1
|
Squire E, Lee HL, Jeong W, Lee S, Ravichandiran V, Limoli CL, Piomelli D, Parihar VK, Jung KM. Targeting dysfunctional endocannabinoid signaling in a mouse model of Gulf War illness. Neuropharmacology 2024; 261:110142. [PMID: 39241906 DOI: 10.1016/j.neuropharm.2024.110142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 07/30/2024] [Accepted: 09/02/2024] [Indexed: 09/09/2024]
Abstract
Gulf War Illness (GWI) is a chronic disorder characterized by a heterogeneous set of symptoms that include pain, fatigue, anxiety, and cognitive impairment. These are thought to stem from damage caused by exposure under unpredictable stress to toxic Gulf War (GW) chemicals, which include pesticides, nerve agents, and prophylactic drugs. We hypothesized that GWI pathogenesis might be rooted in long-lasting disruption of the endocannabinoid (ECB) system, a signaling complex that serves important protective functions in the brain. Using a mouse model of GWI, we found that tissue levels of the ECB messenger, anandamide, were significantly reduced in the brain of diseased mice, compared to healthy controls. In addition, transcription of the Faah gene, which encodes for fatty acid amide hydrolase (FAAH), the enzyme that deactivates anandamide, was significant elevated in prefrontal cortex of GWI mice and brain microglia. Behavioral deficits exhibited by these animals, including heightened anxiety-like and depression-like behaviors, and defective extinction of fearful memories, were corrected by administration of the FAAH inhibitor, URB597, which normalized brain anandamide levels. Furthermore, GWI mice displayed unexpected changes in the microglial transcriptome, implying persistent dampening of homeostatic surveillance genes and abnormal expression of pro-inflammatory genes upon immune stimulation. Together, these results suggest that exposure to GW chemicals produce a deficit in brain ECB signaling which is associated with persistent alterations in microglial function. Pharmacological normalization of anandamide-mediated ECB signaling may offer an effective therapeutic strategy for ameliorating GWI symptomology.
Collapse
Affiliation(s)
- Erica Squire
- Department of Anatomy and Neurobiology, University of California, Irvine, CA, 92697, USA
| | - Hye-Lim Lee
- Department of Anatomy and Neurobiology, University of California, Irvine, CA, 92697, USA
| | - Woojin Jeong
- Department of Anatomy and Neurobiology, University of California, Irvine, CA, 92697, USA
| | - Sumin Lee
- Department of Anatomy and Neurobiology, University of California, Irvine, CA, 92697, USA
| | - V Ravichandiran
- National Institute of Pharmaceutical Education and Research, Hajipur, Bihar, 844102, India
| | - Charles L Limoli
- Radiation Oncology, University of California, Irvine, CA 92697, USA
| | - Daniele Piomelli
- Department of Anatomy and Neurobiology, University of California, Irvine, CA, 92697, USA; Department of Biological Chemistry, University of California, Irvine, CA, 92697, USA; Department of Pharmaceutical Sciences, University of California, Irvine, CA, 92697, USA
| | - Vipan Kumar Parihar
- National Institute of Pharmaceutical Education and Research, Hajipur, Bihar, 844102, India; Radiation Oncology, University of California, Irvine, CA 92697, USA.
| | - Kwang-Mook Jung
- Department of Anatomy and Neurobiology, University of California, Irvine, CA, 92697, USA.
| |
Collapse
|
2
|
Yuan X, Ye W, Chen L, Luo D, Zhou L, Qiu Y, Zhuo R, Zhao Y, Peng L, Yang L, Jin X, Zhou Y. URB597 exerts neuroprotective effects against transient brain ischemia injury in mice by regulating autophagic flux and necroptosis. Eur J Pharmacol 2023; 957:175982. [PMID: 37572942 DOI: 10.1016/j.ejphar.2023.175982] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Revised: 07/14/2023] [Accepted: 08/09/2023] [Indexed: 08/14/2023]
Abstract
Ischemic stroke is a leading cause of death and disability, and medical treatments for ischemic stroke are very limited. URB597 is a potent and selective inhibitor of fatty acid amide hydrolase (FAAH). However, the effect of URB597 on ischemic stroke and the underlying molecular mechanisms remain little known. In this study, focal cerebral ischemia was induced by transient middle cerebral artery occlusion in mice. Our results showed that URB597 dose-dependently improved neurological function and reduced brain infarct volume and brain edema 24 h after brain ischemia. The most effective dose was 1 mg/kg and the therapeutic time window was within 3 h after ischemic stroke. To further investigate the underlying mechanism, necroptosis and autophagy flux were detected by Western blot and/or immunofluorescence staining with or without chloroquine, an autophagic flux inhibitor. Our results showed that URB597 promoted autophagic flux and reduced neuronal necroptosis after brain ischemia and these effects could be abolished by chloroquine. In addition, we found that peroxisome proliferator-activated receptor α (PPARα) antagonist GW6471 partly abolished the effect of URB597 against brain ischemia and URB597 upregulated the expressions of PPARα. In conclusion, URB597 exerts a neuroprotective effect in a dose- and time-dependent manner, and this effect may be related to its restoration of autophagic flux and inhibition of neuronal necroptosis. PPARα is involved in the neuroprotective effect of URB597. This study provides novel evidence that URB597 may be a promising agent for the clinical treatment of ischemic stroke.
Collapse
Affiliation(s)
- Xiaoqian Yuan
- Department of Basic Medical Science, School of Medicine, Xiamen University, Xiamen, 361102, China; Key Laboratory of Chiral Drugs, Xiamen, 361102, China.
| | - Wenxuan Ye
- Department of Basic Medical Science, School of Medicine, Xiamen University, Xiamen, 361102, China; Key Laboratory of Chiral Drugs, Xiamen, 361102, China
| | - Ling Chen
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361102, China; Department of Basic Medical Science, School of Medicine, Xiamen University, Xiamen, 361102, China; Key Laboratory of Chiral Drugs, Xiamen, 361102, China
| | - Doudou Luo
- Department of Basic Medical Science, School of Medicine, Xiamen University, Xiamen, 361102, China; Key Laboratory of Chiral Drugs, Xiamen, 361102, China; State Key Laboratory of Cellular Stress Biology, Xiamen University, Xiamen, 361102, China
| | - Li Zhou
- Department of Basic Medical Science, School of Medicine, Xiamen University, Xiamen, 361102, China
| | - Yan Qiu
- Key Laboratory of Chiral Drugs, Xiamen, 361102, China
| | - Rengong Zhuo
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361102, China; Department of Basic Medical Science, School of Medicine, Xiamen University, Xiamen, 361102, China; Key Laboratory of Chiral Drugs, Xiamen, 361102, China
| | - Yun Zhao
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361102, China; Department of Basic Medical Science, School of Medicine, Xiamen University, Xiamen, 361102, China; Key Laboratory of Chiral Drugs, Xiamen, 361102, China
| | - Lu Peng
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361102, China; Department of Basic Medical Science, School of Medicine, Xiamen University, Xiamen, 361102, China; Key Laboratory of Chiral Drugs, Xiamen, 361102, China
| | - Lichao Yang
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361102, China; Department of Basic Medical Science, School of Medicine, Xiamen University, Xiamen, 361102, China; Key Laboratory of Chiral Drugs, Xiamen, 361102, China
| | - Xin Jin
- Department of Basic Medical Science, School of Medicine, Xiamen University, Xiamen, 361102, China; Key Laboratory of Chiral Drugs, Xiamen, 361102, China
| | - Yu Zhou
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361102, China; Department of Basic Medical Science, School of Medicine, Xiamen University, Xiamen, 361102, China; Key Laboratory of Chiral Drugs, Xiamen, 361102, China; State Key Laboratory of Cellular Stress Biology, Xiamen University, Xiamen, 361102, China.
| |
Collapse
|
3
|
Berg BB, Linhares AFS, Martins DM, Rachid MA, Cau SBDA, Souza GGD, Carvalho JCSD, Sorgi CA, Romero TRL, Pinho V, Teixeira MM, Castor MGME. Anandamide reduces the migration of lymphocytes to the intestine by CB2 activation and reduces TNF-α in the target organs, protecting mice from graft-versus-host disease. Eur J Pharmacol 2023; 956:175932. [PMID: 37536622 DOI: 10.1016/j.ejphar.2023.175932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 07/18/2023] [Accepted: 07/19/2023] [Indexed: 08/05/2023]
Abstract
Graft-versus-host disease (GVHD) is a serious inflammatory illness that often occurs as a secondary complication of bone marrow transplantation. Current therapies have limited effectiveness and fail to achieve a balance between inflammation and the graft-versus-tumor effect. In this study, we investigate the effects of the endocannabinoid anandamide on the complex pathology of GVHD. We assess the effects of an irreversible inhibitor of fatty acid amine hydrolase or exogenous anandamide and find that they increase survival and reduce clinical signs in GVHD mice. In the intestine of GVHD mice, treatment with exogenous anandamide also leads to a reduction in the number of CD3+, CD3+CD4+, and CD3+CD8+ cells, which reduces the activation of CD3+CD4+ and CD3+CD8+ cells, as assessed by enhanced CD28 expression, a T cell co-stimulatory molecule. Exogenous AEA was also able to reduce TNF-α and increase IL-10 in the intestine of GVHD mice. In the liver, exogenous AEA reduces injury, TNF-α levels, and the number of CD3+CD8+ cells. Interestingly, anandamide reduces Mac-1α, which lowers the adhesion of transplanted cells in mesenteric veins. These effects are mimicked by JWH133-a CB2 selective agonist-and abolished by treatment with a CB2 antagonist. Furthermore, the effects caused by anandamide treatment on survival were related to the CB2 receptor, as the CB2 antagonist abolished it. This study shows the critical role of the CB2 receptor in the modulation of the inflammatory response of GVHD by treatment with anandamide, the most prominent endocannabinoid.
Collapse
Affiliation(s)
- Bárbara Betônico Berg
- Graduate Program in Biological Sciences: Physiology and Pharmacology, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Ana Flávia Santos Linhares
- Graduate Program in Biological Sciences: Physiology and Pharmacology, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | | | | | | | | | | | - Carlos Arterio Sorgi
- Chemistry Department, Faculty of Philosophy Sciences and Letters of Ribeirão Preto, São Paulo, Brazil
| | | | - Vanessa Pinho
- Morphology Department, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Mauro Martins Teixeira
- Department of Biochemistry and Immunology, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | | |
Collapse
|
4
|
Alegre-Zurano L, García-Baos A, Castro-Zavala A, Medrano M, Gallego-Landin I, Valverde O. The FAAH inhibitor URB597 reduces cocaine intake during conditioned punishment and mitigates cocaine seeking during withdrawal. Biomed Pharmacother 2023; 165:115194. [PMID: 37499453 DOI: 10.1016/j.biopha.2023.115194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 06/30/2023] [Accepted: 07/18/2023] [Indexed: 07/29/2023] Open
Abstract
The endocannabinoid system is prominently implicated in the control of cocaine reinforcement due to its relevant role in synaptic plasticity and neurotransmitter modulation in the mesocorticolimbic system. The inhibition of fatty acid amide hydrolase (FAAH), and the resulting increase in anandamide and other N-acylethanolamines, represents a promising strategy for reducing drug seeking. In the present study, we aimed to assess the effects of the FAAH inhibitor URB597 (1 mg/kg) on crucial features of cocaine addictive-like behaviour in mice. Therefore, we tested the effects of URB597 on acquisition of cocaine (0.6 mg/kg/inf) self-administration, compulsive-like cocaine intake and cue-induced drug-seeking behaviour during withdrawal. URB597 reduced cocaine intake under conditioned punishment while having no impact on acquisition. This result was associated to increased cannabinoid receptor 1 gene expression in the ventral striatum and medium spiny neurons activation in the nucleus accumbens shell. Moreover, URB597 mitigated cue-induced drug-seeking behaviour during prolonged abstinence and prevented the withdrawal-induced increase in FAAH gene expression in the ventral striatum. In this case, URB597 decreased activation of medium spiny neurons in the nucleus accumbens core. Our findings evidence the prominent role of endocannabinoids in the development of cocaine addictive-like behaviours and support the potential of FAAH inhibition as a therapeutical target for the treatment of cocaine addiction.
Collapse
Affiliation(s)
- Laia Alegre-Zurano
- Neurobiology of Behaviour Research Group (GReNeC-NeuroBio), Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Alba García-Baos
- Neurobiology of Behaviour Research Group (GReNeC-NeuroBio), Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain; Neuroscience Research Program, IMIM-Hospital Del Mar Research Institute, Barcelona, Spain
| | - Adriana Castro-Zavala
- Neurobiology of Behaviour Research Group (GReNeC-NeuroBio), Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Mireia Medrano
- Neurobiology of Behaviour Research Group (GReNeC-NeuroBio), Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Ines Gallego-Landin
- Neurobiology of Behaviour Research Group (GReNeC-NeuroBio), Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Olga Valverde
- Neurobiology of Behaviour Research Group (GReNeC-NeuroBio), Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain; Neuroscience Research Program, IMIM-Hospital Del Mar Research Institute, Barcelona, Spain.
| |
Collapse
|
5
|
Garcia-Baos A, Pastor A, Gallego-Landin I, de la Torre R, Sanz F, Valverde O. The role of PPAR-γ in memory deficits induced by prenatal and lactation alcohol exposure in mice. Mol Psychiatry 2023; 28:3373-3383. [PMID: 37491462 DOI: 10.1038/s41380-023-02191-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 07/11/2023] [Accepted: 07/13/2023] [Indexed: 07/27/2023]
Abstract
Patients diagnosed with fetal alcohol spectrum disorder (FASD) show persistent cognitive disabilities, including memory deficits. However, the neurobiological substrates underlying these deficits remain unclear. Here, we show that prenatal and lactation alcohol exposure (PLAE) in mice induces FASD-like memory impairments. This is accompanied by a reduction of N-acylethanolamines (NAEs) and peroxisome proliferator-activated receptor gamma (PPAR-γ) in the hippocampus specifically in a childhood-like period (at post-natal day (PD) 25). To determine their role in memory deficits, two pharmacological approaches were performed during this specific period of early life. Thus, memory performance was tested after the repeated administration (from PD25 to PD34) of: i) URB597, to increase NAEs, with GW9662, a PPAR-γ antagonist; ii) pioglitazone, a PPAR-γ agonist. We observed that URB597 suppresses PLAE-induced memory deficits through a PPAR-γ dependent mechanism, since its effects are prevented by GW9662. Direct PPAR-γ activation, using pioglitazone, also ameliorates memory impairments. Lastly, to further investigate the region and cellular specificity, we demonstrate that an early overexpression of PPAR-γ, by means of a viral vector, in hippocampal astrocytes mitigates memory deficits induced by PLAE. Together, our data reveal that disruptions of PPAR-γ signaling during neurodevelopment contribute to PLAE-induced memory dysfunction. In turn, PPAR-γ activation during a childhood-like period is a promising therapeutic approach for memory deficits in the context of early alcohol exposure. Thus, these findings contribute to the gaining insight into the mechanisms that might underlie memory impairments in FASD patients.
Collapse
Affiliation(s)
- Alba Garcia-Baos
- Neurobiology of Behavior Research Group (GReNeC-NeuroBio), Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
- Neuroscience Research Program, IMIM-Hospital del Mar Research Institute, Barcelona, Spain
| | - Antoni Pastor
- Neuroscience Research Program, IMIM-Hospital del Mar Research Institute, Barcelona, Spain
| | - Ines Gallego-Landin
- Neurobiology of Behavior Research Group (GReNeC-NeuroBio), Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Rafael de la Torre
- Neurobiology of Behavior Research Group (GReNeC-NeuroBio), Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
- Neuroscience Research Program, IMIM-Hospital del Mar Research Institute, Barcelona, Spain
| | - Ferran Sanz
- Research Program on Biomedical Informatics (GRIB), IMIM-Hospital del Mar Research Institute, Universitat Pompeu Fabra, Barcelona, Spain
| | - Olga Valverde
- Neurobiology of Behavior Research Group (GReNeC-NeuroBio), Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain.
