1
|
Jones JD, Arout CA, Luba R, Murugesan D, Madera G, Gorsuch L, Schusterman R, Martinez S. The influence of drug class on reward in substance use disorders. Pharmacol Biochem Behav 2024; 240:173771. [PMID: 38670466 PMCID: PMC11162950 DOI: 10.1016/j.pbb.2024.173771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 03/26/2024] [Accepted: 04/16/2024] [Indexed: 04/28/2024]
Abstract
In the United States, the societal costs associated with drug use surpass $500 billion annually. The rewarding and reinforcing properties that drive the use of these addictive substances are typically examined concerning the neurobiological effects responsible for their abuse potential. In this review, terms such as "abuse potential," "drug," and "addictive properties" are used due to their relevance to the methodological, theoretical, and conceptual framework for understanding the phenomenon of drug-taking behavior and the associated body of preclinical and clinical literature. The use of these terms is not intended to cast aspersions on individuals with substance use disorders (SUD). Understanding what motivates substance use has been a focus of SUD research for decades. Much of this corpus of work has focused on the shared effects of each drug class to increase dopaminergic transmission within the central reward pathways of the brain, or the "reward center." However, the precise influence of each drug class on dopamine signaling, and the extent thereof, differs considerably. Furthermore, the aforementioned substances have effects on several neurobiological targets that mediate and modulate their addictive properties. The current manuscript sought to review the influence of drug class on the rewarding effects of each of the major pharmacological classes of addictive drugs (i.e., psychostimulants, opioids, nicotine, alcohol, and cannabinoids). Our review suggests that even subtle differences in drug effects can result in significant variability in the subjective experience of the drug, altering rewarding and other reinforcing effects. Additionally, this review will argue that reward (i.e., the attractive and motivational property of a stimulus) alone is not sufficient to explain the abuse liability of these substances. Instead, abuse potential is best examined as a function of both positive and negative reinforcing drug effects (i.e., stimuli that the subject will work to attain and stimuli that the subject will work to end or avoid, respectively). Though reward is central to drug use, the factors that motivate and maintain drug taking are varied and complex, with much to be elucidated.
Collapse
Affiliation(s)
- Jermaine D Jones
- Division on Substance Use Disorders, Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, 1051 Riverside Drive, New York, NY 10032, USA.
| | - Caroline A Arout
- Division on Substance Use Disorders, Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, 1051 Riverside Drive, New York, NY 10032, USA
| | - Rachel Luba
- Division on Substance Use Disorders, Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, 1051 Riverside Drive, New York, NY 10032, USA
| | - Dillon Murugesan
- CUNY School of Medicine, 160 Convent Avenue, New York, NY 10031, USA
| | - Gabriela Madera
- Division on Substance Use Disorders, Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, 1051 Riverside Drive, New York, NY 10032, USA
| | - Liam Gorsuch
- Department of Psychiatry, The University of British Columbia, 430-5950 University Blvd., Vancouver V6T 1Z3, BC, Canada
| | - Rebecca Schusterman
- Division on Substance Use Disorders, Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, 1051 Riverside Drive, New York, NY 10032, USA
| | - Suky Martinez
- Division on Substance Use Disorders, Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, 1051 Riverside Drive, New York, NY 10032, USA
| |
Collapse
|
2
|
The Opioid System in Depression. Neurosci Biobehav Rev 2022; 140:104800. [PMID: 35914624 PMCID: PMC10166717 DOI: 10.1016/j.neubiorev.2022.104800] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 06/20/2022] [Accepted: 07/25/2022] [Indexed: 12/16/2022]
Abstract
Opioid receptors are widely distributed throughout the brain and play an essential role in modulating aspects of human mood, reward, and well-being. Accumulating evidence indicates the endogenous opioid system is dysregulated in depression and that pharmacological modulators of mu, delta, and kappa opioid receptors hold potential for the treatment of depression. Here we review animal and clinical data, highlighting evidence to support: dysregulation of the opioid system in depression, evidence for opioidergic modulation of behavioural processes and brain regions associated with depression, and evidence for opioidergic modulation in antidepressant responses. We evaluate clinical trials that have examined the safety and efficacy of opioidergic agents in depression and consider how the opioid system may be involved in the effects of other treatments, including ketamine, that are currently understood to exert antidepressant effects through non-opioidergic actions. Finally, we explore key neurochemical and molecular mechanisms underlying the potential therapeutic effects of opioid system engagement, that together provides a rationale for further investigation into this relevant target in the treatment of depression.
Collapse
|
3
|
Custodio RJP, Kim M, Sayson LV, Ortiz DM, Buctot D, Lee HJ, Cheong JH, Kim HJ. Regulation of clock and clock-controlled genes during morphine reward and reinforcement: Involvement of the period 2 circadian clock. J Psychopharmacol 2022; 36:875-891. [PMID: 35486444 DOI: 10.1177/02698811221089040] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND Morphine abuse is a devastating disorder that affects millions of people worldwide, and literature evidence indicates a relationship between opioid abuse and the circadian clock. AIM We explored morphine reward and reinforcement using mouse models with Per2 gene modifications (knockout (KO); overexpression (OE)). METHODS Mice were exposed to various behavioral, electroencephalographic, pharmacological, and molecular tests to assess the effects of morphine and identify the underlying mechanisms with a focus on reward and reinforcement and the corresponding involvement of circadian and clock-controlled gene regulation. RESULTS Per2 deletion enhances morphine-induced analgesia, locomotor sensitization, conditioned place preference (CPP), and self-administration (SA) in mice, whereas its overexpression attenuated these effects. In addition, reduced withdrawal was observed in Per2 KO mice, whereas an augmented withdrawal response was observed in Per2 OE mice. Moreover, naloxone and SCH 23390 blocked morphine CPP in Per2 KO and wild-type (WT) mice. The rewarding (CPP) and reinforcing effects (SA) observed in morphine-conditioned and morphine self-administered Per2 KO and WT mice were accompanied by activated μ-opioid and dopamine D1 receptors and TH in the mesolimbic (VTA/NAcc) system. Furthermore, genetic modifications of Per2 in mice innately altered some clock genes in response to morphine. CONCLUSION These findings improve our understanding of the role of Per2 in morphine-induced psychoactive effects. Our data and those obtained in previous studies indicate that targeting Per2 may have applicability in the treatment of substance abuse.
Collapse
Affiliation(s)
- Raly James Perez Custodio
- School of Pharmacy, Jeonbuk National University, Jeonju-si, Republic of Korea.,Uimyung Research Institute for Neuroscience, Department of Pharmacy, Sahmyook University, Seoul, Republic of Korea
| | - Mikyung Kim
- Department of Chemistry & Life Science, Sahmyook University, Seoul, Republic of Korea
| | - Leandro Val Sayson
- Uimyung Research Institute for Neuroscience, Department of Pharmacy, Sahmyook University, Seoul, Republic of Korea
| | - Darlene Mae Ortiz
- Uimyung Research Institute for Neuroscience, Department of Pharmacy, Sahmyook University, Seoul, Republic of Korea
| | - Danilo Buctot
- Uimyung Research Institute for Neuroscience, Department of Pharmacy, Sahmyook University, Seoul, Republic of Korea
| | - Hyun Jun Lee
- Uimyung Research Institute for Neuroscience, Department of Pharmacy, Sahmyook University, Seoul, Republic of Korea
| | - Jae Hoon Cheong
- School of Pharmacy, Jeonbuk National University, Jeonju-si, Republic of Korea
| | - Hee Jin Kim
- Uimyung Research Institute for Neuroscience, Department of Pharmacy, Sahmyook University, Seoul, Republic of Korea
| |
Collapse
|
4
|
Gutridge AM, Chakraborty S, Varga BR, Rhoda ES, French AR, Blaine AT, Royer QH, Cui H, Yuan J, Cassell RJ, Szabó M, Majumdar S, van Rijn RM. Evaluation of Kratom Opioid Derivatives as Potential Treatment Option for Alcohol Use Disorder. Front Pharmacol 2021; 12:764885. [PMID: 34803709 PMCID: PMC8596301 DOI: 10.3389/fphar.2021.764885] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 10/08/2021] [Indexed: 12/13/2022] Open
Abstract
Background and Purpose:Mitragyna speciosa extract and kratom alkaloids decrease alcohol consumption in mice at least in part through actions at the δ-opioid receptor (δOR). However, the most potent opioidergic kratom alkaloid, 7-hydroxymitragynine, exhibits rewarding properties and hyperlocomotion presumably due to preferred affinity for the mu opioid receptor (µOR). We hypothesized that opioidergic kratom alkaloids like paynantheine and speciogynine with reduced µOR potency could provide a starting point for developing opioids with an improved therapeutic window to treat alcohol use disorder. Experimental Approach: We characterized paynantheine, speciociliatine, and four novel kratom-derived analogs for their ability to bind and activate δOR, µOR, and κOR. Select opioids were assessed in behavioral assays in male C57BL/6N WT and δOR knockout mice. Key Results: Paynantheine (10 mg∙kg−1, i.p.) produced aversion in a limited conditioned place preference (CPP) paradigm but did not produce CPP with additional conditioning sessions. Paynantheine did not produce robust antinociception but did block morphine-induced antinociception and hyperlocomotion. Yet, at 10 and 30 mg∙kg−1 doses (i.p.), paynantheine did not counteract morphine CPP. 7-hydroxypaynantheine and 7-hydroxyspeciogynine displayed potency at δOR but limited µOR potency relative to 7-hydroxymitragynine in vitro, and dose-dependently decreased voluntary alcohol consumption in WT but not δOR in KO mice. 7-hydroxyspeciogynine has a maximally tolerated dose of at least 10 mg∙kg−1 (s.c.) at which it did not produce significant CPP neither alter general locomotion nor induce noticeable seizures. Conclusion and Implications: Derivatizing kratom alkaloids with the goal of enhancing δOR potency and reducing off-target effects could provide a pathway to develop novel lead compounds to treat alcohol use disorder with an improved therapeutic window.
Collapse
Affiliation(s)
- Anna M Gutridge
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, United States
| | - Soumen Chakraborty
- Center for Clinical Pharmacology, University of Heath Sciences and Pharmacy at St. Louis and Washington University School of Medicine, St. Louis, MO, United States
| | - Balazs R Varga
- Center for Clinical Pharmacology, University of Heath Sciences and Pharmacy at St. Louis and Washington University School of Medicine, St. Louis, MO, United States
| | - Elizabeth S Rhoda
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, United States
| | - Alexander R French
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, United States.,Purdue Institute for Integrative Neuroscience, West Lafayette, IN, United States
| | - Arryn T Blaine
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, United States
| | - Quinten H Royer
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, United States
| | - Haoyue Cui
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, United States
| | - Jinling Yuan
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, United States
| | - Robert J Cassell
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, United States.,Purdue Institute for Drug Discovery, West Lafayette, IN, United States
| | | | - Susruta Majumdar
- Center for Clinical Pharmacology, University of Heath Sciences and Pharmacy at St. Louis and Washington University School of Medicine, St. Louis, MO, United States
| | - Richard M van Rijn
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, United States.,Purdue Institute for Integrative Neuroscience, West Lafayette, IN, United States.,Purdue Institute for Drug Discovery, West Lafayette, IN, United States
| |
Collapse
|
5
|
Kaski SW, White AN, Gross JD, Siderovski DP. Potential for Kappa-Opioid Receptor Agonists to Engineer Nonaddictive Analgesics: A Narrative Review. Anesth Analg 2021; 132:406-419. [PMID: 33332902 DOI: 10.1213/ane.0000000000005309] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
A serious adverse effect of prescription opioid analgesics is addiction, both to these analgesics and to illicit drugs like heroin that also activate the µ-opioid receptor (MOR). Opioid use disorder (OUD) and opioid overdose deaths represent a current American health crisis, and the prescription of opioid analgesics has contributed significantly to this crisis. While prescription opioids are highly effective analgesics, there currently exists no facile way to use them for extended periods without the risk of addiction. If addiction caused by MOR-targeting analgesics could be blocked by blending in a new "antiaddiction" ingredient that does not diminish analgesia and does not introduce its own therapeutically limiting side effects, then continued clinical use of prescription opioids for treating pain could be maintained (or even enhanced) instead of curtailed. In this narrative review, we contextualize this hypothesis, first with a brief overview of the current American opioid addiction crisis. The neurobiology of 2 key receptors in OUD development, MOR and the κ-opioid receptor (KOR), is then discussed to highlight the neuroanatomical features and circuitry in which signal transduction from these receptors lie in opposition-creating opportunities for pharmacological intervention in curtailing the addictive potential of MOR agonism. Prior findings with mixed MOR/KOR agonists are considered before exploring new potential avenues such as biased KOR agonists. New preclinical data are highlighted, demonstrating that the G protein-biased KOR agonist nalfurafine reduces the rewarding properties of MOR-targeting analgesics and enhances MOR-targeting analgesic-induced antinociception. Finally, we discuss the recent discovery that a regulator of G protein signaling (namely, RGS12) is a key component of signaling bias at KOR, presenting another drug discovery target toward identifying a single agent or adjuvant to be added to traditional opioid analgesics that could reduce or eliminate the addictive potential of the latter drug.
