1
|
Richardson B, Clarke C, Blundell J, Bambico FR. Therapeutic-like activity of cannabidiolic acid methyl ester in the MK-801 mouse model of schizophrenia: Role for cannabinoid CB1 and serotonin-1A receptors. Eur J Neurosci 2024; 59:2403-2415. [PMID: 38385841 DOI: 10.1111/ejn.16278] [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: 08/10/2023] [Revised: 01/15/2024] [Accepted: 01/27/2024] [Indexed: 02/23/2024]
Abstract
Schizophrenia is a psychotic disorder with an increasing prevalence and incidence over the last two decades. The condition presents with a diverse array of positive, negative, and cognitive impairments. Conventional treatments often yield unsatisfactory outcomes, especially with negative symptoms. We investigated the role of prefrontocortical (PFC) N-methyl-D-aspartate receptors (NMDARs) in the pathophysiology and development of schizophrenia. We explored the potential therapeutic effects of cannabidiolic acid (CBDA) methyl ester (HU-580), an analogue of CBDA known to act as an agonist of the serotonin-1A receptor (5-HT1AR) and an antagonist of cannabinoid type 1 receptor (CB1R). C57BL/6 mice were intraperitoneally administered the NMDAR antagonist, dizocilpine (MK-801, .3 mg/kg) once daily for 17 days. After 7 days, they were concurrently given HU-580 (.01 or .05 μg/kg) for 10 days. Behavioural deficits were assessed at two time points. We conducted enzyme-linked immunosorbent assays to measure the concentration of PFC 5-HT1AR and CB1R. We found that MK-801 effectively induced schizophrenia-related behaviours including hyperactivity, social withdrawal, increased forced swim immobility, and cognitive deficits. We discovered that low-dose HU-580 (.01 μg/kg), but not the high dose (.05 μg/kg), attenuated hyperactivity, forced swim immobility and cognitive deficits, particularly in female mice. Our results revealed that MK-801 downregulated both CB1R and 5-HT1AR, an effect that was blocked by both low- and high-dose HU-580. This study sheds light on the potential antipsychotic properties of HU-580, particularly in the context of NMDAR-induced dysfunction. Our findings could contribute significantly to our understanding of schizophrenia pathophysiology and offer a promising avenue for exploring the therapeutic potential of HU-580 and related compounds in alleviating symptoms.
Collapse
MESH Headings
- Animals
- Schizophrenia/drug therapy
- Schizophrenia/chemically induced
- Schizophrenia/metabolism
- Dizocilpine Maleate/pharmacology
- Receptor, Serotonin, 5-HT1A/metabolism
- Receptor, Serotonin, 5-HT1A/drug effects
- Male
- Mice
- Female
- Receptor, Cannabinoid, CB1/metabolism
- Receptor, Cannabinoid, CB1/agonists
- Mice, Inbred C57BL
- Disease Models, Animal
- Cannabinoids/pharmacology
- Receptors, N-Methyl-D-Aspartate/metabolism
- Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors
- Antipsychotic Agents/pharmacology
Collapse
Affiliation(s)
- Brandon Richardson
- Memorial University of Newfoundland and Labrador, St. John's, Newfoundland, Canada
| | - Courtney Clarke
- Memorial University of Newfoundland and Labrador, St. John's, Newfoundland, Canada
| | - Jacqueline Blundell
- Memorial University of Newfoundland and Labrador, St. John's, Newfoundland, Canada
| | - Francis R Bambico
- Memorial University of Newfoundland and Labrador, St. John's, Newfoundland, Canada
| |
Collapse
|
2
|
Raghav JG, Kumar H, Ji L, Vemuri K, Makriyannis A, Suh J, Leonard MZ, Dang V, Ty C, Marandola S, Kane N, Witt AS, Shaqour S, Miczek KA. The neutral CB1 antagonist AM6527 reduces ethanol seeking, binge-like consumption, reinforcing, and withdrawal effects in male and female mice. Psychopharmacology (Berl) 2024; 241:427-443. [PMID: 38001264 DOI: 10.1007/s00213-023-06500-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 11/06/2023] [Indexed: 11/26/2023]
Abstract
RATIONALE Alcohol use disorder (AUD) is a debilitating physiological and psychiatric disorder which affects individuals globally. The current pharmacological interventions to treat AUD are limited, and hence there is an urgent need for a novel pharmacological therapy which can be effective and safe across the population. OBJECTIVE We aimed to investigate a novel neutral cannabinoid receptor-1 (CB1R) antagonist, AM6527, in several preclinical models of ethanol consumption using male and female C57BL6/J mice. METHODS Independent groups of male and female mice were subjected to repeated cycles of drinking in the dark (DID), or intermittent access to alcohol (IAA) procedures. Twenty minutes prior to ethanol access in each procedure, animals were treated with intraperitoneal injections of either 1, 3, and 10 mg/kg of AM6527 or its respective vehicle. Acamprosate (100, 200, 300, and 400 mg/kg) or its respective vehicle was used as a positive control. Separate groups of male mice were subjected to a chain schedule of ethanol reinforcement to gain access to ethanol wherein completion of a fixed interval (FI; 5 min) schedule (link 1: "Seeking") was reinforced with continuous access to ethanol (fixed ratio; FR1) for up to 1.8 g/kg (link 2: "consumption"). All the animals were treated with 1, 3, and 10 mg/kg of AM6527 or its respective vehicle 20 mins prior to the start of the FI chain of the procedure. Separately, AM6527 was also evaluated in male and female mice undergoing acute ethanol withdrawal following 8 weeks of intermittent or continuous access to 20% ethanol drinking. RESULTS In both DID and IAA procedures, AM6527 reduced ethanol consumption in a dose-related manner in both male and female mice. AM6527 produced no tolerance in the DID procedure; mice treated with 3 mg/kg of AM6527 for 3 weeks continuously drank significantly smaller amounts of ethanol as compared to vehicle-treated mice over a period of three DID cycles. Moreover, in the IAA procedure, AM6527 caused an increase in water intake over the 24-h period. Acamprosate transiently reduced ethanol intake in male mice in both the DID and the IAA procedures but failed to produce any significant effect in female mice. AM6527 also produced a decrease in the FI responding ("ethanol seeking") in animals trained to self-administer ethanol. Lastly, AM6527 mitigated neurological withdrawal signs, i.e., handling induced convulsions (HIC) in mice undergoing acute ethanol withdrawal. CONCLUSIONS Current findings support previous studies with CB1R neutral antagonist in reducing voluntary ethanol intake and seeking behavior. Based on results shown in this work, AM6527 can be developed as a first in class CB1R neutral antagonist to treat AUD in both males and females.
Collapse
Affiliation(s)
- Jimit Girish Raghav
- Center for Drug Discovery, Northeastern University, 360 Huntington Avenue, Boston, MA, 02115, USA
| | - Hritik Kumar
- Center for Drug Discovery, Northeastern University, 360 Huntington Avenue, Boston, MA, 02115, USA
| | - Lipin Ji
- Center for Drug Discovery, Northeastern University, 360 Huntington Avenue, Boston, MA, 02115, USA
| | - Kiran Vemuri
- Center for Drug Discovery, Northeastern University, 360 Huntington Avenue, Boston, MA, 02115, USA
| | - Alexandros Makriyannis
- Center for Drug Discovery, Northeastern University, 360 Huntington Avenue, Boston, MA, 02115, USA.
| | - Junghyup Suh
- Division of Depression and Anxiety Disorders, Department of Psychiatry, Harvard Medical School, Mclean Hospital, Belmont, MA, 02478, USA
| | - Michael Z Leonard
- Dept. of Psychology, Tufts University, 530 Boston Ave (Bacon Hall), Medford, MA, 02155, USA
| | - Vivi Dang
- Dept. of Psychology, Tufts University, 530 Boston Ave (Bacon Hall), Medford, MA, 02155, USA
| | - Chelsea Ty
- Dept. of Psychology, Tufts University, 530 Boston Ave (Bacon Hall), Medford, MA, 02155, USA
| | - Stephen Marandola
- Dept. of Psychology, Tufts University, 530 Boston Ave (Bacon Hall), Medford, MA, 02155, USA
| | - Natalie Kane
- Dept. of Psychology, Tufts University, 530 Boston Ave (Bacon Hall), Medford, MA, 02155, USA
| | - Annika S Witt
- Dept. of Psychology, Tufts University, 530 Boston Ave (Bacon Hall), Medford, MA, 02155, USA
| | - Samar Shaqour
- Dept. of Psychology, Tufts University, 530 Boston Ave (Bacon Hall), Medford, MA, 02155, USA
| | - Klaus A Miczek
- Center for Drug Discovery, Northeastern University, 360 Huntington Avenue, Boston, MA, 02115, USA.
- Dept. of Psychology, Tufts University, 530 Boston Ave (Bacon Hall), Medford, MA, 02155, USA.
- Dept. of Neuroscience, Tufts University, Boston, MA, 02111, USA.
| |
Collapse
|
3
|
Haller J. Anxiety Modulation by Cannabinoids-The Role of Stress Responses and Coping. Int J Mol Sci 2023; 24:15777. [PMID: 37958761 PMCID: PMC10650718 DOI: 10.3390/ijms242115777] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 10/25/2023] [Accepted: 10/27/2023] [Indexed: 11/15/2023] Open
Abstract
Endocannabinoids were implicated in a variety of pathological conditions including anxiety and are considered promising new targets for anxiolytic drug development. The optimism concerning the potentials of this system for anxiolysis is probably justified. However, the complexity of the mechanisms affected by endocannabinoids, and discrepant findings obtained with various experimental approaches makes the interpretation of research results difficult. Here, we review the anxiety-related effects of the three main interventions used to study the endocannabinoid system: pharmacological agents active at endocannabinoid-binding sites present on both the cell membrane and in the cytoplasm, genetic manipulations targeting cannabinoid receptors, and function-enhancers represented by inhibitors of endocannabinoid degradation and transport. Binding-site ligands provide inconsistent findings probably because they activate a multitude of mechanisms concomitantly. More robust findings were obtained with genetic manipulations and particularly with function enhancers, which heighten ongoing endocannabinoid activation rather than affecting all mechanisms indiscriminately. The enhancement of ongoing activity appears to ameliorate stress-induced anxiety without consistent effects on anxiety in general. Limited evidence suggests that this effect is achieved by promoting active coping styles in critical situations. These findings suggest that the functional enhancement of endocannabinoid signaling is a promising drug development target for stress-related anxiety disorders.
Collapse
Affiliation(s)
- József Haller
- Drug Research Institute, 1137 Budapest, Hungary;
- Department of Criminal Psychology, University of Public Service, 1082 Budapest, Hungary
| |
Collapse
|
4
|
Maccarrone M, Di Marzo V, Gertsch J, Grether U, Howlett AC, Hua T, Makriyannis A, Piomelli D, Ueda N, van der Stelt M. Goods and Bads of the Endocannabinoid System as a Therapeutic Target: Lessons Learned after 30 Years. Pharmacol Rev 2023; 75:885-958. [PMID: 37164640 PMCID: PMC10441647 DOI: 10.1124/pharmrev.122.000600] [Citation(s) in RCA: 31] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 04/06/2023] [Accepted: 04/10/2023] [Indexed: 05/12/2023] Open
Abstract
The cannabis derivative marijuana is the most widely used recreational drug in the Western world and is consumed by an estimated 83 million individuals (∼3% of the world population). In recent years, there has been a marked transformation in society regarding the risk perception of cannabis, driven by its legalization and medical use in many states in the United States and worldwide. Compelling research evidence and the Food and Drug Administration cannabis-derived cannabidiol approval for severe childhood epilepsy have confirmed the large therapeutic potential of cannabidiol itself, Δ9-tetrahydrocannabinol and other plant-derived cannabinoids (phytocannabinoids). Of note, our body has a complex endocannabinoid system (ECS)-made of receptors, metabolic enzymes, and transporters-that is also regulated by phytocannabinoids. The first endocannabinoid to be discovered 30 years ago was anandamide (N-arachidonoyl-ethanolamine); since then, distinct elements of the ECS have been the target of drug design programs aimed at curing (or at least slowing down) a number of human diseases, both in the central nervous system and at the periphery. Here a critical review of our knowledge of the goods and bads of the ECS as a therapeutic target is presented to define the benefits of ECS-active phytocannabinoids and ECS-oriented synthetic drugs for human health. SIGNIFICANCE STATEMENT: The endocannabinoid system plays important roles virtually everywhere in our body and is either involved in mediating key processes of central and peripheral diseases or represents a therapeutic target for treatment. Therefore, understanding the structure, function, and pharmacology of the components of this complex system, and in particular of key receptors (like cannabinoid receptors 1 and 2) and metabolic enzymes (like fatty acid amide hydrolase and monoacylglycerol lipase), will advance our understanding of endocannabinoid signaling and activity at molecular, cellular, and system levels, providing new opportunities to treat patients.
