1
|
Desmercieres S, Lardeux V, Longueville JE, Dugast E, Thiriet N, Solinas M. Effects of Highly Palatable Diet on motivation for food and resistance to punishment in rats: Role of sex and age of exposure. Appetite 2024; 198:107340. [PMID: 38582135 DOI: 10.1016/j.appet.2024.107340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Revised: 03/05/2024] [Accepted: 04/03/2024] [Indexed: 04/08/2024]
Abstract
Exposure to highly palatable food is believed to induce behavioral and neurobiological changes that may produce addiction-like behavior and increase the risks of obesity and overweight. Studies in rodents have led to conflicting results suggesting that several factors such as sex and age of exposure contribute to the development of maladaptive behaviors towards food. In addition, it is not clear whether effects of exposure to highly palatable diets (HPD) persist after their discontinuation, which would indicate long-term risks to develop addiction-like behavior. In this study, we investigated the persistent effects of an intermittent 8-week exposure to HPD in male and female rats as a function of age of exposure (adult and adolescent). We found that intermittent exposure to HPD did not alter body weight, but it affected consumption of standard food during the time of exposure in all groups. In addition, in adults, HPD produced a decrease in the initial baseline responding in FR1 schedules, an effect that persisted for 4 weeks in males but not in female rats. However, we found that exposure to HPD did not affect resistance to punishment measured by progressive shock strength break points or motivation for food as measured by progressive-ratio break points regardless of sex or age of exposure. Altogether, these results do not provide support for the hypothesis that intermittent exposure to HPD produce persistent increases in the vulnerability to develop addiction-like behaviors towards palatable food.
Collapse
Affiliation(s)
- Stevenson Desmercieres
- Université de Poitiers, INSERM, U-1084, Laboratoire des Neurosciences Expérimentales et Cliniques, Poitiers, France
| | - Virginie Lardeux
- Université de Poitiers, INSERM, U-1084, Laboratoire des Neurosciences Expérimentales et Cliniques, Poitiers, France
| | - Jean-Emmanuel Longueville
- Université de Poitiers, INSERM, U-1084, Laboratoire des Neurosciences Expérimentales et Cliniques, Poitiers, France
| | - Emilie Dugast
- Université de Poitiers, INSERM, U-1084, Laboratoire des Neurosciences Expérimentales et Cliniques, Poitiers, France; CHU de Poitiers, Poitiers, France
| | - Nathalie Thiriet
- Université de Poitiers, INSERM, U-1084, Laboratoire des Neurosciences Expérimentales et Cliniques, Poitiers, France
| | - Marcello Solinas
- Université de Poitiers, INSERM, U-1084, Laboratoire des Neurosciences Expérimentales et Cliniques, Poitiers, France.
| |
Collapse
|
2
|
Solinas M, Lardeux V, Leblanc PM, Longueville JE, Thiriet N, Vandaele Y, Panlilio LV, Jaafari N. Delay of punishment highlights differential vulnerability to developing addiction-like behavior toward sweet food. Transl Psychiatry 2024; 14:155. [PMID: 38509086 PMCID: PMC10954751 DOI: 10.1038/s41398-024-02863-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 03/06/2024] [Accepted: 03/07/2024] [Indexed: 03/22/2024] Open
Abstract
Resistance to punishment is commonly used to measure the difficulty in refraining from rewarding activities when negative consequences ensue, which is a hallmark of addictive behavior. We recently developed a progressive shock strength (PSS) procedure in which individual rats can titrate the amount of punishment that they are willing to tolerate to obtain food rewards. Here, we investigated the effects of a range of delays (0-12 s) on resistance to punishment measured by PSS break points. As expected from delay discounting principles, we found that delayed shock was less effective as a punisher, as revealed by higher PSS breakpoints. However, this discounting effect was not equally distributed in the population of rats, and the introduction of a delay highlighted the existence of two populations: rats that were sensitive to immediate punishment were also sensitive to delayed shock, whereas rats that were resistant to immediate punishment showed strong temporal discounting of delayed punishment. Importantly, shock-sensitive rats suppressed responding even in subsequent non-punishment sessions, and they differed from shock-resistant rats in anxiety-like behavior, but not in sensitivity to pain. These results show that manipulation of temporal contingencies of punishment in the PSS procedure provides a valuable tool to identify individuals with a double vulnerability to addiction: low sensitivity to aversion and excessive discounting of negative future consequences. Conversely, the shock-sensitive population may provide a model of humans who are vulnerable to opportunity loss due to excessive anxiety.
Collapse
Affiliation(s)
- Marcello Solinas
- Université de Poitiers, INSERM, U1084, Laboratoire de Neurosciences Expérimentales et Cliniques, Poitiers, France.
- Unité de Recherche Clinique Intersectorielle en Psychiatrie, Centre Hospitalier Henri-Laborit, Poitiers, France.
| | - Virginie Lardeux
- Université de Poitiers, INSERM, U1084, Laboratoire de Neurosciences Expérimentales et Cliniques, Poitiers, France
| | - Pierre-Marie Leblanc
- Unité de Recherche Clinique Intersectorielle en Psychiatrie, Centre Hospitalier Henri-Laborit, Poitiers, France
| | - Jean-Emmanuel Longueville
- Université de Poitiers, INSERM, U1084, Laboratoire de Neurosciences Expérimentales et Cliniques, Poitiers, France
| | - Nathalie Thiriet
- Université de Poitiers, INSERM, U1084, Laboratoire de Neurosciences Expérimentales et Cliniques, Poitiers, France
| | - Youna Vandaele
- Université de Poitiers, INSERM, U1084, Laboratoire de Neurosciences Expérimentales et Cliniques, Poitiers, France
| | - Leigh V Panlilio
- Real-world Assessment, Prediction, and Treatment Unit, Translational Addiction Medicine Branch, Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD, USA
| | - Nematollah Jaafari
- Unité de Recherche Clinique Intersectorielle en Psychiatrie, Centre Hospitalier Henri-Laborit, Poitiers, France
- Université de Poitiers, CNRS, UMR 7295, Centre de Recherche sur la Cognition et l'apprentissage, Poitiers, France
| |
Collapse
|
3
|
Gaborit M, Massotte D. Therapeutic potential of opioid receptor heteromers in chronic pain and associated comorbidities. Br J Pharmacol 2023; 180:994-1013. [PMID: 34883528 DOI: 10.1111/bph.15772] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 06/07/2021] [Accepted: 07/21/2021] [Indexed: 11/27/2022] Open
Abstract
Chronic pain affects 20% to 45% of the global population and is often associated with the development of anxio-depressive disorders. Treatment of this debilitating condition remains particularly challenging with opioids prescribed to alleviate moderate to severe pain. However, despite strong antinociceptive properties, numerous adverse effects limit opioid use in the clinic. Moreover, opioid misuse and abuse have become a major health concern worldwide. This prompted efforts to design original strategies that would efficiently and safely relieve pain. Targeting of opioid receptor heteromers is one of these. This review summarizes our current knowledge on the role of heteromers involving opioid receptors in the context of chronic pain and anxio-depressive comorbidities. It also examines how heteromerization in native tissue affects ligand binding, receptor signalling and trafficking properties. Finally, the therapeutic potential of ligands designed to specifically target opioid receptor heteromers is considered. LINKED ARTICLES: This article is part of a themed issue on Advances in Opioid Pharmacology at the Time of the Opioid Epidemic. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v180.7/issuetoc.
Collapse
Affiliation(s)
- Marion Gaborit
- Centre National de la Recherche Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives, Strasbourg, France
| | - Dominique Massotte
- Centre National de la Recherche Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives, Strasbourg, France
| |
Collapse
|
4
|
Hersey M, Bacon AK, Bailey LG, Lee MR, Chen AY, Leggio L, Tanda G. Oxytocin receptors mediate oxytocin potentiation of methylphenidate-induced stimulation of accumbens dopamine in rats. J Neurochem 2023; 164:613-623. [PMID: 36420597 PMCID: PMC10766115 DOI: 10.1111/jnc.15730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 11/06/2022] [Accepted: 11/07/2022] [Indexed: 11/25/2022]
Abstract
While the illicit use and misuse of stimulants like cocaine and methylphenidate (MP) has increased, there remains no FDA-approved treatments for psychostimulant use disorders (PSUD). Oxytocin (OT) has shown promise as a potential pharmacotherapy for PSUD. Dopamine (DA) neurotransmission plays a significant role in PSUD. We have recently shown that OT blunts the reinforcing effects of MP but, surprisingly, enhanced MP-induced stimulation of DA levels. Such effects have been suggested as a result of activation of OT receptors or, alternatively, could be mediated by direct actions of OT on MP blockade of the DA transporter. Here, we employed fast scan cyclic voltammetry (FSCV) to investigate the effects of systemic OT on MP-induced changes in the dynamics of DA, phasic release and uptake, in the nucleus accumbens shell (NAS) of Sprague-Dawley rats. We also tested the systemic effects of an antagonist of OT receptors, atosiban, to counteract the OT enhancement of dopaminergic effects of MP under microdialysis procedures in the NAS in rats. Administration of OT alone (2 mg/kg; i.p.) did not significantly modify evoked NAS DA dynamics measured by FSCV, and when administered 10 min before MP (0.1, 0.3, 1.0 mg/kg; i.v.), OT did not potentiate MP-induced increases in phasic DA release and did not alter DA clearance rate, suggesting no direct interactions of OT with the MP-induced blockade of DA uptake. Also, OT alone did not elicit significant changes in tonic, extracellular NAS DA levels measured by microdialysis. However, consistent with previous studies, we observed that OT pretreatments (2 mg/kg; i.p.) potentiated MP-induced (0.1, 0.3, 1.0 mg/kg; i.v.) efflux of extracellular NAS DA levels. This effect was abolished when rats were pretreated with atosiban (2 mg/kg; i.p.), suggesting that OT receptors mediate this OT action. Overall, our results suggest that OT receptors mediated OT potentiation of MP-induced stimulation of extracellular NAS DA levels, likely driven by modulation of DA receptor signaling pathways, without affecting MP blockade of DAT.
Collapse
Affiliation(s)
| | | | | | - Mary R. Lee
- Veterans Affairs Medical Center, Washington, DC
| | - Andy Y. Chen
- Medication Development Program, NIDA IRP, Baltimore, MD
| | - Lorenzo Leggio
- Medication Development Program, NIDA IRP, Baltimore, MD
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, NIDA/NIAAA IRP, Baltimore, MD
| | | |
Collapse
|
5
|
Eraso‐Pichot A, Pouvreau S, Olivera‐Pinto A, Gomez‐Sotres P, Skupio U, Marsicano G. Endocannabinoid signaling in astrocytes. Glia 2023; 71:44-59. [PMID: 35822691 PMCID: PMC9796923 DOI: 10.1002/glia.24246] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 06/28/2022] [Accepted: 07/04/2022] [Indexed: 01/07/2023]
Abstract
The study of the astrocytic contribution to brain functions has been growing in popularity in the neuroscience field. In the last years, and especially since the demonstration of the involvement of astrocytes in synaptic functions, the astrocyte field has revealed multiple functions of these cells that seemed inconceivable not long ago. In parallel, cannabinoid investigation has also identified different ways by which cannabinoids are able to interact with these cells, modify their functions, alter their communication with neurons and impact behavior. In this review, we will describe the expression of different endocannabinoid system members in astrocytes. Moreover, we will relate the latest findings regarding cannabinoid modulation of some of the most relevant astroglial functions, namely calcium (Ca2+ ) dynamics, gliotransmission, metabolism, and inflammation.
Collapse
Affiliation(s)
- Abel Eraso‐Pichot
- U1215 Neurocentre MagendieInstitut national de la santé et de la recherche médicale (INSERM)BordeauxFrance,University of BordeauxBordeauxFrance
| | - Sandrine Pouvreau
- U1215 Neurocentre MagendieInstitut national de la santé et de la recherche médicale (INSERM)BordeauxFrance,University of BordeauxBordeauxFrance
| | - Alexandre Olivera‐Pinto
- U1215 Neurocentre MagendieInstitut national de la santé et de la recherche médicale (INSERM)BordeauxFrance,University of BordeauxBordeauxFrance
| | - Paula Gomez‐Sotres
- U1215 Neurocentre MagendieInstitut national de la santé et de la recherche médicale (INSERM)BordeauxFrance,University of BordeauxBordeauxFrance
| | - Urszula Skupio
- U1215 Neurocentre MagendieInstitut national de la santé et de la recherche médicale (INSERM)BordeauxFrance,University of BordeauxBordeauxFrance
| | - Giovanni Marsicano
- U1215 Neurocentre MagendieInstitut national de la santé et de la recherche médicale (INSERM)BordeauxFrance,University of BordeauxBordeauxFrance
| |
Collapse
|
6
|
Neutral CB1 Receptor Antagonists as Pharmacotherapies for Substance Use Disorders: Rationale, Evidence, and Challenge. Cells 2022; 11:cells11203262. [PMID: 36291128 PMCID: PMC9600259 DOI: 10.3390/cells11203262] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Revised: 10/03/2022] [Accepted: 10/10/2022] [Indexed: 11/17/2022] Open
Abstract
Cannabinoid receptor 1 (CB1R) has been one of the major targets in medication development for treating substance use disorders (SUDs). Early studies indicated that rimonabant, a selective CB1R antagonist with an inverse agonist profile, was highly promising as a therapeutic for SUDs. However, its adverse side effects, such as depression and suicidality, led to its withdrawal from clinical trials worldwide in 2008. Consequently, much research interest shifted to developing neutral CB1R antagonists based on the recognition that rimonabant’s side effects may be related to its inverse agonist profile. In this article, we first review rimonabant’s research background as a potential pharmacotherapy for SUDs. Then, we discuss the possible mechanisms underlying its therapeutic anti-addictive effects versus its adverse effects. Lastly, we discuss the rationale for developing neutral CB1R antagonists as potential treatments for SUDs, the supporting evidence in recent research, and the challenges of this strategy. We conclude that developing neutral CB1R antagonists without inverse agonist profile may represent attractive strategies for the treatment of SUDs.