Collapse
Affiliation(s)
- Shane W Kaski
- From the Departments of Neuroscience and Behavioral Medicine & Psychiatry, West Virginia University, Morgantown, West Virginia
| | - Allison N White
- From the Departments of Neuroscience and Behavioral Medicine & Psychiatry, West Virginia University, Morgantown, West Virginia
| | - Joshua D Gross
- Department of Cell Biology, Duke University Medical Center, Durham, North Carolina
| | - David P Siderovski
- Department of Pharmacology & Neuroscience, University of North Texas Health Science Center, Fort Worth, Texas
| |
Collapse
|
6
|
Townsend EA, Negus SS, Banks ML. Medications Development for Treatment of Opioid Use Disorder. Cold Spring Harb Perspect Med 2021; 11:a039263. [PMID: 31932466 PMCID: PMC7778216 DOI: 10.1101/cshperspect.a039263] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
This review describes methods for preclinical evaluation of candidate medications to treat opioid use disorder (OUD). The review is founded on the propositions that (1) drug self-administration procedures provide the most direct method for assessment of medication effectiveness, (2) procedures that assess choice between opioid and nondrug reinforcers are especially useful, and (3) states of opioid dependence and withdrawal profoundly influence both opioid reinforcement and effects of candidate medications. Effects of opioid medications and vaccines on opioid choice in nondependent and opioid-dependent subjects are reviewed. Various nonopioid medications have also been examined, but none yet have been identified that safely and reliably reduce opioid choice. Future research will focus on (1) strategies for increasing safety and/or effectiveness of opioid medications (e.g., G-protein-biased μ-opioid agonists), and (2) continued development of nonopioid medications (e.g., clonidine) that might serve as adjunctive agents to current opioid medications.
Collapse
Affiliation(s)
- E Andrew Townsend
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia 23298, USA
| | - S Stevens Negus
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia 23298, USA
| | - Matthew L Banks
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia 23298, USA
| |
Collapse
|
7
|
Collins D, Zhang Y, Blendy J, Kreek MJ. Murine model of OPRM1 A118G alters oxycodone self-administration and locomotor activation, but not conditioned place preference. Neuropharmacology 2020; 167:107864. [DOI: 10.1016/j.neuropharm.2019.107864] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 11/03/2019] [Accepted: 11/24/2019] [Indexed: 12/18/2022]
|
8
|
Dextromethorphan and bupropion reduces high level remifentanil self-administration in rats. Pharmacol Biochem Behav 2020; 193:172919. [PMID: 32246985 DOI: 10.1016/j.pbb.2020.172919] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 03/30/2020] [Accepted: 03/30/2020] [Indexed: 02/04/2023]
Abstract
Opiate addiction has risen substantially during the past decade. New treatments to combat opiate addiction are sorely needed. The current study was conducted to determine the acute individual and interactive effects of bupropion and dextromethorphan in a rat model of opiate self-administration using the short-acting synthetic opioid remifentanil. Both of these drugs have been found to reduce self-administration of nicotine. Bupropion and dextromethorphan and their combination had differential effects depending on whether the rats showed higher or lower baseline remifentanil self-administration. The rats with higher initial remifentanil self-administration showed a significant decrease in remifentanil self-administration with bupropion or dextromethorphan treatment, compared to the vehicle control condition. This decrease in self-remifentanil administration was most pronounced when combination of the higher doses of bupropion and dextromethorphan were administered. In contrast, the rats with lower baseline remifentanil self-administration showed the opposite effect of drug treatment with an increase in remifentanil self-administration with bupropion treatment compared to the vehicle control condition. Dextromethorphan had no significant effect inthis group. This study shows that combination bupropion and dextromethorphan affects remifentanil self-administration in a complex fashion with differential effects on low and high baseline responders. In subjects with high baseline remifentanil self-administration, bupropion and dextromethorphan treatment significantly reduced self-administration, whereas in subjects with low baseline remifentanil self-administration, bupropion increased remifentanil self-administration and dextromethorphan had no discernible effect. This finding suggests that combination bupropion-dextromethorphan should be tested in humans, with a focus on treating people with high-level opiate use.
Collapse
|
9
|
Kibaly C, Xu C, Cahill CM, Evans CJ, Law PY. Non-nociceptive roles of opioids in the CNS: opioids' effects on neurogenesis, learning, memory and affect. Nat Rev Neurosci 2019; 20:5-18. [PMID: 30518959 DOI: 10.1038/s41583-018-0092-2] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Mortality due to opioid use has grown to the point where, for the first time in history, opioid-related deaths exceed those caused by car accidents in many states in the United States. Changes in the prescribing of opioids for pain and the illicit use of fentanyl (and derivatives) have contributed to the current epidemic. Less known is the impact of opioids on hippocampal neurogenesis, the functional manipulation of which may improve the deleterious effects of opioid use. We provide new insights into how the dysregulation of neurogenesis by opioids can modify learning and affect, mood and emotions, processes that have been well accepted to motivate addictive behaviours.
Collapse
Affiliation(s)
- Cherkaouia Kibaly
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, Shirley and Stefan Hatos Center for Neuropharmacology, University of California, Los Angeles, CA, USA.
| | - Chi Xu
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Catherine M Cahill
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, Shirley and Stefan Hatos Center for Neuropharmacology, University of California, Los Angeles, CA, USA
| | - Christopher J Evans
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, Shirley and Stefan Hatos Center for Neuropharmacology, University of California, Los Angeles, CA, USA
| | - Ping-Yee Law
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, Shirley and Stefan Hatos Center for Neuropharmacology, University of California, Los Angeles, CA, USA
| |
Collapse
|
10
|
Translational Molecular Approaches in Substance Abuse Research. Handb Exp Pharmacol 2019; 258:31-60. [PMID: 31628598 DOI: 10.1007/164_2019_259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2023]
Abstract
Excessive abuse of psychoactive substances is one of the leading contributors to morbidity and mortality worldwide. In this book chapter, we review translational research strategies that are applied in the pursuit of new and more effective therapeutics for substance use disorder (SUD). The complex, multidimensional nature of psychiatric disorders like SUD presents difficult challenges to investigators. While animal models are critical for outlining the mechanistic relationships between defined behaviors and genetic and/or molecular changes, the heterogeneous pathophysiology of brain diseases is uniquely human, necessitating the use of human studies and translational research schemes. Translational research describes a cross-species approach in which findings from human patient-based data can be used to guide molecular genetic investigations in preclinical animal models in order to delineate the mechanisms of reward circuitry changes in the addicted state. Results from animal studies can then inform clinical investigations toward the development of novel treatments for SUD. Here we describe the strategies that are used to identify and functionally validate genetic variants in the human genome which may contribute to increased risk for SUD, starting from early candidate gene approaches to more recent genome-wide association studies. We will next examine studies aimed at understanding how transcriptional and epigenetic dysregulation in SUD can persistently alter cellular function in the disease state. In our discussion, we then focus on examples from the literature illustrating molecular genetic methodologies that have been applied to studies of different substances of abuse - from alcohol and nicotine to stimulants and opioids - in order to exemplify how these approaches can both delineate the underlying molecular systems driving drug addiction and provide insights into the genetic basis of SUD.
Collapse
|
11
|
Dentate gyrus μ-opioid receptor-mediated neurogenic processes are associated with alterations in morphine self-administration. Sci Rep 2019; 9:1471. [PMID: 30728362 PMCID: PMC6365505 DOI: 10.1038/s41598-018-37083-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 11/30/2018] [Indexed: 02/06/2023] Open
Abstract
Adult hippocampal dentate gyrus (DG) neural stem cells (NSCs) continuously undergo proliferation and differentiation, producing new functional neurons that remodel existing synaptic circuits. Although proliferation of these adult DG NSCs has been implicated in opiate dependence, whether NSC neuronal differentiation and subsequent dendritogenesis are also involved in such addictive behavior remains unknown. Here, we ask whether opiate exposure alters differentiation and dendritogenesis of DG NSCs and investigate the possibility that these alterations contribute to opiate addiction. We show that rat morphine self-administration (MSA), a paradigm that effectively mimics human opiate addiction, increases NSC neuronal differentiation and promotes neuronal dendrite growth in the adult DG. Further, we demonstrate that the μ-opioid receptor (MOR) is expressed on DG NSCs and that MSA leads to a two-fold elevation of endogenous MOR levels in doublecortin expressing (DCX+) NSC progenies in the rat DG. MOR expression is also detected in the cultured rat NSCs and morphine treatment in vitro increases NSC neuronal differentiation and dendritogenesis, suggesting that MOR mediates the effect of morphine on NSC neuronal differentiation and maturation. Finally, we show that conditional overexpression of MOR in DG NSCs under a doxycycline inducible system leads to facilitation of the acquisition of MSA in rats, without affecting the extinction process. We advocate that targeting MOR selectively in the DG NSC population might offer a novel therapeutic intervention for morphine addiction.
Collapse
|
12
|
The new kisspeptin derivative - kissorphin (KSO) - attenuates acute hyperlocomotion and sensitization induced by ethanol and morphine in mice. Alcohol 2017; 64:45-53. [PMID: 28965655 DOI: 10.1016/j.alcohol.2017.04.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Revised: 04/10/2017] [Accepted: 04/24/2017] [Indexed: 12/25/2022]
Abstract
Kissorphin (KSO) is a new peptide derived from kisspeptin-10. This peptide possesses neuropeptide FF (NPFF)-like biological activity in vitro; NPFF, in many cases, inhibits opioid and ethanol effects in rodents. Therefore, the current study explored the influence of KSO on acute ethanol- and morphine-induced hyperactivity, and on the development and expression of locomotor sensitization induced by these drugs. In the present study, sensitization to locomotor effects was induced by repeated exposure to ethanol (2.4 g/kg, intraperitoneally [i.p.], 1 × 4 days) or morphine (10 mg/kg, subcutaneously [s.c.], 1 × 7 days). We found that KSO (1-10 nmol/300 μL, intravenously [i.v.]) did not have an impact on locomotor activity of naïve mice. However, it reduced both acute ethanol- (10 nmol/300 μL) and morphine-induced hyperactivity (3 and 10 nmol/300 μL). Pretreatment of animals with KSO (10 nmol/300 μL), before every ethanol or morphine injection during development of sensitization or before the ethanol or morphine challenge, attenuated the development, as well as the expression of locomotor sensitization to both substances. Moreover, prior administration of the NPFF receptor antagonist RF9 (10 nmol/300 μL, i.v.) inhibited the ability of KSO (10 nmol/300 μL) to reduce the expression of ethanol and morphine sensitization. KSO given alone, at all used doses, did not influence the motor coordination measured via the rotarod test. The results from this study show that KSO effectively attenuated acute and repeated effects of ethanol and morphine. Thus, KSO possesses NPFF-like anti-opioid activity in these behavioral studies.