Collapse
Affiliation(s)
- Mauro Maccarrone
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Italy (M.M.); European Center for Brain Research, Santa Lucia Foundation, Rome, Italy (M.M.); Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, University of Laval, Quebec, Canada (V.D.); Institute of Biochemistry and Molecular Medicine, NCCR TransCure, University of Bern, Bern, Switzerland (J.G.); Roche Pharma Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland (U.G.); Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina (A.C.H.); iHuman Institute, ShanghaiTech University, Shanghai, China (T.H.); Center for Drug Discovery and Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts (A.M.); Departments of Pharmaceutical Sciences and Biological Chemistry, University of California, Irvine, California (D.P.); Department of Biochemistry, Kagawa University School of Medicine, Miki, Kagawa, Japan (N.U.); Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands (M.S.)
| | - Vincenzo Di Marzo
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Italy (M.M.); European Center for Brain Research, Santa Lucia Foundation, Rome, Italy (M.M.); Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, University of Laval, Quebec, Canada (V.D.); Institute of Biochemistry and Molecular Medicine, NCCR TransCure, University of Bern, Bern, Switzerland (J.G.); Roche Pharma Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland (U.G.); Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina (A.C.H.); iHuman Institute, ShanghaiTech University, Shanghai, China (T.H.); Center for Drug Discovery and Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts (A.M.); Departments of Pharmaceutical Sciences and Biological Chemistry, University of California, Irvine, California (D.P.); Department of Biochemistry, Kagawa University School of Medicine, Miki, Kagawa, Japan (N.U.); Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands (M.S.)
| | - Jürg Gertsch
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Italy (M.M.); European Center for Brain Research, Santa Lucia Foundation, Rome, Italy (M.M.); Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, University of Laval, Quebec, Canada (V.D.); Institute of Biochemistry and Molecular Medicine, NCCR TransCure, University of Bern, Bern, Switzerland (J.G.); Roche Pharma Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland (U.G.); Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina (A.C.H.); iHuman Institute, ShanghaiTech University, Shanghai, China (T.H.); Center for Drug Discovery and Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts (A.M.); Departments of Pharmaceutical Sciences and Biological Chemistry, University of California, Irvine, California (D.P.); Department of Biochemistry, Kagawa University School of Medicine, Miki, Kagawa, Japan (N.U.); Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands (M.S.)
| | - Uwe Grether
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Italy (M.M.); European Center for Brain Research, Santa Lucia Foundation, Rome, Italy (M.M.); Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, University of Laval, Quebec, Canada (V.D.); Institute of Biochemistry and Molecular Medicine, NCCR TransCure, University of Bern, Bern, Switzerland (J.G.); Roche Pharma Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland (U.G.); Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina (A.C.H.); iHuman Institute, ShanghaiTech University, Shanghai, China (T.H.); Center for Drug Discovery and Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts (A.M.); Departments of Pharmaceutical Sciences and Biological Chemistry, University of California, Irvine, California (D.P.); Department of Biochemistry, Kagawa University School of Medicine, Miki, Kagawa, Japan (N.U.); Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands (M.S.)
| | - Allyn C Howlett
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Italy (M.M.); European Center for Brain Research, Santa Lucia Foundation, Rome, Italy (M.M.); Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, University of Laval, Quebec, Canada (V.D.); Institute of Biochemistry and Molecular Medicine, NCCR TransCure, University of Bern, Bern, Switzerland (J.G.); Roche Pharma Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland (U.G.); Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina (A.C.H.); iHuman Institute, ShanghaiTech University, Shanghai, China (T.H.); Center for Drug Discovery and Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts (A.M.); Departments of Pharmaceutical Sciences and Biological Chemistry, University of California, Irvine, California (D.P.); Department of Biochemistry, Kagawa University School of Medicine, Miki, Kagawa, Japan (N.U.); Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands (M.S.)
| | - Tian Hua
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Italy (M.M.); European Center for Brain Research, Santa Lucia Foundation, Rome, Italy (M.M.); Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, University of Laval, Quebec, Canada (V.D.); Institute of Biochemistry and Molecular Medicine, NCCR TransCure, University of Bern, Bern, Switzerland (J.G.); Roche Pharma Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland (U.G.); Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina (A.C.H.); iHuman Institute, ShanghaiTech University, Shanghai, China (T.H.); Center for Drug Discovery and Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts (A.M.); Departments of Pharmaceutical Sciences and Biological Chemistry, University of California, Irvine, California (D.P.); Department of Biochemistry, Kagawa University School of Medicine, Miki, Kagawa, Japan (N.U.); Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands (M.S.)
| | - Alexandros Makriyannis
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Italy (M.M.); European Center for Brain Research, Santa Lucia Foundation, Rome, Italy (M.M.); Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, University of Laval, Quebec, Canada (V.D.); Institute of Biochemistry and Molecular Medicine, NCCR TransCure, University of Bern, Bern, Switzerland (J.G.); Roche Pharma Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland (U.G.); Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina (A.C.H.); iHuman Institute, ShanghaiTech University, Shanghai, China (T.H.); Center for Drug Discovery and Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts (A.M.); Departments of Pharmaceutical Sciences and Biological Chemistry, University of California, Irvine, California (D.P.); Department of Biochemistry, Kagawa University School of Medicine, Miki, Kagawa, Japan (N.U.); Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands (M.S.)
| | - Daniele Piomelli
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Italy (M.M.); European Center for Brain Research, Santa Lucia Foundation, Rome, Italy (M.M.); Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, University of Laval, Quebec, Canada (V.D.); Institute of Biochemistry and Molecular Medicine, NCCR TransCure, University of Bern, Bern, Switzerland (J.G.); Roche Pharma Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland (U.G.); Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina (A.C.H.); iHuman Institute, ShanghaiTech University, Shanghai, China (T.H.); Center for Drug Discovery and Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts (A.M.); Departments of Pharmaceutical Sciences and Biological Chemistry, University of California, Irvine, California (D.P.); Department of Biochemistry, Kagawa University School of Medicine, Miki, Kagawa, Japan (N.U.); Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands (M.S.)
| | - Natsuo Ueda
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Italy (M.M.); European Center for Brain Research, Santa Lucia Foundation, Rome, Italy (M.M.); Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, University of Laval, Quebec, Canada (V.D.); Institute of Biochemistry and Molecular Medicine, NCCR TransCure, University of Bern, Bern, Switzerland (J.G.); Roche Pharma Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland (U.G.); Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina (A.C.H.); iHuman Institute, ShanghaiTech University, Shanghai, China (T.H.); Center for Drug Discovery and Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts (A.M.); Departments of Pharmaceutical Sciences and Biological Chemistry, University of California, Irvine, California (D.P.); Department of Biochemistry, Kagawa University School of Medicine, Miki, Kagawa, Japan (N.U.); Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands (M.S.)
| | - Mario van der Stelt
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Italy (M.M.); European Center for Brain Research, Santa Lucia Foundation, Rome, Italy (M.M.); Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, University of Laval, Quebec, Canada (V.D.); Institute of Biochemistry and Molecular Medicine, NCCR TransCure, University of Bern, Bern, Switzerland (J.G.); Roche Pharma Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland (U.G.); Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina (A.C.H.); iHuman Institute, ShanghaiTech University, Shanghai, China (T.H.); Center for Drug Discovery and Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts (A.M.); Departments of Pharmaceutical Sciences and Biological Chemistry, University of California, Irvine, California (D.P.); Department of Biochemistry, Kagawa University School of Medicine, Miki, Kagawa, Japan (N.U.); Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands (M.S.)
| |
Collapse
|
5
|
Zou G, Xia J, Luo H, Xiao D, Jin J, Miao C, Zuo X, Gao Q, Zhang Z, Xue T, You Y, Zhang Y, Zhang L, Xiong W. Combined alcohol and cannabinoid exposure leads to synergistic toxicity by affecting cerebellar Purkinje cells. Nat Metab 2022; 4:1138-1149. [PMID: 36109623 DOI: 10.1038/s42255-022-00633-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 08/03/2022] [Indexed: 11/09/2022]
Abstract
Combined use of cannabis and alcohol results in greater psychoactive toxicity than either substance alone, but the underlying central mechanisms behind this worsened outcome remain unclear. Here we show that the synergistic effect of Δ9-tetrahydrocannabinol (THC) and ethanol on motor incoordination in mice is achieved by activating presynaptic type 1 cannabinoid receptors (CB1R) and potentiating extrasynaptic glycine receptors (GlyR) within cerebellar Purkinje cells (PCs). The combination of ethanol and THC significantly reduces miniature excitatory postsynaptic current frequency in a CB1R-dependent manner, while increasing the extrasynaptic GlyR-mediated chronic chloride current, both leading to decreased PC activity. Ethanol enhances THC actions by boosting the blood-brain-barrier permeability of THC and enriching THC in the cell membrane. Di-desoxy-THC, a designed compound that specifically disrupts THC-GlyR interaction without affecting the basic functions of CB1R and GlyR, is able to restore PC function and motor coordination in mice. Our findings provide potential therapeutic strategies for overcoming the synergistic toxicity caused by combining cannabis and alcohol use.
Collapse
Affiliation(s)
- Guichang Zou
- Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, China
| | - Jing Xia
- Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Heyi Luo
- Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Dan Xiao
- Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Jin Jin
- Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Chenjian Miao
- Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Xin Zuo
- Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Qianqian Gao
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, China
| | - Zhi Zhang
- Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Tian Xue
- Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Yezi You
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, China
| | - Ye Zhang
- Department of Anesthesiology and Perioperative Medicine, The Second Hospital of Anhui Medical University, Hefei, China
| | - Li Zhang
- Laboratory for Integrative Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Wei Xiong
- Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
- Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, China.
- Anhui Province Key Laboratory of Biomedical Aging Research, Hefei, China.
| |
Collapse
|
6
|
Simone JJ, Green MR, McCormick CM. Endocannabinoid system contributions to sex-specific adolescent neurodevelopment. Prog Neuropsychopharmacol Biol Psychiatry 2022; 113:110438. [PMID: 34534603 DOI: 10.1016/j.pnpbp.2021.110438] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 08/13/2021] [Accepted: 09/08/2021] [Indexed: 01/08/2023]
Abstract
With an increasing number of countries and states adopting legislation permitting the use of cannabis for medical purposes, there is a growing interest among health and research professionals into the system through which cannabinoids principally act, the endocannabinoid system (ECS). Much of the seminal research into the ECS dates back only 30 years and, although there has been tremendous development within the field during this time, many questions remain. More recently, investigations have emerged examining the contributions of the ECS to normative development and the effect of altering this system during important critical periods. One such period is adolescence, a unique period during which brain and behaviours are maturing and reorganizing in preparation for adulthood, including shifts in endocannabinoid biology. The purpose of this review is to discuss findings to date regarding the maturation of the ECS during adolescence and the consequences of manipulations of the ECS during this period to normative neurodevelopmental processes, as well as highlight sex differences in ECS function, important technical considerations, and future directions. Because most of what we know is derived from preclinical studies on rodents, we provide relevant background of this model and some commentary on the translational relevance of the research in this area.
Collapse
Affiliation(s)
- Jonathan J Simone
- Department of Biological Sciences, 1812 Sir Isaac Brock Way, Brock University, St. Catharines, ON L2S 3A1, Canada; Centre for Neuroscience, 1812 Sir Isaac Brock Way, Brock University, St. Catharines, ON L2S 3A1, Canada; Huxley Health Inc., 8820 Jane St., Concord, ON, L4K 2M9, Canada; eCB Consulting Inc., PO Box 652, 3 Cameron St. W., Cannington, ON L0E 1E0, Canada; Medical Cannabis Canada, 601-3500 Lakeshore Rd. W., Oakville, ON L6L 0B4, Canada.
| | - Matthew R Green
- eCB Consulting Inc., PO Box 652, 3 Cameron St. W., Cannington, ON L0E 1E0, Canada; Medical Cannabis Canada, 601-3500 Lakeshore Rd. W., Oakville, ON L6L 0B4, Canada.
| | - Cheryl M McCormick
- Department of Biological Sciences, 1812 Sir Isaac Brock Way, Brock University, St. Catharines, ON L2S 3A1, Canada; Centre for Neuroscience, 1812 Sir Isaac Brock Way, Brock University, St. Catharines, ON L2S 3A1, Canada; Department of Psychology, 1812 Sir Isaac Brock Way, Brock University, St. Catharines, ON L2S 3A1, Canada.
| |
Collapse
|
7
|
Ahmed M, Boileau I, Le Foll B, Carvalho AF, Kloiber S. The endocannabinoid system in social anxiety disorder: from pathophysiology to novel therapeutics. ACTA ACUST UNITED AC 2021; 44:81-93. [PMID: 34468550 PMCID: PMC8827369 DOI: 10.1590/1516-4446-2021-1926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 06/15/2021] [Indexed: 11/22/2022]
Abstract
Social anxiety disorder (SAD) is a highly prevalent psychiatric disorder that presents with an early age of onset, chronic disease course, and increased risk of psychiatric comorbidity. Current treatment options for SAD are associated with low response rates, suboptimal efficacy, and possible risk of adverse effects. Investigation of new neurobiological mechanisms may aid in the identification of more specific therapeutic targets for the treatment of this disorder. Emerging evidence suggests that the endogenous cannabinoid system, also referred to as the endocannabinoid system (ECS), could play a potential role in the pathophysiology of SAD. This review discusses the known pathophysiological mechanisms of SAD, the potential role of the ECS in this disorder, current drugs targeting the ECS, and the potential of these novel compounds to enhance the therapeutic armamentarium for SAD. Further investigational efforts, specifically in human populations, are warranted to improve our knowledge of the ECS in SAD.