Collapse
|
7
|
Desmercieres S, Lardeux V, Longueville JE, Hanna M, Panlilio LV, Thiriet N, Solinas M. A self-adjusting, progressive shock strength procedure to investigate resistance to punishment: Characterization in male and female rats. Neuropharmacology 2022; 220:109261. [PMID: 36152690 DOI: 10.1016/j.neuropharm.2022.109261] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 09/06/2022] [Accepted: 09/14/2022] [Indexed: 10/14/2022]
Abstract
Indifference to harmful consequences is one of the main characteristics of compulsive behaviors and addiction. Animal models that provide a rapid and effective measure of resistance to punishment could be critical for the investigation of mechanisms underlying these maladaptive behaviors. Here, analogous to the progressive ratio (PR) procedure widely used to evaluate appetitive motivation as the response requirement is increased, we developed a self-adjusting, progressive shock strength (PSS) procedure. The PSS provides, within a single session, a break point that quantifies the propensity to work for a reward in spite of receiving electric footshock that progressively increases in duration. In both male and female rats, the PSS break point was sensitive to 1) hunger; and 2) changes in the qualitative, but not quantitative, incentive value of the reward. In systematic comparisons between PSS and PR procedures in the same rats, we found that both measures are sensitive to manipulations of motivational states, but they are not intercorrelated, suggesting that they measure overlapping but partially distinct processes. Importantly, the PSS procedure represents a refinement in the 3Rs principles of animal research because animals can control the strength of shock that they are willing to tolerate. This self-adjusting PSS procedure may represent a useful tool to investigate mechanisms underlying maladaptive behavior that persists in certain individuals despite harmful consequences.
Collapse
Affiliation(s)
- Stevenson Desmercieres
- Université de Poitiers, INSERM, U-1084, Laboratoire des Neurosciences Expérimentales et Cliniques, Poitiers, France
| | - Virginie Lardeux
- Université de Poitiers, INSERM, U-1084, Laboratoire des Neurosciences Expérimentales et Cliniques, Poitiers, France
| | - Jean-Emmanuel Longueville
- Université de Poitiers, INSERM, U-1084, Laboratoire des Neurosciences Expérimentales et Cliniques, Poitiers, France
| | - Myriam Hanna
- Université de Poitiers, INSERM, U-1084, Laboratoire des Neurosciences Expérimentales et Cliniques, Poitiers, France
| | - Leigh V Panlilio
- Real-world Assessment, Prediction, and Treatment Unit, Translational Addiction Medicine Branch, Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD, USA
| | - Nathalie Thiriet
- Université de Poitiers, INSERM, U-1084, Laboratoire des Neurosciences Expérimentales et Cliniques, Poitiers, France
| | - Marcello Solinas
- Université de Poitiers, INSERM, U-1084, Laboratoire des Neurosciences Expérimentales et Cliniques, Poitiers, France.
| |
Collapse
|
8
|
Toniolo EF, Gupta A, Franciosi AC, Gomes I, Devi LA, Dale CS. Interactions between cannabinoid and opioid receptors in a mouse model of diabetic neuropathy. Pain 2022; 163:1414-1423. [PMID: 34724682 PMCID: PMC9043031 DOI: 10.1097/j.pain.0000000000002527] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 10/19/2021] [Indexed: 11/25/2022]
Abstract
ABSTRACT Diabetic neuropathy, often associated with diabetes mellitus, is a painful condition with no known effective treatment except glycemic control. Studies with neuropathic pain models report alterations in cannabinoid and opioid receptor expression levels; receptors whose activation induces analgesia. We examined whether interactions between CB1R and opioid receptors could be targeted for the treatment of diabetic neuropathy. For this, we generated antibodies that selectively recognize native CB1R-MOR and CB1R-DOR heteromers using a subtractive immunization strategy. We assessed the levels of CB1R, MOR, DOR, and interacting complexes using a model of streptozotocin-induced diabetic neuropathy and detected increased levels of CB1R, MOR, DOR, and CB1R-MOR complexes compared with those in controls. An examination of G-protein signaling revealed that activity induced by the MOR, but not the DOR agonist, was potentiated by low nanomolar doses of CB1R ligands, including antagonists, suggesting an allosteric modulation of MOR signaling by CB1R ligands within CB1R-MOR complexes. Because the peptide endocannabinoid, hemopressin, caused a significant potentiation of MOR activity, we examined its effect on mechanical allodynia and found that it blocked allodynia in wild-type mice and mice with diabetic neuropathy lacking DOR (but have CB1R-MOR complexes). However, hemopressin does not alter the levels of CB1R-MOR complexes in diabetic mice lacking DOR but increases the levels of CB1R-DOR complexes in diabetic mice lacking MOR. Together, these results suggest the involvement of CB1R-MOR and CB1R-DOR complexes in diabetic neuropathy and that hemopressin could be developed as a potential therapeutic for the treatment of this painful condition.
Collapse
Affiliation(s)
- Elaine F. Toniolo
- Department of Pharmacology, University of Sao Paulo, Sao Paulo, Brazil
- Department of Anatomy, Laboratory of Neuromodulation and Experimental Pain, University of Sao Paulo, Sao Paulo, Brazil
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Hospital Sírio-Libanês, São Paulo, Brasil
| | - Achla Gupta
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Adriano C. Franciosi
- Department of Pharmacology, University of Sao Paulo, Sao Paulo, Brazil
- Department of Anatomy, Laboratory of Neuromodulation and Experimental Pain, University of Sao Paulo, Sao Paulo, Brazil
- Hospital Sírio-Libanês, São Paulo, Brasil
| | - Ivone Gomes
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Lakshmi A. Devi
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Camila S. Dale
- Department of Anatomy, Laboratory of Neuromodulation and Experimental Pain, University of Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
9
|
He XH, Galaj E, Bi GH, He Y, Hempel B, Wang YL, Gardner EL, Xi ZX. β-caryophyllene, an FDA-Approved Food Additive, Inhibits Methamphetamine-Taking and Methamphetamine-Seeking Behaviors Possibly via CB2 and Non-CB2 Receptor Mechanisms. Front Pharmacol 2021; 12:722476. [PMID: 34566647 PMCID: PMC8458938 DOI: 10.3389/fphar.2021.722476] [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: 06/08/2021] [Accepted: 08/23/2021] [Indexed: 01/11/2023] Open
Abstract
Recent research indicates that brain cannabinoid CB2 receptors are involved in drug reward and addiction. However, it is unclear whether β-caryophyllene (BCP), a natural product with a CB2 receptor agonist profile, has therapeutic effects on methamphetamine (METH) abuse and dependence. In this study, we used animal models of self-administration, electrical brain-stimulation reward (BSR) and in vivo microdialysis to explore the effects of BCP on METH-taking and METH-seeking behavior. We found that systemic administration of BCP dose-dependently inhibited METH self-administration under both fixed-ratio and progressive-ratio reinforcement schedules in rats, indicating that BCP reduces METH reward, METH intake, and incentive motivation to seek and take METH. The attenuating effects of BCP were partially blocked by AM 630, a selective CB2 receptor antagonist. Genetic deletion of CB2 receptors in CB2-knockout (CB2-KO) mice also blocked low dose BCP-induced reduction in METH self-administration, suggesting possible involvement of a CB2 receptor mechanism. However, at high doses, BCP produced a reduction in METH self-administration in CB2-KO mice in a manner similar as in WT mice, suggesting that non-CB2 receptor mechanisms underlie high dose BCP-produced effects. In addition, BCP dose-dependently attenuated METH-enhanced electrical BSR and inhibited METH-primed and cue-induced reinstatement of drug-seeking in rats. In vivo microdialysis assays indicated that BCP alone did not produce a significant reduction in extracellular dopamine (DA) in the nucleus accumbens (NAc), while BCP pretreatment significantly reduced METH-induced increases in extracellular NAc DA in a dose-dependent manner, suggesting a DA-dependent mechanism involved in BCP action. Together, the present findings suggest that BCP might be a promising therapeutic candidate for the treatment of METH use disorder.
Collapse
Affiliation(s)
- Xiang-Hu He
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, United States.,Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Hubei, China
| | - Ewa Galaj
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, United States
| | - Guo-Hua Bi
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, United States
| | - Yi He
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, United States
| | - Briana Hempel
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, United States
| | - Yan-Lin Wang
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Hubei, China
| | - Eliot L Gardner
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, United States
| | - Zheng-Xiong Xi
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, United States
| |
Collapse
|
10
|
Engi SA, Beebe EJ, Ayvazian VM, Cruz FC, Cheer JF, Wenzel JM, Zlebnik NE. Cocaine-induced increases in motivation require 2-arachidonoylglycerol mobilization and CB1 receptor activation in the ventral tegmental area. Neuropharmacology 2021; 193:108625. [PMID: 34058192 PMCID: PMC8312311 DOI: 10.1016/j.neuropharm.2021.108625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 05/13/2021] [Accepted: 05/24/2021] [Indexed: 11/15/2022]
Abstract
A wide body of evidence supports an integral role for mesolimbic dopamine (DA) in motivated behavior. In brief, drugs that increase DA in mesolimbic terminal regions, like cocaine, enhance motivation, while drugs that decrease DA concentration reduce motivation. Data from our laboratory and others shows that phasic activation of mesolimbic DA requires signaling at cannabinoid type-1 (CB1) receptors in the ventral tegmental area (VTA), and systemic delivery of CB1 receptor antagonists reduces DA cell activity and attenuates motivated behaviors. Recent findings demonstrate that cocaine mobilizes the endocannabinoid 2-arachidonoylglycerol (2-AG) in the VTA to cause phasic activation of DA neurons and terminal DA release. It remains unclear, however, if cocaine-induced midbrain 2-AG signaling contributes to the motivation-enhancing effects of cocaine. To examine this, we trained male and female rats on a progressive ratio (PR) task for a food reinforcer. Each rat underwent a series of tests in which they were pretreated with cocaine alone or in combination with systemic or intra-VTA administration of the CB1 receptor antagonist rimonabant or the 2-AG synthesis inhibitor tetrahydrolipstatin (THL). Cocaine increased motivation, measured by augmented PR breakpoints, while rimonabant dose-dependently decreased motivation. Importantly, intra-VTA administration of rimonabant or THL, at doses that did not decrease breakpoints on their own, blocked systemic cocaine administration from increasing breakpoints in male and female rats. These data suggest that cocaine-induced increases in motivation require 2-AG signaling at CB1 receptors in the VTA and may provide critical insight into cannabinoid-based pharmacotherapeutic targets for the successful treatment of substance abuse.
Collapse
Affiliation(s)
- Sheila A Engi
- Dept. of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA; Dept. of Pharmacology, Universidade Federal de São Paulo - UNIFESP, São Paulo, SP, Brazil
| | - Erin J Beebe
- Dept. of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Victoria M Ayvazian
- Dept. of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Fabio C Cruz
- Dept. of Pharmacology, Universidade Federal de São Paulo - UNIFESP, São Paulo, SP, Brazil
| | - Joseph F Cheer
- Dept. of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA; Dept. of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Jennifer M Wenzel
- Dept. of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA.
| | - Natalie E Zlebnik
- Dept. of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
11
|
Wilkerson JL, Bilbrey JA, Felix JS, Makriyannis A, McMahon LR. Untapped endocannabinoid pharmacological targets: Pipe dream or pipeline? Pharmacol Biochem Behav 2021; 206:173192. [PMID: 33932409 DOI: 10.1016/j.pbb.2021.173192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 04/18/2021] [Accepted: 04/21/2021] [Indexed: 10/21/2022]
Abstract
It has been established that the endogenous cannabinoid (endocannabinoid) system plays key modulatory roles in a wide variety of pathological conditions. The endocannabinoid system comprises both cannabinoid receptors, their endogenous ligands including 2-arachidonoylglycerol (2-AG), N-arachidonylethanolamine (anandamide, AEA), and enzymes that regulate the synthesis and degradation of endogenous ligands which include diacylglycerol lipase alpha (DAGL-α), diacylglycerol lipase beta (DAGL-β), fatty acid amide hydrolase (FAAH), monoacylglycerol lipase (MAGL), α/β hydrolase domain 6 (ABHD6). As the endocannabinoid system exerts considerable involvement in the regulation of homeostasis and disease, much effort has been made towards understanding endocannabinoid-related mechanisms of action at cellular, physiological, and pathological levels as well as harnessing the various components of the endocannabinoid system to produce novel therapeutics. However, drug discovery efforts within the cannabinoid field have been slower than anticipated to reach satisfactory clinical endpoints and raises an important question into the validity of developing novel ligands that therapeutically target the endocannabinoid system. To answer this, we will first examine evidence that supports the existence of an endocannabinoid system role within inflammatory diseases, neurodegeneration, pain, substance use disorders, mood disorders, as well as metabolic diseases. Next, this review will discuss recent clinical studies, within the last 5 years, of cannabinoid compounds in context to these diseases. We will also address some of the challenges and considerations within the cannabinoid field that may be important in the advancement of therapeutics into the clinic.
Collapse
Affiliation(s)
- Jenny L Wilkerson
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, USA.
| | - Joshua A Bilbrey
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Jasmine S Felix
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Alexandros Makriyannis
- Center for Drug Discovery and Department of Pharmaceutical Sciences, Northeastern University, Boston, MA 02115, USA; Departments of Chemistry and Chemical Biology, Northeastern University, Boston, MA 02115, USA
| | - Lance R McMahon
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
12
|
Zhang H, Lipinski AA, Liktor-Busa E, Smith AF, Moutal A, Khanna R, Langlais PR, Largent-Milnes TM, Vanderah TW. The Effects of Repeated Morphine Treatment on the Endogenous Cannabinoid System in the Ventral Tegmental Area. Front Pharmacol 2021; 12:632757. [PMID: 33953672 PMCID: PMC8090348 DOI: 10.3389/fphar.2021.632757] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 02/26/2021] [Indexed: 12/18/2022] Open
Abstract
The therapeutic utility of opioids is diminished by their ability to induce rewarding behaviors that may lead to opioid use disorder. Recently, the endogenous cannabinoid system has emerged as a hot topic in the study of opioid reward but relatively little is known about how repeated opioid exposure may affect the endogenous cannabinoid system in the mesolimbic reward circuitry. In the present study, we investigated how sustained morphine may modulate the endogenous cannabinoid system in the ventral tegmental area (VTA) of Sprague Dawley rats, a critical region in the mesolimbic reward circuitry. Studies here using proteomic analysis and quantitative real-time PCR (qRT-PCR) found that the VTA expresses 32 different proteins or genes related to the endogenous cannabinoid system; three of these proteins or genes (PLCγ2, ABHD6, and CB2R) were significantly affected after repeated morphine exposure (CB2R was only detected by qRT-PCR but not proteomics). We also identified that repeated morphine treatment does not alter either anandamide (AEA) or 2-arachidonoylglycerol (2-AG) levels in the VTA compared to saline treatment; however, there may be diminished levels of anandamide (AEA) production in the VTA 4 h after a single morphine injection in both chronic saline and morphine pretreated cohorts. Treating the animals with an inhibitor of 2-AG degradation significantly decreased repeated opioid rewarding behavior. Taken together, our studies reveal a potential influence of sustained opioids on the endocannabinoid system in the VTA, suggesting that the endogenous cannabinoid system may participate in the opioid-induced reward.
