1
|
Li S, Xiao L, Sun Y, Hu S, Hu D. A TRPV1 common missense variant affected the prognosis of ischemic cardiomyopathy. Medicine (Baltimore) 2022; 101:e29892. [PMID: 35905222 PMCID: PMC9333512 DOI: 10.1097/md.0000000000029892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
The purpose was to identify the Transient receptor potential (TRP) superfamily gene variants associate with the prognosis of ischemic cardiomyopathy (ICM). A whole-exome sequencing study involving 252 ICM and 252 healthy controls participants enrolled from March 2003 to November 2017. Optimal sequence kernel association test and Cox regression dominant was conducted to identify the cause genes of TRP with ICM and association of common SNPs with prognosis of ICM. Rs224534 was verified in the replication population. Besides, the expression of TRPV1 was detectable in human failed heart ventricular tissues. The TRPs was not associated with the risk of ICM (P > .05). Rs224534 was significantly associated with the prognosis of ICM (Hazard ratio, 2.27, 95%CI: 1.31-3.94; P = 3.7 × 10-3), in the replication cohort, (hazard ratio 1.47, 95%CI: 1.04-2.07; P = 2.9 × 10-2), and in combined cohort hazard ratio 1.62 (95%CI: 1.21-2.18; P = 1.1 × 10-3). The common SNP of TRPV1 (rs224534) is associated with the prognosis of ICM, and homozygote rs224534-AA showed an unfavorable prognosis of ICM in the dominant model tested. Genotyping the variant may benefit to further progress judgment of ICM.
Collapse
Affiliation(s)
- Shiyang Li
- Division of Cardiology, Panzhihua Central Hospital, Panzhihua, China
- *Correspondence: Panzhihua Central Hospital, 34# Yi kang Ave., Panzhihua 617000, China (e-mail: )
| | - Lei Xiao
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Huazhong University of Science and Technology, Hubei Province, Wuhan, China
| | - Yang Sun
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Huazhong University of Science and Technology, Hubei Province, Wuhan, China
| | - Senlin Hu
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Huazhong University of Science and Technology, Hubei Province, Wuhan, China
| | - Dong Hu
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Huazhong University of Science and Technology, Hubei Province, Wuhan, China
| |
Collapse
|
2
|
Xiao T, Sun M, Kang J, Zhao C. Transient Receptor Potential Vanilloid1 (TRPV1) Channel Opens Sesame of T Cell Responses and T Cell-Mediated Inflammatory Diseases. Front Immunol 2022; 13:870952. [PMID: 35634308 PMCID: PMC9130463 DOI: 10.3389/fimmu.2022.870952] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 04/19/2022] [Indexed: 12/11/2022] Open
Abstract
Transient receptor potential vanilloid1 (TRPV1) was primarily expressed in sensory neurons, and could be activated by various physical and chemical factors, resulting in the flow of extracellular Ca2+ into cells. Accumulating data suggest that the TRPV1 is expressed in some immune cells and is a novel regulator of the immune system. In this review, we highlight the structure and biological features of TRPV1 channel. We also summarize recent findings on its role in modulating T cell activation and differentiation as well as its protective effect in T cell-mediated inflammatory diseases and potential mechanisms.
Collapse
Affiliation(s)
- Tengfei Xiao
- Department of Clinical Laboratory, The Sixth Affiliated Hospital of Nantong University, Yancheng Third People’s Hospital, Yancheng, China
| | - Mingzhong Sun
- Department of Clinical Laboratory, The Sixth Affiliated Hospital of Nantong University, Yancheng Third People’s Hospital, Yancheng, China
| | - Jingjing Kang
- Department of Clinical Laboratory, Affiliated Hospital of Nanjing University Medical School, Yancheng First People’s Hospital, Yancheng, China
| | - Chuanxiang Zhao
- Institute of Medical Genetics and Reproductive Immunity, School of Medical Science and Laboratory Medicine, Jiangsu College of Nursing, Huai’an, China
| |
Collapse
|
3
|
TRPV1 channel mediates NLRP3 inflammasome-dependent neuroinflammation in microglia. Cell Death Dis 2021; 12:1159. [PMID: 34907173 PMCID: PMC8671551 DOI: 10.1038/s41419-021-04450-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 11/06/2021] [Accepted: 11/17/2021] [Indexed: 12/12/2022]
Abstract
Multiple sclerosis (MS) is a chronic inflammatory autoimmune disease in the central nervous system (CNS). The NLRP3 inflammasome is considered an important regulator of immunity and inflammation, both of which play a critical role in MS. However, the underlying mechanism of NLRP3 inflammasome activation is not fully understood. Here we identified that the TRPV1 (transient receptor potential vanilloid type 1) channel in microglia, as a Ca2+ influx-regulating channel, played an important role in NLRP3 inflammasome activation. Deletion or pharmacological blockade of TRPV1 inhibited NLRP3 inflammasome activation in microglia in vitro. Further research revealed that TRPV1 channel regulated ATP-induced NLRP3 inflammasome activation through mediating Ca2+ influx and phosphorylation of phosphatase PP2A in microglia. In addition, TRPV1 deletion could alleviate mice experimental autoimmune encephalomyelitis (EAE) and reduce neuroinflammation by inhibiting NLRP3 inflammasome activation. These data suggested that the TRPV1 channel in microglia can regulate NLRP3 inflammasome activation and consequently mediate neuroinflammation. Meanwhile, our study indicated that TRPV1-Ca2+-PP2A pathway may be a novel regulator of NLRP3 inflammasome activation, pointing to TRPV1 as a potential target for CNS inflammatory diseases.
