1
|
Iyer V, Saberi SA, Pacheco R, Sizemore EF, Stockman S, Kulkarni A, Cantwell L, Thakur GA, Hohmann AG. Negative allosteric modulation of CB 1 cannabinoid receptor signaling suppresses opioid-mediated tolerance and withdrawal without blocking opioid antinociception. Neuropharmacology 2024; 257:110052. [PMID: 38936657 PMCID: PMC11261750 DOI: 10.1016/j.neuropharm.2024.110052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 06/02/2024] [Accepted: 06/21/2024] [Indexed: 06/29/2024]
Abstract
The direct blockade of CB1 cannabinoid receptors produces therapeutic effects as well as adverse side-effects that limit their clinical potential. CB1 negative allosteric modulators (NAMs) represent an indirect approach to decrease the affinity and/or efficacy of orthosteric cannabinoid ligands or endocannabinoids at CB1. We recently reported that GAT358, a CB1-NAM, blocked opioid-induced mesocorticolimbic dopamine release and reward via a CB1-allosteric mechanism of action. Whether a CB1-NAM dampens opioid-mediated therapeutic effects such as analgesia or alters other unwanted opioid side-effects remain unknown. Here, we characterized the effects of GAT358 on nociceptive behaviors in the presence and absence of morphine in male rats. We examined the impact of GAT358 on formalin-evoked pain behavior and Fos protein expression, a marker of neuronal activation, in the lumbar spinal cord. We also assessed the impact of GAT358 on morphine-induced slowing of colonic transit, tolerance, and withdrawal behaviors in male mice. GAT358 attenuated morphine antinociceptive tolerance without blocking acute antinociception and reduced morphine-induced slowing of colonic motility without impacting fecal boli production. GAT358 also produced antinociception in the presence and absence of morphine in the formalin model of inflammatory nociception and reduced the number of formalin-evoked Fos protein-like immunoreactive cells in the lumbar spinal cord. Finally, GAT358 mitigated the somatic signs of naloxone-precipitated, but not spontaneous, opioid withdrawal following chronic morphine dosing. Our results support the therapeutic potential of CB1-NAMs as novel drug candidates aimed at preserving opioid-mediated analgesia while preventing their unwanted side-effects. Our studies also uncover previously unrecognized antinociceptive properties associated with an arrestin-biased CB1-NAM.
Collapse
Affiliation(s)
- Vishakh Iyer
- Program in Neuroscience, Indiana University, Bloomington, IN, USA; Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA
| | - Shahin A Saberi
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA
| | - Romario Pacheco
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA
| | - Emily Fender Sizemore
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA
| | - Sarah Stockman
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA
| | - Abhijit Kulkarni
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, USA
| | - Lucas Cantwell
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, USA
| | - Ganesh A Thakur
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, USA
| | - Andrea G Hohmann
- Program in Neuroscience, Indiana University, Bloomington, IN, USA; Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA; Gill Center for Biomolecular Science, Indiana University, Bloomington, IN, USA.
| |
Collapse
|
2
|
Iyer V, Saberi SA, Pacheco R, Sizemore EF, Stockman S, Kulkarni A, Cantwell L, Thakur GA, Hohmann AG. Negative allosteric modulation of cannabinoid CB 1 receptor signaling suppresses opioid-mediated tolerance and withdrawal without blocking opioid antinociception. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.06.574477. [PMID: 38260598 PMCID: PMC10802405 DOI: 10.1101/2024.01.06.574477] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
The direct blockade of CB 1 cannabinoid receptors produces therapeutic effects as well as adverse side-effects that limit their clinical potential. CB 1 negative allosteric modulators (NAMs) represent an indirect approach to decrease the affinity and/or efficacy of orthosteric cannabinoid ligands or endocannabinoids at CB 1 . We recently reported that GAT358, a CB 1 -NAM, blocked opioid-induced mesocorticolimbic dopamine release and reward via a CB 1 -allosteric mechanism of action. Whether a CB 1 -NAM dampens opioid-mediated therapeutic effects such as analgesia or alters other unwanted side-effects of opioids remain unknown. Here, we characterized the effects of GAT358 on nociceptive behaviors in the presence and absence of morphine. We examined the impact of GAT358 on formalin-evoked pain behavior and Fos protein expression, a marker of neuronal activation, in the lumbar dorsal horn. We also assessed the impact of GAT358 on morphine-induced slowing of colonic transit, tolerance, and withdrawal behaviors. GAT358 attenuated morphine antinociceptive tolerance without blocking acute antinociception. GAT358 also reduced morphine-induced slowing of colonic motility without impacting fecal boli production. GAT358 produced antinociception in the presence and absence of morphine in the formalin model of inflammatory nociception and reduced the number of formalin-evoked Fos protein-like immunoreactive cells in the lumbar spinal dorsal horn. Finally, GAT358 mitigated the somatic signs of naloxone-precipitated, but not spontaneous, opioid withdrawal following chronic morphine dosing in mice. Our results support the therapeutic potential of CB 1 -NAMs as novel drug candidates aimed at preserving opioid-mediated analgesia while preventing their unwanted side-effects. Our studies also uncover previously unrecognized antinociceptive properties associated with an arrestin-biased CB 1 -NAMs. Highlights CB 1 negative allosteric modulator (NAM) GAT358 attenuated morphine tolerance GAT358 reduced morphine-induced slowing of colonic motility but not fecal productionGAT358 was antinociceptive for formalin pain alone and when combined with morphineGAT358 reduced formalin-evoked Fos protein expression in the lumbar spinal cordGAT358 mitigated naloxone precipitated withdrawal after chronic morphine dosing.
Collapse
|
3
|
Low ZXB, Lee XR, Soga T, Goh BH, Alex D, Kumari Y. Cannabinoids: Emerging sleep modulator. Biomed Pharmacother 2023; 165:115102. [PMID: 37406510 DOI: 10.1016/j.biopha.2023.115102] [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: 04/20/2023] [Revised: 06/28/2023] [Accepted: 06/28/2023] [Indexed: 07/07/2023] Open
Abstract
Sleep is an essential biological phase of our daily life cycle and is necessary for maintaining homeostasis, alertness, metabolism, cognition, and other key functions across the animal kingdom. Dysfunctional sleep leads to deleterious effects on health, mood, and cognition, including memory deficits and an increased risk of diabetes, stroke, and neurological disorders. Sleep is regulated by several brain neuronal circuits, neuromodulators, and neurotransmitters, where cannabinoids have been increasingly found to play a part in its modulation. Cannabinoids, a group of lipid metabolites, are regulatory molecules that bind mainly to cannabinoid receptors (CB1 and CB2). Much evidence supports the role of cannabinoid receptors in the modulation of sleep, where their alteration exhibits sleep-promoting effects, including an increase in non-rapid-eye movement sleep and a reduction in sleep latency. However, the pharmacological alteration of CB1 receptors is associated with adverse psychotropic effects, which are not exhibited in CB2 receptor alteration. Hence, selective alteration of CB2 receptors is also of clinical importance, where it could potentially be used in treating sleep disorders. Thus, it is crucial to understand the neurobiological basis of cannabinoids in sleep physiology. In this review article, the alteration of the endocannabinoid system by various cannabinoids and their respective effects on the sleep-wake cycle are discussed based on recent findings. The mechanisms of the cannabinoid receptors on sleep and wakefulness are also explored for their clinical implications and potential therapeutic use on sleep disorders.
Collapse
Affiliation(s)
- Zhen Xuen Brandon Low
- Neurological Disorder and Aging Research Group (NDA), Neuroscience Research Strength (NRS), Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, 47500 Selangor, Malaysia
| | - Xin Ru Lee
- Neurological Disorder and Aging Research Group (NDA), Neuroscience Research Strength (NRS), Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, 47500 Selangor, Malaysia
| | - Tomoko Soga
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, 47500 Selangor, Malaysia
| | - Bey Hing Goh
- Biofunctional Molecule Exploratory Research Group, School of Pharmacy, Monash University Malaysia, Bandar Sunway, Selangor Darul Ehsan 47500, Malaysia; College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Deepa Alex
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, 47500 Selangor, Malaysia
| | - Yatinesh Kumari
- Neurological Disorder and Aging Research Group (NDA), Neuroscience Research Strength (NRS), Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, 47500 Selangor, Malaysia.
| |
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
|
Luo ML, Zhao Q, He XH, Xie X, Zhu HP, You FM, Peng C, Zhan G, Huang W. Research progress of indole-fused derivatives as allosteric modulators: Opportunities for drug development. Biomed Pharmacother 2023; 162:114574. [PMID: 36996677 DOI: 10.1016/j.biopha.2023.114574] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 03/12/2023] [Accepted: 03/21/2023] [Indexed: 03/30/2023] Open
Abstract
Allosteric modulation is a direct and effective method for regulating the function of biological macromolecules, which play vital roles in various cellular activities. Unlike orthosteric modulators, allosteric modulators bind to sites distant from the protein's orthosteric/active site and can have specific effects on the protein's function or activity without competing with endogenous ligands. Compared to traditional orthosteric modulators, allosteric modulators offer several advantages, including reduced side effects, greater specificity, and lower toxicity, making them a promising strategy for developing novel drugs. Indole-fused architectures are widely distributed in natural products and bioactive drug leads, displaying diverse biological activities that attract the interest of both chemists and biologists in drug discovery. Currently, an increasing number of indole-fused compounds have exhibited potent activities in allosteric modulation. In this review, we provide a brief summary of examples of allosteric modulators based on the indole-fused complex architecture, highlighting the strategies for drug design/discovery and the structure-activity relationships of allosteric modulators from the perspective of medicinal chemistry.
Collapse
|
6
|
Gharbi KA, Bonomo YA, Hallinan CM. Evidence from Human Studies for Utilising Cannabinoids for the Treatment of Substance-Use Disorders: A Scoping Review with a Systematic Approach. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:4087. [PMID: 36901098 PMCID: PMC10001982 DOI: 10.3390/ijerph20054087] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 02/02/2023] [Accepted: 02/10/2023] [Indexed: 06/08/2023]
Abstract
Substance-use disorders are pervasive, comorbid with a plethora of disease and possess limited treatment options. Medicinal cannabinoids have been proposed as a novel potential treatment based on preclinical/animal trials. The objective of this study was to examine the efficacy and safety of potential therapeutics targeting the endocannabinoid system in the treatment of substance-use disorders. We performed a scoping review using a systematic approach of systematic reviews, narrative reviews, and randomised control trials that utilised cannabinoids as treatment for substance-use disorders. For this scoping review we used the PRISMA guidelines, a framework for systematic reviews and meta-analyses, to inform our methodology. We conducted a manual search of Medline, Embase, and Scopus databases in July 2022. Of the 253 results returned by the databases, 25 studies including reviews were identified as relevant, from which 29 randomised controlled trials were derived and analysed via a primary study decomposition. This review captured a small volume of highly heterogenous primary literature investing the therapeutic effect of cannabinoids for substance-use disorders. The most promising findings appeared to be for cannabis-use disorder. Cannabidiol appeared to be the cannabinoid showing the most promise for the treatment of multiple-substance-use disorders.
Collapse
Affiliation(s)
- Kayvan Ali Gharbi
- Department of General Practice, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, VIC 3010, Australia
- Department of Medicine, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Yvonne Ann Bonomo
- Department of Medicine, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, VIC 3010, Australia
- St Vincent’s Health—Department of Addiction Medicine, Fitzroy, VIC 3065, Australia
| | - Christine Mary Hallinan
- Department of General Practice, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, VIC 3010, Australia
- Health & Biomedical Research Information Technology Unit (HaBIC R2), Department of General Practice, Melbourne Medical School, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, VIC 3010, Australia
| |
Collapse
|
7
|
Raux PL, Vallée M. Cross-talk between neurosteroid and endocannabinoid systems in cannabis addiction. J Neuroendocrinol 2023; 35:e13191. [PMID: 36043319 DOI: 10.1111/jne.13191] [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: 05/17/2022] [Revised: 07/01/2022] [Accepted: 07/15/2022] [Indexed: 11/30/2022]
Abstract
Steroids and endocannabinoids are part of two modulatory systems and some evidence has shown their interconnections in several functions. Homeostasis is a common steady-state described in the body, which is settled by regulatory systems to counterbalance deregulated or allostatic set points towards an equilibrium. This regulation is of primary significance in the central nervous system for maintaining neuronal plasticity and preventing brain-related disorders. In this context, the recent discovery of the shutdown of the endocannabinoid system (ECS) overload by the neurosteroid pregnenolone has highlighted new endogenous mechanisms of ECS regulation related to cannabis-induced intoxication. These mechanisms involve a regulatory loop mediated by overactivation of the central type-1 cannabinoid receptor (CB1R), which triggers the production of its own regulator, pregnenolone. Therefore, this highlights a new process of regulation of steroidogenesis in the brain. Pregnenolone, long considered an inactive precursor of neurosteroids, can then act as an endogenous negative allosteric modulator of CB1R. The present review aims to shed light on a new framework for the role of ECS in the addictive characteristics of cannabis with the novel endogenous mechanism of ECS involving the neurosteroid pregnenolone. In addition, this new endogenous regulatory loop could provide a relevant therapeutic model in the current context of increasing recreational and medical use of cannabis.