Collapse
Affiliation(s)
- Hong Zhang
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, United States
| | - Austin A. Lipinski
- Department of Medicine, Division of Endocrinology, College of Medicine, University of Arizona, Tucson, AZ, United States
| | - Erika Liktor-Busa
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, United States
| | - Angela F. Smith
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, United States
| | - Aubin Moutal
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, United States
| | - Rajesh Khanna
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, United States
| | - Paul R. Langlais
- Department of Medicine, Division of Endocrinology, College of Medicine, University of Arizona, Tucson, AZ, United States
| | - Tally M. Largent-Milnes
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, United States
| | - Todd W. Vanderah
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, United States
| |
Collapse
|
13
|
Mohammadkhani A, Borgland SL. Cellular and behavioral basis of cannabinioid and opioid interactions: Implications for opioid dependence and withdrawal. J Neurosci Res 2020; 100:278-296. [PMID: 33352618 DOI: 10.1002/jnr.24770] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 11/25/2020] [Indexed: 01/22/2023]
Abstract
The brain's endogenous opioid and endocannabinoid systems are neuromodulatory of synaptic transmission, and play key roles in pain, memory, reward, and addiction. Recent clinical and pre-clinical evidence suggests that opioid use may be reduced with cannabinoid intake. This suggests the presence of a functional interaction between these two systems. Emerging research indicates that cannabinoids and opioids can functionally interact at different levels. At the cellular level, opioid and cannabinoids can have direct receptor associations, alterations in endogenous opioid peptide or cannabinoid release, or post-receptor activation interactions via shared signal transduction pathways. At the systems level, the nature of cannabinoid and opioid interaction might differ in brain circuits underlying different behavioral phenomenon, including reward-seeking or antinociception. Given the rising use of opioid and cannabinoid drugs, a better understanding of how these endogenous signaling systems interact in the brain is of significant interest. This review focuses on the potential relationship of these neural systems in addiction-related processes.
Collapse
Affiliation(s)
- Aida Mohammadkhani
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, The University of Calgary, Calgary, AB, Canada
| | - Stephanie L Borgland
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, The University of Calgary, Calgary, AB, Canada
| |
Collapse
|
14
|
Moreno-Rius J. Opioid addiction and the cerebellum. Neurosci Biobehav Rev 2019; 107:238-251. [DOI: 10.1016/j.neubiorev.2019.09.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 09/06/2019] [Accepted: 09/10/2019] [Indexed: 01/10/2023]
|
15
|
Fitzgerald ML, Mackie K, Pickel VM. Ultrastructural localization of cannabinoid CB1 and mGluR5 receptors in the prefrontal cortex and amygdala. J Comp Neurol 2019; 527:2730-2741. [PMID: 31008528 DOI: 10.1002/cne.24704] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 04/01/2019] [Accepted: 04/17/2019] [Indexed: 12/29/2022]
Abstract
Stimulation of the postsynaptic metabotropic glutamate receptor mGluR5 triggers retrograde signaling of endocannabinoids that activate presynaptic cannabinoid CB1 receptors on juxtaposing axon terminals. To better understand the synaptic structure that supports mGluR5 mediation of CB1 activation in the prefrontal cortex (PFC) and basolateral amygdala (BLA), we examined electron microscopic dual immunolabeling of these receptors in the prelimbic PFC (prPFC) and BLA of adult male rats. CB1 immunoreactivity was detected in axon terminals that were typically large, complex, and contained dense-core and clear synaptic vesicles. Of terminals forming discernible synaptic specializations, 95% were symmetric inhibitory-type in the prPFC and 90% were inhibitory in the BLA. CB1-immunoreactive terminals frequently contacted dendrites containing mGluR5 adjacent to unlabeled terminals forming excitatory-type synapses. Because most CB1-containing terminals form inhibitory-type synapses, the unlabeled axon terminals forming asymmetric synapses are the likely source of the mGluR5 ligand glutamate. In the prPFC, serial section analysis revealed that GABAergic CB1-containing axon terminals targeted dendrites adjacent to glutamatergic axon terminals, often near dendritic bifurcations. These observations provide ultrastructural evidence that cortical CB1 receptors are strategically positioned for integration of synaptic signaling in response to stimulation of postsynaptic mGluR5 receptors and facilitation of heterosynaptic communication between multiple neurons.
Collapse
Affiliation(s)
- Megan L Fitzgerald
- Weill Cornell Medicine, Feil Family Brain and Mind Research Institute, New York, New York
| | - Ken Mackie
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, Indiana
| | - Virginia M Pickel
- Weill Cornell Medicine, Feil Family Brain and Mind Research Institute, New York, New York
| |
Collapse
|
16
|
Cannabinoid CB 1 receptor neutral antagonist AM4113 inhibits heroin self-administration without depressive side effects in rats. Acta Pharmacol Sin 2019; 40:365-373. [PMID: 29967454 DOI: 10.1038/s41401-018-0059-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2018] [Accepted: 05/31/2018] [Indexed: 11/08/2022] Open
Abstract
Cannabinoid CB1 receptors (CB1Rs) have been shown to be a promising target in medication development for the treatment of addiction. However, clinical trials with SR141716A (rimonabant, a selective CB1R antagonist/inverse agonist) for the treatment of obesity and smoking cessation failed due to unwanted side effects, such as depression, anxiety, and suicidal tendencies. Recent preclinical studies suggest that the neutral CB1R antagonist AM4113 may retain the therapeutic anti-addictive effects of SR141716A in nicotine self-administration models and possibly has fewer unwanted side effects. However, little is known about whether AM4113 is also effective for other drugs of abuse, such as opioids and psychostimulants, and whether it produces depressive side effects similar to SR141716A in experimental animals. In this study, we demonstrated that systemic administration of AM4113 (3 and 10 mg/kg) dose-dependently inhibited the self-administration of intravenous heroin but not cocaine or methamphetamine, whereas SR141716A (3 and 10 mg/kg) dose-dependently inhibited the self-administration of heroin and methamphetamine but not cocaine. In the electrical brain-stimulation reward (BSR) paradigm, SR141716A (3 and 10 mg/kg) dose-dependently increased the BSR stimulation threshold (i.e., decreased the stimulation reward), but AM4113 had no effect on BSR at the same doses, suggesting that SR141716A may produce aversive effects while AM4113 may not. Together, these findings show that neutral CB1R antagonists such as AM4113 deserve further research as a new class of CB1R-based medications for the treatment of opioid addiction without SR141716A-like aversive effects.
Collapse
|
17
|
Hay GL, Baracz SJ, Everett NA, Roberts J, Costa PA, Arnold JC, McGregor IS, Cornish JL. Cannabidiol treatment reduces the motivation to self-administer methamphetamine and methamphetamine-primed relapse in rats. J Psychopharmacol 2018; 32:1369-1378. [PMID: 30260267 DOI: 10.1177/0269881118799954] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND Methamphetamine is an addictive stimulant that can cause many adverse physical, psychological and psychosocial effects. Preliminary evidence shows cannabidiol, a non-intoxicating constituent of the cannabis plant, may have efficacy in treating opioid and nicotine dependence. However, no study has yet examined whether cannabidiol treatment might impact on methamphetamine addiction. AIMS The current study investigated whether cannabidiol administration reduces the motivation to self-administer methamphetamine and relapse to methamphetamine-seeking behavior following abstinence. METHODS Thirty-two male Sprague Dawley rats with implanted jugular vein catheters were initially trained to self-administer methamphetamine via lever press during two-hour sessions on a fixed ratio 1 schedule of reinforcement. Rats in experiment 1 ( n=16) then advanced to a progressive ratio reinforcement schedule to examine the effects of cannabidiol (0, 20, 40, and 80 mg/kg intraperitoneal) on motivation to self-administer methamphetamine. Rats in experiment 2 ( n=16) were tested for cannabidiol effects on methamphetamine-primed reinstatement following extinction. RESULTS Cannabidiol (80 mg/kg, but not 40 mg/kg, or 20 mg/kg) reduced the motivation to self-administer methamphetamine and attenuated methamphetamine-primed relapse to methamphetamine-seeking behavior after extinction. CONCLUSION This is the first demonstration that cannabidiol can reduce the motivation to seek and consume methamphetamine, and suggests that cannabidiol might be worth trialing as a novel pharmacotherapy for methamphetamine dependence.
Collapse
Affiliation(s)
- Gracie L Hay
- 1 Department of Psychology, Macquarie University, North Ryde, NSW, Australia
| | - Sarah J Baracz
- 1 Department of Psychology, Macquarie University, North Ryde, NSW, Australia.,2 School of Psychology, University of Sydney, Sydney, NSW, Australia
| | - Nicholas A Everett
- 1 Department of Psychology, Macquarie University, North Ryde, NSW, Australia
| | - Jessica Roberts
- 1 Department of Psychology, Macquarie University, North Ryde, NSW, Australia
| | - Priscila A Costa
- 1 Department of Psychology, Macquarie University, North Ryde, NSW, Australia
| | - Jonathon C Arnold
- 3 Department of Pharmacology, University of Sydney, Sydney, NSW, Australia.,4 Lambert Initiative for Cannabinoid Therapeutics, University of Sydney, Sydney, NSW, Australia
| | - Iain S McGregor
- 2 School of Psychology, University of Sydney, Sydney, NSW, Australia.,4 Lambert Initiative for Cannabinoid Therapeutics, University of Sydney, Sydney, NSW, Australia
| | - Jennifer L Cornish
- 1 Department of Psychology, Macquarie University, North Ryde, NSW, Australia
| |
Collapse
|
18
|
De Luca MA, Buczynski MW, Di Chiara G. Loren Parsons' contribution to addiction neurobiology. Addict Biol 2018; 23:1207-1222. [PMID: 29949237 DOI: 10.1111/adb.12642] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 05/17/2018] [Indexed: 11/29/2022]
Abstract
Loren (Larry) H. Parsons passed away at the age of 51. In spite of his premature departure, Larry much contributed to the drug abuse field. Since his graduate studies for the Ph.D. in Chemistry in J.B. Justice lab, microdialysis is the tread that links Larry's research topics, namely, the role of dopamine (DA), serotonin (5-HT), gamma-aminobutyric acid (GABA), glutamate and endocannabinoids (eCBs) in drug reinforcement and dependence. Larry was the first to show that abstinence from chronic cocaine reduces extracellular DA in the NAc, consistent with the so called 'dopamine depletion hypothesis' of cocaine addiction. Another Larry's major contributions are the studies on 5-HT and 5-HT receptors' role in cocaine stimulant actions, which resulted in the identification of 5-HT1B receptors as a critical substrate of cocaine reinforcement. By applying mass spectrometry to eCBs analysis in brain dialysates, Larry's lab showed that ethanol, heroin, nicotine and cocaine differentially affect anandamide and 2-arachidonoylglicerol overflow in the NAc shell, a critical site of drugs of abuse DA stimulant actions. Larry also applied microdialysis to study GABA and glutamate's role in ethanol dependence and heroin reinforcement, providing in vivo evidence for a sensitization of corticotropin-releasing factor-dependent release of GABA in the central amygdala in withdrawal from chronic ethanol and for a reduction of GABA transmission in the ventral pallidum in heroin but not cocaine intravenous self-administration. Larry showed the wide possibilities of microdialysis as a general purpose methodology for monitoring neurotransmitters and neuromodulators in the brain extracellular compartment. From this viewpoint, he stands as the best advocate for microdialysis.
Collapse
Affiliation(s)
- Maria A. De Luca
- Department of Biomedical Sciences, Section of Neuropsychopharmacology; University of Cagliari; Cagliari Italy
- National Institute of Neuroscience (INN); University of Cagliari; Cagliari Italy
| | - Matthew W. Buczynski
- School of Neuroscience; Virginia Polytechnic Institute and State University; Blacksburg VA 24061 USA
| | - Gaetano Di Chiara
- Department of Biomedical Sciences, Section of Neuropsychopharmacology; University of Cagliari; Cagliari Italy
- National Institute of Neuroscience (INN); University of Cagliari; Cagliari Italy
- National Research Council of Italy; Institute of Neuroscience; Cagliari Italy
| |
Collapse
|
19
|
Spechler PA, Allgaier N, Chaarani B, Whelan R, Watts R, Orr C, Albaugh MD, D'Alberto N, Higgins ST, Hudson KE, Mackey S, Potter A, Banaschewski T, Bokde ALW, Bromberg U, Büchel C, Cattrell A, Conrod PJ, Desrivières S, Flor H, Frouin V, Gallinat J, Gowland P, Heinz A, Ittermann B, Martinot JL, Paillère Martinot ML, Nees F, Papadopoulos Orfanos D, Paus T, Poustka L, Smolka MN, Walter H, Schumann G, Althoff RR, Garavan H. The initiation of cannabis use in adolescence is predicted by sex-specific psychosocial and neurobiological features. Eur J Neurosci 2018; 50:2346-2356. [PMID: 29889330 DOI: 10.1111/ejn.13989] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 05/03/2018] [Accepted: 05/21/2018] [Indexed: 12/14/2022]
Abstract
Cannabis use initiated during adolescence might precipitate negative consequences in adulthood. Thus, predicting adolescent cannabis use prior to any exposure will inform the aetiology of substance abuse by disentangling predictors from consequences of use. In this prediction study, data were drawn from the IMAGEN sample, a longitudinal study of adolescence. All selected participants (n = 1,581) were cannabis-naïve at age 14. Those reporting any cannabis use (out of six ordinal use levels) by age 16 were included in the outcome group (N = 365, males n = 207). Cannabis-naïve participants at age 14 and 16 were included in the comparison group (N = 1,216, males n = 538). Psychosocial, brain and genetic features were measured at age 14 prior to any exposure. Cross-validated regularized logistic regressions for each use level by sex were used to perform feature selection and obtain prediction error statistics on independent observations. Predictors were probed for sex- and drug-specificity using post-hoc logistic regressions. Models reliably predicted use as indicated by satisfactory prediction error statistics, and contained psychosocial features common to both sexes. However, males and females exhibited distinct brain predictors that failed to predict use in the opposite sex or predict binge drinking in independent samples of same-sex participants. Collapsed across sex, genetic variation on catecholamine and opioid receptors marginally predicted use. Using machine learning techniques applied to a large multimodal dataset, we identified a risk profile containing psychosocial and sex-specific brain prognostic markers, which were likely to precede and influence cannabis initiation.