Collapse
Affiliation(s)
- Mashal Ahmed
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada.,Brain Health Imaging Centre, Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada
| | - Isabelle Boileau
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada.,Brain Health Imaging Centre, Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada.,Campbell Family Mental Health Research Institute, CAMH, Toronto, ON, Canada.,Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Bernard Le Foll
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada.,Campbell Family Mental Health Research Institute, CAMH, Toronto, ON, Canada.,Department of Psychiatry, University of Toronto, Toronto, ON, Canada.,Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - Andre F Carvalho
- Campbell Family Mental Health Research Institute, CAMH, Toronto, ON, Canada.,Department of Psychiatry, University of Toronto, Toronto, ON, Canada.,Innovation in Mental and Physical Health and Clinical Treatment (IMPACT) Strategic Research Centre, Deakin University, Geelong, VIC, Australia, 3216
| | - Stefan Kloiber
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada.,Campbell Family Mental Health Research Institute, CAMH, Toronto, ON, Canada.,Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
8
|
Parihar VK, Syage A, Flores L, Lilagan A, Allen BD, Angulo MC, Song J, Smith SM, Arechavala RJ, Giedzinski E, Limoli CL. The Cannabinoid Receptor 1 Reverse Agonist AM251 Ameliorates Radiation-Induced Cognitive Decrements. Front Cell Neurosci 2021; 15:668286. [PMID: 34262437 PMCID: PMC8273551 DOI: 10.3389/fncel.2021.668286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 05/26/2021] [Indexed: 11/13/2022] Open
Abstract
Despite advancements in the radiotherapeutic management of brain malignancies, resultant sequelae include persistent cognitive dysfunction in the majority of survivors. Defining the precise causes of normal tissue toxicity has proven challenging, but the use of preclinical rodent models has suggested that reductions in neurogenesis and microvascular integrity, impaired synaptic plasticity, increased inflammation, and alterations in neuronal structure are contributory if not causal. As such, strategies to reverse these persistent radiotherapy-induced neurological disorders represent an unmet medical need. AM251, a cannabinoid receptor 1 reverse agonist known to facilitate adult neurogenesis and synaptic plasticity, may help to ameliorate radiation-induced CNS impairments. To test this hypothesis, three treatment paradigms were used to evaluate the efficacy of AM251 to ameliorate radiation-induced learning and memory deficits along with disruptions in mood at 4 and 12 weeks postirradiation. Results demonstrated that acute (four weekly injections) and chronic (16 weekly injections) AM251 treatments (1 mg/kg) effectively alleviated cognitive and mood dysfunction in cranially irradiated mice. The beneficial effects of AM251 were exemplified by improved hippocampal- and cortical-dependent memory function on the novel object recognition and object in place tasks, while similar benefits on mood were shown by reductions in depressive- and anxiety-like behaviors on the forced swim test and elevated plus maze. The foregoing neurocognitive benefits were associated with significant increases in newly born (doublecortin+) neurons (1.7-fold), hippocampal neurogenesis (BrdU+/NeuN+mature neurons, 2.5-fold), and reduced expression of the inflammatory mediator HMGB (1.2-fold) in the hippocampus of irradiated mice. Collectively, these findings indicate that AM251 ameliorates the effects of clinically relevant cranial irradiation where overall neurological benefits in memory and mood coincided with increased hippocampal cell proliferation, neurogenesis, and reduced expression of proinflammatory markers.
Collapse
Affiliation(s)
- Vipan K Parihar
- Department of Radiation Oncology, University of California, Irvine, Irvine, CA, United States
| | - Amber Syage
- Department of Radiation Oncology, University of California, Irvine, Irvine, CA, United States
| | - Lidia Flores
- Department of Radiation Oncology, University of California, Irvine, Irvine, CA, United States
| | - Angelica Lilagan
- Department of Radiation Oncology, University of California, Irvine, Irvine, CA, United States
| | - Barrett D Allen
- Department of Radiation Oncology, University of California, Irvine, Irvine, CA, United States
| | - Maria C Angulo
- Department of Radiation Oncology, University of California, Irvine, Irvine, CA, United States
| | - Joseph Song
- Department of Radiation Oncology, University of California, Irvine, Irvine, CA, United States
| | - Sarah M Smith
- Department of Radiation Oncology, University of California, Irvine, Irvine, CA, United States
| | - Rebecca J Arechavala
- Department of Radiation Oncology, University of California, Irvine, Irvine, CA, United States
| | - Erich Giedzinski
- Department of Radiation Oncology, University of California, Irvine, Irvine, CA, United States
| | - Charles L Limoli
- Department of Radiation Oncology, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
9
|
Klein PM, Parihar VK, Szabo GG, Zöldi M, Angulo MC, Allen BD, Amin AN, Nguyen QA, Katona I, Baulch JE, Limoli CL, Soltesz I. Detrimental impacts of mixed-ion radiation on nervous system function. Neurobiol Dis 2021; 151:105252. [PMID: 33418069 DOI: 10.1016/j.nbd.2021.105252] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 12/02/2020] [Accepted: 01/02/2021] [Indexed: 12/11/2022] Open
Abstract
Galactic cosmic radiation (GCR), composed of highly energetic and fully ionized atomic nuclei, produces diverse deleterious effects on the body. In researching the neurological risks of GCR exposures, including during human spaceflight, various ground-based single-ion GCR irradiation paradigms induce differential disruptions of cellular activity and overall behavior. However, it remains less clear how irradiation comprising a mix of multiple ions, more accurately recapitulating the space GCR environment, impacts the central nervous system. We therefore examined how mixed-ion GCR irradiation (two similar 5-6 beam combinations of protons, helium, oxygen, silicon and iron ions) influenced neuronal connectivity, functional generation of activity within neural circuits and cognitive behavior in mice. In electrophysiological recordings we find that space-relevant doses of mixed-ion GCR preferentially alter hippocampal inhibitory neurotransmission and produce related disruptions in the local field potentials of hippocampal oscillations. Such underlying perturbation in hippocampal network activity correspond with perturbed learning, memory and anxiety behavior.
Collapse
Affiliation(s)
- Peter M Klein
- Department of Neurosurgery, Stanford University, Palo Alto, CA 94305, United States of America.
| | - Vipan K Parihar
- Department of Radiation Oncology, University of California, Irvine, CA 92697, United States of America
| | - Gergely G Szabo
- Department of Neurosurgery, Stanford University, Palo Alto, CA 94305, United States of America
| | - Miklós Zöldi
- Momentum Laboratory of Molecular Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences, 1083 Budapest, Hungary
| | - Maria C Angulo
- Department of Radiation Oncology, University of California, Irvine, CA 92697, United States of America
| | - Barrett D Allen
- Department of Radiation Oncology, University of California, Irvine, CA 92697, United States of America
| | - Amal N Amin
- Department of Radiation Oncology, University of California, Irvine, CA 92697, United States of America
| | - Quynh-Anh Nguyen
- Department of Neurosurgery, Stanford University, Palo Alto, CA 94305, United States of America
| | - István Katona
- Momentum Laboratory of Molecular Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences, 1083 Budapest, Hungary; Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN 47405, United States of America
| | - Janet E Baulch
- Department of Radiation Oncology, University of California, Irvine, CA 92697, United States of America
| | - Charles L Limoli
- Department of Radiation Oncology, University of California, Irvine, CA 92697, United States of America
| | - Ivan Soltesz
- Department of Neurosurgery, Stanford University, Palo Alto, CA 94305, United States of America; Department of Neurology & Neurological Sciences, Stanford University, Palo Alto, CA 94305, United States of America
| |
Collapse
|
10
|
Chen C, Chang Z, Tsai F, Chen S. Cannabinoid receptor type 1 antagonist inhibits progression of obesity-associated nonalcoholic steatohepatitis in a mouse model by remodulating immune system disturbances. Immun Inflamm Dis 2020; 8:544-558. [PMID: 32798334 PMCID: PMC7654409 DOI: 10.1002/iid3.338] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 07/23/2020] [Accepted: 07/31/2020] [Indexed: 01/15/2023] Open
Abstract
SCOPE This study investigated whether AM251, a cannabinoid receptor type 1 (CB1) antagonist, ameliorates hepatic levels of metabolic abnormalities and inflammatory responses in a murine nonalcoholic steatohepatitis (NASH) model via reversal of disturbances in the immune system. METHODS AND RESULTS Fifteen-week-old male obese db/db mice were randomly assigned to the following two groups: no treatment and treatment with AM251 at 5 mg/kg for 15 days. C57BL/6J-Lean mice were utilized as the control group. Plasma parameters, liver histopathology, and hepatic status were measured. For the in vitro study, macrophage-derived RAW264.7 cells were cultured with AM251 or CB1 small interfering RNA (siRNA) before challenge with arachidonyl-2'-chloroethylamide (ACEA) or a high concentration of fatty acids (HFFAs). The db/db mice exhibited an increase in CB1 levels, lipid droplet accumulation, mitogen-activated protein kinase-related inflammatory responses, and macrophage and neutrophil infiltration in the liver tissues. Flow cytometry analysis revealed an elevation in macrophages and T helper cells, plus a decrease in natural killer T cells and regulatory T cells in the liver tissues of the db/db mice; treatment with 5 mg/kg AM251 reversed these changes. Moreover, in vitro experiments revealed that administration of 3.3 μM AM251 or CB1 siRNA prevented 1 mM HFFA- and 1 μΜ ACEA-induced inflammatory cytokine protein expression in the RAW264.7 cells. CONCLUSION These findings suggested that a blockade caused by CB1 reduced obesity-associated NASH progression via correction of immune system dysregulations and elevated inflammatory responses in the liver tissues.
Collapse
Affiliation(s)
- Chin‐Chang Chen
- Department of Traditional Chinese MedicineChang Gung Memorial HospitalKeelungTaiwan, ROC
- Department of Anatomy, School of MedicineChina Medical UniversityTaichungTaiwan, ROC
| | - Zi‐Yu Chang
- Department of Traditional Chinese MedicineChang Gung Memorial HospitalKeelungTaiwan, ROC
- Institute of Traditional Medicine, School of MedicineNational Yang‐Ming UniversityTaipeiTaiwan, ROC
| | - Fuu‐Jen Tsai
- School of Chinese MedicineChina Medical UniversityTaichungTaiwan, ROC
- Department of Medical Research, Genetics CenterChina Medical University HospitalTaichungTaiwan, ROC
- Department of Medical GeneticsChina Medical University HospitalTaichungTaiwan, ROC
| | - Shih‐Yin Chen
- School of Chinese MedicineChina Medical UniversityTaichungTaiwan, ROC
- Department of Medical Research, Genetics CenterChina Medical University HospitalTaichungTaiwan, ROC
| |
Collapse
|
11
|
Effects of the CB1 Receptor Antagonists AM6545 and AM4113 on Insulin Resistance in a High-Fructose High-Salt Rat Model of Metabolic Syndrome. ACTA ACUST UNITED AC 2020; 56:medicina56110573. [PMID: 33138155 PMCID: PMC7692885 DOI: 10.3390/medicina56110573] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 10/22/2020] [Accepted: 10/27/2020] [Indexed: 02/07/2023]
Abstract
Background and Objectives: Insulin resistance (IR) is a serious condition leading to development of diabetes and cardiovascular complications. Hyper-activation of cannabinoid receptors-1 (CB1) has been linked to the development of metabolic disorders such as IR. Therefore, the effect of blocking CB1 on the development of IR was investigated in the present study. Materials and Methods: A 12-week high-fructose/high-salt feeding model of metabolic syndrome was used to induce IR in male Wistar rats. For this purpose, two different CB1-antagonists were synthesized and administered to the rats during the final four weeks of the study, AM6545, the peripheral neutral antagonist and AM4113, the central neutral antagonist. Results: High-fructose/salt feeding for 12 weeks led to development of IR while both AM6545 and AM4113, administered in the last 4 weeks, significantly inhibited IR. This was correlated with increased animal body weight wherein both AM6545 and AM4113 decreased body weight in IR animals but with loss of IR/body weight correlation. While IR animals showed significant elevations in serum cholesterol and triglycerides with no direct correlation with IR, both AM6545 and AM4113 inhibited these elevations, with direct IR/cholesterol correlation in case of AM6545. IR animals had elevated serum uric acid, which was reduced by both AM6545 and AM4113. In addition, IR animals had decreased adiponectin levels and elevated liver TNFα content with strong IR/adiponectin and IR/TNFα correlations. AM6545 inhibited the decreased adiponectin and the increased TNFα levels and retained the strong IR/adiponectin correlation. However, AM4113 inhibited the decreased adiponectin and the increased TNFα levels, but with loss of IR/adiponectin and IR/TNFα correlations. Conclusions: Both CB1 neutral antagonists alleviated IR peripherally, and exerted similar effects on rats with metabolic syndrome. They also displayed anti-dyslipidemic, anti-hyperurecemic and anti-inflammatory effects. Overall, these results should assist in the development of CB1 neutral antagonists with improved safety profiles for managing metabolic disorders.
Collapse
|
12
|
La-Vu M, Tobias BC, Schuette PJ, Adhikari A. To Approach or Avoid: An Introductory Overview of the Study of Anxiety Using Rodent Assays. Front Behav Neurosci 2020; 14:145. [PMID: 33005134 PMCID: PMC7479238 DOI: 10.3389/fnbeh.2020.00145] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 07/28/2020] [Indexed: 12/21/2022] Open
Abstract
Anxiety is a widely studied phenomenon in behavioral neuroscience, but the recent literature lacks an overview of the major conceptual framework underlying anxiety research to introduce young researchers to the field. In this mini-review article, which is aimed toward new undergraduate and graduate students, we discuss how researchers exploit the approach-avoidance conflict, an internal conflict rodents face between exploration of novel environments and avoidance of danger, to inform rodent assays that allow for the measurement of anxiety-related behavior in the laboratory. We review five widely-used rodent anxiety assays, consider the pharmacological validity of these assays, and discuss neural circuits that have recently been shown to modulate anxiety using the assays described. Finally, we offer related lines of inquiry and comment on potential future directions.