- Neuroscience Research Program, IMIM-Hospital del Mar Research Institute, Barcelona, Spain.
| |
Collapse
|
6
|
Kruk-Slomka M, Adamski B, Slomka T, Biala G. Inhibitors of Endocannabinoids' Enzymatic Degradation as a Potential Target of the Memory Disturbances in an Acute N-Methyl-D-Aspartate (NMDA) Receptor Hypofunction Model of Schizophrenia in Mice. Int J Mol Sci 2023; 24:11400. [PMID: 37511157 PMCID: PMC10380236 DOI: 10.3390/ijms241411400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 07/08/2023] [Accepted: 07/11/2023] [Indexed: 07/30/2023] Open
Abstract
Treating schizophrenia with the available pharmacotherapy is difficult. One possible strategy is focused on the modulation of the function of the endocannabinoid system (ECS). The ECS is comprised of cannabinoid (CB) receptors, endocannabinoids and enzymes responsible for the metabolism of endocannabinoids (fatty acid hydrolase (FAAH) and monoacylglycerol lipase (MAGL)). Here, the aim of the experiments was to evaluate the impact of inhibitors of endocannabinoids' enzymatic degradation in the brain: KML-29 (MAGL inhibitor), JZL-195 (MAGL/FAAH inhibitor) and PF-3845 (FAAH inhibitor), on the memory disturbances typical for schizophrenia in an acute N-methyl-D-aspartate (NMDA) receptor hypofunction animal model of schizophrenia (i.e., injection of MK-801, an NMDA receptor antagonist). The memory-like responses were assessed in the passive avoidance (PA) test. A single administration of KML-29 or PF-3845 had a positive effect on the memory processes, but an acute administration of JZL-195 impaired cognition in mice in the PA test. Additionally, the combined administration of a PA-ineffective dose of KML-29 (5 mg/kg) or PF-3845 (3 mg/kg) attenuated the MK-801-induced cognitive impairment (0.6 mg/kg). Our results suggest that the indirect regulation of endocannabinoids' concentration in the brain through the use of selected inhibitors may positively affect memory disorders, and thus increase the effectiveness of modern pharmacotherapy of schizophrenia.
Collapse
Affiliation(s)
- Marta Kruk-Slomka
- Department of Pharmacology and Pharmacodynamics, Medical University of Lublin, 4a Chodzki Street, 20-093 Lublin, Poland
| | - Bartlomiej Adamski
- Department of Pharmacology and Pharmacodynamics, Medical University of Lublin, 4a Chodzki Street, 20-093 Lublin, Poland
| | - Tomasz Slomka
- Department of Medical Informatics and Statistics with E-Health Lab, Medical University of Lublin, Jaczewskiego 4 Street, 20-954 Lublin, Poland
| | - Grazyna Biala
- Department of Pharmacology and Pharmacodynamics, Medical University of Lublin, 4a Chodzki Street, 20-093 Lublin, Poland
| |
Collapse
|
7
|
Greco R, Francavilla M, Demartini C, Zanaboni AM, Facchetti S, Palmisani M, Franco V, Tassorelli C. Activity of FAAH-Inhibitor JZP327A in an Experimental Rat Model of Migraine. Int J Mol Sci 2023; 24:10102. [PMID: 37373250 PMCID: PMC10299064 DOI: 10.3390/ijms241210102] [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: 05/21/2023] [Revised: 06/09/2023] [Accepted: 06/12/2023] [Indexed: 06/29/2023] Open
Abstract
Increased anandamide levels via fatty acid amide hydrolase (FAAH) inhibition can decrease the pronociceptive responses and inflammatory mediators in animal models of migraine. Here, we profile the pharmacological activity of the FAAH inhibitor JZP327A, a chiral 1,3,4-oxadiazol-2(3H)-one compound, in the mediation of spontaneous and nocifensive behaviour in the animal models of migraine based on nitroglycerin (NTG) administration. JZP327A (0.5 mg/kg, i.p.) or vehicle was administered to male rats 3 h after NTG (10 mg/kg, i.p.) or NTG vehicle injection. The rats were then exposed to the open field test and an orofacial formalin test 1 h later. The levels of endocannabinoids and lipid-related substances, and the expression of pain and inflammatory mediators were evaluated in cranial tissues and serum. The findings show that JZP327A did not affect NTG-induced changes in the spontaneous behaviour of rats, while it inhibited NTG-induced hyperalgesia at the orofacial formalin test. Furthermore, JZP327A dramatically decreased the gene expression of calcitonin gene-related peptide (CGRP), tumor necrosis factor alpha (TNF-alpha) and interleukin 6 (IL-6) in the trigeminal ganglia and medulla-pons, while it did not change endocannabinoids or lipids levels nor CGRP serum levels in the same tissues. These data suggest an anti-hyperalgesic role for JZP327A in the NTG model, which is mediated by the inhibition of the inflammatory cascade of events. This activity does not seem mediated by a change in the levels of endocannabinoids and lipid amides.
Collapse
Affiliation(s)
- Rosaria Greco
- Section of Translational Neurovascular Research, IRCCS Mondino Foundation, Via Mondino 2, 27100 Pavia, Italy; (M.F.); (C.D.); (A.M.Z.); (S.F.); (M.P.); (V.F.); (C.T.)
| | - Miriam Francavilla
- Section of Translational Neurovascular Research, IRCCS Mondino Foundation, Via Mondino 2, 27100 Pavia, Italy; (M.F.); (C.D.); (A.M.Z.); (S.F.); (M.P.); (V.F.); (C.T.)
- Department of Brain and Behavioral Sciences, University of Pavia, Via Bassi 21, 27100 Pavia, Italy
| | - Chiara Demartini
- Section of Translational Neurovascular Research, IRCCS Mondino Foundation, Via Mondino 2, 27100 Pavia, Italy; (M.F.); (C.D.); (A.M.Z.); (S.F.); (M.P.); (V.F.); (C.T.)
- Department of Brain and Behavioral Sciences, University of Pavia, Via Bassi 21, 27100 Pavia, Italy
| | - Anna Maria Zanaboni
- Section of Translational Neurovascular Research, IRCCS Mondino Foundation, Via Mondino 2, 27100 Pavia, Italy; (M.F.); (C.D.); (A.M.Z.); (S.F.); (M.P.); (V.F.); (C.T.)
- Department of Brain and Behavioral Sciences, University of Pavia, Via Bassi 21, 27100 Pavia, Italy
| | - Sara Facchetti
- Section of Translational Neurovascular Research, IRCCS Mondino Foundation, Via Mondino 2, 27100 Pavia, Italy; (M.F.); (C.D.); (A.M.Z.); (S.F.); (M.P.); (V.F.); (C.T.)
| | - Michela Palmisani
- Section of Translational Neurovascular Research, IRCCS Mondino Foundation, Via Mondino 2, 27100 Pavia, Italy; (M.F.); (C.D.); (A.M.Z.); (S.F.); (M.P.); (V.F.); (C.T.)
- Clinical and Experimental Pharmacology Unit, Department of Internal Medicine and Therapeutics, University of Pavia, Via Ferrata 9/A, 27100 Pavia, Italy
| | - Valentina Franco
- Section of Translational Neurovascular Research, IRCCS Mondino Foundation, Via Mondino 2, 27100 Pavia, Italy; (M.F.); (C.D.); (A.M.Z.); (S.F.); (M.P.); (V.F.); (C.T.)
- Clinical and Experimental Pharmacology Unit, Department of Internal Medicine and Therapeutics, University of Pavia, Via Ferrata 9/A, 27100 Pavia, Italy
| | - Cristina Tassorelli
- Section of Translational Neurovascular Research, IRCCS Mondino Foundation, Via Mondino 2, 27100 Pavia, Italy; (M.F.); (C.D.); (A.M.Z.); (S.F.); (M.P.); (V.F.); (C.T.)
- Department of Brain and Behavioral Sciences, University of Pavia, Via Bassi 21, 27100 Pavia, Italy
| |
Collapse
|
8
|
ElShebiney S, Elgohary R, El-Shamarka M, Mowaad N, Abulseoud OA. Natural Polyphenols-Resveratrol, Quercetin, Magnolol, and β-Catechin-Block Certain Aspects of Heroin Addiction and Modulate Striatal IL-6 and TNF-α. TOXICS 2023; 11:379. [PMID: 37112606 PMCID: PMC10145039 DOI: 10.3390/toxics11040379] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 04/04/2023] [Accepted: 04/14/2023] [Indexed: 06/19/2023]
Abstract
We have examined the effects of four different polyphenols in attenuating heroin addiction using a conditioned place preference (CPP) paradigm. Adult male Sprague Dawley rats received heroin (alternating with saline) in escalating doses starting from 10 mg/kg, i.p. up to 80 mg/kg/d for 14 consecutive days. The rats were treated with distilled water (1 mL), quercetin (50 mg/kg/d), β-catechin (100 mg/kg/d), resveratrol (30 mg/kg/d), or magnolol (50 mg/kg/d) through oral gavage for 7 consecutive days, 30 min before heroin administration, starting on day 8. Heroin withdrawal manifestations were assessed 24 h post last heroin administration following the administration of naloxone (1 mg/kg i.p). Heroin CPP reinstatement was tested following a single dose of heroin (10 mg/kg i.p.) administration. Striatal interleukin 6 (IL-6) and tumor necrosis factor alpha (TNF-α) were quantified (ELISA) after naloxone-precipitated heroin withdrawal. Compared to the vehicle, the heroin-administered rats spent significantly more time in the heroin-paired chamber (p < 0.0001). Concomitant administration of resveratrol and quercetin prevented the acquisition of heroin CPP, while resveratrol, quercetin, and magnolol blocked heroin-triggered reinstatement. Magnolol, quercetin, and β-catechin blocked naloxone-precipitated heroin withdrawal and increased striatal IL-6 concentration (p < 0.01). Resveratrol administration was associated with significantly higher withdrawal scores compared to those of the control animals (p < 0.0001). The results of this study show that different polyphenols target specific behavioral domains of heroin addiction in a CPP model and modulate the increase in striatal inflammatory cytokines TNF-α and IL-6 observed during naloxone-precipitated heroin withdrawal. Further research is needed to study the clinical utility of polyphenols and to investigate the intriguing finding that resveratrol enhances, rather than attenuates naloxone-precipitated heroin withdrawal.
Collapse
Affiliation(s)
- Shaimaa ElShebiney
- Department of Narcotics, Ergogenics, and Poisons, National Research Centre, Dokki, Cairo 12622, Egypt
| | - Rania Elgohary
- Department of Narcotics, Ergogenics, and Poisons, National Research Centre, Dokki, Cairo 12622, Egypt
| | - Marwa El-Shamarka
- Department of Narcotics, Ergogenics, and Poisons, National Research Centre, Dokki, Cairo 12622, Egypt
| | - Noha Mowaad
- Department of Narcotics, Ergogenics, and Poisons, National Research Centre, Dokki, Cairo 12622, Egypt
| | - Osama A. Abulseoud
- Department of Psychiatry and Psychology, Mayo Clinic, Phoenix, AZ 85001, USA
- Department of Neuroscience, Graduate School of Biomedical Sciences, Mayo Clinic College of Medicine, Phoenix, AZ 85001, USA
| |
Collapse
|
9
|
Gomes AKC, Alves Soares M, Miranda ALP, Tributino JLM, Goetze Fiorot R, Kuster RM, Gomes ACC, Simas NK. Flavonoids and fractions from Saccharum officinarum L. juice: antinociceptive agents and molecular docking evaluations with µ-opioid receptor. Nat Prod Res 2023; 37:592-597. [PMID: 35422173 DOI: 10.1080/14786419.2022.2063854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Opioid receptors mediate antinociceptive effects. Methanolic fractions from sugarcane varieties (MFSCf) were evaluated in classic nociception models. Interactions between bioactive compounds and the µ-opioid receptor (µOR) through docking analysis were also studied. Five methanolic fractions of sugarcane juice were obtained and analysed by LC-ESI-MS/MS. The fractions and standards of phenolic compounds were evaluated in a nociception model using the formalin test. All MFSCfs exhibited antinociceptive activity in the first phase of the formalin test. Docking analyses corroborates with the in vivo test results, suggesting that the phenolic substances are able to activate µOR. These results, for the first time, implicate phenolic constituents from sugarcane juice and other phenolic compounds in the activation of µOR. The antinociceptive activity of fractions from sugarcane juice suggests the potential pharmacological use of this species, widely cultivated in Brazil.
Collapse
Affiliation(s)
- Anne Katherine C Gomes
- Programa de Pós-graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Mariana Alves Soares
- Instituto de Ciências Biomédicas, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Ana Luísa P Miranda
- Programa de Pós-graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | | | - Rodolfo Goetze Fiorot
- Departmento de Química Orgânica, Instituto de Química, Fluminense Federal University (UFF), Niterói, RJ, Brazil
| | | | - Anne Caroline C Gomes
- Instituto Federal de Educação, Ciência e Tecnologia do Rio de Janeiro (IFRJ), Rio de Janeiro, Brazil
| | - Naomi Kato Simas
- Programa de Pós-graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| |
Collapse
|
10
|
Fateh ST, Salehi-Najafabadi A. Repurposing of substances with lactone moiety for the treatment of γ-Hydroxybutyric acid and γ-Butyrolactone intoxication through modulating paraoxonase and PPARγ. Front Pharmacol 2022; 13:909460. [PMID: 35935832 PMCID: PMC9354891 DOI: 10.3389/fphar.2022.909460] [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: 04/01/2022] [Accepted: 06/30/2022] [Indexed: 11/13/2022] Open
Abstract
GHB and GBL are highly accessible recreational drugs of abuse with a high risk of adverse effects and mortality while no specific antidotes exist. These components can also be found in the clinical setting, beverages, and cosmetic products, leading to unwanted exposures and further intoxications. As the structural analogue of GABA, GHB is suggested as the primary mediator of GHB/GBL effects. We further suggest that GBL might be as critical as GHB in this process, acting through PPARγ as its receptor. Moreover, PPARγ and PON (i.e., the GHB-GBL converting enzyme) can be targeted for GHB/GBL addiction and intoxication, leading to modulation of the GHB-GBL balance and blockage of their effects. We suggest that repurposing substances with lactone moiety such as bacterial lactones, sesquiterpene lactones, and statins might lead to potential therapeutic options as they occupy the active sites of PPARγ and PON and interfere with the GHB-GBL balance. In conclusion, this hypothesis improves the GHB/GBL mechanism of action, suggests potential therapeutic options, and highlights the necessity of classifying GBL as a controlled substance.
Collapse
Affiliation(s)
- Sepand Tehrani Fateh
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amir Salehi-Najafabadi
- Department of Microbiology, School of Biology, University College of Science, University of Tehran, Tehran, Iran
- *Correspondence: Amir Salehi-Najafabadi,
| |
Collapse
|
11
|
Greco R, Demartini C, Zanaboni AM, Francavilla M, Reggiani A, Realini N, Scarpelli R, Piomelli D, Tassorelli C. Potentiation of endocannabinoids and other lipid amides prevents hyperalgesia and inflammation in a pre-clinical model of migraine. J Headache Pain 2022; 23:79. [PMID: 35799128 PMCID: PMC9264488 DOI: 10.1186/s10194-022-01449-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 06/22/2022] [Indexed: 11/10/2022] Open
Abstract
Targeting fatty acid amide hydrolase (FAAH) is a promising therapeutic strategy to combat certain forms of pain, including migraine headache. FAAH inhibitors, such as the O-biphenyl-3-yl carbamate URB597, have been shown to produce anti-hyperalgesic effects in animal models of migraine. The objective of this study was to investigate the behavioral and biochemical effects of compounds ARN14633 and ARN14280, two URB597 analogs with improved solubility and bioavailability, in a migraine-specific rat model in which trigeminal hyperalgesia is induced by nitroglycerin (NTG) administration. ARN14633 (1 mg/kg, i.p.) and ARN14280 (3 mg/kg, i.p.) were administered to adult male Sprague-Dawley rats 3 hours after NTG injection. One hour after the administration of either compound, rats were subjected to the orofacial formalin test. ARN14633 and ARN14280 attenuated NTG-induced nocifensive behavior and reduced transcription of genes encoding neuronal nitric oxide synthase, pain mediators peptides (calcitonin gene-related peptide, substance P) and pro-inflammatory cytokines (tumor necrosis factor-alpha, interleukin-1beta and 6) in the trigeminal ganglion, cervical spinal cord and medulla. Finally, both compounds strongly elevated levels of endocannabinoids and/or other FAAH substrates in cervical spinal cord and medulla, and, to a lesser extent, in the trigeminal ganglia. The results indicate that the novel global FAAH inhibitors ARN14633 and ARN14280 elicit significant anti-hyperalgesic effects in a migraine-specific animal model and inhibit the associated peptidergic-inflammatory response. Although the precise mechanism underlying these effects remains to be elucidated, our results support further investigational studies of FAAH blockade as a potential therapeutic strategy to treat migraine conditions.
