301
|
Mortimer TL, Mabin T, Engelbrecht AM. Cannabinoids: the lows and the highs of chemotherapy-induced nausea and vomiting. Future Oncol 2019; 15:1035-1049. [PMID: 30720344 DOI: 10.2217/fon-2018-0530] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Despite remaining one of the most widely abused drugs worldwide, Cannabis sativa exhibits remarkable medicinal properties. The phytocannabinoids, cannabidiol and Δ-9-tetrahydrocannabinol, reduce nausea and vomiting, particularly during chemotherapy. This is attributed to their ability to reduce the release of serotonin from enterochromaffin cells in the small intestine, which would otherwise orchestrate the vomiting reflex. Although there are many preclinical and clinical studies on the effects of Δ-9-tetrahydrocannabinol during nausea and vomiting, little is known about the role that cannabidiol plays in this scenario. Since cannabidiol does not induce psychotropic effects, in contrast to other cannabinoids, its use as an anti-emetic is of great interest. This review aims to summarize the available literature on cannabinoid use, with a specific focus on the nonpsychotropic drug cannabidiol, as well as the roles that cannabinoids play in preventing several other adverse side effects of chemotherapy including organ toxicity, pain and loss of appetite.
Collapse
Affiliation(s)
- Toni Leigh Mortimer
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch, 7600, South Africa
| | - Tom Mabin
- Department of Medicine, Division of Cardiology, University of Cape Town, Observatory, 7925, South Africa
| | - Anna-Mart Engelbrecht
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch, 7600, South Africa
| |
Collapse
|
302
|
Kotlar I, Rangel-López E, Colonnello A, Aguilera-Portillo G, Serratos IN, Galván-Arzate S, Pedraza-Chaverri J, Túnez I, Wajner M, Santamaría A. Anandamide Reduces the Toxic Synergism Exerted by Quinolinic Acid and Glutaric Acid in Rat Brain Neuronal Cells. Neuroscience 2019; 401:84-95. [PMID: 30668975 DOI: 10.1016/j.neuroscience.2019.01.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 01/07/2019] [Accepted: 01/11/2019] [Indexed: 01/26/2023]
Abstract
The endocannabinoid system (ECS) regulates several physiological processes in the Central Nervous System, including the modulation of neuronal excitability via activation of cannabinoid receptors (CBr). Both glutaric acid (GA) and quinolinic acid (QUIN) are endogenous metabolites that, under pathological conditions, recruit common toxic mechanisms. A synergistic effect between them has already been demonstrated, supporting potential implications for glutaric acidemia type I (GA I). Here we investigated the possible involvement of a cannabinoid component in the toxic model exerted by QUIN + GA in rat cortical slices and primary neuronal cell cultures. The effects of the CB1 receptor agonist anandamide (AEA), and the fatty acid amide hydrolase inhibitor URB597, were tested on cell viability in cortical brain slices and primary neuronal cultures exposed to QUIN, GA, or QUIN + GA. As a pre-treatment to the QUIN + GA condition, AEA prevented the loss of cell viability in both preparations. URB597 only protected in a moderate manner the cultured neuronal cells against the QUIN + GA-induced damage. The use of the CB1 receptor reverse agonist AM251 in both biological preparations prevented partially the protective effects exerted by AEA, thus suggesting a partial role of CB1 receptors in this toxic model. AEA also prevented the cell damage and apoptotic death induced by the synergic model in cell cultures. Altogether, these findings demonstrate a modulatory role of the ECS on the synergic toxic actions exerted by QUIN + GA, thus providing key information for the understanding of the pathophysiological events occurring in GA I.
Collapse
Affiliation(s)
- Ilan Kotlar
- Laboratorio de Aminoácidos Excitadores, Instituto Nacional de Neurología y Neurocirugía, S.S.A., Mexico City, Mexico
| | - Edgar Rangel-López
- Laboratorio de Aminoácidos Excitadores, Instituto Nacional de Neurología y Neurocirugía, S.S.A., Mexico City, Mexico
| | - Aline Colonnello
- Laboratorio de Aminoácidos Excitadores, Instituto Nacional de Neurología y Neurocirugía, S.S.A., Mexico City, Mexico
| | - Gabriela Aguilera-Portillo
- Laboratorio de Aminoácidos Excitadores, Instituto Nacional de Neurología y Neurocirugía, S.S.A., Mexico City, Mexico
| | - Iris N Serratos
- Departamento de Química, Universidad Autónoma Metropolitana-Iztapalapa, Mexico City, Mexico
| | - Sonia Galván-Arzate
- Departamento de Neuroquímica, Instituto Nacional de Neurología y Neurocirugía, S.S.A., Mexico City, Mexico
| | - José Pedraza-Chaverri
- Departamento de Biología, Facultad de Química, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Isaac Túnez
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina y Enfermería, Universidad de Córdoba, Cordoba, Spain
| | - Moacir Wajner
- Departamento de Bioquímica, Instituto de Ciências Básicas da Sáude, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil; Serviço de Genética Médica, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
| | - Abel Santamaría
- Laboratorio de Aminoácidos Excitadores, Instituto Nacional de Neurología y Neurocirugía, S.S.A., Mexico City, Mexico.
| |
Collapse
|
303
|
Luo D, Zhang Y, Yuan X, Pan Y, Yang L, Zhao Y, Zhuo R, Chen C, Peng L, Li W, Jin X, Zhou Y. Oleoylethanolamide inhibits glial activation via moudulating PPARα and promotes motor function recovery after brain ischemia. Pharmacol Res 2019; 141:530-540. [PMID: 30660821 DOI: 10.1016/j.phrs.2019.01.027] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 12/29/2018] [Accepted: 01/14/2019] [Indexed: 02/06/2023]
Abstract
Glial activation and scar formation impede the neurological function recovery after cerebral ischemia. Oleoylethanolamide (OEA), a bioactive lipid mediator, shows neuroprotection against acute brain ischemia, however, its long-term effect, especially on glial scar formation, has not been characterized. In this research, we investigate the effect of OEA on glial activation and scar formation after cerebral ischemia in vitro and in vivo experiments. Glial scar formation in vitro model was induced by transforming growth factor β1 (TGF-β1) in C6 glial cell culture, and experiment model in vivo was induced by middle cerebral artery occlusion (MCAO) in mice. The protein expressions of the markers of glial activation (S100β, GFAP, or pSmads) and glial scar (neurocan) were detected by Western blot and/or immunofluorescence staining; To evaluate the role of PPARɑ in the effect of OEA on glial activation, the PPARɑ antagonist GW6471 was used. Behavior tests were used to assay the effect of OEA on motor function recovery 14 days after brain ischemia in mice. Our results show that OEA (10-50 μM) concentration-dependently inhibited the upregulation of S100β, GFAP, pSmads and neurocan induced by TGF-β1 in C6 glial cells. At the same time, OEA promoted the protein expression and nuclear transportation of PPARɑ in glial cells. PPARα antagonist GW6471 abolished the effect of OEA on glial activation. In addition, we found that delay administration of OEA inhibited the astrocyte activation and promoted the recovery of motor function after brain ischemia in mice. These results indicate that OEA may be developed into a new candidate for attenuating astrocytic scar formation and improving motor function after ischemic stroke.
Collapse
Affiliation(s)
- Doudou Luo
- Department of Basic Medical Science, School of Medicine, Xiamen University, Xiamen, 361102, PR China; Key Laboratory of Chiral Drugs, Xiamen, 361102, PR China
| | - Yali Zhang
- Department of Basic Medical Science, School of Medicine, Xiamen University, Xiamen, 361102, PR China; Key Laboratory of Chiral Drugs, Xiamen, 361102, PR China; Medical College, Xuchang University, Xuchang, 461000, PR China
| | - Xiaoqian Yuan
- Department of Basic Medical Science, School of Medicine, Xiamen University, Xiamen, 361102, PR China; Key Laboratory of Chiral Drugs, Xiamen, 361102, PR China
| | - Yilin Pan
- Department of Basic Medical Science, School of Medicine, Xiamen University, Xiamen, 361102, PR China
| | - Lichao Yang
- Department of Basic Medical Science, School of Medicine, Xiamen University, Xiamen, 361102, PR China; Key Laboratory of Chiral Drugs, Xiamen, 361102, PR China
| | - Yun Zhao
- Department of Basic Medical Science, School of Medicine, Xiamen University, Xiamen, 361102, PR China; Key Laboratory of Chiral Drugs, Xiamen, 361102, PR China
| | - Rengong Zhuo
- Department of Basic Medical Science, School of Medicine, Xiamen University, Xiamen, 361102, PR China; Key Laboratory of Chiral Drugs, Xiamen, 361102, PR China
| | - Caixia Chen
- Department of Basic Medical Science, School of Medicine, Xiamen University, Xiamen, 361102, PR China; Key Laboratory of Chiral Drugs, Xiamen, 361102, PR China
| | - Lu Peng
- Department of Basic Medical Science, School of Medicine, Xiamen University, Xiamen, 361102, PR China; Key Laboratory of Chiral Drugs, Xiamen, 361102, PR China
| | - Wenjun Li
- Department of Basic Medical Science, School of Medicine, Xiamen University, Xiamen, 361102, PR China; Key Laboratory of Chiral Drugs, Xiamen, 361102, PR China
| | - Xin Jin
- Department of Basic Medical Science, School of Medicine, Xiamen University, Xiamen, 361102, PR China; Key Laboratory of Chiral Drugs, Xiamen, 361102, PR China.