Collapse
Affiliation(s)
- Mimi La-Vu
- Department of Psychology, University of California, Los Angeles, Los Angeles, CA, United States
| | - Brooke C Tobias
- Department of Psychology, University of California, Los Angeles, Los Angeles, CA, United States
| | - Peter J Schuette
- Department of Psychology, University of California, Los Angeles, Los Angeles, CA, United States
| | - Avishek Adhikari
- Department of Psychology, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
13
|
Kunos G. Interactions Between Alcohol and the Endocannabinoid System. Alcohol Clin Exp Res 2020; 44:790-805. [PMID: 32056226 DOI: 10.1111/acer.14306] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 02/05/2020] [Indexed: 12/12/2022]
Abstract
Endocannabinoids are lipid mediators that interact with the same cannabinoid receptors that recognize Δ9 -tetrahydrocannabinol (THC), the psychoactive constituent of marijuana, to induce similar effects in the brain and periphery. Alcohol and THC are both addictive substances whose acute use elicits rewarding effects that can lead to chronic and compulsive use via engaging similar signaling pathways in the brain. In the liver, both alcohol and endocannabinoids activate lipogenic gene expression leading to fatty liver disease. This review focuses on evidence accumulated over the last 2 decades to indicate that both the addictive neural effects of ethanol and its organ toxic effects in the liver and elsewhere are mediated, to a large extent, by endocannabinoids signaling via cannabinoid-1 receptors (CB1 R). The therapeutic potential of CB1 R blockade globally or in peripheral tissues only is also discussed.
Collapse
Affiliation(s)
- George Kunos
- From the, Division of Clinical and Biological Research, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
14
|
Navarrete F, García-Gutiérrez MS, Jurado-Barba R, Rubio G, Gasparyan A, Austrich-Olivares A, Manzanares J. Endocannabinoid System Components as Potential Biomarkers in Psychiatry. Front Psychiatry 2020; 11:315. [PMID: 32395111 PMCID: PMC7197485 DOI: 10.3389/fpsyt.2020.00315] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 03/30/2020] [Indexed: 12/19/2022] Open
Abstract
The high heterogeneity of psychiatric disorders leads to a lack of diagnostic precision. Therefore, the search of biomarkers is a fundamental aspect in psychiatry to reach a more personalized medicine. The endocannabinoid system (ECS) has gained increasing interest due to its involvement in many different functional processes in the brain, including the regulation of emotions, motivation, and cognition. This article reviews the role of the main components of the ECS as biomarkers in certain psychiatric disorders. Studies carried out in rodents evaluating the effects of pharmacological and genetic manipulation of cannabinoid receptors or endocannabinoids (eCBs) degrading enzymes were included. Likewise, the ECS-related alterations occurring at the molecular level in animal models reproducing some behavioral and/or neuropathological aspects of psychiatric disorders were reviewed. Furthermore, clinical studies evaluating gene or protein alterations in post-mortem brain tissue or in vivo blood, plasma, and cerebrospinal fluid (CSF) samples were analyzed. Also, the results from neuroimaging studies using positron emission tomography (PET) or functional magnetic resonance (fMRI) were included. This review shows the close involvement of cannabinoid receptor 1 (CB1r) in stress regulation and the development of mood disorders [anxiety, depression, bipolar disorder (BD)], in post-traumatic stress disorder (PTSD), as well as in the etiopathogenesis of schizophrenia, attention deficit hyperactivity disorder (ADHD), or eating disorders (i.e. anorexia and bulimia nervosa). On the other hand, recent results reveal the potential therapeutic action of the endocannabinoid tone manipulation by inhibition of eCBs degrading enzymes, as well as by the modulation of cannabinoid receptor 2 (CB2r) activity on anxiolytic, antidepressive, or antipsychotic associated effects. Further clinical research studies are needed; however, current evidence suggests that the components of the ECS may become promising biomarkers in psychiatry to improve, at least in part, the diagnosis and pharmacological treatment of psychiatric disorders.
Collapse
Affiliation(s)
- Francisco Navarrete
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Alicante, Spain.,Red Temática de Investigación Cooperativa en Salud (RETICS), Red de Trastornos Adictivos, Instituto de Salud Carlos III, MICINN and FEDER, Madrid, Spain
| | - María Salud García-Gutiérrez
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Alicante, Spain.,Red Temática de Investigación Cooperativa en Salud (RETICS), Red de Trastornos Adictivos, Instituto de Salud Carlos III, MICINN and FEDER, Madrid, Spain
| | - Rosa Jurado-Barba
- Instituto de Investigación i+12, Hospital Universitario 12 de Octubre, Madrid, Spain.,Servicio de Psiquiatría, Hospital Universitario 12 de Octubre, Madrid, Spain.,Departamento de Psicología, Facultad de Educación y Salud, Universidad Camilo José Cela, Madrid, Spain
| | - Gabriel Rubio
- Red Temática de Investigación Cooperativa en Salud (RETICS), Red de Trastornos Adictivos, Instituto de Salud Carlos III, MICINN and FEDER, Madrid, Spain.,Instituto de Investigación i+12, Hospital Universitario 12 de Octubre, Madrid, Spain.,Servicio de Psiquiatría, Hospital Universitario 12 de Octubre, Madrid, Spain.,Department of Psychiatry, Complutense University of Madrid, Madrid, Spain
| | - Ani Gasparyan
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Alicante, Spain.,Red Temática de Investigación Cooperativa en Salud (RETICS), Red de Trastornos Adictivos, Instituto de Salud Carlos III, MICINN and FEDER, Madrid, Spain
| | | | - Jorge Manzanares
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Alicante, Spain.,Red Temática de Investigación Cooperativa en Salud (RETICS), Red de Trastornos Adictivos, Instituto de Salud Carlos III, MICINN and FEDER, Madrid, Spain
| |
Collapse
|
15
|
Shallcross J, Hámor P, Bechard AR, Romano M, Knackstedt L, Schwendt M. The Divergent Effects of CDPPB and Cannabidiol on Fear Extinction and Anxiety in a Predator Scent Stress Model of PTSD in Rats. Front Behav Neurosci 2019; 13:91. [PMID: 31133832 PMCID: PMC6523014 DOI: 10.3389/fnbeh.2019.00091] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 04/16/2019] [Indexed: 01/12/2023] Open
Abstract
Post-traumatic stress disorder (PTSD) currently has no FDA-approved treatments that reduce symptoms in the majority of patients. The ability to extinguish fear memory associations is impaired in PTSD individuals. As such, the development of extinction-enhancing pharmacological agents to be used in combination with exposure therapies may benefit the treatment of PTSD. Both mGlu5 and CB1 receptors have been implicated in contextual fear extinction. Thus, here we tested the ability of the mGlu5 positive allosteric modulator 3-Cyano-N-(1,3-diphenyl-1H-pyrazol-5-yl)benzamide (CDPPB) and cannabidiol (CBD) to reduce both conditioned and unconditioned fear. We used a predator-threat animal model of PTSD which we and others have previously shown to capture the heterogeneity of anxiety responses observed in humans exposed to trauma. Here, 1 week following a 10-min exposure to predator scent stress, rats were classified into stress-Susceptible and stress-Resilient phenotypes using behavioral criteria for elevated plus maze and acoustic startle response performance. Two weeks after classification, rats underwent 3 days of contextual fear extinction and were treated with vehicle, CDPPB or CBD prior to each session. Finally, the light-dark box test was employed to assess phenotypic differences and the effects of CDPPB and CBD on unconditioned anxiety. CDPBB but not CBD, reduced freezing in Susceptible rats relative to vehicle. In the light-dark box test for unconditioned anxiety, CBD, but not CDPPB, reduced anxiety in Susceptible rats. Resilient rats displayed reduced anxiety in the light-dark box relative to Susceptible rats. Taken together, the present data indicate that enhancement of mGlu5 receptor signaling in populations vulnerable to stress may serve to offset a resistance to fear memory extinction without producing anxiogenic effects. Furthermore, in a susceptible population, CBD attenuates unconditioned but not conditioned fear. Taken together, these findings support the use of predator-threat stress exposure in combination with stress-susceptibility phenotype classification as a model for examining the unique drug response profiles and altered neuronal function that emerge as a consequence of the heterogeneity of psychophysiological response to stress.
Collapse
Affiliation(s)
- John Shallcross
- Department of Psychology, University of Florida, Gainesville, FL, United States.,Center for Addiction Research & Education, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Peter Hámor
- Department of Psychology, University of Florida, Gainesville, FL, United States.,Center for Addiction Research & Education, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Allison R Bechard
- Department of Psychology, University of Florida, Gainesville, FL, United States
| | - Madison Romano
- Department of Psychology, University of Florida, Gainesville, FL, United States
| | - Lori Knackstedt
- Department of Psychology, University of Florida, Gainesville, FL, United States.,Center for Addiction Research & Education, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Marek Schwendt
- Department of Psychology, University of Florida, Gainesville, FL, United States.,Center for Addiction Research & Education, College of Medicine, University of Florida, Gainesville, FL, United States
| |
Collapse
|
16
|
Micale V, Drago F, Noerregaard PK, Elling CE, Wotjak CT. The Cannabinoid CB1 Antagonist TM38837 With Limited Penetrance to the Brain Shows Reduced Fear-Promoting Effects in Mice. Front Pharmacol 2019; 10:207. [PMID: 30949045 PMCID: PMC6435594 DOI: 10.3389/fphar.2019.00207] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2018] [Accepted: 02/19/2019] [Indexed: 12/26/2022] Open
Abstract
Rimonabant was the first selective CB1 antagonist/inverse agonist introduced into clinical practice to treat obesity and metabolic-related disorders. It was withdrawn from market due to the notably increased rates of psychiatric side effects. We have evaluated TM38837, a novel, largely peripherally restricted CB1 antagonist, in terms of fear-promoting consequences of systemic vs. intracerebral injections. Different groups of male C57BL/6 N mice underwent auditory fear conditioning, followed by re-exposure to the tone. Mice were treated per os (p.o.) with TM38837 (10, 30, or 100 mg/kg), rimonabant (10 mg/kg; a brain penetrating CB1 antagonist/inverse agonist which served as a positive control), or vehicle, 2 h prior the tone presentation. Only the high dose of TM38837 (100 mg/kg) induced a significant increase in freezing behavior, similar to that induced by rimonabant (10 mg/kg) (p < 0.001). If injected into the brain both TM38837 (10 or 30 μg/mouse) and rimonabant (1 or 10 μg/mouse) caused a sustained fear response to the tone, which was more pronounced after rimonabant treatment. Taken together, TM38837 was at least one order of magnitude less effective in promoting fear responses than rimonabant. Given the equipotency of the two CB1 antagonists with regard to weight loss and metabolic syndrome-like symptoms in rodent obesity models, our results point to a critical dose range in which TM3887 might be beneficial for indications such as obesity and metabolic disorders with limited risk of fear-promoting effects.
Collapse
Affiliation(s)
- Vincenzo Micale
- Research Group "Neuronal Plasticity", Max Planck Institute of Psychiatry, Munich, Germany.,Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, Catania, Italy.,National Institute Mental Health, Klecany, Czechia
| | - Filippo Drago
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, Catania, Italy
| | | | | | - Carsten T Wotjak
- Research Group "Neuronal Plasticity", Max Planck Institute of Psychiatry, Munich, Germany
| |
Collapse
|
17
|
Simone JJ, Baumbach JL, McCormick CM. Sex-specific effects of CB1 receptor antagonism and stress in adolescence on anxiety, corticosterone concentrations, and contextual fear in adulthood in rats. Int J Dev Neurosci 2018; 69:119-131. [PMID: 30063953 DOI: 10.1016/j.ijdevneu.2018.07.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 07/23/2018] [Accepted: 07/26/2018] [Indexed: 02/07/2023] Open
Abstract
There is a paucity of research regarding the role of endogenous cannabinoid signalling in adolescence on brain and behaviour development. We previously demonstrated effects of repeated CB1 receptor antagonism in adolescence on socioemotional behaviours and neural protein expression 24-48 h after the last drug administration in female rats, with no effect in males. Here we investigate whether greater effects would be manifested after a lengthier delay. In Experiment 1, male and female rats were administered either 1 mg / kg of the CB1 receptor-selective antagonist AM251, vehicle (VEH), or did not receive injections (NoINJ) daily on postnatal days (PND) 30-44 either alone (no adolescent confinement stress; noACS), or in tandem with 1 h ACS. On PND 70, adolescent AM251 exposure reduced anxiety in an elevated plus maze in males, irrespective of ACS, with no effects in females. On PND 73, there were no group differences in either sex in plasma corticosterone concentrations before or after 30 min of restraint stress, although injection stress resulted in higher baseline concentrations in males. Brains were collected on PND 74, with negligible effects of either AM251 or ACS on protein markers of synaptic plasticity and of the endocannabinoid system in the hippocampus and medial prefrontal cortex. In Experiment 2, rats from both sexes were treated with vehicle or AM251 on PND 30-44 and were tested for contextual fear conditioning and extinction in adulthood. AM251 females had greater fear recall than VEH females 24 h after conditioning, with no group differences in within- or between-session fear extinction. There were no group differences in long-term extinction memory, although AM251 females froze more during a reconditioning trial compared with VEH females. There were no group differences on any of the fear conditioning measures in males. Together, these findings indicate a modest, sex-specific role of CB1 receptor signalling in adolescence on anxiety-like behaviour in males and conditioned fear behaviour in females.