Collapse
|
13
|
Vázquez López JL, Schild L, Günther T, Schulz S, Neurath H, Becker A. The effects of kratom on restraint-stress-induced analgesia and its mechanisms of action. JOURNAL OF ETHNOPHARMACOLOGY 2017; 205:178-185. [PMID: 28501425 DOI: 10.1016/j.jep.2017.05.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 05/08/2017] [Accepted: 05/09/2017] [Indexed: 06/07/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Mitragyna speciosa and its extracts are called kratom (dried leaves, extract). They contain several alkaloids with an affinity for different opioid receptors. They are used in traditional medicine for the treatment of different diseases, as a substitute by opiate addicts, and to mitigate opioid withdrawal symptoms. Apart from their medical properties, they are used to enhance physical endurance and as a means of overcoming stress. PURPOSE The aim of this study was to determine the mechanisms underlying the effects of kratom on restraint-stress-induced analgesia which occurs during or following exposure to a stressful or fearful stimulus. METHODS To gain further insights into the action of kratom on stress, we conducted experiments using restraint stress as a test system and stress-induced analgesia as a test parameter. Using transgenic mu opioid-receptor (MOR) deficient mice, we studied the involvement of this receptor type. We used nor-binaltorphimine (BNT), an antagonist at kappa opioid receptors (KOR), to study functions of this type of receptor. Membrane potential assay was also employed to measure the intrinsic activity of kratom in comparison to U50,488, a highly selective kappa agonist. RESULTS Treatment with kratom diminished stress-induced analgesia in wildtype and MOR knockout animals. Pretreatment of MOR deficient mice with BNT resulted in similar effects. In comparison to U50,488, kratom exhibited negligible intrinsic activity at KOR alone. CONCLUSIONS The results suggest that the use of kratom as a pharmacological tool to mitigate withdrawal symptoms is related to its action on KOR.
Collapse
Affiliation(s)
- José Luis Vázquez López
- Otto-von-Guericke University, Faculty of Medicine, Institute of Pharmacology and Toxicology, Leipziger Str. 44, 39120 Magdeburg, Germany
| | - Lorenz Schild
- Otto-von-Guericke-University, Faculty of Medicine, Department of Pathobiochemistry, Leipziger Strasse 44, 39120 Magdeburg, Germany
| | - Thomas Günther
- Friedrich Schiller University Jena, Jena University Hospital, Institute of Pharmacology and Toxicology, Drackendorfer Str. 1, 07747 Jena, Germany
| | - Stefan Schulz
- Friedrich Schiller University Jena, Jena University Hospital, Institute of Pharmacology and Toxicology, Drackendorfer Str. 1, 07747 Jena, Germany
| | - Hartmud Neurath
- Center of Pharmacology and Toxicology, Georg August University, Robert-Koch-Str. 40, 37075 Göttingen, Germany
| | - Axel Becker
- Otto-von-Guericke University, Faculty of Medicine, Institute of Pharmacology and Toxicology, Leipziger Str. 44, 39120 Magdeburg, Germany.
| |
Collapse
|
14
|
Genetic dissociation of morphine analgesia from hyperalgesia in mice. Psychopharmacology (Berl) 2017; 234:1891-1900. [PMID: 28343361 PMCID: PMC5520541 DOI: 10.1007/s00213-017-4600-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Accepted: 03/10/2017] [Indexed: 01/10/2023]
Abstract
RATIONALE Morphine is the prototypic mu opioid, producing its analgesic actions through traditional 7 transmembrane domain (7TM) G-protein-coupled receptors generated by the mu opioid receptor gene (Oprm1). However, the Oprm1 gene undergoes extensive alternative splicing to yield three structurally distinct sets of splice variants. In addition to the full-length 7TM receptors, it produces a set of truncated variants comprised of only 6 transmembrane domains (6TM). OBJECTIVES This study explored the relative contributions of 7TM and 6TM variants in a range of morphine actions. METHODS Groups of male and mixed-gender wild-type and exon 11 Oprm1 knockout mice were examined in a series of behavioral assays measuring analgesia, hyperalgesia, respiration, and reward in conditioned place preference assays. RESULTS Loss of the 6TM variants in an exon 11 knockout (E11 KO) mouse did not affect morphine analgesia, reward, or respiratory depression. However, E11 KO mice lacking 6TM variants failed to show morphine-induced hyperalgesia, developed tolerance more slowly than wild-type mice, and did not display hyperlocomotion. CONCLUSIONS Together, our findings confirm the established role of 7TM mu receptor variants in morphine analgesia, reward, and respiratory depression, but reveal an unexpected obligatory role for 6TM variants in morphine-induced hyperalgesia and a modulatory role in morphine tolerance and dependence.
Collapse
|
15
|
Charbogne P, Gardon O, Martín-García E, Keyworth HL, Matsui A, Mechling AE, Bienert T, Nasseef T, Robé A, Moquin L, Darcq E, Ben Hamida S, Robledo P, Matifas A, Befort K, Gavériaux-Ruff C, Harsan LA, Von Everfeldt D, Hennig J, Gratton A, Kitchen I, Bailey A, Alvarez VA, Maldonado R, Kieffer BL. Mu Opioid Receptors in Gamma-Aminobutyric Acidergic Forebrain Neurons Moderate Motivation for Heroin and Palatable Food. Biol Psychiatry 2017; 81:778-788. [PMID: 28185645 PMCID: PMC5386808 DOI: 10.1016/j.biopsych.2016.12.022] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 11/12/2016] [Accepted: 12/12/2016] [Indexed: 12/13/2022]
Abstract
BACKGROUND Mu opioid receptors (MORs) are central to pain control, drug reward, and addictive behaviors, but underlying circuit mechanisms have been poorly explored by genetic approaches. Here we investigate the contribution of MORs expressed in gamma-aminobutyric acidergic forebrain neurons to major biological effects of opiates, and also challenge the canonical disinhibition model of opiate reward. METHODS We used Dlx5/6-mediated recombination to create conditional Oprm1 mice in gamma-aminobutyric acidergic forebrain neurons. We characterized the genetic deletion by histology, electrophysiology, and microdialysis; probed neuronal activation by c-Fos immunohistochemistry and resting-state functional magnetic resonance imaging; and investigated main behavioral responses to opiates, including motivation to obtain heroin and palatable food. RESULTS Mutant mice showed MOR transcript deletion mainly in the striatum. In the ventral tegmental area, local MOR activity was intact, and reduced activity was only observed at the level of striatonigral afferents. Heroin-induced neuronal activation was modified at both sites, and whole-brain functional networks were altered in live animals. Morphine analgesia was not altered, and neither was physical dependence to chronic morphine. In contrast, locomotor effects of heroin were abolished, and heroin-induced catalepsy was increased. Place preference to heroin was not modified, but remarkably, motivation to obtain heroin and palatable food was enhanced in operant self-administration procedures. CONCLUSIONS Our study reveals dissociable MOR functions across mesocorticolimbic networks. Thus, beyond a well-established role in reward processing, operating at the level of local ventral tegmental area neurons, MORs also moderate motivation for appetitive stimuli within forebrain circuits that drive motivated behaviors.
Collapse
Affiliation(s)
- Pauline Charbogne
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS/INSERM/Université de Strasbourg, 1 rue Laurent Fries, 67404 Illkirch, France,Douglas Mental Health Institute, Department of Psychiatry, McGill University, 6875 boulevard LaSalle, H4H 1R3 Montreal, QC, Canada
| | - Olivier Gardon
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS/INSERM/Université de Strasbourg, 1 rue Laurent Fries, 67404 Illkirch, France
| | - Elena Martín-García
- Departament de Ciencies Experimentals i de la Salut, Universitat Pompeu Fabra, PRBB, C/Dr. Aiguader 88, 08003 Barcelona, Spain
| | - Helen L. Keyworth
- Faculty of Health and Medical Sciences, AY Building, University of Surrey, Guildford, Surrey GU2 7XH, UK
| | - Aya Matsui
- Section on Neuronal Structure, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Anna E. Mechling
- Department of Radiology, Medical Physics, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Germany,Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Thomas Bienert
- Department of Radiology, Medical Physics, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Taufiq Nasseef
- Douglas Mental Health Institute, Department of Psychiatry, McGill University, 6875 boulevard LaSalle, H4H 1R3 Montreal, QC, Canada
| | - Anne Robé
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS/INSERM/Université de Strasbourg, 1 rue Laurent Fries, 67404 Illkirch, France
| | - Luc Moquin
- Douglas Mental Health Institute, Department of Psychiatry, McGill University, 6875 boulevard LaSalle, H4H 1R3 Montreal, QC, Canada
| | - Emmanuel Darcq
- Douglas Mental Health Institute, Department of Psychiatry, McGill University, 6875 boulevard LaSalle, H4H 1R3 Montreal, QC, Canada
| | - Sami Ben Hamida
- Douglas Mental Health Institute, Department of Psychiatry, McGill University, 6875 boulevard LaSalle, H4H 1R3 Montreal, QC, Canada
| | - Patricia Robledo
- Departament de Ciencies Experimentals i de la Salut, Universitat Pompeu Fabra, PRBB, C/Dr. Aiguader 88, 08003 Barcelona, Spain
| | - Audrey Matifas
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS/INSERM/Université de Strasbourg, 1 rue Laurent Fries, 67404 Illkirch, France
| | - Katia Befort
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS/INSERM/Université de Strasbourg, 1 rue Laurent Fries, 67404 Illkirch, France
| | - Claire Gavériaux-Ruff
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS/INSERM/Université de Strasbourg, 1 rue Laurent Fries, 67404 Illkirch, France
| | - Laura-Adela Harsan
- Department of Radiology, Medical Physics, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Germany,Laboratory of Engineering, Informatics and Imaging (ICube), Integrative multimodal imaging in healthcare (IMIS), UMR 7357, University of Strasbourg, France,University Hospital Strasbourg, Department of Biophysics and Nuclear Medicine, Strasbourg, France
| | - Dominik Von Everfeldt
- Department of Radiology, Medical Physics, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Jurgen Hennig
- Department of Radiology, Medical Physics, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Alain Gratton
- Douglas Mental Health Institute, Department of Psychiatry, McGill University, 6875 boulevard LaSalle, H4H 1R3 Montreal, QC, Canada
| | - Ian Kitchen
- Faculty of Health and Medical Sciences, AY Building, University of Surrey, Guildford, Surrey GU2 7XH, UK
| | - Alexis Bailey
- Faculty of Health and Medical Sciences, AY Building, University of Surrey, Guildford, Surrey GU2 7XH, UK
| | - Veronica A. Alvarez
- Section on Neuronal Structure, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Rafael Maldonado
- Departament de Ciencies Experimentals i de la Salut, Universitat Pompeu Fabra, PRBB, C/Dr. Aiguader 88, 08003 Barcelona, Spain
| | - Brigitte L. Kieffer
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS/INSERM/Université de Strasbourg, 1 rue Laurent Fries, 67404 Illkirch, France,Douglas Mental Health Institute, Department of Psychiatry, McGill University, 6875 boulevard LaSalle, H4H 1R3 Montreal, QC, Canada,Corresponding author. Douglas Mental Health Institute, Department of Psychiatry, McGill, University, 6875 boulevard LaSalle, H4H 1R3 Montreal, QC, Canada, Phone: 514 761-6131 ext.: 3175; fax: 514 762-3033,
| |
Collapse
|
16
|
Reakkamnuan C, Cheaha D, Kumarnsit E. Nucleus accumbens local field potential power spectrums, phase-amplitude couplings and coherences following morphine treatment. Acta Neurobiol Exp (Wars) 2017. [DOI: 10.21307/ane-2017-055] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
17
|
Muelbl MJ, Nawarawong NN, Clancy PT, Nettesheim CE, Lim YW, Olsen CM. Responses to drugs of abuse and non-drug rewards in leptin deficient ob/ob mice. Psychopharmacology (Berl) 2016; 233:2799-811. [PMID: 27256358 PMCID: PMC5095929 DOI: 10.1007/s00213-016-4323-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 05/09/2016] [Indexed: 12/28/2022]
Abstract
RATIONALE Although leptin receptors are found in hypothalamic nuclei classically associated with homeostatic feeding mechanisms, they are also present in brain regions known to regulate hedonic-based feeding, natural reward processing, and responses to drugs of abuse. The ob/ob mouse is deficient in leptin signaling, and previous work has found altered mesolimbic dopamine signaling and sensitivity to the locomotor activating effects of amphetamine in these mice. OBJECTIVES We directly assessed responses to three drugs of abuse and non-drug rewards in the leptin-deficient ob/ob mouse. METHODS Ob/ob mice were tested in assays of sweet preference, novelty seeking, and drug reward/reinforcement. RESULTS In assays of novelty seeking, novel open field activity and operant sensation seeking were reduced in ob/ob mice, although novel object interaction and novel environment preference were comparable to wild types. We also found that ob/ob mice had specific phenotypes in regard to cocaine: conditioned place preference for 2.5 mg/kg was increased, while the locomotor response to 10 mg/kg was reduced, and cocaine self-administration was the same as wild types. Ob/ob mice also acquired self-administration of the potent opioid remifentanil, but breakpoints for the drug were significantly reduced. Finally, we found significant differences in ethanol drinking in ob/ob mice that correlated negatively with body weight and positively with operant sensation seeking. CONCLUSIONS In conclusion, ob/ob mice displayed task-specific deficits in novelty seeking and dissociable differences in reward/reinforcement associated with cocaine, remifentanil, and ethanol.