Collapse
Affiliation(s)
- Pierre-Louis Raux
- INSERM U1215, Neurocentre Magendie, Group "Physiopathology and Therapeutic Approaches of Stress-Related Disease", Bordeaux, France
- University of Bordeaux, Bordeaux, France
| | - Monique Vallée
- INSERM U1215, Neurocentre Magendie, Group "Physiopathology and Therapeutic Approaches of Stress-Related Disease", Bordeaux, France
- University of Bordeaux, Bordeaux, France
| |
Collapse
|
8
|
Scheyer A, Yasmin F, Naskar S, Patel S. Endocannabinoids at the synapse and beyond: implications for neuropsychiatric disease pathophysiology and treatment. Neuropsychopharmacology 2023; 48:37-53. [PMID: 36100658 PMCID: PMC9700791 DOI: 10.1038/s41386-022-01438-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 08/10/2022] [Accepted: 08/18/2022] [Indexed: 11/09/2022]
Abstract
Endocannabinoids (eCBs) are lipid neuromodulators that suppress neurotransmitter release, reduce postsynaptic excitability, activate astrocyte signaling, and control cellular respiration. Here, we describe canonical and emerging eCB signaling modes and aim to link adaptations in these signaling systems to pathological states. Adaptations in eCB signaling systems have been identified in a variety of biobehavioral and physiological process relevant to neuropsychiatric disease states including stress-related disorders, epilepsy, developmental disorders, obesity, and substance use disorders. These insights have enhanced our understanding of the pathophysiology of neurological and psychiatric disorders and are contributing to the ongoing development of eCB-targeting therapeutics. We suggest future studies aimed at illuminating how adaptations in canonical as well as emerging cellular and synaptic modes of eCB signaling contribute to disease pathophysiology or resilience could further advance these novel treatment approaches.
Collapse
Affiliation(s)
| | - Farhana Yasmin
- Northwestern Center for Psychiatric Neuroscience, Chicago, IL, USA
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Saptarnab Naskar
- Northwestern Center for Psychiatric Neuroscience, Chicago, IL, USA
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Sachin Patel
- Northwestern Center for Psychiatric Neuroscience, Chicago, IL, USA.
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA.
| |
Collapse
|
9
|
Iyer V, Rangel-Barajas C, Woodward TJ, Kulkarni A, Cantwell L, Crystal JD, Mackie K, Rebec GV, Thakur GA, Hohmann AG. Negative allosteric modulation of CB 1 cannabinoid receptor signaling suppresses opioid-mediated reward. Pharmacol Res 2022; 185:106474. [PMID: 36179954 PMCID: PMC9948526 DOI: 10.1016/j.phrs.2022.106474] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 09/15/2022] [Accepted: 09/25/2022] [Indexed: 01/18/2023]
Abstract
Blockade of cannabinoid type 1 (CB1)-receptor signaling decreases the rewarding properties of many drugs of abuse and has been proposed as an anti-addiction strategy. However, psychiatric side-effects limit the clinical potential of orthosteric CB1 antagonists. Negative allosteric modulators (NAMs) represent a novel and indirect approach to attenuate CB1 signaling by decreasing affinity and/or efficacy of CB1 ligands. We hypothesized that a CB1-NAM would block opioid reward while avoiding the unwanted effects of orthosteric CB1 antagonists. GAT358, a CB1-NAM, failed to elicit cardinal signs of direct CB1 activation or inactivation when administered by itself. GAT358 decreased catalepsy and hypothermia but not antinociception produced by the orthosteric CB1 agonist CP55,940, suggesting that a CB1-NAM blocked cardinal signs of CB1 activation. Next, GAT358 was evaluated using in vivo assays of opioid-induced dopamine release and reward in male rodents. In the nucleus accumbens shell, a key component of the mesocorticolimbic reward pathway, morphine increased electrically-evoked dopamine efflux and this effect was blocked by a dose of GAT358 that lacked intrinsic effects on evoked dopamine efflux. Moreover, GAT358 blocked morphine-induced reward in a conditioned place preference (CPP) assay without producing reward or aversion alone. GAT358-induced blockade of morphine CPP was also occluded by GAT229, a CB1 positive allosteric modulator (CB1-PAM), and absent in CB1-knockout mice. Finally, GAT358 also reduced oral oxycodone (but not water) consumption in a two-bottle choice paradigm. Our results support the therapeutic potential of CB1-NAMs as novel drug candidates aimed at preventing opioid reward and treating opioid abuse while avoiding unwanted side-effects.
Collapse
Affiliation(s)
- Vishakh Iyer
- Program in Neuroscience, Indiana University, Bloomington, IN, USA,Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA
| | | | - Taylor J. Woodward
- Program in Neuroscience, Indiana University, Bloomington, IN, USA,Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA
| | - Abhijit Kulkarni
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, USA
| | - Lucas Cantwell
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, USA
| | - Jonathon D. Crystal
- Program in Neuroscience, Indiana University, Bloomington, IN, USA,Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA
| | - Ken Mackie
- Program in Neuroscience, Indiana University, Bloomington, IN, USA,Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA,Gill Center for Biomolecular Science, Indiana University, Bloomington, IN, USA
| | - George V. Rebec
- Program in Neuroscience, Indiana University, Bloomington, IN, USA,Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA
| | - Ganesh A. Thakur
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, USA
| | - Andrea G. Hohmann
- Program in Neuroscience, Indiana University, Bloomington, IN, USA,Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA,Gill Center for Biomolecular Science, Indiana University, Bloomington, IN, USA,Corresponding Author: Andrea G. Hohmann, Psychological and Brain Sciences, Gill Center for Biomolecular Science, Indiana University, Bloomington, IN 47405-7007,
| |
Collapse
|
10
|
Synthesis and Biological Evaluation of Indole-2-Carboxamides with Potent Apoptotic Antiproliferative Activity as EGFR/CDK2 Dual Inhibitors. Pharmaceuticals (Basel) 2022; 15:ph15081006. [PMID: 36015154 PMCID: PMC9414584 DOI: 10.3390/ph15081006] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 08/10/2022] [Accepted: 08/10/2022] [Indexed: 11/28/2022] Open
Abstract
The apoptotic antiproliferative actions of our previously reported CB1 allosteric modulators 5-chlorobenzofuran-2-carboxamide derivatives VIIa–j prompted us to develop and synthesise a novel series of indole-2-carboxamide derivatives 5a–k, 6a–c, and 7. Different spectroscopic methods of analysis were used to validate the novel compounds. Using the MTT assay method, the novel compounds were examined for antiproliferative activity against four distinct cancer cell lines. Compounds 5a–k, 6a–c, and 7 demonstrated greater antiproliferative activity against the breast cancer cell line (MCF-7) than other tested cancer cell lines, and 5a–k (which contain the phenethyl moiety in their backbone structure) demonstrated greater potency than 6a–c and 7, indicating the importance of the phenethyl moiety for antiproliferative action. Compared to reference doxorubicin (GI50 = 1.10 µM), compounds 5d, 5e, 5h, 5i, 5j, and 5k were the most effective of the synthesised derivatives, with GI50 ranging from 0.95 µM to 1.50 µM. Compounds 5d, 5e, 5h, 5i, 5j, and 5k were tested for their inhibitory impact on EGFR and CDK2, and the results indicated that the compounds tested had strong antiproliferative activity and are effective at suppressing both CDK2 and EGFR. Moreover, the studied compounds induced apoptosis with high potency, as evidenced by their effects on apoptotic markers such as Caspases 3, 8, 9, Cytochrome C, Bax, Bcl2, and p53.
Collapse
|
11
|
Nguyen T, Gamage TF, Finlay DB, Decker AM, Langston TL, Barrus D, Glass M, Li JX, Kenakin TP, Zhang Y. Development of 3-(4-Chlorophenyl)-1-(phenethyl)urea Analogues as Allosteric Modulators of the Cannabinoid Type-1 Receptor: RTICBM-189 is Brain Penetrant and Attenuates Reinstatement of Cocaine-Seeking Behavior. J Med Chem 2021; 65:257-270. [PMID: 34929081 DOI: 10.1021/acs.jmedchem.1c01432] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
We have shown that CB1 receptor negative allosteric modulators (NAMs) attenuated the reinstatement of cocaine-seeking behaviors in rats. In an effort to further define the structure-activity relationships and assess the druglike properties of the 3-(4-chlorophenyl)-1-(phenethyl)urea-based CB1 NAMs that we recently reported, we introduced substituents of different electronic properties and sizes to the phenethyl group and evaluated their potency in CB1 calcium mobilization, cAMP, and GTPγS assays. We found that 3-position substitutions such as Cl, F, and Me afforded enhanced CB1 potency, whereas 4-position analogues were generally less potent. The 3-chloro analogue (31, RTICBM-189) showed no activity at >50 protein targets and excellent brain permeation but relatively low metabolic stability in rat liver microsomes. Pharmacokinetic studies in rats confirmed the excellent brain exposure of 31 with a brain/plasma ratio Kp of 2.0. Importantly, intraperitoneal administration of 31 significantly and selectively attenuated the reinstatement of the cocaine-seeking behavior in rats without affecting locomotion.
Collapse
Affiliation(s)
- Thuy Nguyen
- Research Triangle Institute, Research Triangle Park, Research Triangle Park, North Carolina 27709, United States
| | - Thomas F Gamage
- Research Triangle Institute, Research Triangle Park, Research Triangle Park, North Carolina 27709, United States
| | - David B Finlay
- Department of Pharmacology and Toxicology, School of Biomedical Sciences, University of Otago, Dunedin 9054, New Zealand
| | - Ann M Decker
- Research Triangle Institute, Research Triangle Park, Research Triangle Park, North Carolina 27709, United States
| | - Tiffany L Langston
- Research Triangle Institute, Research Triangle Park, Research Triangle Park, North Carolina 27709, United States
| | - Daniel Barrus
- Research Triangle Institute, Research Triangle Park, Research Triangle Park, North Carolina 27709, United States
| | - Michelle Glass
- Department of Pharmacology and Toxicology, School of Biomedical Sciences, University of Otago, Dunedin 9054, New Zealand
| | - Jun-Xu Li
- Department of Pharmacology and Toxicology, University at Buffalo, the State University of New York, Buffalo, New York 14214, United States
| | - Terry P Kenakin
- Department of Pharmacology, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| | - Yanan Zhang
- Research Triangle Institute, Research Triangle Park, Research Triangle Park, North Carolina 27709, United States
| |
Collapse
|
12
|
Minh Le T, Szakonyi Z. Enantiomeric Isopulegol as the Chiral Pool in the Total Synthesis of Bioactive Agents. CHEM REC 2021; 22:e202100194. [PMID: 34553822 DOI: 10.1002/tcr.202100194] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 09/05/2021] [Indexed: 11/12/2022]
Abstract
Isopulegol, a pool of abundant chiral terpene, has long served as the starting material for the total synthesis of isopulegol-based drugs. As an inexpensive and versatile starting material, this compound continues to serve modern synthetic chemistry. This review highlights the total syntheses of terpenoids in the period from 1980 to 2020 in which with isopulegol applied as a building block.
Collapse
Affiliation(s)
- Tam Minh Le
- Institute of Pharmaceutical Chemistry, University of Szeged, Interdisciplinary Excellent Center, Eötvös utca 6, H-6720, Szeged, Hungary.,Stereochemistry Research Group of the Hungarian Academy Science, Eötvös utca 6, H-6720, Szeged, Hungary
| | - Zsolt Szakonyi
- Institute of Pharmaceutical Chemistry, University of Szeged, Interdisciplinary Excellent Center, Eötvös utca 6, H-6720, Szeged, Hungary.,Interdisciplinary Centre of Natural Products, University of Szeged, Eötvös utca 6, H-6720, Szeged, Hungary
| |
Collapse
|
13
|
Cuesta SA, Meneses L. The Role of Organic Small Molecules in Pain Management. Molecules 2021; 26:4029. [PMID: 34279369 PMCID: PMC8271912 DOI: 10.3390/molecules26134029] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/25/2021] [Accepted: 06/27/2021] [Indexed: 12/28/2022] Open
Abstract
In this review, a timeline starting at the willow bark and ending in the latest discoveries of analgesic and anti-inflammatory drugs will be discussed. Furthermore, the chemical features of the different small organic molecules that have been used in pain management will be studied. Then, the mechanism of different types of pain will be assessed, including neuropathic pain, inflammatory pain, and the relationship found between oxidative stress and pain. This will include obtaining insights into the cyclooxygenase action mechanism of nonsteroidal anti-inflammatory drugs (NSAID) such as ibuprofen and etoricoxib and the structural difference between the two cyclooxygenase isoforms leading to a selective inhibition, the action mechanism of pregabalin and its use in chronic neuropathic pain, new theories and studies on the analgesic action mechanism of paracetamol and how changes in its structure can lead to better characteristics of this drug, and cannabinoid action mechanism in managing pain through a cannabinoid receptor mechanism. Finally, an overview of the different approaches science is taking to develop more efficient molecules for pain treatment will be presented.