Collapse
Affiliation(s)
- Rosaria Greco
- Unit of Translational Neurovascular Research, IRCCS Mondino Foundation, Pavia, Italy.
| | - Chiara Demartini
- Unit of Translational Neurovascular Research, IRCCS Mondino Foundation, Pavia, Italy
| | - Anna Maria Zanaboni
- Unit of Translational Neurovascular Research, IRCCS Mondino Foundation, Pavia, Italy.,Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Miriam Francavilla
- Unit of Translational Neurovascular Research, IRCCS Mondino Foundation, Pavia, Italy
| | - Angelo Reggiani
- Drug Discovery and Development (D3)-Validation, Fondazione Istituto Italiano di Tecnologia, Genoa, Italy
| | - Natalia Realini
- Drug Discovery and Development (D3)-Validation, Fondazione Istituto Italiano di Tecnologia, Genoa, Italy
| | - Rita Scarpelli
- Drug Discovery and Development (D3)-Validation, Fondazione Istituto Italiano di Tecnologia, Genoa, Italy
| | - Daniele Piomelli
- Department of Anatomy and Neurobiology, University of California, Irvine, CA, USA
| | - Cristina Tassorelli
- Unit of Translational Neurovascular Research, IRCCS Mondino Foundation, Pavia, Italy.,Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| |
Collapse
|
12
|
Lunerti V, Li H, Benvenuti F, Shen Q, Domi A, Soverchia L, Concetta Di Martino RM, Bottegoni G, Haass-Koffler CL, Cannella N. The multitarget FAAH inhibitor/D3 partial agonist ARN15381 decreases nicotine self-administration in male rats. Eur J Pharmacol 2022; 928:175088. [PMID: 35690082 DOI: 10.1016/j.ejphar.2022.175088] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 05/18/2022] [Accepted: 06/03/2022] [Indexed: 11/03/2022]
Abstract
Tobacco use disorder is a worldwide health problem for which available medications show limited efficacy. Nicotine is the psychoactive component of tobacco responsible for its addictive liability. Similar to other addictive drugs, nicotine enhances mesolimbic dopamine transmission. Inhibition of the fatty acid amide hydrolase (FAAH), the enzyme responsible for the degradation of the endocannabinoid anandamide (AEA), palmitoylethanolamide (PEA) and oleoylethanolamide (OEA), reduces nicotine-enhanced dopamine transmission and acquisition of nicotine self-administration in rats. Down-regulation of dopamine transmission by antagonists or partial agonists of the dopamine D3 receptor (DRD3) also reduced nicotine self-administration and conditioned place preference. Based on these premises, we evaluated the effect of ARN15381, a multitarget compound showing FAAH inhibition and DRD3 partial agonist activity in the low nanomolar range, on nicotine self-administration in rats. Pretreatment with ARN15381 dose dependently decreased self-administration of a nicotine dose at the top of the nicotine dose/response (D/R) curve, while it did not affect self-administration of a nicotine dose laying on the descending limb of the D/R curve. Conversely, pretreatment with the selective FAAH inhibitor URB597 and the DRD3 partial agonist CJB090 failed to modify nicotine self-administration independent of the nicotine dose self-administered. Our data indicates that the concomitant FAAH inhibition and DRD3 partial agonism produced by ARN15381 is key to the observed reduction of nicotine self-administration, demonstrating that a multitarget approach may hold clinical importance for the treatment of tobacco use disorder.
Collapse
Affiliation(s)
- Veronica Lunerti
- School of Pharmacy, Pharmacology Unit, University of Camerino, Italy
| | - Hongwu Li
- School of Pharmacy, Pharmacology Unit, University of Camerino, Italy; School of Chemical Engineering, Changchun University of Changchung, 130012, China
| | | | - Qianwei Shen
- School of Pharmacy, Pharmacology Unit, University of Camerino, Italy
| | - Ana Domi
- School of Pharmacy, Pharmacology Unit, University of Camerino, Italy
| | - Laura Soverchia
- School of Pharmacy, Pharmacology Unit, University of Camerino, Italy
| | | | - Giovanni Bottegoni
- School of Pharmacy, University of Birmingham, Edgbaston, B15 2TT, Birmingham, United Kingdom; Dipartimento di Scienze Biomolecolari, Università degli Studi di Urbino "Carlo Bo", Urbino, Italy
| | - Carolina L Haass-Koffler
- Center for Alcohol and Addiction Studies, Department of Psychiatry and Human Behavior, Warren Alpert Medical School, Department of Behavioral and Social Sciences, School of Public Health, Carney Institute for Brain Science, Brown University, USA
| | | |
Collapse
|
13
|
Leuci R, Brunetti L, Laghezza A, Piemontese L, Carrieri A, Pisani L, Tortorella P, Catto M, Loiodice F. A New Series of Aryloxyacetic Acids Endowed with Multi-Target Activity towards Peroxisome Proliferator-Activated Receptors (PPARs), Fatty Acid Amide Hydrolase (FAAH), and Acetylcholinesterase (AChE). Molecules 2022; 27:molecules27030958. [PMID: 35164223 PMCID: PMC8839882 DOI: 10.3390/molecules27030958] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/20/2022] [Accepted: 01/27/2022] [Indexed: 02/04/2023] Open
Abstract
A new series of aryloxyacetic acids was prepared and tested as peroxisome proliferator-activated receptors (PPARs) agonists and fatty acid amide hydrolase (FAAH) inhibitors. Some compounds exhibited an interesting dual activity that has been recently proposed as a new potential therapeutic strategy for the treatment of Alzheimer’s disease (AD). AD is a multifactorial pathology, hence multi-target agents are currently one of the main lines of research for the therapy and prevention of this disease. Given that cholinesterases represent one of the most common targets of recent research, we decided to also evaluate the effects of our compounds on the inhibition of these specific enzymes. Interestingly, two of these compounds, (S)-5 and 6, showed moderate activity against acetylcholinesterase (AChE) and even some activity, although at high concentration, against Aβ peptide aggregation, thus demonstrating, in agreement with the preliminary dockings carried out on the different targets, the feasibility of a simultaneous multi-target activity towards PPARs, FAAH, and AChE. As far as we know, these are the first examples of molecules endowed with this pharmacological profile that might represent a promising line of research for the identification of novel candidates for the treatment of AD.
Collapse
|
14
|
Papa A, Pasquini S, Contri C, Gemma S, Campiani G, Butini S, Varani K, Vincenzi F. Polypharmacological Approaches for CNS Diseases: Focus on Endocannabinoid Degradation Inhibition. Cells 2022; 11:cells11030471. [PMID: 35159280 PMCID: PMC8834510 DOI: 10.3390/cells11030471] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/21/2022] [Accepted: 01/27/2022] [Indexed: 01/27/2023] Open
Abstract
Polypharmacology breaks up the classical paradigm of “one-drug, one target, one disease” electing multitarget compounds as potential therapeutic tools suitable for the treatment of complex diseases, such as metabolic syndrome, psychiatric or degenerative central nervous system (CNS) disorders, and cancer. These diseases often require a combination therapy which may result in positive but also negative synergistic effects. The endocannabinoid system (ECS) is emerging as a particularly attractive therapeutic target in CNS disorders and neurodegenerative diseases including Parkinson’s disease (PD), Alzheimer’s disease (AD), Huntington’s disease (HD), multiple sclerosis (MS), amyotrophic lateral sclerosis (ALS), stroke, traumatic brain injury (TBI), pain, and epilepsy. ECS is an organized neuromodulatory network, composed by endogenous cannabinoids, cannabinoid receptors type 1 and type 2 (CB1 and CB2), and the main catabolic enzymes involved in the endocannabinoid inactivation such as fatty acid amide hydrolase (FAAH) and monoacylglycerol lipase (MAGL). The multiple connections of the ECS with other signaling pathways in the CNS allows the consideration of the ECS as an optimal source of inspiration in the development of innovative polypharmacological compounds. In this review, we focused our attention on the reported polypharmacological examples in which FAAH and MAGL inhibitors are involved.
Collapse
Affiliation(s)
- Alessandro Papa
- Department of Biotechnology, Chemistry and Pharmacy, DoE Department of Excellence 2018-2022, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy; (A.P.); (S.G.); (G.C.)
| | - Silvia Pasquini
- Department of Translational Medicine, University of Ferrara, Via Fossato di Mortara 17-19, 44121 Ferrara, Italy; (S.P.); (C.C.); (K.V.); (F.V.)
| | - Chiara Contri
- Department of Translational Medicine, University of Ferrara, Via Fossato di Mortara 17-19, 44121 Ferrara, Italy; (S.P.); (C.C.); (K.V.); (F.V.)
| | - Sandra Gemma
- Department of Biotechnology, Chemistry and Pharmacy, DoE Department of Excellence 2018-2022, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy; (A.P.); (S.G.); (G.C.)
| | - Giuseppe Campiani
- Department of Biotechnology, Chemistry and Pharmacy, DoE Department of Excellence 2018-2022, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy; (A.P.); (S.G.); (G.C.)
| | - Stefania Butini
- Department of Biotechnology, Chemistry and Pharmacy, DoE Department of Excellence 2018-2022, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy; (A.P.); (S.G.); (G.C.)
- Correspondence: ; Tel.: +39-0577-234161
| | - Katia Varani
- Department of Translational Medicine, University of Ferrara, Via Fossato di Mortara 17-19, 44121 Ferrara, Italy; (S.P.); (C.C.); (K.V.); (F.V.)
| | - Fabrizio Vincenzi
- Department of Translational Medicine, University of Ferrara, Via Fossato di Mortara 17-19, 44121 Ferrara, Italy; (S.P.); (C.C.); (K.V.); (F.V.)
| |
Collapse
|
15
|
Yunusoğlu O. Evaluation of the effects of quercetin on the rewarding property of ethanol in mice. Neurosci Lett 2022; 768:136383. [PMID: 34864087 DOI: 10.1016/j.neulet.2021.136383] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 11/26/2021] [Accepted: 11/29/2021] [Indexed: 01/27/2023]
Abstract
BACKGROUND The flavonoid quercetin has several pharmacological effects on the nervous system. Previous research showed that quercetin has useful influences on some mechanisms that are relevant in drug and substance addiction. Alcohol addiction, also known as alcoholism, is a disorder that influences the population in all walks of life. The purpose of the current study was to investigate whether quercetin affects the acquisition, extinction, and reinstatement of ethanol-induced conditioned place preference (ethanol-CPP) in adolescent mice. METHODS CPP was established by administration of intraperitoneal (i.p.) ethanol (2.0 g/kg) in a conditioning trial. The mice were pretreated with quercetin (at doses of 10, 30, and 100 mg/kg, i.p.) 30 minutes before each ethanol injection to test the effects of quercetin on the reward properties of ethanol. Ethanol-CPP was extinguished (13-days) by repeated testing, during which conditioned mice were given different doses of quercetin every day. Lastly, efficacy of quercetin in preventing reinstatement of ethanol-CPP triggers was also assessed by the administration of single dose ethanol (0.4 g/kg, i.p.). RESULTS Quercetin pretreatment attenuated the acquisition and reinstatement. In addition, quercetin administration accelerated the extinction of ethanol-CPP. CONCLUSIONS In conclusion, these results may cast a novel light on quercetin as an agent that could be potentially useful to attenuate different effects of ethanol and as adjuvant pharmacotherapy for ethanol addiction. However, future studies are needed to demonstrate the detailed underlying mechanisms of quercetin on ethanol addiction.
Collapse
Affiliation(s)
- Oruç Yunusoğlu
- Bolu Abant Izzet Baysal University, Faculty of Medicine, Medical Pharmacology, 14030 Bolu, Turkey.
| |
Collapse
|
16
|
Yunusoğlu O. Linalool attenuates acquisition and reinstatement and accelerates the extinction of nicotine-induced conditioned place preference in male mice. THE AMERICAN JOURNAL OF DRUG AND ALCOHOL ABUSE 2021; 47:422-432. [PMID: 33852814 DOI: 10.1080/00952990.2021.1898627] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Background: Nicotine is the addictive agent in tobacco products. The monoterpene linalool is the main ingredient in the essential oils of various aromatic plants. It has previously been demonstrated that linalool has beneficial effects on some mechanisms that are important in drug addiction.Objectives: The goal of the current study was to investigate the effect of linalool on nicotine-induced conditioned place preference (CPP) in male mice.Methods: CPP was induced by administering intraperitoneal (i.p.) injection of nicotine (0.5 mg/kg) during the conditioning phase. The effects of nicotinic acetylcholine receptor partial agonist varenicline and linalool on the rewarding characteristics of nicotine were tested in mice with administration of linalool (12.5, 25, and 50 mg/kg, i.p.), varenicline (2 mg/kg, i.p.) or saline 30 minutes before nicotine injection. CPP was extinguished by repeated testing, during which conditioned mice were administered varenicline and linalool every day. One day after the last extinction trial, mice that received linalool, varenicline or saline 30 minutes before a priming injection of nicotine (0.1 mg/kg, i.p.) were immediately tested for reinstatement of CPP.Results: Linalool attenuated nicotine acquisition (50 mg/kg, p < .01) and reinstatement (25 and 50 mg/kg, respectively p < .05, p < .01) and accelerated the extinction of nicotine-induced CPP (50 mg/kg, p < .05). Linalool exhibited similar effects on the reference drug varenicline in the CPP phases.Conclusion: These results suggest that linalool may be helpful as an adjuvant for the treatment of nicotine use disorder.
Collapse
Affiliation(s)
- Oruç Yunusoğlu
- Department of Pharmacology, Faculty of Medicine, Van Yuzuncu Yil University, Van, Turkey
| |
Collapse
|
17
|
Potential and Limits of Cannabinoids in Alzheimer's Disease Therapy. BIOLOGY 2021; 10:biology10060542. [PMID: 34204237 PMCID: PMC8234911 DOI: 10.3390/biology10060542] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 06/13/2021] [Accepted: 06/14/2021] [Indexed: 11/16/2022]
Abstract
Simple Summary This review was aimed at exploring the potentiality of drugging the endocannabinoid system as a therapeutic option for Alzheimer’s disease (AD). Recent discoveries have demonstrated how the modulation of cannabinoid receptor 1 (CB1) and receptor 2 (CB2) can exert neuroprotective effects without the recreational and pharmacological properties of Cannabis sativa. Thus, this review explores the potential of cannabinoids in AD, also highlighting their limitations in perspective to point out the need for further research on cannabinoids in AD therapy. Abstract Alzheimer’s disease (AD) is a detrimental brain disorder characterized by a gradual cognitive decline and neuronal deterioration. To date, the treatments available are effective only in the early stage of the disease. The AD etiology has not been completely revealed, and investigating new pathological mechanisms is essential for developing effective and safe drugs. The recreational and pharmacological properties of marijuana are known for centuries, but only recently the scientific community started to investigate the potential use of cannabinoids in AD therapy—sometimes with contradictory outcomes. Since the endocannabinoid system (ECS) is highly expressed in the hippocampus and cortex, cannabis use/abuse has often been associated with memory and learning dysfunction in vulnerable individuals. However, the latest findings in AD rodent models have shown promising effects of cannabinoids in reducing amyloid plaque deposition and stimulating hippocampal neurogenesis. Beneficial effects on several dementia-related symptoms have also been reported in clinical trials after cannabinoid treatments. Accordingly, future studies should address identifying the correct therapeutic dosage and timing of treatment from the perspective of using cannabinoids in AD therapy. The present paper aims to summarize the potential and limitations of cannabinoids as therapeutics for AD, focusing on recent pre-clinical and clinical evidence.