Collapse
Affiliation(s)
- Jonathan J Simone
- Department of Biological Sciences, Brock University, 1812 Sir Isaac Brock Way, St. Catharines, Ontario, L2S 3A1, Canada.
| | - Jennet L Baumbach
- Department of Psychology, Brock University, 1812 Sir Isaac Brock Way, St. Catharines, Ontario, L2S 3A1, Canada.
| | - Cheryl M McCormick
- Department of Biological Sciences, Brock University, 1812 Sir Isaac Brock Way, St. Catharines, Ontario, L2S 3A1, Canada; Department of Psychology, Brock University, 1812 Sir Isaac Brock Way, St. Catharines, Ontario, L2S 3A1, Canada; Center for Neuroscience, Brock University, 1812 Sir Isaac Brock Way, St. Catharines, Ontario, L2S 3A1, Canada.
| |
Collapse
|
18
|
Integrating Endocannabinoid Signaling and Cannabinoids into the Biology and Treatment of Posttraumatic Stress Disorder. Neuropsychopharmacology 2018; 43:80-102. [PMID: 28745306 PMCID: PMC5719095 DOI: 10.1038/npp.2017.162] [Citation(s) in RCA: 154] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 07/17/2017] [Accepted: 07/20/2017] [Indexed: 01/21/2023]
Abstract
Exposure to stress is an undeniable, but in most cases surmountable, part of life. However, in certain individuals, exposure to severe or cumulative stressors can lead to an array of pathological conditions including posttraumatic stress disorder (PTSD), characterized by debilitating trauma-related intrusive thoughts, avoidance behaviors, hyperarousal, as well as depressed mood and anxiety. In the context of the rapidly changing political and legal landscape surrounding use of cannabis products in the USA, there has been a surge of public and research interest in the role of cannabinoids in the regulation of stress-related biological processes and in their potential therapeutic application for stress-related psychopathology. Here we review the current state of knowledge regarding the effects of cannabis and cannabinoids in PTSD and the preclinical and clinical literature on the effects of cannabinoids and endogenous cannabinoid signaling systems in the regulation of biological processes related to the pathogenesis of PTSD. Potential therapeutic implications of the reviewed literature are also discussed. Finally, we propose that a state of endocannabinoid deficiency could represent a stress susceptibility endophenotype predisposing to the development of trauma-related psychopathology and provide biologically plausible support for the self-medication hypotheses used to explain high rates of cannabis use in patients with trauma-related disorders.
Collapse
|
19
|
Panlilio LV, Justinova Z. Preclinical Studies of Cannabinoid Reward, Treatments for Cannabis Use Disorder, and Addiction-Related Effects of Cannabinoid Exposure. Neuropsychopharmacology 2018; 43:116-141. [PMID: 28845848 PMCID: PMC5719102 DOI: 10.1038/npp.2017.193] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 08/17/2017] [Accepted: 08/22/2017] [Indexed: 12/21/2022]
Abstract
Cannabis use has become increasingly accepted socially and legally, for both recreational and medicinal purposes. Without reliable information about the effects of cannabis, people cannot make informed decisions regarding its use. Like alcohol and tobacco, cannabis can have serious adverse effects on health, and some people have difficulty discontinuing their use of the drug. Many cannabis users progress to using and becoming addicted to other drugs, but the reasons for this progression are unclear. The natural cannabinoid system of the brain is complex and involved in many functions, including brain development, reward, emotion, and cognition. Animal research provides an objective and controlled means of obtaining information about: (1) how cannabis affects the brain and behavior, (2) whether medications can be developed to treat cannabis use disorder, and (3) whether cannabis might produce lasting changes in the brain that increase the likelihood of becoming addicted to other drugs. This review explains the tactics used to address these issues, evaluates the progress that has been made, and offers some directions for future research.
Collapse
Affiliation(s)
- Leigh V Panlilio
- Preclinical Pharmacology Section, Behavioral Neuroscience Research Branch, Intramural Research Program, National Institute on Drug Abuse, NIH, DHHS, Baltimore, MD, USA
| | - Zuzana Justinova
- Preclinical Pharmacology Section, Behavioral Neuroscience Research Branch, Intramural Research Program, National Institute on Drug Abuse, NIH, DHHS, Baltimore, MD, USA
| |
Collapse
|
20
|
Balla A, Dong B, Shilpa BM, Vemuri K, Makriyannis A, Pandey SC, Sershen H, Suckow RF, Vinod KY. Cannabinoid-1 receptor neutral antagonist reduces binge-like alcohol consumption and alcohol-induced accumbal dopaminergic signaling. Neuropharmacology 2017; 131:200-208. [PMID: 29109060 DOI: 10.1016/j.neuropharm.2017.10.040] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 10/27/2017] [Accepted: 10/30/2017] [Indexed: 01/16/2023]
Abstract
Binge alcohol (ethanol) drinking is associated with profound adverse effects on our health and society. Rimonabant (SR141716A), a CB1 receptor inverse agonist, was previously shown to be effective for nicotine cessation and obesity. However, studies using rimonabant were discontinued as it was associated with an increased risk of depression and anxiety. In the present study, we examined the pharmacokinetics and effects of AM4113, a novel CB1 receptor neutral antagonist on binge-like ethanol drinking in C57BL/6J mice using a two-bottle choice drinking-in-dark (DID) paradigm. The results indicated a slower elimination of AM4113 in the brain than in plasma. AM4113 suppressed ethanol consumption and preference without having significant effects on body weight, ambulatory activity, preference for tastants (saccharin and quinine) and ethanol metabolism. AM4113 pretreatment reduced ethanol-induced increase in dopamine release in nucleus accumbens. Collectively, these data suggest an important role of CB1 receptor-mediated regulation of binge-like ethanol consumption and mesolimbic dopaminergic signaling, and further points to the potential utility of CB1 neutral antagonists for the treatment of binge ethanol drinking.
Collapse
Affiliation(s)
- Andrea Balla
- Department of Neurochemistry, Nathan Kline Institute for Psychiatric Research, Orangeburg, New York, NY, United States
| | - Bin Dong
- Division of Analytical Psychopharmacology, Nathan Kline Institute for Psychiatric Research, Orangeburg, New York, NY, United States
| | - Borehalli M Shilpa
- Division of Analytical Psychopharmacology, Nathan Kline Institute for Psychiatric Research, Orangeburg, New York, NY, United States
| | - Kiran Vemuri
- Center for Drug Discovery, Northeastern University, Boston, MA, United States
| | | | - Subhash C Pandey
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois at Chicago and Jesse Brown VA Medical Center, Chicago, IL, United States
| | - Henry Sershen
- Department of Neurochemistry, Nathan Kline Institute for Psychiatric Research, Orangeburg, New York, NY, United States; Department of Psychiatry, NYU Langone Medical Center, New York, NY, United States
| | - Raymond F Suckow
- Division of Analytical Psychopharmacology, Nathan Kline Institute for Psychiatric Research, Orangeburg, New York, NY, United States; New York State Psychiatric Institute, New York, United States; Department of Psychiatry, Columbia University College of Physicians and Surgeons, New York, NY, United States
| | - K Yaragudri Vinod
- Division of Analytical Psychopharmacology, Nathan Kline Institute for Psychiatric Research, Orangeburg, New York, NY, United States; Emotional Brain Institute, Orangeburg, New York, NY, United States; Department of Child and Adolescent Psychiatry, NYU Langone Medical Center, New York, NY, United States.
| |
Collapse
|
21
|
Sloan ME, Gowin JL, Ramchandani VA, Hurd YL, Le Foll B. The endocannabinoid system as a target for addiction treatment: Trials and tribulations. Neuropharmacology 2017; 124:73-83. [PMID: 28564576 DOI: 10.1016/j.neuropharm.2017.05.031] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 05/24/2017] [Accepted: 05/26/2017] [Indexed: 12/19/2022]
Abstract
Addiction remains a major public health concern, and while pharmacotherapies can be effective, clinicians are limited by the paucity of existing interventions. Endocannabinoid signaling is involved in reward and addiction, which raises the possibility that drugs targeting this system could be used to treat substance use disorders. This review discusses findings from randomized controlled trials evaluating cannabinergic medications for addiction. Current evidence suggests that pharmacotherapies containing delta-9-tetrahydrocannabinol, such as dronabinol and nabiximols, are effective for cannabis withdrawal. Dronabinol may also reduce symptoms of opioid withdrawal. The cannabinoid receptor 1 (CB1) inverse agonist rimonabant showed promising effects for smoking cessation but also caused psychiatric side effects and currently lacks regulatory approval. Few trials have investigated cannabinergic medications for alcohol use disorder. Overall, the endocannabinoid system remains a promising target for addiction treatment. Development of novel medications such as fatty acid amide hydrolase inhibitors and neutral CB1 antagonists promises to extend the range of available interventions. This article is part of the Special Issue entitled "A New Dawn in Cannabinoid Neurobiology".
Collapse
Affiliation(s)
- Matthew E Sloan
- Section on Human Psychopharmacology, National Institute on Alcohol Abuse and Alcoholism, 10 Center Drive (10CRC, 2-2352), Bethesda, MD, 20892-1540, USA
| | - Joshua L Gowin
- Section on Human Psychopharmacology, National Institute on Alcohol Abuse and Alcoholism, 10 Center Drive (10CRC, 2-2352), Bethesda, MD, 20892-1540, USA
| | - Vijay A Ramchandani
- Section on Human Psychopharmacology, National Institute on Alcohol Abuse and Alcoholism, 10 Center Drive (10CRC, 2-2352), Bethesda, MD, 20892-1540, USA
| | - Yasmin L Hurd
- Departments of Psychiatry, Neuroscience, Pharmacology, and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, Hess CSM Building, Floor 10, Rm 105, Office 1470, Madison Avenue, New York, NY, 10029, USA
| | - Bernard Le Foll
- Translational Addiction Research Laboratory, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, 33 Russell Street, Toronto, Ontario, M5S 2S1, Canada; Addiction Medicine Service, Ambulatory Care and Structured Treatments, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Departments of Family and Community Medicine, Pharmacology and Toxicology, and Psychiatry, Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada; Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada.
| |
Collapse
|
22
|
Salort G, Álvaro-Bartolomé M, García-Sevilla JA. Regulation of cannabinoid CB 2 receptor constitutive activity in vivo: repeated treatments with inverse agonists reverse the acute activation of JNK and associated apoptotic signaling in mouse brain. Psychopharmacology (Berl) 2017; 234:925-941. [PMID: 28127623 DOI: 10.1007/s00213-017-4537-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 01/07/2017] [Indexed: 01/29/2023]
Abstract
RATIONALE CB2 receptors express constitutive activity and inverse agonists regulate receptor basal activity, which might be involved in death mechanisms. This study assessed the effects of a selective CB2 agonist (JWH133) and different CB2 inverse agonists (AM630, JTE907, raloxifene) on death pathways in brain. OBJECTIVES The acute (JWH13) and the acute/chronic effects (AM630, JTE907, raloxifene) of CB2 ligands regulating pro-apoptotic c-Jun NH2-terminal kinase (p-JNK/JNK ratio) and associated signaling of extrinsic (Fas receptor, Fas-Associated death domain protein, FADD) and intrinsic (Bax, cytochrome c) death pathways (nuclear poly (ADP-ribose) polymerase PARP) were investigated in mouse brain. METHODS Mice were treated with CB2 drugs and target protein contents were assessed by western blot analysis. RESULTS JWH133 reduced cortical JNK (-27-45%) whereas AM630 acutely increased JNK in cortex (+61-148%), cerebellum (+34-40%), and striatum (+33-42%). JTE907 and raloxifene also increased cortical JNK (+31%-57%). Acute AM630, but not JWH133, increased cortical FADD, Bax, cytochrome c, and PARP cleavage. Repeated treatments with the three CB2 inverse agonists were associated with a reversal of the acute effects resulting in decreases in cortical JNK (AM630: -36%; JTE907: -25%; raloxifene: -11%). Chronic treatments also induced a reversal with down-regulation (AM630) or only tolerance (JTE907 and raloxifene) on other apoptotic markers (FADD, Bax, cytochrome c, PARP). CONCLUSIONS AM630 and JTE907 are CB2 protean ligands. Thus, chronic inverse agonists abolished CB2 constitutive activity and then the ligands behaved as agonists reducing (like JWH133) JNK activity. Acute and chronic treatments with CB2 inverse agonists regulate in opposite directions brain death markers.