Collapse
Affiliation(s)
| | - Lorena Meneses
- Laboratorio de Química Computacional, Facultad de Ciencias Exactas y Naturales, Escuela de Ciencias Químicas, Pontificia Universidad Católica del Ecuador, Av. 12 de Octubre 1076 Apartado, Quito 17-01-2184, Ecuador;
| |
Collapse
|
14
|
Finn DP, Haroutounian S, Hohmann AG, Krane E, Soliman N, Rice ASC. Cannabinoids, the endocannabinoid system, and pain: a review of preclinical studies. Pain 2021; 162:S5-S25. [PMID: 33729211 PMCID: PMC8819673 DOI: 10.1097/j.pain.0000000000002268] [Citation(s) in RCA: 75] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 03/10/2021] [Indexed: 12/13/2022]
Abstract
ABSTRACT This narrative review represents an output from the International Association for the Study of Pain's global task force on the use of cannabis, cannabinoids, and cannabis-based medicines for pain management, informed by our companion systematic review and meta-analysis of preclinical studies in this area. Our aims in this review are (1) to describe the value of studying cannabinoids and endogenous cannabinoid (endocannabinoid) system modulators in preclinical/animal models of pain; (2) to discuss both pain-related efficacy and additional pain-relevant effects (adverse and beneficial) of cannabinoids and endocannabinoid system modulators as they pertain to animal models of pathological or injury-related persistent pain; and (3) to identify important directions for future research. In service of these goals, this review (1) provides an overview of the endocannabinoid system and the pharmacology of cannabinoids and endocannabinoid system modulators, with specific relevance to animal models of pathological or injury-related persistent pain; (2) describes pharmacokinetics of cannabinoids in rodents and humans; and (3) highlights differences and discrepancies between preclinical and clinical studies in this area. Preclinical (rodent) models have advanced our understanding of the underlying sites and mechanisms of action of cannabinoids and the endocannabinoid system in suppressing nociceptive signaling and behaviors. We conclude that substantial evidence from animal models supports the contention that cannabinoids and endocannabinoid system modulators hold considerable promise for analgesic drug development, although the challenge of translating this knowledge into clinically useful medicines is not to be underestimated.
Collapse
Affiliation(s)
- David P Finn
- Pharmacology and Therapeutics, School of Medicine, Galway Neuroscience Centre and Centre for Pain Research, Human Biology Building, National University of Ireland Galway, University Road, Galway, Ireland
| | - Simon Haroutounian
- Department of Anesthesiology and Washington University Pain Center, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Andrea G Hohmann
- Psychological and Brain Sciences, Program in Neuroscience, and Gill Center for Biomolecular Science, Indiana University, Bloomington, IN, USA
| | - Elliot Krane
- Departments of Anesthesiology, Perioperative, and Pain Medicine, & Pediatrics, Stanford University School of Medicine, Stanford, California, USA
| | - Nadia Soliman
- Pain Research, Department of Surgery & Cancer, Faculty of Medicine, Imperial College London, UK
| | - Andrew SC Rice
- Pain Research, Department of Surgery & Cancer, Faculty of Medicine, Imperial College London, UK
| |
Collapse
|
15
|
Nguyen T, Gamage TF, Decker AM, Finlay DB, Langston TL, Barrus D, Glass M, Harris DL, Zhang Y. Rational design of cannabinoid type-1 receptor allosteric modulators: Org27569 and PSNCBAM-1 hybrids. Bioorg Med Chem 2021; 41:116215. [PMID: 34015703 DOI: 10.1016/j.bmc.2021.116215] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 04/22/2021] [Accepted: 05/07/2021] [Indexed: 11/25/2022]
Abstract
Allosteric modulation offers an alternate approach to target the cannabinoid type-1 receptor (CB1) for therapeutic benefits. Examination of the two widely studied prototypic CB1 negative allosteric modulators (NAMs) Org27569 and PSNCBAM-1 revealed structural resemblance and similar structure-activity relationships (SARs). In silico docking and dynamics simulation studies using the crystal structure of CB1 co-bound with CP55,940 and Org27569 suggested that Org27569 and PSNCBAM-1 occupied the same binding pocket and several common interactions were present in both series with the CB1 receptor. A new scaffold was therefore designed by merging the key structural features from the two series and the hybrids retained these binding features in the in silico docking studies. In addition, one such hybrid displayed similar functions to Org27569 in dynamic simulations by preserving a key R2143.50-D3386.30 salt bridge and maintaining an antagonist-like Helix3-Helix6 interhelical distance. Based on these results, a series of hybrids were synthesized and assessed in calcium mobilization, [35S]GTPγS binding and cAMP assays. Several compounds displayed comparable potencies to Org27569 and PSNCBAM-1 in these assays. This work offers new insight of the SAR requirement at the allosteric site of the CB1 receptor and provides a new scaffold that can be optimized for the development of future CB1 allosteric modulators.
Collapse
Affiliation(s)
- Thuy Nguyen
- Research Triangle Institute, Research Triangle Park, NC 27709, USA
| | - Thomas F Gamage
- Research Triangle Institute, Research Triangle Park, NC 27709, USA
| | - Ann M Decker
- Research Triangle Institute, Research Triangle Park, NC 27709, USA
| | - David B Finlay
- Department of Pharmacology and Toxicology, University of Otago, Dunedin 9054, New Zealand
| | | | - Daniel Barrus
- Research Triangle Institute, Research Triangle Park, NC 27709, USA
| | - Michelle Glass
- Department of Pharmacology and Toxicology, University of Otago, Dunedin 9054, New Zealand
| | - Danni L Harris
- Research Triangle Institute, Research Triangle Park, NC 27709, USA.
| | - Yanan Zhang
- Research Triangle Institute, Research Triangle Park, NC 27709, USA.
| |
Collapse
|
16
|
Vitale RM, Iannotti FA, Amodeo P. The (Poly)Pharmacology of Cannabidiol in Neurological and Neuropsychiatric Disorders: Molecular Mechanisms and Targets. Int J Mol Sci 2021; 22:4876. [PMID: 34062987 PMCID: PMC8124847 DOI: 10.3390/ijms22094876] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 04/29/2021] [Accepted: 05/03/2021] [Indexed: 12/14/2022] Open
Abstract
Cannabidiol (CBD), the major nonpsychoactive Cannabis constituent, has been proposed for the treatment of a wide panel of neurological and neuropsychiatric disorders, including anxiety, schizophrenia, epilepsy and drug addiction due to the ability of its versatile scaffold to interact with diverse molecular targets that are not restricted to the endocannabinoid system. Albeit the molecular mechanisms responsible for the therapeutic effects of CBD have yet to be fully elucidated, many efforts have been devoted in the last decades to shed light on its complex pharmacological profile. In particular, an ever-increasing number of molecular targets linked to those disorders have been identified for this phytocannabinoid, along with the modulatory effects of CBD on their cascade signaling. In this view, here we will try to provide a comprehensive and up-to-date overview of the molecular basis underlying the therapeutic effects of CBD involved in the treatment of neurological and neuropsychiatric disorders.
Collapse
Affiliation(s)
- Rosa Maria Vitale
- Institute of Biomolecular Chemistry, National Research Council (ICB-CNR), Via Campi Flegrei 34, 80078 Pozzuoli, NA, Italy; (F.A.I.); (P.A.)
| | - Fabio Arturo Iannotti
- Institute of Biomolecular Chemistry, National Research Council (ICB-CNR), Via Campi Flegrei 34, 80078 Pozzuoli, NA, Italy; (F.A.I.); (P.A.)
- Endocannabinoid Research Group (ERG), Institute of Biomolecular Chemistry, National Research Council (ICB-CNR), Via Campi Flegrei 34, 80078 Pozzuoli, NA, Italy
| | - Pietro Amodeo
- Institute of Biomolecular Chemistry, National Research Council (ICB-CNR), Via Campi Flegrei 34, 80078 Pozzuoli, NA, Italy; (F.A.I.); (P.A.)
| |
Collapse
|
17
|
Shapiro L, Gado F, Manera C, Escayg A. Allosteric modulation of the cannabinoid 2 receptor confers seizure resistance in mice. Neuropharmacology 2021; 188:108448. [PMID: 33450277 DOI: 10.1016/j.neuropharm.2021.108448] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 12/18/2020] [Accepted: 12/30/2020] [Indexed: 02/07/2023]
Abstract
Mounting evidence suggests that modulation of cannabinoid 2 receptors (CB2Rs) is therapeutic in mouse models of neurological disorders, including neuropathic pain, neurodegenerative disease, and stroke. We previously showed that reducing CB2R activity increases seizure susceptibility in mice. In the present study, we evaluated the therapeutic potential of the CB2R positive allosteric modulator, Ec21a, against induced seizures in mice. The pharmacokinetic profile of Ec21 demonstrated a similar distribution in brain and plasma, with detection up to 12 h following injection. Ec21a increased resistance to induced seizures in CF1 wild-type mice and mice harboring the SCN1A R1648H human epilepsy mutation. A rotarod test provided evidence that Ec21a does not cause neurotoxicity-induced motor deficits at its therapeutic dose, and seizure protection was maintained with repeated drug administration. The selectivity of Ec21a for CB2R was supported by the ability of the CB2R antagonist AM630, but not the CB1R antagonist AM251, to block Ec21a-conferred seizure protection in mice, and a lack of significant binding of Ec21a to 34 brain-expressed receptors and transporters in vitro. These results identify allosteric modulation of CB2Rs as a promising therapeutic approach for the treatment of epilepsy.
Collapse
Affiliation(s)
- Lindsey Shapiro
- Emory University, Department of Human Genetics, Atlanta, 30322, Georgia
| | - Francesca Gado
- University of Pisa, Department of Pharmacy, Pisa, 56126, Italy
| | | | - Andrew Escayg
- Emory University, Department of Human Genetics, Atlanta, 30322, Georgia.
| |
Collapse
|
18
|
Mielnik CA, Lam VM, Ross RA. CB 1 allosteric modulators and their therapeutic potential in CNS disorders. Prog Neuropsychopharmacol Biol Psychiatry 2021; 106:110163. [PMID: 33152384 DOI: 10.1016/j.pnpbp.2020.110163] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 09/30/2020] [Accepted: 10/29/2020] [Indexed: 01/05/2023]
Abstract
CB1 is the most abundant GPCR found in the mammalian brain. It has garnered considerable attention as a potential therapeutic drug target. CB1 is involved in a wide range of physiological and psychiatric processes and has the potential to be targeted in a wide range of disease states. However, most of the selective and non-selective synthetic CB1 agonists and antagonists/inverse agonists developed to date are primarily used as research tools. No novel synthetic cannabinoids are currently in the clinic for use in psychiatric illness; synthetic analogues of the phytocannabinoid THC are on the market to treat nausea and vomiting caused by cancer chemotherapy, along with off-label use for pain. Novel strategies are being explored to target CB1, but with emphasis on the elimination or mitigation of the potential psychiatric adverse effects that are observed by central agonism/antagonism of CB1. New pharmacological options are being pursued that may avoid these adverse effects while preserving the potential therapeutic benefits of CB1 modulation. Allosteric modulation of CB1 is one such approach. In this review, we will summarize and critically analyze both the in vitro characterization and in vivo validation of CB1 allosteric modulators developed to date, with a focus on CNS therapeutic effects.
Collapse
Affiliation(s)
- Catharine A Mielnik
- Department of Pharmacology & Toxicology, University of Toronto, ON M5S 1A8, Canada
| | - Vincent M Lam
- Department of Pharmacology & Toxicology, University of Toronto, ON M5S 1A8, Canada
| | - Ruth A Ross
- Department of Pharmacology & Toxicology, University of Toronto, ON M5S 1A8, Canada.
| |
Collapse
|
19
|
Aderibigbe AO, Pandey P, Doerksen RJ. Negative allosteric modulators of cannabinoid receptor 1: Ternary complexes including CB1, orthosteric CP55940 and allosteric ORG27569. J Biomol Struct Dyn 2021; 40:5729-5747. [PMID: 33480332 DOI: 10.1080/07391102.2021.1873187] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
In October 2019, the first X-ray crystal structure of a ternary cannabinoid receptor 1 (CB1) complex (PDB ID: 6KQI) was published, including the well-known orthosteric agonist, CP55940, and the well-studied negative allosteric modulator, ORG27569. Prior to the release of 6KQI, we applied binding pocket analysis and molecular docking to carefully prepared computational models of the ternary CB1 complex, in order to predict the binding site for ORG27569 with the CP55940-bound CB1 receptor. We carefully studied the binding pose of agonist ligands in the CB1 orthosteric pocket, including CP55940. Our computational studies identified the most favorable binding site for ORG27569, in the CP55940-CB1 complex, to be at the intracellular end of the receptor. However, in the 6KQI structure, ORG27569 was found at an extrahelical, intramembrane site on the complex, a site that partially overlaps with the site predicted in our calculations to be second-best. We performed molecular dynamics simulations of the CP55940-bound CB1 complex with ORG27569 at different binding sites. Our analysis of the simulations indicated that ORG27569 bound favorably and stably in each simulation, but, as in the earlier calculations, bound best at the intracellular site, which is different than that found in the crystal structure. These results suggest that the intracellular site might serve as an alternative binding site in CB1. Our studies show that the computational techniques we used are valuable in identifying ligand-binding pockets in proteins, and could be useful for the study of the interaction mode of other allosteric modulators.