Collapse
|
18
|
Malamas MS, Pavlopoulos S, Alapafuja SO, Farah SI, Zvonok A, Mohammad KA, West J, Perry NT, Pelekoudas DN, Rajarshi G, Shields C, Chandrashekhar H, Wood J, Makriyannis A. Design and Structure-Activity Relationships of Isothiocyanates as Potent and Selective N-Acylethanolamine-Hydrolyzing Acid Amidase Inhibitors. J Med Chem 2021; 64:5956-5972. [PMID: 33900772 DOI: 10.1021/acs.jmedchem.1c00076] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
N-Acylethanolamines are signaling lipid molecules implicated in pathophysiological conditions associated with inflammation and pain. N-Acylethanolamine acid amidase (NAAA) favorably hydrolyzes lipid palmitoylethanolamide, which plays a key role in the regulation of inflammatory and pain processes. The synthesis and structure-activity relationship studies encompassing the isothiocyanate pharmacophore have produced potent low nanomolar inhibitors for hNAAA, while exhibiting high selectivity (>100-fold) against other serine hydrolases and cysteine peptidases. We have followed a target-based structure-activity relationship approach, supported by computational methods and known cocrystals of hNAAA. We have identified systemically active inhibitors with good plasma stability (t1/2 > 2 h) and microsomal stability (t1/2 ∼ 15-30 min) as pharmacological tools to investigate the role of NAAA in inflammation, pain, and drug addiction.
Collapse
Affiliation(s)
| | - Spiro Pavlopoulos
- Center for Drug Discovery, Northeastern University, Boston, Massachusetts 02115, United States
| | - Shakiru O Alapafuja
- MAK Scientific LLC, 151 South Bedford Street, Burlington, Massachusetts 01803, United States
| | - Shrouq I Farah
- Center for Drug Discovery, Northeastern University, Boston, Massachusetts 02115, United States
| | - Alexander Zvonok
- Center for Drug Discovery, Northeastern University, Boston, Massachusetts 02115, United States
| | - Khadijah A Mohammad
- Center for Drug Discovery, Northeastern University, Boston, Massachusetts 02115, United States
| | - Jay West
- Center for Drug Discovery, Northeastern University, Boston, Massachusetts 02115, United States
| | - Nicholas Thomas Perry
- Center for Drug Discovery, Northeastern University, Boston, Massachusetts 02115, United States
| | - Dimitrios N Pelekoudas
- Center for Drug Discovery, Northeastern University, Boston, Massachusetts 02115, United States
| | - Girija Rajarshi
- Center for Drug Discovery, Northeastern University, Boston, Massachusetts 02115, United States
| | - Christina Shields
- Center for Drug Discovery, Northeastern University, Boston, Massachusetts 02115, United States
| | - Honrao Chandrashekhar
- Center for Drug Discovery, Northeastern University, Boston, Massachusetts 02115, United States
| | - Jodi Wood
- Center for Drug Discovery, Northeastern University, Boston, Massachusetts 02115, United States
| | - Alexandros Makriyannis
- Center for Drug Discovery and Departments of Chemistry and Chemical Biology and Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts 02115, United States
| |
Collapse
|
19
|
Niemela G, Terry GE. Contribution of Fatty Acid Amide Hydrolase to Alcohol Use Disorder: A Systematic Review. Cannabis Cannabinoid Res 2021; 6:105-118. [PMID: 33989054 DOI: 10.1089/can.2020.0158] [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] [Indexed: 12/25/2022] Open
Abstract
Purpose: Recent research has suggested that chronic alcohol exposure induces changes in the endocannabinoid system within the central nervous system and therefore could be an attractive target for better understanding and treating alcohol use disorder (AUD). Much of this research has centered around the CB1 receptor and its endogenous partial agonist, the endocannabinoid anandamide, as the CB1 receptor is densely expressed in brain regions involved in development and maintenance of addictive behaviors. In addition, recent evidence has suggested that chronic alcohol exposure induces changes in the modulation of endocannabinoid concentration and suggests that these changes may contribute to the motivation to abuse alcohol. Therefore, we performed a systematic literature review to evaluate how fatty acid amide hydrolase (FAAH), an enzyme that degrades anandamide, relates to the characteristics and biology of AUD, as well as how modulating FAAH through pharmacologic inhibition or genetic manipulation affects outcomes related to alcohol use and consumption. Method: A search strategy was developed using the terms "endocannabinoids" or "drug delivery systems" and "alcohol dependence" or "alcohol use disorder" or "alcoholism" and "Fatty Acid Amide Hydrolase" and "FAAH" as text words and Medical Subject Headings (i.e., MeSH and EMTREE). We then used this search strategy on the electronic databases PubMed, Embase, and Web of Science. Results: We found 224 records; after removing repeated records (37%), articles that did not fit the topic question (47%), or were not primary research (4%), we included 26 for qualitative synthesis (12%). Discussion: The literature clearly suggests that FAAH has a role in the biology and characteristics of AUD. FAAH inhibition seems especially promising as a target for alcohol withdrawal as it may lead to a reduction in symptoms, including anxiety and a reduction of alcohol intake reinstatement. However, decreased FAAH may also lead to reduced sensitivity to alcohol along with increased preference and intake. Conclusions: Modulation of FAAH is promising for therapeutic intervention of AUD, but requires more research. Pre-clinical studies have indicated that FAAH inhibition may reduce withdrawal characteristics, but may also exacerbate other characteristics of AUD outside of that period.
Collapse
Affiliation(s)
- Greta Niemela
- University of Washington School of Medicine, Seattle, Washington, USA
| | - Garth E Terry
- Department of Psychiatry & Behavioral Sciences, and Radiology, University of Washington School of Medicine, Seattle, Washington, USA.,Department of Veterans Affairs, Puget Sound Health Care System, Mental Illness Research, Education, and Clinical Center (MIRECC), Seattle, Washington, USA
| |
Collapse
|
20
|
Cannabinoid-Induced Conditioned Place Preference, Intravenous Self-Administration, and Behavioral Stimulation Influenced by Ghrelin Receptor Antagonism in Rats. Int J Mol Sci 2021; 22:ijms22052397. [PMID: 33673659 PMCID: PMC7957642 DOI: 10.3390/ijms22052397] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 02/13/2021] [Accepted: 02/21/2021] [Indexed: 11/24/2022] Open
Abstract
Cannabis/cannabinoids are widely used for recreational and therapy purposes, but their risks are largely disregarded. However, cannabinoid-associated use disorders and dependence are alarmingly increasing and an effective treatment is lacking. Recently, the growth hormone secretagogue receptor (GHSR1A) antagonism was proposed as a promising mechanism for drug addiction therapy. However, the role of GHS-R1A and its endogenous ligand ghrelin in cannabinoid abuse remains unclear. Therefore, the aim of our study was to investigate whether the GHS-R1A antagonist JMV2959 could reduce the tetrahydrocannabinol (THC)-induced conditioned place preference (CPP) and behavioral stimulation, the WIN55,212-2 intravenous self-administration (IVSA), and the tendency to relapse. Following an ongoing WIN55,212-2 self-administration, JMV2959 3 mg/kg was administered intraperitoneally 20 min before three consequent daily 120-min IVSA sessions under a fixed ratio FR1, which significantly reduced the number of the active lever-pressing, the number of infusions, and the cannabinoid intake. Pretreatment with JMV2959 suggested reduction of the WIN55,212-2-seeking/relapse-like behavior tested in rats on the twelfth day of the forced abstinence period. On the contrary, pretreatment with ghrelin significantly increased the cannabinoid IVSA as well as enhanced the relapse-like behavior. Co-administration of ghrelin with JMV2959 abolished/reduced the significant efficacy of the GHS-R1A antagonist in the cannabinoid IVSA. Pretreatment with JMV2959 significantly and dose-dependently reduced the manifestation of THC-induced CPP. The THC-CPP development was reduced after the simultaneous administration of JMV2959 with THC during conditioning. JMV2959 also significantly reduced the THC-induced behavioral stimulation in the LABORAS cage. Our findings suggest that GHS-R1A importantly participates in the rewarding/reinforcing effects of cannabinoids.
Collapse
|
21
|
Asadi Akbarabadi E, Rajabi Vardanjani H, Molavinia S, Pashmforoosh M, Khodayar MJ. PMSF Attenuates Morphine Antinociceptive Tolerance and Dependence in Mice: Its Association with the Oxidative Stress Suppression. IRANIAN JOURNAL OF PHARMACEUTICAL RESEARCH : IJPR 2021; 20:300-309. [PMID: 34903990 PMCID: PMC8653646 DOI: 10.22037/ijpr.2020.112936.14038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Opioids use has been limited due to tolerance and dependence as major unwanted effects. Previous evidence has shown that targeting endocannabinoid signaling can prevent the development of opioid tolerance and dependence. This study was designed to evaluate the effect of phenylmethylsulfonyl fluoride (PMSF), an inhibitor of fatty acid amide hydrolase (FAAH), on morphine antinociceptive tolerance and physical dependence in mice. The antinociceptive effects of PMSF at the doses 60, 120, and 300 mg/kg were investigated. Results showed that PMSF has a notable antinociceptive effect at doses 120 and 300 mg/kg. The dose of (60 mg/kg, i.p.) PMSF was considered as a sub-antinociceptive dose. Morphine tolerance and dependence were induced by twice-daily injection of morphine (10 mg/kg, s.c.) for 10 consecutive days and the last dose on day 11. Tolerance was assessed by the hot-plate test and dependence by naloxone-precipitated morphine withdrawal signs. In the brain, oxidative stress markers include activities of glutathione peroxidase, catalase, superoxide dismutase, and levels of malondialdehyde and glutathione were determined. A sub-antinociceptive dose (60 mg/kg) of PMSF could reduce tolerance in both acute and chronic methods of administration. However, alleviation of dependence and suppression of oxidative stress markers occurred in the chronic administration of PMSF. In conclusion, it seems that PMSF can suppress morphine tolerance and dependence. However, more studies are needed to clarify its mechanism.
Collapse
Affiliation(s)
- Ehsan Asadi Akbarabadi
- Student Research Committee, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| | - Hossein Rajabi Vardanjani
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran.
| | - Shahrzad Molavinia
- Toxicology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran. ,Department of Toxicology, School of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| | | | - Mohammad Javad Khodayar
- Toxicology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran. ,Department of Toxicology, School of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran. ,Corresponding author: E-mail:
| |
Collapse
|
22
|
Charalambous C, Lapka M, Havlickova T, Syslova K, Sustkova-Fiserova M. Alterations in Rat Accumbens Dopamine, Endocannabinoids and GABA Content During WIN55,212-2 Treatment: The Role of Ghrelin. Int J Mol Sci 2020; 22:ijms22010210. [PMID: 33379212 PMCID: PMC7795825 DOI: 10.3390/ijms22010210] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 12/21/2020] [Accepted: 12/24/2020] [Indexed: 01/22/2023] Open
Abstract
The endocannabinoid/CB1R system as well as the central ghrelin signalling with its growth hormone secretagogoue receptors (GHS-R1A) are importantly involved in food intake and reward/reinforcement processing and show distinct overlaps in distribution within the relevant brain regions including the hypothalamus (food intake), the ventral tegmental area (VTA) and the nucleus accumbens (NAC) (reward/reinforcement). The significant mutual interaction between these systems in food intake has been documented; however, the possible role of ghrelin/GHS-R1A in the cannabinoid reinforcement effects and addiction remain unclear. Therefore, the principal aim of the present study was to investigate whether pretreatment with GHS-R1A antagonist/JMV2959 could reduce the CB1R agonist/WIN55,212-2–induced dopamine efflux in the nucleus accumbens shell (NACSh), which is considered a crucial trigger impulse of the addiction process. The synthetic aminoalklylindol cannabinoid WIN55,212-2 administration into the posterior VTA induced significant accumbens dopamine release, which was significantly reduced by the 3 mg/kg i.p. JMV2959 pretreatment. Simultaneously, the cannabinoid-increased accumbens dopamine metabolic turnover was significantly augmented by the JMV2959 pretreament. The intracerebral WIN55,212-2 administration also increased the endocannabinoid arachidonoylethanolamide/anandamide and the 2-arachidonoylglycerol/2-AG extracellular levels in the NACSh, which was moderately but significantly attenuated by the JMV2959 pretreatment. Moreover, the cannabinoid-induced decrease in accumbens γ-aminobutyric acid/gamma-aminobutyric acid levels was reversed by the JMV2959 pretreatment. The behavioural study in the LABORAS cage showed that 3 mg/kg JMV2959 pretreatment also significantly reduced the systemic WIN55,212-2-induced behavioural stimulation. Our results demonstrate that the ghrelin/GHS-R1A system significantly participates in the rewarding/reinforcing effects of the cannabinoid/CB1 agonist that are involved in cannabinoid addiction processing.
Collapse
Affiliation(s)
- Chrysostomos Charalambous
- Department of Addictology, First Faculty of Medicine, Charles University, Apolinarska 4, 128 00 Prague 2, Czech Republic;
| | - Marek Lapka
- Department of Pharmacology, Third Faculty of Medicine, Charles University, Ruska 87, 100 34 Prague 10, Czech Republic; (M.L.); (T.H.)
| | - Tereza Havlickova
- Department of Pharmacology, Third Faculty of Medicine, Charles University, Ruska 87, 100 34 Prague 10, Czech Republic; (M.L.); (T.H.)
| | - Kamila Syslova
- Laboratory of Medicinal Diagnostics, Department of Organic Technology, University of Chemistry and Technology Prague, Technicka 5, 166 28 Prague 6, Czech Republic;
| | - Magdalena Sustkova-Fiserova
- Department of Pharmacology, Third Faculty of Medicine, Charles University, Ruska 87, 100 34 Prague 10, Czech Republic; (M.L.); (T.H.)
- Correspondence: ; Tel.: +420-267-102-450; Fax: +420-267-102-461
| |
Collapse
|
23
|
Greco R, Demartini C, Zanaboni A, Casini I, De Icco R, Reggiani A, Misto A, Piomelli D, Tassorelli C. Characterization of the peripheral FAAH inhibitor, URB937, in animal models of acute and chronic migraine. Neurobiol Dis 2020; 147:105157. [PMID: 33129939 DOI: 10.1016/j.nbd.2020.105157] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 10/14/2020] [Accepted: 10/27/2020] [Indexed: 12/27/2022] Open
Abstract
Inhibiting the activity of fatty-acid amide hydrolase (FAAH), the enzyme that deactivates the endocannabinoid anandamide, enhances anandamide-mediated signaling and holds promise as a molecular target for the treatment of human pathologies such as anxiety and pain. We have previously shown that the peripherally restricted FAAH inhibitor, URB937, prevents nitroglycerin-induced hyperalgesia - an animal model of migraine - and attenuates the activation of brain areas that are relevant for migraine pain, e.g. trigeminal nucleus caudalis and locus coeruleus. The current study is aimed at profiling the behavioral and biochemical effects of URB937 in animal models of acute and chronic migraine. We evaluated the effects of URB937 in two rat models that capture aspects of acute and chronic migraine, and are based on single or repeated administration of the vasodilating drug, nitroglycerin (NTG). In addition to nocifensive behavior, in trigeminal ganglia and medulla, we measured mRNA levels of neuropeptides and pro-inflammatory cytokines along with tissue levels of anandamide and palmitoylethanolamide (PEA), an endogenous agonist of peroxisome proliferator-activated receptor type-a (PPAR-a), which is also a FAAH substrate. In the acute migraine model, we also investigated the effect of subtype-selective antagonist for cannabinoid receptors 1 and 2 (AM251 and AM630, respectively) on nocifensive behavior and on levels of neuropeptides and pro-inflammatory cytokines. In the acute migraine paradigm, URB937 significantly reduced hyperalgesia in the orofacial formalin test when administered either before or after NTG. This effect was accompanied by an increase in anandamide and PEA levels in target neural tissue, depended upon CB1 receptor activation, and was associated with a decrease in calcitonin gene-related peptide (CGRP), substance P and cytokines TNF-alpha and IL-6 mRNA. Similar effects were observed in the chronic migraine paradigm, where URB937 counteracted NTG-induced trigeminal hyperalgesia and prevented the increase in neuropeptide and cytokine transcription. The results show that peripheral FAAH inhibition by URB937 effectively reduces both acute and chronic NTG-induced trigeminal hyperalgesia, likely via augmented anandamide-mediated CB1 receptor activation. These effects are associated with inhibition of neuropeptidergic and inflammatory pathways.
