)
- Samata R Sharma
- Department of Psychiatry, Harvard Medical School, Brigham and Women's Hospital, Boston, MA 02115, USA.
| | - Xenia Gonda
- Department of Psychiatry and Psychotherapy, Semmelweis University, Budapest, Hungary; MTA-SE Neurochemistry and Neuropsychopharmacology Research Group, Hungarian Academy of Sciences and Semmelweis University, Budapest, Hungary; NAP-2-SE New Antidepressant Target Research Group, Hungarian Brain Research Program, Semmelweis University, Budapest, Hungary; Laboratory of Suicide Prevention and Research, National Institute for Psychiatry and Addictions, Budapest, Hungary
| | - Peter Dome
- Department of Psychiatry and Psychotherapy, Semmelweis University, Budapest, Hungary; Laboratory of Suicide Prevention and Research, National Institute for Psychiatry and Addictions, Budapest, Hungary
| | - Frank I Tarazi
- Department of Psychiatry and Neuroscience Program, Harvard Medical School, McLean Hospital, Belmont, MA 02478, USA
| |
Collapse
|
48
|
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
|
49
|
Braidy N, Villalva MD, van Eeden S. Sobriety and Satiety: Is NAD+ the Answer? Antioxidants (Basel) 2020; 9:antiox9050425. [PMID: 32423100 PMCID: PMC7278809 DOI: 10.3390/antiox9050425] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 05/03/2020] [Accepted: 05/05/2020] [Indexed: 02/07/2023] Open
Abstract
Nicotinamide adenine dinucleotide (NAD+) is an essential pyridine nucleotide that has garnered considerable interest in the last century due to its critical role in cellular processes associated with energy production, cellular protection against stress and longevity. Research in NAD+ has been reinvigorated by recent findings that components of NAD+ metabolism and NAD-dependent enzymes can influence major signalling processes associated with the neurobiology of addiction. These studies implicate raising intracellular NAD+ levels as a potential target for managing and treating addictive behaviour and reducing cravings and withdrawal symptoms in patients with food addiction and/or substance abuse. Since clinical studies showing the use of NAD+ for the treatment of addiction are limited, this review provides literature evidence that NAD+ can influence the neurobiology of addiction and may have benefits as an anti-addiction intervention.
Collapse
Affiliation(s)
- Nady Braidy
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, NSW 2052, Australia;
- Correspondence:
| | - Maria D. Villalva
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, NSW 2052, Australia;
| | - Sam van Eeden
- Centre for Cutaneous Research, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 4NS, UK;
| |
Collapse
|
50
|
Ferrer-Pérez C, Reguilón MD, Miñarro J, Rodríguez-Arias M. Endogenous oxytocin is essential for the buffering effects of pair housing against the increase in cocaine reward induced by social stress. Physiol Behav 2020; 221:112913. [PMID: 32298668 DOI: 10.1016/j.physbeh.2020.112913] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 04/02/2020] [Accepted: 04/06/2020] [Indexed: 12/11/2022]
Abstract
Social factors have a dual influence on addictive disorders. While social defeat stress in rodents increases the response to drug reward, positive social conditions, such as pair housing, increase stress resilience. The objective of the present study was to confirm whether oxytocin (OT) mediates this social buffering. To this end, male mice were housed in pairs and administered the OT receptor antagonist atosiban prior to each stress episode or for ten days after the stress protocol. The response to cocaine was assessed using a conditioned place preference paradigm. Our results confirmed that OT activity mediates the protective effect of pair housing and highlights its therapeutic potential.
Collapse
Affiliation(s)
- Carmen Ferrer-Pérez
- Department of Psychobiology, Faculty of Psychology, Universitat de València, Valencia, Spain
| | - Marina D Reguilón
- Department of Psychobiology, Faculty of Psychology, Universitat de València, Valencia, Spain
| | - José Miñarro
- Department of Psychobiology, Faculty of Psychology, Universitat de València, Valencia, Spain
| | - Marta Rodríguez-Arias
- Department of Psychobiology, Faculty of Psychology, Universitat de València, Valencia, Spain.
| |
Collapse
|