Collapse
Affiliation(s)
- Glòria Salort
- Laboratori de Neurofarmacologia, Institut Universitari d'Investigació en Ciències de la Salut (IUNICS/IdisPa), Universitat de les Illes Balears, Cra. Valldemossa km 7.5, E-07122, Palma de Mallorca, Spain.,Redes Temáticas de Investigación Cooperativa en Salud-Red de Trastornos Adictivos (RETICS-RTA), ISCIII, Madrid, Spain
| | - María Álvaro-Bartolomé
- Laboratori de Neurofarmacologia, Institut Universitari d'Investigació en Ciències de la Salut (IUNICS/IdisPa), Universitat de les Illes Balears, Cra. Valldemossa km 7.5, E-07122, Palma de Mallorca, Spain.,Redes Temáticas de Investigación Cooperativa en Salud-Red de Trastornos Adictivos (RETICS-RTA), ISCIII, Madrid, Spain
| | - Jesús A García-Sevilla
- Laboratori de Neurofarmacologia, Institut Universitari d'Investigació en Ciències de la Salut (IUNICS/IdisPa), Universitat de les Illes Balears, Cra. Valldemossa km 7.5, E-07122, Palma de Mallorca, Spain. .,Redes Temáticas de Investigación Cooperativa en Salud-Red de Trastornos Adictivos (RETICS-RTA), ISCIII, Madrid, Spain.
| |
Collapse
|
23
|
Chen CC, Lee TY, Kwok CF, Hsu YP, Shih KC, Lin YJ, Ho LT. Using proteomics to discover novel biomarkers for fatty liver development and response to CB1R antagonist treatment in an obese mouse model. Proteomics 2017; 17. [DOI: 10.1002/pmic.201600292] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 11/23/2016] [Accepted: 11/25/2016] [Indexed: 12/12/2022]
Affiliation(s)
- Chin-Chang Chen
- Institute of Physiology; National Yang-Ming University; Taipei Taiwan
- Graduate Institute of Traditional Chinese Medicine; Chang Gung University; Taoyuan Taiwan
| | - Tzung-Yan Lee
- Graduate Institute of Traditional Chinese Medicine; Chang Gung University; Taoyuan Taiwan
| | - Ching-Fai Kwok
- Division of Endocrinology and Metabolism; Department of Medicine; Taipei Veterans General Hospital; Taipei Taiwan
| | - Yung-Pei Hsu
- Department of Medical Research; Taipei Veterans General Hospital; Taipei Taiwan
| | - Kuang-Chung Shih
- Department of Medicine-Metabolism; Cheng Hsin General Hospital; Taipei Taiwan
| | - Yan-Jie Lin
- Department of Research Planning and Development; National Health Research Institutes; Miaoli Taiwan
| | - Low-Tone Ho
- Institute of Physiology; National Yang-Ming University; Taipei Taiwan
- Division of Endocrinology and Metabolism; Department of Medicine; Taipei Veterans General Hospital; Taipei Taiwan
- Department of Medical Research; Taipei Veterans General Hospital; Taipei Taiwan
- School of Medicine; National Yang-Ming University; Taipei Taiwan
| |
Collapse
|
24
|
|
25
|
Gueye AB, Pryslawsky Y, Trigo JM, Poulia N, Delis F, Antoniou K, Loureiro M, Laviolette SR, Vemuri K, Makriyannis A, Le Foll B. The CB1 Neutral Antagonist AM4113 Retains the Therapeutic Efficacy of the Inverse Agonist Rimonabant for Nicotine Dependence and Weight Loss with Better Psychiatric Tolerability. Int J Neuropsychopharmacol 2016; 19:pyw068. [PMID: 27493155 PMCID: PMC5203757 DOI: 10.1093/ijnp/pyw068] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 07/31/2016] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Multiple studies suggest a pivotal role of the endocannabinoid system in regulating the reinforcing effects of various substances of abuse. Rimonabant, a CB1 inverse agonist found to be effective for smoking cessation, was associated with an increased risk of anxiety and depression. Here we evaluated the effects of the CB1 neutral antagonist AM4113 on the abuse-related effects of nicotine and its effects on anxiety and depressive-like behavior in rats. METHODS Rats were trained to self-administer nicotine under a fixed-ratio 5 or progressive-ratio schedules of reinforcement. A control group was trained to self-administer food. The acute/chronic effects of AM4113 pretreatment were evaluated on nicotine taking, motivation for nicotine, and cue-, nicotine priming- and yohimbine-induced reinstatement of nicotine-seeking. The effects of AM4113 in the basal firing and bursting activity of midbrain dopamine neurons were evaluated in a separate group of animals treated with nicotine. Anxiety/depression-like effects of AM4113 and rimonabant were evaluated 24h after chronic (21 days) pretreatment (0, 1, 3, and 10mg/kg, 1/d). RESULTS AM4113 significantly attenuated nicotine taking, motivation for nicotine, as well as cue-, priming- and stress-induced reinstatement of nicotine-seeking behavior. These effects were accompanied by a decrease of the firing and burst rates in the ventral tegmental area dopamine neurons in response to nicotine. On the other hand, AM4113 pretreatment did not have effects on operant responding for food. Importantly, AM4113 did not have effects on anxiety and showed antidepressant-like effects. CONCLUSION Our results indicate that AM4113 could be a promising therapeutic option for the prevention of relapse to nicotine-seeking while lacking anxiety/depression-like side effects.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Bernard Le Foll
- Translational Addiction Research Laboratory (Dr Gueye, Mr Pryslawsky, Dr Trigo, and Dr Le Foll), Alcohol Research and Treatment Clinic, Addiction Medicine Services, Ambulatory Care and Structured Treatments (Dr Le Foll), and Campbell Family Mental Health Research Institute (Dr Le Foll), Centre for Addiction and Mental Health, Toronto, ON, Canada; Department of Family and Community Medicine, Department of Pharmacology, and Department of Psychiatry, Division of Brain and Therapeutics, University of Toronto, Toronto, ON, Canada (Dr Le Foll); Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada (Dr Le Foll); Department of Pharmacology, Faculty of Medicine, School of Health Sciences, University of Ioannina, Greece (Ms Poulia and Drs Delis and Antoniou); Department of Anatomy and Cell Biology, The Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada (Drs Loureiro and Laviolette); Center for Drug Discovery, Department of Pharmaceutical Sciences and Department of Chemistry and Chemical Biology, Northeastern University, Boston, MA (Drs Vemuri and Makriyannis).
| |
Collapse
|
26
|
Blockade of Nicotine and Cannabinoid Reinforcement and Relapse by a Cannabinoid CB1-Receptor Neutral Antagonist AM4113 and Inverse Agonist Rimonabant in Squirrel Monkeys. Neuropsychopharmacology 2016; 41:2283-93. [PMID: 26888056 PMCID: PMC4946059 DOI: 10.1038/npp.2016.27] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Revised: 02/08/2016] [Accepted: 02/16/2016] [Indexed: 01/08/2023]
Abstract
Nicotine, the main psychoactive component of tobacco, and (-)-Δ(9)-tetrahydrocannabinol (THC), the main psychoactive ingredient in cannabis, play major roles in tobacco and marijuana dependence as reinforcers of drug-seeking and drug-taking behavior. Drugs that act as inverse agonists of cannabinoid CB1 receptors in the brain can attenuate the rewarding and abuse-related effects of nicotine and THC, but their clinical use is hindered by potentially serious side effects. The recently developed CB1-receptor neutral antagonists may provide an alternative therapeutic approach to nicotine and cannabinoid dependence. Here we compare attenuation of nicotine and THC reinforcement and reinstatement in squirrel monkeys by the CB1-receptor inverse agonist rimonabant and by the recently developed CB1-receptor neutral antagonist AM4113. Both rimonabant and AM4113 reduced two effects of nicotine and THC that play major roles in tobacco and marijuana dependence: (1) maintenance of high rates of drug-taking behavior, and (2) priming- or cue-induced reinstatement of drug-seeking behavior in abstinent subjects (models of relapse). In contrast, neither rimonabant nor AM4113 modified cocaine-reinforced or food-reinforced operant behavior under similar experimental conditions. However, both rimonabant and AM4113 reduced cue-induced reinstatement in monkeys trained to self-administer cocaine, suggesting the involvement of a common cannabinoid-mediated mechanism in the cue-induced reinstatement for different drugs of abuse. These findings point to CB1-receptor neutral antagonists as a new class of medications for treatment of both tobacco dependence and cannabis dependence.
Collapse
|
27
|
Dickens MJ, Vecchiarelli HA, Hill MN, Bentley GE. Endocannabinoid Signaling in the Stress Response of Male and Female Songbirds. Endocrinology 2015; 156:4649-59. [PMID: 26431225 PMCID: PMC4655215 DOI: 10.1210/en.2015-1425] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Endocannabinoid (eCB) signaling plays an important role in the stress response pathways of the mammalian brain, yet its role in the avian stress response has not been described. Understanding eCB signaling in avian species (such as the European starling, Sturnus vulgaris) allows a model system that exhibits natural attenuation of hypothalamic-pituitary-adrenal (HPA) responsiveness to stressors. Specifically, seasonally breeding birds exhibit the highest HPA activity during the breeding season and subsequently exhibit a robust HPA down-regulation during molt. Because eCB signaling in mammals has an overall inhibitory effect on HPA activity, we expected shifts in eCB signaling to regulate the seasonal HPA down-regulation during molt. However, our data did not support a role for eCB signaling in the molt-related suppression of HPA activity. For example, injection of the cannabinoid receptor (CB1) antagonist, AM251, did not potentiate molt-suppressed HPA activity. Instead, our data suggest eCB regulation of HPA plasticity as birds transition from breeding to molt. In support of this hypothesis, birds in the late breeding season demonstrated a more dynamic response at the level of avian amygdala eCB content in response to acute stress. The response and directionality of this effect match that seen in mammals. Overall, our data suggest that eCB signaling may allow for a dynamic range in HPA responsiveness (eg, breeding), but the signaling pathway's role may be limited when the HPA response is restrained (eg, molt). This first characterization of eCB signaling in the avian stress response also emphasizes that although the system functions similarly to other species, its exact role may be species specific.
Collapse
Affiliation(s)
- Molly J Dickens
- Department of Integrative Biology (M.J.D., G.E.B.), University of California, Berkeley, Berkeley, California 94609; Department of Cell Biology and Anatomy (H.A.V., M.N.H.), Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada T2N 1N4; and Helen Wills Neuroscience Institute (G.E.B.), University of California, Berkeley, Berkeley, California 94720-1650
| | - Haley A Vecchiarelli
- Department of Integrative Biology (M.J.D., G.E.B.), University of California, Berkeley, Berkeley, California 94609; Department of Cell Biology and Anatomy (H.A.V., M.N.H.), Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada T2N 1N4; and Helen Wills Neuroscience Institute (G.E.B.), University of California, Berkeley, Berkeley, California 94720-1650
| | - Matthew N Hill
- Department of Integrative Biology (M.J.D., G.E.B.), University of California, Berkeley, Berkeley, California 94609; Department of Cell Biology and Anatomy (H.A.V., M.N.H.), Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada T2N 1N4; and Helen Wills Neuroscience Institute (G.E.B.), University of California, Berkeley, Berkeley, California 94720-1650
| | - George E Bentley
- Department of Integrative Biology (M.J.D., G.E.B.), University of California, Berkeley, Berkeley, California 94609; Department of Cell Biology and Anatomy (H.A.V., M.N.H.), Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada T2N 1N4; and Helen Wills Neuroscience Institute (G.E.B.), University of California, Berkeley, Berkeley, California 94720-1650
| |
Collapse
|
28
|
Simone JJ, Malivoire BL, McCormick CM. Effects of CB1 receptor agonism and antagonism on behavioral fear and physiological stress responses in adult intact, ovariectomized, and estradiol-replaced female rats. Neuroscience 2015; 306:123-37. [PMID: 26311003 DOI: 10.1016/j.neuroscience.2015.08.032] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Revised: 07/29/2015] [Accepted: 08/15/2015] [Indexed: 12/16/2022]
Abstract
There is growing interest in the development of cannabis-based therapies for the treatment of fear and anxiety disorders. There are a few studies, but none in females, of the effects of the highly selective cannabinoid receptor type 1 (CB1) agonist, arachidonyl 2'-chlorethylamide (ACEA), on behavioral fear. In experiment 1 involving gonadally-intact females, ACEA (either 0.1 or 0.01 mg/kg) was without effect in the elevated plus maze (EPM), and the lower dose decreased anxiety in the open field test (OFT). AM251 increased anxiety in the EPM and decreased locomotor activity in the OFT. Twenty-four hours after fear conditioning, neither ACEA nor AM251 affected generalized fear or conditioned fear recall. AM251 and 0.1 mg/kg ACEA impaired, and 0.01 mg/kg ACEA enhanced, within-session fear extinction. AM251 increased plasma corticosterone concentrations after the fear extinction session, whereas ACEA was without effect. Based on evidence that estradiol may moderate the effects of CB1 receptor signaling in females, experiment 2 involved ovariectomized (OVX) rats provided with 10-μg 17β-Estradiol and compared with OVX rats without hormone replacement (oil vehicle). Irrespective of hormone treatment, AM251 increased anxiety in the EPM, whereas ACEA (0.01 mg/kg) was without effect. Neither hormone nor drug altered anxiety in the OFT, but estradiol increased and AM251 decreased distance traveled. After fear conditioning, AM251 decreased generalized fear. Neither hormone nor drug had any effect on recall or extinction of conditioned fear, however, ACEA and AM251 increased fear-induced plasma corticosterone concentrations. Further, when results with intact rats were compared with those from OVX rats, gonadal status did not moderate the effects of either AM251 or ACEA, although OVX displayed greater anxiety and fear than did intact rats. Thus, the effects of CB1 receptor antagonism and agonism in adult female rats do not depend on ovarian estradiol.