Collapse
Affiliation(s)
- Rosaria Greco
- Translational Neurovascular Research Unit, Headache Science Centre, IRCCS Mondino Foundation, via Mondino 2, 27100 Pavia, Italy.
| | - Chiara Demartini
- Translational Neurovascular Research Unit, Headache Science Centre, IRCCS Mondino Foundation, via Mondino 2, 27100 Pavia, Italy
| | - Annamaria Zanaboni
- Translational Neurovascular Research Unit, Headache Science Centre, IRCCS Mondino Foundation, via Mondino 2, 27100 Pavia, Italy; Department of Brain and Behavioral Sciences, University of Pavia, via Bassi 21, 27100 Pavia, Italy
| | - Ilenia Casini
- Translational Neurovascular Research Unit, Headache Science Centre, IRCCS Mondino Foundation, via Mondino 2, 27100 Pavia, Italy
| | - Roberto De Icco
- Translational Neurovascular Research Unit, Headache Science Centre, IRCCS Mondino Foundation, via Mondino 2, 27100 Pavia, Italy
| | - Angelo Reggiani
- Dept. of Drug Discovery and Development, Istituto Italiano di Tecnologia, Genova, Italy
| | - Alessandra Misto
- Dept. of Drug Discovery and Development, Istituto Italiano di Tecnologia, Genova, Italy
| | - Daniele Piomelli
- Department of Anatomy and Neurobiology, University of California, Irvine, CA, USA
| | - Cristina Tassorelli
- Translational Neurovascular Research Unit, Headache Science Centre, IRCCS Mondino Foundation, via Mondino 2, 27100 Pavia, Italy; Department of Brain and Behavioral Sciences, University of Pavia, via Bassi 21, 27100 Pavia, Italy
| |
Collapse
|
24
|
Brunetti L, Carrieri A, Piemontese L, Tortorella P, Loiodice F, Laghezza A. Beyond the Canonical Endocannabinoid System. A Screening of PPAR Ligands as FAAH Inhibitors. Int J Mol Sci 2020; 21:ijms21197026. [PMID: 32987725 PMCID: PMC7582602 DOI: 10.3390/ijms21197026] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 09/21/2020] [Accepted: 09/22/2020] [Indexed: 12/16/2022] Open
Abstract
In recent years, Peroxisome Proliferator-Activated Receptors (PPARs) have been connected to the endocannabinoid system. These nuclear receptors indeed mediate the effects of anandamide and similar substances such as oleoyl-ethanolamide and palmitoyl-ethanolamide. An increasing body of literature describing the interactions between the endocannabinoid system and PPARs has slowly but surely been accumulating over the past decade, and a multitarget approach involving these receptors and endocannabinoid degrading enzyme FAAH has been proposed for the treatment of inflammatory states, cancer, and Alzheimer’s disease. The lack of knowledge about compounds endowed with such an activity profile therefore led us to investigate a library of readily available, well-characterized PPAR agonists that we had synthesized over the years in order to find a plausible lead compound for further development. Moreover, we propose a rationalization of our results via a docking study, which sheds some light on the binding mode of these PPAR agonists to FAAH and opens the way for further research in this field.
Collapse
|
25
|
Gomes ACC, Soares MA, Gomes AKC, Silva NLC, Miranda ALP, Tributino JLM, Luz DA, de Amorim GC, Simas NK, Kuster RM. Antinociceptive constituents from Saccharum officinarum L. juice. Nat Prod Res 2020; 35:5392-5396. [PMID: 32515612 DOI: 10.1080/14786419.2020.1771708] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
This work aimed to investigate the main components of methanol fractions (MFSC and MFSCf) from Saccharum officinarum L. juice and their in vivo antinociceptive potential. After LC-ESI-MS and ESI-MS/MS analysis, phenolic compounds, such as dicaffeoylquinic acid, schaftoside, vicenin-2, stilbene glycoside and the major compound tricin-7-O-(2″- α-L-rhamnopyranosyl)-α-D-galacturonide (1), were identified. MFSC and MFSCf significantly inhibited nociceptive responses in classical mice pain models. The isolated flavone, 1, inhibited strongly the neurogenic phase in formalin test without interfering with the inflammatory one. The co-administration of the opioid antagonist, naloxone, significantly reversed the antinociceptive effects on the neurogenic phase of both methanol fractions and 1, demonstrating the involvement of the opioid system on the antinociceptive effect. This work describes for the first time the antinociceptive effect of flavonoids present in sugarcane juice, highlighting the isolation and the structural elucidation of tricin-7-O-(2″-α-L-rhamnopyranosyl)-α-D-galacturonide through ESI-MS/MS, 1D- and 2D-NMR.
Collapse
Affiliation(s)
- Anne C C Gomes
- Instituto de Pesquisas de Produtos Naturais, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brasil.,Instituto Federal de Educação, Ciência e Tecnologia do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
| | - Mariana A Soares
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
| | - Anne K C Gomes
- Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
| | - Natália L C Silva
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
| | - Ana L P Miranda
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
| | - Jorge L M Tributino
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
| | - Dandara A Luz
- Departamento de Química, Universidade Federal do Espírito Santo, Espírito Santo, ES, Brasil
| | - Gisele C de Amorim
- Instituto de Bioquímica Médica, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
| | - Naomi K Simas
- Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
| | - Ricardo M Kuster
- Instituto de Pesquisas de Produtos Naturais, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brasil.,Departamento de Química, Universidade Federal do Espírito Santo, Espírito Santo, ES, Brasil
| |
Collapse
|
26
|
Matheson J, Le Foll B. Therapeutic Potential of Peroxisome Proliferator-Activated Receptor (PPAR) Agonists in Substance Use Disorders: A Synthesis of Preclinical and Human Evidence. Cells 2020; 9:cells9051196. [PMID: 32408505 PMCID: PMC7291117 DOI: 10.3390/cells9051196] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 05/06/2020] [Accepted: 05/08/2020] [Indexed: 12/17/2022] Open
Abstract
Targeting peroxisome proliferator-activated receptors (PPARs) has received increasing interest as a potential strategy to treat substance use disorders due to the localization of PPARs in addiction-related brain regions and the ability of PPAR ligands to modulate dopamine neurotransmission. Robust evidence from animal models suggests that agonists at both the PPAR-α and PPAR-γ isoforms can reduce both positive and negative reinforcing properties of ethanol, nicotine, opioids, and possibly psychostimulants. A reduction in the voluntary consumption of ethanol following treatment with PPAR agonists seems to be the most consistent finding. However, the human evidence is limited in scope and has so far been less promising. There have been no published human trials of PPAR agonists for treatment of alcohol use disorder, despite the compelling preclinical evidence. Two trials of PPAR-α agonists as potential smoking cessation drugs found no effect on nicotine-related outcomes. The PPAR-γ agonist pioglitazone showed some promise in reducing heroin, nicotine, and cocaine craving in two human laboratory studies and one pilot trial, yet other outcomes were unaffected. Potential explanations for the discordance between the animal and human evidence, such as the potency and selectivity of PPAR ligands and sex-related variability in PPAR physiology, are discussed.
Collapse
Affiliation(s)
- Justin Matheson
- Department of Pharmacology and Toxicology, Faculty of Medicine, University of Toronto, 27 King’s College Circle, Toronto, ON M5S 3H7, Canada;
- Institute for Mental Health Policy Research, Centre for Addiction and Mental Health, 33 Russell Street, Toronto, ON M5S 2S1, Canada
- Correspondence: ; Tel.: +1-416-535-8501 (ext. 34727)
| | - Bernard Le Foll
- Department of Pharmacology and Toxicology, Faculty of Medicine, University of Toronto, 27 King’s College Circle, Toronto, ON M5S 3H7, Canada;
- Translational Addiction Research Laboratory, Centre for Addiction and Mental Health, 33 Russell Street, Toronto, ON M5S 2S1, Canada
- Addictions Division, Centre for Addiction and Mental Health, 100 Stokes Street, Toronto, ON M6J 1H4, Canada
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, 250 College Street, Toronto, ON M5T 1R8, Canada
- Department of Psychiatry, Faculty of Medicine, University of Toronto, 250 College Street, Toronto, ON M5T 1R8, Canada
- Institute of Medical Sciences, University of Toronto, 1 King’s College Circle, Room 2374, Toronto, ON M5S 1A8, Canada
- Department of Family and Community Medicine, University of Toronto, 500 University Avenue, 5th Floor, Toronto, ON M5G 1V7, Canada
| |
Collapse
|
27
|
Wu HF, Lu TY, Chu MC, Chen PS, Lee CW, Lin HC. Targeting the inhibition of fatty acid amide hydrolase ameliorate the endocannabinoid-mediated synaptic dysfunction in a valproic acid-induced rat model of Autism. Neuropharmacology 2020; 162:107736. [DOI: 10.1016/j.neuropharm.2019.107736] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 08/02/2019] [Accepted: 08/05/2019] [Indexed: 10/26/2022]
|
28
|
Brunetti L, Laghezza A, Loiodice F, Tortorella P, Piemontese L. Combining fatty acid amide hydrolase (FAAH) inhibition with peroxisome proliferator-activated receptor (PPAR) activation: a new potential multi-target therapeutic strategy for the treatment of Alzheimer's disease. Neural Regen Res 2020; 15:67-68. [PMID: 31535650 PMCID: PMC6862411 DOI: 10.4103/1673-5374.264458] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Affiliation(s)
- Leonardo Brunetti
- Dipartimento di Farmacia - Scienze del Farmaco, Università degli Studi di Bari "Aldo Moro", Bari, Italy
| | - Antonio Laghezza
- Dipartimento di Farmacia - Scienze del Farmaco, Università degli Studi di Bari "Aldo Moro", Bari, Italy
| | - Fulvio Loiodice
- Dipartimento di Farmacia - Scienze del Farmaco, Università degli Studi di Bari "Aldo Moro", Bari, Italy
| | - Paolo Tortorella
- Dipartimento di Farmacia - Scienze del Farmaco, Università degli Studi di Bari "Aldo Moro", Bari, Italy
| | - Luca Piemontese
- Dipartimento di Farmacia - Scienze del Farmaco, Università degli Studi di Bari "Aldo Moro", Bari, Italy
| |
Collapse
|
29
|
Tripathi RKP. A perspective review on fatty acid amide hydrolase (FAAH) inhibitors as potential therapeutic agents. Eur J Med Chem 2019; 188:111953. [PMID: 31945644 DOI: 10.1016/j.ejmech.2019.111953] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Revised: 12/02/2019] [Accepted: 12/04/2019] [Indexed: 02/06/2023]
Abstract
Fatty acid amide hydrolase (FAAH) is an important enzyme creditworthy of hydrolyzing endocannabinoids and related-amidated signalling lipids, discovery of which has pioneered novel arena of pharmacological canvasses to unwrap its curative potency in various diseased circumstances. It presents contemporary basis for understanding molecules regulating and mediating inflammatory reactions, pain, anxiety, depression, and neurodegeneration. FAAH inhibitors form vital approach for discovery of therapeutic agents that are concerned with local elevation of endocannabinoids under certain stimuli, debarring adverse/unwanted secondary effects from global activation of cannabinoid receptors by exogenous cannabimimetics. During past decades, several molecules with excellent potency developed through tailor-made approaches entered into clinical trials, but none could reach market. Hence, hunt for novel, non-toxic and selective FAAH inhibitors are on horizon. This review summarizes present perception on FAAH in conjunction with its structure, mechanism of catalysis and biological functions. It also foregrounds recent development of molecules belonging to diverse chemical classes as potential FAAH inhibitors bobbing up from in-depth chemical, mechanistic and computational studies published since 2015-November 2019, focusing on their potency. This review will assist readers to obtain rationale on FAAH as potential target for addressing various disease conditions, acquiring significant knowledge on recently established inhibitor scaffolds and their development potentials. New technologies including MD-MM simulations and 3D-QSAR studies allow mechanistic characterization of enzyme. Assessment of in-vitro and in-vivo efficacy of existing FAAH inhibitors will facilitate researchers to design novel ligands utilizing modern drug design methods. The discussions will also impose precaution in decision making process, quashing possibility of late stage failure.
Collapse
Affiliation(s)
- Rati Kailash Prasad Tripathi
- Department of Pharmaceutical Science, Sushruta School of Medical and Paramedical Sciences, Assam University (A Central University), Silchar, Assam, 788011, India; Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology, Banaras Hindu University, Varanasi, Uttar Pradesh, 221005, India.
| |
Collapse
|
30
|
Abstract
Substance use disorder (SUD) is a major public health crisis worldwide, and effective treatment options are limited. During the past 2 decades, researchers have investigated the impact of a variety of pharmacological approaches to treat SUD, one of which is the use of medical cannabis or cannabinoids. Significant progress was made with the discovery of rimonabant, a selective CB1 receptor (CB1R) antagonist (also an inverse agonist), as a promising therapeutic for SUDs and obesity. However, serious adverse effects such as depression and suicidality led to the withdrawal of rimonabant (and almost all other CB1R antagonists/inverse agonists) from clinical trials worldwide in 2008. Since then, much research interest has shifted to other cannabinoid-based strategies, such as peripheral CB1R antagonists/inverse agonists, neutral CB1R antagonists, allosteric CB1R modulators, CB2R agonists, fatty acid amide hydrolase (FAAH) inhibitors, monoacylglycerol lipase (MAGL) inhibitors, fatty acid binding protein (FABP) inhibitors, or nonaddictive phytocannabinoids with CB1R or CB2R-binding profiles, as new therapeutics for SUDs. In this article, we first review recent progress in research regarding the endocannabinoid systems, cannabis reward versus aversion, and the underlying receptor mechanisms. We then review recent progress in cannabinoid-based medication development for the treatment of SUDs. As evidence continues to accumulate, neutral CB1R antagonists (such as AM4113), CB2R agonists (JWH133, Xie2-64), and nonselective phytocannabinoids (cannabidiol, β-caryophyllene, ∆9-tetrahydrocannabivarin) have shown great therapeutic potential for SUDs, as shown in experimental animals. Several cannabinoid-based medications (e.g., dronabinol, nabilone, PF-04457845) that entered clinical trials have shown promising results in reducing withdrawal symptoms in cannabis and opioid users.