Collapse
|
4
|
Schmaul S, Hanuscheck N, Bittner S. Astrocytic potassium and calcium channels as integrators of the inflammatory and ischemic CNS microenvironment. Biol Chem 2021; 402:1519-1530. [PMID: 34455729 DOI: 10.1515/hsz-2021-0256] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 08/13/2021] [Indexed: 12/24/2022]
Abstract
Astrocytes are key regulators of their surroundings by receiving and integrating stimuli from their local microenvironment, thereby regulating glial and neuronal homeostasis. Cumulating evidence supports a plethora of heterogenic astrocyte subpopulations that differ morphologically and in their expression patterns of receptors, transporters and ion channels, as well as in their functional specialisation. Astrocytic heterogeneity is especially relevant under pathological conditions. In experimental autoimmune encephalomyelitis (EAE), a mouse model of multiple sclerosis (MS), morphologically distinct astrocytic subtypes were identified and could be linked to transcriptome changes during different disease stages and regions. To allow for continuous awareness of changing stimuli across age and diseases, astrocytes are equipped with a variety of receptors and ion channels allowing the precise perception of environmental cues. Recent studies implicate the diverse repertoire of astrocytic ion channels - including transient receptor potential channels, voltage-gated calcium channels, inwardly rectifying K+ channels, and two-pore domain potassium channels - in sensing the brain state in physiology, inflammation and ischemia. Here, we review current evidence regarding astrocytic potassium and calcium channels and their functional contribution in homeostasis, neuroinflammation and stroke.
Collapse
Affiliation(s)
- Samantha Schmaul
- Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2), University Medical Centre of the Johannes Gutenberg University Mainz, Langenbeckstraße 1, D-55131 Mainz, Germany
| | - Nicholas Hanuscheck
- Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2), University Medical Centre of the Johannes Gutenberg University Mainz, Langenbeckstraße 1, D-55131 Mainz, Germany
| | - Stefan Bittner
- Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2), University Medical Centre of the Johannes Gutenberg University Mainz, Langenbeckstraße 1, D-55131 Mainz, Germany
| |
Collapse
|
5
|
Çakır M, Saçmacı H, Sabah-Özcan S. Selected transient receptor potential channel genes' expression in peripheral blood mononuclear cells of multiple sclerosis. Hum Exp Toxicol 2021; 40:S406-S413. [PMID: 34569347 DOI: 10.1177/09603271211043476] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Transient receptor potential channels have responsibilities in many cellular processes such as cytokine production, cell differentiation, and cytotoxicity by affecting intracellular cation levels or intracellular signal pathways. Multiple sclerosis is a chronic autoimmune central nervous system (CNS) disease caused by environmental and genetic factors. In this study, we aim to investigate TRPV1-TRPV4, TRPM2, TRPM4, TRPM7, TRPC6, and TRPA1 mRNA expression levels, which are associated with the inflammatory process, in the peripheral blood mononuclear cells (PBMCs) of relapsing-remitting multiple sclerosis (RRMS) patients. Thirty-five healthy controls and age-gender matched thirty patients with RRMS were involved in the study. TRPC6, TRPA1, TRPM2, TRPM4, TRPM7, TRPV1, TRPV2, TRPV3, and TRPV4 PBMCs mRNA expression levels were determined by qPCR. In the present study, the TRPC6, TRPM7, TRPV1, TRPV3, and TRPV4 mRNA expressions of RRMS patients in PBMCs decreased at a significant level compared to the healthy control group (p = .000, p = .000, p = .044, p = .000, p = .004, respectively). The decreased expression of TRPC6, TRPM7, TRPV1, TRPV3, and TRPV4 in PBMCs may be associated with the pathogenesis of MS. Further studies are required to understand the mechanism of the relation between these TRP channels and MS and other autoimmune diseases.
Collapse
Affiliation(s)
- Murat Çakır
- Department of Physiology, Faculty of Medicine, 162338University of Yozgat Bozok, Yozgat, Turkey
| | - Hikmet Saçmacı
- Department of Neurology, Faculty of Medicine, 162338University of Yozgat Bozok, Yozgat, Turkey
| | - Seda Sabah-Özcan
- Department of Medical Biology, Faculty of Medicine, 64230University of Manisa Celal Bayar, Manisa, Turkey
| |
Collapse
|
6
|
Bottemanne P, Guillemot-Legris O, Paquot A, Masquelier J, Malamas M, Makriyannis A, Alhouayek M, Muccioli GG. N-Acylethanolamine-Hydrolyzing Acid Amidase Inhibition, but Not Fatty Acid Amide Hydrolase Inhibition, Prevents the Development of Experimental Autoimmune Encephalomyelitis in Mice. Neurotherapeutics 2021; 18:1815-1833. [PMID: 34235639 PMCID: PMC8609003 DOI: 10.1007/s13311-021-01074-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/13/2021] [Indexed: 02/06/2023] Open
Abstract
N-acylethanolamines (NAEs) are endogenous bioactive lipids reported to exert anti-inflammatory and neuroprotective effects mediated by cannabinoid receptors and peroxisome proliferator-activated receptors (PPARs), among others. Therefore, interfering with NAE signaling could be a promising strategy to decrease inflammation in neurological disorders such as multiple sclerosis (MS). Fatty acid amide hydrolase (FAAH) and N-acylethanolamine-hydrolyzing acid amidase (NAAA) are key modulators of NAE levels. This study aims to investigate and compare the effect of NAAA inhibition, FAAH inhibition, and dual inhibition of both enzymes in a mouse model of MS, namely the experimental autoimmune encephalomyelitis (EAE). Our data show that NAAA inhibition strongly decreased the hallmarks of the pathology. Interestingly, FAAH inhibition was less efficient in decreasing inflammatory hallmarks despite the increased NAE levels. Moreover, the inhibition of both NAAA and FAAH, using a dual-inhibitor or the co-administration of NAAA and FAAH inhibitors, did not show an added value compared to NAAA inhibition. Furthermore, our data suggest an important role of decreased activation of astrocytes and microglia in the effects of NAAA inhibition on EAE, while NAAA inhibition did not affect T cell recall. This work highlights the beneficial effects of NAAA inhibition in the context of central nervous system inflammation and suggests that the simultaneous inhibition of NAAA and FAAH has no additional beneficial effect in EAE.