Collapse
Affiliation(s)
- J J Simone
- Department of Biological Sciences, Brock University, 500 Glenridge Avenue, St. Catharines, ON L2S 3A1, Canada.
| | - B L Malivoire
- Department of Biological Sciences, Brock University, 500 Glenridge Avenue, St. Catharines, ON L2S 3A1, Canada; Department of Psychology, Brock University, 500 Glenridge Avenue, St. Catharines, ON L2S 3A1, Canada.
| | - C M McCormick
- Department of Biological Sciences, Brock University, 500 Glenridge Avenue, St. Catharines, ON L2S 3A1, Canada; Department of Psychology, Brock University, 500 Glenridge Avenue, St. Catharines, ON L2S 3A1, Canada; Centre for Neuroscience, Brock University, 500 Glenridge Avenue, St. Catharines, ON L2S 3A1, Canada.
| |
Collapse
|
29
|
Lee TTY, Hill MN, Hillard CJ, Gorzalka BB. Disruption of peri-adolescent endocannabinoid signaling modulates adult neuroendocrine and behavioral responses to stress in male rats. Neuropharmacology 2015; 99:89-97. [PMID: 26192544 DOI: 10.1016/j.neuropharm.2015.07.021] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Revised: 07/14/2015] [Accepted: 07/16/2015] [Indexed: 10/23/2022]
Abstract
The endocannabinoid (eCB) system is known to regulate neural, endocrine and behavioral responses to stress in adults; however there is little knowledge regarding how this system governs the development and maturation of these responses. Previous work has reported dynamic and time-specific changes in CB1 receptor expression, N-arachidonylethanolamine (AEA) content and fatty acid amide hydrolase (FAAH) activity within corticolimbic structures throughout the peri-adolescent period. To examine whether fluctuations in adolescent eCB activity contribute to the development of adult stress responsivity and emotionality, we treated male Sprague-Dawley rats daily with the CB1R antagonist, AM-251 (5 mg/kg), or vehicle between post-natal days (PND) 35-45. Following this treatment, emotional behavior, HPA axis stress reactivity and habituation to repeated restraint stress, as well as corticolimbic eCB content were examined in adulthood (PND 75). Behaviorally, AM-251-treated males exhibited more active stress-coping behavior in the forced swim test, greater risk assessment behavior in the elevated plus maze and no significant differences in general motor activity. Peri-adolescent AM-251 treatment modified corticosterone habituation to repeated restraint exposure compared to vehicle. Peri-adolescent CB1R antagonism induced moderate changes in adult corticolimbic eCB signaling, with a significant decrease in amygdalar AEA, an increase in hypothalamic AEA and an increase in prefrontal cortical CB1R expression. Together, these data indicate that peri-adolescent endocannabinoid signaling contributes to the maturation of adult neurobehavioral responses to stress.
Collapse
Affiliation(s)
- Tiffany T-Y Lee
- Dept. of Psychology, University of British Columbia, Vancouver, V6T 1Z4, Canada
| | - Matthew N Hill
- Hotchkiss Brain Institute, Dept. of Cell Biology and Anatomy, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Cecilia J Hillard
- Dept. of Pharmacology and Toxicology and Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Boris B Gorzalka
- Dept. of Psychology, University of British Columbia, Vancouver, V6T 1Z4, Canada.
| |
Collapse
|
30
|
Krug RG, Clark KJ. Elucidating cannabinoid biology in zebrafish (Danio rerio). Gene 2015; 570:168-79. [PMID: 26192460 DOI: 10.1016/j.gene.2015.07.036] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2015] [Accepted: 07/11/2015] [Indexed: 02/01/2023]
Abstract
The number of annual cannabinoid users exceeds 100,000,000 globally and an estimated 9% of these individuals will suffer from dependency. Although exogenous cannabinoids, like those contained in marijuana, are known to exert their effects by disrupting the endocannabinoid system, a dearth of knowledge exists about the potential toxicological consequences on public health. Conversely, the endocannabinoid system represents a promising therapeutic target for a plethora of disorders because it functions to endogenously regulate a vast repertoire of physiological functions. Accordingly, the rapidly expanding field of cannabinoid biology has sought to leverage model organisms in order to provide both toxicological and therapeutic insights about altered endocannabinoid signaling. The primary goal of this manuscript is to review the existing field of cannabinoid research in the genetically tractable zebrafish model-focusing on the cannabinoid receptor genes, cnr1 and cnr2, and the genes that produce enzymes for synthesis and degradation of the cognate ligands anandamide and 2-arachidonylglycerol. Consideration is also given to research that has studied the effects of exposure to exogenous phytocannabinoids and synthetic cannabinoids that are known to interact with cannabinoid receptors. These results are considered in the context of either endocannabinoid gene expression or endocannabinoid gene function, and are integrated with findings from rodent studies. This provides the framework for a discussion of how zebrafish may be leveraged in the future to provide novel toxicological and therapeutic insights in the field of cannabinoid biology, which has become increasingly significant given recent trends in cannabis legislation.
Collapse
Affiliation(s)
- Randall G Krug
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA; Mayo Graduate School, Neurobiology of Disease Track, Mayo Clinic, Rochester, MN, USA
| | - Karl J Clark
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
31
|
Differential effects of CB1 receptor agonism in behavioural tests of unconditioned and conditioned fear in adult male rats. Behav Brain Res 2015; 279:9-16. [DOI: 10.1016/j.bbr.2014.11.012] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Revised: 10/07/2014] [Accepted: 11/05/2014] [Indexed: 11/24/2022]
|
32
|
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
|
33
|
Abstract
Preclinical and clinical data fully support the involvement of the endocannabinoid system in the etiopathogenesis of several mental diseases. In this review we will briefly summarize the most common alterations in the endocannabinoid system, in terms of cannabinoid receptors and endocannabinoid levels, present in mood disorders (anxiety, posttraumatic stress disorder, depression, bipolar disorder, and suicidality) as well as psychosis (schizophrenia) and autism. The arising picture for each pathology is not always straightforward; however, both animal and human studies seem to suggest that pharmacological modulation of this system might represent a novel approach for treatment.
Collapse
Affiliation(s)
- Tiziana Rubino
- Department of Theoretical and Applied Sciences and Neuroscience Centre, University of Insubria, Via Manara 7, 21052, Busto Arsizio, VA, Italy
| | - Erica Zamberletti
- Department of Theoretical and Applied Sciences and Neuroscience Centre, University of Insubria, Via Manara 7, 21052, Busto Arsizio, VA, Italy
- Fondazione Zardi Gori, Milan, Italy
| | - Daniela Parolaro
- Department of Theoretical and Applied Sciences and Neuroscience Centre, University of Insubria, Via Manara 7, 21052, Busto Arsizio, VA, Italy.
- Fondazione Zardi Gori, Milan, Italy.
| |
Collapse
|
34
|
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
|
35
|
Rock EM, Sticht MA, Duncan M, Stott C, Parker LA. Evaluation of the potential of the phytocannabinoids, cannabidivarin (CBDV) and Δ(9) -tetrahydrocannabivarin (THCV), to produce CB1 receptor inverse agonism symptoms of nausea in rats. Br J Pharmacol 2014; 170:671-8. [PMID: 23902479 DOI: 10.1111/bph.12322] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Revised: 07/08/2013] [Accepted: 07/24/2013] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND AND PURPOSE The cannabinoid 1 (CB1 ) receptor inverse agonists/antagonists, rimonabant (SR141716, SR) and AM251, produce nausea and potentiate toxin-induced nausea by inverse agonism (rather than antagonism) of the CB1 receptor. Here, we evaluated two phytocannabinoids, cannabidivarin (CBDV) and Δ(9) -tetrahydrocannabivarin (THCV), for their ability to produce these behavioural effect characteristics of CB1 receptor inverse agonism in rats. EXPERIMENTAL APPROACH In experiment 1, we investigated the potential of THCV and CBDV to produce conditioned gaping (measure of nausea-induced behaviour) in the same manner as SR and AM251. In experiment 2, we investigated the potential of THCV and CBDV to enhance conditioned gaping produced by a toxin in the same manner as CB1 receptor inverse agonists. KEY RESULTS SR (10 and 20 mg·kg(-1) ) and AM251 (10 mg·kg(-1) ) produced conditioned gaping; however, THCV (10 or 20 mg·kg(-1) ) and CBDV (10 or 200 mg·kg(-1) ) did not. At a subthreshold dose for producing nausea, SR (2.5 mg·kg(-1) ) enhanced lithium chloride (LiCl)-induced conditioned gaping, whereas Δ(9) -tetrahydrocannabinol (THC, 2.5 and 10 mg·kg(-1) ), THCV (2.5 or 10 mg·kg(-1) ) and CBDV (2.5 or 200 mg·kg(-1) ) did not; in fact, THC (2.5 and 10 mg·kg(-1) ), THCV (10 mg·kg(-1) ) and CBDV (200 mg·kg(-1) ) suppressed LiCl-induced conditioned gaping, suggesting anti-nausea potential. CONCLUSIONS AND IMPLICATIONS The pattern of findings indicates that neither THCV nor CBDV produced a behavioural profile characteristic of CB1 receptor inverse agonists. As well, these compounds may have therapeutic potential in reducing nausea.
Collapse
Affiliation(s)
- Erin M Rock
- Department of Psychology and Neuroscience Graduate Program, University of Guelph, Guelph, ON, Canada
| | | | | | | | | |
Collapse
|
36
|
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
|
37
|
The microinjection of a cannabinoid agonist into the accumbens shell induces anxiogenesis in the elevated plus-maze. Pharmacol Biochem Behav 2014; 124:160-6. [PMID: 24887448 DOI: 10.1016/j.pbb.2014.05.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Revised: 05/19/2014] [Accepted: 05/22/2014] [Indexed: 11/23/2022]
Abstract
This study investigated the effect of a cannabinoid agonist injected into the shell region of the nucleus accumbens (nAcb shell) on anxiety-related behaviors. The animals (male Wistar rats) were unilaterally microinjected with either ACEA (arachidonyl-2'-chloroethylamide a CB1 receptor agonist) at doses of 0.005, 0.05 or 0.5 pmol, or vehicle (ethanol 0.04% in saline 0.9%) and submitted to the elevated plus-maze (EPM), a pre-clinical test of anxiety. The data showed that rats microinjected with ACEA (0.05 pmol/0.2 μl) into the nAcb shell exhibited decreased % open arm time and open arm entries in comparison with the control group, which is compatible with an anxiogenic-like effect. To rule out the hypothesis that spread of the drug into the ventricle was responsible for the observed anxiogenic effect, 0.05 pmol ACEA was injected into the lateral ventricle and shown not to alter the responses representative of fear/anxiety and locomotion. The locomotor activity was not changed at the dose of 0.05 pmol ACEA microinjected into the nAcb shell. The present data suggest that activation of cannabinoid receptors in the nAcb shell may modulate fear/anxiety in the EPM.
Collapse
|
38
|
Makriyannis A. 2012 Division of medicinal chemistry award address. Trekking the cannabinoid road: a personal perspective. J Med Chem 2014; 57:3891-911. [PMID: 24707904 DOI: 10.1021/jm500220s] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
My involvement with the field of cannabinoids spans close to 3 decades and covers a major part of my scientific career. It also reflects the robust progress in this initially largely unexplored area of biology. During this period of time, I have witnessed the growth of modern cannabinoid biology, starting from the discovery of its two receptors and followed by the characterization of its endogenous ligands and the identification of the enzyme systems involved in their biosynthesis and biotransformation. I was fortunate enough to start at the beginning of this new era and participate in a number of the new discoveries. It has been a very exciting journey. With coverage of some key aspects of my work during this period of "modern cannabinoid research," this Award Address, in part historical, intends to give an account of how the field grew, the key discoveries, and the most promising directions for the future.
Collapse
Affiliation(s)
- Alexandros Makriyannis
- Center for Drug Discovery and Departments of Chemistry and Chemical Biology and Pharmaceutical Sciences, Northeastern University , 360 Huntington Avenue, Boston, Massachusetts 02115, United States
| |
Collapse
|
39
|
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
|
40
|
Neutral antagonism at the cannabinoid 1 receptor: a safer treatment for obesity. Mol Psychiatry 2013; 18:1294-301. [PMID: 23070073 DOI: 10.1038/mp.2012.145] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2012] [Revised: 08/21/2012] [Accepted: 09/04/2012] [Indexed: 01/10/2023]
Abstract
Obesity is a global problem with often strong neurobiological underpinnings. The cannabinoid 1 receptor (CB1R) was put forward as a promising drug target for antiobesity medication. However, the first marketed CB1R antagonist/inverse agonist rimonabant was discontinued, as its use was occasionally associated with negative affect and suicidality. In artificial cell systems, CB1Rs can become constitutively active in the absence of ligands. Here, we show that such constitutive CB1R activity also regulates GABAergic and glutamatergic neurotransmission in the ventral tegmental area and basolateral amygdala, regions which regulate motivation and emotions. We show that CB1R inverse agonists like rimonabant suppress the constitutive CB1R activity in such regions, and cause anxiety and reduced motivation for reward. The neutral CB1R antagonist NESS0327 does not suppress constitutive activity and lacks these negative effects. Importantly, however, both rimonabant and NESS0327 equally reduce weight gain and food intake. Together, these findings suggest that neutral CB1R antagonists can treat obesity efficiently and more safely than inverse agonists.
Collapse
|
41
|
Gobira PH, Aguiar DC, Moreira FA. Effects of compounds that interfere with the endocannabinoid system on behaviors predictive of anxiolytic and panicolytic activities in the elevated T-maze. Pharmacol Biochem Behav 2013; 110:33-9. [DOI: 10.1016/j.pbb.2013.05.013] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2012] [Revised: 05/10/2013] [Accepted: 05/18/2013] [Indexed: 02/06/2023]
|
42
|
Akhtar MT, Ali S, Rashidi H, van der Kooy F, Verpoorte R, Richardson MK. Developmental effects of cannabinoids on zebrafish larvae. Zebrafish 2013; 10:283-93. [PMID: 23789728 DOI: 10.1089/zeb.2012.0785] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Cannabinoids are natural or synthetic compounds related chemically to (-)-(6aR,10aR)-6,6,9-trimethyl-3-pentyl-6a,7,8,10a-tetrahydro-6H-benzo[c]chromen-1-ol (Δ(9)-THC), the principle psychotropic constituent of the hemp plant, Cannabis sativa L. Here we examine the effects of the cannabinoids Δ(9)-THC, (R)-(+)-[2,3-dihydro-5-methyl-3-(4-morpholinylmethyl)pyrrolo [1,2,3-de]-1,4-benzoxazin-6-yl]-1-napthalenylmethanone and 2-[(1R,2R,5R)-5-hydroxy-2-(3-hydroxypropyl) cyclohexyl]-5-(2-methyloctan-2-yl)phenol, and the cannabinoid antagonist (AM 251). Exposures were either acute (1-12-h exposure at 108 hours of postfertilization [hpf]) or chronic (96-h exposure starting at 24 hpf). Geometric range finding was used to determine the experimental concentrations. The concentration of the chemical that kills 50% of the test animals in a given time (LC50) was determined based on cumulative mortality at 5 days of postfertilization. At day 5, behavioral analysis (visual motor response test) was carried out in which movement of individual larvae was analysed using automated video-tracking. With acute exposure, embryos showed a biphasic response to the dark challenge with all three cannabinoids tested. This response consisted of stimulation of the locomotor activity at low concentrations, suppression at high doses. With chronic exposure, embryos habituated to the effects of all three cannabinoids when assayed with the dark challenge phase. Further, the excitation was ameliorated when the antagonist AM 251 was coadministered with the cannabinoid. When AM 251 was administered on its own (chronically or acutely), the locomotor activity was suppressed at high concentrations. We examined the embryos for a range of malformations after chronic exposure to cannabinoid. Only Δ(9)-THC was associated with a significant increase in malformations at 5d (yolk sac and pericardial edema, bent tail/body axis). We conclude that cannabinoids have behavioral effects in zebrafish that are comparable to some of those reported in the literature for mammals. In particular, the acute exposure response resembles behavioral effects reported for adult rodents. Our data are consistent with these behavioral effects being mediated, at least in part, by the CB1 receptor.