Collapse
Affiliation(s)
- Matthew J. Muelbl
- Neuroscience Research Center and Department of Pharmacology & Toxicology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Natalie N. Nawarawong
- Neuroscience Research Center and Department of Pharmacology & Toxicology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Patrick T. Clancy
- Neuroscience Research Center and Department of Pharmacology & Toxicology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Catherine E. Nettesheim
- Neuroscience Research Center and Department of Pharmacology & Toxicology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Yi Wei Lim
- Neuroscience Research Center and Department of Pharmacology & Toxicology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Christopher M. Olsen
- Neuroscience Research Center and Department of Pharmacology & Toxicology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| |
Collapse
|
18
|
Zhang Y, Picetti R, Butelman ER, Ho A, Blendy JA, Kreek MJ. Mouse model of the OPRM1 (A118G) polymorphism: differential heroin self-administration behavior compared with wild-type mice. Neuropsychopharmacology 2015; 40:1091-100. [PMID: 25336208 PMCID: PMC4367451 DOI: 10.1038/npp.2014.286] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Revised: 08/20/2014] [Accepted: 08/26/2014] [Indexed: 12/16/2022]
Abstract
Mu-opioid receptors (MOPRs) are the target of heroin and other prescription opioids, which are currently responsible for massive addiction morbidity in the US. The gene coding for the human MOPR (OPRM1) has an important functional single nucleotide polymorphism (SNP), A118G. The OPRM1 A118G genotype results in substantially increased risk of heroin addiction in humans; however, the neurobiological mechanism for this increased risk is not fully understood. This study examined heroin self-administration (SA) behavior in A112G (G/G) mice, harboring a functionally equivalent SNP in Oprm1 with a similar amino acid substitution, in extended (4 h) SA sessions. Adult male and female G/G mice and 'wild-type' litter mates (A/A) were allowed to self-administer heroin (0.25 mg/kg/unit dose, FR1 with a nose poke response) for 4 h/day, for 10 consecutive days. Half of the mice then continued in a heroin dose-response study, while extinction from heroin SA was studied in the other half. In vivo microdialysis was used to measure acute heroin-induced increases of striatal dopamine in the GG vs AA genotypes. Male and female G/G mice responded for heroin significantly more (and thus had greater intake) than A/A mice, in the initial 10 days of heroin SA, and in the subsequent dose-response study. There were no significant differences in extinction of SA between the A/A and G/G mice. Heroin-induced increases in striatal dopamine levels are higher in the GG mice than in the AA mice. Both male and female G/G mice self-administered more heroin than did A/A mice over a 10-day period, possibly because of the greater increases of heroin-induced striatal dopamine in the GG mice. Furthermore, G/G male mice escalated the amount of heroin self-administration across 10 extended-access sessions more than A/A male mice did. These are the first studies to examine the acquisition of heroin SA in this mouse model. These studies may lead to a better understanding of the neurobiological and behavioral mechanisms that underlie greater risk of heroin addiction in carriers of the A118G SNP.
Collapse
Affiliation(s)
- Yong Zhang
- The Laboratory of the Biology of Addictive Diseases, The Rockefeller University, New York, NY, USA,The Laboratory of the Biology of Addictive Diseases, The Rockefeller University, 1230 York Avenue, Box 171, New York, NY 10065, USA, Tel: +212 327 8490, Fax: +212 327 8574, E-mail:
| | - Roberto Picetti
- The Laboratory of the Biology of Addictive Diseases, The Rockefeller University, New York, NY, USA
| | - Eduardo R Butelman
- The Laboratory of the Biology of Addictive Diseases, The Rockefeller University, New York, NY, USA
| | - Ann Ho
- The Laboratory of the Biology of Addictive Diseases, The Rockefeller University, New York, NY, USA
| | - Julie A Blendy
- Department of Pharmacology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mary Jeanne Kreek
- The Laboratory of the Biology of Addictive Diseases, The Rockefeller University, New York, NY, USA
| |
Collapse
|
19
|
Behavioral and neurochemical characterization of kratom (Mitragyna speciosa) extract. Psychopharmacology (Berl) 2014; 231:13-25. [PMID: 23846544 DOI: 10.1007/s00213-013-3201-y] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2013] [Accepted: 06/24/2013] [Indexed: 10/26/2022]
Abstract
OBJECTIVE Mitragyna speciosa and its extracts are named kratom (dried leaves, extract). It contains several alkaloids and is used in traditional medicine to alleviate musculoskeletal pain, hypertension, coughing, diarrhea, and as an opiate substitute for addicts. Abuse and addiction to kratom is described, and kratom has attracted increasing interest in Western countries. Individual effects of kratom on opioidergic, adrenergic, serotonergic, and dopaminergic receptors are known, but not all of the effects have been explained. Pharmacokinetic and pharmacodynamic data are needed. METHODS The effects of kratom extract on mice behavior were investigated following oral (po), intraperitoneal (ip), and intracerebroventricular (icv) application. Receptor-binding studies were performed. RESULTS In μ opioid receptor knockout mice (-/-) and wild type (+/+) animals, the extract reduced locomotor activity after ip and low po doses in +/+ animals, but not after icv administration. The ip effect was counteracted by 0.3 mg/kg of apomorphine sc, suggesting dopaminergic presynaptic activity. An analgesic effect was only found in -/- mice after icv application. Norbinaltorphimine abolished the analgesic effect, but not the inhibitory effect, on locomotor activity, indicating that the analgesic effect is mediated via κ opioid receptors. Oral doses, which did not diminish locomotor activity, impaired the acquisition of shuttle box avoidance learning. There was no effect on consolidation. Binding studies showed affinity of kratom to μ, δ, and κ opioid receptors and to dopamine D1 receptors. CONCLUSIONS The results obtained in drug-naïve mice demonstrate weak behavioral effects mediated via μ and κ opioid receptors.
Collapse
|
20
|
Charbogne P, Kieffer BL, Befort K. 15 years of genetic approaches in vivo for addiction research: Opioid receptor and peptide gene knockout in mouse models of drug abuse. Neuropharmacology 2013; 76 Pt B:204-17. [PMID: 24035914 DOI: 10.1016/j.neuropharm.2013.08.028] [Citation(s) in RCA: 110] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2013] [Revised: 08/19/2013] [Accepted: 08/23/2013] [Indexed: 12/21/2022]
Abstract
The endogenous opioid system is expressed throughout the brain reinforcement circuitry, and plays a major role in reward processing, mood control and the development of addiction. This neuromodulator system is composed of three receptors, mu, delta and kappa, interacting with a family of opioid peptides derived from POMC (β-endorphin), preproenkephalin (pEnk) and preprodynorphin (pDyn) precursors. Knockout mice targeting each gene of the opioid system have been created almost two decades ago. Extending classical pharmacology, these mutant mice represent unique tools to tease apart the specific role of each opioid receptor and peptide in vivo, and a powerful approach to understand how the opioid system modulates behavioral effects of drugs of abuse. The present review summarizes these studies, with a focus on major drugs of abuse including morphine/heroin, cannabinoids, psychostimulants, nicotine or alcohol. Genetic data, altogether, set the mu receptor as the primary target for morphine and heroin. In addition, this receptor is essential to mediate rewarding properties of non-opioid drugs of abuse, with a demonstrated implication of β-endorphin for cocaine and nicotine. Delta receptor activity reduces levels of anxiety and depressive-like behaviors, and facilitates morphine-context association. pEnk is involved in these processes and delta/pEnk signaling likely regulates alcohol intake. The kappa receptor mainly interacts with pDyn peptides to limit drug reward, and mediate dysphoric effects of cannabinoids and nicotine. Kappa/dynorphin activity also increases sensitivity to cocaine reward under stressful conditions. The opioid system remains a prime candidate to develop successful therapies in addicted individuals, and understanding opioid-mediated processes at systems level, through emerging genetic and imaging technologies, represents the next challenging goal and a promising avenue in addiction research. This article is part of a Special Issue entitled 'NIDA 40th Anniversary Issue'.
Collapse
Affiliation(s)
- Pauline Charbogne
- IGBMC Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS UMR 7104 - Inserm U964, Illkirch F-67404, France; CNRS, UMR7104, Illkirch F-67404, France; UdS Université de Strasbourg, CNRS UMR 7104 - Inserm U964, Illkirch F-67404, France; Inserm U964, Illkirch F-67404, France
| | | | | |
Collapse
|
21
|
Hall FS, Drgonova J, Jain S, Uhl GR. Implications of genome wide association studies for addiction: are our a priori assumptions all wrong? Pharmacol Ther 2013; 140:267-79. [PMID: 23872493 DOI: 10.1016/j.pharmthera.2013.07.006] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2013] [Accepted: 07/11/2013] [Indexed: 11/24/2022]
Abstract
Substantial genetic contributions to addiction vulnerability are supported by data from twin studies, linkage studies, candidate gene association studies and, more recently, Genome Wide Association Studies (GWAS). Parallel to this work, animal studies have attempted to identify the genes that may contribute to responses to addictive drugs and addiction liability, initially focusing upon genes for the targets of the major drugs of abuse. These studies identified genes/proteins that affect responses to drugs of abuse; however, this does not necessarily mean that variation in these genes contributes to the genetic component of addiction liability. One of the major problems with initial linkage and candidate gene studies was an a priori focus on the genes thought to be involved in addiction based upon the known contributions of those proteins to drug actions, making the identification of novel genes unlikely. The GWAS approach is systematic and agnostic to such a priori assumptions. From the numerous GWAS now completed several conclusions may be drawn: (1) addiction is highly polygenic; each allelic variant contributing in a small, additive fashion to addiction vulnerability; (2) unexpected, compared to our a priori assumptions, classes of genes are most important in explaining addiction vulnerability; (3) although substantial genetic heterogeneity exists, there is substantial convergence of GWAS signals on particular genes. This review traces the history of this research; from initial transgenic mouse models based upon candidate gene and linkage studies, through the progression of GWAS for addiction and nicotine cessation, to the current human and transgenic mouse studies post-GWAS.
Collapse
Affiliation(s)
- F Scott Hall
- Molecular Neurobiology Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD 21224, United States.
| | | | | | | |
Collapse
|
22
|
Oddi D, Crusio WE, D'Amato FR, Pietropaolo S. Monogenic mouse models of social dysfunction: implications for autism. Behav Brain Res 2013; 251:75-84. [PMID: 23327738 DOI: 10.1016/j.bbr.2013.01.002] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2012] [Revised: 12/14/2012] [Accepted: 01/05/2013] [Indexed: 12/21/2022]
Abstract
Autism is a pervasive disorder characterized by a complex symptomatology, based principally on social dysfunction. The disorder has a highly complex, largely genetic etiology, involving an impressive variety of genes, the precise contributions of which still remain to be determined. For this reason, a reductionist approach to the study of autism has been proposed, employing monogenic animal models of social dysfunction, either by targeting a candidate gene, or by mimicking a single-gene disorder characterized by autistic symptoms. In the present review, we discuss this monogenic approach by comparing examples of each strategy: the mu opioid receptor knock-out (KO) mouse line, which targets the opioid system (known to be involved in the control of social behaviors); and the Fmr1-KO mouse, a model for Fragile X syndrome (a neurodevelopmental syndrome that includes autistic symptoms). The autistic-relevant behavioral phenotypes of the mu-opioid and Fmr1-KO mouse lines are described here, summarizing previous work by our research group and others, but also providing novel experimental evidence. Relevant factors influencing the validity of the two models, such as sex differences and age at testing, are also addressed, permitting an extensive evaluation of the advantages and limits of monogenic mouse models for autism.