Collapse
Affiliation(s)
- Pauline Bottemanne
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, B1.72.01, Av. E. Mounier 72, 1200, Bruxelles, Belgium
| | - Owein Guillemot-Legris
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, B1.72.01, Av. E. Mounier 72, 1200, Bruxelles, Belgium
| | - Adrien Paquot
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, B1.72.01, Av. E. Mounier 72, 1200, Bruxelles, Belgium
| | - Julien Masquelier
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, B1.72.01, Av. E. Mounier 72, 1200, Bruxelles, Belgium
| | - Michael Malamas
- Center for Drug Discovery and Departments of Chemistry and Chemical Biology and Pharmaceutical Sciences, Northeastern University, Boston, MA, 02115, USA
| | - Alexandros Makriyannis
- Center for Drug Discovery and Departments of Chemistry and Chemical Biology and Pharmaceutical Sciences, Northeastern University, Boston, MA, 02115, USA
| | - Mireille Alhouayek
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, B1.72.01, Av. E. Mounier 72, 1200, Bruxelles, Belgium
| | - Giulio G Muccioli
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, B1.72.01, Av. E. Mounier 72, 1200, Bruxelles, Belgium.
| |
Collapse
|
7
|
Boscia F, Elkjaer ML, Illes Z, Kukley M. Altered Expression of Ion Channels in White Matter Lesions of Progressive Multiple Sclerosis: What Do We Know About Their Function? Front Cell Neurosci 2021; 15:685703. [PMID: 34276310 PMCID: PMC8282214 DOI: 10.3389/fncel.2021.685703] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Accepted: 05/23/2021] [Indexed: 12/19/2022] Open
Abstract
Despite significant advances in our understanding of the pathophysiology of multiple sclerosis (MS), knowledge about contribution of individual ion channels to axonal impairment and remyelination failure in progressive MS remains incomplete. Ion channel families play a fundamental role in maintaining white matter (WM) integrity and in regulating WM activities in axons, interstitial neurons, glia, and vascular cells. Recently, transcriptomic studies have considerably increased insight into the gene expression changes that occur in diverse WM lesions and the gene expression fingerprint of specific WM cells associated with secondary progressive MS. Here, we review the ion channel genes encoding K+, Ca2+, Na+, and Cl- channels; ryanodine receptors; TRP channels; and others that are significantly and uniquely dysregulated in active, chronic active, inactive, remyelinating WM lesions, and normal-appearing WM of secondary progressive MS brain, based on recently published bulk and single-nuclei RNA-sequencing datasets. We discuss the current state of knowledge about the corresponding ion channels and their implication in the MS brain or in experimental models of MS. This comprehensive review suggests that the intense upregulation of voltage-gated Na+ channel genes in WM lesions with ongoing tissue damage may reflect the imbalance of Na+ homeostasis that is observed in progressive MS brain, while the upregulation of a large number of voltage-gated K+ channel genes may be linked to a protective response to limit neuronal excitability. In addition, the altered chloride homeostasis, revealed by the significant downregulation of voltage-gated Cl- channels in MS lesions, may contribute to an altered inhibitory neurotransmission and increased excitability.
