1
|
Liu Y, Wang S, Han M, Luo S, Chu P, Tang X, Zhao C, Han S, Yin S, Wang T. 17α-methyltestosterone exposure disrupted growth, liver physiology and intestinal microbial on fish: A case study on migratory bony fishes (Takifugu fasciatus). MARINE POLLUTION BULLETIN 2025; 212:117533. [PMID: 39778389 DOI: 10.1016/j.marpolbul.2025.117533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 01/02/2025] [Accepted: 01/03/2025] [Indexed: 01/11/2025]
Abstract
17α-methyltestosterone (17α-MT) is prevalent in the aqueous environment, but its toxicological profile remains incomplete. This study analyzed the effects of different 17α-MT concentrations on the growth performance, mortality, sex ratio, liver physiological metabolism, and intestinal microorganisms of Takifugu fasciatus, and on the microorganisms composition of its culture environment. Results showed that 17α-MT increased the male ratio and mortality but inhibited the growth of T. fasciatus. Meanwhile, 17α-MT induced oxidative stress and mitochondrial autophagy, changed lipid metabolism and inhibited triglyceride (TG) accumulation in the liver. 17α-MT altered the composition of the intestinal microorganisms, notably increasing the number of pathogenic bacteria. Additionally, the Firmicutes to the Bacteroidetes (F/B) ratio, associated with lipid metabolism, decreased with increasing 17α-MT concentration. Furthermore, 17α-MT disrupted the connection between the intestine microorganisms in T. fasciatus and the water column. This study enhances understanding of the effect of 17α-MT on T. fasciatus and the toxicological profile of 17α-MT.
Collapse
Affiliation(s)
- Yuxi Liu
- College of Marine Science and Engineering, Jiangsu Province Engineering Research Center for Aquatic Animals Breeding and Green Efficient Aquacultural Technology, Nanjing Normal University, Nanjing 210023, China; Co-Innovation Center for Marine Bio-Industry Technology of Jiangsu Province, Lianyungang, Jiangsu 222005, China
| | - Sijin Wang
- College of Marine Science and Engineering, Jiangsu Province Engineering Research Center for Aquatic Animals Breeding and Green Efficient Aquacultural Technology, Nanjing Normal University, Nanjing 210023, China; Co-Innovation Center for Marine Bio-Industry Technology of Jiangsu Province, Lianyungang, Jiangsu 222005, China
| | - Mengbo Han
- College of Marine Science and Engineering, Jiangsu Province Engineering Research Center for Aquatic Animals Breeding and Green Efficient Aquacultural Technology, Nanjing Normal University, Nanjing 210023, China; Co-Innovation Center for Marine Bio-Industry Technology of Jiangsu Province, Lianyungang, Jiangsu 222005, China
| | - Sheng Luo
- College of Marine Science and Engineering, Jiangsu Province Engineering Research Center for Aquatic Animals Breeding and Green Efficient Aquacultural Technology, Nanjing Normal University, Nanjing 210023, China; Co-Innovation Center for Marine Bio-Industry Technology of Jiangsu Province, Lianyungang, Jiangsu 222005, China
| | - Peng Chu
- College of Marine Science and Engineering, Jiangsu Province Engineering Research Center for Aquatic Animals Breeding and Green Efficient Aquacultural Technology, Nanjing Normal University, Nanjing 210023, China; Co-Innovation Center for Marine Bio-Industry Technology of Jiangsu Province, Lianyungang, Jiangsu 222005, China
| | - Xiaodong Tang
- College of Marine Science and Engineering, Jiangsu Province Engineering Research Center for Aquatic Animals Breeding and Green Efficient Aquacultural Technology, Nanjing Normal University, Nanjing 210023, China; Co-Innovation Center for Marine Bio-Industry Technology of Jiangsu Province, Lianyungang, Jiangsu 222005, China
| | - Cheng Zhao
- College of Marine Science and Engineering, Jiangsu Province Engineering Research Center for Aquatic Animals Breeding and Green Efficient Aquacultural Technology, Nanjing Normal University, Nanjing 210023, China; Co-Innovation Center for Marine Bio-Industry Technology of Jiangsu Province, Lianyungang, Jiangsu 222005, China.