| | - Yu Zhou
- Department of Basic Medical Science, School of Medicine, Xiamen University, Xiamen, 361102, PR China; Key Laboratory of Chiral Drugs, Xiamen, 361102, PR China.
| |
Collapse
|
304
|
Tung CW, Ho C, Hsu YC, Huang SC, Shih YH, Lin CL. MicroRNA-29a Attenuates Diabetic Glomerular Injury through Modulating Cannabinoid Receptor 1 Signaling. Molecules 2019; 24:molecules24020264. [PMID: 30642005 PMCID: PMC6359641 DOI: 10.3390/molecules24020264] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 01/01/2019] [Accepted: 01/07/2019] [Indexed: 12/25/2022] Open
Abstract
Diabetic nephropathy often leads to end-stage renal disease and life-threatening morbidities. Simple control of risk factors is insufficient to prevent the progression of diabetic nephropathy, hence the need for discovering new treatments is of paramount importance. Recently, the dysregulation of microRNAs or the cannabinoid signaling pathway has been implicated in the pathogenesis of various renal tubulointerstitial fibrotic damages and thus novel therapeutic targets for chronic kidney diseases have emerged; however, the role of microRNAs or cannabinoid receptors on diabetes-induced glomerular injuries remains to be elucidated. In high-glucose-stressed renal mesangial cells, transfection of a miR-29a precursor sufficiently suppressed the mRNA and protein expressions of cannabinoid type 1 receptor (CB1R). Our data also revealed upregulated CB1R, interleukin-1β, interleukin-6, tumor necrosis factor-α, c-Jun, and type 4 collagen in the glomeruli of streptozotocin (STZ)-induced diabetic mice, whereas the expression of peroxisome proliferator-activated receptor-γ (PPAR-γ) was decreased. Importantly, using gain-of-function transgenic mice, we demonstrated that miR-29a acts as a negative regulator of CB1R, blocks the expressions of these proinflammatory and profibrogenic mediators, and attenuates renal hypertrophy. We also showed that overexpression of miR-29a restored PPAR-γ signaling in the renal glomeruli of diabetic animals. Collectively, our findings indicate that the interaction between miR-29a, CB1R, and PPAR-γ may play an important role in protecting diabetic renal glomeruli from fibrotic injuries.
Collapse
Affiliation(s)
- Chun-Wu Tung
- Department of Nephrology, Chang Gung Memorial Hospital, Chiayi 61363, Taiwan.
- Kidney and Diabetic Complications Research Team (KDCRT), Chang Gung Memorial Hospital, Chiayi 61363, Taiwan.
| | - Cheng Ho
- Kidney and Diabetic Complications Research Team (KDCRT), Chang Gung Memorial Hospital, Chiayi 61363, Taiwan.
- Division of Endocrinology and Metabolism, Chang Gung Memorial Hospital, Chiayi 61363, Taiwan.
| | - Yung-Chien Hsu
- Department of Nephrology, Chang Gung Memorial Hospital, Chiayi 61363, Taiwan.
- Kidney and Diabetic Complications Research Team (KDCRT), Chang Gung Memorial Hospital, Chiayi 61363, Taiwan.
| | - Shun-Chen Huang
- Department of Anatomic Pathology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University, College of Medicine, Kaohsiung 83301, Taiwan.
| | - Ya-Hsueh Shih
- Department of Nephrology, Chang Gung Memorial Hospital, Chiayi 61363, Taiwan.
- Kidney and Diabetic Complications Research Team (KDCRT), Chang Gung Memorial Hospital, Chiayi 61363, Taiwan.
| | - Chun-Liang Lin
- Department of Nephrology, Chang Gung Memorial Hospital, Chiayi 61363, Taiwan.
- Kidney and Diabetic Complications Research Team (KDCRT), Chang Gung Memorial Hospital, Chiayi 61363, Taiwan.
- 10507, Taiwan.
- College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan.
- Center for Shockwave Medicine and Tissue Engineering, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University, College of Medicine, Kaohsiung 83301, Taiwan.
| |
Collapse
|
305
|
Mitchell CM, El Jordi O, Yamamoto BK. Inflammatory mechanisms of abused drugs. ROLE OF INFLAMMATION IN ENVIRONMENTAL NEUROTOXICITY 2019. [DOI: 10.1016/bs.ant.2018.10.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
306
|
Machado MMF, Bassani TB, Cóppola-Segovia V, Moura ELR, Zanata SM, Andreatini R, Vital MABF. PPAR-γ agonist pioglitazone reduces microglial proliferation and NF-κB activation in the substantia nigra in the 6-hydroxydopamine model of Parkinson's disease. Pharmacol Rep 2018; 71:556-564. [PMID: 31132685 DOI: 10.1016/j.pharep.2018.11.005] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 09/14/2018] [Accepted: 11/29/2018] [Indexed: 01/29/2023]
Abstract
BACKGROUND Peroxisome proliferator-activated receptor γ (PPAR-γ) agonists have received much attention in research because of their neuroprotective and anti-inflammatory effects that reduce cell death and halt the progression of neurodegeneration. Thus, this study observed the pioglitazone effects on the main inflammatory markers after 6-hydroxydopamine (6-OHDA) lesion. METHODS The effects of a 5-day administration of the PPAR-γ agonist pioglitazone (30 mg/kg) in male Wistar rats that received bilateral intranigral infusions of 6-OHDA. After surgery, the rats were evaluated in the open-field test on days 1,7,14, and 21. Immediately after the behavioral tests on day 21, the rats were euthanized, and the substantia nigra was removed to analyze the expression of nuclear factor κB (NF-κB) and IκB by western blot. To immunohistochemical, animals were intracardially perfused, with brain removal that was frozen and sectioned, being selected slices of the SNc region to detect tyrosine hydroxylase (TH) immunoreactivity, microglia activation (Iba-1) and NF-κB translocation in the nucleus. RESULTS Pioglitazone protected rats against hypolocomotion and 6-OHDA-induced dopaminergic neurodegeneration on day 7. Decreases in the microglial activation and the NF-κB expression were observed, and the p65 activation was inhibited. CONCLUSIONS These results suggest that pioglitazone may be a potential adjuvant for the treatment of Parkinson`s disease because of its effects on pathological markers of the progression of neurodegeneration.
Collapse
Affiliation(s)
| | | | | | | | | | - Roberto Andreatini
- Department of Pharmacology, Federal University of Paraná, Curitiba, PR, Brazil
| | | |
Collapse
|
307
|
Vučković S, Srebro D, Vujović KS, Vučetić Č, Prostran M. Cannabinoids and Pain: New Insights From Old Molecules. Front Pharmacol 2018; 9:1259. [PMID: 30542280 PMCID: PMC6277878 DOI: 10.3389/fphar.2018.01259] [Citation(s) in RCA: 178] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 10/15/2018] [Indexed: 01/11/2023] Open
Abstract
Cannabis has been used for medicinal purposes for thousands of years. The prohibition of cannabis in the middle of the 20th century has arrested cannabis research. In recent years there is a growing debate about the use of cannabis for medical purposes. The term ‘medical cannabis’ refers to physician-recommended use of the cannabis plant and its components, called cannabinoids, to treat disease or improve symptoms. Chronic pain is the most commonly cited reason for using medical cannabis. Cannabinoids act via cannabinoid receptors, but they also affect the activities of many other receptors, ion channels and enzymes. Preclinical studies in animals using both pharmacological and genetic approaches have increased our understanding of the mechanisms of cannabinoid-induced analgesia and provided therapeutical strategies for treating pain in humans. The mechanisms of the analgesic effect of cannabinoids include inhibition of the release of neurotransmitters and neuropeptides from presynaptic nerve endings, modulation of postsynaptic neuron excitability, activation of descending inhibitory pain pathways, and reduction of neural inflammation. Recent meta-analyses of clinical trials that have examined the use of medical cannabis in chronic pain present a moderate amount of evidence that cannabis/cannabinoids exhibit analgesic activity, especially in neuropathic pain. The main limitations of these studies are short treatment duration, small numbers of patients, heterogeneous patient populations, examination of different cannabinoids, different doses, the use of different efficacy endpoints, as well as modest observable effects. Adverse effects in the short-term medical use of cannabis are generally mild to moderate, well tolerated and transient. However, there are scant data regarding the long-term safety of medical cannabis use. Larger well-designed studies of longer duration are mandatory to determine the long-term efficacy and long-term safety of cannabis/cannabinoids and to provide definitive answers to physicians and patients regarding the risk and benefits of its use in the treatment of pain. In conclusion, the evidence from current research supports the use of medical cannabis in the treatment of chronic pain in adults. Careful follow-up and monitoring of patients using cannabis/cannabinoids are mandatory.