Collapse
Affiliation(s)
- Francesca Boscia
- Division of Pharmacology, Department of Neuroscience, Reproductive and Dentistry Sciences, School of Medicine, University of Naples "Federico II", Naples, Italy
| | - Maria Louise Elkjaer
- Neurology Research Unit, Department of Clinical Research, University of Southern Denmark, Odense, Denmark.,Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Zsolt Illes
- Neurology Research Unit, Department of Clinical Research, University of Southern Denmark, Odense, Denmark.,Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark.,Department of Neurology, Odense University Hospital, Odense, Denmark
| | - Maria Kukley
- Achucarro Basque Center for Neuroscience, Leioa, Spain.,Ikerbasque Basque Foundation for Science, Bilbao, Spain
| |
Collapse
|
8
|
Dalenogare DP, Theisen MC, Peres DS, Fialho MFP, Lückemeyer DD, Antoniazzi CTDD, Kudsi SQ, Ferreira MDA, Ritter CDS, Ferreira J, Oliveira SM, Trevisan G. TRPA1 activation mediates nociception behaviors in a mouse model of relapsing-remitting experimental autoimmune encephalomyelitis. Exp Neurol 2020; 328:113241. [DOI: 10.1016/j.expneurol.2020.113241] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 01/31/2020] [Accepted: 02/07/2020] [Indexed: 12/16/2022]
|
9
|
Stampanoni Bassi M, Gentile A, Iezzi E, Zagaglia S, Musella A, Simonelli I, Gilio L, Furlan R, Finardi A, Marfia GA, Guadalupi L, Bullitta S, Mandolesi G, Centonze D, Buttari F. Transient Receptor Potential Vanilloid 1 Modulates Central Inflammation in Multiple Sclerosis. Front Neurol 2019; 10:30. [PMID: 30761069 PMCID: PMC6361812 DOI: 10.3389/fneur.2019.00030] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 01/10/2019] [Indexed: 12/04/2022] Open
Abstract
Introduction: Disease course of multiple sclerosis (MS) is negatively influenced by proinflammatory molecules released by activated T and B lymphocytes and local immune cells. The endovanilloid system plays different physiological functions, and preclinical data suggest that transient receptor potential vanilloid type 1 (TRPV1) could modulate neuroinflammation in this disorder. Methods: The effect of TRPV1 activation on the release of two main proinflammatory cytokines, tumor necrosis factor (TNF) and interleukin (IL)-6, was explored in activated microglial cells. Furthermore, in a group of 132 MS patients, the association between the cerebrospinal fluid (CSF) levels of TNF and IL-6 and a single nucleotide polymorphisms (SNP) influencing TRPV1 protein expression and function (rs222747) was assessed. Results: In in vitro experiments, TRPV1 stimulation by capsaicin significantly reduced TNF and IL-6 release by activated microglial cells. Moreover, the anti-inflammatory effect of TRPV1 activation was confirmed by another TRPV1 agonist, the resiniferatoxin (RTX), whose effects were significantly inhibited by the TRPV1 antagonist, 5-iodoresiniferatoxin (5-IRTX). Vice versa, BV2 pre-treatment with 5-IRTX increased the inflammatory response induced by LPS. Moreover, in MS patients, a significant association emerged between TRPV1 SNP rs222747 and CSF TNF levels. In particular, the presence of a G allele, known to result in increased TRPV1 protein expression and function, was associated to lower CSF levels of TNF. Conclusions: Our results indicate that TRPV1 influences central inflammation in MS by regulating cytokine release by activated microglial cells. The modulation of the endovanilloid system may represent a useful approach to contrast neuroinflammation in MS.
Collapse
Affiliation(s)
| | - Antonietta Gentile
- Laboratory of Synaptic Immunopathology, Department of Systems Medicine, Tor Vergata University, Rome, Italy
| | - Ennio Iezzi
- Unit of Neurology and Neurorehabilitation, IRCCS Neuromed, Pozzilli, Italy
| | - Sara Zagaglia
- Clinica Neurologica, Università Politecnica delle Marche, Ancona, Italy
| | - Alessandra Musella
- Laboratory of Neuroimmunology and Synaptic Plasticity, University & IRCCS San Raffaele, Rome, Italy
| | - Ilaria Simonelli
- Servizio di Statistica Medica & Information Technology, Fondazione Fatebenefratelli per la Ricerca e la Formazione Sanitaria e Sociale, Rome, Italy
| | - Luana Gilio
- Laboratory of Synaptic Immunopathology, Department of Systems Medicine, Tor Vergata University, Rome, Italy
| | - Roberto Furlan
- Neuroimmunology Unit, Division of Neuroscience, Institute of Experimental Neurology, San Raffaele Scientific Institute, Milan, Italy
| | - Annamaria Finardi
- Neuroimmunology Unit, Division of Neuroscience, Institute of Experimental Neurology, San Raffaele Scientific Institute, Milan, Italy
| | - Girolama A Marfia
- Laboratory of Synaptic Immunopathology, Department of Systems Medicine, Tor Vergata University, Rome, Italy
| | - Livia Guadalupi
- Laboratory of Neuroimmunology and Synaptic Plasticity, University & IRCCS San Raffaele, Rome, Italy
| | - Silvia Bullitta
- Laboratory of Synaptic Immunopathology, Department of Systems Medicine, Tor Vergata University, Rome, Italy
| | - Georgia Mandolesi
- Laboratory of Neuroimmunology and Synaptic Plasticity, University & IRCCS San Raffaele, Rome, Italy
| | - Diego Centonze
- Unit of Neurology and Neurorehabilitation, IRCCS Neuromed, Pozzilli, Italy.,Laboratory of Synaptic Immunopathology, Department of Systems Medicine, Tor Vergata University, Rome, Italy
| | - Fabio Buttari
- Unit of Neurology and Neurorehabilitation, IRCCS Neuromed, Pozzilli, Italy
| |
Collapse
|
10
|
Motte J, Ambrosius B, Grüter T, Bachir H, Sgodzai M, Pedreiturria X, Pitarokoili K, Gold R. Capsaicin-enriched diet ameliorates autoimmune neuritis in rats. J Neuroinflammation 2018; 15:122. [PMID: 29690884 PMCID: PMC5916583 DOI: 10.1186/s12974-018-1165-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 04/18/2018] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Autoimmune neuropathies are common PNS disorders and effective treatment is challenging. Environmental influence and dietary components are known to affect the course of autoimmune diseases. Capsaicin as pungent component of chili-peppers is common in human nutrition. An influence of capsaicin on autoimmune diseases has been postulated. METHODS We tested capsaicin in the animal model of experimental autoimmune neuritis (EAN) in Lewis rat. Rats were immunized with P2-peptide and were treated with capsaicin in different preventive settings. Electrophysiological, histological, and molecular biological analyses of the sciatic nerve were performed to analyze T-cell and macrophage cell count, TRPV1, and cytokine expression. Moreover, FACS analyses including the intestinal immune system were executed. RESULTS We observed an immunomodulatory effect of an early preventive diet-concept, where a physiological dosage of oral capsaicin was given 10 days before immunization in EAN. A reduced inflammation of the sciatic nerve was significant detectable clinically, electrophysiologically (CMAPs reduced in control group p < 0.01; increase of nerve conduction blocks in control group p < 0.05), histologically (significant reduction of T-cells, macrophages and demyelination), and at cytokine level. In contrast, this therapeutic effect was missing with capsaicin given from the day of immunization onwards. As possible underlying mechanism, we were able to show changes in the expression of the capsaicin receptor in the sciatic nerve and the small intestine, as well as altered immune cell populations in the small intestine. CONCLUSION This is the first report about the immunomodulatory effect of the common nutrient, capsaicin, in an experimental model for autoimmune neuropathies.