Collapse
|
43
|
Litvin Y, Phan A, Hill MN, Pfaff DW, McEwen BS. CB1receptor signaling regulates social anxiety and memory. GENES BRAIN AND BEHAVIOR 2013; 12:479-89. [DOI: 10.1111/gbb.12045] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Revised: 03/15/2013] [Accepted: 04/27/2013] [Indexed: 01/31/2023]
Affiliation(s)
| | | | - M. N. Hill
- Laboratory of Neuroendocrinology; The Rockefeller University; New York; NY; USA
| | | | - B. S. McEwen
- Laboratory of Neuroendocrinology; The Rockefeller University; New York; NY; USA
| |
Collapse
|
44
|
Micale V, Di Marzo V, Sulcova A, Wotjak CT, Drago F. Endocannabinoid system and mood disorders: Priming a target for new therapies. Pharmacol Ther 2013; 138:18-37. [DOI: 10.1016/j.pharmthera.2012.12.002] [Citation(s) in RCA: 159] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
45
|
O'Brien LD, Wills KL, Segsworth B, Dashney B, Rock EM, Limebeer CL, Parker LA. Effect of chronic exposure to rimonabant and phytocannabinoids on anxiety-like behavior and saccharin palatability. Pharmacol Biochem Behav 2012; 103:597-602. [PMID: 23103902 DOI: 10.1016/j.pbb.2012.10.008] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2012] [Revised: 10/11/2012] [Accepted: 10/17/2012] [Indexed: 01/14/2023]
Abstract
The acute effects of cannabinoid compounds have been investigated in animal models of anxiety-like behavior and palatability processing. However, the chronic effects of cannabinoids in such models are poorly understood. Experiment 1 compared the effects of both acute and chronic (14 days) exposure to the CB(1) receptor inverse agonist/antagonist, rimonabant, and the cannabis-derived CB(1) receptor neutral antagonist, tetrahydrocannabivarin (THCV), on: 1) time spent in the open, lit box in the Light-Dark (LD) immersion model of anxiety-like behavior and 2) saccharin hedonic reactions in the taste reactivity (TR) test of palatability processing. Experiment 2 compared the effects of chronic administration of cannabis-derived Δ(9)-tetrahydrocannabinol (Δ(9)-THC), cannabidiol (CBD) and cannabigerol (CBG) in these models. Tests were administered on Days 1, 7 and 14 of drug administration. In Experiment 1, rimonabant, but not THCV, produced an anxiogenic-like reaction in the LD immersion test and reduced saccharin palatability in the TR test; both of these effects occurred acutely and were not enhanced by chronic exposure. In Experiment 2, Δ(9)-THC also produced an acute anxiogenic-like reaction in the LD immersion test, without enhancement by chronic exposure. However, Δ(9)-THC enhanced saccharin palatability in the TR test on Day 1 of drug exposure only. CBD and CBG did not modify anxiety-like responding, but CBG produced a weak enhancement of saccharin palatability on Day 1 only. The results suggest that the anxiogenic-like reactions and the suppression of hedonic responding produced by rimonabant, are mediated by inverse agonism of the CB(1) receptor and these effects are not enhanced with chronic exposure.
Collapse
Affiliation(s)
- Lesley D O'Brien
- Department of Psychology and Collaborative Neuroscience Program, University of Guelph, Guelph, ON, Canada N1G 2W1
| | | | | | | | | | | | | |
Collapse
|
46
|
Aydin C, Oztan O, Isgor C. Nicotine-induced anxiety-like behavior in a rat model of the novelty-seeking phenotype is associated with long-lasting neuropeptidergic and neuroplastic adaptations in the amygdala: effects of the cannabinoid receptor 1 antagonist AM251. Neuropharmacology 2012; 63:1335-45. [PMID: 22959963 DOI: 10.1016/j.neuropharm.2012.08.016] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2011] [Revised: 08/15/2012] [Accepted: 08/20/2012] [Indexed: 01/21/2023]
Abstract
A rat model of the novelty-seeking phenotype predicts vulnerability to the expression of behavioral sensitization to nicotine, where locomotor reactivity to novelty is used to screen experimentally-naïve rats for high (HR) versus low (LR) responders. The present study examines the long-term neuropeptidergic and neuroplastic adaptations associated with the expression of locomotor sensitization to a low dose nicotine challenge and social anxiety-like behavior following chronic intermittent nicotine exposure during adolescence in the LRHR phenotype. Our data show that the expression of behavioral sensitization to nicotine and abstinence-related anxiety are detected in nicotine pre-exposed HRs even across a long (3 wks) abstinence. Moreover, these behavioral effects of nicotine are accompanied by a persistent imbalance between neuropeptide Y and corticotrophin releasing factor systems, and a persistent increase in brain-derived neurotrophic factor (BDNF) and spinophilin mRNA levels in the amygdala. Furthermore, treatment with the cannabinoid receptor 1 antagonist, AM251 (5 mg/kg) during a short (1 wk) abstinence is ineffective in reversing nicotine-induced anxiety, fluctuations in BDNF and spinophilin mRNAs, and the neuropeptidergic dysregulations in the amygdala; although this treatment is effective in reversing the expression of locomotor sensitization to challenge nicotine even after a long abstinence. Interestingly, the identical AM251 treatment administered during the late phase of a long abstinence further augments anxiety and associated changes in BDNF and spinophilin mRNA in the basolateral nucleus of the amygdala in nicotine pre-exposed HRs. These findings implicate long-lasting neuropeptidergic and neuroplastic changes in the amygdala in vulnerability to the behavioral effects of nicotine in the novelty-seeking phenotype.
Collapse
Affiliation(s)
- Cigdem Aydin
- Charles E. Schmidt College of Medicine, Department of Biomedical Science, Florida Atlantic University, 777 Glades Road, Boca Raton, FL 33431, USA
| | | | | |
Collapse
|
47
|
Sciolino NR, Dishman RK, Holmes PV. Voluntary exercise offers anxiolytic potential and amplifies galanin gene expression in the locus coeruleus of the rat. Behav Brain Res 2012; 233:191-200. [PMID: 22580167 PMCID: PMC3409590 DOI: 10.1016/j.bbr.2012.05.001] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2012] [Revised: 04/05/2012] [Accepted: 05/01/2012] [Indexed: 01/04/2023]
Abstract
Although exercise improves anxiety in humans, it is controversial whether exercise is anxiolytic in rodents. We tested the hypothesis that stress influences the effect of exercise on anxiety-like and defensive behaviors. To explore the neurobiological mechanisms of exercise, we also examined whether exercise alters gene expression for the stress-related peptide galanin. Rats were housed in the presence or absence of a running wheel for 21 d. A subset of these rats were (1) not injected or received a single high, dose of the β-carboline FG7142 (inverse agonist at the benzodiazepine receptor site) immediately prior to testing or (2) were injected repeatedly with vehicle or FG7142 during the last 10d of exercise. On day 22, anxiety-like and defensive behaviors were measured in the elevated plus maze, shock probe defensive burying, and defensive withdrawal tests. Locus coeruleus prepro-galanin mRNA was measured by in situ hybridization. Exercise and sedentary rats that were not injected exhibited similar behavior in all tests, whereas FG7142 injected immediately prior to the test battery produced intense avoidance and immobility consistent with an anxiety-like response. However, exercise produced anxiolytic-like and active defensive behaviors in the test battery relative to the sedentary condition in rats injected repeatedly with vehicle or FG7142. Exercise also increased prepro-galanin mRNA in the locus coeruleus relative to sedentary controls. These data suggest that the emergence of enhanced adaptive behavior after chronic voluntary exercise is influenced by stress. Our data support a role for galanin in the beneficial consequences of wheel running.
Collapse
Affiliation(s)
- Natale R. Sciolino
- Interdisciplinary Neuroscience Program, Biomedical and Health Sciences Institute, University of Georgia
| | | | - Philip V. Holmes
- Interdisciplinary Neuroscience Program, Biomedical and Health Sciences Institute, University of Georgia
- Department of Psychology, University of Georgia
| |
Collapse
|
48
|
Silvestri C, Di Marzo V. Second generation CB1 receptor blockers and other inhibitors of peripheral endocannabinoid overactivity and the rationale of their use against metabolic disorders. Expert Opin Investig Drugs 2012; 21:1309-22. [DOI: 10.1517/13543784.2012.704019] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Cristoforo Silvestri
- Institute of Biomolecular Chemistry – CNR, Endocannabinoid Research Group,
Via Campi Flegrei 34, 80078, Pozzuoli (NA), Italy ;
| | - Vincenzo Di Marzo
- Institute of Biomolecular Chemistry – CNR, Endocannabinoid Research Group,
Via Campi Flegrei 34, 80078, Pozzuoli (NA), Italy ;
| |
Collapse
|
49
|
Segovia KN, Correa M, Lennington JB, Conover JC, Salamone JD. Changes in nucleus accumbens and neostriatal c-Fos and DARPP-32 immunoreactivity during different stages of food-reinforced instrumental training. Eur J Neurosci 2012; 35:1354-67. [PMID: 22462413 DOI: 10.1111/j.1460-9568.2012.08036.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
Nucleus accumbens is involved in several aspects of instrumental behavior, motivation and learning. Recent studies showed that dopamine (DA) release in the accumbens shell was significantly increased on the first day of training on a fixed ratio (FR) 5 schedule (i.e. the transition from FR1 to FR5) compared with those rats that continued FR1 training, even though the rats on their first day of FR5 training received less food reinforcement than rats continuing on the FR1 schedule. Additionally, the second day of FR5 responding was marked by a significant increase in DA release in accumbens core. The present studies employed immunohistochemical methods to characterize the changes in cellular markers of accumbens and neostriatal neural activity that occur during various stages of food-reinforced FR5 training. c-Fos and DARPP-32 immunoreactivity in accumbens shell was significantly increased on the first day of FR5 training, while core c-Fos and DARPP-32 expression showed large increases on the second day of FR5 training. Additional studies showed that c-Fos and DARPP-32 expression in neostriatum increased after more extensive training. Double-labeling studies with immunofluorescence methods indicated that increases in accumbens c-Fos and DARPP-32 expression were primarily seen in substance-P-positive neurons. These increases in accumbens c-Fos and DARPP-32 immunoreactivity seen during the initial phases of FR training may reflect several factors, including novelty, learning, stress or the presentation of a work-related challenge to the organism. Moreover, it appears that the separate subregions of the striatal complex are differentially activated at distinct phases of instrumental training.
Collapse
Affiliation(s)
- Kristen N Segovia
- Department of Psychology, University of Connecticut, Storrs, CT 06269-1020, USA
| | | | | | | | | |
Collapse
|
50
|
Cornélio AM, Nunes-de-Souza RL, Morgan MM. Contribution of the rostral ventromedial medulla to post-anxiety induced hyperalgesia. Brain Res 2012; 1450:80-6. [PMID: 22418057 DOI: 10.1016/j.brainres.2012.02.037] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2011] [Revised: 02/06/2012] [Accepted: 02/14/2012] [Indexed: 10/28/2022]
Abstract
Rats exposed to an elevated plus maze (EPM) with four open arms display antinociception while on the maze and hyperalgesia immediately upon removal. Little is known about the neural mechanisms underlying EPM-induced antinociception and the subsequent hyperalgesia except that the antinociception is not mediated by endogenous opioids. The objective of the present study was to test the hypothesis that endogenous cannabinoids and/or the rostral ventromedial medulla (RVM) contributes to EPM-induced antinociception. Administration of the CB1 receptor antagonist AM251 (1mg/kg, i.p.) had no effect on baseline nociception to formalin administration into the hindpaw or on the antinociception produced by placing a rat on the open EPM. Likewise, inactivation of the RVM by microinjecting the GABA(A) receptor agonist muscimol (10 ng/0.5 μL) had no effect on the antinociceptive effect of placing a rat in the EPM. However, RVM inactivation blocked the hyperalgesia produced upon removal from the EPM. Although distinct classes of RVM neurons inhibit and facilitate nociception, the present data demonstrate that the antinociception induced by the EPM and the subsequent hyperalgesia is mediated by distinct neural pathways.
Collapse
Affiliation(s)
- Alianda Maira Cornélio
- Programa Interinstitucional de Pós-Graduação em Ciências Fisiológicas, UFSCar-UNESP, Brazil.
| | | | | |
Collapse
|