Collapse
Affiliation(s)
- Ewa Galaj
- Addiction Biology Unit, Molecular Targets and Medication Discoveries Branch, Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD, 21224, USA
| | - Zheng-Xiong Xi
- Addiction Biology Unit, Molecular Targets and Medication Discoveries Branch, Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD, 21224, USA.
| |
Collapse
|
31
|
Brunetti L, Loiodice F, Piemontese L, Tortorella P, Laghezza A. New Approaches to Cancer Therapy: Combining Fatty Acid Amide Hydrolase (FAAH) Inhibition with Peroxisome Proliferator-Activated Receptors (PPARs) Activation. J Med Chem 2019; 62:10995-11003. [PMID: 31407888 DOI: 10.1021/acs.jmedchem.9b00885] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Over the course of the past decade, peroxisome proliferator-activated receptors (PPARs) have been identified as part of the cannabinoid signaling system: both phytocannabinoids and endocannabinoids are capable of binding and activating these nuclear receptors. Fatty acid amide hydrolase (FAAH) hydrolyzes the endocannabinoid anandamide and other N-acylethanolamines. These substances have been shown to have numerous anticancer effects, and indeed the inhibition of FAAH has multiple beneficial effects that are mediated by PPARα subtype and by PPARγ subtype, especially antiproliferation and activation of apoptosis. The substrates of FAAH are also PPAR agonists, which explains the PPAR-mediated effects of FAAH inhibitors. Much like cannabinoid ligands and FAAH inhibitors, PPARγ agonists show antiproliferative effects on cancer cells, suggesting that additive or synergistic effects may be achieved through the positive modulation of both signaling systems. In this Miniperspective, we discuss the development of novel FAAH inhibitors able to directly act as PPAR agonists and their promising utilization as leads for the discovery of highly effective anticancer compounds.
Collapse
Affiliation(s)
- Leonardo Brunetti
- Dipartimento di Farmacia-Scienze del Farmaco , Università degli Studi di Bari "Aldo Moro" , via Orabona 4 , 70125 Bari , Italy
| | - Fulvio Loiodice
- Dipartimento di Farmacia-Scienze del Farmaco , Università degli Studi di Bari "Aldo Moro" , via Orabona 4 , 70125 Bari , Italy
| | - Luca Piemontese
- Dipartimento di Farmacia-Scienze del Farmaco , Università degli Studi di Bari "Aldo Moro" , via Orabona 4 , 70125 Bari , Italy
| | - Paolo Tortorella
- Dipartimento di Farmacia-Scienze del Farmaco , Università degli Studi di Bari "Aldo Moro" , via Orabona 4 , 70125 Bari , Italy
| | - Antonio Laghezza
- Dipartimento di Farmacia-Scienze del Farmaco , Università degli Studi di Bari "Aldo Moro" , via Orabona 4 , 70125 Bari , Italy
| |
Collapse
|
32
|
Contarini G, Ferretti V, Papaleo F. Acute Administration of URB597 Fatty Acid Amide Hydrolase Inhibitor Prevents Attentional Impairments by Distractors in Adolescent Mice. Front Pharmacol 2019; 10:787. [PMID: 31379568 PMCID: PMC6658611 DOI: 10.3389/fphar.2019.00787] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 06/18/2019] [Indexed: 11/18/2022] Open
Abstract
The maturation of attentional control during adolescence might influence later functional outcome or predisposition to psychiatric disorders. During adolescence, the cannabinoid system is particularly sensitive to pharmacological challenges, with potential impact on cognitive functions. Here, we used a recently validated five-choice serial reaction time task protocol to test adolescent C57BL/6J mice. We showed that the pharmacological inhibition (by URB597) of the fatty acid amide hydrolase (FAAH), the major enzyme implicated in anandamide degradation, prevented cognitive disruptions induced by distracting cues in adolescent mice. In particular, these protective effects were indicated by increased accuracy and correct responses and decreased premature responses selectively in the distractor trials. Notably, at the relatively low dose used, we detected no effects in other cognitive, motor, or incentive measures nor long-lasting or rebound effects of FAAH inhibition in cognitive functions. Overall, these data provide initial evidence of selective procognitive effects of FAAH inhibition in measures of attentional control in adolescent mice.
Collapse
Affiliation(s)
- Gabriella Contarini
- Department of Neuroscience and Brain Technologies, Genetics of Cognition Laboratory, Istituto Italiano di Tecnologia, Genova, Italy
| | - Valentina Ferretti
- Department of Neuroscience and Brain Technologies, Genetics of Cognition Laboratory, Istituto Italiano di Tecnologia, Genova, Italy
| | - Francesco Papaleo
- Department of Neuroscience and Brain Technologies, Genetics of Cognition Laboratory, Istituto Italiano di Tecnologia, Genova, Italy
| |
Collapse
|
33
|
Jacobson MR, Watts JJ, Boileau I, Tong J, Mizrahi R. A systematic review of phytocannabinoid exposure on the endocannabinoid system: Implications for psychosis. Eur Neuropsychopharmacol 2019; 29:330-348. [PMID: 30635160 DOI: 10.1016/j.euroneuro.2018.12.014] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 07/17/2018] [Accepted: 12/20/2018] [Indexed: 12/18/2022]
Abstract
Cannabis, the most widely used illicit drug worldwide, produces psychoactive effects through its component cannabinoids, which act on the endocannabinoid system. Research on how cannabinoid exposure affects the endocannabinoid system is limited. Substantial evidence indicates cannabis use as a risk factor for psychosis, and the mechanism(s) by which this is occurring is/are currently unknown. Here, we conduct the first review of the effects of exogenous cannabinoids on the endocannabinoid system in humans with and without psychotic disorders. The most well established finding is the down-regulation of cannabinoid CB1 receptors (CB1R) after chronic and recent cannabis exposure, but it remains uncertain whether this effect is present in cannabis users with schizophrenia. We highlight where cannabis exposure affects the endocannabinoid system in a pattern that may mirror what is seen in psychosis, and how further research can push this field forward. In these times of changing cannabis legislation, research highlighting the biological effects of cannabinoids is greatly needed.
Collapse
Affiliation(s)
- Maya R Jacobson
- Research Imaging Centre, Centre for Addiction and Mental Health, 250 College St., Toronto, Ontario M5T 1R8, Canada; Department of Pharmacology and Toxicology, Faculty of Medicine, 1 King's College Circle, University of Toronto, Toronto, Ontario M5S 1A8, Canada.
| | - Jeremy J Watts
- Research Imaging Centre, Centre for Addiction and Mental Health, 250 College St., Toronto, Ontario M5T 1R8, Canada; Department of Pharmacology and Toxicology, Faculty of Medicine, 1 King's College Circle, University of Toronto, Toronto, Ontario M5S 1A8, Canada.
| | - Isabelle Boileau
- Research Imaging Centre, Centre for Addiction and Mental Health, 250 College St., Toronto, Ontario M5T 1R8, Canada; Centre for Addiction and Mental Health, Campbell Family Mental Health Research Institute, 250 College St., Toronto, Ontario M5T 1R8, Canada; Department of Psychiatry, University of Toronto, 250 College St., Toronto, Ontario M5T 1R8, Canada; Institute of Medical Science, Faculty of Medicine, 1 King's College Circle, University of Toronto, Ontario M5S 1A8, Canada.
| | - Junchao Tong
- Research Imaging Centre, Centre for Addiction and Mental Health, 250 College St., Toronto, Ontario M5T 1R8, Canada; Centre for Addiction and Mental Health, Campbell Family Mental Health Research Institute, 250 College St., Toronto, Ontario M5T 1R8, Canada; Department of Psychiatry, University of Toronto, 250 College St., Toronto, Ontario M5T 1R8, Canada.
| | - Romina Mizrahi
- Research Imaging Centre, Centre for Addiction and Mental Health, 250 College St., Toronto, Ontario M5T 1R8, Canada; Centre for Addiction and Mental Health, Campbell Family Mental Health Research Institute, 250 College St., Toronto, Ontario M5T 1R8, Canada; Department of Psychiatry, University of Toronto, 250 College St., Toronto, Ontario M5T 1R8, Canada; Department of Pharmacology and Toxicology, Faculty of Medicine, 1 King's College Circle, University of Toronto, Toronto, Ontario M5S 1A8, Canada; Institute of Medical Science, Faculty of Medicine, 1 King's College Circle, University of Toronto, Ontario M5S 1A8, Canada.
| |
Collapse
|
34
|
Zhou P, Xiang L, Zhao D, Ren J, Qiu Y, Li Y. Synthesis, biological evaluation, and structure activity relationship (SAR) study of pyrrolidine amide derivatives as N-acylethanolamine acid amidase (NAAA) inhibitors. MEDCHEMCOMM 2018; 10:252-262. [PMID: 30931090 DOI: 10.1039/c8md00432c] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 12/14/2018] [Indexed: 01/15/2023]
Abstract
N-Acylethanolamine acid amidase (NAAA) is one of the key enzymes involved in the degradation of fatty acid ethanolamides (FAEs), especially for palmitoylethanolamide (PEA). Pharmacological blockage of NAAA restores PEA levels, providing therapeutic benefits in the management of inflammation and pain. In the current work, we showed the structure-activity relationship (SAR) studies for pyrrolidine amide derivatives as NAAA inhibitors. A series of aromatic replacements or substituents for the terminal phenyl group of pyrrolidine amides were examined. SAR data showed that small lipophilic 3-phenyl substituents were preferable for optimal potency. The conformationally flexible linkers increased the inhibitory potency of pyrrolidine amide derivatives but reduced their selectivity toward fatty acid amide hydrolase (FAAH). The conformationally restricted linkers did not enhance the inhibitor potency toward NAAA but improved the selectivity over FAAH. Several low micromolar potent NAAA inhibitors were developed, including 4g bearing a rigid 4-phenylcinnamoyl group. Dialysis and kinetic analysis suggested that 4g inhibited NAAA via a competitive and reversible mechanism. Furthermore, 4g showed high anti-inflammatory activities in lipopolysaccharide (LPS) induced acute lung injury (ALI) model, and this effect was blocked by pre-treatment with the PPAR-α antagonist MK886. We anticipate that 4g (E93) will enable a new agent to treat inflammation and related diseases.
Collapse
Affiliation(s)
- Pan Zhou
- Eye Institute of Xiamen University , Xiamen , Fujian 361102 , China.,Medical College , Xiamen University , Xiamen , Fujian 361102 , China
| | - Lei Xiang
- Medical College , Xiamen University , Xiamen , Fujian 361102 , China
| | - Dongsheng Zhao
- Department of Pharmacy , Quanzhou Medical College , China . Tel: Quanzhou 362100
| | - Jie Ren
- Eye Institute of Xiamen University , Xiamen , Fujian 361102 , China.,Medical College , Xiamen University , Xiamen , Fujian 361102 , China
| | - Yan Qiu
- Eye Institute of Xiamen University , Xiamen , Fujian 361102 , China.,Medical College , Xiamen University , Xiamen , Fujian 361102 , China
| | - Yuhang Li
- Xiamen Institute of Rare-earth Materials , Haixi Institutes , Chinese Academy of Sciences , Fujian 361005 , China.,CAS Key Laboratory of Design and Assembly of Functional Nanostructures , and Fujian Provincial Key Laboratory of Nanomaterials , Fujian Institute of Research on the Structure of Matter , Chinese Academy of Sciences , China .
| |
Collapse
|
35
|
Horid’ko TM, Kosiakova HV, Berdyshev AG. Preventive effect of N-stearoylethanolamine on memory disorders, blood and brain biochemical parameters in rats with experimental scopolamine-induced cognitive impairment. UKRAINIAN BIOCHEMICAL JOURNAL 2018. [DOI: 10.15407/ubj90.06.097] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
36
|
De Luca MA, Buczynski MW, Di Chiara G. Loren Parsons' contribution to addiction neurobiology. Addict Biol 2018; 23:1207-1222. [PMID: 29949237 DOI: 10.1111/adb.12642] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 05/17/2018] [Indexed: 11/29/2022]
Abstract
Loren (Larry) H. Parsons passed away at the age of 51. In spite of his premature departure, Larry much contributed to the drug abuse field. Since his graduate studies for the Ph.D. in Chemistry in J.B. Justice lab, microdialysis is the tread that links Larry's research topics, namely, the role of dopamine (DA), serotonin (5-HT), gamma-aminobutyric acid (GABA), glutamate and endocannabinoids (eCBs) in drug reinforcement and dependence. Larry was the first to show that abstinence from chronic cocaine reduces extracellular DA in the NAc, consistent with the so called 'dopamine depletion hypothesis' of cocaine addiction. Another Larry's major contributions are the studies on 5-HT and 5-HT receptors' role in cocaine stimulant actions, which resulted in the identification of 5-HT1B receptors as a critical substrate of cocaine reinforcement. By applying mass spectrometry to eCBs analysis in brain dialysates, Larry's lab showed that ethanol, heroin, nicotine and cocaine differentially affect anandamide and 2-arachidonoylglicerol overflow in the NAc shell, a critical site of drugs of abuse DA stimulant actions. Larry also applied microdialysis to study GABA and glutamate's role in ethanol dependence and heroin reinforcement, providing in vivo evidence for a sensitization of corticotropin-releasing factor-dependent release of GABA in the central amygdala in withdrawal from chronic ethanol and for a reduction of GABA transmission in the ventral pallidum in heroin but not cocaine intravenous self-administration. Larry showed the wide possibilities of microdialysis as a general purpose methodology for monitoring neurotransmitters and neuromodulators in the brain extracellular compartment. From this viewpoint, he stands as the best advocate for microdialysis.
Collapse
Affiliation(s)
- Maria A. De Luca
- Department of Biomedical Sciences, Section of Neuropsychopharmacology; University of Cagliari; Cagliari Italy
- National Institute of Neuroscience (INN); University of Cagliari; Cagliari Italy
| | - Matthew W. Buczynski
- School of Neuroscience; Virginia Polytechnic Institute and State University; Blacksburg VA 24061 USA
| | - Gaetano Di Chiara
- Department of Biomedical Sciences, Section of Neuropsychopharmacology; University of Cagliari; Cagliari Italy
- National Institute of Neuroscience (INN); University of Cagliari; Cagliari Italy
- National Research Council of Italy; Institute of Neuroscience; Cagliari Italy
| |
Collapse
|
37
|
Danandeh A, Vozella V, Lim J, Oveisi F, Ramirez GL, Mears D, Wynn G, Piomelli D. Effects of fatty acid amide hydrolase inhibitor URB597 in a rat model of trauma-induced long-term anxiety. Psychopharmacology (Berl) 2018; 235:3211-3221. [PMID: 30251159 DOI: 10.1007/s00213-018-5020-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 08/31/2018] [Indexed: 12/20/2022]
Abstract
RATIONALE The endocannabinoid neurotransmitter, anandamide, has been implicated in the central modulation of stress responses. Previous animal experiments have shown that inhibitors of the anandamide-degrading enzyme, fatty acid amide hydrolase (FAAH), enhance the ability to cope with acute and chronic stress. OBJECTIVES Here, we investigated the effects of the globally active FAAH inhibitor URB597 in a rat model of predator stress-induced long-term anxiety. RESULTS Rats exposed to 2,5-dihydro-2,4,5-trimethylthiazoline (TMT), a chemical constituent of fox feces, developed a persistent anxiety-like state, which was assessed 7 days after exposure using the elevated plus maze (EPM) test. Systemic administration of URB597 [0.03-0.1-0.3 mg/kg, intraperitoneal (ip)] 2 h before testing suppressed TMT-induced behaviors with a median effective dose (IC50) of 0.075 mg/kg. This effect was strongly correlated with inhibition of brain FAAH activity (r2 = 1.0) and was accompanied by increased brain levels of three FAAH substrates: the endocannabinoid anandamide and the endogenous peroxisome proliferator-activated receptor-α (PPAR-α) agonists, oleoylethanolamide (OEA), and palmitoylethanolamide (PEA). The anxiolytic-like effects of URB597 were blocked by co-administration of the CB1 receptor antagonist rimonabant (1 mg/kg, ip), but not of the PPAR-α antagonist GW6471 (1 mg/kg, ip). Finally, when administered 18 h after TMT exposure (i.e., 6 days before the EPM test), URB597 (0.3 mg/kg, ip) prevented the consolidation of anxiety-like behavior in a CB1-dependent manner. CONCLUSIONS The results support the hypothesis that anandamide-mediated signaling at CB1 receptors serves an important regulatory function in the stress response, and confirm that FAAH inhibition may offer a potential therapeutic strategy for post-traumatic stress disorder.