Collapse
Affiliation(s)
- Philip A Spechler
- Vermont Center on Behavior and Health, University of Vermont, Burlington, VT, USA.,Department of Psychological Science, University of Vermont, Burlington, VT, 05401, USA.,Department of Psychiatry, University of Vermont, Burlington, VT, USA
| | - Nicholas Allgaier
- Department of Psychiatry, University of Vermont, Burlington, VT, USA
| | - Bader Chaarani
- Vermont Center on Behavior and Health, University of Vermont, Burlington, VT, USA.,Department of Psychiatry, University of Vermont, Burlington, VT, USA
| | - Robert Whelan
- School of Psychology and Global Brain Health Institute, Trinity College Dublin, Dublin, Ireland
| | - Richard Watts
- Department of Radiology, University of Vermont, Burlington, VT, USA
| | - Catherine Orr
- Department of Psychiatry, University of Vermont, Burlington, VT, USA
| | - Matthew D Albaugh
- Department of Psychiatry, University of Vermont, Burlington, VT, USA
| | | | - Stephen T Higgins
- Vermont Center on Behavior and Health, University of Vermont, Burlington, VT, USA.,Department of Psychological Science, University of Vermont, Burlington, VT, 05401, USA.,Department of Psychiatry, University of Vermont, Burlington, VT, USA
| | - Kelsey E Hudson
- Department of Psychological Science, University of Vermont, Burlington, VT, 05401, USA
| | - Scott Mackey
- Department of Psychiatry, University of Vermont, Burlington, VT, USA
| | - Alexandra Potter
- Department of Psychiatry, University of Vermont, Burlington, VT, USA
| | - Tobias Banaschewski
- Medical Faculty Mannheim, Department of Child and Adolescent Psychiatry and Psychotherapy, Central Institute of Mental Health, Heidelberg University, Mannheim, Germany
| | - Arun L W Bokde
- Discipline of Psychiatry, School of Medicine and Trinity College Institute of Neurosciences, Trinity College Dublin, Dublin, Ireland
| | - Uli Bromberg
- University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | | | - Anna Cattrell
- Centre for Population Neuroscience and Stratified Medicine (PONS) and MRC-SGDP Centre, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Patricia J Conrod
- Department of Psychiatry, Universite de Montreal, CHU Ste Justine Hospital, Montreal, Canada
| | - Sylvane Desrivières
- Centre for Population Neuroscience and Stratified Medicine (PONS) and MRC-SGDP Centre, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Herta Flor
- Medical Faculty Mannheim, Department of Cognitive and Clinical Neuroscience, Central Institute of Mental Health, Heidelberg University, Mannheim, Germany.,Department of Psychology, School of Social Sciences, University of Mannheim, Mannheim, Germany
| | - Vincent Frouin
- NeuroSpin, CEA, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Jürgen Gallinat
- Department of Psychiatry and Psychotherapy, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Penny Gowland
- Sir Peter Mansfield Imaging Centre School of Physics and Astronomy, University of Nottingham, University Park, Nottingham, UK
| | - Andreas Heinz
- Department of Psychiatry and Psychotherapy, Charité, Universitätsmedizin Berlin, Campus Charité Mitte, Berlin, Germany
| | - Bernd Ittermann
- Physikalisch-Technische Bundesanstalt (PTB), Braunschweig, Germany
| | - Jean-Luc Martinot
- DIGITEO Labs, Institut National de la Santé et de la Recherche Médicale, INSERM Unit 1000 "Neuroimaging & Psychiatry", University Paris Sud - University Paris Saclay, Gif sur Yvette, France
| | - Marie-Laure Paillère Martinot
- Institut National de la Santé et de la Recherche Médicale, INSERM Unit 1000 "Neuroimaging & Psychiatry", University Paris Sud - Paris Saclay, University Paris Descartes, Paris, France.,Department of Adolescent Psychopathology and Medicine, AP-HP, Maison de Solenn, Cochin Hospital, Paris, France
| | - Frauke Nees
- Medical Faculty Mannheim, Department of Child and Adolescent Psychiatry and Psychotherapy, Central Institute of Mental Health, Heidelberg University, Mannheim, Germany.,Medical Faculty Mannheim, Department of Cognitive and Clinical Neuroscience, Central Institute of Mental Health, Heidelberg University, Mannheim, Germany
| | | | - Tomáš Paus
- Baycrest and Departments of Psychology and Psychiatry, Rotman Research Institute, University of Toronto, Toronto, ON, M6A 2E1, Canada
| | - Luise Poustka
- Department of Child and Adolescent Psychiatry and Psychotherapy, University Medical Centre Göttingen, 37075, Göttingen, Germany.,Clinic for Child and Adolescent Psychiatry, Medical University of Vienna, Vienna, Austria
| | - Michael N Smolka
- Department of Psychiatry and Neuroimaging Center, Technische Universität Dresden, Dresden, Germany
| | - Henrik Walter
- Department of Psychiatry and Psychotherapy, Charité, Universitätsmedizin Berlin, Campus Charité Mitte, Berlin, Germany
| | - Gunter Schumann
- Centre for Population Neuroscience and Stratified Medicine (PONS) and MRC-SGDP Centre, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Robert R Althoff
- Department of Psychological Science, University of Vermont, Burlington, VT, 05401, USA.,Department of Psychiatry, University of Vermont, Burlington, VT, USA
| | - Hugh Garavan
- Vermont Center on Behavior and Health, University of Vermont, Burlington, VT, USA.,Department of Psychological Science, University of Vermont, Burlington, VT, 05401, USA.,Department of Psychiatry, University of Vermont, Burlington, VT, USA
| | | |
Collapse
|
20
|
Abstract
Introduction: The opioid epidemic has become an immense problem in North America, and despite decades of research on the most effective means to treat opioid use disorder (OUD), overdose deaths are at an all-time high, and relapse remains pervasive. Discussion: Although there are a number of FDA-approved opioid replacement therapies and maintenance medications to help ease the severity of opioid withdrawal symptoms and aid in relapse prevention, these medications are not risk free nor are they successful for all patients. Furthermore, there are legal and logistical bottlenecks to obtaining traditional opioid replacement therapies such as methadone or buprenorphine, and the demand for these services far outweighs the supply and access. To fill the gap between efficacious OUD treatments and the widespread prevalence of misuse, relapse, and overdose, the development of novel, alternative, or adjunct OUD treatment therapies is highly warranted. In this article, we review emerging evidence that suggests that cannabis may play a role in ameliorating the impact of OUD. Herein, we highlight knowledge gaps and discuss cannabis' potential to prevent opioid misuse (as an analgesic alternative), alleviate opioid withdrawal symptoms, and decrease the likelihood of relapse. Conclusion: The compelling nature of these data and the relative safety profile of cannabis warrant further exploration of cannabis as an adjunct or alternative treatment for OUD.
Collapse
Affiliation(s)
- Beth Wiese
- Department of Psychology, University of Missouri–St. Louis, St. Louis, Missouri
- Department of Anesthesiology, Pain Center, Washington University School of Medicine, St. Louis, Missouri
| | - Adrianne R. Wilson-Poe
- Department of Anesthesiology, Pain Center, Washington University School of Medicine, St. Louis, Missouri
| |
Collapse
|
21
|
Sustkova-Fiserova M, Charalambous C, Havlickova T, Lapka M, Jerabek P, Puskina N, Syslova K. Alterations in Rat Accumbens Endocannabinoid and GABA Content during Fentanyl Treatment: The Role of Ghrelin. Int J Mol Sci 2017; 18:E2486. [PMID: 29165386 PMCID: PMC5713452 DOI: 10.3390/ijms18112486] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 11/07/2017] [Accepted: 11/17/2017] [Indexed: 12/20/2022] Open
Abstract
The opioid-induced rise of extracellular dopamine, endocannabinoid anandamide and γ-aminobutyric acid (GABA) concentrations triggered by opioids in the nucleus accumbens shell (NACSh) most likely participate in opioid reward. We have previously demonstrated that systemic administration of ghrelin antagonist (JMV2959) significantly decreased morphine-induced dopamine and anandamide (N-arachidonoylethanolamine, AEA) increase in the NACSh. Fentanyl is considered as a µ-receptor-selective agonist. The aim of this study was to test whether JMV2959, a growth hormone secretagogue receptor (GHS-R1A) antagonist, can influence the fentanyl-induced effects on anandamide, 2-arachidonoylglycerol (2-AG) and GABA in the NACSh and specify the involvement of GHS-R1A located in the ventral tegmental area (VTA) and nucleus accumbens (NAC). Using in vivo microdialysis in rats, we have found that pre-treatment with JMV2959 reversed dose dependently fentanyl-induced anandamide increases in the NACSh, resulting in a significant AEA decrease and intensified fentanyl-induced decreases in accumbens 2-AG levels, with both JMV2959 effects more expressed when administered into the NACSh in comparison to the VTA. JMV2959 pre-treatment significantly decreased the fentanyl-evoked accumbens GABA efflux and reduced concurrently monitored fentanyl-induced behavioural stimulation. Our current data encourage further investigation to assess if substances affecting GABA or endocannabinoid concentrations and action, such as GHS-R1A antagonists, can be used to prevent opioid-seeking behaviour.
Collapse
Affiliation(s)
- Magdalena Sustkova-Fiserova
- Department of Pharmacology, Third Faculty of Medicine, Charles University, Ruska 87, 100 34 Prague 10, Czech Republic; (C.C.); (T.H.); (M.L.); (P.J.)
| | - Chrysostomos Charalambous
- Department of Pharmacology, Third Faculty of Medicine, Charles University, Ruska 87, 100 34 Prague 10, Czech Republic; (C.C.); (T.H.); (M.L.); (P.J.)
| | - Tereza Havlickova
- Department of Pharmacology, Third Faculty of Medicine, Charles University, Ruska 87, 100 34 Prague 10, Czech Republic; (C.C.); (T.H.); (M.L.); (P.J.)
| | - Marek Lapka
- Department of Pharmacology, Third Faculty of Medicine, Charles University, Ruska 87, 100 34 Prague 10, Czech Republic; (C.C.); (T.H.); (M.L.); (P.J.)
| | - Pavel Jerabek
- Department of Pharmacology, Third Faculty of Medicine, Charles University, Ruska 87, 100 34 Prague 10, Czech Republic; (C.C.); (T.H.); (M.L.); (P.J.)
| | - Nina Puskina
- Department of Addictology, First Faculty of Medicine, Charles University, Apolinarska 4, 128 00 Prague 2, Czech Republic;
| | - Kamila Syslova
- Laboratory of Medicinal Diagnostics, Department of Organic Technology ICT, Technicka 5, 166 28 Prague 6, Czech Republic;
| |
Collapse
|
22
|
Manduca A, Lassalle O, Sepers M, Campolongo P, Cuomo V, Marsicano G, Kieffer B, Vanderschuren LJMJ, Trezza V, Manzoni OJJ. Interacting Cannabinoid and Opioid Receptors in the Nucleus Accumbens Core Control Adolescent Social Play. Front Behav Neurosci 2016; 10:211. [PMID: 27899885 PMCID: PMC5110529 DOI: 10.3389/fnbeh.2016.00211] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Accepted: 10/18/2016] [Indexed: 12/31/2022] Open
Abstract
Social play behavior is a highly rewarding, developmentally important form of social interaction in young mammals. However, its neurobiological underpinnings remain incompletely understood. Previous work has suggested that opioid and endocannabinoid neurotransmission interact in the modulation of social play. Therefore, we combined behavioral, pharmacological, electrophysiological, and genetic approaches to elucidate the role of the endocannabinoid 2-arachidonoylglycerol (2-AG) in social play, and how cannabinoid and opioid neurotransmission interact to control social behavior in adolescent rodents. Systemic administration of the 2-AG hydrolysis inhibitor JZL184 or the opioid receptor agonist morphine increased social play behavior in adolescent rats. These effects were blocked by systemic pretreatment with either CB1 cannabinoid receptor (CB1R) or mu-opioid receptor (MOR) antagonists. The social play-enhancing effects of systemic morphine or JZL184 treatment were also prevented by direct infusion of the CB1R antagonist SR141716 and the MOR antagonist naloxone into the nucleus accumbens core (NAcC). Searching for synaptic correlates of these effects in adolescent NAcC excitatory synapses, we observed that CB1R antagonism blocked the effect of the MOR agonist DAMGO and, conversely, that naloxone reduced the effect of a cannabinoid agonist. These results were recapitulated in mice, and completely abolished in CB1R and MOR knockout mice, suggesting that the functional interaction between CB1R and MOR in the NAcC in the modulation of social behavior is widespread in rodents. The data shed new light on the mechanism by which endocannabinoid lipids and opioid peptides interact to orchestrate rodent socioemotional behaviors.
Collapse
Affiliation(s)
- Antonia Manduca
- Institut National De La Santé Et De La Recherche Médicale U901Marseille, France; Université de la Méditerranée UMR S901 Aix-Marseille 2Marseille, France; INMEDMarseille, France
| | - Olivier Lassalle
- Institut National De La Santé Et De La Recherche Médicale U901Marseille, France; Université de la Méditerranée UMR S901 Aix-Marseille 2Marseille, France; INMEDMarseille, France
| | - Marja Sepers
- Institut National De La Santé Et De La Recherche Médicale U901Marseille, France; Université de la Méditerranée UMR S901 Aix-Marseille 2Marseille, France; INMEDMarseille, France
| | - Patrizia Campolongo
- Department of Physiology and Pharmacology, Sapienza University of Rome Rome, Italy
| | - Vincenzo Cuomo
- Department of Physiology and Pharmacology, Sapienza University of Rome Rome, Italy
| | - Giovanni Marsicano
- NeuroCentre Magendie, Endocannabinoids and Neuroadaptation, Institut National De La Santé Et De La Recherche Médicale U862Bordeaux, France; NeuroCentre Magendie U862, University of BordeauxBordeaux, France
| | - Brigitte Kieffer
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Centre National de la Recherche Scientifique/Institut National de la Santé et de la Recherche Médicale/Université de Strasbourg Illkirch, France
| | - Louk J M J Vanderschuren
- Division of Behavioural Neuroscience, Department of Animals in Science and Society, Faculty of Veterinary Medicine, Utrecht University Utrecht, Netherlands
| | - Viviana Trezza
- Section of Biomedical Sciences and Technologies, Department of Science, University Roma Tre Rome, Italy
| | - Olivier J J Manzoni
- Institut National De La Santé Et De La Recherche Médicale U901Marseille, France; Université de la Méditerranée UMR S901 Aix-Marseille 2Marseille, France; INMEDMarseille, France
| |
Collapse
|
23
|
Johnson KA, Lovinger DM. Presynaptic G Protein-Coupled Receptors: Gatekeepers of Addiction? Front Cell Neurosci 2016; 10:264. [PMID: 27891077 PMCID: PMC5104741 DOI: 10.3389/fncel.2016.00264] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 10/31/2016] [Indexed: 12/21/2022] Open
Abstract
Drug abuse and addiction cause widespread social and public health problems, and the neurobiology underlying drug actions and drug use and abuse is an area of intensive research. Drugs of abuse alter synaptic transmission, and these actions contribute to acute intoxication as well as the chronic effects of abused substances. Transmission at most mammalian synapses involves neurotransmitter activation of two receptor subtypes, ligand-gated ion channels that mediate fast synaptic responses and G protein-coupled receptors (GPCRs) that have slower neuromodulatory actions. The GPCRs represent a large proportion of neurotransmitter receptors involved in almost all facets of nervous system function. In addition, these receptors are targets for many pharmacotherapeutic agents. Drugs of abuse directly or indirectly affect neuromodulation mediated by GPCRs, with important consequences for intoxication, drug taking and responses to prolonged drug exposure, withdrawal and addiction. Among the GPCRs are several subtypes involved in presynaptic inhibition, most of which are coupled to the Gi/o class of G protein. There is increasing evidence that these presynaptic Gi/o-coupled GPCRs have important roles in the actions of drugs of abuse, as well as behaviors related to these drugs. This topic will be reviewed, with particular emphasis on receptors for three neurotransmitters, Dopamine (DA; D1- and D2-like receptors), Endocannabinoids (eCBs; CB1 receptors) and glutamate (group II metabotropic glutamate (mGlu) receptors). The focus is on recent evidence from laboratory animal models (and some evidence in humans) implicating these receptors in the acute and chronic effects of numerous abused drugs, as well as in the control of drug seeking and taking. The ability of drugs targeting these receptors to modify drug seeking behavior has raised the possibility of using compounds targeting these receptors for addiction pharmacotherapy. This topic is also discussed, with emphasis on development of mGlu2 positive allosteric modulators (PAMs).