Collapse
Affiliation(s)
- Sonja Vučković
- Department of Pharmacology, Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Dragana Srebro
- Department of Pharmacology, Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Katarina Savić Vujović
- Department of Pharmacology, Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Čedomir Vučetić
- Clinic of Orthopaedic Surgery and Traumatology, Clinical Center of Serbia, Belgrade, Serbia.,Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Milica Prostran
- Department of Pharmacology, Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
308
|
Antón M, Rodríguez-González A, Rodríguez-Rojo IC, Pastor A, Correas Á, Serrano A, Ballesta A, Alén F, Gómez de Heras R, de la Torre R, Rodríguez de Fonseca F, Orio L. Increased plasma oleoylethanolamide and palmitoleoylethanolamide levels correlate with inflammatory changes in alcohol binge drinkers: the case of HMGB1 in women. Addict Biol 2018; 23:1242-1250. [PMID: 29178411 DOI: 10.1111/adb.12580] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Accepted: 10/19/2017] [Indexed: 01/15/2023]
Abstract
Alcohol binge drinking is a heavy pattern of alcohol consumption increasingly used by young people. In a previous study, we reported that young drinkers with a 2-year history of binge alcohol consumption had an overactivation of the innate immune system and peripheral inflammation when compared with controls. In the present study, we measured several biolipids that are fatty acid derivatives belonging to the acylethanolamide or 2-acylglycerol families in the plasma of the same subjects (n = 42; 20 men and 22 women). We found that during abstinence, alcohol binge drinkers had elevated plasma levels of oleoylethanolamide, palmitoleoylethanolamide, arachidonoylethanolamide, dihomo-γ-linolenoyl ethanolamide and linoleoyl ethanolamide, which positively correlated with changes in the mRNA expression of key inflammatory markers in peripheral blood mononuclear cells, such as toll-like receptors (TLR4), pro-inflammatory cytokines/chemokines interleukin-1 beta, interleukin-6 and monocyte chemoattractant protein-1, and cyclooxygenase-2. Additionally, plasma oleoylethanolamide positively correlated with plasma levels of high mobility group box-1, which is a danger-associated molecular pattern and an endogenous TLR4 agonist, specifically in female alcohol binge drinkers. No changes were observed in 2-acylglycerols in alcohol binge drinkers, although sex-related differences in these bioactive lipids as well as in palmitoleoylethanolamide and docosatetraenoylethanolamide levels were detected. These results extend the previous clinical findings observed in patients diagnosed with long-term alcohol use disorder to young users and suggest a prominent role for these lipids in the response to acute alcohol exposure.
Collapse
Affiliation(s)
- María Antón
- Departamento de Psicobiología, Facultad de Psicología; Universidad Complutense de Madrid; Spain
| | | | | | - Antoni Pastor
- Programa de Neurociencias; Institut Hospital del Mar d'Investigacions Mèdiques (IMIM); Spain
| | - Ángeles Correas
- Laboratorio de Neurociencia Cognitiva y Computacional; Centro de Tecnología Biomédica (CTB); Spain
| | - Antonia Serrano
- Red de Trastornos Adictivos (RTA) del Instituto de Salud Carlos III (ISCIII); Spain
- Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica (IBIMA); Hospital Regional Universitario de Málaga; Spain
| | - Antonio Ballesta
- Departamento de Psicobiología, Facultad de Psicología; Universidad Complutense de Madrid; Spain
| | - Francisco Alén
- Departamento de Psicobiología, Facultad de Psicología; Universidad Complutense de Madrid; Spain
| | - Raquel Gómez de Heras
- Departamento de Psicobiología, Facultad de Psicología; Universidad Complutense de Madrid; Spain
| | - Rafael de la Torre
- Programa de Neurociencias; Institut Hospital del Mar d'Investigacions Mèdiques (IMIM); Spain
| | - Fernando Rodríguez de Fonseca
- Departamento de Psicobiología, Facultad de Psicología; Universidad Complutense de Madrid; Spain
- Red de Trastornos Adictivos (RTA) del Instituto de Salud Carlos III (ISCIII); Spain
- Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica (IBIMA); Hospital Regional Universitario de Málaga; Spain
| | - Laura Orio
- Departamento de Psicobiología, Facultad de Psicología; Universidad Complutense de Madrid; Spain
- Red de Trastornos Adictivos (RTA) del Instituto de Salud Carlos III (ISCIII); Spain
| |
Collapse
|
309
|
Huang H, McIntosh AL, Martin GG, Dangott LJ, Kier AB, Schroeder F. Structural and Functional Interaction of Δ 9-Tetrahydrocannabinol with Liver Fatty Acid Binding Protein (FABP1). Biochemistry 2018; 57:6027-6042. [PMID: 30232874 DOI: 10.1021/acs.biochem.8b00744] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Although serum Δ9-tetrahydrocannabinol (Δ9-THC) undergoes rapid hepatic clearance and metabolism, almost nothing is known regarding the mechanism(s) whereby this highly lipophilic phytocannabinoid is transported for metabolism/excretion. A novel NBD-arachidonoylethanolamide (NBD-AEA) fluorescence displacement assay showed that liver fatty acid binding protein (FABP1), the major hepatic endocannabinoid (EC) binding protein, binds the first major metabolite of Δ9-THC (Δ9-THC-OH) as well as Δ9-THC itself. Circular dichroism (CD) confirmed that not only Δ9-THC and Δ9-THC-OH but also downstream metabolites Δ9-THC-COOH and Δ9-THC-CO-glucuronide directly interact with FABP1. Δ9-THC and metabolite interaction differentially altered the FABP1 secondary structure, increasing total α-helix (all), decreasing total β-sheet (Δ9-THC-COOH, Δ9-THC-CO-glucuronide), increasing turns (Δ9-THC-OH, Δ9-THC-COOH, Δ9-THC-CO-glucuronide), and decreasing unordered structure (Δ9-THC, Δ9-THC-OH). Cultured primary hepatocytes from wild-type (WT) mice took up and converted Δ9-THC to the above metabolites. Fabp1 gene ablation (LKO) dramatically increased hepatocyte accumulation of Δ9-THC and even more so its primary metabolites Δ9-THC-OH and Δ9-THC-COOH. Concomitantly, rtPCR and Western blotting indicated that LKO significantly increased Δ9-THC's ability to regulate downstream nuclear receptor transcription of genes important in both EC ( Napepld > Daglb > Dagla, Naaa, Cnr1) and lipid ( Cpt1A > Fasn, FATP4) metabolism. Taken together, the data indicated that FABP1 may play important roles in Δ9-THC uptake and elimination as well as Δ9-THC induction of genes regulating hepatic EC levels and downstream targets in lipid metabolism.
Collapse
Affiliation(s)
- Huan Huang
- Department of Physiology and Pharmacology , Texas A&M University , College Station , Texas 77843-4466 , United States
| | - Avery L McIntosh
- Department of Physiology and Pharmacology , Texas A&M University , College Station , Texas 77843-4466 , United States
| | - Gregory G Martin
- Department of Physiology and Pharmacology , Texas A&M University , College Station , Texas 77843-4466 , United States
| | - Lawrence J Dangott
- Protein Chemistry Laboratory , Texas A&M University , College Station , Texas 77843-2128 , United States
| | - Ann B Kier
- Department of Pathobiology , Texas A&M University , College Station , Texas 77843-4467 , United States
| | - Friedhelm Schroeder
- Department of Physiology and Pharmacology , Texas A&M University , College Station , Texas 77843-4466 , United States
| |
Collapse
|
310
|
Hamid Z, Summa M, Armirotti A. A Swath Label-Free Proteomics insight into the Faah -/- Mouse Liver. Sci Rep 2018; 8:12142. [PMID: 30108271 PMCID: PMC6092373 DOI: 10.1038/s41598-018-30553-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 08/01/2018] [Indexed: 12/31/2022] Open
Abstract
Fatty acid amide hydrolase (FAAH) is an important enzyme for lipid metabolism and an interesting pharmacological target, given its role in anandamide breakdown. The FAAH−/− genotype is the most widely used mouse model to investigate the effects of a complete pharmacological inhibition of this enzyme. In this paper, we explore, by means of label-free SWATH proteomics, the changes in protein expression occurring in the liver of FAAH−/− knockout (KO) mice. We identified several altered biological processes and pathways, like fatty acid synthesis and glycolysis, which explain the observed phenotype of this mouse. We also observed the alteration of other proteins, like carboxylesterases and S-methyltransferases, apparently not immediately related to FAAH, but known to have important biological roles. Our study, reporting more than 3000 quantified proteins, offers an in-depth analysis of the liver proteome of this model.
Collapse
Affiliation(s)
- Zeeshan Hamid
- D3Validation, Fondazione Istituto Italiano di Tecnologia, via Morego 30, 16163, Genova, Italy.,Scuola Superiore Sant'Anna. via Piazza Martiri della Libertà, 33, 56127, Pisa, Italy
| | - Maria Summa
- Analytical Chemistry and In-vivo Facility, Fondazione Istituto Italiano di Tecnologia, via Morego 30, 16163, Genova, Italy
| | - Andrea Armirotti
- Analytical Chemistry and In-vivo Facility, Fondazione Istituto Italiano di Tecnologia, via Morego 30, 16163, Genova, Italy.
| |
Collapse
|
311
|
Gugliandolo A, Pollastro F, Grassi G, Bramanti P, Mazzon E. In Vitro Model of Neuroinflammation: Efficacy of Cannabigerol, a Non-Psychoactive Cannabinoid. Int J Mol Sci 2018; 19:ijms19071992. [PMID: 29986533 PMCID: PMC6073490 DOI: 10.3390/ijms19071992] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 07/02/2018] [Accepted: 07/06/2018] [Indexed: 12/20/2022] Open
Abstract
Inflammation and oxidative stress play main roles in neurodegeneration. Interestingly, different natural compounds may be able to exert neuroprotective actions against inflammation and oxidative stress, protecting from neuronal cell loss. Among these natural sources, Cannabis sativa represents a reservoir of compounds exerting beneficial properties, including cannabigerol (CBG), whose antioxidant properties have already been demonstrated in macrophages. Here, we aimed to evaluate the ability of CBG to protect NSC-34 motor neurons against the toxicity induced from the medium of LPS-stimulated RAW 264.7 macrophages. Using MTT assay, we observed that CBG pre-treatment was able to reduce the loss of cell viability induced by the medium of LPS-stimulated macrophages in NSC-34 cells. Indeed, CBG pre-treatment inhibited apoptosis, as shown by the reduction of caspase 3 activation and Bax expression, while Bcl-2 levels increased. Furthermore, CBG pre-treatment counteracted not only inflammation, as demonstrated by the reduction of IL-1β, TNF-α, IFN-γ and PPARγ protein levels assessed by immunocytochemistry, but also oxidative stress in NSC-34 cells treated with the medium of LPS-stimulated RAW 264.7. Indeed, immunocytochemistry showed that CBG pre-treatment reduced nitrotyrosine, SOD1 and iNOS protein levels and restored Nrf-2 levels. All together, these results indicated the neuroprotective effects of CBG, that may be a potential treatment against neuroinflammation and oxidative stress.