Collapse
MESH Headings
- Animals
- Capsaicin/therapeutic use
- Cell Movement/drug effects
- Cytokines/metabolism
- Diet/methods
- Disease Models, Animal
- Evoked Potentials, Motor/drug effects
- Female
- Freund's Adjuvant/toxicity
- Gene Expression Regulation/drug effects
- Gene Expression Regulation/physiology
- Leukocytes, Mononuclear/metabolism
- Leukocytes, Mononuclear/pathology
- Macrophages/drug effects
- Macrophages/pathology
- Neural Conduction/drug effects
- Neuritis, Autoimmune, Experimental/diet therapy
- Neuritis, Autoimmune, Experimental/pathology
- Neuritis, Autoimmune, Experimental/physiopathology
- Rats
- Rats, Inbred Lew
- S100 Proteins/metabolism
- Sciatic Nerve/drug effects
- Sciatic Nerve/metabolism
- Sciatic Nerve/pathology
- T-Lymphocytes/drug effects
- TRPV Cation Channels/metabolism
Collapse
Affiliation(s)
- Jeremias Motte
- Department of Neurology, Ruhr University Bochum, St. Josef- Hospital, Gudrunstrasse 56, 44791 Bochum, Germany
| | - Björn Ambrosius
- Department of Neurology, Ruhr University Bochum, St. Josef- Hospital, Gudrunstrasse 56, 44791 Bochum, Germany
| | - Thomas Grüter
- Department of Neurology, Ruhr University Bochum, St. Josef- Hospital, Gudrunstrasse 56, 44791 Bochum, Germany
| | - Hussein Bachir
- Department of Neurology, Ruhr University Bochum, St. Josef- Hospital, Gudrunstrasse 56, 44791 Bochum, Germany
| | - Melissa Sgodzai
- Department of Neurology, Ruhr University Bochum, St. Josef- Hospital, Gudrunstrasse 56, 44791 Bochum, Germany
| | - Xiomara Pedreiturria
- Department of Neurology, Ruhr University Bochum, St. Josef- Hospital, Gudrunstrasse 56, 44791 Bochum, Germany
| | - Kalliopi Pitarokoili
- Department of Neurology, Ruhr University Bochum, St. Josef- Hospital, Gudrunstrasse 56, 44791 Bochum, Germany
| | - Ralf Gold
- Department of Neurology, Ruhr University Bochum, St. Josef- Hospital, Gudrunstrasse 56, 44791 Bochum, Germany
| |
Collapse
|
11
|
Ewanchuk BW, Allan ERO, Warren AL, Ramachandran R, Yates RM. The cooling compound icilin attenuates autoimmune neuroinflammation through modulation of the T-cell response. FASEB J 2017; 32:1236-1249. [PMID: 29114087 DOI: 10.1096/fj.201700552r] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The synthetic supercooling drug, icilin, and its primary receptor target, the cation channel transient receptor potential (TRP) melastatin-8 (TRPM8), have been described as potent negative regulators of inflammation in the colon. The aim of this study was to determine whether the anti-inflammatory action of icilin could potentially be used to treat autoimmune neuroinflammatory disorders, such as multiple sclerosis (MS). During experimental autoimmune encephalomyelitis (EAE)-a CD4+ T cell-driven murine model of MS-we found that both wild-type (WT) and TRPM8-deficient EAE mice were protected from disease progression during icilin treatment, as evidenced by delays in clinical onset and reductions in neuroinflammation. In vitro, icilin potently inhibited the proliferation of murine and human CD4+ T cells, with the peripheral expansion of autoantigen-restricted T cells similarly diminished by the administration of icilin in mice. Attenuation of both TRPM8-/- and TRP ankyrin-1-/- T-cell proliferation by icilin was consistent with the WT phenotype, which suggests a mechanism that is independent of these channels. In addition, icilin treatment altered the expressional profile of activated CD4+ T cells to one that was indicative of restricted effector function and limited neuroinflammatory potential. These findings identify a potent anti-inflammatory role for icilin in lymphocyte-mediated neuroinflammation and highlight clear pleiotropic effects of the compound beyond classic TRP channel activation.-Ewanchuk, B. W., Allan, E. R. O., Warren, A. L., Ramachandran, R., Yates, R. M. The cooling compound icilin attenuates autoimmune neuroinflammation through modulation of the T-cell response.