| | - Shiqun Han
- Institution of Agricultural Resource and Environmental Sciences, Jiangsu Academy of Agricultural Sciences, Nanjing, Jiangsu 210014, China
| | - Shaowu Yin
- College of Marine Science and Engineering, Jiangsu Province Engineering Research Center for Aquatic Animals Breeding and Green Efficient Aquacultural Technology, Nanjing Normal University, Nanjing 210023, China; Co-Innovation Center for Marine Bio-Industry Technology of Jiangsu Province, Lianyungang, Jiangsu 222005, China
| | - Tao Wang
- College of Marine Science and Engineering, Jiangsu Province Engineering Research Center for Aquatic Animals Breeding and Green Efficient Aquacultural Technology, Nanjing Normal University, Nanjing 210023, China; Co-Innovation Center for Marine Bio-Industry Technology of Jiangsu Province, Lianyungang, Jiangsu 222005, China.
| |
Collapse
|
2
|
Guy AT, Kamiguchi H. Lipids as new players in axon guidance and circuit development. Curr Opin Neurobiol 2020; 66:22-29. [PMID: 33039927 DOI: 10.1016/j.conb.2020.09.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 07/27/2020] [Accepted: 09/01/2020] [Indexed: 11/24/2022]
Abstract
The formation of functional neuronal circuitry depends on axon guidance, in which extracellular chemotropic cues provide directional instructions to developing axons in the embryonic nervous system. Recently lipids, in particular lysolipids, are being appraised as a new class of axon guidance cues. Most lysolipids are formed by enzymatic hydrolysis of membrane phospholipids, and signal via a wide variety of mechanisms including specific G protein-coupled receptors. For example, lysophosphatidylglucoside released from a specific type of glia activates neuronal GPR55 to regulate axon tract patterning. However, demonstrating the in vivo mechanisms of lysolipid axon guidance is often challenging and complex. Here we will review in detail lysolipids that have been identified or proposed as axon guidance cues in the developing nervous system.
Collapse
Affiliation(s)
- Adam T Guy
- RIKEN Center for Brain Science, 2-1 Hirosawa, Wako City, Saitama, 351-0198 Japan
| | - Hiroyuki Kamiguchi
- RIKEN Center for Brain Science, 2-1 Hirosawa, Wako City, Saitama, 351-0198 Japan.
| |
Collapse
|
3
|
Yan Y, Du Z, Chen C, Li J, Xiong X, Zhang Y, Jiang H. Lysophospholipid profiles of apolipoprotein E-deficient mice reveal potential lipid biomarkers associated with atherosclerosis progression using validated UPLC-QTRAP-MS/MS-based lipidomics approach. J Pharm Biomed Anal 2019; 171:148-157. [PMID: 30999225 DOI: 10.1016/j.jpba.2019.03.062] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 03/25/2019] [Accepted: 03/27/2019] [Indexed: 01/19/2023]
Abstract
Lysophospholipids (Lyso-PLs) are lipid-derived signaling molecules which were demonstrated to have a strong correlation with the progression of atherosclerosis. In this study, we investigated the influence of high-fat diet on Lyso-PL profiles of atherosclerosis-prone apolipoprotein E-deficient (ApoE-/-) mice and wild type C57BL/6 J mice to find out the potential biomarkers associated with atherosclerosis. Firstly, the quantitative profiling method for Lyso-PLs based on ultra-performance liquid chromatography-quadrupole linear ion trap mass spectrometry (UPLC-QTRAP-MS/MS) was established and validated. Secondly, this method was utilized to quantify 169 targeted Lyso-PLs in plasma samples of ApoE-/- mice and wild type C57BL/6 J mice collected at different time points. Finally, 12 of 37 differential Lyso-PLs were identified as more reliable biomarkers by integrating static metabolomics and time-dependent analyses, among which Lyso-PC/15:0, 18:1/Lyso-PI, 22:5/Lyso-PI and 22:4/Lyso-PI were highly correlated with TCand LDL-C levels. Meanwhile, we found that the Lyso-PL profiles of ApoE-/- mice and C57BL/6 J mice were distinguished by altered metabolism of different Lyso-PLs classes, while C57BL/6 J mice fed with high-fat diet and normal diet were discriminated by the content differences of Lyso-PLs with same fatty acid composition. In conclusion, these results provided detailed changes of Lyso-PL profiles associated with atherosclerosis and the differential Lyso-PLs with reasonable change trends may serve as promising biomarkers for atherosclerosis progression.