Collapse
Affiliation(s)
- Kari A. Johnson
- Section on Synaptic Pharmacology, Laboratory for Integrative Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of HealthBethesda, MD, USA
| | - David M. Lovinger
- Section on Synaptic Pharmacology, Laboratory for Integrative Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of HealthBethesda, MD, USA
| |
Collapse
|
24
|
Faison SL, Schindler CW, Goldberg SR, Wang JB. l-tetrahydropalmatine reduces nicotine self-administration and reinstatement in rats. BMC Pharmacol Toxicol 2016; 17:49. [PMID: 27817750 PMCID: PMC5098281 DOI: 10.1186/s40360-016-0093-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 10/04/2016] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND The negative consequences of nicotine use are well known and documented, however, abstaining from nicotine use and achieving abstinence poses a major challenge for the majority of nicotine users trying to quit. l-Tetrahydropalmatine (l-THP), a compound extracted from the Chinese herb Corydalis, displayed utility in the treatment of cocaine and heroin addiction via reduction of drug-intake and relapse. The present study examined the effects of l-THP on abuse-related effects of nicotine. METHODS Self-administration and reinstatement testing was conducted. Rats trained to self-administer nicotine (0.03 mg/kg/injection) under a fixed-ratio 5 schedule (FR5) of reinforcement were pretreated with l-THP (3 or 5 mg/kg), varenicline (1 mg/kg), bupropion (40 mg/kg), or saline before daily 2-h sessions. Locomotor, food, and microdialysis assays were also conducted in separate rats. RESULTS l-THP significantly reduced nicotine self-administration (SA). l-THP's effect was more pronounced than the effect of varenicline and similar to the effect of bupropion. In reinstatement testing, animals were pretreated with the same compounds, challenged with nicotine (0.3 mg/kg, s.c.), and reintroduced to pre-extinction conditions. l-THP blocked reinstatement of nicotine seeking more effectively than either varenicline or bupropion. Locomotor data revealed that therapeutic doses of l-THP had no inhibitory effects on ambulatory ability and that l-THP (3 and 5 mg/kg) significantly blocked nicotine induced hyperactivity when administered before nicotine. In in-vivo microdialysis experiments, l-THP, varenicline, and bupropion alone elevated extracellular dopamine (DA) levels in the nucleus accumbens shell (nAcb). CONCLUSIONS Since l-THP reduces nicotine taking and blocks relapse it could be a useful alternative to varenicline and bupropion as a treatment for nicotine addiction.
Collapse
Affiliation(s)
- Shamia L. Faison
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, MD USA
- Preclinical Pharmacology Section, Behavioral Neuroscience Research Branch, National Institute on Drug Abuse, National Institutes of Health, DHHS, Baltimore, MD USA
| | - Charles W. Schindler
- Preclinical Pharmacology Section, Behavioral Neuroscience Research Branch, National Institute on Drug Abuse, National Institutes of Health, DHHS, Baltimore, MD USA
| | - Steven R. Goldberg
- Preclinical Pharmacology Section, Behavioral Neuroscience Research Branch, National Institute on Drug Abuse, National Institutes of Health, DHHS, Baltimore, MD USA
| | - Jia Bei Wang
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, MD USA
| |
Collapse
|
25
|
Ghrelin and endocannabinoids participation in morphine-induced effects in the rat nucleus accumbens. Psychopharmacology (Berl) 2016; 233:469-84. [PMID: 26507196 DOI: 10.1007/s00213-015-4119-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2015] [Accepted: 10/15/2015] [Indexed: 12/11/2022]
Abstract
RATIONALE AND OBJECTIVES In addition to dopamine, endocannabinoids are thought to participate in neural reward mechanisms of opioids. Number of recent studies suggests crucial involvement of ghrelin in some addictive drugs effects. Our previous results showed that ghrelin participates in morphine-induced changes in the mesolimbic dopaminergic system associated with reward processing. The goal of the present study was to test whether the growth hormone secretagogue receptor (GHS-R1A) antagonist JMV2959 was able to influence morphine-induced effects on anandamide (N-arachidonoylethanolamine, AEA) and 2-arachidonoylglycerol (2-AG) in the nucleus accumbens shell (NACSh). METHODS We used in vivo microdialysis to determine changes in levels of AEA and 2-AG in the NACSh in rats following (i) an acute morphine dose (5, 10 mg/kg s.c.) with and without JMV2959 pretreatment (3, 6 mg/kg i.p.) or (ii) a morphine challenge dose (5 mg/kg s.c.) with and without JMV2959 (3, 6 mg/kg i.p.) pretreatment, administered during abstinence following repeated doses of morphine (5 days, 10-40 mg/kg). Co-administration of ghrelin (40 ug/kg i.p.) was used to verify the ghrelin mechanisms involvement. RESULTS Pretreatment with JMV2959 significantly and dose-dependently reversed morphine-induced anandamide increases in the NACSh in both the acute and longer-term models, resulting in a significant AEA decrease. JMV2959 significantly intensified acute morphine-induced decreases in accumbens 2-AG levels and attenuated morphine challenge-induced 2-AG decreases. JMV2959 pretreatment significantly reduced concurrent morphine challenge-induced behavioral sensitization. JMV2959 pretreatment effects were abolished by co-administration of ghrelin. CONCLUSIONS Our results indicate significant involvement of ghrelin signaling in morphine-induced endocannabinoid changes in the NACSh.
Collapse
|
26
|
Rygula R, Golebiowska J, Kregiel J, Kubik J, Popik P. Effects of optimism on motivation in rats. Front Behav Neurosci 2015; 9:32. [PMID: 25762910 PMCID: PMC4340205 DOI: 10.3389/fnbeh.2015.00032] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Accepted: 01/30/2015] [Indexed: 11/13/2022] Open
Abstract
In humans, optimism is a cognitive construct related to motivation; optimists exert effort, whereas pessimists disengage from effort. In this study, using a recently developed ambiguous-cue interpretation (ACI) paradigm we took the unique opportunity to investigate whether "optimism" as a trait is correlated with motivation in rodents. In a series of ACI tests (cognitive bias screening, CBS), we identified rats displaying "pessimistic" and "optimistic" traits. Subsequently, we investigated the trait differences in the motivation of these rats to gain reward and to avoid punishment using a progressive ratio (PR) schedule of reinforcement paradigm. Although "optimistic" and "pessimistic" animals did not differ in their motivation to avoid punishment, the "optimistic" rats were significantly more motivated to gain reward than their "pessimistic" conspecifics. For the first time, we showed an association between cognitive judgment bias and motivation in an animal model. Because both investigated processes are closely related to mental health and wellbeing, our results may be valuable for preclinical modeling of many psychiatric disorders.
Collapse
Affiliation(s)
- Rafal Rygula
- Affective Cognitive Neuroscience Lab, Department of Behavioral Neuroscience and Drug Development, Institute of Pharmacology Polish Academy of Sciences Krakow, Poland
| | - Joanna Golebiowska
- Affective Cognitive Neuroscience Lab, Department of Behavioral Neuroscience and Drug Development, Institute of Pharmacology Polish Academy of Sciences Krakow, Poland
| | - Jakub Kregiel
- Affective Cognitive Neuroscience Lab, Department of Behavioral Neuroscience and Drug Development, Institute of Pharmacology Polish Academy of Sciences Krakow, Poland
| | - Jakub Kubik
- Affective Cognitive Neuroscience Lab, Department of Behavioral Neuroscience and Drug Development, Institute of Pharmacology Polish Academy of Sciences Krakow, Poland
| | - Piotr Popik
- Affective Cognitive Neuroscience Lab, Department of Behavioral Neuroscience and Drug Development, Institute of Pharmacology Polish Academy of Sciences Krakow, Poland ; Faculty of Health Sciences, Collegium Medicum, Jagiellonian University Krakow, Poland
| |
Collapse
|
27
|
Moreira FA, Jupp B, Belin D, Dalley JW. Endocannabinoids and striatal function: implications for addiction-related behaviours. Behav Pharmacol 2015; 26:59-72. [PMID: 25369747 PMCID: PMC5398317 DOI: 10.1097/fbp.0000000000000109] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Accepted: 09/26/2014] [Indexed: 12/24/2022]
Abstract
Since the identification and cloning of the major cannabinoid receptor expressed in the brain almost 25 years ago research has highlighted the potential of drugs that target the endocannabinoid system for treating addiction. The endocannabinoids, anandamide and 2-arachidonoyl glycerol, are lipid-derived metabolites found in abundance in the basal ganglia and other brain areas innervated by the mesocorticolimbic dopamine systems. Cannabinoid CB1 receptor antagonists/inverse agonists reduce reinstatement of responding for cocaine, alcohol and opiates in rodents. However, compounds acting on the endocannabinoid system may have broader application in treating drug addiction by ameliorating associated traits and symptoms such as impulsivity and anxiety that perpetuate drug use and interfere with rehabilitation. As a trait, impulsivity is known to predispose to addiction and facilitate the emergence of addiction to stimulant drugs. In contrast, anxiety and elevated stress responses accompany extended drug use and may underlie the persistence of drug intake in dependent individuals. In this article we integrate and discuss recent findings in rodents showing selective pharmacological modulation of impulsivity and anxiety by cannabinoid agents. We highlight the potential of selective inhibitors of endocannabinoid metabolism, directed at fatty acid amide hydrolase and monoacylglycerol lipase, to reduce anxiety and stress responses, and discuss novel mechanisms underlying the modulation of the endocannabinoid system, including the attenuation of impulsivity, anxiety, and drug reward by selective CB2 receptor agonists.
Collapse
Affiliation(s)
- Fabricio A. Moreira
- Department of Pharmacology, Institute of Biological Science, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- Departments of Psychology
| | | | | | - Jeffrey W. Dalley
- Departments of Psychology
- Department of Psychiatry, Addenbrookes’s Hospital University of Cambridge, Cambridge, UK
| |
Collapse
|
28
|
CB1 antagonism: interference with affective properties of acute naloxone-precipitated morphine withdrawal in rats. Psychopharmacology (Berl) 2014; 231:4291-300. [PMID: 24770676 PMCID: PMC4209202 DOI: 10.1007/s00213-014-3575-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Accepted: 04/04/2014] [Indexed: 12/18/2022]
Abstract
RATIONALE Modulation of the endocannabinoid system has been found to interfere with opiate withdrawal. The potential of activation and blockade of the endocannabinoid system to prevent the aversive-affective state of naloxone-precipitated morphine withdrawal (MWD) was investigated in a one-trial conditioned place aversion (CPA) paradigm. OBJECTIVE CPA provides a sensitive measure of the motivational effects of acute MWD. The potential of the fatty acid amide hydrolase (FAAH) inhibitors, URB597 and PF-3845, the CB1 antagonist/inverse agonist, AM251, and the neutral CB1 antagonists, AM4113 and AM6527 (oral), to interfere with establishment of a MWD-induced CPA was investigated. As well, the potential of AM251 and AM4113 to interfere with reinstatement of a previously established MWD-induced CPA was investigated. MATERIALS AND METHODS Using a one-trial place conditioning paradigm, rats were administered naloxone (1 mg/kg, subcutaneous (s.c.)) 24 h after receiving a high dose of morphine (20 mg/kg, s.c.) and were placed on the conditioning floor. To determine the effect of each pretreatment drug on the establishment of the MWD-induced CPA, URB597 (0.3 mg/kg, intraperitoneally (i.p.)), PF-3845 (10 mg/kg, i.p.), AM251 (1 or 2.5 mg/kg, i.p.), AM4113 (1 or 2.5 mg/kg, i.p.), and AM6527 (5 mg/kg, oral) were administered prior to conditioning. RESULTS AM251 (2.5, but not 1 mg/k), AM4113, and AM6527, but not URB597 or PF-3845, interfered with the establishment of the MWD-induced CPA. AM251 and AM4113 did not prevent reinstatement of the CPA. CONCLUSIONS Neutral antagonism of the CB1 receptor reduces the aversive affective properties of morphine withdrawal.
Collapse
|
29
|
Jing L, Qiu Y, Zhang Y, Li JX. Effects of the cannabinoid CB₁ receptor allosteric modulator ORG 27569 on reinstatement of cocaine- and methamphetamine-seeking behavior in rats. Drug Alcohol Depend 2014; 143:251-6. [PMID: 25169627 PMCID: PMC4161648 DOI: 10.1016/j.drugalcdep.2014.08.004] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Revised: 07/31/2014] [Accepted: 08/02/2014] [Indexed: 12/25/2022]
Abstract
BACKGROUND Cannabinoid CB1 receptors play an essential role in drug addiction. Given the side effect profiles of orthosteric CB1 antagonism, new strategies have been attempted to modulate this target, such as CB1 receptor allosteric modulation. However, the effect of CB1 allosteric modulation in drug addiction is unknown. The present study examined the effects of the CB1 receptor allosteric modulator ORG27569 on the reinstatement of cocaine- and methamphetamine-seeking behavior in rats. METHODS Rats were trained to self-administer 0.75 mg/kg cocaine or 0.05 mg/kg methamphetamine in 2-h daily sessions for 14 days which was followed by 7 days of extinction sessions in which rats responded on the levers with no programmed consequences. On reinstatement test sessions, rats were administered ORG27569 (1.0, 3.2, 5.6 mg/kg, i.p.) or SR141716A (3.2 mg/kg, i.p.) 10 min prior to re-exposure to cocaine- or methamphetamine-paired cues or a priming injection of cocaine (10mg/kg, i.p.) or methamphetamine (1mg/kg, i.p.). RESULTS Both cues and a priming injection of cocaine or methamphetamine significantly reinstated the extinguished active lever responding. Pretreatment with ORG27569 resulted in a dose-related attenuation of both cue- and drug-induced reinstatement of cocaine- and methamphetamine-seeking behavior. SR141716A also exhibited similar inhibitory action on reinstatement of drug-seeking behavior. CONCLUSION Negative allosteric modulation of CB1 receptors can produce similar functional antagonism as orthosteric CB1 receptor antagonists on reinstatement of drug-seeking behavior. Thus, ORG27569 or other negative allosteric modulators deserve further study as potentially effective pharmacotherapy for drug addiction.