Collapse
Affiliation(s)
- D Oddi
- CNR, Cell Biology and Neurobiology Institute, Rome, Italy; IRCCS, Santa Lucia Foundation, Rome, Italy
| | | | | | | |
Collapse
|
23
|
Yoo JH, Kitchen I, Bailey A. The endogenous opioid system in cocaine addiction: what lessons have opioid peptide and receptor knockout mice taught us? Br J Pharmacol 2012; 166:1993-2014. [PMID: 22428846 DOI: 10.1111/j.1476-5381.2012.01952.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Cocaine addiction has become a major concern in the UK as Britain tops the European 'league table' for cocaine abuse. Despite its devastating health and socio-economic consequences, no effective pharmacotherapy for treating cocaine addiction is available. Identifying neurochemical changes induced by repeated drug exposure is critical not only for understanding the transition from recreational drug use towards compulsive drug abuse but also for the development of novel targets for the treatment of the disease and especially for relapse prevention. This article focuses on the effects of chronic cocaine exposure and withdrawal on each of the endogenous opioid peptides and receptors in rodent models. In addition, we review the studies that utilized opioid peptide or receptor knockout mice in order to identify and/or clarify the role of different components of the opioid system in cocaine-addictive behaviours and in cocaine-induced alterations of brain neurochemistry. The review of these studies indicates a region-specific activation of the µ-opioid receptor system following chronic cocaine exposure, which may contribute towards the rewarding effect of the drug and possibly towards cocaine craving during withdrawal followed by relapse. Cocaine also causes a region-specific activation of the κ-opioid receptor/dynorphin system, which may antagonize the rewarding effect of the drug, and at the same time, contribute to the stress-inducing properties of the drug and the triggering of relapse. These conclusions have important implications for the development of effective pharmacotherapy for the treatment of cocaine addiction and the prevention of relapse.
Collapse
Affiliation(s)
- Ji Hoon Yoo
- Division of Biochemistry, Faculty of Health & Medical Sciences, University of Surrey, Guildford, Surrey, UK
| | | | | |
Collapse
|
24
|
Nagaya D, Ramanathan S, Ravichandran M, Navaratnam V. A118G mu opioid receptor polymorphism among drug addicts in Malaysia. J Integr Neurosci 2012; 11:117-22. [DOI: 10.1142/s0219635212500082] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2011] [Accepted: 01/06/2012] [Indexed: 11/18/2022] Open
|
25
|
Hadjiconstantinou M, Neff NH. Nicotine and endogenous opioids: Neurochemical and pharmacological evidence. Neuropharmacology 2011; 60:1209-20. [DOI: 10.1016/j.neuropharm.2010.11.010] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2010] [Revised: 11/03/2010] [Accepted: 11/11/2010] [Indexed: 10/18/2022]
|
26
|
UMB-3, a novel rabbit monoclonal antibody, for assessing μ-opioid receptor expression in mouse, rat and human formalin-fixed and paraffin-embedded tissues. ACTA ACUST UNITED AC 2010; 167:9-13. [PMID: 20851148 DOI: 10.1016/j.regpep.2010.09.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2010] [Revised: 08/26/2010] [Accepted: 09/13/2010] [Indexed: 11/22/2022]
Abstract
BACKGROUND The immunohistochemical localization of the μ-opioid receptor (MOR, MOP) has been studied in detail in mouse and rat brain using a variety of polyclonal antibodies. However, biochemical analysis of the MOR signaling complex in vivo has been hampered by the lack of suitable monoclonal antibodies for efficient immunoprecipitation of the receptor protein from native sources. Moreover, previous immunohistochemical investigations were restricted to frozen sections from perfusion-fixed rodent brain, largely due to the limited availability of MOR antibodies that effectively stain paraffin-embedded tissues. METHODS Here, we extensively characterized the novel rabbit monoclonal anti-MOR antibody UMB-3 using transfected cells and MOR-deficient mice. UMB-3 was also subjected to a comparative immunohistochemical study of formalin-fixed, paraffin-embedded mouse and rat organ samples as well as human normal and neoplastic tissues. RESULTS Specificity of UMB-3 was demonstrated by detection of a broad band migrating at M(r) 70,000-80,000 in immunoprecipitates from crude brain homogenates of MOR+/+ mice but not of MOR⁻/⁻ mice; cell surface staining of MOR-transfected cells; translocation of MOR receptor immunostaining after agonist exposure; distinct immunostaining of neuronal cell bodies and fibers in MOR-expressing brain regions; absence of staining in MOR-deficient mice; and abolition of tissue immunostaining by preadsorption of UMB-3 with its immunizing peptide. CONCLUSIONS The rabbit monoclonal antibody UMB-3 is an excellent tool for immunoprecipitation of MOR from native sources as well as for immunohistochemical staining of MOR in paraffin-embedded tissue samples of rodent and human origin.
Collapse
|
27
|
Khokhar JY, Ferguson CS, Zhu AZX, Tyndale RF. Pharmacogenetics of drug dependence: role of gene variations in susceptibility and treatment. Annu Rev Pharmacol Toxicol 2010; 50:39-61. [PMID: 20055697 DOI: 10.1146/annurev.pharmtox.010909.105826] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Drug dependency is a highly prevalent mental health disorder that imposes a significant burden on those directly affected, health care systems, and society in general. There is substantial heritability in the susceptibility to drug addiction, which indicates that there are genetic risk factors. Variation in the human genome is abundant and can directly affect drug dependency phenotypes, for example, by altering the function of a gene product or by altering gene expression. Pharmacogenetic studies can assess the effects of genetic variation on the risk for a particular phenotype (e.g., being an alcoholic). In addition, pharmacogenetic variability in treatment efficacy and adverse reactions can be investigated to identify particular genetic variants associated with altered responses. This review highlights examples of genetic variations that are important in the development and maintenance of specific drug dependencies as well as those that affect the response to treatment.
Collapse
Affiliation(s)
- Jibran Y Khokhar
- The Center for Addiction and Mental Health and the Department of Pharmacology and Toxicology, University of Toronto, Ontario, Canada
| | | | | | | |
Collapse
|
28
|
Forebrain PENK and PDYN gene expression levels in three inbred strains of mice and their relationship to genotype-dependent morphine reward sensitivity. Psychopharmacology (Berl) 2010; 208:291-300. [PMID: 19997907 DOI: 10.1007/s00213-009-1730-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2009] [Accepted: 11/10/2009] [Indexed: 12/18/2022]
Abstract
RATIONALE Vulnerability to drug abuse disorders is determined not only by environmental but also by genetic factors. A body of evidence suggests that endogenous opioid peptide systems may influence rewarding effects of addictive substances, and thus, their individual expression levels may contribute to drug abuse liability. OBJECTIVES The aim of our study was to assess whether basal genotype-dependent brain expression of opioid propeptides genes can influence sensitivity to morphine reward. METHODS Experiments were performed on inbred mouse strains C57BL/6J, DBA/2J, and SWR/J, which differ markedly in responses to morphine administration: DBA/2J and SWR/J show low and C57BL/6J high sensitivity to opioid reward. Proenkephalin (PENK) and prodynorphin (PDYN) gene expression was measured by in situ hybridization in brain regions implicated in addiction. The influence of the kappa opioid receptor antagonist nor-binaltorphimine (nor-BNI), which attenuates effects of endogenous PDYN-derived peptides, on rewarding actions of morphine was studied using the conditioned place preference (CPP) paradigm. RESULTS DBA/2J and SWR/J mice showed higher levels of PDYN and lower levels of PENK messenger RNA in the nucleus accumbens than the C57BL/6J strain. Pretreatment with nor-BNI enhanced morphine-induced CPP in the opioid-insensitive DBA/2J and SWR/J strains. CONCLUSIONS Our results demonstrate that inter-strain differences in PENK and PDYN genes expression in the nucleus accumbens parallel sensitivity of the selected mouse strains to rewarding effects of morphine. They suggest that high expression of PDYN may protect against drug abuse by limiting drug-produced reward, which may be due to dynorphin-mediated modulation of dopamine release in the nucleus accumbens.
Collapse
|
29
|
A gene-based method for detecting gene-gene co-association in a case-control association study. Eur J Hum Genet 2009; 18:582-7. [PMID: 20029457 DOI: 10.1038/ejhg.2009.223] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Association study (especially the genome-wide association study) now has a key function in identification and characterization of disease-predisposing genetic variant(s), which customarily involve multiple single nucleotide polymorphisms (SNPs) in a candidate region or across the genome. Case-control association design remains the most popular and a challenging issue in the statistical analysis is the optimal use of all information contained in these SNPs. Previous approaches often treated gene-gene interaction as deviation from additive genetic effects or replaced it with SNP-SNP interaction. However, these approaches are limited for their failure of consideration of gene-gene interaction or gene-gene co-association at gene level. Although the co-association of the SNPs within a candidate gene can be detected by principal component analysis-based logistic regression model, the detection of co-association between genes in genome remains uncertain. Here, we proposed a canonical correlation-based U statistic (CCU) for detecting gene-based gene-gene co-association in the case-control design. We explored its type I error rates and power through simulation and analyzed two real data sets. By treating gene as a functional unit in analysis, we found that CCU was a strong alternative to previous approaches. We discussed the performance of CCU as a gene-based gene-gene co-association statistic and the prospect of further improvement.
Collapse
|
30
|
Le Merrer J, Becker JAJ, Befort K, Kieffer BL. Reward processing by the opioid system in the brain. Physiol Rev 2009; 89:1379-412. [PMID: 19789384 DOI: 10.1152/physrev.00005.2009] [Citation(s) in RCA: 679] [Impact Index Per Article: 45.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The opioid system consists of three receptors, mu, delta, and kappa, which are activated by endogenous opioid peptides processed from three protein precursors, proopiomelanocortin, proenkephalin, and prodynorphin. Opioid receptors are recruited in response to natural rewarding stimuli and drugs of abuse, and both endogenous opioids and their receptors are modified as addiction develops. Mechanisms whereby aberrant activation and modifications of the opioid system contribute to drug craving and relapse remain to be clarified. This review summarizes our present knowledge on brain sites where the endogenous opioid system controls hedonic responses and is modified in response to drugs of abuse in the rodent brain. We review 1) the latest data on the anatomy of the opioid system, 2) the consequences of local intracerebral pharmacological manipulation of the opioid system on reinforced behaviors, 3) the consequences of gene knockout on reinforced behaviors and drug dependence, and 4) the consequences of chronic exposure to drugs of abuse on expression levels of opioid system genes. Future studies will establish key molecular actors of the system and neural sites where opioid peptides and receptors contribute to the onset of addictive disorders. Combined with data from human and nonhuman primate (not reviewed here), research in this extremely active field has implications both for our understanding of the biology of addiction and for therapeutic interventions to treat the disorder.