Collapse
Affiliation(s)
| | - Federica Pollastro
- Department of Pharmaceutical Sciences, University of Eastern Piedmont "Amedeo Avogadro", 28100 Novara, Italy.
| | - Gianpaolo Grassi
- Research Centre for Industrial Crops, Council for Agricultural Research and Economics (CREA-CIN), 45100 Rovigo, Italy.
| | | | - Emanuela Mazzon
- IRCCS Centro Neurolesi "Bonino Pulejo", 98124 Messina, Italy.
| |
Collapse
|
312
|
Grill M, Hasenoehrl C, Storr M, Schicho R. Medical Cannabis and Cannabinoids: An Option for the Treatment of Inflammatory Bowel Disease and Cancer of the Colon? Med Cannabis Cannabinoids 2018; 1:28-35. [PMID: 34676319 DOI: 10.1159/000489036] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 04/09/2018] [Indexed: 12/11/2022] Open
Abstract
In the past few years, we have witnessed a surge of new reports dealing with the role of cannabinoids, synthetic as well as herbal, in the mechanisms of inflammation and carcinogenesis. However, despite the wealth of in vitro data and anecdotal reports, evidence that cannabinoids could act as beneficial drugs in inflammatory bowel disease (IBD) or in neoplastic development of the human gastrointestinal tract is lacking. Some insight into the effects of medical Cannabis (usually meaning dried flowers) and cannabinoids in IBD has been gained through questionnaires and small pilot studies. As to colorectal cancer, only preclinical data are available. Currently, Δ9-tetrahydrocannabinol (THC) and its synthetic forms, dronabinol and nabilone, are used as an add-on treatment to alleviate chronic pain and spasticity in multiple sclerosis patients as well as chemotherapy-induced nausea. The use of medical Cannabis is authorized only in a limited number of countries. None of the mentioned substances are currently indicated for IBD. This review is an update of our knowledge on the role of cannabinoids in intestinal inflammation and carcinogenesis and a discussion on their potential therapeutic use.
Collapse
Affiliation(s)
- Magdalena Grill
- Otto Loewi Research Center, Pharmacology Section, Medical University of Graz, Graz, Austria
| | - Carina Hasenoehrl
- Otto Loewi Research Center, Pharmacology Section, Medical University of Graz, Graz, Austria
| | - Martin Storr
- Department of Medicine 2, Ludwig-Maximilians University, Munich, Germany.,Zentrum für Endoskopie, Starnberg, Germany
| | - Rudolf Schicho
- Otto Loewi Research Center, Pharmacology Section, Medical University of Graz, Graz, Austria.,BioTechMed, Graz, Austria
| |
Collapse
|
313
|
Localization of cannabinoid receptors CB1, CB2, GPR55, and PPARα in the canine gastrointestinal tract. Histochem Cell Biol 2018; 150:187-205. [DOI: 10.1007/s00418-018-1684-7] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/02/2018] [Indexed: 12/26/2022]
|
314
|
Zhang J, Chen C. Alleviation of Neuropathology by Inhibition of Monoacylglycerol Lipase in APP Transgenic Mice Lacking CB2 Receptors. Mol Neurobiol 2018; 55:4802-4810. [PMID: 28733897 PMCID: PMC5776068 DOI: 10.1007/s12035-017-0689-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Accepted: 07/12/2017] [Indexed: 01/25/2023]
Abstract
Inhibition of monoacylglycerol lipase (MAGL), the primary enzyme that hydrolyzes the endocannabinoid 2-arachidonoylglycerol (2-AG) in the brain, produces profound anti-inflammatory and neuroprotective effects and improves synaptic and cognitive functions in animal models of Alzheimer's disease (AD). However, the molecular mechanisms underlying the beneficial effects produced by inhibition of 2-AG metabolism are still not clear. The cannabinoid receptor type 2 (CB2R) has been thought to be a therapeutic target for AD. Here, we provide evidence, however, that CB2R does not play a role in ameliorating AD neuropathology produced by inactivation of MAGL in 5XFAD APP transgenic mice, an animal model of AD. We observed that expression of APP and β-secretase as well as production of total Aβ and Aβ42 were significantly reduced in APP transgenic mice lacking CB2R (TG-CB2-KO) treated with JZL184, a selective and potent inhibitor for MAGL. Inactivation of MAGL also alleviated neuroinflammation and neurodegeneration in TG-CB2-KO mice. Importantly, TG-CB2-KO mice treated with JZL184 still exhibited improvements in spatial learning and memory. In addition, MAGL inhibition prevented deterioration in expression of important synaptic proteins in TG-CB2-KO mice. Our results suggest that CB2R is not required in ameliorating neuropathology and preventing cognitive decline by inhibition of 2-AG metabolism in AD model animals.
Collapse
Affiliation(s)
- Jian Zhang
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health Sciences Center, 2020 Gravier Street, Suite D, New Orleans, LA, 70112, USA
| | - Chu Chen
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health Sciences Center, 2020 Gravier Street, Suite D, New Orleans, LA, 70112, USA.
- Department of Otolaryngology-Head and Neck Surgery, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, 70112, USA.
| |
Collapse
|
315
|
Aguilera-Portillo G, Rangel-López E, Villeda-Hernández J, Chavarría A, Castellanos P, Elmazoglu Z, Karasu Ç, Túnez I, Pedraza G, Königsberg M, Santamaría A. The Pharmacological Inhibition of Fatty Acid Amide Hydrolase Prevents Excitotoxic Damage in the Rat Striatum: Possible Involvement of CB1 Receptors Regulation. Mol Neurobiol 2018; 56:844-856. [DOI: 10.1007/s12035-018-1129-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 05/14/2018] [Indexed: 02/06/2023]
|
316
|
Chen HJC, Spiers JG, Sernia C, Lavidis NA. Inhibition of Fatty Acid Amide Hydrolase by PF-3845 Alleviates the Nitrergic and Proinflammatory Response in Rat Hippocampus Following Acute Stress. Int J Neuropsychopharmacol 2018; 21:786-795. [PMID: 29579222 PMCID: PMC6070085 DOI: 10.1093/ijnp/pyy033] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 03/15/2018] [Accepted: 03/21/2018] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Long-term exposure to stress has been demonstrated to cause neuroinflammation through a sustained overproduction of free radicals, including nitric oxide, via an increased inducible nitric oxide synthase activity. We previously demonstrated that inducible nitric oxide synthase activity and mRNA are significantly upregulated in the rat hippocampus following just 4 hours of restraint stress. Similar to nitric oxide, endocannabinoids are synthesized on demand, with preclinical observations suggesting that cannabinoid receptor agonists and endocannabinoid enhancers inhibit nitrergic activity. Specifically, previous work has shown that enhancement of endocannabinoids via inhibition of fatty acid amide hydrolase with PF-3845 reduced inducible nitric oxide synthase-expressing microglia following traumatic brain injury. However, this describes cannabinoid modulation following physical injury, and therefore the present study aimed to examine the effects of PF-3845 in the modulation of nitrergic and inflammatory-related genes within the hippocampus after acute stress exposure. METHODS Following vehicle or PF-3845 injections (5 mg/kg; i.p.), male Wistar rats were exposed to 0 (control), 60, 240, or 360 minutes of restraint stress after which plasma and dorsal hippocampus were isolated for further biochemical and gene expression analysis. RESULTS The results demonstrate that pretreatment with PF-3845 rapidly ameliorates plasma corticosterone release at 60 minutes of stress. An increase in endocannabinoid signalling also induces an overall attenuation in inducible nitric oxide synthase, tumor necrosis factor-alpha convertase, interleukin-6, cyclooxygenase-2, peroxisome proliferator-activated receptor gamma mRNA, and the transactivation potential of nuclear factor kappa-light-chain-enhancer of activated B cells in the hippocampus. CONCLUSIONS These results suggest that enhanced endocannabinoid levels in the dorsal hippocampus have an overall antinitrosative and antiinflammatory effect following acute stress exposure.