Collapse
Affiliation(s)
- Li Jing
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, New York, USA
,Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Yanyan Qiu
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, New York, USA
| | - Yanan Zhang
- Research Triangle Institute, Research Triangle Park, North Carolina, USA
| | - Jun-Xu Li
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, NY, USA.
| |
Collapse
|
30
|
Le Foll B, Pushparaj A, Pryslawsky Y, Forget B, Vemuri K, Makriyannis A, Trigo JM. Translational strategies for therapeutic development in nicotine addiction: rethinking the conventional bench to bedside approach. Prog Neuropsychopharmacol Biol Psychiatry 2014; 52:86-93. [PMID: 24140878 PMCID: PMC4002666 DOI: 10.1016/j.pnpbp.2013.10.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2013] [Revised: 10/08/2013] [Accepted: 10/09/2013] [Indexed: 12/16/2022]
Abstract
Tobacco produces an impressive burden of disease resulting in premature death in half of users. Despite effective smoking cessation medications (nicotine replacement therapies, bupropion and varenicline), there is a very high rate of relapse following quit attempts. The use of efficient strategies for the development of novel treatments is a necessity. A 'bench to bedside strategy' was initially used to develop cannabinoid CB1 receptor antagonists for the treatment of nicotine addiction. Unfortunately, after being tested on experimental animals, what seemed to be an interesting approach for the treatment of nicotine addiction resulted in serious unwanted side effects when tested in humans. Current research is focusing again on pre-clinical models in an effort to eliminate unwanted side effects while preserving the initially observed efficacy. A 'bed side to bench strategy' was used to study the role of the insula (part of the frontal cortex) in nicotine addiction. This line of research started based on clinical observations that patients suffering stroke-induced lesions to the insula showed a greater likelihood to report immediate smoking cessation without craving or relapse. Subsequently, animal models of addiction are used to explore the role of insula in addiction. Due to the inherent limitations existing in clinical versus preclinical studies, the possibility of close interaction between both models seems to be critical for the successful development of novel therapeutic strategies for nicotine dependence.
Collapse
Affiliation(s)
- Bernard Le Foll
- Translational Addiction Research Laboratory, Centre for Addiction and Mental Health, 33 Russell Street, Toronto, ON M5S 2S1, Canada; Alcohol Research and Treatment Clinic, Addiction Medicine Services, Ambulatory Care and Structured Treatments, Centre for Addiction and Mental Health, Toronto, ON, Canada; Department of Family and Community Medicine, University of Toronto, Toronto, Canada; Department of Pharmacology and Toxicology, University of Toronto, Toronto, Canada; Department of Psychiatry and Institute of Medical Sciences, University of Toronto, Toronto, Canada.
| | - Abhiram Pushparaj
- Translational Addiction Research Laboratory, Centre for Addiction and Mental Health, 33 Russell Street, Toronto, ON M5S 2S1, Canada
| | - Yaroslaw Pryslawsky
- Translational Addiction Research Laboratory, Centre for Addiction and Mental Health, 33 Russell Street, Toronto, ON M5S 2S1, Canada
| | - Benoit Forget
- Integrative Neurobiology of Cholinergic Systems, Department of Neuroscience, Pasteur Institute, 25 rue du Dr. Roux, Paris 75724, France
| | - Kiran Vemuri
- Center for Drug Discovery, Northeastern University, Boston, MA 02115-5005, United States; Department of Pharmaceutical Sciences, Northeastern University, Boston, MA 02115-5005, United States; Department of Chemistry and Chemical Biology, Northeastern University, Boston, MA 02115-5005, United States
| | - Alexandros Makriyannis
- Center for Drug Discovery, Northeastern University, Boston, MA 02115-5005, United States; Department of Pharmaceutical Sciences, Northeastern University, Boston, MA 02115-5005, United States; Department of Chemistry and Chemical Biology, Northeastern University, Boston, MA 02115-5005, United States
| | - Jose M Trigo
- Translational Addiction Research Laboratory, Centre for Addiction and Mental Health, 33 Russell Street, Toronto, ON M5S 2S1, Canada
| |
Collapse
|
31
|
Nader J, Rapino C, Gennequin B, Chavant F, Francheteau M, Makriyannis A, Duranti A, Maccarrone M, Solinas M, Thiriet N. Prior stimulation of the endocannabinoid system prevents methamphetamine-induced dopaminergic neurotoxicity in the striatum through activation of CB2 receptors. Neuropharmacology 2014; 87:214-21. [PMID: 24709540 DOI: 10.1016/j.neuropharm.2014.03.014] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Revised: 03/19/2014] [Accepted: 03/27/2014] [Indexed: 12/30/2022]
Abstract
Methamphetamine toxicity is associated with cell death and loss of dopamine neuron terminals in the striatum similar to what is found in some neurodegenerative diseases. Conversely, the endocannabinoid system (ECS) has been suggested to be neuroprotective in the brain, and new pharmacological tools have been developed to increase their endogenous tone. In this study, we evaluated whether ECS stimulation could reduce the neurotoxicity of high doses of methamphetamine on the dopamine system. We found that methamphetamine alters the levels of the major endocannabinoids, anandamide (AEA) and 2-arachidonoyl glycerol (2-AG) in the striatum, suggesting that the ECS participates in the brain responses to methamphetamine. Δ(9)-tetrahydrocannabinol (THC), a cannabis-derived agonist of both CB1 and CB2 cannabinoid receptors, or inhibitors of the main enzymes responsible for the degradation of AEA and 2-AG (URB597 and JZL184, respectively), blunted the decrease in striatal protein levels of tyrosine hydroxylase induced by methamphetamine. In addition, antagonists of CB2, but not of CB1, blocked the preventive effects of URB597 and JZL184, suggesting that only the former receptor subtype is engaged in neuroprotection exerted by ECS stimulation. Finally, we found that methamphetamine increases striatal levels of the cytokine tumor necrosis factor alpha, an effect that was blocked by ECS stimulation. Altogether, our results indicate that stimulation of ECS prior to the administration of an overdose of methamphetamine considerably reduces the neurotoxicity of the drug through CB2 receptor activation and highlight a protective function for the ECS against the toxicity induced by drugs and other external insults to the brain. This article is part of the Special Issue entitled 'CNS Stimulants'.
Collapse
MESH Headings
- Animals
- Arachidonic Acids/metabolism
- Benzamides/pharmacology
- Benzodioxoles/pharmacology
- Cannabinoid Receptor Modulators/pharmacology
- Carbamates/pharmacology
- Central Nervous System Stimulants/toxicity
- Dronabinol/pharmacology
- Endocannabinoids/metabolism
- Enzyme Inhibitors/pharmacology
- Glycerides/metabolism
- Male
- Methamphetamine/toxicity
- Mice, Inbred C57BL
- Neostriatum/drug effects
- Neostriatum/metabolism
- Neurotoxicity Syndromes/metabolism
- Neurotoxicity Syndromes/prevention & control
- Piperidines/pharmacology
- Polyunsaturated Alkamides/metabolism
- Random Allocation
- Receptor, Cannabinoid, CB1/agonists
- Receptor, Cannabinoid, CB1/antagonists & inhibitors
- Receptor, Cannabinoid, CB1/metabolism
- Receptor, Cannabinoid, CB2/agonists
- Receptor, Cannabinoid, CB2/antagonists & inhibitors
- Receptor, Cannabinoid, CB2/metabolism
- Tumor Necrosis Factor-alpha/metabolism
- Tyrosine 3-Monooxygenase/metabolism
Collapse
Affiliation(s)
- Joëlle Nader
- INSERM, U1084, Experimental and Clinical Neurosciences Laboratory, Neurobiology and Neuropharmacology of Addiction, F-86022 Poitiers, France; University of Poitiers, U1084, F-86022 Poitiers, France
| | - Cinzia Rapino
- Faculty of Veterinary Medicine, University of Teramo, Teramo, Italy
| | - Benjamin Gennequin
- INSERM, U1084, Experimental and Clinical Neurosciences Laboratory, Neurobiology and Neuropharmacology of Addiction, F-86022 Poitiers, France; University of Poitiers, U1084, F-86022 Poitiers, France
| | - Francois Chavant
- University of Poitiers, U1084, F-86022 Poitiers, France; Pharmacology Department, Poitiers University Hospital, Poitiers, France
| | - Maureen Francheteau
- INSERM, U1084, Experimental and Clinical Neurosciences Laboratory, Neurobiology and Neuropharmacology of Addiction, F-86022 Poitiers, France; University of Poitiers, U1084, F-86022 Poitiers, France
| | - Alexandros Makriyannis
- Center for Drug Discovery, Department of Pharmaceutical Sciences, and Chemistry and Chemical Biology, Northeastern University, Boston, USA
| | - Andrea Duranti
- Dipartimento di Scienze Biomolecolari, Università degli Studi di Urbino "Carlo Bo", Urbino, Italy
| | - Mauro Maccarrone
- Center of Integrated Research, Campus Bio-Medico University of Rome, Via Alvaro del Portillo 21, 00128 Rome, Italy; European Center for Brain Research/IRCCS Santa Lucia Foundation, Via del Fosso di Fiorano 35, 00146 Rome, Italy.
| | - Marcello Solinas
- INSERM, U1084, Experimental and Clinical Neurosciences Laboratory, Neurobiology and Neuropharmacology of Addiction, F-86022 Poitiers, France; University of Poitiers, U1084, F-86022 Poitiers, France
| | - Nathalie Thiriet
- INSERM, U1084, Experimental and Clinical Neurosciences Laboratory, Neurobiology and Neuropharmacology of Addiction, F-86022 Poitiers, France; University of Poitiers, U1084, F-86022 Poitiers, France.
| |
Collapse
|
32
|
The vital role of constitutive GPCR activity in the mesolimbic dopamine system. Transl Psychiatry 2014; 4:e361. [PMID: 24518399 PMCID: PMC3944632 DOI: 10.1038/tp.2013.130] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2013] [Revised: 11/13/2013] [Accepted: 12/07/2013] [Indexed: 12/28/2022] Open
Abstract
The midbrain dopamine system has an important role in processing rewards and the stimuli associated with them, and is implicated in various psychiatric disorders. This system is tightly regulated by various G protein-coupled receptors (GPCRs). It is becoming increasingly clear that these receptors are not only activated by (endogenous) agonists but that they also exhibit agonist-independent intrinsic constitutive activity. In this review we highlight the evidence for the physiological role of such constitutive GPCR activity (in particular for cannabinoid 1, serotonin 2C and mu-opioid receptors) in the ventral tegmental area and in its output regions like the nucleus accumbens. We also address the behavioral relevance of constitutive GPCR signaling and discuss the repercussions of its abolition in dopamine-related psychiatric diseases.
Collapse
|
33
|
De Luca MA, Valentini V, Bimpisidis Z, Cacciapaglia F, Caboni P, Di Chiara G. Endocannabinoid 2-Arachidonoylglycerol Self-Administration by Sprague-Dawley Rats and Stimulation of in vivo Dopamine Transmission in the Nucleus Accumbens Shell. Front Psychiatry 2014; 5:140. [PMID: 25368584 PMCID: PMC4201088 DOI: 10.3389/fpsyt.2014.00140] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2014] [Accepted: 09/23/2014] [Indexed: 01/09/2023] Open
Abstract
2-Arachidonoylglycerol (2-AG) is the most potent endogenous ligand of brain cannabinoid CB1 receptors and is synthesized on demand from 2-arachidonate-containing phosphoinositides by the action of diacylglycerol lipase in response to increased intracellular calcium. Several studies indicate that the endocannabinoid (eCB) system is involved in the mechanism of reward and that diverse drugs of abuse increase brain eCB levels. In addition, eCB are self-administered (SA) by squirrel monkeys, and anandamide increases nucleus accumbens (NAc) shell dopamine (DA) in rats. To date, there is no evidence on the reinforcing effects of 2-AG and its effects on DA transmission in rodents. In order to fill this gap, we studied intravenous 2-AG SA and monitored the effect of 2-AG on extracellular DA in the NAc shell and core via microdialysis in male Sprague-Dawley rats. Rats were implanted with jugular catheters and trained to self-administer 2-AG [25 mg/kg/inf intravenously (iv)] in single daily 1 h sessions for 5 weeks under initial fixed ratio (FR) 1 schedule. The ratio was subsequently increased to FR2. Active nose poking increased from the 6th SA session (acquisition phase) but no significant increase of nose pokes was observed after FR2. When 2-AG was substituted for vehicle (25th SA session, extinction phase), rate responding as well as number of injections slowly decreased. When vehicle was replaced with 2-AG, SA behavior immediately recovered (reacquisition phase). The reinforcing effects of 2-AG in SA behavior were fully blocked by the CB1 receptor inverse agonist/antagonist rimonabant (1 mg/kg intraperitoneally, 30 min before SA session). In the microdialysis studies, we observed that 2-AG (0.1-1.0 mg/kg iv) preferentially stimulates NAc shell as compared to the NAc core. NAc shell DA increased by about 25% over basal value at the highest doses tested (0.5 and 1.0 mg/kg iv). The results obtained suggest that the eCB system, via 2-AG, plays an important role in reward.