Collapse
Affiliation(s)
- AyoOluwa O Aderibigbe
- Department of BioMolecular Sciences, Division of Medicinal Chemistry, School of Pharmacy, University of Mississippi, University, MS, USA
| | - Pankaj Pandey
- Department of BioMolecular Sciences, Division of Medicinal Chemistry, School of Pharmacy, University of Mississippi, University, MS, USA.,National Center for Natural Products Research, University of Mississippi, University, Mississippi, USA
| | - Robert J Doerksen
- Department of BioMolecular Sciences, Division of Medicinal Chemistry, School of Pharmacy, University of Mississippi, University, MS, USA.,Research Institute of Pharmaceutical Sciences, School of Pharmacy, University of Mississippi, University, MS, USA
| |
Collapse
|
20
|
Hamilton AJ, Payne AD, Mocerino M, Gunosewoyo H. Imaging Cannabinoid Receptors: A Brief Collection of Covalent and Fluorescent Probes for CB1 and CB2 Receptors. Aust J Chem 2021. [DOI: 10.1071/ch21007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
There has been an expanding public interest towards the notion that modulation of the sophisticated endocannabinoid system can lead to various therapeutic benefits that are yet to be fully explored. In recent years, the drug discovery paradigm in this field has been largely based on the development of selective CB2 receptor agonists, avoiding the unwanted CB1 receptor-mediated psychoactive side effects. Mechanistically, target engagement studies are crucial for confirming the ligand–receptor interaction and the subsequent biological cascades that lead to the observed therapeutic effects. Concurrently, imaging techniques for visualisation of cannabinoid receptors are increasingly reported in the literature. Small molecule imaging tools ranging from phytocannabinoids such as tetrahydrocannabinol (THC) and cannabidiol (CBD) to the endocannabinoids as well as the purely synthetic cannabimimetics, have been explored to date with varying degrees of success. This Review will cover currently known photoactivatable, electrophilic, and fluorescent ligands for both the CB1 and CB2 receptors. Structural insights from techniques such as ligand-assisted protein structure (LAPS) and the discovery of novel allosteric modulators are significant additions for better understanding of the endocannabinoid system. There has also been a plethora of fluorescent conjugates that have been assessed for their binding to cannabinoid receptors as well as their potential for cellular imaging. More recently, bifunctional probes containing either fluorophores or electrophilic tags are becoming more prevalent in the literature. Collectively, these molecular tools are invaluable in demonstrating target engagement within the human endocannabinoid system.
Collapse
|
21
|
A novel allosteric modulator of the cannabinoid CB 1 receptor ameliorates hyperdopaminergia endophenotypes in rodent models. Neuropsychopharmacology 2021; 46:413-422. [PMID: 33036015 PMCID: PMC7852560 DOI: 10.1038/s41386-020-00876-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 08/24/2020] [Accepted: 09/11/2020] [Indexed: 02/06/2023]
Abstract
The endocannabinoid system (eCBs) encompasses the endocannabinoids, their synthetic and degradative enzymes, and cannabinoid (CB) receptors. The eCBs mediates inhibition of neurotransmitter release and acts as a major homeostatic system. Many aspects of the eCBs are altered in a number of psychiatric disorders including schizophrenia, which is characterized by dysregulation of dopaminergic signaling. The GluN1-Knockdown (GluN1KD) and Dopamine Transporter Knockout (DATKO) mice are models of hyperdopaminergia, which display abnormal psychosis-related behaviors, including hyperlocomotion and changes in pre-pulse inhibition (PPI). Here, we investigate the ability of a novel CB1 receptor (CB1R) allosteric modulator, ABM300, to ameliorate these dysregulated behaviors. ABM300 was characterized in vitro (receptor binding, β-arrestin2 recruitment, ERK1/2 phosphorylation, cAMP inhibition) and in vivo (anxiety-like behaviors, cannabimimetic effects, novel environment exploratory behavior, pre-pulse inhibition, conditioned avoidance response) to assess the effects of the compound in dysregulated behaviors within the transgenic models. In vitro, ABM300 increased CB1R agonist binding but acted as an inhibitor of CB1R agonist induced signaling, including β-arrestin2 translocation, ERK phosphorylation and cAMP inhibition. In vivo, ABM300 did not elicit anxiogenic-like or cannabimimetic effects, but it decreased novelty-induced hyperactivity, exaggerated stereotypy, and vertical exploration in both transgenic models of hyperdopaminergia, as well as normalizing PPI in DATKO mice. The data demonstrate for the first time that a CB1R allosteric modulator ameliorates the behavioral deficits in two models of increased dopamine, warranting further investigation as a potential therapeutic target in psychiatry.
Collapse
|
22
|
Hou L, Rong J, Haider A, Ogasawara D, Varlow C, Schafroth MA, Mu L, Gan J, Xu H, Fowler CJ, Zhang MR, Vasdev N, Ametamey S, Cravatt BF, Wang L, Liang SH. Positron Emission Tomography Imaging of the Endocannabinoid System: Opportunities and Challenges in Radiotracer Development. J Med Chem 2020; 64:123-149. [PMID: 33379862 DOI: 10.1021/acs.jmedchem.0c01459] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The endocannabinoid system (ECS) is involved in a wide range of biological functions and comprises cannabinoid receptors and enzymes responsible for endocannabinoid synthesis and degradation. Over the past 2 decades, significant advances toward developing drugs and positron emission tomography (PET) tracers targeting different components of the ECS have been made. Herein, we summarized the recent development of PET tracers for imaging cannabinoid receptors 1 (CB1R) and 2 (CB2R) as well as the key enzymes monoacylglycerol lipase (MAGL) and fatty acid amide hydrolase (FAAH), particularly focusing on PET neuroimaging applications. State-of-the-art PET tracers for the ECS will be reviewed including their chemical design, pharmacological properties, radiolabeling, as well as preclinical and human PET imaging. In addition, this review addresses the current challenges for ECS PET biomarker development and highlights the important role of PET ligands to study disease pathophysiology as well as to facilitate drug discovery.
Collapse
Affiliation(s)
- Lu Hou
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, The First Affiliated Hospital of Jinan University, 613 West Huangpu Road, Tianhe District, Guangzhou 510630, China
| | - Jian Rong
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital, and Department of Radiology, Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Ahmed Haider
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital, and Department of Radiology, Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Daisuke Ogasawara
- The Skaggs Institute for Chemical Biology and Department of Chemical Physiology, The Scripps Research Institute, SR107, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Cassis Varlow
- Azrieli Centre for Neuro-Radiochemistry, Brain Health Imaging Centre, Centre for Addiction and Mental Health, and Department of Psychiatry/Institute of Medical Science, University of Toronto, 250 College Street, Toronto, M5T 1R8 ON, Canada
| | - Michael A Schafroth
- The Skaggs Institute for Chemical Biology and Department of Chemical Physiology, The Scripps Research Institute, SR107, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Linjing Mu
- Center for Radiopharmaceutical Sciences of ETH, PSI, and USZ, and Institute of Pharmaceutical Sciences, ETH Zurich, Vladimir-Prelog-Weg 4, CH-8093 Zurich, Switzerland
| | - Jiefeng Gan
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, The First Affiliated Hospital of Jinan University, 613 West Huangpu Road, Tianhe District, Guangzhou 510630, China
| | - Hao Xu
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, The First Affiliated Hospital of Jinan University, 613 West Huangpu Road, Tianhe District, Guangzhou 510630, China
| | - Christopher J Fowler
- Department of Pharmacology and Clinical Neuroscience, Umeå University, SE-901 87 Umeå, Sweden
| | - Ming-Rong Zhang
- Department of Radiopharmaceuticals Development, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| | - Neil Vasdev
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital, and Department of Radiology, Harvard Medical School, Boston, Massachusetts 02114, United States.,Azrieli Centre for Neuro-Radiochemistry, Brain Health Imaging Centre, Centre for Addiction and Mental Health, and Department of Psychiatry/Institute of Medical Science, University of Toronto, 250 College Street, Toronto, M5T 1R8 ON, Canada
| | - Simon Ametamey
- Center for Radiopharmaceutical Sciences of ETH, PSI, and USZ, and Institute of Pharmaceutical Sciences, ETH Zurich, Vladimir-Prelog-Weg 4, CH-8093 Zurich, Switzerland
| | - Benjamin F Cravatt
- The Skaggs Institute for Chemical Biology and Department of Chemical Physiology, The Scripps Research Institute, SR107, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Lu Wang
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, The First Affiliated Hospital of Jinan University, 613 West Huangpu Road, Tianhe District, Guangzhou 510630, China.,Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital, and Department of Radiology, Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Steven H Liang
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital, and Department of Radiology, Harvard Medical School, Boston, Massachusetts 02114, United States
| |
Collapse
|
23
|
Aalkjær C, Nilsson H, De Mey JGR. Sympathetic and Sensory-Motor Nerves in Peripheral Small Arteries. Physiol Rev 2020; 101:495-544. [PMID: 33270533 DOI: 10.1152/physrev.00007.2020] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Small arteries, which play important roles in controlling blood flow, blood pressure, and capillary pressure, are under nervous influence. Their innervation is predominantly sympathetic and sensory motor in nature, and while some arteries are densely innervated, others are only sparsely so. Innervation of small arteries is a key mechanism in regulating vascular resistance. In the second half of the previous century, the physiology and pharmacology of this innervation were very actively investigated. In the past 10-20 yr, the activity in this field was more limited. With this review we highlight what has been learned during recent years with respect to development of small arteries and their innervation, some aspects of excitation-release coupling, interaction between sympathetic and sensory-motor nerves, cross talk between endothelium and vascular nerves, and some aspects of their role in vascular inflammation and hypertension. We also highlight what remains to be investigated to further increase our understanding of this fundamental aspect of vascular physiology.
Collapse
Affiliation(s)
| | - Holger Nilsson
- Department Physiology, Gothenburg University, Gothenburg, Sweden
| | - Jo G R De Mey
- Deptartment Pharmacology and Personalized Medicine, Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
24
|
Positive allosteric modulation of the cannabinoid type-1 receptor (CB1R) in periaqueductal gray (PAG) antagonizes anti-nociceptive and cellular effects of a mu-opioid receptor agonist in morphine-withdrawn rats. Psychopharmacology (Berl) 2020; 237:3729-3739. [PMID: 32857187 PMCID: PMC7687722 DOI: 10.1007/s00213-020-05650-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 08/20/2020] [Indexed: 12/24/2022]
Abstract
Opioid drugs are a first-line treatment for severe acute pain and other chronic pain conditions, but long-term opioid drug use produces opioid-induced hyperalgesia (OIH). Co-administration of cannabinoids with opioid receptor agonists produce anti-nociceptive synergy, but cannabinoid receptor agonists may also produce undesirable side effects. Therefore, positive allosteric modulators (PAM) of cannabinoid type-1 receptors (CB1R) may provide an option reducing pain and/or enhancing the anti-hyperalgesic effects of opioids without the side effects, tolerance, and dependence observed with the use of ligands that target the orthosteric binding sites. This study tested GAT211, a PAM of cannabinoid type-1 receptors (CB1R), for its ability to enhance the anti-hyperalgesic effects of the mu-opioid receptor (MOR) agonist DAMGO in rats treated chronically with morphine (or saline) and tested during withdrawal. We tested the effects of intra-periaqueductal gray (PAG) injections of (1) DAMGO, (2) GAT211, or (3) DAMGO + GAT211 on thermal nociception in chronic morphine-treated rats that were hyperalgesic and also in saline-treated control rats. We used slice electrophysiology to test the effects of DAMGO/GAT211 bath application on synaptic transmission in the vlPAG. Intra-PAG DAMGO infusions dose-dependently reversed chronic morphine-induced hyperalgesia, but intra-PAG GAT211 did not alter nociception at the doses we tested. When co-administered into the PAG, GAT211 antagonized the anti-nociceptive effects of DAMGO in morphine-withdrawn rats. DAMGO suppressed synaptic inhibition in the vlPAG of brain slices taken from saline- and morphine-treated rats, and GAT211 attenuated DAMGO-induced suppression of synaptic inhibition in vlPAG neurons via actions at CB1R. These findings show that positive allosteric modulation of CB1R antagonizes the behavioral and cellular effects of a MOR agonist in the PAG of rats.