Collapse
Affiliation(s)
- Yingfei Yan
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan, China
| | - Zhifeng Du
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan, China
| | - Chang Chen
- Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Jiaxin Li
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan, China
| | - Xiang Xiong
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan, China
| | - Yang Zhang
- Department of Pharmacy, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China.
| | - Hongliang Jiang
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
4
|
Guerrero-Alba R, Barragán-Iglesias P, González-Hernández A, Valdez-Moráles EE, Granados-Soto V, Condés-Lara M, Rodríguez MG, Marichal-Cancino BA. Some Prospective Alternatives for Treating Pain: The Endocannabinoid System and Its Putative Receptors GPR18 and GPR55. Front Pharmacol 2019; 9:1496. [PMID: 30670965 PMCID: PMC6331465 DOI: 10.3389/fphar.2018.01496] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 12/07/2018] [Indexed: 12/12/2022] Open
Abstract
Background: Marijuana extracts (cannabinoids) have been used for several millennia for pain treatment. Regarding the site of action, cannabinoids are highly promiscuous molecules, but only two cannabinoid receptors (CB1 and CB2) have been deeply studied and classified. Thus, therapeutic actions, side effects and pharmacological targets for cannabinoids have been explained based on the pharmacology of cannabinoid CB1/CB2 receptors. However, the accumulation of confusing and sometimes contradictory results suggests the existence of other cannabinoid receptors. Different orphan proteins (e.g., GPR18, GPR55, GPR119, etc.) have been proposed as putative cannabinoid receptors. According to their expression, GPR18 and GPR55 could be involved in sensory transmission and pain integration. Methods: This article reviews select relevant information about the potential role of GPR18 and GPR55 in the pathophysiology of pain. Results: This work summarized novel data supporting that, besides cannabinoid CB1 and CB2 receptors, GPR18 and GPR55 may be useful for pain treatment. Conclusion: There is evidence to support an antinociceptive role for GPR18 and GPR55.
Collapse
Affiliation(s)
- Raquel Guerrero-Alba
- Departamento de Fisiología y Farmacología, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes, Aguascalientes, Mexico
| | - Paulino Barragán-Iglesias
- School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, United States
| | - Abimael González-Hernández
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Santiago de Querétaro, Mexico
| | - Eduardo E Valdez-Moráles
- Cátedras CONACYT, Departamento de Cirugía, Centro de Ciencias Biomédicas, Universidad Autónoma de Aguascalientes, Aguascalientes, Mexico
| | - Vinicio Granados-Soto
- Neurobiology of Pain Laboratory, Departamento de Farmacobiología, Cinvestav, Mexico City, Mexico
| | - Miguel Condés-Lara
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Santiago de Querétaro, Mexico
| | - Martín G Rodríguez
- Departamento de Fisiología y Farmacología, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes, Aguascalientes, Mexico
| | - Bruno A Marichal-Cancino
- Departamento de Fisiología y Farmacología, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes, Aguascalientes, Mexico
| |
Collapse
|
5
|
Abstract
Cannabinoids influence cardiovascular variables in health and disease via multiple mechanisms. The chapter covers the impact of cannabinoids on cardiovascular function in physiology and pathology and presents a critical analysis of the proposed signalling pathways governing regulation of cardiovascular function by endogenously produced and exogenous cannabinoids. We know that endocannabinoid system is overactivated under pathological conditions and plays both a protective compensatory role, such as in some forms of hypertension, atherosclerosis and other inflammatory conditions, and a pathophysiological role, such as in disease states associated with excessive hypotension. This chapter focuses on the mechanisms affecting hemodynamics and vasomotor effects of cannabinoids in health and disease states, highlighting mismatches between some studies. The chapter will first review the effects of marijuana smoking on cardiovascular system and then describe the impact of exogenous cannabinoids on cardiovascular parameters in humans and experimental animals. This will be followed by analysis of the impact of cannabinoids on reactivity of isolated vessels. The article critically reviews current knowledge on cannabinoid induction of vascular relaxation by cannabinoid receptor-dependent and -independent mechanisms and dysregulation of vascular endocannabinoid signaling in disease states.