Collapse
Affiliation(s)
- Maria Antonietta De Luca
- Neuropsychopharmacology Section, Department of Biomedical Sciences, University of Cagliari , Cagliari , Italy ; National Institute of Neuroscience (INN) , Cagliari , Italy
| | - Valentina Valentini
- Neuropsychopharmacology Section, Department of Biomedical Sciences, University of Cagliari , Cagliari , Italy ; National Institute of Neuroscience (INN) , Cagliari , Italy ; Centre of Excellence for Studies on the Neurobiology of Addiction , Cagliari , Italy
| | - Zisis Bimpisidis
- Neuropsychopharmacology Section, Department of Biomedical Sciences, University of Cagliari , Cagliari , Italy
| | - Fabio Cacciapaglia
- Neuropsychopharmacology Section, Department of Biomedical Sciences, University of Cagliari , Cagliari , Italy
| | - Pierluigi Caboni
- Department of Life and Environmental Sciences, University of Cagliari , Cagliari , Italy
| | - Gaetano Di Chiara
- Neuropsychopharmacology Section, Department of Biomedical Sciences, University of Cagliari , Cagliari , Italy ; National Institute of Neuroscience (INN) , Cagliari , Italy ; Centre of Excellence for Studies on the Neurobiology of Addiction , Cagliari , Italy ; Cagliari Section, Neuroscience Institute, National Research Council of Italy , Cagliari , Italy
| |
Collapse
|
34
|
Justinova Z, Mascia P, Wu HQ, Secci ME, Redhi GH, Panlilio LV, Scherma M, Barnes C, Parashos A, Zara T, Fratta W, Solinas M, Pistis M, Bergman J, Kangas BD, Ferré S, Tanda G, Schwarcz R, Goldberg SR. Reducing cannabinoid abuse and preventing relapse by enhancing endogenous brain levels of kynurenic acid. Nat Neurosci 2013; 16:1652-61. [PMID: 24121737 PMCID: PMC3835353 DOI: 10.1038/nn.3540] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2013] [Accepted: 09/11/2013] [Indexed: 02/06/2023]
Abstract
In the reward circuitry of the brain, α-7-nicotinic acetylcholine receptors (α7nAChRs) modulate effects of Δ(9)-tetrahydrocannabinol (THC), marijuana's main psychoactive ingredient. Kynurenic acid (KYNA) is an endogenous negative allosteric modulator of α7nAChRs. Here we report that the kynurenine 3-monooxygenase (KMO) inhibitor Ro 61-8048 increases brain KYNA levels and attenuates cannabinoid-induced increases in extracellular dopamine in reward-related brain areas. In the self-administration model of drug abuse, Ro 61-8048 reduced the rewarding effects of THC and the synthetic cannabinoid WIN 55,212-2 in squirrel monkeys and rats, respectively, and it also prevented relapse to drug-seeking induced by reexposure to cannabinoids or cannabinoid-associated cues. The effects of enhancing endogenous KYNA levels with Ro 61-8048 were prevented by positive allosteric modulators of α7nAChRs. Despite a clear need, there are no medications approved for treatment of marijuana dependence. Modulation of KYNA offers a pharmacological strategy for achieving abstinence from marijuana and preventing relapse.
Collapse
Affiliation(s)
- Zuzana Justinova
- 1] Preclinical Pharmacology Section, Behavioral Neuroscience Research Branch, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Department of Health and Human Services, Baltimore, Maryland, USA. [2] Department of Psychiatry, Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore, Maryland, USA. [3]
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Interactions between Δ(9)-tetrahydrocannabinol and heroin: self-administration in rhesus monkeys. Behav Pharmacol 2013; 23:754-61. [PMID: 23044830 DOI: 10.1097/fbp.0b013e32835a3907] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The cannabinoid receptor agonist Δ(9)-tetrahydrocannabinol (THC) enhances the antinociceptive effects of µ-opioid receptor agonists, raising the possibility of using a combination of THC and opioids for treating pain. This study examined the effects of noncontingent and contingent administration of THC on intravenous heroin self-administration in rhesus monkeys. Self-administration of different unit doses of heroin (0.0001-0.1 mg/kg/infusion) generated a typical inverted U-shaped dose-response curve. In one experiment (n=4), noncontingent THC (0.1-1.0 mg/kg) dose dependently shifted the heroin dose-response curve downward in three monkeys and slightly leftward in one monkey. In a second experiment (n=4), monkeys could self-administer THC alone (0.0032-0.032 mg/kg/infusion), heroin alone, or a mixture of THC and heroin. THC alone did not maintain responding above that obtained with saline; however, increasing the THC dose with heroin dose dependently decreased the number of infusions received and the rate of responding, as compared with data that were obtained with heroin alone. These results indicate that THC does not significantly enhance the positive reinforcing effects of heroin, further supporting the view that combining cannabinoid and opioid receptor agonists (e.g. for treating pain) does not increase, and might decrease, the abuse liability of individual drugs.
Collapse
|
36
|
Maguire DR, Yang W, France CP. Interactions between μ-opioid receptor agonists and cannabinoid receptor agonists in rhesus monkeys: antinociception, drug discrimination, and drug self-administration. J Pharmacol Exp Ther 2013; 345:354-62. [PMID: 23536317 DOI: 10.1124/jpet.113.204099] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Cannabinoid receptor agonists enhance the antinociceptive effects of μ-opioid receptor agonists, which suggests that combinations of these drugs might enhance therapeutic effectiveness (e.g., analgesia). However, it is not clear whether combinations of these drugs also enhance abuse or dependence liability. This experiment examined whether combinations of cannabinoids and opioids that enhance antinociception also increase abuse-related effects by studying the effects of the cannabinoid receptor agonists 2-[(1R,2R,5R)-5-hydroxy-2-(3-hydroxypropyl)cyclohexyl]-5-(2-methyloctan-2-yl)phenol (CP 55,940) and (R)-(+)-[2,3-dihydro-5-methyl-3-(4-morpholinylmethyl)pyrrolo[1,2,3-de]-1,4-benzoxazin-6-yl]-1-naphthalenylmethanone mesylate (WIN 55,212) on the antinociceptive, discriminative stimulus, and positive reinforcing effects of μ-opioid receptor agonists in rhesus monkeys. In one group of monkeys (n = 3), morphine (0.1-5.6 mg/kg s.c.), CP 55,940 (0.0032-0.032 mg/kg s.c.), and WIN 55,212 (0.1-1.0 mg/kg s.c.) dose-dependently increased tail withdrawal latency from 50°C water, and pretreatment with small, otherwise ineffective, doses of CP 55,940 and WIN 55,212 shifted the morphine dose-effect curve to the left. In monkeys (n = 3) discriminating 3.2 mg/kg morphine, CP 55,940 (0.01-0.032 mg/kg s.c.) and WIN 55,212 (0.1-1.78 mg/kg s.c.) attenuated the discriminative stimulus effects of morphine, shifting the dose-effect curve to the right. In monkeys (n = 4) self-administering heroin (0.32-32.0 µg/kg/infusion i.v.), CP 55,940 (0.001-0.032 mg/kg s.c.), and WIN 55,212 (0.1-1.0 mg/kg s.c.) shifted the heroin dose-effect curve rightward and downward. Cannabinoid receptor agonists CP 55,940 and WIN 55,212 enhanced the antinociceptive effects but not the discriminative stimulus or positive reinforcing effects of μ-opioid receptor agonists in rhesus monkeys, supporting the view that combining cannabinoid and opioid receptor agonists might result in enhanced treatment effectiveness for pain without similarly enhancing abuse and dependence liability.
Collapse
Affiliation(s)
- David R Maguire
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Dr., San Antonio, TX 78229-3900, USA
| | | | | |
Collapse
|
37
|
Panlilio LV, Justinova Z, Goldberg SR. Inhibition of FAAH and activation of PPAR: new approaches to the treatment of cognitive dysfunction and drug addiction. Pharmacol Ther 2013; 138:84-102. [PMID: 23333350 DOI: 10.1016/j.pharmthera.2013.01.003] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Accepted: 12/21/2012] [Indexed: 12/16/2022]
Abstract
Enhancing the effects of endogenously-released cannabinoid ligands in the brain might provide therapeutic effects more safely and effectively than administering drugs that act directly at the cannabinoid receptor. Inhibitors of fatty acid amide hydrolase (FAAH) prevent the breakdown of endogenous ligands for cannabinoid receptors and peroxisome proliferator-activated receptors (PPAR), prolonging and enhancing the effects of these ligands when they are naturally released. This review considers recent research on the effects of FAAH inhibitors and PPAR activators in animal models of addiction and cognition (specifically learning and memory). These studies show that FAAH inhibitors can produce potentially therapeutic effects, some through cannabinoid receptors and some through PPAR. These effects include enhancing certain forms of learning, counteracting the rewarding effects of nicotine and alcohol, relieving symptoms of withdrawal from cannabis and other drugs, and protecting against relapse-like reinstatement of drug self-administration. Since FAAH inhibition might have a wide range of therapeutic actions but might also share some of the adverse effects of cannabis, it is noteworthy that at least one FAAH-inhibiting drug (URB597) has been found to have potentially beneficial effects but no indication of liability for abuse or dependence. Although these areas of research are new, the preliminary evidence indicates that they might lead to improved therapeutic interventions and a better understanding of the brain mechanisms underlying addiction and memory.
Collapse
Affiliation(s)
- Leigh V Panlilio
- Preclinical Pharmacology Section, Behavioral Neuroscience Research Branch, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Department of Health and Human Services, Baltimore, MD 21224, USA
| | | | | |
Collapse
|
38
|
Possible involvement of endocannabinoids in the increase of morphine consumption in maternally deprived rat. Neuropharmacology 2012; 65:193-9. [PMID: 23089638 DOI: 10.1016/j.neuropharm.2012.10.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2012] [Revised: 10/11/2012] [Accepted: 10/13/2012] [Indexed: 11/21/2022]
Abstract
Whether adolescent exposure to chronic delta-9-tetrahydrocannabinol (THC) facilitates progression to opioid consumption is still controversial. In a maternal deprivation model (3 h daily from postnatal day 1-14), we previously reported that adolescent exposure to chronic THC blocks morphine dependence in maternally deprived (D) rats. Owing to the existence of a functional cross-interaction between the opioid and cannabinoid systems in reward, we evaluated if the vulnerability to opiate reward in D rats, may involve an alteration of the endocannabinoid system. Anandamide and 2-arachidonoylglycerol (2-AG), were quantified in the striatum and mesencephalon of adolescent and adult D and non-deprived (animal facility rearing, AFR) rats by isotope dilution liquid chromatography-mass spectrometry. Oral morphine self-administration behavior was analyzed for 14 weeks, 24 days after chronic injection of the cannabinoid CB1 receptor antagonist/inverse agonist, SR141716A (3 mg/kg) for 2 weeks during adolescence (PND 35-48). Adolescent D rats exhibited higher basal levels of anandamide than adolescent AFR rats in the nucleus accumbens (38%), the caudate-putamen nucleus (62%) and the mesencephalon (320%), whereas adult D rats showed an increase of anandamide and 2-AG levels in the nucleus accumbens (50% and 24%, respectively) and of 2-AG in the caudate-putamen nucleus (48%), compared to adult AFR rats. Chronic administration of SR141716A to adolescent D rats blocked the escalation behavior in the morphine consumption test. Our data suggest that altered brain endocannabinoid levels may contribute to the escalation behavior in the morphine consumption test in a maternal deprivation model.
Collapse
|
39
|
Chauvet C, Goldberg SR, Jaber M, Solinas M. Effects of environmental enrichment on the incubation of cocaine craving. Neuropharmacology 2012; 63:635-41. [PMID: 22634364 PMCID: PMC3392425 DOI: 10.1016/j.neuropharm.2012.05.014] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2012] [Revised: 04/13/2012] [Accepted: 05/13/2012] [Indexed: 11/23/2022]
Abstract
Recent studies have demonstrated that exposure to environmental enrichment (EE) during withdrawal periods reduces the risks of relapse to drug-seeking behavior. In this study, we investigated whether EE could prevent the development of time-dependent increases in cocaine-seeking behavior (incubation of craving). In addition, we investigated whether EE could eliminate already developed incubation and whether the effects of EE would last when enrichment is discontinued. For this, we allowed rats to self-administer cocaine for 10 daily 6 h sessions and measured cocaine-seeking 1, 30 and 60 days after the last self-administration session. In between these tests, rats were kept in forced abstinence and housed either in EE or standard environments (SE). Between day 30 and 60 of withdrawal, half of the rats in each group were maintained in their original environmental condition and the other half was switched to the other environmental condition. We found that exposure to EE prevents development of incubation of cocaine craving and eliminates already developed incubation. In addition, contrary to our expectations, when EE was discontinued, its positive effects on incubation of craving disappeared. These results indicate that EE can reduce cocaine seeking but only temporarily and questions the hypothesis that EE can permanently eliminate the neural consequences of exposure to drugs of abuse. Therefore, stimulating environments could have positive effects on the treatment of cocaine addiction only if they are maintained for long periods of abstinence that encompass the time-frame during which addicts are most vulnerable to relapse.
Collapse
Affiliation(s)
- Claudia Chauvet
- University of Poitiers; 1 Rue George Bonnet, 86022, Poitiers-FRANCE
| | - Steven R. Goldberg
- Preclinical Pharmacology Section, Behavioral Neuroscience Research Branch, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Department of Health and Human Services, Baltimore, Maryland 21224
| | - Mohamed Jaber
- University of Poitiers; 1 Rue George Bonnet, 86022, Poitiers-FRANCE
- INSERM U-1084, Experimental and Clinical Neurosciences Laboratory; Poitiers-FRANCE
| | - Marcello Solinas
- University of Poitiers; 1 Rue George Bonnet, 86022, Poitiers-FRANCE
- INSERM U-1084, Experimental and Clinical Neurosciences Laboratory; Poitiers-FRANCE
| |
Collapse
|
40
|
Oleson EB, Beckert MV, Morra JT, Lansink CS, Cachope R, Abdullah RA, Loriaux AL, Schetters D, Pattij T, Roitman MF, Lichtman AH, Cheer JF. Endocannabinoids shape accumbal encoding of cue-motivated behavior via CB1 receptor activation in the ventral tegmentum. Neuron 2012; 73:360-73. [PMID: 22284189 DOI: 10.1016/j.neuron.2011.11.018] [Citation(s) in RCA: 121] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/15/2011] [Indexed: 01/01/2023]
Abstract
Transient increases in nucleus accumbens (NAc) dopamine concentration are observed when animals are presented with motivationally salient stimuli and are theorized to energize reward seeking. They arise from high-frequency firing of dopamine neurons in the ventral tegmental area (VTA), which also results in the release of endocannabinoids from dopamine cell bodies. In this context, endocannabinoids are thought to regulate reward seeking by modulating dopamine signaling, although a direct link has never been demonstrated. To test this, we pharmacologically manipulated endocannabinoid neurotransmission in the VTA while measuring transient changes in dopamine concentration in the NAc during reward seeking. Disrupting endocannabinoid signaling dramatically reduced, whereas augmenting levels of the endocannabinoid 2-arachidonoylglycerol (2AG) increased, cue-evoked dopamine concentrations and reward seeking. These data suggest that 2AG in the VTA regulates reward seeking by sculpting ethologically relevant patterns of dopamine release during reward-directed behavior.