Collapse
|
25
|
Bian Y, Jun JJ, Cuyler J, Xie XQ. Covalent allosteric modulation: An emerging strategy for GPCRs drug discovery. Eur J Med Chem 2020; 206:112690. [PMID: 32818870 PMCID: PMC9948676 DOI: 10.1016/j.ejmech.2020.112690] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 07/10/2020] [Accepted: 07/24/2020] [Indexed: 12/13/2022]
Abstract
Designing covalent allosteric modulators brings new opportunities to the field of drug discovery towards G-protein-coupled receptors (GPCRs). Targeting an allosteric binding pocket can allow a modulator to have protein subtype selectivity and low drug resistance. Utilizing covalent warheads further enables the modulator to increase the binding potency and extend the duration of action. This review starts with GPCR allosteric modulation to discuss the structural biology of allosteric binding pockets, the different types of allosteric modulators, as well as the advantages of employing allosteric modulation. This is followed by a discussion on covalent modulators to clarify how covalent ligands can benefit the receptor modulation and to illustrate moieties that can commonly be used as covalent warheads. Finally, case studies are presented on designing class A, B, and C GPCR covalent allosteric modulators to demonstrate successful stories on combining allosteric modulation and covalent binding. Limitations and future perspectives are also covered.
Collapse
Affiliation(s)
- Yuemin Bian
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy,NIH National Center of Excellence for Computational Drug Abuse Research
| | - Jaden Jungho Jun
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy,NIH National Center of Excellence for Computational Drug Abuse Research
| | - Jacob Cuyler
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy,NIH National Center of Excellence for Computational Drug Abuse Research
| | - Xiang-Qun Xie
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy, Pittsburgh, PA, 15261, United States; NIH National Center of Excellence for Computational Drug Abuse Research, Pittsburgh, PA, 15261, United States; Drug Discovery Institute, Pittsburgh, PA, 15261, United States; Departments of Computational Biology and Structural Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15261, United States.
| |
Collapse
|
26
|
The role of cannabinoid 1 receptor in the nucleus accumbens on tramadol induced conditioning and reinstatement. Life Sci 2020; 260:118430. [PMID: 32931800 DOI: 10.1016/j.lfs.2020.118430] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 08/27/2020] [Accepted: 09/08/2020] [Indexed: 12/21/2022]
Abstract
AIMS Previous investigations demonstrated that tramadol, as a painkiller, similar to morphine induces tolerance and dependence. Furthermore, the cannabinoid receptor 1 (CB1R) located in the nucleus accumbens (NAc) plays a critical role in morphine-induced conditioning. Therefore, the main objective of this study was to evaluate the role of NAc CB1R in tramadol induced conditioning and reinstatement. MAIN METHODS In the present experiment, the effect of NAc CB1 receptors on tramadol induced conditioning was tested by microinjecting of arachidonylcyclopropylamide (ACPA, CB1R agonist) and AM 251 (CB1R inverse agonist) in the NAc during tramadol-induced conditioning in the adult male Wistar rats. In addition, the role of NAc CB1R in the reinstatement was also evaluated by injecting ACPA and AM 251 after a 10-days extinction period. KEY FINDINGS The obtained data revealed that the administration of tramadol (1,2, and 4 mg/kg, ip) dose-dependently produced conditioned place preference (CPP). Moreover, intra-NAc administration of ACPA (0.25, 0.5, and 1 μg/rat) dose-dependently induced conditioning, while the administration of AM-251 (30, 60, and 120 ng/rat) induced a significant aversion. In addition, the administration of a non-effective dose of AM251 during tramadol conditioning inhibited conditioning induced by tramadol. On the other hand, the administration of ACPA after extinction induced a significant reinstatement. Notably, the locomotor activity did not change among groups. SIGNIFICANCE Previous studies have shown that tramadol-induced CPP occurs through μ-opioid receptors. The data obtained in the current study indicated that CB1R located in the NAc is involved in mediating conditioning induced by tramadol. Besides, CB1R also plays a vital role in the reinstatement of tramadol-conditioned animals. It might be due to the effect of opioids on enhancing the level of CB1R.
Collapse
|
27
|
Rodríguez-Rodríguez I, Kalafut J, Czerwonka A, Rivero-Müller A. A novel bioassay for quantification of surface Cannabinoid receptor 1 expression. Sci Rep 2020; 10:18191. [PMID: 33097803 PMCID: PMC7584592 DOI: 10.1038/s41598-020-75331-y] [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: 05/02/2020] [Accepted: 10/14/2020] [Indexed: 12/04/2022] Open
Abstract
The cannabinoid receptor type 1 (CB1) plays critical roles in multiple physiological processes such as pain perception, brain development and body temperature regulation. Mutations on this gene (CNR1), results in altered functionality and/or biosynthesis such as reduced membrane expression, changes in mRNA stability or changes in downstream signaling that act as triggers for diseases such as obesity, Parkinson’s, Huntington’s, among others; thus, it is considered as a potential pharmacological target. To date, multiple quantification methods have been employed to determine how these mutations affect receptor expression and localization; however, they present serious disadvantages that may arise quantifying errors. Here, we describe a sensitive bioassay to quantify receptor surface expression; in this bioassay the Gaussia Luciferase (GLuc) was fused to the extracellular portion of the CB1. The GLuc activity was assessed by coelenterazine addition to the medium followed by immediate readout. Based on GLuc activity assay, we show that the GLuc signals corelate with CB1 localization, besides, we showed the assay’s functionality and reliability by comparing its results with those generated by previously reported mutations on the CNR1 gene and by using flow cytometry to determine the cell surface receptor expression. Detection of membrane-bound CB1, and potentially other GPCRs, is able to quickly screen for receptor levels and help to understand the effect of clinically relevant mutations or polymorphisms.
Collapse
Affiliation(s)
| | - Joanna Kalafut
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, Lublin, Poland
| | - Arkadiusz Czerwonka
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, Lublin, Poland.,Department of Virology and Immunology, Faculty of Biology and Biotechnology, Maria Curie-Skłodowska University, Lublin, Poland
| | - Adolfo Rivero-Müller
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, Lublin, Poland.
| |
Collapse
|
28
|
Abstract
The endocannabinoid system (ECS) is a highly versatile signaling system within the nervous system. Despite its widespread localization, its functions within the context of distinct neural processes are very well discernable and specific. This is remarkable, and the question remains as to how such specificity is achieved. One key player in the ECS is the cannabinoid type 1 receptor (CB1), a G protein-coupled receptor characterized by the complexity of its cell-specific expression, cellular and subcellular localization, and its adaptable regulation of intracellular signaling cascades. CB1 receptors are involved in different synaptic and cellular plasticity processes and in the brain's bioenergetics in a context-specific manner. CB2 receptors are also important in several processes in neurons, glial cells, and immune cells of the brain. As polymorphisms in ECS components, as well as external impacts such as stress and metabolic challenges, can both lead to dysregulated ECS activity and subsequently to possible neuropsychiatric disorders, pharmacological intervention targeting the ECS is a promising therapeutic approach. Understanding the neurobiology of cannabinoid receptor signaling in depth will aid optimal design of therapeutic interventions, minimizing unwanted side effects.
.
Collapse
Affiliation(s)
- Beat Lutz
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| |
Collapse
|
29
|
Morales P, Jagerovic N. Novel approaches and current challenges with targeting the endocannabinoid system. Expert Opin Drug Discov 2020; 15:917-930. [PMID: 32336154 PMCID: PMC7502221 DOI: 10.1080/17460441.2020.1752178] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 04/02/2020] [Indexed: 12/12/2022]
Abstract
INTRODUCTION The pathophysiological relevance of the endocannabinoid system has been widely demonstrated in a variety of diseases including cancer, neurological disorders, and metabolic issues. Therefore, targeting the receptors and the endogenous machinery involved in this system can provide a successful therapeutic outcome. Ligands targeting the canonical cannabinoid receptors, CB1 and CB2, along with inhibitors of the endocannabinoid enzymes have been thoroughly studied in diverse disease models. In fact, phytocannabinoids such as cannabidiol or Δ9-tetrahydrocannabinol are currently on the market for the management of neuropathic pain due to spasticity in multiple sclerosis or seizures in children epilepsy amongst others. AREAS COVERED Challenges in the pharmacology of cannabinoids arise from its pharmacokinetics, off-target effects, and psychoactive effects. In this context, the current review outlines the novel molecular approaches emerging in the field discussing their clinical potential. EXPERT OPINION Even if orthosteric CB1 and CB2 ligands are on the forefront in cannabinoid clinical research, emerging strategies such as allosteric or biased modulation of these receptors along with controlled off-targets effects may increase the therapeutic potential of cannabinoids.
Collapse
Affiliation(s)
- Paula Morales
- Instituto de Química Médica, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Nadine Jagerovic
- Instituto de Química Médica, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| |
Collapse
|
30
|
Butler K, Le Foll B. Novel therapeutic and drug development strategies for tobacco use disorder: endocannabinoid modulation. Expert Opin Drug Discov 2020; 15:1065-1080. [DOI: 10.1080/17460441.2020.1767581] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Kevin Butler
- Translational Addiction Research Laboratory, Centre for Addiction and Mental Health, University of Toronto, Toronto, ON, Canada
| | - Bernard Le Foll
- Translational Addiction Research Laboratory, Centre for Addiction and Mental Health, University of Toronto, Toronto, ON, Canada
- Acute Care Program, Centre for Addiction and Mental Health, Centre for Addiction and Mental Health, Toronto, ON, Canada
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada
- Department of Family and Community Medicine, University of Toronto, Toronto, ON, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
31
|
Stasiulewicz A, Znajdek K, Grudzień M, Pawiński T, Sulkowska JI. A Guide to Targeting the Endocannabinoid System in Drug Design. Int J Mol Sci 2020; 21:ijms21082778. [PMID: 32316328 PMCID: PMC7216112 DOI: 10.3390/ijms21082778] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 04/07/2020] [Accepted: 04/14/2020] [Indexed: 12/11/2022] Open
Abstract
The endocannabinoid system (ECS) is one of the most crucial systems in the human organism, exhibiting multi-purpose regulatory character. It is engaged in a vast array of physiological processes, including nociception, mood regulation, cognitive functions, neurogenesis and neuroprotection, appetite, lipid metabolism, as well as cell growth and proliferation. Thus, ECS proteins, including cannabinoid receptors and their endogenous ligands’ synthesizing and degrading enzymes, are promising therapeutic targets. Their modulation has been employed in or extensively studied as a treatment of multiple diseases. However, due to a complex nature of ECS and its crosstalk with other biological systems, the development of novel drugs turned out to be a challenging task. In this review, we summarize potential therapeutic applications for ECS-targeting drugs, especially focusing on promising synthetic compounds and preclinical studies. We put emphasis on modulation of specific proteins of ECS in different pathophysiological areas. In addition, we stress possible difficulties and risks and highlight proposed solutions. By presenting this review, we point out information pivotal in the spotlight of ECS-targeting drug design, as well as provide an overview of the current state of knowledge on ECS-related pharmacodynamics and show possible directions for needed research.
Collapse
Affiliation(s)
- Adam Stasiulewicz
- Department of Drug Chemistry, Faculty of Pharmacy, Medical University of Warsaw, Banacha 1, 02-097 Warsaw, Poland; (M.G.); (T.P.)