Collapse
Affiliation(s)
- Julie Le Merrer
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Département Neurobiologie et Génétique, Illkirch, France
| | | | | | | |
Collapse
|
31
|
How to find non-dependent opiate users: a comparison of sampling methods in a field study of opium and heroin users. THE INTERNATIONAL JOURNAL OF DRUG POLICY 2009; 21:215-21. [PMID: 19747812 DOI: 10.1016/j.drugpo.2009.08.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2009] [Revised: 08/13/2009] [Accepted: 08/15/2009] [Indexed: 11/24/2022]
Abstract
BACKGROUND/AIM The first aim is to better understand the potentials and limitations of different sampling methods for reaching a specific, rarely studied population of drug users and for persuading them to take part in a multidisciplinary study. The second is to determine the extent to which these different methods reach similar or dissimilar segments of the non-dependent opiate-using population. METHOD Using ethnographic fieldwork (EFW) and targeted canvassing (TARC; small newspaper advertisements and website announcements), supplemented by snowball referrals, we recruited and interviewed 127 non-dependent opiate users (lifetime prevalence of use 5-100 times; 86.6% had used heroin and 56.7% opium). Average age was 39.0; 66.1% were male and 33.9% female. RESULTS In addition to opiates, many respondents had wide experience with other illicit drugs. The majority had non-conventional lifestyles. Both EFW and TARC yielded only limited numbers of snowball referrals. EFW requires specific skills, is labour-intensive, thus expensive, but allows unsuitable candidates to be excluded faster. Respondents recruited through EFW were significantly more likely to have experience with opium and various drugs other than opiates. TARC resulted in larger percentages of women and respondents with conventional lifestyles. TARC is less labour-intensive but requires more time for screening candidates; its cost-effectiveness depends on the price of advertising for the recruitment. CONCLUSION Different methods reach different segments of the population of non-dependent opiate users. It is useful to employ a multi-method approach to reduce selectivity.
Collapse
|
32
|
Zhang Y, Landthaler M, Schlussman SD, Yuferov V, Ho A, Tuschl T, Kreek MJ. Mu opioid receptor knockdown in the substantia nigra/ventral tegmental area by synthetic small interfering RNA blocks the rewarding and locomotor effects of heroin. Neuroscience 2008; 158:474-83. [PMID: 18938225 DOI: 10.1016/j.neuroscience.2008.09.039] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2008] [Revised: 09/19/2008] [Accepted: 10/13/2008] [Indexed: 10/21/2022]
Abstract
Mu opioid receptors (MOP-r) play an important role in the rewarding and locomotor stimulatory effects of heroin. The aim of the current study was to determine whether infusion of small interfering RNAs (siRNA) targeting MOP-r into the midbrain could knock down MOP-r mRNA and affect heroin-induced locomotor activity or heroin-induced conditioned place preference. Ten-week-old male C57BL/6J mice were surgically implanted bilaterally with guide cannulae directed between the substantia nigra and ventral tegmental area. After 4 days' recovery, mice were infused bilaterally with siRNAs that target the MOP-r (2 mMx0.75 microl/side/day for 3 days) or control siRNA. Seven days after the last infusion, a procedure for conditioned place preference was begun with four heroin (3 mg/kg i.p.) administration sessions alternating with four saline sessions. While heroin induced an increase in locomotor activity in all groups, siRNAs targeting specific regions of MOP-r significantly attenuated this effect. Of particular interest, mice infused with specific siRNAs targeting the MOP-r failed to develop and express conditioned place preference to heroin, or showed a significantly attenuated preference. These alterations in reward-related behaviors are likely due to the reduction in MOP-r mRNA and protein, shown in separate studies by in situ hybridization and autoradiography using the same MOP-r- siRNA infusions. Taken together, these studies demonstrate the utility of siRNA in the neurobiological study of specific components of the reward system and should contribute to the study of other complex behaviors.
Collapse
Affiliation(s)
- Y Zhang
- The Laboratory of the Biology of Addictive Diseases, The Rockefeller University, 1230 York Avenue, Box 171, New York, NY 10065, USA.
| | | | | | | | | | | | | |
Collapse
|
33
|
Befort K, Filliol D, Darcq E, Ghate A, Matifas A, Lardenois A, Muller J, Thibault C, Dembele D, Poch O, Kieffer BL. Gene expression is altered in the lateral hypothalamus upon activation of the mu opioid receptor. Ann N Y Acad Sci 2008; 1129:175-84. [PMID: 18591478 DOI: 10.1196/annals.1417.028] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The lateral hypothalamus (LH) is a brain structure that controls hedonic properties of both natural rewards and drugs of abuse. Mu opioid receptors are known to mediate drug reward, but whether overstimulation of these receptors impacts on LH function has not been studied. Here we have used a genome-wide microarray approach to identify LH responses to chronic mu opioid receptor activation at the transcriptional level. We have subjected wild-type and mu opioid receptor knockout mice to an escalating morphine regimen, which produces severe physical dependence in wild-type but not mutant animals. We have analyzed gene profiles in LH samples using the 430A.2 Affymetrix array and identified a set of 25 genes whose expression is altered by morphine in wild-type mice only. The regulation was confirmed for a subset of these genes using real-time quantitative PCR on samples from independent treatments. Altered expression of aquaporin 4, apolipoprotein D, and prostaglandin synthase is indicative of modified LH physiology. The regulation of two signaling genes (the serum glucocorticoid kinase and the regulator of G protein signaling 4) suggests that neurotransmission is altered in LH circuitry. Finally, the downregulation of apelin may indicate a potential role for this neuropeptide in opioid signaling and hedonic homeostasis. Altogether, our study shows that chronic mu opioid receptor stimulation induces gene expression plasticity in the LH and provides a unique collection of mu opioid receptor-dependent genes that potentially contribute to alter reward processes in addictive diseases.
Collapse
Affiliation(s)
- K Befort
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Département Neurobiologie et Génétique, Illkirch, France.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
David V, Matifas A, Gavello-Baudy S, Decorte L, Kieffer BL, Cazala P. Brain regional Fos expression elicited by the activation of mu- but not delta-opioid receptors of the ventral tegmental area: evidence for an implication of the ventral thalamus in opiate reward. Neuropsychopharmacology 2008; 33:1746-59. [PMID: 17895918 DOI: 10.1038/sj.npp.1301529] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Both mu-opioid receptors (MORs) and delta-opioid receptors (DORs) are expressed in the ventral tegmental area (VTA) and are thought to be involved in the addictive properties of opiates. However, their respective contributions to opiate reward remain unclear. We used intracranial self-administration (ICSA) to study the rewarding effects of morphine microinjections into the VTA of male and female MOR-/- and DOR-/- mice. In brains of mice tested for intra-VTA morphine self-administration, we analyzed regional Fos protein expression to investigate the neural circuitry underlying this behavior. Male and female WT and DOR-/- mice exhibited similar self-administration performances, whereas knockout of the MOR gene abolished intra-VTA morphine self-administration at all doses tested. Naloxone (4 mg/kg) disrupted this behavior in WT and DOR mutants, without triggering physical signs of withdrawal. Morphine ICSA was associated with an increase in Fos within the nucleus accumbens, striatum, limbic cortices, amygdala, hippocampus, the lateral mammillary nucleus (LM), and the ventral posteromedial thalamus (VPM). This latter structure was found to express high levels of Fos exclusively in self-administering WT and DOR-/- mice. Abolition of morphine reward in MOR-/- mice was associated with a decrease in Fos-positive neurons in the mesocorticolimbic dopamine system, amygdala, hippocampus (CA1), LM, and a complete absence within the VPM. We conclude that (i) VTA MORs, but not DORs, are critical for morphine reward and (ii) the role of VTA-thalamic projections in opiate reward deserves to be further explored.
Collapse
MESH Headings
- Analysis of Variance
- Animals
- Behavior, Animal/drug effects
- Brain/anatomy & histology
- Brain/drug effects
- Brain/metabolism
- Cell Count/methods
- Conditioning, Operant/drug effects
- Female
- Gene Expression Regulation/drug effects
- Gene Expression Regulation/physiology
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Morphine/administration & dosage
- Naloxone/pharmacology
- Narcotic Antagonists/pharmacology
- Narcotics/administration & dosage
- Neurons/drug effects
- Neurons/metabolism
- Oncogene Proteins v-fos/genetics
- Oncogene Proteins v-fos/metabolism
- Reaction Time/drug effects
- Reaction Time/physiology
- Receptors, Opioid, delta/deficiency
- Receptors, Opioid, delta/metabolism
- Receptors, Opioid, mu/deficiency
- Receptors, Opioid, mu/metabolism
- Self Administration
- Ventral Tegmental Area/cytology
- Ventral Tegmental Area/drug effects
- Ventral Tegmental Area/metabolism
Collapse
Affiliation(s)
- Vincent David
- Centre de Neurosciences Intégratives et Cognitives, CNRS UMR 5228/Universités de Bordeaux 1 et 2, Talence, France.
| | | | | | | | | | | |
Collapse
|
35
|
Fichna J, Janecka A, Costentin J, Do Rego JC. The endomorphin system and its evolving neurophysiological role. Pharmacol Rev 2007; 59:88-123. [PMID: 17329549 DOI: 10.1124/pr.59.1.3] [Citation(s) in RCA: 180] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Endomorphin-1 (Tyr-Pro-Trp-Phe-NH2) and endomorphin-2 (Tyr-Pro-Phe-Phe-NH2) are two endogenous opioid peptides with high affinity and remarkable selectivity for the mu-opioid receptor. The neuroanatomical distribution of endomorphins reflects their potential endogenous role in many major physiological processes, which include perception of pain, responses related to stress, and complex functions such as reward, arousal, and vigilance, as well as autonomic, cognitive, neuroendocrine, and limbic homeostasis. In this review we discuss the biological effects of endomorphin-1 and endomorphin-2 in relation to their distribution in the central and peripheral nervous systems. We describe the relationship between these two mu-opioid receptor-selective peptides and endogenous neurohormones and neurotransmitters. We also evaluate the role of endomorphins from the physiological point of view and report selectively on the most important findings in their pharmacology.
Collapse
Affiliation(s)
- Jakub Fichna
- Laboratory of Experimental Neuropsychopharmacology, CNRS FRE 2735, IFRMP 23, Faculty of Medicine & Pharmacy, University of Rouen, 22, Boulevard Gambetta, 76183 Rouen cedex, France
| | | | | | | |
Collapse
|
36
|
Sánchez-Cardoso P, Higuera-Matas A, Martín S, del Olmo N, Miguéns M, García-Lecumberri C, Ambrosio E. Modulation of the endogenous opioid system after morphine self-administration and during its extinction: a study in Lewis and Fischer 344 rats. Neuropharmacology 2006; 52:931-48. [PMID: 17161852 DOI: 10.1016/j.neuropharm.2006.10.011] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2006] [Revised: 10/16/2006] [Accepted: 10/18/2006] [Indexed: 11/29/2022]
Abstract
Lewis (LEW) and Fischer 344 (F344) rats show differential morphine self-administration rates. In this study, after animals of both strains self-administered morphine (1mg/kg) or extinguished this behaviour for 3, 7 or 15days, we measured the binding to, and functional state of mu opioid receptors (MORs) as well as proenkephalin (PENK) mRNA content in several brain regions. The results showed that in most brain areas: 1) LEW rats had less binding to MORs in basal conditions than F344 rats; 2) after morphine self-administration, either one of the strains or both (depending on the brain area) showed increased levels of binding to MORs as compared to basal groups; and 3) these binding levels in morphine self-administration animals came down in each extinction group. Moreover, F344 rats exhibited, in general, an increased functionality of MORs after morphine self-administration, as compared to basal groups, which also went down during extinction. Finally, the basal content of PENK mRNA was lower in LEW rats than in F344 rats and it decreased more after self-administration; during extinction, the levels of PENK mRNA got normalized in this strain. This differential modulation of the endogenous opioid system might be related to the different rates of morphine self-administration behavior exhibited by both inbred rat strains.