Collapse
Affiliation(s)
- Erik B Oleson
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Antypa M, Faux C, Eichele G, Parnavelas JG, Andrews WD. Differential gene expression in migratory streams of cortical interneurons. Eur J Neurosci 2012; 34:1584-94. [PMID: 22103416 PMCID: PMC3401901 DOI: 10.1111/j.1460-9568.2011.07896.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Cortical interneurons originate in the ganglionic eminences of the subpallium and migrate into the cortex in well-defined tangential streams. At the start of corticogenesis, two streams of migrating neurons are evident: a superficial one at the level of the preplate (PPL), and a deeper one at the level of the intermediate zone (IZ). Currently, little is known about the signalling mechanisms that regulate interneuron migration, and almost nothing is known about the molecules that may be involved in their choice of migratory stream. Here, we performed a microarray analysis, comparing the changes in gene expression between cells migrating in the PPL and those migrating in the IZ at embryonic day 13.5. This analysis identified genes, many of them novel, that were upregulated in one of the two streams. Moreover, polymerase chain reaction, in situ hybridization experiments and immunohistochemistry showed the expression of these genes in interneurons migrating within the PPL or IZ, suggesting that they play a role in their migration and choice of stream.
Collapse
Affiliation(s)
- Mary Antypa
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
| | | | | | | | | |
Collapse
|
42
|
Janero DR. Cannabinoid-1 receptor (CB1R) blockers as medicines: beyond obesity and cardiometabolic disorders to substance abuse/drug addiction with CB1R neutral antagonists. Expert Opin Emerg Drugs 2012; 17:17-29. [DOI: 10.1517/14728214.2012.660916] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
43
|
Fattore L, Spano M, Melis V, Fadda P, Fratta W. Differential effect of opioid and cannabinoid receptor blockade on heroin-seeking reinstatement and cannabinoid substitution in heroin-abstinent rats. Br J Pharmacol 2012; 163:1550-62. [PMID: 21518339 DOI: 10.1111/j.1476-5381.2011.01459.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND AND PURPOSE Opioids and cannabinoids interact in drug addiction and relapse. We investigated the effect of the opioid receptor antagonist naloxone and/or the cannabinoid CB(1) receptor antagonist rimonabant on cannabinoid-induced reinstatement of heroin seeking and on cannabinoid substitution in heroin-abstinent rats. EXPERIMENTAL APPROACH Rats were trained to self-administer heroin (30 µg·kg(-1) per infusion) under a fixed-ratio 1 reinforcement schedule. After extinction of self-administration (SA) behaviour, we confirmed the effect of naloxone (0.1-1 mg·kg(-1)) and rimonabant (0.3-3 mg·kg(-1)) on the reinstatement of heroin seeking induced by priming with the CB(1) receptor agonist WIN55,212-2 (WIN, 0.15-0.3 mg·kg(-1)). Then, in a parallel set of heroin-trained rats, we evaluated whether WIN (12.5 µg·kg(-1) per infusion) SA substituted for heroin SA after different periods of extinction. In groups of rats in which substitution occurred, we studied the effect of both antagonists on cannabinoid intake. KEY RESULTS Cannabinoid-induced reinstatement of heroin seeking was significantly attenuated by naloxone (1 mg·kg(-1)) and rimonabant (3 mg·kg(-1)) and fully blocked by co-administration of sub-threshold doses of the two antagonists. Moreover, contrary to immediate (1 day) or delayed (90 days) drug substitution, rats readily self-administered WIN when access was given after 7, 14 or 21 days of extinction from heroin, and showed a response rate that was positively correlated with the extinction period. In these animals, cannabinoid intake was increased by naloxone (1 mg·kg(-1)) and decreased by rimonabant (3 mg·kg(-1)). CONCLUSIONS AND IMPLICATIONS Our findings extend previous research on the crosstalk between cannabinoid and opioid receptors in relapse mechanisms, which suggests a differential role in heroin-seeking reinstatement and cannabinoid substitution in heroin-abstinent rats.
Collapse
Affiliation(s)
- L Fattore
- Institute of Neuroscience-Cagliari, CNR National Research Council of Italy, Cagliari, Italy.
| | | | | | | | | |
Collapse
|
44
|
Järbe TUC, LeMay BJ, Vemuri VK, Vadivel SK, Zvonok A, Makriyannis A. Central mediation and differential blockade by cannabinergics of the discriminative stimulus effects of the cannabinoid CB1 receptor antagonist rimonabant in rats. Psychopharmacology (Berl) 2011; 216:355-65. [PMID: 21369753 PMCID: PMC3727221 DOI: 10.1007/s00213-011-2226-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2010] [Accepted: 02/06/2011] [Indexed: 10/18/2022]
Abstract
RATIONALE Discovery of an endocannabinoid signaling system launched the development of the blocker rimonabant, a cannabinoid CB1 receptor (CB(1)R) antagonist/inverse agonist. Due to untoward effects, this medication was withdrawn and efforts have been directed towards discovering chemicals with more benign profiles. OBJECTIVE This study aims to comparatively evaluate new ligands using a rimonabant discriminated drinking aversion procedure. METHODS Rats discriminated between rimonabant (5.6 mg/kg) and vehicle. The 30 min saccharin (0.1%) drinking after rimonabant pretreatment was followed by injection of lithium chloride (120 mg/kg) in the experimental animals. After vehicle pretreatment, experimental animals were given i.p. NaCl (10 ml/kg). Postdrinking treatment for controls was NaCl, irrespective of pretreatment condition (rimonabant or vehicle). RESULTS The centrally acting neutral CB(1)R antagonist AM4113, but not the limited brain penetrating CB(1)R neutral antagonist AM6545, substituted for rimonabant. The CB(1)R agonists THC (1-10 mg/kg), AM1346 (1-10 mg/kg) did not substitute. The rimonabant-induced conditioned suppression of saccharin drinking was attenuated when CB(1)R agonists AM5983 (0.01-1 mg/kg) and THC (10 mg/kg), but not the CB(1)R agonist AM1346 (0.1-18 mg/kg), were combined with rimonabant (5.6 mg/kg). By varying the injection-to-test interval, we gauged the relative duration of the cueing effects of rimonabant, and the in vivo functional half-life was estimated to be approximately 1.5 h. CONCLUSION A neutral CB(1)R antagonist (AM4113) produced cueing effects similar to those of rimonabant and generalization likely was centrally mediated. The functional cueing effects of rimonabant are relatively short-acting, pharmacologically selective, and differentially blocked by cannabinergics.
Collapse
|
45
|
Serrano A, Parsons LH. Endocannabinoid influence in drug reinforcement, dependence and addiction-related behaviors. Pharmacol Ther 2011; 132:215-41. [PMID: 21798285 DOI: 10.1016/j.pharmthera.2011.06.005] [Citation(s) in RCA: 140] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2011] [Accepted: 06/17/2011] [Indexed: 12/12/2022]
Abstract
The endogenous cannabinoid system is an important regulatory system involved in physiological homeostasis. Endocannabinoid signaling is known to modulate neural development, immune function, metabolism, synaptic plasticity and emotional state. Accumulating evidence also implicates brain endocannabinoid signaling in the etiology of drug addiction which is characterized by compulsive drug seeking, loss of control in limiting drug intake, emergence of a negative emotional state in the absence of drug use and a persistent vulnerability toward relapse to drug use during protracted abstinence. In this review we discuss the effects of drug intake on brain endocannabinoid signaling, evidence implicating the endocannabinoid system in the motivation for drug consumption, and drug-induced alterations in endocannabinoid function that may contribute to various aspects of addiction including dysregulated synaptic plasticity, increased stress responsivity, negative affective states, drug craving and relapse to drug taking. Current knowledge of genetic variants in endocannabinoid signaling associated with addiction is also discussed.
Collapse
Affiliation(s)
- Antonia Serrano
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA 92037, USA
| | | |
Collapse
|
46
|
Ward SJ, Raffa RB. Rimonabant redux and strategies to improve the future outlook of CB1 receptor neutral-antagonist/inverse-agonist therapies. Obesity (Silver Spring) 2011; 19:1325-34. [PMID: 21475141 DOI: 10.1038/oby.2011.69] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Affiliation(s)
- Sara Jane Ward
- Department of Pharmaceutical Sciences, Temple University School of Pharmacy, Philadelphia, Pennsylvania, USA.
| | | |
Collapse
|
47
|
Miller LL, Picker MJ, Schmidt KT, Dykstra LA. Effects of morphine on pain-elicited and pain-suppressed behavior in CB1 knockout and wildtype mice. Psychopharmacology (Berl) 2011; 215:455-65. [PMID: 21373789 PMCID: PMC3160632 DOI: 10.1007/s00213-011-2232-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2010] [Accepted: 02/13/2011] [Indexed: 12/21/2022]
Abstract
RATIONALE Pharmacological manipulations of the type 1 cannabinoid receptor (CB1) suggest a role for CB1 in morphine-induced antinociception, but studies utilizing CB1 knockout (KO) mice do not support this conclusion. Since studies using CB1 KO mice to study morphine's antinociceptive effects have only examined thermal nociception, this study examines these interactions in models that employ a chemical stimulus. OBJECTIVES To determine whether the findings obtained with thermal pain models extend to other models, the effects of morphine on acetic acid-induced writhing were examined in CB1 KO and wildtype (WT) mice. Behaviors that decrease in response to acid injection, feeding and wheel running, were also examined, and investigations were carried out in the thermal hotplate assay. The CB1 antagonist SR141716A was also examined in these assays. RESULTS Morphine completely blocked acid-induced writhing (1.0-10.0 mg/kg) and increased response latencies in the hotplate (10.0-32.0 mg/kg) in both genotypes. Morphine (3.2 mg/kg) significantly attenuated the suppression of wheel running but did not completely prevent this effect in either genotype. Morphine did not alter pain-suppressed feeding. In each of these assays, morphine's effects were not altered in CB1 KO mice compared with WT mice; however, SR141716A attenuated morphine's effects in C57BL/6 mice. CONCLUSIONS The effects of morphine do not differ in CB1 KO and WT mice in preclinical pain models using thermal and chemical stimuli. Since SR141716A did attenuate the effects of morphine, it is possible that CB1 KO mice undergo developmental changes that mask the role of CB1 receptors in morphine's antinociceptive effects.
Collapse
Affiliation(s)
- Laurence L Miller
- Department of Psychology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | | | | | | |
Collapse
|
48
|
Sex and cannabinoid CB1 genotype differentiate palatable food and cocaine self-administration behaviors in mice. Behav Pharmacol 2011; 20:605-13. [PMID: 19745722 DOI: 10.1097/fbp.0b013e328331ba30] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Both cannabinoid CB1 receptor knockout and antagonism produce well-established attenuation of palatable food and drug self-administration behavior. Although cannabinoid drugs have received attention as pharmacotherapeutics for various disorders, including obesity and addiction, it is unclear whether these agents produce equivalent behavioral effects in females and males. In this study, acquisition of 32% corn oil or 10% Ensure self-administration, and maintenance of corn oil, Ensure, or 0.56 mg/kg/infusion cocaine self-administration under both fixed ratio (FR)-1 and progressive ratio (PR) schedule of reinforcement, was compared in male and female wild type (WT) and CB1 knockout (KO) mice. Furthermore, the effect of pretreatment with the CB1 antagonist SR141716 (0.3-3.0) on Ensure self-administration in male and female WT and CB1 KO mice was assessed. CB1 genotype and sex significantly interacted to produce an attenuation of acquisition and maintenance of Ensure self-administration and PR self-administration for both Ensure and cocaine in male CB1 KO mice. In contrast, male CB1 KO mice showed no deficit in acquisition and maintenance of FR-1 responding or in PR responding maintained by corn oil. Sex differences also arose within genotypes for responding maintained under all three reinforcers. Lastly, pretreatment with SR141716 attenuated Ensure self-administration in WT and CB1 KO mice but was approximately five-fold more potent in WT mice than in CB1 KOs. The present data add to a small but growing literature suggesting that the cannabinoid system may be differentially sensitive in its modulation of appetitive behavior in males versus females.
Collapse
|
49
|
Scrima M, Di Marino S, Grimaldi M, Mastrogiacomo A, Novellino E, Bifulco M, D'Ursi AM. Binding of the hemopressin peptide to the cannabinoid CB1 receptor: structural insights. Biochemistry 2010; 49:10449-57. [PMID: 21062041 DOI: 10.1021/bi1011833] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Hemopressin, a bioactive nonapeptide derived from the α1 chain of hemoglobin, was recently shown to possess selective antagonist activity at the cannabinoid CB(1) receptor [Heimann, A. S., et al. (2007) Proc. Natl. Acad. Sci. U.S.A. 104, 20588-20593]. CB(1) receptor antagonists have been extensively studied for their possible therapeutic use in the treatment of obesity, drug abuse, and heroin addiction. In particular, many compounds acting as CB(1) receptor antagonists have been synthesized and subjected to experiments as possible anti-obesity drugs, but their therapeutic application is still complicated by important side effects. Using circular dichroism and nuclear magnetic resonance spectroscopy, this work reports the conformational analysis of hemopressin and its truncated, biologically active fragment hemopressin(1-6). The binding modes of both hemopressin and hemopressin(1-6) are investigated by molecular docking calculations. Our conformational data indicate that regular turn structures in the central portion of hemopressin and hemopressin(1-6) are critical for an effective interaction with the receptor. The results of molecular docking calculations, indicating similarities and differences in comparison to the most accepted CB(1) pharmacophore model, suggest the possibility of new chemical scaffolds for the design of new CB(1) antagonist lead compounds.
Collapse
Affiliation(s)
- Mario Scrima
- Department of Pharmaceutical Sciences, University of Salerno, I-84084 Fisciano, Italy
| | | | | | | | | | | | | |
Collapse
|
50
|
Effects of 2-AG on the reinforcing properties of wheel activity in obese and lean Zucker rats. Behav Pharmacol 2010; 21:292-300. [PMID: 20512031 DOI: 10.1097/fbp.0b013e32833aec4d] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The endocannabinoid system plays a role in obesity, primarily by its role in food reward. Activity, also involved in obesity, seems to be at least partially controlled by the endocannabinoid system, but the relevant behavioral and neurochemical mechanisms have not been well established. This study represents an attempt to begin elucidating these mechanisms by examining the effects of an endogenous cannabinoid ligand, 2-arachidonoylglycerol (2-AG), on the reinforcing properties of exercise reinforcement in lean and obese Zucker rats. Ten obese and 10 lean Zucker rats pressed a locked door under a progressive ratio schedule of reinforcement that, when unlocked, provided access to a running wheel for 2-min periods. After baseline breakpoints were established, doses of 2-AG (0.3-3 mg/kg) were administered before experimental sessions. Obese rats exhibited lower breakpoints for wheel activity, lower response rates, and fewer revolutions compared with lean rats. 2-AG decreased breakpoints, response rates, and revolutions for obese rats, and revolutions only for lean rats. These data suggest that 2-AG may reduce the reinforcing properties of activity, and that obese Zuckers may show a greater sensitivity to 2-AG. The data also suggest that endocannabinoids may play a role in the reinforcing properties of exercise.
Collapse
|