- Interdisciplinary Laboratory of Biological Systems Modelling, Centre of New Technologies, University of Warsaw, Banacha 2c, 02-097 Warsaw, Poland;
- Correspondence: (A.S.); (J.I.S.)
| | - Katarzyna Znajdek
- Interdisciplinary Laboratory of Biological Systems Modelling, Centre of New Technologies, University of Warsaw, Banacha 2c, 02-097 Warsaw, Poland;
- Faculty of Pharmacy, Medical University of Warsaw, Banacha 1, 02-097 Warsaw, Poland
| | - Monika Grudzień
- Department of Drug Chemistry, Faculty of Pharmacy, Medical University of Warsaw, Banacha 1, 02-097 Warsaw, Poland; (M.G.); (T.P.)
| | - Tomasz Pawiński
- Department of Drug Chemistry, Faculty of Pharmacy, Medical University of Warsaw, Banacha 1, 02-097 Warsaw, Poland; (M.G.); (T.P.)
| | - Joanna I. Sulkowska
- Interdisciplinary Laboratory of Biological Systems Modelling, Centre of New Technologies, University of Warsaw, Banacha 2c, 02-097 Warsaw, Poland;
- Faculty of Chemistry, University of Warsaw, Pasteura 1, 02-093 Warsaw, Poland
- Materials and Process Simulation Center, California Institute of Technology, Pasadena, CA 91125, USA
- Correspondence: (A.S.); (J.I.S.)
| |
Collapse
|
32
|
Thapa D, Cairns EA, Szczesniak AM, Kulkarni PM, Straiker AJ, Thakur GA, Kelly MEM. Allosteric Cannabinoid Receptor 1 (CB1) Ligands Reduce Ocular Pain and Inflammation. Molecules 2020; 25:E417. [PMID: 31968549 PMCID: PMC7024337 DOI: 10.3390/molecules25020417] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 01/16/2020] [Indexed: 01/08/2023] Open
Abstract
Cannabinoid receptor 1 (CB1) activation has been reported to reduce transient receptor potential cation channel subfamily V member 1 (TRPV1)-induced inflammatory responses and is anti-nociceptive and anti-inflammatory in corneal injury. We examined whether allosteric ligands, can modulate CB1 signaling to reduce pain and inflammation in corneal hyperalgesia. Corneal hyperalgesia was generated by chemical cauterization of cornea in wildtype and CB2 knockout (CB2-/-) mice. The novel racemic CB1 allosteric ligand GAT211 and its enantiomers GAT228 and GAT229 were examined alone or in combination with the orthosteric CB1 agonist Δ8-tetrahydrocannabinol (Δ8-THC). Pain responses were assessed following capsaicin (1 µM) stimulation of injured corneas at 6 h post-cauterization. Corneal neutrophil infiltration was also analyzed. GAT228, but not GAT229 or GAT211, reduced pain scores in response to capsaicin stimulation. Combination treatments of 0.5% GAT229 or 1% GAT211 with subthreshold Δ8-THC (0.4%) significantly reduced pain scores following capsaicin stimulation. The anti-nociceptive effects of both GAT229 and GAT228 were blocked with CB1 antagonist AM251, but remained unaffected in CB2-/- mice. Two percent GAT228, or the combination of 0.2% Δ8-THC with 0.5% GAT229 also significantly reduced corneal inflammation. CB1 allosteric ligands could offer a novel approach for treating corneal pain and inflammation.
Collapse
Affiliation(s)
- Dinesh Thapa
- Department of Pharmacology, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Elizabeth A. Cairns
- Department of Pharmacology, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | | | - Pushkar M. Kulkarni
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA 02115, USA
| | - Alex J. Straiker
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN 47405, USA
| | - Ganesh A. Thakur
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA 02115, USA
| | - Melanie E. M. Kelly
- Department of Pharmacology, Dalhousie University, Halifax, NS B3H 4R2, Canada
- Department of Anesthesia, Pain Management & Perioperative Medicine, Dalhousie University, Halifax, NS B3H 4R2, Canada
| |
Collapse
|
33
|
Schurman LD, Lu D, Kendall DA, Howlett AC, Lichtman AH. Molecular Mechanism and Cannabinoid Pharmacology. Handb Exp Pharmacol 2020; 258:323-353. [PMID: 32236882 PMCID: PMC8637936 DOI: 10.1007/164_2019_298] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Since antiquity, Cannabis has provoked enormous intrigue for its potential medicinal properties as well as for its unique pharmacological effects. The elucidation of its major cannabinoid constituents, Δ9-tetrahydrocannabinol (THC) and cannabidiol (CBD), led to the synthesis of new cannabinoids (termed synthetic cannabinoids) to understand the mechanisms underlying the pharmacology of Cannabis. These pharmacological tools were instrumental in the ultimate discovery of the endogenous cannabinoid system, which consists of CB1 and CB2 cannabinoid receptors and endogenously produced ligands (endocannabinoids), which bind and activate both cannabinoid receptors. CB1 receptors mediate the cannabimimetic effects of THC and are highly expressed on presynaptic neurons in the nervous system, where they modulate neurotransmitter release. In contrast, CB2 receptors are primarily expressed on immune cells. The endocannabinoids are tightly regulated by biosynthetic and hydrolytic enzymes. Accordingly, the endocannabinoid system plays a modulatory role in many physiological processes, thereby generating many promising therapeutic targets. An unintended consequence of this research was the emergence of synthetic cannabinoids sold for human consumption to circumvent federal laws banning Cannabis use. Here, we describe research that led to the discovery of the endogenous cannabinoid system and show how knowledge of this system benefitted as well as unintentionally harmed human health.
Collapse
Affiliation(s)
- Lesley D Schurman
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, USA
| | - Dai Lu
- Rangel College of Pharmacy, Health Science Center, Texas A&M University, Kingsville, TX, USA
| | - Debra A Kendall
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT, USA
| | - Allyn C Howlett
- Department of Physiology and Pharmacology and Center for Research on Substance Use and Addiction, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Aron H Lichtman
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, USA.
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA, USA.
| |
Collapse
|
34
|
Allosteric Modulation of Cannabinoid Receptor 1-Current Challenges and Future Opportunities. Int J Mol Sci 2019; 20:ijms20235874. [PMID: 31771126 PMCID: PMC6928801 DOI: 10.3390/ijms20235874] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 11/19/2019] [Accepted: 11/20/2019] [Indexed: 02/07/2023] Open
Abstract
The cannabinoid receptor type 1 (CB1R), a G protein-coupled receptor (GPCR), plays an essential role in the control of many physiological processes such as hunger, memory loss, gastrointestinal activity, catalepsy, fear, depression, and chronic pain. Therefore, it is an attractive target for drug discovery to manage pain, neurodegenerative disorders, obesity, and substance abuse. However, the psychoactive adverse effects, generated by CB1R activation in the brain, limit the use of the orthosteric CB1R ligands as drugs. The discovery of CB1R allosteric modulators during the last decade provided new tools to target the CB1R. Moreover, application of the site-directed mutagenesis in combination with advanced physical methods, especially X-ray crystallography and computational modeling, has opened new horizons for understanding the complexity of the structure, function, and activity of cannabinoid receptors. In this paper, we present the latest advances in research on the CB1R, its allosteric modulation and allosteric ligands, and their translational potential. We focused on structural essentials of the cannabinoid 1 receptor- ligand (drug) interactions, as well as modes of CB1R signaling regulation.
Collapse
|
35
|
Structure of an allosteric modulator bound to the CB1 cannabinoid receptor. Nat Chem Biol 2019; 15:1199-1205. [PMID: 31659318 DOI: 10.1038/s41589-019-0387-2] [Citation(s) in RCA: 113] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Accepted: 09/09/2019] [Indexed: 02/05/2023]
Abstract
The CB1 receptor mediates the central nervous system response to cannabinoids, and is a drug target for pain, anxiety and seizures. CB1 also responds to allosteric modulators, which influence cannabinoid binding and efficacy. To understand the mechanism of these compounds, we solved the crystal structure of CB1 with the negative allosteric modulator (NAM) ORG27569 and the agonist CP55940. The structure reveals that the NAM binds to an extrahelical site within the inner leaflet of the membrane, which overlaps with a conserved site of cholesterol interaction in many G protein-coupled receptors (GPCRs). The ternary structure with ORG27569 and CP55940 captures an intermediate state of the receptor, in which aromatic residues at the base of the agonist-binding pocket adopt an inactive conformation despite the large contraction of the orthosteric pocket. The structure illustrates a potential strategy for drug modulation of CB1 and other class A GPCRs.
Collapse
|
36
|
Páez JA, Campillo NE. Innovative Therapeutic Potential of Cannabinoid Receptors as Targets in Alzheimer’s Disease and Less Well-Known Diseases. Curr Med Chem 2019; 26:3300-3340. [DOI: 10.2174/0929867325666180226095132] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 02/08/2018] [Accepted: 02/15/2018] [Indexed: 02/07/2023]
Abstract
:
The discovery of cannabinoid receptors at the beginning of the 1990s, CB1 cloned
in 1990 and CB2 cloned in 1993, and the availability of selective and potent cannabimimetics
could only be justified by the existence of endogenous ligands that are capable of binding to
them. Thus, the characterisation and cloning of the first cannabinoid receptor (CB1) led to the
isolation and characterisation of the first endocannabinoid, arachidonoylethanolamide (AEA),
two years later and the subsequent identification of a family of lipid transmitters known as the
fatty acid ester 2-arachidonoylglycerol (2-AG).
:
The endogenous cannabinoid system is a complex signalling system that comprises transmembrane
endocannabinoid receptors, their endogenous ligands (the endocannabinoids), the
specific uptake mechanisms and the enzymatic systems related to their biosynthesis and degradation.
:
The endocannabinoid system has been implicated in a wide diversity of biological processes,
in both the central and peripheral nervous systems, including memory, learning, neuronal development,
stress and emotions, food intake, energy regulation, peripheral metabolism, and
the regulation of hormonal balance through the endocrine system.
:
In this context, this article will review the current knowledge of the therapeutic potential of
cannabinoid receptor as a target in Alzheimer’s disease and other less well-known diseases
that include, among others, multiple sclerosis, bone metabolism, and Fragile X syndrome.
:
The therapeutic applications will be addressed through the study of cannabinoid agonists acting
as single drugs and multi-target drugs highlighting the CB2 receptor agonist.
Collapse
Affiliation(s)
- Juan A. Páez
- Instituto de Quimica Medica (IQM-CSIC). C/ Juan de la Cierva, 3, 28006, Madrid, Spain
| | - Nuria E. Campillo
- Centro de Investigaciones Biologicas (CIB-CSIC). C/ Ramiro de Maeztu, 9, 28040, Madrid, Spain
| |
Collapse
|
37
|
Youssif BG, Mohamed AM, Osman EEA, Abou-Ghadir OF, Elnaggar DH, Abdelrahman MH, Treamblu L, Gomaa HA. 5-Chlorobenzofuran-2-carboxamides: From allosteric CB1 modulators to potential apoptotic antitumor agents. Eur J Med Chem 2019; 177:1-11. [DOI: 10.1016/j.ejmech.2019.05.040] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 04/05/2019] [Accepted: 05/13/2019] [Indexed: 01/09/2023]
|
38
|
Bukiya AN, Dopico AM. Cannabinoid Interactions with Proteins: Insights from Structural Studies. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1162:39-50. [PMID: 31332733 DOI: 10.1007/978-3-030-21737-2_3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Cannabinoids have been widely used for recreational and medicinal purposes. The increasing legalization of cannabinoid use and the growing success in Medicinal Chemistry of cannabinoids have fueled recent interest in cannabinoid-sensing sites in receptor proteins. Here, we review structural data from high-resolution cryo-EM and crystallography studies that depict phytocannabinoid, endocannabinoid, and synthetic cannabinoid molecules bound to various proteins. The latter include antigen-binding fragment (Fab), cellular retinol binding protein 2 (CRBP2), fatty acid-binding protein 5 (FABP5), peroxisome proliferator-activated receptor γ (PPAR γ), and cannabinoid receptor types 1 and 2 (CB1 and CB2). Cannabinoid-protein complexes reveal the complex design of cannabinoid binding sites that are usually presented by conventional ligand-binding pockets on respective proteins. However, subtle differences in cannabinoid interaction with amino acids within the binding pocket often result in diverse consequences for protein function. The rapid increase in available structural data on cannabinoid-protein interactions will ultimately direct drug design efforts toward rendering highly potent cannabinoid-related pharmacotherapies that are devoid of side effects.
Collapse
Affiliation(s)
- Anna N Bukiya
- Department of Pharmacology, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN, USA.
| | - Alex M Dopico
- Department of Pharmacology, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN, USA
| |
Collapse
|
39
|
Bian Y, Jing Y, Wang L, Ma S, Jun JJ, Xie XQ. Prediction of Orthosteric and Allosteric Regulations on Cannabinoid Receptors Using Supervised Machine Learning Classifiers. Mol Pharm 2019; 16:2605-2615. [PMID: 31013097 DOI: 10.1021/acs.molpharmaceut.9b00182] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Designing highly selective compounds to protein subtypes and developing allosteric modulators targeting them are critical considerations to both drug discovery and mechanism studies for cannabinoid receptors. It is challenging but in demand to have classifiers to identify active ligands from inactive or random compounds and distinguish allosteric modulators from orthosteric ligands. In this study, supervised machine learning classifiers were built for two subtypes of cannabinoid receptors, CB1 and CB2. Three types of features, including molecular descriptors, MACCS fingerprints, and ECFP6 fingerprints, were calculated to evaluate the compound sets from diverse aspects. Deep neural networks, as well as conventional machine learning algorithms including support vector machine, naïve Bayes, logistic regression, and ensemble learning, were applied. Their performances on the classification with different types of features were compared and discussed. According to the receiver operating characteristic curves and the calculated metrics, the advantages and drawbacks of each algorithm were investigated. The feature ranking was followed to help extract useful knowledge about critical molecular properties, substructural keys, and circular fingerprints. The extracted features will then facilitate the research on cannabinoid receptors by providing guidance on preferred properties for compound modification and novel scaffold design. Besides using conventional molecular docking studies for compound virtual screening, machine-learning-based decision-making models provide alternative options. This study can be of value to the application of machine learning in the area of drug discovery and compound development.