Collapse
Affiliation(s)
- Pilar Sánchez-Cardoso
- Departamento de Psicobiología, Facultad de Psicología, UNED, Ciudad Universitaria, Madrid 28040, Spain
| | | | | | | | | | | | | |
Collapse
|
37
|
Sahraei H, Barzegari AA, Shams J, Zarrindast MR, Haeri-Rohani A, Ghoshooni H, Sepehri H, Salimi SH. Theophylline inhibits tolerance and sensitization induced by morphine: a conditioned place preference paradigm study in female mice. Behav Pharmacol 2006; 17:621-8. [PMID: 17021395 DOI: 10.1097/01.fbp.0000236274.18042.54] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The effect of theophylline on reward properties of morphine was examined in the present study. A biased conditioned place preference paradigm was used to study the effects of theophylline on the development of conditioned place preference by morphine in sensitized and tolerant female mice. Subcutaneous injection of morphine (0.5-10 mg/kg) induced conditioned place preference in mice, while intraperitoneal administration of theophylline (2.5-100 mg/kg) did not induce conditioned place preference or conditioned place aversion. Theophylline (2.5-100 mg/kg) in combination with morphine (5 mg/kg), during conditioning sessions, decreased the acquisition of morphine conditioned place preference dose independently. Administration of theophylline (2.5-100 mg/kg) before testing also caused a significant reduction of the expression of morphine-induced conditioned place preference in a dose-independent manner. Administration of morphine (12.5, 25 or 50 mg/kg) daily, for 3 days, produced tolerance to conditioned place preference induced by the drug (5 mg/kg). Administration of theophylline (2.5 and 10 mg/kg) 1 h before morphine (12.5, 25 mg/kg), during development of tolerance, abolished morphine tolerance. A higher dose of theophylline (100 mg/kg), however, did not alter morphine tolerance. In addition, theophylline (2.5, 10 and 100 mg/kg) failed to reduce tolerance to a higher dose of morphine (50 mg/kg). Daily administration of morphine (5 mg/kg) for 3 days followed by a 5-day interval caused sensitization to morphine place conditioning. When theophylline was administered (2.5, 10 and 100 mg/kg) 1 h before morphine (5 mg/kg), during development of sensitization, inhibition of morphine-induced sensitization was demonstrated. The effect of theophylline was dose independent. It is concluded that while theophylline has no effect by itself, it reduced both the acquisition and expression of morphine conditioned place preference. In addition, theophylline reduced the acquisition of morphine conditioned place preference in morphine-sensitized and morphine-tolerant mice.
Collapse
Affiliation(s)
- Hedayat Sahraei
- Department of Physiology and Biophysics, Baqiyatallah, University of Medical Sciences, School of Medicine, Baqiyatallah.
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Bart G, LaForge KS, Borg L, Lilly C, Ho A, Kreek MJ. Altered levels of basal cortisol in healthy subjects with a 118G allele in exon 1 of the Mu opioid receptor gene. Neuropsychopharmacology 2006; 31:2313-7. [PMID: 16794569 DOI: 10.1038/sj.npp.1301128] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The mu opioid receptor is centrally involved in the development of the addictive diseases. It also modulates the stress responsive hypothalamic-pituitary-adrenal axis. Receptors encoded by the variant 118G polymorphism in exon 1 of the mu opioid receptor gene have a threefold increase in beta-endorphin binding and beta-endorphin is three times more potent in receptor-mediated activation of G protein-coupled inwardly rectifying potassium channels. Humans with this variant have increased stress response following opioid antagonism. Here, we study basal levels of adrenocorticotropic hormone and cortisol in subjects with this variant. In all, 59 healthy adults were genotyped and had morning levels of adrenocorticotropic hormone and cortisol measured following intravenous administration of saline placebo. Subjects with a 118G allele had significantly greater levels of cortisol than subjects with the prototype gene. Groups did not differ in levels of adrenocorticotropic hormone. A planned comparison revealed significantly greater cortisol in females with at least one copy of the 118G allele compared to females with the prototype gene. There was no significant effect of gender alone, nor was there a significant interaction between gender and genotype, on ACTH or cortisol. Subjects with at least one copy of the 118G allele have increased basal levels of cortisol, which may influence the susceptibility to and treatment of the stress responsive dyscrasia.
Collapse
Affiliation(s)
- Gavin Bart
- The Laboratory of the Biology of Addictive Diseases, The Rockefeller University, New York, NY 10021, USA
| | | | | | | | | | | |
Collapse
|
39
|
Ammon-Treiber S, Stolze D, Schröder H, Loh H, Höllt V. Effects of opioid antagonists and morphine in a hippocampal hypoxia/hypoglycemia model. Neuropharmacology 2005; 49:1160-9. [PMID: 16098996 DOI: 10.1016/j.neuropharm.2005.06.016] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2005] [Revised: 06/24/2005] [Accepted: 06/27/2005] [Indexed: 11/18/2022]
Abstract
The influence of opioid antagonists and of morphine on rat hippocampal slices in a model of reversible hypoxia/hypoglycemia was investigated by assessment of evoked field potentials (population spike amplitude). In control slices, a brief hypoxia/hypoglycemia led to a loss of field potentials followed by an impaired recovery (40-50% of baseline) during reperfusion. In contrast, restoration was significantly improved when the opioid receptor antagonists funaltrexamine (mu) or naltrindole (delta) were administered prior to and during hypoxia/hypoglycemia. In addition, recovery was improved in brain slices derived from mu-opioid receptor-deficient mice as compared to wild-type mice, indicating a deleterious role of endogenous opioids in hypoxia/hypoglycemia. Exogenous opiate exposure with morphine (0.1, 1.0, 10 microM) prior to hypoxia/hypoglycemia caused a slight concentration dependent increase of evoked field potentials. When morphine exposure was terminated after 1h and immediately followed by hypoxia/hypoglycemia, an impaired recovery of population spike amplitude was obtained, dependent on morphine concentration during preincubation. These results demonstrate that morphine aggravates neurotoxic effects of hypoxia/hypoglycemia. Conversely, when onset of hypoxia/hypoglycemia was delayed for 3h after morphine termination, a significantly improved recovery was observed. Similarly, in vivo administration of morphine 12h prior to slice preparation resulted in a dose dependent improved recovery of field potentials after hypoxia/hypoglycemia. These results provide evidence that preconditioning with morphine is able to induce neuroprotective effects.
Collapse
Affiliation(s)
- S Ammon-Treiber
- Institute of Pharmacology and Toxicology, Otto-von-Guericke University Magdeburg, Germany
| | | | | | | | | |
Collapse
|
40
|
Szumlinski KK, Lominac KD, Frys KA, Middaugh LD. Genetic variation in heroin-induced changes in behaviour: effects of B6 strain dose on conditioned reward and locomotor sensitization in 129-B6 hybrid mice. GENES BRAIN AND BEHAVIOR 2005; 4:324-36. [PMID: 16011579 DOI: 10.1111/j.1601-183x.2004.00111.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Substantial interindividual variability exists in the propensity to develop opiate addiction. Genetic variation in opiate reward may contribute to this variability. A large body of evidence indicates genetic variation in mice for several effects of opiate drugs. The present study examined heroin-induced place conditioning and locomotor sensitization in the two strains of mice employed most frequently in the generation of transgenic animals, C57BL/6J (B6) and 129X1/sVJ (129), as well as in groups of B6-129 hybrid mice, differing in their amount of B6 genetic background. Four pairings of 100 microg/kg of heroin elicited robust place conditioning and locomotor sensitization in B6 controls and in N(10) congenic B6-129 hybrid mice. In comparison, the identical treatment produced no locomotor sensitization and induced place aversion in 129 controls. No heroin-induced changes in the behaviour of N(3) congenic B6-129 hybrid mice or F5-8 non-congenic B6-129 hybrid mice were observed. The expression of place conditioning was not facilitated in any group by the administration of a heroin-priming injection prior to testing. These data indicate that genetic variation exists in mice for the rewarding and locomotor-sensitizing effects of heroin and that the capacity of heroin to induce conditioned reward and locomotor sensitization can be modulated in a B6 strain dose-dependent manner in B6-129 hybrid mice. Thus, strain differences in heroin responsiveness should be considered when examining transgenic lines on B6-129 backgrounds for opiate-induced changes in behaviour that may be relevant for addiction.
Collapse
Affiliation(s)
- K K Szumlinski
- Department of Physiology and Neuroscience, and Department of Psychiatry and Behavioral Sciences, Center for Drug and Alcohol Programs, Medical University of South Carolina, Charleston, SC, USA.
| | | | | | | |
Collapse
|
41
|
Mathon DS, Lesscher HMB, Gerrits MAFM, Kamal A, Pintar JE, Schuller AGP, Spruijt BM, Burbach JPH, Smidt MP, van Ree JM, Ramakers GMJ. Increased gabaergic input to ventral tegmental area dopaminergic neurons associated with decreased cocaine reinforcement in mu-opioid receptor knockout mice. Neuroscience 2005; 130:359-67. [PMID: 15664692 DOI: 10.1016/j.neuroscience.2004.10.002] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/01/2004] [Indexed: 11/18/2022]
Abstract
There is general agreement that dopaminergic neurons projecting from the ventral tegmental area (VTA) to the nucleus accumbens and prefrontal cortex play a key role in drug reinforcement. The activity of these neurons is strongly modulated by the inhibitory and excitatory input they receive. Activation of mu-opioid receptors, located on GABAergic neurons in the VTA, causes hyperpolarization of these GABAergic neurons, thereby causing a disinhibition of VTA dopaminergic neurons. This effect of mu-opioid receptors upon GABA neurotransmission is a likely mechanism for mu-opioid receptor modulation of drug reinforcement. We studied mu-opioid receptor signaling in relation to cocaine reinforcement in wild-type and mu-opioid receptor knockout mice using a cocaine self-administration paradigm and in vitro electrophysiology. Cocaine self-administration was reduced in mu-opioid receptor knockout mice, suggesting a critical role of mu-opioid receptors in cocaine reinforcement. The frequency of spontaneous inhibitory post-synaptic currents onto dopaminergic neurons in the ventral tegmental area was increased in mu-opioid receptor knockout mice compared with wild-type controls, while the frequency of spontaneous excitatory post-synaptic currents was unaltered. The reduced cocaine self-administration and increased GABAergic input to VTA dopaminergic neurons in mu-opioid receptor knockout mice supports the notion that suppression of GABAergic input onto dopaminergic neurons in the VTA contributes to mu-opioid receptor modulation of cocaine reinforcement.
Collapse
Affiliation(s)
- D S Mathon
- Rudolf Magnus Institute of Neuroscience, Department of Pharmacology and Anatomy, University Medical Centre Utrecht, Stratenum, Universiteitsweg 100, 3584 CG Utrecht, The Netherlands
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Ferraro TN, Golden GT, Smith GG, Martin JF, Schwebel CL, Doyle GA, Buono RJ, Berrettini WH. Confirmation of a major QTL influencing oral morphine intake in C57 and DBA mice using reciprocal congenic strains. Neuropsychopharmacology 2005; 30:742-6. [PMID: 15508023 DOI: 10.1038/sj.npp.1300592] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
C57BL/6 (B6) and DBA/2 (D2) mice exhibit disparate behavior when tested for voluntary morphine intake in a two-bottle choice drinking paradigm with B6 mice consuming 10 times more drug than D2 mice. Previous genetic mapping studies identified a locus, Mop2, on the proximal part of chromosome 10 that explained over half of the genetic variance in this mouse model of opioid self-administration. We constructed a set of reciprocal congenic strains between B6 and D2 mice in which the proximal portion of chromosome 10 has been introgressed from one strain onto the background of the other. We tested mice from this pair of reciprocal strains together with progenitor B6 and D2 mice in a two-bottle choice drinking paradigm with morphine and quinine. The results showed that introgression of chromosome 10 alleles from the B6 strain onto a D2 genetic background increased voluntary morphine intake four-fold compared to progenitor D2 mice. Preference for morphine was also increased significantly in D2.B6-Mop2 mice compared to progenitor D2 mice. Conversely, introgression of chromosome 10 alleles from the D2 strain onto a B6 genetic background decreased morphine intake by half compared to progenitor B6 mice in B6.D2 -Mop2 mice; however, high morphine preference was maintained in this congenic strain most likely due to strong quinine aversion. When quinine was eliminated from the control bottle, morphine preference in B6.D2-Mop2 mice was decreased significantly relative to B6 and D2.B6-Mop2 mice. Overall, these data confirm the existence of a gene(s) on chromosome 10 proximal to D10Mit124 that has a strong influence on the difference in morphine drinking behavior between B6 and D2 mice.
Collapse
Affiliation(s)
- Thomas N Ferraro
- Department of Psychiatry, Center for Neurobiology and Behavior, University of Pennsylvania, Philadelphia, PA 19104-6140, USA.