Collapse
Affiliation(s)
- Benjamin W Ewanchuk
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Euan R O Allan
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Amy L Warren
- Department of Veterinary Clinical and Diagnostic Sciences, Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Rithwik Ramachandran
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Robin M Yates
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
12
|
Parenti A, De Logu F, Geppetti P, Benemei S. What is the evidence for the role of TRP channels in inflammatory and immune cells? Br J Pharmacol 2016; 173:953-69. [PMID: 26603538 DOI: 10.1111/bph.13392] [Citation(s) in RCA: 109] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Revised: 10/25/2015] [Accepted: 11/10/2015] [Indexed: 12/11/2022] Open
Abstract
A complex network of many interacting mechanisms orchestrates immune and inflammatory responses. Among these, the cation channels of the transient receptor potential (TRP) family expressed by resident tissue cells, inflammatory and immune cells and distinct subsets of primary sensory neurons, have emerged as a novel and interrelated system to detect and respond to harmful agents. TRP channels, by means of their direct effect on the intracellular levels of cations and/or through the indirect modulation of a large series of intracellular pathways, orchestrate a range of cellular processes, such as cytokine production, cell differentiation and cytotoxicity. The contribution of TRP channels to the transition of inflammation and immune responses from a defensive early response to a chronic and pathological condition is also emerging as a possible underlying mechanism in various diseases. This review discusses the roles of TRP channels in inflammatory and immune cell function and provides an overview of the effects of inflammatory and immune TRP channels on the pathogenesis of human diseases.
Collapse
Affiliation(s)
- A Parenti
- Clinical Pharmacology and Oncology Unit, Department of Health Sciences, University of Florence, Florence, Italy
| | - F De Logu
- Clinical Pharmacology and Oncology Unit, Department of Health Sciences, University of Florence, Florence, Italy
| | - P Geppetti
- Clinical Pharmacology and Oncology Unit, Department of Health Sciences, University of Florence, Florence, Italy
| | - S Benemei
- Clinical Pharmacology and Oncology Unit, Department of Health Sciences, University of Florence, Florence, Italy
| |
Collapse
|
13
|
Deng Y, Huang X, Wu H, Zhao M, Lu Q, Israeli E, Dahan S, Blank M, Shoenfeld Y. Some like it hot: The emerging role of spicy food (capsaicin) in autoimmune diseases. Autoimmun Rev 2016; 15:451-6. [PMID: 26812350 DOI: 10.1016/j.autrev.2016.01.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 01/20/2016] [Indexed: 12/28/2022]
Abstract
Autoimmune diseases refer to a spectrum of diseases characterized by an active immune response against the host, which frequently involves increased autoantibody production. The pathogenesis of autoimmune diseases is multifactorial and the exploitation of novel effective treatment is urgent. Capsaicin is a nutritional factor, the active component of chili peppers, which is responsible for the pungent component of chili pepper. As a stimuli, capsaicin selectively activate transient receptor potential vanilloid subfamily 1(TRPV1) and exert various biological effects. This review discusses the effect of capsaicin through its receptor on the development and modulation of autoimmune diseases, which may shed light upon potential therapies in capsaicin-targeted approaches.
Collapse
Affiliation(s)
- Yaxiong Deng
- Hunan Key Laboratory of Medical Epigenetics, Department of Dermatology, Second Xiangya Hospital, Central South University, #139 Renmin Middle Rd, Changsha, Hunan 410011, PR China
| | - Xin Huang
- Hunan Key Laboratory of Medical Epigenetics, Department of Dermatology, Second Xiangya Hospital, Central South University, #139 Renmin Middle Rd, Changsha, Hunan 410011, PR China
| | - Haijing Wu
- Hunan Key Laboratory of Medical Epigenetics, Department of Dermatology, Second Xiangya Hospital, Central South University, #139 Renmin Middle Rd, Changsha, Hunan 410011, PR China
| | - Ming Zhao
- Hunan Key Laboratory of Medical Epigenetics, Department of Dermatology, Second Xiangya Hospital, Central South University, #139 Renmin Middle Rd, Changsha, Hunan 410011, PR China
| | - Qianjin Lu
- Hunan Key Laboratory of Medical Epigenetics, Department of Dermatology, Second Xiangya Hospital, Central South University, #139 Renmin Middle Rd, Changsha, Hunan 410011, PR China.
| | - Eitan Israeli
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Hashomer, Israel; Sackler Faculty of Medicine, Tel-Aviv University, Israel
| | - Shani Dahan
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Hashomer, Israel; Sackler Faculty of Medicine, Tel-Aviv University, Israel
| | - Miri Blank
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Hashomer, Israel; Sackler Faculty of Medicine, Tel-Aviv University, Israel
| | - Yehuda Shoenfeld
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Hashomer, Israel; Sackler Faculty of Medicine, Tel-Aviv University, Israel
| |
Collapse
|
14
|
Majhi RK, Sahoo SS, Yadav M, Pratheek BM, Chattopadhyay S, Goswami C. Functional expression of TRPV channels in T cells and their implications in immune regulation. FEBS J 2015; 282:2661-81. [PMID: 25903376 DOI: 10.1111/febs.13306] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Revised: 03/02/2015] [Accepted: 04/20/2015] [Indexed: 12/11/2022]
Abstract
The importance of Ca(2+) signalling and temperature in the context of T cell activation is well known. However, the molecular identities of key players involved in such critical regulations are still unknown. In this work we explored the endogenous expression of transient receptor potential vanilloid (TRPV) channels, a group of thermosensitive and non-selective cation channels, in T cells. Using flow cytometry and confocal microscopy, we demonstrate that members belonging to the TRPV subfamily are expressed endogenously in the human T cell line Jurkat, in primary human T cells and in primary murine splenic T cells. We also demonstrate that TRPV1- and TRPV4-specific agonists, namely resiniferatoxin and 4α-phorbol-12,13-didecanoate, can cause Ca(2+) influx in T cells. Moreover, our results show that expression of these channels can be upregulated in T cells during concanavalin A-driven mitogenic and anti-CD3/CD28 stimulated TCR activation of T cells. By specific blocking of TRPV1 and TRPV4 channels, we found that these TRPV inhibitors may regulate mitogenic and T cell receptor mediated T cell activation and effector cytokine(s) production by suppressing tumour necrosis factor, interleukin-2 and interferon-γ release. These results may have broad implications in the context of cell-mediated immunity, especially T cell responses and their regulations, neuro-immune interactions and molecular understanding of channelopathies.