Collapse
Affiliation(s)
- Hsiao-Jou Cortina Chen
- School of Biomedical Sciences, The University of Queensland, St Lucia, Australia,Correspondence: Hsiao-Jou Cortina Chen, PhD, School of Biomedical Sciences, The University of Queensland, St Lucia, 4072, Australia ()
| | - Jereme G Spiers
- MRC Toxicology Unit, University of Leicester, Leicester, United Kingdom
| | - Conrad Sernia
- School of Biomedical Sciences, The University of Queensland, St Lucia, Australia
| | - Nickolas A Lavidis
- School of Biomedical Sciences, The University of Queensland, St Lucia, Australia
| |
Collapse
|
317
|
Bottemanne P, Muccioli GG, Alhouayek M. N-acylethanolamine hydrolyzing acid amidase inhibition: tools and potential therapeutic opportunities. Drug Discov Today 2018; 23:1520-1529. [PMID: 29567427 DOI: 10.1016/j.drudis.2018.03.007] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Revised: 03/06/2018] [Accepted: 03/15/2018] [Indexed: 01/12/2023]
Abstract
N-acylethanolamines (NAEs) (e.g., N-palmitoylethanolamine, N-arachidonoylethanolamine, N-oleoylethanolamine) are bioactive lipids involved in many physiological processes including pain, inflammation, anxiety, cognition and food intake. Two enzymes are responsible for the hydrolysis of NAEs and therefore regulate their endogenous levels and effects: fatty acid amide hydrolase (FAAH) and N-acylethanolamine-hydrolyzing acid amidase (NAAA). As discussed here, extensive biochemical characterization of NAAA was carried out over the years that contributed to a better understanding of NAAA enzymology. An increasing number of studies describe the synthesis and pharmacological characterization of NAAA inhibitors. Recent medicinal chemistry efforts have led to the development of potent and stable inhibitors that enable studying the effects of NAAA inhibition in preclinical disease models, notably in the context of pain and inflammation.
Collapse
Affiliation(s)
- Pauline Bottemanne
- BPBL Research Group, Louvain Drug Research Institute, Université catholique de Louvain, Av. E. Mounier 72, B1.72.01, B-1200 Bruxelles, Belgium
| | - Giulio G Muccioli
- BPBL Research Group, Louvain Drug Research Institute, Université catholique de Louvain, Av. E. Mounier 72, B1.72.01, B-1200 Bruxelles, Belgium
| | - Mireille Alhouayek
- BPBL Research Group, Louvain Drug Research Institute, Université catholique de Louvain, Av. E. Mounier 72, B1.72.01, B-1200 Bruxelles, Belgium.
| |
Collapse
|
318
|
Ullah I, Subhan F, Alam J, Shahid M, Ayaz M. Suppression of Cisplatin-Induced Vomiting by Cannabis sativa in Pigeons: Neurochemical Evidences. Front Pharmacol 2018; 9:231. [PMID: 29615907 PMCID: PMC5865282 DOI: 10.3389/fphar.2018.00231] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 02/28/2018] [Indexed: 01/03/2023] Open
Abstract
Cannabis sativa (CS, family Cannabinaceae) has been reported for its anti-emetic activity against cancer chemotherapy-induced emesis in animal models and in clinics. The current study was designed to investigate CS for potential effectiveness to attenuate cisplatin-induced vomiting in healthy pigeons and to study the impact on neurotransmitters involved centrally and peripherally in the act of vomiting. High-performance liquid chromatography system coupled with electrochemical detector was used for the quantification of neurotransmitters 5-hydroxytryptamine (5HT), dopamine (DA) and their metabolites; Di-hydroxy Phenyl Acetic acid (Dopac), Homovanillic acid (HVA), and 5-hydroxy indole acetic acid (5HIAA) centrally in specific brain areas (area postrema and brain stem) while, peripherally in small intestine. Cisplatin (7 mg/kg i.v.) induce emesis without lethality across the 24 h observation period. CS hexane fraction (CS-HexFr; 10 mg/kg) attenuated cisplatin-induced emesis ∼ 65.85% (P < 0.05); the reference anti-emetic drug, metoclopramide (MCP; 30 mg/kg), produced ∼43.90% reduction (P < 0.05). At acute time point (3rd h), CS-HexFr decreased (P < 0.001) the concentration of 5HT and 5HIAA in the area postrema, brain stem and intestine, while at 18th h (delayed time point) CS-HexFr attenuated (P < 0.001) the upsurge of 5HT caused by cisplatin in the brain stem and intestine and dopamine in the area postrema. CS-HexFr treatment alone did not alter the basal neurotransmitters and their metabolites in the brain areas and intestine except 5HIAA and HVA, which were decreased significantly. In conclusion the anti-emetic effect of CS-HexFr is mediated by anti-serotonergic and anti-dopaminergic components in a blended manner at the two different time points, i.e., 3rd and 18th h in pigeons.
Collapse
Affiliation(s)
- Ihsan Ullah
- Department of Pharmacy, University of Swabi, Swabi, Pakistan.,Department of Pharmacy, University of Peshawar, Peshawar, Pakistan
| | - Fazal Subhan
- Department of Pharmacy, University of Peshawar, Peshawar, Pakistan
| | - Javaid Alam
- Department of Pharmacy, University of Peshawar, Peshawar, Pakistan.,Drug and Herbal Research Centre, Faculty of Pharmacy, University Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Muhammad Shahid
- Department of Pharmacy, University of Peshawar, Peshawar, Pakistan
| | - Muhammad Ayaz
- Department of Pharmacy, University of Malakand, Chakdara, Pakistan
| |
Collapse
|
319
|
Gugnani KS, Vu N, Rondón-Ortiz AN, Böhlke M, Maher TJ, Pino-Figueroa AJ. Neuroprotective activity of macamides on manganese-induced mitochondrial disruption in U-87 MG glioblastoma cells. Toxicol Appl Pharmacol 2017; 340:67-76. [PMID: 29288688 DOI: 10.1016/j.taap.2017.12.014] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Revised: 12/09/2017] [Accepted: 12/23/2017] [Indexed: 02/07/2023]
Abstract
Macamides are a distinct class of secondary metabolites, benzylamides of long chain fatty acids, which were isolated from the Peruvian plant Lepidium meyenii (Maca). As structural analogues of the endocannabinoid anandamide (AEA), they have demonstrated neuroprotective effects in vitro and in vivo. The purpose of this study was to demonstrate the neuroprotective activity of the macamides: N-(3-methoxybenzyl)oleamide (MAC 18:1), N-(3-methoxybenzyl)linoleamide (MAC 18:2) and N-(3-methoxybenzyl)linolenamide (MAC 18:3) in a neurotoxic environment caused by exposure of U-87 MG glioblastoma cells to manganese chloride (MnCl2). The neuroprotective effects of these macamides were reversed by the CB1 antagonist AM251. The mechanism by which manganese (Mn) induces cell damage was investigated by studying its effects on mitochondria. Reactive oxygen species (ROS) increase intracellular calcium and enhance the opening of mitochondrial permeability transition pores (MPTP), which leads to decreased mitochondrial membrane potential (MMP), to disruption of mitochondria and to neuron death in neurodegenerative disorders. In this study, MnCl2 at 50μM was responsible for mitochondrial disruption, which was attenuated by all three of the macamides tested. Human peroxisome proliferator-activated receptor gamma (PPARγ) has been proposed to be a cannabinoid target, and PPARγ has also been demonstrated to mediate some of the longer-term vascular effects of the plant cannabinoid, ∆9-tetrahydrocannabinol. PPARγ activation was observed in response to exposures of cells to MAC 18:2 and MAC 18:3. These findings suggest that macamides achieve their neuroprotective effects by binding to CB1 receptors to protect against Mn-induced toxicity in U-87 MG glioblastoma cells. Additionally these macamides, in a manner similar to the analogous endocannabinoid AEA, interact with other targets such as PPARγ to regulate metabolism and energy homeostasis, cell differentiation and inflammation.
Collapse
Affiliation(s)
- Kuljeet S Gugnani
- Department of Pharmaceutical Sciences, MCPHS University, 179 Longwood Avenue, Boston, MA, USA
| | - Nguyen Vu
- School of Pharmacy, MCPHS University, 179 Longwood Avenue, Boston, MA, USA
| | | | - Mark Böhlke
- Department of Pharmaceutical Sciences, MCPHS University, 179 Longwood Avenue, Boston, MA, USA
| | - Timothy J Maher
- Department of Pharmaceutical Sciences, MCPHS University, 179 Longwood Avenue, Boston, MA, USA
| | | |
Collapse
|
320
|
Sierra S, Luquin N, Navarro-Otano J. The endocannabinoid system in cardiovascular function: novel insights and clinical implications. Clin Auton Res 2017; 28:35-52. [PMID: 29222605 DOI: 10.1007/s10286-017-0488-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 11/28/2017] [Indexed: 12/18/2022]
Abstract
RATIONALE Cardiovascular disease is now recognized as the number one cause of death in the world, and the size of the population at risk continues to increase rapidly. The dysregulation of the endocannabinoid (eCB) system plays a central role in a wide variety of conditions including cardiovascular disorders. Cannabinoid receptors, their endogenous ligands, as well as enzymes conferring their synthesis and degradation, exhibit overlapping distributions in the cardiovascular system. Furthermore, the pharmacological manipulation of the eCB system has effects on blood pressure, cardiac contractility, and endothelial vasomotor control. Growing evidence from animal studies supports the significance of the eCB system in cardiovascular disorders. OBJECTIVE To summarize the literature surrounding the eCB system in cardiovascular function and disease and the new compounds that may potentially extend the range of available interventions. RESULTS Drugs targeting CB1R, CB2R, TRPV1 and PPARs are proven effective in animal models mimicking cardiovascular disorders such as hypertension, atherosclerosis and myocardial infarction. Despite the setback of two clinical trials that exhibited unexpected harmful side-effects, preclinical studies are accelerating the development of more selective drugs with promising results devoid of adverse effects. CONCLUSION Over the last years, increasing evidence from basic and clinical research supports the role of the eCB system in cardiovascular function. Whereas new discoveries are paving the way for the identification of novel drugs and therapeutic targets, the close cooperation of researchers, clinicians and pharmaceutical companies is needed to achieve successful outcomes.