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Kreek MJ, Bart G, Lilly C, LaForge KS, Nielsen DA. Pharmacogenetics and Human Molecular Genetics of Opiate and Cocaine Addictions and Their Treatments. Pharmacol Rev 2005; 57:1-26. [PMID: 15734726 DOI: 10.1124/pr.57.1.1] [Citation(s) in RCA: 238] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Opiate and cocaine addictions are major social and medical problems that impose a significant burden on society. Despite the size and scope of these problems, there are few effective treatments for these addictions. Methadone maintenance is an effective and most widely used treatment for opiate addiction, allowing normalization of many physiological abnormalities caused by chronic use of short-acting opiates. There are no pharmacological treatments for cocaine addiction. Epidemiological, linkage, and association studies have demonstrated a significant contribution of genetic factors to the addictive diseases. This article reviews the molecular genetics and pharmacogenetics of opiate and cocaine addictions, focusing primarily on genes of the opioid and monoaminergic systems that have been associated with or have evidence for linkage to opiate or cocaine addiction. This evidence has been marshalled either through identification of variant alleles that lead to functional alterations of gene products, altered gene expression, or findings of linkage or association studies. Studies of polymorphisms in the mu opioid receptor gene, which encodes the receptor target of some endogenous opioids, heroin, morphine, and synthetic opioids, have contributed substantially to knowledge of genetic influences on opiate and cocaine addiction. Other genes of the endogenous opioid and monoaminergic systems, particularly genes encoding dopamine beta-hydroxylase, and the dopamine, serotonin, and norepinephrine transporters have also been implicated. Variants in genes encoding proteins involved in metabolism or biotransformation of drugs of abuse and also of treatment agents are reviewed.
Collapse
Affiliation(s)
- Mary Jeanne Kreek
- The Laboratory of the Biology of Addictive Diseases, The Rockefeller University, Box 171, 1230 York Avenue, New York, New York 10021, USA.
| | | | | | | | | |
Collapse
|
44
|
Abstract
Mu opioid receptors mediate positive reinforcement following direct (morphine) or indirect (alcohol, cannabinoids, nicotine) activation, and our understanding of mu receptor function is central to the development of addiction therapies. Recent data obtained in native neurons confirm that mu receptor signaling and regulation are strongly agonist-dependent. Current functional mapping reveals morphine-activated neurons in the extended amygdala and early genomic approaches have identified novel mu receptor-associated proteins. A classification of about 30 genes either promoting or counteracting the addictive properties of morphine is proposed from the analysis of knockout mice data. The targeting of effectors or regulatory proteins, beyond the mu receptor itself, might provide valuable strategies to treat addictive disorders.
Collapse
Affiliation(s)
- Candice Contet
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS/INSERM/ULP, UMR7104, Parc d'Innovation, 1 rue Laurent Fries BP 10142, 67404 Illkirch Cedex, Strasbourg, France
| | | | | |
Collapse
|
45
|
Kreek MJ, Nielsen DA, LaForge KS. Genes associated with addiction: alcoholism, opiate, and cocaine addiction. Neuromolecular Med 2004; 5:85-108. [PMID: 15001815 DOI: 10.1385/nmm:5:1:085] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2003] [Accepted: 08/15/2003] [Indexed: 11/11/2022]
Abstract
Drug addiction is a complex disorder that has a large spectrum of causes. Vulnerability to addiction has been shown in twin studies to have a robust genetic component. This genetic basis for addiction has general and specific components for each drug abused. Although many genes have been implicated in drug addiction, only a handful have either been replicated to have an association or to have an identified functional mechanism related to specific effects of abused drugs. A few selected genetic variants that currently look promising for the study of alcohol, opiate, and cocaine addiction are discussed in this article.
Collapse
Affiliation(s)
- Mary Jeanne Kreek
- Laboratory of the Biology of Addictive Diseases, The Rockefeller University, 1230 York Avenue, New York, NY 10021, USA.
| | | | | |
Collapse
|
46
|
Bart G, Heilig M, LaForge KS, Pollak L, Leal SM, Ott J, Kreek MJ. Substantial attributable risk related to a functional mu-opioid receptor gene polymorphism in association with heroin addiction in central Sweden. Mol Psychiatry 2004; 9:547-9. [PMID: 15037869 PMCID: PMC6141020 DOI: 10.1038/sj.mp.4001504] [Citation(s) in RCA: 127] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- G Bart
- The Laboratory of the Biology of Addictive Diseases, The Rockefeller University, New York, NY, USA
| | - M Heilig
- Division of Psychiatry, NEUROTEC, Karolinska Institute, M57 Huddinge University Hospital, Stockholm, Sweden
| | - KS LaForge
- The Laboratory of the Biology of Addictive Diseases, The Rockefeller University, New York, NY, USA
| | - L Pollak
- Division of Psychiatry, NEUROTEC, Karolinska Institute, M57 Huddinge University Hospital, Stockholm, Sweden
| | - SM Leal
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - J Ott
- The Laboratory of Statistical Genetics, The Rockefeller University, New York, NY, USA
| | - MJ Kreek
- The Laboratory of the Biology of Addictive Diseases, The Rockefeller University, New York, NY, USA
- Division of Psychiatry, NEUROTEC, Karolinska Institute, M57 Huddinge University Hospital, Stockholm, Sweden
| |
Collapse
|
47
|
Hall FS, Goeb M, Li XF, Sora I, Uhl GR. mu-Opioid receptor knockout mice display reduced cocaine conditioned place preference but enhanced sensitization of cocaine-induced locomotion. ACTA ACUST UNITED AC 2004; 121:123-30. [PMID: 14969743 DOI: 10.1016/j.molbrainres.2003.10.024] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/21/2003] [Indexed: 01/04/2023]
Abstract
The mu-opioid receptor (OPRM1) is expressed in brain regions implicated in reward and locomotor processes. Reduced reward, not only from opiates, but also from several other abused substances has been observed in mice with lifelong deletions of the OPRM1 gene. To further define the roles of mu-opioid receptors in psychostimulant actions, cocaine psychomotor stimulant and rewarding effects were examined in wild-type (WT), heterozygous and homozygous mu-opioid receptor knockout mice. While mu-opioid receptor knockout did not affect basal locomotion, locomotor stimulant effects of cocaine were enhanced in a within-subjects dose-response experiment. However, further study revealed that in mice injected with 20 mg/kg for the first time, there was no difference in the locomotor-stimulating effects of cocaine between knockout and wild-type mice. In a sensitization study (modeled after the conditions in the dose-response experiment) although not observed in WT mice, OPRM1-/- mice did exhibit cocaine sensitization. By stark contrast, and similar to the effects of other rewarding drugs in OPRM1 KO mice, cocaine reward, as assessed by conditioned place preference, was reduced in both homozygous and heterozygous OPRM1 KO mice. The present results confirm a central role of the mu-opioid receptor in drug reward but opposing effects on locomotor sensitization. The reduced cocaine reward identified in heterozygous mu-opioid receptor knockout mice supports the possibility that humans with fewer available mu-opioid receptors might experience less cocaine reward.
Collapse
MESH Headings
- Analysis of Variance
- Anesthetics, Local/pharmacology
- Animals
- Behavior, Animal
- Cocaine/pharmacology
- Conditioning, Operant/drug effects
- Dose-Response Relationship, Drug
- Heterozygote
- Homozygote
- Locomotion/drug effects
- Locomotion/genetics
- Mice
- Mice, Knockout
- Receptors, Opioid, mu/deficiency
- Receptors, Opioid, mu/genetics
- Receptors, Opioid, mu/metabolism
Collapse
Affiliation(s)
- F Scott Hall
- Molecular Neurobiology Branch, National Institute on Drug Abuse-IRP, NIH/DHHS, 5500 Nathan Shock Drive, Baltimore, MD 21224, USA.
| | | | | | | | | |
Collapse
|
48
|
Abstract
The discovery of endogenous opioids has markedly influenced the research on the biology of drug dependence. Evidence has been presented that these brain substances are self-administered by laboratory animals. This finding, among others, has led to the hypothesis that endogenous opioids are involved in reinforcing habits, including dependence on drugs of abuse. The course of drug dependence is presented as a continuum from no drug use via controlled use to an actual dependence on the drug. Specific brain opioid systems belonging to four conceptualized brain circuits are described to be involved during the different phases of the drug dependence continuum. More recent research to delineate the role of endogenous opioid systems in drug dependence has focussed on genetic research in humans and animals. Among others, the findings obtained from studies of opioid receptor and opioid peptide precursor knockout mice provided further support for a role of endogenous opioid systems in drug dependence, in agreement with previous pharmacological studies.
Collapse
Affiliation(s)
- Mirjam A F M Gerrits
- Rudolf Magnus Institute of Neuroscience, Department of Pharmacology and Anatomy, University Medical Center Utrecht, Universiteitsweg 100, 3584 CG Utrecht, The Netherlands.
| | | | | |
Collapse
|
49
|
Compan V, Scearce-Levie K, Crosson C, Daszuta A, Hen R. Enkephalin contributes to the locomotor stimulating effects of 3,4-methylenedioxy-N-methylamphetamine. Eur J Neurosci 2003; 18:383-90. [PMID: 12887420 DOI: 10.1046/j.1460-9568.2003.02767.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
3,4-methylenedioxy-N-methylamphetamine (MDMA, 'Ecstasy') is a potent inhibitor of serotonin uptake, which induces both an increase in locomotion and a decrease in exploratory activity in rodents. Serotonin 5-HT1B receptors, located on the terminals of striatal efferent neurons, have been suggested to mediate these motor effects of MDMA. Striatal neurons projecting to the globus pallidus contain met-enkephalin, whilst those projecting to the substantia nigra contain substance P. We therefore analysed the levels of both peptides using radioimmunocytochemistry after MDMA administration (10 mg/kg, 3 h) in wild-type and 5-HT1B receptor knockout mice. Our results demonstrate that MDMA induces a decrease in pallidal met-enkephalin immunolabelling in wild-type, but not in 5-HT1B receptor knockout mice. Similar results were obtained following treatment with the 5-HT1A/1B agonist RU24969 (5 mg/kg, 3 h), suggesting that activation of 5-HT1B receptors leads to a reduction in met-enkephalin levels in the globus pallidus. In contrast, MDMA had no effect on the nigral substance P levels. We have previously shown that both MDMA and RU24969 fail to stimulate locomotor activity in 5-HT1B receptor knockout mice. Our present data indicate that the opioid antagonist naloxone suppressed the locomotor effects of MDMA. This study is the first to demonstrate that Enk contributes to MDMA-induced increases in locomotor activity. Such an effect may be related to the 5-HT control of pallidal met-enkephalin levels via the 5-HT1B receptors.
Collapse
Affiliation(s)
- V Compan
- Columbia University, Center for Neurobiology and Behaviour, N.Y.S.P.I. Kolb Research Annex, Room 732, 1051 Riverside Drive, Unit 87, New York, NY 10032-2695, USA.
| | | | | | | | | |
Collapse
|
50
|
Yoo JH, Yang EM, Lee SY, Loh HH, Ho IK, Jang CG. Differential effects of morphine and cocaine on locomotor activity and sensitization in mu-opioid receptor knockout mice. Neurosci Lett 2003; 344:37-40. [PMID: 12781916 DOI: 10.1016/s0304-3940(03)00410-5] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
The present study was undertaken to investigate the hypothesis that the mu-opioid receptors play a crucial role in locomotor activity and sensitization to cocaine and morphine in wild-type and mu-opioid receptor knockout mice. Our results show that morphine and cocaine increased locomotor activity in wild-type mice, but failed to increase locomotor activity in mu-opioid receptor knockout mice, suggesting a contribution of mu-opioid receptor. Repeated morphine treatment induced sensitization in wild-type mice, but this was not observed in mu-opioid receptor knockout mice. In contrast repeated cocaine treatment produced sensitization in mu-opioid receptor knockout mice, but not in wild-type mice on day 6. However, the sensitization to cocaine was observed in mu-opioid receptor knockout and wild-type mice on day 12. These results suggest that the expression of mu-opioid receptor may contribute to locomotor sensitization induced by morphine, but that mu-opioid receptor does not play an important role in mediating sensitization to cocaine.
Collapse
Affiliation(s)
- Ji-Hoon Yoo
- Department of Pharmacology, College of Pharmacy, Sungkyunkwan University, Cheoncheon-dong, Jangan-ku, Suwon, 440-746, South Korea
| | | | | | | | | | | |
Collapse
|