Collapse
Affiliation(s)
- Rakesh K Majhi
- School of Biological Sciences, National Institute of Science Education and Research, Bhubaneswar, Orissa, India
| | - Subhransu S Sahoo
- School of Biological Sciences, National Institute of Science Education and Research, Bhubaneswar, Orissa, India
| | - Manoj Yadav
- School of Biological Sciences, National Institute of Science Education and Research, Bhubaneswar, Orissa, India
| | - Belluru M Pratheek
- School of Biological Sciences, National Institute of Science Education and Research, Bhubaneswar, Orissa, India
| | - Subhasis Chattopadhyay
- School of Biological Sciences, National Institute of Science Education and Research, Bhubaneswar, Orissa, India
| | - Chandan Goswami
- School of Biological Sciences, National Institute of Science Education and Research, Bhubaneswar, Orissa, India
| |
Collapse
|
15
|
Ehling P, Cerina M, Budde T, Meuth SG, Bittner S. The CNS under pathophysiologic attack--examining the role of K₂p channels. Pflugers Arch 2014; 467:959-72. [PMID: 25482672 DOI: 10.1007/s00424-014-1664-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Revised: 11/17/2014] [Accepted: 11/28/2014] [Indexed: 10/24/2022]
Abstract
Members of the two-pore domain K(+) channel (K2P) family are increasingly recognized as being potential targets for therapeutic drugs and could play a role in the diagnosis and treatment of neurologic disorders. Their broad and diverse expression pattern in pleiotropic cell types, importance in cellular function, unique biophysical properties, and sensitivity toward pathophysiologic parameters represent the basis for their involvement in disorders of the central nervous system (CNS). This review will focus on multiple sclerosis (MS) and stroke, as there is growing evidence for the involvement of K2P channels in these two major CNS disorders. In MS, TASK1-3 channels are expressed on T lymphocytes and are part of a signaling network regulating Ca(2+)- dependent pathways that are mandatory for T cell activation, differentiation, and effector functions. In addition, TASK1 channels are involved in neurodegeneration, resulting in autoimmune attack of CNS cells. On the blood-brain barrier, TREK1 channels regulate immune cell trafficking under autoinflammatory conditions. Cerebral ischemia shares some pathophysiologic similarities with MS, including hypoxia and extracellular acidosis. On a cellular level, K2P channels can have both proapoptotic and antiapoptotic effects, either promoting neurodegeneration or protecting neurons from ischemic cell death. TASK1 and TREK1 channels have a neuroprotective effect on stroke development, whereas TASK2 channels have a detrimental effect on neuronal survival under ischemic conditions. Future research in preclinical models is needed to provide a more detailed understanding of the contribution of K2P channel family members to neurologic disorders, before translation to the clinic is an option.
Collapse
Affiliation(s)
- Petra Ehling
- Department of Neurology, University of Münster, Münster, Germany,
| | | | | | | | | |
Collapse
|
16
|
Cahill LS, Laliberté CL, Liu XJ, Bishop J, Nieman BJ, Mogil JS, Sorge RE, Jones CD, Salter MW, Henkelman RM. Quantifying blood-spinal cord barrier permeability after peripheral nerve injury in the living mouse. Mol Pain 2014; 10:60. [PMID: 25216623 PMCID: PMC4190293 DOI: 10.1186/1744-8069-10-60] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2014] [Accepted: 09/10/2014] [Indexed: 11/16/2022] Open
Abstract
Background Genetic polymorphisms, gender and age all influence the risk of developing chronic neuropathic pain following peripheral nerve injury (PNI). It is known that there are significant inter-strain differences in pain hypersensitivity in strains of mice after PNI. In response to PNI, one of the earliest events is thought to be the disruption of the blood-spinal cord barrier (BSCB). The study of BSCB integrity after PNI may lead to a better understanding of the mechanisms that contribute to chronic pain. Results Here we used in vivo dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) to establish a timecourse for BSCB permeability following PNI, produced by performing a spared nerve injury (SNI). From this longitudinal study, we found that the SNI group had a significant increase in BSCB permeability over time throughout the entire spinal cord. The BSCB opening had a delayed onset and the increase in permeability was transient, returning to control levels just over one day after the surgery. We also examined inter-strain differences in BSCB permeability using five mouse strains (B10, C57BL/6J, CD-1, A/J and BALB/c) that spanned the range of pain hypersensitivity. We found a significant increase in BSCB permeability in the SNI group that was dependent on strain but that did not correlate with the reported strain differences in PNI-induced tactile hypersensitivity. These results were consistent with a previous experiment using Evans Blue dye to independently assess the status of the BSCB permeability. Conclusions DCE-MRI provides a sensitive and non-invasive method to follow BSCB permeability in the same group of mice over time. Examining differences between mouse strains, we demonstrated that there is an important genetically-based control of the PNI-induced increase in BSCB permeability and that the critical genetic determinants of BSCB opening after PNI are distinct from those that determine genetic variability in PNI-induced pain hypersensitivity. Electronic supplementary material The online version of this article (doi:10.1186/1744-8069-10-60) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Lindsay S Cahill
- Mouse Imaging Centre, Hospital for Sick Children, 25 Orde Street, Toronto, Ontario, Canada.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Nilius B, Szallasi A. Transient Receptor Potential Channels as Drug Targets: From the Science of Basic Research to the Art of Medicine. Pharmacol Rev 2014; 66:676-814. [DOI: 10.1124/pr.113.008268] [Citation(s) in RCA: 348] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
|
18
|
Schattling B, Eggert B, Friese MA. Acquired channelopathies as contributors to development and progression of multiple sclerosis. Exp Neurol 2014; 262 Pt A:28-36. [PMID: 24656770 DOI: 10.1016/j.expneurol.2013.12.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Accepted: 12/13/2013] [Indexed: 12/18/2022]
Abstract
Multiple sclerosis (MS), the most frequent inflammatory disease of the central nervous system (CNS), affects about two and a half million individuals worldwide and causes major burdens to the patients, which develop the disease usually at the age of 20 to 40. MS is likely referable to a breakdown of immune cell tolerance to CNS self-antigens resulting in focal immune cell infiltration, activation of microglia and astrocytes, demyelination and axonal and neuronal loss. Here we discuss how altered expression patterns and dysregulated functions of ion channels contribute on a molecular level to nearly all pathophysiological steps of the disease. In particular the detrimental redistribution of ion channels along axons, as well as neuronal excitotoxicity with regard to imbalanced glutamate homeostasis during chronic CNS inflammation will be discussed in detail. Together, we describe which ion channels in the immune and nervous system commend as attractive future drugable targets in MS treatment.