Collapse
Affiliation(s)
- Andalib Danandeh
- Department of Anatomy and Neurobiology, University of California, 3101 Gillespie NRF, Irvine, CA, 92697-4625, USA
| | - Valentina Vozella
- Department of Anatomy and Neurobiology, University of California, 3101 Gillespie NRF, Irvine, CA, 92697-4625, USA
| | - James Lim
- Department of Anatomy and Neurobiology, University of California, 3101 Gillespie NRF, Irvine, CA, 92697-4625, USA.,Department of Molecular & Cellular Biology, The University of Arizona, Tucson, AZ, USA
| | - Fariba Oveisi
- Department of Anatomy and Neurobiology, University of California, 3101 Gillespie NRF, Irvine, CA, 92697-4625, USA
| | - Gina L Ramirez
- Department of Anatomy and Neurobiology, University of California, 3101 Gillespie NRF, Irvine, CA, 92697-4625, USA
| | - David Mears
- Department of Anatomy, Physiology, and Genetics, Uniformed Service University of the Health Sciences, Bethesda, MD, USA.,Center for the Study of Traumatic Stress, Department of Psychiatry, Uniformed Service University of the Health Sciences, Bethesda, MD, USA
| | - Gary Wynn
- Center for the Study of Traumatic Stress, Department of Psychiatry, Uniformed Service University of the Health Sciences, Bethesda, MD, USA
| | - Daniele Piomelli
- Department of Anatomy and Neurobiology, University of California, 3101 Gillespie NRF, Irvine, CA, 92697-4625, USA. .,Department of Pharmacology, University of California, Irvine, CA, 92697, USA. .,Department of Biological Chemistry, University of California, Irvine, CA, 92697, USA.
| |
Collapse
|
38
|
Ferguson LB, Zhang L, Wang S, Bridges C, Harris RA, Ponomarev I. Peroxisome Proliferator Activated Receptor Agonists Modulate Transposable Element Expression in Brain and Liver. Front Mol Neurosci 2018; 11:331. [PMID: 30283300 PMCID: PMC6156381 DOI: 10.3389/fnmol.2018.00331] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 08/27/2018] [Indexed: 12/17/2022] Open
Abstract
Peroxisome proliferator activated receptors (PPARs) are nuclear hormone receptors that act as transcription factors in response to endogenous lipid messengers. The fibrates and thiazolidinediones are synthetic PPAR agonists used clinically to treat dyslipidemia and Type 2 Diabetes Mellitus, respectively, but also improve symptoms of several other diseases. Transposable elements (TEs), repetitive sequences in mammalian genomes, are implicated in many of the same conditions for which PPAR agonists are therapeutic, including neurodegeneration, schizophrenia, and drug addiction. We tested the hypothesis that there is a link between actions of PPAR agonists and TE expression. We developed an innovative application of microarray data by mapping Illumina mouse WG-6 microarray probes to areas of the mouse genome that contain TEs. Using this information, we assessed the effects of systemic administration of three PPAR agonists with different PPAR subtype selectivity: fenofibrate, tesaglitazar, and bezafibrate, on TE probe expression in mouse brain [prefrontal cortex (PFC) and amygdala] and liver. We found that fenofibrate, and bezafibrate to a lesser extent, up-regulated probes mapped to retrotransposons: Short-Interspersed Elements (SINEs) and Long-Interspersed Elements (LINEs), in the PFC. Conversely, all PPAR agonists down-regulated LINEs and tesaglitazar and bezafibrate also down-regulated SINEs in liver. We built gene coexpression networks that partitioned the diverse transcriptional response to PPAR agonists into groups of probes with highly correlated expression patterns (modules). Most of the differentially expressed retrotransposons were within the same module, suggesting coordinated regulation of their expression, possibly by PPAR signaling. One TE module was conserved across tissues and was enriched with genes whose products participate in epigenetic regulation, suggesting that PPAR agonists affect TE expression via epigenetic mechanisms. Other enriched functional categories included phenotypes related to embryonic development and learning and memory, suggesting functional links between these biological processes and TE expression. In summary, these findings suggest mechanistic relationships between retrotransposons and PPAR agonists and provide a basis for future exploration of their functional roles in brain and liver.
Collapse
Affiliation(s)
- Laura B. Ferguson
- Waggoner Center for Alcohol & Addiction Research, The University of Texas at Austin, Austin, TX, United States
| | - Lingling Zhang
- MOE Key Laboratory of Marine Genetics and Breeding, Ocean University of China, Qingdao, China
| | - Shi Wang
- MOE Key Laboratory of Marine Genetics and Breeding, Ocean University of China, Qingdao, China
- Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Courtney Bridges
- Waggoner Center for Alcohol & Addiction Research, The University of Texas at Austin, Austin, TX, United States
| | - R. Adron Harris
- Waggoner Center for Alcohol & Addiction Research, The University of Texas at Austin, Austin, TX, United States
| | - Igor Ponomarev
- Waggoner Center for Alcohol & Addiction Research, The University of Texas at Austin, Austin, TX, United States
| |
Collapse
|
39
|
Kubilius RA, Kaplick PM, Wotjak CT. Highway to hell or magic smoke? The dose-dependence of Δ 9-THC in place conditioning paradigms. ACTA ACUST UNITED AC 2018; 25:446-454. [PMID: 30115766 PMCID: PMC6097764 DOI: 10.1101/lm.046870.117] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 07/09/2018] [Indexed: 02/06/2023]
Abstract
The prerequisites for responsible cannabis use are at the heart of current inquiries into cannabis decriminalization by policy makers as well as academic and nonacademic stakeholders at a global scale. Δ9-tetrahydrocannabinol (Δ9-THC), the prime psychoactive compound of the cannabis sativa, as well as cannabimimetics that resemble the pharmacological properties and psychological effects of Δ9-THC, lend themselves handsomely to the preclinical scrutiny of reward-related behavior because they carry marked translational value. Although a functional dichotomy of the psychological effects of Δ9-THC (rewarding versus aversive) has been abundantly reported in place conditioning (PC) paradigms, and might be best attributed to a dose-dependence of Δ9-THC, most PC studies with Δ9-THC feature no significant effects at all. Therefore, after decades of rigorous research, it still remains undetermined whether Δ9-THC generally exerts rewarding or aversive effects in rodents. Here, we set out to extrapolate the commonly alleged dose-dependence of the rewarding and aversive effects of Δ9-THC from the existing literature, at the behavioral pharmacological level of analysis. Specifically, our meta-analysis investigated: (i) the alleged bidirectional effects and dose-dependence of Δ9-THC in the PC test; (ii) methodological inconsistencies between PC studies; and (iii) other pharmacological studies on cannabinoids (i.e., dopamine release, anxiety, stress, conditioned taste aversion, catalepsy) to substantiate the validity of PC findings. Our findings suggest that: (i) Δ9-THC dose-dependently generates rewarding (1 mg/kg) and aversive (5 mg/kg) effects in PC; (ii) an inconsistent use of priming injections hampers a clear establishment of the rewarding effects of Δ9-THC in PC tests and might explain the seemingly contradictory plethora of nonsignificant THC studies in the PC test; and (iii) other pharmacological studies on Δ9-THC substantiate the dose-dependent biphasic effects of Δ9-THC in PC. A standardized experimental design would advance evidence-based practice in future PC studies with Δ9-THC and facilitate the pointed establishment of rewarding and aversive effects of the substance.
Collapse
Affiliation(s)
- Rimas A Kubilius
- Neuronal Plasticity Research Group, Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, 80804 Munich, Germany.,Faculty of Biology, Ludwig Maximilian University of Munich, 82152 Planegg-Martinsried, Germany
| | - Paul M Kaplick
- Neuronal Plasticity Research Group, Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, 80804 Munich, Germany.,Structural and Functional Plasticity of the Nervous System Group, Swammerdam Institute for Life Sciences, University of Amsterdam, 1098 XH Amsterdam, The Netherlands.,Institute for Interdisciplinary Studies, University of Amsterdam, 1098 XH Amsterdam, The Netherlands
| | - Carsten T Wotjak
- Neuronal Plasticity Research Group, Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| |
Collapse
|
40
|
Zhou Y, Kreek MJ. Involvement of Activated Brain Stress Responsive Systems in Excessive and "Relapse" Alcohol Drinking in Rodent Models: Implications for Therapeutics. J Pharmacol Exp Ther 2018; 366:9-20. [PMID: 29669731 PMCID: PMC5988024 DOI: 10.1124/jpet.117.245621] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 04/16/2018] [Indexed: 02/06/2023] Open
Abstract
Addictive diseases, including addiction to alcohol, pose massive public health costs. Addiction is a chronic relapsing disease caused by both the direct effects induced by drugs and persistent neuroadaptations at the molecular, cellular, and behavioral levels. These drug-type specific neuroadaptations are brought on largely by the reinforcing effects of drugs on the central nervous system and environmental stressors. Results from animal experiments have demonstrated important interactions between alcohol and stress-responsive systems. Addiction to specific drugs such as alcohol, psychostimulants, and opioids shares some common direct or downstream effects on the brain's stress-responsive systems, including arginine vasopressin and its V1b receptors, dynorphin and the κ-opioid receptors, pro-opiomelanocortin/β-endorphin and the μ-opioid receptors, and the endocannabinoids. Further study of these systems through laboratory-based and translational research could lead to the discovery of novel treatment targets and the early optimization of interventions (for example, combination) for the pharmacologic therapy of alcoholism.
Collapse
Affiliation(s)
- Yan Zhou
- Laboratory of Biology of Addictive Diseases, Rockefeller University, New York, New York
| | - Mary Jeanne Kreek
- Laboratory of Biology of Addictive Diseases, Rockefeller University, New York, New York
| |
Collapse
|
41
|
Hindocha C, Freeman TP, Grabski M, Stroud JB, Crudgington H, Davies AC, Das RK, Lawn W, Morgan CJA, Curran HV. Cannabidiol reverses attentional bias to cigarette cues in a human experimental model of tobacco withdrawal. Addiction 2018; 113:1696-1705. [PMID: 29714034 PMCID: PMC6099309 DOI: 10.1111/add.14243] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 01/15/2018] [Accepted: 04/06/2018] [Indexed: 01/01/2023]
Abstract
BACKGROUND AND AIMS Cannabidiol (CBD), a non-intoxicating cannabinoid found in cannabis, may be a promising novel smoking cessation treatment due to its anxiolytic properties, minimal side effects and research showing that it may modify drug cue salience. We used an experimental medicine approach with dependent cigarette smokers to investigate if (1) overnight nicotine abstinence, compared with satiety, will produce greater attentional bias (AB), higher pleasantness ratings of cigarette-related stimuli and increased craving and withdrawal; and (2) CBD in comparison to placebo, would attenuate AB, pleasantness of cigarette-related stimuli, craving and withdrawal and not produce any side effects. DESIGN Randomized, double-blind cross-over study with a fixed satiated session followed by two overnight abstinent sessions. SETTING UK laboratory. PARTICIPANTS Thirty non-treatment-seeking, dependent cigarette smokers recruited from the community. INTERVENTION AND COMPARATOR 800 mg oral CBD, or matched placebo (PBO) in a counterbalanced order MEASUREMENTS: AB to pictorial tobacco cues was recorded using a visual probe task and an explicit rating task. Withdrawal, craving, side effects, heart rate and blood pressure were assessed repeatedly. FINDINGS When participants received PBO, tobacco abstinence increased AB (P = 0.001, d = 0.789) compared with satiety. However, CBD reversed this effect, such that automatic AB was directed away from cigarette cues (P = 0.007, d = 0.704) and no longer differed from satiety (P = 0.82). Compared with PBO, CBD also reduced explicit pleasantness of cigarette images (P = 0.011; d = 0.514). Craving (Bayes factor = 7.08) and withdrawal (Bayes factor = 6.95) were unaffected by CBD, but greater in abstinence compared with satiety. Systolic blood pressure decreased under CBD during abstinence. CONCLUSIONS A single 800-mg oral dose of cannabidiol reduced the salience and pleasantness of cigarette cues, compared with placebo, after overnight cigarette abstinence in dependent smokers. Cannabidiol did not influence tobacco craving or withdrawal or any subjectively rated side effects.
Collapse
Affiliation(s)
- Chandni Hindocha
- Clinical Psychopharmacology UnitUniversity College LondonLondonUK
| | - Tom P. Freeman
- Clinical Psychopharmacology UnitUniversity College LondonLondonUK
- National Addiction Centre, Institute of PsychiatryPsychology and Neuroscience, King's College LondonLondonUK
| | - Meryem Grabski
- Clinical Psychopharmacology UnitUniversity College LondonLondonUK
- School of Experimental PsychologyUniversity of BristolBristolUK
| | - Jack B. Stroud
- Clinical Psychopharmacology UnitUniversity College LondonLondonUK
| | | | - Alan C. Davies
- Clinical Psychopharmacology UnitUniversity College LondonLondonUK
| | - Ravi K. Das
- Clinical Psychopharmacology UnitUniversity College LondonLondonUK
| | - William Lawn
- Clinical Psychopharmacology UnitUniversity College LondonLondonUK
| | - Celia J. A. Morgan
- Clinical Psychopharmacology UnitUniversity College LondonLondonUK
- Psychopharmacology and Addiction Research CentreUniversity of ExeterExeterUK
| | | |
Collapse
|
42
|
Reexamining Dis/Similarity-Based Tests for Rare-Variant Association with Case-Control Samples. Genetics 2018; 209:105-113. [PMID: 29545466 DOI: 10.1534/genetics.118.300769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 03/02/2018] [Indexed: 11/18/2022] Open
Abstract
A properly designed distance-based measure can capture informative genetic differences among individuals with different phenotypes and can be used to detect variants responsible for the phenotypes. To detect associated variants, various tests have been designed to contrast genetic dissimilarity or similarity scores of certain subject groups in different ways, among which the most widely used strategy is to quantify the difference between the within-group genetic dissimilarity/similarity (i.e., case-case and control-control similarities) and the between-group dissimilarity/similarity (i.e., case-control similarities). While it has been noted that for common variants, the within-group and the between-group measures should all be included; in this work, we show that for rare variants, comparison based on the two within-group measures can more effectively quantify the genetic difference between cases and controls. The between-group measure tends to overlap with one of the two within-group measures for rare variants, although such overlap is not present for common variants. Consequently, a dissimilarity or similarity test that includes the between-group information tends to attenuate the association signals and leads to power loss. Based on these findings, we propose a dissimilarity test that compares the degree of SNP dissimilarity within cases to that within controls to better characterize the difference between two disease phenotypes. We provide the statistical properties, asymptotic distribution, and computation details for a small sample size of the proposed test. We use simulated and real sequence data to assess the performance of the proposed test, comparing it with other rare-variant methods including those similarity-based tests that use both within-group and between-group information. As similarity-based approaches serve as one of the dominating approaches in rare-variant analysis, our results provide some insight for the effective detection of rare variants.