Collapse
|
40
|
Al-Zoubi R, Morales P, Reggio PH. Structural Insights into CB1 Receptor Biased Signaling. Int J Mol Sci 2019; 20:E1837. [PMID: 31013934 PMCID: PMC6515405 DOI: 10.3390/ijms20081837] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 04/09/2019] [Accepted: 04/11/2019] [Indexed: 12/21/2022] Open
Abstract
The endocannabinoid system has emerged as a promising target for the treatment of numerous diseases, including cancer, neurodegenerative disorders, and metabolic syndromes. Thus far, two cannabinoid receptors, CB1 and CB2, have been discovered, which are found predominantly in the central nervous system (CB1) or the immune system (CB2), among other organs and tissues. CB1 receptor ligands have been shown to induce a complex pattern of intracellular effects. The binding of a ligand induces distinct conformational changes in the receptor, which will eventually translate into distinct intracellular signaling pathways through coupling to specific intracellular effector proteins. These proteins can mediate receptor desensitization, trafficking, or signaling. Ligand specificity and selectivity, complex cellular components, and the concomitant expression of other proteins (which either regulate the CB1 receptor or are regulated by the CB1 receptor) will affect the therapeutic outcome of its targeting. With an increased interest in G protein-coupled receptors (GPCR) research, in-depth studies using mutations, biological assays, and spectroscopic techniques (such as NMR, EPR, MS, FRET, and X-ray crystallography), as well as computational modelling, have begun to reveal a set of concerted structural features in Class A GPCRs which relate to signaling pathways and the mechanisms of ligand-induced activation, deactivation, or activity modulation. This review will focus on the structural features of the CB1 receptor, mutations known to bias its signaling, and reported studies of CB1 receptor ligands to control its specific signaling.
Collapse
Affiliation(s)
- Rufaida Al-Zoubi
- Department of Medicinal Chemistry and Pharmacognosy, Faculty of Pharmacy, Jordan University of Science & Technology, P.O.BOX 3030, Irbid 22110, Jordan.
| | - Paula Morales
- Departamento de Química-Física Biológica, Instituto de Química Física Rocasolano (IQFR-CSIC), Serrano 119, 28006 Madrid, Spain.
| | - Patricia H Reggio
- Chemistry and Biochemistry Department, UNC Greensboro, Greensboro, NC 27412, USA.
| |
Collapse
|
41
|
Li-Zhulanov NS, Il’ina IV, Chicca A, Schenker P, Patrusheva OS, Nazimova EV, Korchagina DV, Krasavin M, Volcho KP, Salakhutdinov NF. Effect of chiral polyhydrochromenes on cannabinoid system. Med Chem Res 2019. [DOI: 10.1007/s00044-019-02294-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
42
|
Trexler KR, Eckard ML, Kinsey SG. CB 1 positive allosteric modulation attenuates Δ 9-THC withdrawal and NSAID-induced gastric inflammation. Pharmacol Biochem Behav 2019; 177:27-33. [PMID: 30597181 PMCID: PMC6405226 DOI: 10.1016/j.pbb.2018.12.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 12/06/2018] [Accepted: 12/27/2018] [Indexed: 02/07/2023]
Abstract
Recently, multiple compounds have been synthesized that target the allosteric binding site(s) of CB1. These CB1 positive allosteric modulators may capture the benefits of cannabinoid receptor activation without unwanted psychoactive effects, such as sedation. For example, ZCZ011 blocks neuropathic pain, absent the catalepsy, sedation, and hypothermia caused by CB1 orthosteric modulators, including Δ9-tetrahydrocannabinol (THC). The primary goal of the present study was to evaluate the potential of ZCZ011 to attenuate somatic signs of cannabinoid withdrawal in mice. Mice were repeatedly administered THC (10 mg/kg, s.c.) or vehicle, and withdrawal was either precipitated using the CB1 antagonist rimonabant (3 mg/kg, i.p.) or elicited spontaneously via THC abstinence. ZCZ011 (≥10 mg/kg, i.p.) significantly attenuated somatic signs of withdrawal, including head twitches and paw tremors, but had no effect on locomotor activity or conditioned place preference. We next tested the antiulcerogenic properties of CB1 positive allosteric modulation. Mice were fasted for 22 h, administered ZCZ011, and gastric hemorrhages were induced with the nonsteroidal anti-inflammatory drug diclofenac sodium (100 mg/kg, p.o.). ZCZ011 alone had no effect on gastric ulceration, but ZCZ011 (≥10 mg/kg) blocked ulcer formation when combined with a subthreshold MAGL inhibitor (JZL184; 1 mg/kg, i.p.). Thus, CB1 positive allosteric modulation is a novel approach to treat cannabinoid dependence and gastric inflammation.
Collapse
Affiliation(s)
- K R Trexler
- Department of Psychology, West Virginia University, Morgantown, WV, USA
| | - M L Eckard
- Department of Psychology, West Virginia University, Morgantown, WV, USA
| | - S G Kinsey
- Department of Psychology, West Virginia University, Morgantown, WV, USA; Department of Neuroscience, West Virginia University, Morgantown, WV, USA.
| |
Collapse
|
43
|
Nguyen T, Gamage TF, Decker AM, German N, Langston TL, Farquhar CE, Kenakin TP, Wiley JL, Thomas BF, Zhang Y. Diarylureas Containing 5-Membered Heterocycles as CB 1 Receptor Allosteric Modulators: Design, Synthesis, and Pharmacological Evaluation. ACS Chem Neurosci 2019; 10:518-527. [PMID: 30188693 DOI: 10.1021/acschemneuro.8b00396] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Allosteric modulators have attracted significant interest as an alternate strategy to modulate CB1 receptor signaling for therapeutic benefits that may avoid the adverse effects associated with orthosteric ligands. Here we extended our previous structure-activity relationship studies on the diarylurea-based CB1 negative allosteric modulators (NAMs) by introducing five-membered heterocycles to replace the 5-pyrrolidinylpyridinyl group in PSNCBAM-1 (1), one of the first generation CB1 allosteric modulators. Many of these compounds had comparable potency to 1 in blocking the CB1 agonist CP55,940 stimulated calcium mobilization and [35S]GTP-γ-S binding. Similar to 1, most compounds showed positive cooperativity by increasing [3H]CP55,940 binding, consistent with the positive allosteric modulator (PAM)-antagonist mechanism. Interestingly, these compounds exhibited differences in ability to increase specific binding of [3H]CP55,940 and decrease binding of the antagonist [3H]SR141716. In saturation binding studies, only increases in [3H]CP55,940 Bmax, but not Kd, were observed, suggesting that these compounds stabilize low affinity receptors into a high affinity state. Among the series, the 2-pyrrolyl analogue (13) exhibited greater potency than 1 in the [35S]GTP-γ-S binding assay and significantly enhanced the maximum binding level in the [3H]CP5,5940 binding assay, indicating greater CB1 receptor affinity and/or cooperativity.
Collapse
Affiliation(s)
- Thuy Nguyen
- Research Triangle Institute, Research Triangle Park, North Carolina 27709, United States
| | - Thomas F. Gamage
- Research Triangle Institute, Research Triangle Park, North Carolina 27709, United States
| | - Ann M. Decker
- Research Triangle Institute, Research Triangle Park, North Carolina 27709, United States
| | - Nadezhda German
- Research Triangle Institute, Research Triangle Park, North Carolina 27709, United States
| | - Tiffany L. Langston
- Research Triangle Institute, Research Triangle Park, North Carolina 27709, United States
| | - Charlotte E. Farquhar
- Research Triangle Institute, Research Triangle Park, North Carolina 27709, United States
| | - Terry P. Kenakin
- Department of Pharmacology, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| | - Jenny L. Wiley
- Research Triangle Institute, Research Triangle Park, North Carolina 27709, United States
| | - Brian F. Thomas
- Research Triangle Institute, Research Triangle Park, North Carolina 27709, United States
| | - Yanan Zhang
- Research Triangle Institute, Research Triangle Park, North Carolina 27709, United States
| |
Collapse
|
44
|
Emendato A, Guerrini R, Marzola E, Wienk H, Boelens R, Leone S, Picone D. Disordered Peptides Looking for Their Native Environment: Structural Basis of CB1 Endocannabinoid Receptor Binding to Pepcans. Front Mol Biosci 2018; 5:100. [PMID: 30505835 PMCID: PMC6250848 DOI: 10.3389/fmolb.2018.00100] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 10/26/2018] [Indexed: 01/15/2023] Open
Abstract
Endocannabinoid peptides, or “pepcans,” are endogenous ligands of the CB1 cannabinoid receptor. Depending on their length, they display diverse activity: For instance, the nona-peptide Pepcan-9, also known as hemopressin, is a powerful inhibitor of CB1, whereas the longer variant Pepcan-12, which extends by only three amino acid residues at the N-terminus, acts on both CB1 and CB2 as an allosteric modulator, although with diverse effects. Despite active research on their pharmacological applications, very little is known about structure-activity relationships of pepcans. Different structures have been proposed for the nona-peptide, which has also been reported to form fibrillar aggregates. This might have affected the outcome and reproducibility of bioactivity studies. In an attempt of elucidating the determinants of both biological activity and aggregation propensity of Pepcan-9 and Pepcan-12, we have performed their structure characterization in solvent systems characterized by different polarity and pH. We have found that, while disordered in aqueous environment, both peptides display helical structure in less polar environment, mimicking the proteic receptor milieu. In the case of Pepcan-9, this structure is fully consistent with the observed modulation of the CB1. For Pepcan-12, whose allosteric binding site is still unknown, the presented structure is compatible with the binding at one of the previously proposed allosteric sites on CB1. These findings open the way to structure-driven design of selective peptide modulators of CB1.
Collapse
Affiliation(s)
- Alessandro Emendato
- Department of Chemical Sciences, University of Naples Federico II, Naples, Italy
| | - Remo Guerrini
- Department of Chemical and Pharmaceutical Sciences, University of Ferrara, Ferrara, Italy
| | - Erika Marzola
- Department of Chemical and Pharmaceutical Sciences, University of Ferrara, Ferrara, Italy
| | - Hans Wienk
- Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, Netherlands
| | - Rolf Boelens
- Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, Netherlands
| | - Serena Leone
- Department of Chemical Sciences, University of Naples Federico II, Naples, Italy
| | - Delia Picone
- Department of Chemical Sciences, University of Naples Federico II, Naples, Italy
| |
Collapse
|
45
|
De Aquino JP, Ross DA. Cannabinoids and Pain: Weeding Out Undesired Effects With a Novel Approach to Analgesia. Biol Psychiatry 2018; 84:e67-e69. [PMID: 30360776 PMCID: PMC6697049 DOI: 10.1016/j.biopsych.2018.09.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 09/11/2018] [Accepted: 09/11/2018] [Indexed: 01/27/2023]
|
46
|
Vučković S, Srebro D, Vujović KS, Vučetić Č, Prostran M. Cannabinoids and Pain: New Insights From Old Molecules. Front Pharmacol 2018; 9:1259. [PMID: 30542280 PMCID: PMC6277878 DOI: 10.3389/fphar.2018.01259] [Citation(s) in RCA: 178] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 10/15/2018] [Indexed: 01/11/2023] Open
Abstract
Cannabis has been used for medicinal purposes for thousands of years. The prohibition of cannabis in the middle of the 20th century has arrested cannabis research. In recent years there is a growing debate about the use of cannabis for medical purposes. The term ‘medical cannabis’ refers to physician-recommended use of the cannabis plant and its components, called cannabinoids, to treat disease or improve symptoms. Chronic pain is the most commonly cited reason for using medical cannabis. Cannabinoids act via cannabinoid receptors, but they also affect the activities of many other receptors, ion channels and enzymes. Preclinical studies in animals using both pharmacological and genetic approaches have increased our understanding of the mechanisms of cannabinoid-induced analgesia and provided therapeutical strategies for treating pain in humans. The mechanisms of the analgesic effect of cannabinoids include inhibition of the release of neurotransmitters and neuropeptides from presynaptic nerve endings, modulation of postsynaptic neuron excitability, activation of descending inhibitory pain pathways, and reduction of neural inflammation. Recent meta-analyses of clinical trials that have examined the use of medical cannabis in chronic pain present a moderate amount of evidence that cannabis/cannabinoids exhibit analgesic activity, especially in neuropathic pain. The main limitations of these studies are short treatment duration, small numbers of patients, heterogeneous patient populations, examination of different cannabinoids, different doses, the use of different efficacy endpoints, as well as modest observable effects. Adverse effects in the short-term medical use of cannabis are generally mild to moderate, well tolerated and transient. However, there are scant data regarding the long-term safety of medical cannabis use. Larger well-designed studies of longer duration are mandatory to determine the long-term efficacy and long-term safety of cannabis/cannabinoids and to provide definitive answers to physicians and patients regarding the risk and benefits of its use in the treatment of pain. In conclusion, the evidence from current research supports the use of medical cannabis in the treatment of chronic pain in adults. Careful follow-up and monitoring of patients using cannabis/cannabinoids are mandatory.