Collapse
Affiliation(s)
- Salvador Sierra
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA. .,Department of Physiology and Biophysics, Molecular Medicine Research Building, Virginia Commonwealth University, 1220 East Broad Street, Richmond, VA, 23298, USA.
| | - Natasha Luquin
- Department of Medical Genomics, Royal Prince Alfred Hospital, Sydney, Australia
| | - Judith Navarro-Otano
- Neurology Service, Electromyography, Motor Control and Neuropathic Pain Unit, Hospital Clínic de Barcelona, University of Barcelona, Barcelona, Spain
| |
Collapse
|
321
|
Oláh A, Szekanecz Z, Bíró T. Targeting Cannabinoid Signaling in the Immune System: "High"-ly Exciting Questions, Possibilities, and Challenges. Front Immunol 2017; 8:1487. [PMID: 29176975 PMCID: PMC5686045 DOI: 10.3389/fimmu.2017.01487] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Accepted: 10/23/2017] [Indexed: 12/21/2022] Open
Abstract
It is well known that certain active ingredients of the plants of Cannabis genus, i.e., the "phytocannabinoids" [pCBs; e.g., (-)-trans-Δ9-tetrahydrocannabinol (THC), (-)-cannabidiol, etc.] can influence a wide array of biological processes, and the human body is able to produce endogenous analogs of these substances ["endocannabinoids" (eCB), e.g., arachidonoylethanolamine (anandamide, AEA), 2-arachidonoylglycerol (2-AG), etc.]. These ligands, together with multiple receptors (e.g., CB1 and CB2 cannabinoid receptors, etc.), and a complex enzyme and transporter apparatus involved in the synthesis and degradation of the ligands constitute the endocannabinoid system (ECS), a recently emerging regulator of several physiological processes. The ECS is widely expressed in the human body, including several members of the innate and adaptive immune system, where eCBs, as well as several pCBs were shown to deeply influence immune functions thereby regulating inflammation, autoimmunity, antitumor, as well as antipathogen immune responses, etc. Based on this knowledge, many in vitro and in vivo studies aimed at exploiting the putative therapeutic potential of cannabinoid signaling in inflammation-accompanied diseases (e.g., multiple sclerosis) or in organ transplantation, and to dissect the complex immunological effects of medical and "recreational" marijuana consumption. Thus, the objective of the current article is (i) to summarize the most recent findings of the field; (ii) to highlight the putative therapeutic potential of targeting cannabinoid signaling; (iii) to identify open questions and key challenges; and (iv) to suggest promising future directions for cannabinoid-based drug development.
Collapse
Affiliation(s)
- Attila Oláh
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zoltán Szekanecz
- Department of Internal Medicine, Division of Rheumatology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Tamás Bíró
- Department of Immunology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
322
|
Woodhams SG, Chapman V, Finn DP, Hohmann AG, Neugebauer V. The cannabinoid system and pain. Neuropharmacology 2017; 124:105-120. [PMID: 28625720 PMCID: PMC5785108 DOI: 10.1016/j.neuropharm.2017.06.015] [Citation(s) in RCA: 188] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Revised: 05/31/2017] [Accepted: 06/14/2017] [Indexed: 01/20/2023]
Abstract
Chronic pain states are highly prevalent and yet poorly controlled by currently available analgesics, representing an enormous clinical, societal, and economic burden. Existing pain medications have significant limitations and adverse effects including tolerance, dependence, gastrointestinal dysfunction, cognitive impairment, and a narrow therapeutic window, making the search for novel analgesics ever more important. In this article, we review the role of an important endogenous pain control system, the endocannabinoid (EC) system, in the sensory, emotional, and cognitive aspects of pain. Herein, we briefly cover the discovery of the EC system and its role in pain processing pathways, before concentrating on three areas of current major interest in EC pain research; 1. Pharmacological enhancement of endocannabinoid activity (via blockade of EC metabolism or allosteric modulation of CB1receptors); 2. The EC System and stress-induced modulation of pain; and 3. The EC system & medial prefrontal cortex (mPFC) dysfunction in pain states. Whilst we focus predominantly on the preclinical data, we also include extensive discussion of recent clinical failures of endocannabinoid-related therapies, the future potential of these approaches, and important directions for future research on the EC system and pain. This article is part of the Special Issue entitled "A New Dawn in Cannabinoid Neurobiology".
Collapse
Affiliation(s)
- Stephen G Woodhams
- Arthritis UK Pain Centre, School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham, United Kingdom.
| | - Victoria Chapman
- Arthritis UK Pain Centre, School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham, United Kingdom
| | - David P Finn
- Pharmacology & Therapeutics, School of Medicine, Galway Neuroscience Centre and Centre for Pain Research, NCBES, National University of Ireland Galway, University Road, Galway, Ireland
| | - Andrea G Hohmann
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA; Program in Neuroscience, Indiana University, Bloomington, IN, USA; Interdisciplinary Biochemistry Graduate Program, Department of Molecular and Cellular Biochemistry, Indiana University, Bloomington, IN, USA; Gill Center for Biomolecular Science, Indiana University, Bloomington, IN, USA
| | - Volker Neugebauer
- Department of Pharmacology and Neuroscience, Center of Excellence for Translational Neuroscience and Therapeutics, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| |
Collapse
|
323
|
Ho WSV, Kelly MEM. Cannabinoids in the Cardiovascular System. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2017; 80:329-366. [PMID: 28826540 DOI: 10.1016/bs.apha.2017.05.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Cannabinoids are known to modulate cardiovascular functions including heart rate, vascular tone, and blood pressure in humans and animal models. Essential components of the endocannabinoid system, namely, the production, degradation, and signaling pathways of endocannabinoids have been described not only in the central and peripheral nervous system but also in myocardium, vasculature, platelets, and immune cells. The mechanisms of cardiovascular responses to endocannabinoids are often complex and may involve cannabinoid CB1 and CB2 receptors or non-CB1/2 receptor targets. Preclinical and some clinical studies have suggested that targeting the endocannabinoid system can improve cardiovascular functions in a number of pathophysiological conditions, including hypertension, metabolic syndrome, sepsis, and atherosclerosis. In this chapter, we summarize the local and systemic cardiovascular effects of cannabinoids and highlight our current knowledge regarding the therapeutic potential of endocannabinoid signaling and modulation.
Collapse
Affiliation(s)
- Wing S V Ho
- Vascular Biology Research Centre, St George's University of London, London, United Kingdom.
| | | |
Collapse
|
324
|
Molecular Targets of the Phytocannabinoids: A Complex Picture. PROGRESS IN THE CHEMISTRY OF ORGANIC NATURAL PRODUCTS 2017; 103:103-131. [PMID: 28120232 DOI: 10.1007/978-3-319-45541-9_4] [Citation(s) in RCA: 224] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
For centuries, hashish and marihuana, both derived from the Indian hemp Cannabis sativa L., have been used for their medicinal, as well as, their psychotropic effects. These effects are associated with the phytocannabinoids which are oxygen containing C21 aromatic hydrocarbons found in Cannabis sativa L. To date, over 120 phytocannabinoids have been isolated from Cannabis. For many years, it was assumed that the beneficial effects of the phytocannabinoids were mediated by the cannabinoid receptors, CB1 and CB2. However, today we know that the picture is much more complex, with the same phytocannabinoid acting at multiple targets. This contribution focuses on the molecular pharmacology of the phytocannabinoids, including Δ9-THC and CBD, from the prospective of the targets at which these important compounds act.
Collapse
|
325
|
The Role of Nuclear Hormone Receptors in Cannabinoid Function. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2017; 80:291-328. [PMID: 28826538 DOI: 10.1016/bs.apha.2017.03.008] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Since the early 2000s, evidence has been accumulating that most cannabinoid compounds interact with the nuclear hormone family peroxisome proliferator-activated receptors (PPARs). This can be through direct binding of these compounds to PPARs, metabolism of cannabinoid to other PPAR-activating chemicals, or indirect activation of PPAR through cell signaling pathways. Delivery of cannabinoids to the nucleus may be facilitated by fatty acid-binding proteins and carrier proteins. All PPAR isoforms appear to be activated by cannabinoids, but the majority of evidence is for PPARα and γ. To date, little is known about the potential interaction of cannabinoids with other nuclear hormones. At least some (but not all) of the well-known biological actions of cannabinoids including neuroprotection, antiinflammatory action, and analgesic effects are partly mediated by PPAR-activation, often in combination with activation of the more traditional target sites of action. This has been best investigated for the endocannabinoid-like compounds palmitoylethanolamide and oleoylethanolamine acting at PPARα, and for phytocannabinoids or their derivatives activation acting at PPARγ. However, there are still many aspects of cannabinoid activation of PPAR and the role it plays in the biological and therapeutic effects of cannabinoids that remain to be investigated.