Collapse
|
43
|
Pavon FJ, Serrano A, Sidhpura N, Polis I, Stouffer D, de Fonseca FR, Cravatt BF, Martin-Fardon R, Parsons LH. Fatty acid amide hydrolase (FAAH) inactivation confers enhanced sensitivity to nicotine-induced dopamine release in the mouse nucleus accumbens. Addict Biol 2018; 23:723-734. [PMID: 28660730 DOI: 10.1111/adb.12531] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 05/29/2017] [Indexed: 01/06/2023]
Abstract
Nicotine exerts its rewarding effects by promoting an increase in dopamine (DA) release in the nucleus accumbens (NAc), and this process is influenced by the endocannabinoid system. Fatty acid amide hydrolase (FAAH) is the main enzyme responsible for the degradation of the endocannabinoid anandamide and other non-cannabinoid N-acylethanolamines. Previous research has reported that both genetic deletion and pharmacological inhibition of FAAH enhance nicotine-induced conditioned place preference at low doses. We conducted a microdialysis study to characterize nicotine-induced changes in DA and serotonin (5-HT) levels in the NAc of FAAH knockout (KO) mice using a conditioned place preference-like paradigm with three nicotine doses (0.1, 1 and 10 mg/kg, s.c.). Additionally, the effects of the selective FAAH inhibitor PF-3845 (10 mg/kg, i.p.) were also examined. Our data indicated that compared with wild-type mice, genetic deletion of FAAH selectively enhanced the effect of low-dose nicotine on DA release (p < 0.001) and resulted in a strong post-nicotine elevation in DA levels (p < 0.01). However, there were no differences between the genotypes at higher doses. Furthermore, FAAH KO mice displayed a moderate enhancement of the effect of low-dose nicotine on NAc 5-HT release (p < 0.05), with no differences between the genotypes at higher doses. Compared with vehicle-pretreated mice, mice pretreated with PF-3845 displayed an enhancement of the effect of low-dose nicotine on NAc DA release (p < 0.001), which resulted in a sustained increase in DA levels (p < 0.05). Similar to FAAH KO mice, PF-3845-pretreated mice displayed a moderate enhancement of the effect of low-dose nicotine on NAc 5-HT release (p < 0.01). These observations in mice suggest that enhanced nicotine-induced NAc DA release might contribute to increased sensitivity to the conditioned rewarding effects of low-dose nicotine following FAAH inhibition, which has been previously reported. Future studies combining behavioral and neurochemical approaches are needed to elucidate the precise mechanism of these effects.
Collapse
Affiliation(s)
- Francisco J. Pavon
- Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA); Hospital Regional Universitario de Málaga/Universidad de Málaga; Malaga Spain
- Department of Neuroscience; The Scripps Research Institute; La Jolla California USA
| | - Antonia Serrano
- Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA); Hospital Regional Universitario de Málaga/Universidad de Málaga; Malaga Spain
- Department of Neuroscience; The Scripps Research Institute; La Jolla California USA
| | - Nimish Sidhpura
- Department of Neuroscience; The Scripps Research Institute; La Jolla California USA
| | - Ilham Polis
- Department of Neuroscience; The Scripps Research Institute; La Jolla California USA
| | - David Stouffer
- Department of Neuroscience; The Scripps Research Institute; La Jolla California USA
| | - Fernando Rodriguez de Fonseca
- Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA); Hospital Regional Universitario de Málaga/Universidad de Málaga; Malaga Spain
| | - Benjamin F. Cravatt
- Department of Chemical Physiology, Skaggs Institute for Chemical Biology; The Scripps Research Institute; La Jolla California USA
| | - Rémi Martin-Fardon
- Department of Neuroscience; The Scripps Research Institute; La Jolla California USA
| | - Loren H. Parsons
- Department of Neuroscience; The Scripps Research Institute; La Jolla California USA
| |
Collapse
|
44
|
Blockade of alcohol escalation and "relapse" drinking by pharmacological FAAH inhibition in male and female C57BL/6J mice. Psychopharmacology (Berl) 2017; 234:2955-2970. [PMID: 28730283 PMCID: PMC5693682 DOI: 10.1007/s00213-017-4691-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Accepted: 07/03/2017] [Indexed: 01/09/2023]
Abstract
BACKGROUND Anandamide (AEA)-dependent signaling is regulated by the catabolic enzyme fatty acid amide hydrolase (FAAH). Several lines of evidence have demonstrated that FAAH and AEA are involved in the behavioral effects of alcohol. Therefore, we investigated whether a selective FAAH inhibitor, URB597 (cyclohexylcarbamic acid 3'-[aminocarbonyl]-[1,1'-biphenyl]-3-yl ester), altered alcohol intake in mice in a voluntary alcohol drinking model. METHODS Mice, subjected to 3 weeks of chronic intermittent access (IA) in a two-bottle choice paradigm with 24-h access every other day, developed rapid escalation of alcohol intake and high preference. We evaluated the pharmacological effects of URB597 after both acute (1-day) withdrawal from chronic IA and 1-week withdrawal using the alcohol deprivation effect (ADE) model. AEA and N-acyl ethanolamide (NAE) abundances were determined after chronic IA, acute (1-day), or long-term (1 and 2 weeks) withdrawal in four brain regions. RESULTS Acute pretreatment with URB597 reduced alcohol intake and preference after acute withdrawal. This effect was blocked by pretreatment with a selective type 1 cannabinoid receptor (CB1) antagonist, suggesting a CB1-mediated mechanism. Both single- and multiple-dosing regimens with an effective dose of URB597 prevented the ADE, with no tolerance development after the multi-dosing regimen. AEA and NAE levels were transiently increased in all brain regions measured after acute withdrawal, indicating that the endocannabinoid system is involved in acute alcohol withdrawal stress response. CONCLUSION FAAH inhibitors reduce alcohol escalation and "relapse" drinking in mice.
Collapse
|
45
|
Antistress effects of N-stearoylethanolamine in rats with chronic social stress. UKRAINIAN BIOCHEMICAL JOURNAL 2017. [DOI: 10.15407/ubj89.04.068] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
|
46
|
Lipids in psychiatric disorders and preventive medicine. Neurosci Biobehav Rev 2017; 76:336-362. [DOI: 10.1016/j.neubiorev.2016.06.002] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Revised: 05/06/2016] [Accepted: 06/06/2016] [Indexed: 01/12/2023]
|
47
|
De Simone A, Russo D, Ruda GF, Micoli A, Ferraro M, Di Martino RMC, Ottonello G, Summa M, Armirotti A, Bandiera T, Cavalli A, Bottegoni G. Design, Synthesis, Structure–Activity Relationship Studies, and Three-Dimensional Quantitative Structure–Activity Relationship (3D-QSAR) Modeling of a Series of O-Biphenyl Carbamates as Dual Modulators of Dopamine D3 Receptor and Fatty Acid Amide Hydrolase. J Med Chem 2017; 60:2287-2304. [DOI: 10.1021/acs.jmedchem.6b01578] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
48
|
Figueiredo A, Hamilton J, Marion M, Blum K, Kaczocha M, Haj-Dahmane S, Deutsch D, Thanos PK. Pharmacological Inhibition of Brain Fatty Acid Binding Protein Reduces Ethanol Consumption in Mice. JOURNAL OF REWARD DEFICIENCY SYNDROME AND ADDICTION SCIENCE 2017; 3:21-27. [PMID: 29367955 PMCID: PMC5777574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The endocannabinoid (eCB) system is involved in a wide range of behavioral disorders including alcoholism. Inhibition of fatty acid amide hydrolase (FAAH), the principal enzyme that degrades the eCB anandamide (AEA), which enhances AEA levels in the brain, significantly increases ethanol consumption and preference. In the present study, we examined whether pharmacological inhibition of fatty acid binding proteins (FABPs) 5 and 7, which blocks the transport of AEA to FAAH, and increase AEA levels in vivo also alters ethanol consumption and preference. Using a limited access two-bottle choice paradigm, we evaluated ethanol consumption in both male and female C57Bl/6 mice. Results showed a significant decrease in ethanol consumption in both males and females treated with SBFI26, an inhibitor of FABPs. Specifically, male and female mice treated with SBFI26 consumed 24% and 42% less compared to mice receiving no injections, respectively. Subsequently, corticosterone was examined to evaluate the effects FABP5/7 inhibition upon the stress response. We observed a significant elevation in corticosterone levels following restraint stress in SBFI26 treated females, with a weak effect seen in males as compared to vehicle. Based on our results, targeting of FABPs appears to play an important role in ethanol consumption that is differentially regulated in males and females, which is mediated by the stress response.
Collapse
Affiliation(s)
- Antonio Figueiredo
- Behavioral Neuropharmacology and Neuroimaging Laboratory on Addictions, Research Institute on Addictions, University at Buffalo, Buffalo, NY, USA
| | - John Hamilton
- Behavioral Neuropharmacology and Neuroimaging Laboratory on Addictions, Research Institute on Addictions, University at Buffalo, Buffalo, NY, USA
| | - Matthew Marion
- Behavioral Neuropharmacology and Neuroimaging Laboratory on Addictions, Research Institute on Addictions, University at Buffalo, Buffalo, NY, USA
| | - Kenneth Blum
- Department of Psychiatry and McKnight Brain Institute, University of Florida College of Medicine, Gainesville, FL, USA
| | - Martin Kaczocha
- Department of Anesthesiology, Stony Brook University, Stony Brook, NY, USA
| | - Samir Haj-Dahmane
- Behavioral Neuropharmacology and Neuroimaging Laboratory on Addictions, Research Institute on Addictions, University at Buffalo, Buffalo, NY, USA
| | - Dale Deutsch
- Department of Biochemistry, Stony Brook University, Stony Brook, NY, USA
| | - Panayotis K. Thanos
- Behavioral Neuropharmacology and Neuroimaging Laboratory on Addictions, Research Institute on Addictions, University at Buffalo, Buffalo, NY, USA,Correspondence to: Dr. Panayotis K. Thanos, Behavioral Neuropharmacology and Neuroimaging Laboratory on Addictions, Research Institute on Addictions, University at Buffalo, Buffalo, New York, 14203, USA Tel: (716) 881-7520,
| |
Collapse
|
49
|
Endocannabinoid System: the Direct and Indirect Involvement in the Memory and Learning Processes-a Short Review. Mol Neurobiol 2016; 54:8332-8347. [PMID: 27924524 PMCID: PMC5684264 DOI: 10.1007/s12035-016-0313-5] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 11/21/2016] [Indexed: 11/24/2022]
Abstract
The endocannabinoid system via cannabinoid (CB: CB1 and CB2) receptors and their endogenous ligands is directly and indirectly involved in many physiological functions, especially in memory and learning processes. Extensive studies reported that this system strictly modulates cognition-related processes evaluated in various animal models. However, the effects of cannabinoids on the cognition have been contradictory. The cannabinoid compounds were able to both impair or improve different phases of memory processes through direct (receptor related) or indirect (non-receptor related) mechanism. The memory-related effects induced by the cannabinoids can be depended on the kind of cannabinoid compound used, dosage, and route of administration as well as on the memory task chosen. Therefore, the objectives of this paper are to review and summarize the results describing the role of endocannabinoid system in cognition, including various stages of memory.
Collapse
|
50
|
Boileau I, Mansouri E, Williams B, Le Foll B, Rusjan P, Mizrahi R, Tyndale RF, Huestis MA, Payer DE, Wilson AA, Houle S, Kish SJ, Tong J. Fatty Acid Amide Hydrolase Binding in Brain of Cannabis Users: Imaging With the Novel Radiotracer [ 11C]CURB. Biol Psychiatry 2016; 80:691-701. [PMID: 27345297 PMCID: PMC5050070 DOI: 10.1016/j.biopsych.2016.04.012] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Revised: 03/24/2016] [Accepted: 04/18/2016] [Indexed: 01/01/2023]
Abstract
BACKGROUND One of the major mechanisms for terminating the actions of the endocannabinoid anandamide is hydrolysis by fatty acid amide hydrolase (FAAH), and inhibitors of the enzyme were suggested as potential treatment for human cannabis dependence. However, the status of brain FAAH in cannabis use disorder is unknown. METHODS Brain FAAH binding was measured with positron emission tomography and [11C]CURB in 22 healthy control subjects and ten chronic cannabis users during early abstinence. The FAAH genetic polymorphism (rs324420) and blood, urine, and hair levels of cannabinoids and metabolites were determined. RESULTS In cannabis users, FAAH binding was significantly lower by 14%-20% across the brain regions examined than in matched control subjects (overall Cohen's d = 0.96). Lower binding was negatively correlated with cannabinoid concentrations in blood and urine and was associated with higher trait impulsiveness. CONCLUSIONS Lower FAAH binding levels in the brain may be a consequence of chronic and recent cannabis exposure and could contribute to cannabis withdrawal. This effect should be considered in the development of novel treatment strategies for cannabis use disorder that target FAAH and endocannabinoids. Further studies are needed to examine possible changes in FAAH binding during prolonged cannabis abstinence and whether lower FAAH binding predates drug use.
Collapse
Affiliation(s)
- Isabelle Boileau
- Addiction Imaging Research Group, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Human Brain Lab, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Psychiatry, University of Toronto, Toronto, Ontario, Canada; Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada.
| | - Esmaeil Mansouri
- Addiction Imaging Research Group, Centre for Addiction and Mental Health, 250 College Street, Toronto, ON, Canada,Human Brain Lab, Centre for Addiction and Mental Health, 250 College Street, Toronto, ON, Canada,Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, 250 College Street, Toronto, ON, Canada,Research Imaging Centre, Centre for Addiction and Mental Health, 250 College Street, Toronto, ON, Canada
| | - Belinda Williams
- Addiction Imaging Research Group, Centre for Addiction and Mental Health, 250 College Street, Toronto, ON, Canada,Human Brain Lab, Centre for Addiction and Mental Health, 250 College Street, Toronto, ON, Canada,Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, 250 College Street, Toronto, ON, Canada,Research Imaging Centre, Centre for Addiction and Mental Health, 250 College Street, Toronto, ON, Canada
| | - Bernard Le Foll
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, 250 College Street, Toronto, ON, Canada,Department Pharmacology & Toxicology, University of Toronto, Toronto, Canada,Department Psychiatry, University of Toronto, Toronto, Canada,Institute of Medical Sciences, University of Toronto, Toronto, Canada
| | - Pablo Rusjan
- Research Imaging Centre, Centre for Addiction and Mental Health, 250 College Street, Toronto, ON, Canada,Department Psychiatry, University of Toronto, Toronto, Canada
| | - Romina Mizrahi
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, 250 College Street, Toronto, ON, Canada,Research Imaging Centre, Centre for Addiction and Mental Health, 250 College Street, Toronto, ON, Canada,Department Psychiatry, University of Toronto, Toronto, Canada,Institute of Medical Sciences, University of Toronto, Toronto, Canada
| | - Rachel F. Tyndale
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, 250 College Street, Toronto, ON, Canada,Department Pharmacology & Toxicology, University of Toronto, Toronto, Canada,Department Psychiatry, University of Toronto, Toronto, Canada
| | - Marilyn A. Huestis
- Chemistry and Drug Metabolism, IRP, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, USA
| | - Doris E. Payer
- Addiction Imaging Research Group, Centre for Addiction and Mental Health, 250 College Street, Toronto, ON, Canada,Human Brain Lab, Centre for Addiction and Mental Health, 250 College Street, Toronto, ON, Canada,Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, 250 College Street, Toronto, ON, Canada,Research Imaging Centre, Centre for Addiction and Mental Health, 250 College Street, Toronto, ON, Canada,Department Psychiatry, University of Toronto, Toronto, Canada
| | - Alan A. Wilson
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, 250 College Street, Toronto, ON, Canada,Research Imaging Centre, Centre for Addiction and Mental Health, 250 College Street, Toronto, ON, Canada
| | - Sylvain Houle
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, 250 College Street, Toronto, ON, Canada,Research Imaging Centre, Centre for Addiction and Mental Health, 250 College Street, Toronto, ON, Canada
| | - Stephen J. Kish
- Human Brain Lab, Centre for Addiction and Mental Health, 250 College Street, Toronto, ON, Canada,Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, 250 College Street, Toronto, ON, Canada,Research Imaging Centre, Centre for Addiction and Mental Health, 250 College Street, Toronto, ON, Canada,Department Pharmacology & Toxicology, University of Toronto, Toronto, Canada,Department Psychiatry, University of Toronto, Toronto, Canada,Institute of Medical Sciences, University of Toronto, Toronto, Canada
| | - Junchao Tong
- Human Brain Lab, Centre for Addiction and Mental Health, 250 College Street, Toronto, ON, Canada,Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, 250 College Street, Toronto, ON, Canada,Research Imaging Centre, Centre for Addiction and Mental Health, 250 College Street, Toronto, ON, Canada,Department Psychiatry, University of Toronto, Toronto, Canada
| |
Collapse
|