Collapse
Affiliation(s)
- Benjamin Schattling
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Falkenried 94, D-20251 Hamburg, Germany
| | - Britta Eggert
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Falkenried 94, D-20251 Hamburg, Germany
| | - Manuel A Friese
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Falkenried 94, D-20251 Hamburg, Germany.
| |
Collapse
|
19
|
Sisay S, Pryce G, Jackson SJ, Tanner C, Ross RA, Michael GJ, Selwood DL, Giovannoni G, Baker D. Genetic background can result in a marked or minimal effect of gene knockout (GPR55 and CB2 receptor) in experimental autoimmune encephalomyelitis models of multiple sclerosis. PLoS One 2013; 8:e76907. [PMID: 24130809 PMCID: PMC3793915 DOI: 10.1371/journal.pone.0076907] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Accepted: 08/26/2013] [Indexed: 12/20/2022] Open
Abstract
Endocannabinoids and some phytocannabinoids bind to CB1 and CB2 cannabinoid receptors, transient receptor potential vanilloid one (TRPV1) receptor and the orphan G protein receptor fifty-five (GPR55). Studies using C57BL/10 and C57BL/6 (Cnr2tm1Zim) CB2 cannabinoid receptor knockout mice have demonstrated an immune-augmenting effect in experimental autoimmune encephalomyelitis (EAE) models of multiple sclerosis. However, other EAE studies in Biozzi ABH mice often failed to show any treatment effect of either CB2 receptor agonism or antagonism on inhibition of T cell autoimmunity. The influence of genetic background on the induction of EAE in endocannabinoid system-related gene knockout mice was examined. It was found that C57BL/6.GPR55 knockout mice developed less severe disease, notably in female mice, following active induction with myelin oligodendrocyte glycoprotein 35-55 peptide. In contrast C57BL/6.CB2 (Cnr2Dgen) receptor knockout mice developed augmented severity of disease consistent with the genetically and pharmacologically-distinct, Cnr2tm1Zim mice. However, when the knockout gene was bred into the ABH mouse background and EAE induced with spinal cord autoantigens the immune-enhancing effect of CB2 receptor deletion was lost. Likewise CB1 receptor and transient receptor potential vanilloid one knockout mice on the ABH background demonstrated no alteration in immune-susceptibility, in terms of disease incidence and severity of EAE, in contrast to that reported in some C57BL/6 mouse studies. Furthermore the immune-modulating influence of GPR55 was marginal on the ABH mouse background. Whilst sedative doses of tetrahydrocannabinol could induce immunosuppression, this was associated with a CB1 receptor rather than a CB2 receptor-mediated effect. These data support the fact that non-psychoactive doses of medicinal cannabis have a marginal influence on the immune response in MS. Importantly, it adds a note of caution for the translational value of some transgenic/gene knockout and other studies on low-EAE susceptibility backgrounds with inconsistent disease course and susceptibility.
Collapse
MESH Headings
- Animals
- Disease Models, Animal
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Female
- Gene Deletion
- Gene Knockout Techniques
- Immunomodulation/genetics
- Male
- Mice
- Multiple Sclerosis/genetics
- Multiple Sclerosis/immunology
- Phenotype
- Receptor, Cannabinoid, CB2/deficiency
- Receptor, Cannabinoid, CB2/genetics
- Receptors, Cannabinoid/deficiency
- Receptors, Cannabinoid/genetics
- Species Specificity
Collapse
Affiliation(s)
- Sofia Sisay
- Neuroimmunology, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Gareth Pryce
- Neuroimmunology, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Samuel J. Jackson
- Neuroimmunology, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Carolyn Tanner
- School of Medical Science, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, United Kingdom
| | - Ruth A. Ross
- School of Medical Science, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, United Kingdom
- Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Gregory J. Michael
- Neuroimmunology, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - David L. Selwood
- Biological and Medical Chemistry, the Wolfson Institute for Biomedical Research, University College London, London, United Kingdom
| | - Gavin Giovannoni
- Neuroimmunology, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - David Baker
- Neuroimmunology, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
- * E-mail:
| |
Collapse
|