Collapse
Affiliation(s)
- Sonja Vučković
- Department of Pharmacology, Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Dragana Srebro
- Department of Pharmacology, Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Katarina Savić Vujović
- Department of Pharmacology, Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Čedomir Vučetić
- Clinic of Orthopaedic Surgery and Traumatology, Clinical Center of Serbia, Belgrade, Serbia.,Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Milica Prostran
- Department of Pharmacology, Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
47
|
Jagla CAD, Scott CE, Tang Y, Qiao C, Mateo-Semidey GE, Yudowski GA, Lu D, Kendall DA. Pyrimidinyl Biphenylureas Act as Allosteric Modulators to Activate Cannabinoid Receptor 1 and Initiate β-Arrestin-Dependent Responses. Mol Pharmacol 2018; 95:1-10. [PMID: 30322873 DOI: 10.1124/mol.118.112854] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 10/12/2018] [Indexed: 01/14/2023] Open
Abstract
Cannabinoid receptor 1 (CB1) is a G-protein-coupled receptor that is abundant in the central nervous system. It binds several compounds in its orthosteric site, including the endocannabinoids, arachidonoyl ethanolamide (anandamide) and 2-arachidonoyl glycerol, and the plant-derived Δ9-tetrahydrocannabinol, one of the main psychoactive components of marijuana. It primarily couples to Gi/o proteins to inhibit adenylate cyclase activity and typically induces downstream signaling that is Gi-dependent. Since this receptor is implicated in several maladies, such as obesity, pain, and neurodegenerative disorders, there is interest in developing therapeutics that selectively target this receptor. Allosteric modulators of CB1 offer one new approach that has tremendous therapeutic potential. Here, we reveal receptor- and cellular-level properties consistent with receptor activation by a series of pyrimidinyl biphenylureas (LDK1285, LDK1288, LDK1305, and PSNCBAM1), including promoting binding of the agonist CP55940 with positive cooperativity and inhibiting binding of the inverse agonist SR141716A with negative cooperativity, demonstrated via radioligand binding studies. Consistent with these findings, the allosteric modulators induced cellular internalization of the receptor and recruitment of β-arrestin 2 in human embryonic kidney cell line 293 cells monitored with confocal and total internal reflective fluorescence microscopy, respectively. These allosteric modulators, however, caused G-protein-independent but β-arrestin 1-dependent phosphorylation of the downstream kinases extracellular signal-regulated kinase 1/2, mitogen-activated protein kinase, and Src, shown by immunoblotting studies. These results are consistent with the involvement of β-arrestin and suggest that these allosteric modulators induce biased signaling.
Collapse
Affiliation(s)
- Caitlin A D Jagla
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, Connecticut (C.A.D.J., C.E.S., Y.T., D.A.K.); Department of Anatomy and Neurobiology (G.E.M.-S., G.A.Y.) and Institute of Neurobiology (G.E.M.-S., G.A.Y.), University of Puerto Rico, San Juan, Puerto Rico; and Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M Health Science Center, Kingsville, Texas (C.Q., D.L.)
| | - Caitlin E Scott
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, Connecticut (C.A.D.J., C.E.S., Y.T., D.A.K.); Department of Anatomy and Neurobiology (G.E.M.-S., G.A.Y.) and Institute of Neurobiology (G.E.M.-S., G.A.Y.), University of Puerto Rico, San Juan, Puerto Rico; and Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M Health Science Center, Kingsville, Texas (C.Q., D.L.)
| | - Yaliang Tang
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, Connecticut (C.A.D.J., C.E.S., Y.T., D.A.K.); Department of Anatomy and Neurobiology (G.E.M.-S., G.A.Y.) and Institute of Neurobiology (G.E.M.-S., G.A.Y.), University of Puerto Rico, San Juan, Puerto Rico; and Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M Health Science Center, Kingsville, Texas (C.Q., D.L.)
| | - Changjiang Qiao
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, Connecticut (C.A.D.J., C.E.S., Y.T., D.A.K.); Department of Anatomy and Neurobiology (G.E.M.-S., G.A.Y.) and Institute of Neurobiology (G.E.M.-S., G.A.Y.), University of Puerto Rico, San Juan, Puerto Rico; and Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M Health Science Center, Kingsville, Texas (C.Q., D.L.)
| | - Gabriel E Mateo-Semidey
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, Connecticut (C.A.D.J., C.E.S., Y.T., D.A.K.); Department of Anatomy and Neurobiology (G.E.M.-S., G.A.Y.) and Institute of Neurobiology (G.E.M.-S., G.A.Y.), University of Puerto Rico, San Juan, Puerto Rico; and Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M Health Science Center, Kingsville, Texas (C.Q., D.L.)
| | - Guillermo A Yudowski
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, Connecticut (C.A.D.J., C.E.S., Y.T., D.A.K.); Department of Anatomy and Neurobiology (G.E.M.-S., G.A.Y.) and Institute of Neurobiology (G.E.M.-S., G.A.Y.), University of Puerto Rico, San Juan, Puerto Rico; and Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M Health Science Center, Kingsville, Texas (C.Q., D.L.)
| | - Dai Lu
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, Connecticut (C.A.D.J., C.E.S., Y.T., D.A.K.); Department of Anatomy and Neurobiology (G.E.M.-S., G.A.Y.) and Institute of Neurobiology (G.E.M.-S., G.A.Y.), University of Puerto Rico, San Juan, Puerto Rico; and Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M Health Science Center, Kingsville, Texas (C.Q., D.L.)
| | - Debra A Kendall
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, Connecticut (C.A.D.J., C.E.S., Y.T., D.A.K.); Department of Anatomy and Neurobiology (G.E.M.-S., G.A.Y.) and Institute of Neurobiology (G.E.M.-S., G.A.Y.), University of Puerto Rico, San Juan, Puerto Rico; and Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M Health Science Center, Kingsville, Texas (C.Q., D.L.)
| |
Collapse
|
48
|
Lupica CR, Hoffman AF. Cannabinoid disruption of learning mechanisms involved in reward processing. ACTA ACUST UNITED AC 2018; 25:435-445. [PMID: 30115765 PMCID: PMC6097761 DOI: 10.1101/lm.046748.117] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 07/06/2018] [Indexed: 02/06/2023]
Abstract
The increasing use of cannabis, its derivatives, and synthetic cannabinoids for medicinal and recreational purposes has led to burgeoning interest in understanding the addictive potential of this class of molecules. It is estimated that ∼10% of marijuana users will eventually show signs of dependence on the drug, and the diagnosis of cannabis use disorder (CUD) is increasing in the United States. The molecule that sustains the use of cannabis is Δ9-tetrahydrocannabinol (Δ9-THC), and our knowledge of its effects, and those of other cannabinoids on brain function has expanded rapidly in the past two decades. Additionally, the identification of endogenous cannabinoid (endocannabinoid) systems in brain and their roles in physiology and behavior, demonstrate extensive involvement of these lipid signaling molecules in regulating CNS function. Here, we examine roles for endogenous cannabinoids in shaping synaptic activity in cortical and subcortical brain circuits, and we discuss mechanisms in which exogenous cannabinoids, such as Δ9-THC, interact with endocannabinoid systems to disrupt neuronal network oscillations. We then explore how perturbation of the interaction of this activity within brain reward circuits may lead to impaired learning. Finally, we propose that disruption of cellular plasticity mechanisms by exogenous cannabinoids in cortical and subcortical circuits may explain the difficulty in establishing viable cannabinoid self-administration models in animals.
Collapse
Affiliation(s)
- Carl R Lupica
- Electrophysiology Research Section, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, Maryland 21224, USA
| | - Alexander F Hoffman
- Electrophysiology Research Section, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, Maryland 21224, USA
| |
Collapse
|
49
|
Gil-Ordóñez A, Martín-Fontecha M, Ortega-Gutiérrez S, López-Rodríguez ML. Monoacylglycerol lipase (MAGL) as a promising therapeutic target. Biochem Pharmacol 2018; 157:18-32. [PMID: 30059673 DOI: 10.1016/j.bcp.2018.07.036] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 07/25/2018] [Indexed: 12/31/2022]
Abstract
Monoacylglycerol lipase (MAGL) has been characterized as the main enzyme responsible for the inactivation of the most abundant brain endocannabinoid, 2-arachidonoylglycerol (2-AG). Besides this role, MAGL has progressively acquired a growing importance as an integrative metabolic hub that controls not only the in vivo levels of 2-AG but also of other monoacylglycerides and, indirectly, the levels of free fatty acids derived from their hydrolysis as well as other lipids with pro-inflammatory or pro-tumorigenic effects, coming from the further metabolism of fatty acids. All these functions have only started to be elucidated in the last years due to the progress made in the knowledge of the structure of MAGL and in the development of genetic and chemical tools. In this review we report the advances made in the field with a special focus on the last decade and how MAGL has become a promising therapeutic target for the treatment of several diseases that currently lack appropriate therapies.
Collapse
Affiliation(s)
- Ana Gil-Ordóñez
- Department of Organic Chemistry, School of Chemistry, Universidad Complutense de Madrid, Av. Complutense s/n, E-28040 Madrid, Spain
| | - Mar Martín-Fontecha
- Department of Organic Chemistry, School of Chemistry, Universidad Complutense de Madrid, Av. Complutense s/n, E-28040 Madrid, Spain
| | - Silvia Ortega-Gutiérrez
- Department of Organic Chemistry, School of Chemistry, Universidad Complutense de Madrid, Av. Complutense s/n, E-28040 Madrid, Spain
| | - María L López-Rodríguez
- Department of Organic Chemistry, School of Chemistry, Universidad Complutense de Madrid, Av. Complutense s/n, E-28040 Madrid, Spain.
| |
Collapse
|
50
|
Gado F, Di Cesare Mannelli L, Lucarini E, Bertini S, Cappelli E, Digiacomo M, Stevenson LA, Macchia M, Tuccinardi T, Ghelardini C, Pertwee RG, Manera C. Identification of the First Synthetic Allosteric Modulator of the CB 2 Receptors and Evidence of Its Efficacy for Neuropathic Pain Relief. J Med Chem 2018; 62:276-287. [PMID: 29990428 DOI: 10.1021/acs.jmedchem.8b00368] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The direct activation of cannabinoid receptors (CBRs) results in several beneficial effects; therefore several CBRs ligands have been synthesized and tested in vitro and in vivo. However, none of them reached an advanced phase of clinical development due mainly to side effects on the CNS. Medicinal chemistry approaches are now engaged to develop allosteric modulators that might offer a novel therapeutic approach to achieve potential therapeutic benefits avoiding inherent side effects of orthosteric ligands. Here we identify the first ever synthesized positive allosteric modulator (PAM) that targets CB2Rs. The evidence for this was obtained using [3H]CP55940 and [35S]GTPγS binding assays. This finding will be useful for the characterization of allosteric binding site(s) on CB2Rs which will be essential for the further development of CB2R allosteric modulators. Moreover, the new CB2R PAM displayed antinociceptive activity in vivo in an experimental mouse model of neuropathic pain, raising the possibility that it might be a good candidate for clinical development.
Collapse
Affiliation(s)
- Francesca Gado
- Department of Pharmacy , University of Pisa , 56126 Pisa , Italy
| | - Lorenzo Di Cesare Mannelli
- Department of Neuroscience, Psychology, Drug Research and Child Health, Section of Pharmacology and Toxicology , University of Florence , 50139 Florence , Italy
| | - Elena Lucarini
- Department of Neuroscience, Psychology, Drug Research and Child Health, Section of Pharmacology and Toxicology , University of Florence , 50139 Florence , Italy
| | - Simone Bertini
- Department of Pharmacy , University of Pisa , 56126 Pisa , Italy
| | - Elena Cappelli
- Department of Pharmacy , University of Pisa , 56126 Pisa , Italy
| | - Maria Digiacomo
- Department of Pharmacy , University of Pisa , 56126 Pisa , Italy
| | - Lesley A Stevenson
- School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences , University of Aberdeen , AB25 2ZD Aberdeen , Scotland, U.K
| | - Marco Macchia
- Department of Pharmacy , University of Pisa , 56126 Pisa , Italy
| | - Tiziano Tuccinardi
- Department of Pharmacy , University of Pisa , 56126 Pisa , Italy.,Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology , Temple University , Philadelphia , Pennsylvania 19122 , United States
| | - Carla Ghelardini
- Department of Neuroscience, Psychology, Drug Research and Child Health, Section of Pharmacology and Toxicology , University of Florence , 50139 Florence , Italy
| | - Roger G Pertwee
- School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences , University of Aberdeen , AB25 2ZD Aberdeen , Scotland, U.K
| | | |
Collapse
|