Collapse
|
326
|
Ratano P, Palmery M, Trezza V, Campolongo P. Cannabinoid Modulation of Memory Consolidation in Rats: Beyond the Role of Cannabinoid Receptor Subtype 1. Front Pharmacol 2017; 8:200. [PMID: 28446875 PMCID: PMC5388693 DOI: 10.3389/fphar.2017.00200] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2017] [Accepted: 03/28/2017] [Indexed: 12/30/2022] Open
Abstract
The effects induced by exogenous manipulation of endocannabinoid neurotransmission on emotion and memory are often contradictory. Among the different factors involved, of particular interest is the binding affinity of endocannabinoids, and their analogs, for other receptor families beyond cannabinoid receptors, such as the peroxisome proliferator-activated receptors (PPARs), and the transient receptor potential cation channel subfamily V member 1 (TRPV1). The aim of this study was to investigate which receptor subtype mediates cannabinoid effects on memory consolidation for emotionally arousing experiences. We tested two cannabinoid compounds with different pharmacological properties in the inhibitory avoidance task, and evaluated whether the observed effects are mediated by cannabinoid, PPARα or TRPV1 receptor activation. We found that the synthetic cannabinoid agonist WIN55,212-2 and the FAAH inhibitor URB597 both enhanced memory consolidation for inhibitory avoidance training. WIN55,212-22 effects on memory consolidation were predominantly mediated by CB1 receptor activation but CB2 receptors were involved as well. The URB597-induced memory enhancement was dependent on the activation not only of CB1 and CB2 receptors but, notwithstanding, PPAR-α and TRPV1 receptors were involved as well. Our findings drive beyond the classical hypothesis centered on the unique role of CB1 receptor activation for cannabinoid effects on memory, and reveal new insights in the neural mechanisms of memory consolidation.
Collapse
Affiliation(s)
- Patrizia Ratano
- Department of Physiology and Pharmacology, Sapienza University of RomeRome, Italy
| | - Maura Palmery
- Department of Physiology and Pharmacology, Sapienza University of RomeRome, Italy
| | - Viviana Trezza
- Department of Science, Section of Biomedical Sciences and Technologies, Roma Tre UniversityRome, Italy
| | - Patrizia Campolongo
- Department of Physiology and Pharmacology, Sapienza University of RomeRome, Italy
| |
Collapse
|
327
|
Mouro FM, Batalha VL, Ferreira DG, Coelho JE, Baqi Y, Müller CE, Lopes LV, Ribeiro JA, Sebastião AM. Chronic and acute adenosine A 2A receptor blockade prevents long-term episodic memory disruption caused by acute cannabinoid CB 1 receptor activation. Neuropharmacology 2017; 117:316-327. [PMID: 28235548 DOI: 10.1016/j.neuropharm.2017.02.021] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Revised: 01/17/2017] [Accepted: 02/19/2017] [Indexed: 11/19/2022]
Abstract
Cannabinoid-mediated memory impairment is a concern in cannabinoid-based therapies. Caffeine exacerbates cannabinoid CB1 receptor (CB1R)-induced memory deficits through an adenosine A1 receptor-mediated mechanism. We now evaluated how chronic or acute blockade of adenosine A2A receptors (A2ARs) affects long-term episodic memory deficits induced by a single injection of a selective CB1R agonist. Long-term episodic memory was assessed by the novel object recognition (NOR) test. Mice received an intraperitoneal (i.p.) injection of the CB1/CB2 receptor agonist WIN 55,212-2 (1 mg/kg) immediately after the NOR training, being tested for novelty recognition 24 h later. Anxiety levels were assessed by the Elevated Plus Maze test, immediately after the NOR. Mice were also tested for exploratory behaviour at the Open Field. For chronic A2AR blockade, KW-6002 (istradefylline) (3 mg/kg/day) was administered orally for 30 days; acute blockade of A2ARs was assessed by i.p. injection of SCH 58261 (1 mg/kg) administered either together with WIN 55,212-2 or only 30 min before the NOR test phase. The involvement of CB1Rs was assessed by using the CB1R antagonist, AM251 (3 mg/kg, i.p.). WIN 55,212-2 caused a disruption in NOR, an action absent in mice also receiving AM251, KW-6002 or SCH 58261 during the encoding/consolidation phase; SCH 58251 was ineffective if present during retrieval only. No effects were detected in the Elevated Plus maze or Open Field Test. The finding that CB1R-mediated memory disruption is prevented by antagonism of adenosine A2ARs, highlights a possibility to prevent cognitive side effects when therapeutic application of CB1R drugs is desired.
Collapse
MESH Headings
- Adenosine A2 Receptor Antagonists/administration & dosage
- Animals
- Benzoxazines/pharmacology
- Calcium Channel Blockers/pharmacology
- Cannabinoid Receptor Agonists/toxicity
- Exploratory Behavior/drug effects
- Exploratory Behavior/physiology
- Male
- Maze Learning/drug effects
- Maze Learning/physiology
- Memory Disorders/chemically induced
- Memory Disorders/metabolism
- Memory Disorders/prevention & control
- Memory, Episodic
- Memory, Long-Term/drug effects
- Memory, Long-Term/physiology
- Mice, Inbred C57BL
- Morpholines/pharmacology
- Naphthalenes/pharmacology
- Piperidines/pharmacology
- Purines/administration & dosage
- Pyrazoles/pharmacology
- Pyrimidines/administration & dosage
- Receptor, Adenosine A2A/metabolism
- Receptor, Cannabinoid, CB1/agonists
- Receptor, Cannabinoid, CB1/metabolism
- Recognition, Psychology/drug effects
- Recognition, Psychology/physiology
- Triazoles/administration & dosage
Collapse
Affiliation(s)
- Francisco M Mouro
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Portugal; Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Portugal
| | - Vânia L Batalha
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Portugal
| | - Diana G Ferreira
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Portugal
| | - Joana E Coelho
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Portugal
| | - Younis Baqi
- Pharma-Zentrum Bonn, Pharmazeutisches Institut, Pharmazeutische Chemie I, University of Bonn, Germany; Department of Chemistry, Faculty of Science, Sultan Qaboos University, Muscat, Oman
| | - Christa E Müller
- Pharma-Zentrum Bonn, Pharmazeutisches Institut, Pharmazeutische Chemie I, University of Bonn, Germany
| | - Luísa V Lopes
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Portugal
| | - Joaquim A Ribeiro
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Portugal; Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Portugal
| | - Ana M Sebastião
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Portugal; Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Portugal.
| |
Collapse
|
328
|
Grabner GF, Zimmermann R, Schicho R, Taschler U. Monoglyceride lipase as a drug target: At the crossroads of arachidonic acid metabolism and endocannabinoid signaling. Pharmacol Ther 2017; 175:35-46. [PMID: 28213089 DOI: 10.1016/j.pharmthera.2017.02.033] [Citation(s) in RCA: 108] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Monoglyerides (MGs) are short-lived, intermediary lipids deriving from the degradation of phospho- and neutral lipids, and monoglyceride lipase (MGL), also designated as monoacylglycerol lipase (MAGL), is the major enzyme catalyzing the hydrolysis of MGs into glycerol and fatty acids. This distinct function enables MGL to regulate a number of physiological and pathophysiological processes since both MGs and fatty acids can act as signaling lipids or precursors thereof. The most prominent MG species acting as signaling lipid is 2-arachidonoyl glycerol (2-AG) which is the most abundant endogenous agonist of cannabinoid receptors in the body. Importantly, recent observations demonstrate that 2-AG represents a quantitatively important source for arachidonic acid, the precursor of prostaglandins and other inflammatory mediators. Accordingly, MGL-mediated 2-AG degradation affects lipid signaling by cannabinoid receptor-dependent and independent mechanisms. Recent genetic and pharmacological studies gave important insights into MGL's role in (patho-)physiological processes, and the enzyme is now considered as a promising drug target for a number of disorders including cancer, neurodegenerative and inflammatory diseases. This review summarizes the basics of MG (2-AG) metabolism and provides an overview on the therapeutic potential of MGL.
Collapse
Affiliation(s)
- Gernot F Grabner
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Robert Zimmermann
- Institute of Molecular Biosciences, University of Graz, Graz, Austria; BioTechMed Graz, Graz, Austria
| | - Rudolf Schicho
- Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Graz, Austria; BioTechMed Graz, Graz, Austria.
| | - Ulrike Taschler
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| |
Collapse
|
329
|
Le Boisselier R, Alexandre J, Lelong-Boulouard V, Debruyne D. Focus on cannabinoids and synthetic cannabinoids. Clin Pharmacol Ther 2016; 101:220-229. [PMID: 27861784 DOI: 10.1002/cpt.563] [Citation(s) in RCA: 105] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Revised: 11/03/2016] [Accepted: 11/06/2016] [Indexed: 02/06/2023]
Abstract
The recent emergence of a multitude of synthetic cannabinoids (SCs) has generated a wealth of new information, suggesting the usefulness of state-of-the-art on lato sensu cannabinoids. By modulating a plurality of neurotransmission pathways, the endocannabinoid system is involved in many physiological processes that are increasingly explored. SCs desired and adverse effects are considered to be more intense than those observed with cannabis smoking, which is partly explained by the full agonist activity and higher affinity for cannabinoid receptors. Neurological and cardiovascular side effects observed after cannabinoid poisoning generally respond to conventional supportive care, but severe outcomes may occur in a minority of cases, mainly observed with SCs. The likelihood of severe abuse and addiction produced by SCs are of concern for the scientific community also interested in the potential therapeutic value of cannabinoids.
Collapse
Affiliation(s)
- R Le Boisselier
- University Hospital Centre Côte de Nacre, Centre for Evaluation and Information on Pharmacodependance - Addictovigilance (CEIP-A), F-14033, Caen, France.,University Hospital Centre Côte de Nacre, Department of Pharmacology, Caen, France
| | - J Alexandre
- University Hospital Centre Côte de Nacre, Department of Pharmacology, Caen, France.,Normandy University, UNICAEN, University Hospital Centre Côte de Nacre, Caen, France
| | - V Lelong-Boulouard
- University Hospital Centre Côte de Nacre, Department of Pharmacology, Caen, France.,Normandy University, UNICAEN, University Hospital Centre Côte de Nacre, Inserm U 1075 COMETE Caen, France
| | - D Debruyne
- University Hospital Centre Côte de Nacre, Centre for Evaluation and Information on Pharmacodependance - Addictovigilance (CEIP-A), F-14033, Caen, France.,University Hospital Centre Côte de Nacre, Department of Pharmacology, Caen, France.,Medical School, Normandy University, Caen, France
| |
Collapse
|
330
|
Hasenoehrl C, Taschler U, Storr M, Schicho R. The gastrointestinal tract - a central organ of cannabinoid signaling in health and disease. Neurogastroenterol Motil 2016; 28:1765-1780. [PMID: 27561826 PMCID: PMC5130148 DOI: 10.1111/nmo.12931] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2016] [Accepted: 08/01/2016] [Indexed: 12/18/2022]
Abstract
BACKGROUND In ancient medicine, extracts of the marijuana plant Cannabis sativa were used against diseases of the gastrointestinal (GI) tract. Today, our knowledge of the ingredients of the Cannabis plant has remarkably advanced enabling us to use a variety of herbal and synthetic cannabinoid (CB) compounds to study the endocannabinoid system (ECS), a physiologic entity that controls tissue homeostasis with the help of endogenously produced CBs and their receptors. After many anecdotal reports suggested beneficial effects of Cannabis in GI disorders, it was not surprising to discover that the GI tract accommodates and expresses all the components of the ECS. Cannabinoid receptors and their endogenous ligands, the endocannabinoids, participate in the regulation of GI motility, secretion, and the maintenance of the epithelial barrier integrity. In addition, other receptors, such as the transient receptor potential cation channel subfamily V member 1 (TRPV1), the peroxisome proliferator-activated receptor alpha (PPARα) and the G-protein coupled receptor 55 (GPR55), are important participants in the actions of CBs in the gut and critically determine the course of bowel inflammation and colon cancer. PURPOSE The following review summarizes important and recent findings on the role of CB receptors and their ligands in the GI tract with emphasis on GI disorders, such as irritable bowel syndrome, inflammatory bowel disease, and colon cancer.
Collapse
Affiliation(s)
- Carina Hasenoehrl
- Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Graz, Austria
| | - Ulrike Taschler
- Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Graz, Austria
| | - Martin Storr
- Department of Medicine, Ludwig-Maximilians University, Munich, Germany and Zentrum für Endoskopie, Starnberg, Germany
| | - Rudolf Schicho
- Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Graz, Austria
| |
Collapse
|
331
|
Navarro-Dorado J, Villalba N, Prieto D, Brera B, Martín-Moreno AM, Tejerina T, de Ceballos ML. Vascular Dysfunction in a Transgenic Model of Alzheimer's Disease: Effects of CB1R and CB2R Cannabinoid Agonists. Front Neurosci 2016; 10:422. [PMID: 27695396 PMCID: PMC5025475 DOI: 10.3389/fnins.2016.00422] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 08/29/2016] [Indexed: 01/21/2023] Open
Abstract
There is evidence of altered vascular function, including cerebrovascular, in Alzheimer's disease (AD) and transgenic models of the disease. Indeed vasoconstrictor responses are increased, while vasodilation is reduced in both conditions. β-Amyloid (Aβ) appears to be responsible, at least in part, of alterations in vascular function. Cannabinoids, neuroprotective and anti-inflammatory agents, induce vasodilation both in vivo and in vitro. We have demonstrated a beneficial effect of cannabinoids in models of AD by preventing glial activation. In this work we have studied the effects of these compounds on vessel density in amyloid precursor protein (APP) transgenic mice, line 2576, and on altered vascular responses in aortae isolated ring. First we showed increased collagen IV positive vessels in AD brain compared to control subjects, with a similar increase in TgAPP mice, which was normalized by prolonged oral treatment with the CB1/CB2 mixed agonist WIN 55,212-2 (WIN) and the CB2 selective agonist JWH-133 (JWH). In Tg APP mice the vasoconstriction induced by phenylephrine and the thromboxane agonist U46619 was significantly increased, and no change in the vasodilation to acetylcholine (ACh) was observed. Tg APP displayed decreased vasodilation to both cannabinoid agonists, which were able to prevent decreased ACh relaxation in the presence of Aβ. In summary, we have confirmed and extended the existence of altered vascular responses in Tg APP mice. Moreover, our results suggest that treatment with cannabinoids may ameliorate the vascular responses in AD-type pathology.
Collapse
Affiliation(s)
- Jorge Navarro-Dorado
- Department of Pharmacology, School of Medicine, Complutense University of Madrid Madrid, Spain
| | - Nuria Villalba
- Department of Physiology, Faculty of Pharmacy, Complutense University of Madrid Madrid, Spain
| | - Dolores Prieto
- Department of Physiology, Faculty of Pharmacy, Complutense University of Madrid Madrid, Spain
| | - Begoña Brera
- Neurodegeneration Group, Cellular, Molecular and Developmental Neurobiology and CIBERNED, Cajal Institute, CSIC Madrid, Spain
| | - Ana M Martín-Moreno
- Neurodegeneration Group, Cellular, Molecular and Developmental Neurobiology and CIBERNED, Cajal Institute, CSIC Madrid, Spain
| | - Teresa Tejerina
- Department of Pharmacology, School of Medicine, Complutense University of Madrid Madrid, Spain
| | - María L de Ceballos
- Neurodegeneration Group, Cellular, Molecular and Developmental Neurobiology and CIBERNED, Cajal Institute, CSIC Madrid, Spain
| |
Collapse
|
332
|
Agarwal S, Yadav A, Chaturvedi RK. Peroxisome proliferator-activated receptors (PPARs) as therapeutic target in neurodegenerative disorders. Biochem Biophys Res Commun 2016; 483:1166-1177. [PMID: 27514452 DOI: 10.1016/j.bbrc.2016.08.043] [Citation(s) in RCA: 133] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 07/21/2016] [Accepted: 08/07/2016] [Indexed: 01/06/2023]
Abstract
Peroxisome proliferator-activated receptors (PPARs) are nuclear receptors and they serve to be a promising therapeutic target for several neurodegenerative disorders, which includes Parkinson disease, Alzheimer's disease, Huntington disease and Amyotrophic Lateral Sclerosis. PPARs play an important role in the downregulation of mitochondrial dysfunction, proteasomal dysfunction, oxidative stress, and neuroinflammation, which are the major causes of the pathogenesis of neurodegenerative disorders. In this review, we discuss about the role of PPARs as therapeutic targets in neurodegenerative disorders. Several experimental approaches suggest potential application of PPAR agonist as well as antagonist in the treatment of neurodegenerative disorders. Several epidemiological studies found that the regular usage of PPAR activating non-steroidal anti-inflammatory drugs is effective in decreasing the progression of neurodegenerative diseases including PD and AD. We also reviewed the neuroprotective effects of PPAR agonists and associated mechanism of action in several neurodegenerative disorders both in vitro as well as in vivo animal models.
Collapse
Affiliation(s)
- Swati Agarwal
- Developmental Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, 226001, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), CSIR-IITR Lucknow Campus, Lucknow, India
| | - Anuradha Yadav
- Developmental Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, 226001, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), CSIR-IITR Lucknow Campus, Lucknow, India
| | - Rajnish Kumar Chaturvedi
- Developmental Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, 226001, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), CSIR-IITR Lucknow Campus, Lucknow, India.
| |
Collapse
|
333
|
O'Sullivan SE. An update on PPAR activation by cannabinoids. Br J Pharmacol 2016; 173:1899-910. [PMID: 27077495 PMCID: PMC4882496 DOI: 10.1111/bph.13497] [Citation(s) in RCA: 322] [Impact Index Per Article: 35.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Revised: 03/16/2016] [Accepted: 04/04/2016] [Indexed: 02/06/2023] Open
Abstract
Some cannabinoids activate the different isoforms of PPARs (α, β and γ), as shown through the use of reporter gene assays, binding studies, selective antagonists and knockout studies. Activation of all isoforms, but primarily PPARα and γ, mediates some (but not all) of the analgesic, neuroprotective, neuronal function modulation, anti-inflammatory, metabolic, anti-tumour, gastrointestinal and cardiovascular effects of some cannabinoids, often in conjunction with activation of the more traditional target sites of action such as the cannabinoid CB1 and CB2 receptors and the TRPV1 ion channel. PPARs also mediate some of the effects of inhibitors of endocannabinoid degradation or transport. Cannabinoids may be chaperoned to the PPARs by fatty acid binding proteins. The aims of this review are to update the evidence supporting PPAR activation by cannabinoids and to review the physiological responses to cannabinoids that are mediated, and not mediated, by PPAR activation.
Collapse
|