Collapse
Affiliation(s)
- Alexander I Bondarenko
- Circulatory Physiology Department, Bogomoletz Institute of Physiology National Academy of Sciences of Ukraine, Kiev, Ukraine.
| |
Collapse
|
6
|
Alhouayek M, Masquelier J, Muccioli GG. Lysophosphatidylinositols, from Cell Membrane Constituents to GPR55 Ligands. Trends Pharmacol Sci 2018; 39:586-604. [PMID: 29588059 DOI: 10.1016/j.tips.2018.02.011] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Revised: 02/20/2018] [Accepted: 02/28/2018] [Indexed: 12/25/2022]
Abstract
Lysophosphatidylinositols (LPIs) are membrane constituents that alter the properties of said membranes. However, recent data showing that the once orphan receptor, GPR55, can act as a receptor for LPIs has sparked a renewed interest in LPIs as bioactive lipids. As evidence supporting the importance of LPIs and/or GPR55 is continuously accumulating and because LPI levels are altered in a number of pathologies such as obesity and cancer, the coming years should bring new, exciting discoveries to this field. In this review, we discuss the recent work on LPIs and on their molecular target, the GPR55 receptor. First, we summarize the metabolism of LPIs before outlining the cellular pathways activated by GPR55. Then, we review the actions of LPIs and GPR55 that could have potential pharmacological or therapeutic applications in several pathophysiological settings, such as cancer, obesity, pain, and inflammation.
Collapse
Affiliation(s)
- Mireille Alhouayek
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, Université catholique de Louvain, 1200 Bruxelles, Belgium; These authors contributed equally to this work
| | - Julien Masquelier
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, Université catholique de Louvain, 1200 Bruxelles, Belgium; These authors contributed equally to this work
| | - Giulio G Muccioli
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, Université catholique de Louvain, 1200 Bruxelles, Belgium.
| |
Collapse
|
7
|
Bondarenko AI, Panasiuk O, Drachuk K, Montecucco F, Brandt KJ, Mach F. The quest for endothelial atypical cannabinoid receptor: BK Ca channels act as cellular sensors for cannabinoids in in vitro and in situ endothelial cells. Vascul Pharmacol 2018; 102:44-55. [PMID: 29355732 PMCID: PMC6481560 DOI: 10.1016/j.vph.2018.01.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 01/10/2018] [Accepted: 01/16/2018] [Indexed: 12/20/2022]
Abstract
Endothelium-dependent component of cannabinoid-induced vasodilation has been postulated to require G-protein-coupled non-CB1/CB2 endothelial cannabinoid (eCB) receptor. GPR18 was proposed as a candidate for eCBR. To address the hypothesis that the effects attributed to eCBR are mediated by G-protein-coupled receptor (GPCR)-independent targets, we studied the electrical responses in endothelial cells, focusing on BKCa channels. In patches excised from endothelial-derived EA.hy926 cells, N-arachidonoyl glycine (NAGly) and abnormal cannabidiol (abn-cbd), prototypical agonists for eCB receptor, stimulate single BKCa activity in a concentration- and Ca2+-dependent manner. The postulated eCB receptor inhibitors rimonabant and AM251 were found to inhibit basal and stimulated by NAGly- and abn-cbd BKCa activity in cell-free patches. In isolated mice aortas, abn-cbd and NAGly produced endothelial cell hyperpolarization that was sensitive to paxilline, a selective BKCa inhibitor, but not to GPR18 antibody, and mimicked by NS1619, a direct BKCa opener. In excised patches from mice aortic endothelium, single channel activity with characteristics similar to BKCa was established by the addition of abn-cbd and NAGly. We conclude that the two cannabinoids abn-cbd and NAGly initiate a GPR18-independent activation of BKCa channels in mice aortic endothelial cells that might contribute to vasodilation to cannabinoids.
Collapse
Affiliation(s)
- Alexander I Bondarenko
- Circulatory Physiology Department, Bogomoletz Institute of Physiology NAS of Ukraine, Bogomoletz Str 4, Kiev 01024, Ukraine; Institute of Molecular Biology and Biochemistry, Medical University of Graz, Harrachgasse 21/III, Graz, 8010, Austria.
| | - Olga Panasiuk
- Circulatory Physiology Department, Bogomoletz Institute of Physiology NAS of Ukraine, Bogomoletz Str 4, Kiev 01024, Ukraine
| | - Konstantin Drachuk
- Circulatory Physiology Department, Bogomoletz Institute of Physiology NAS of Ukraine, Bogomoletz Str 4, Kiev 01024, Ukraine
| | - Fabrizio Montecucco
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy; Ospedale Policlinico San Martino, largo Benzi 10, 16132 Genoa, Italy; Centre of Excellence for Biomedical Research (CEBR), University of Genoa, 9 viale Benedetto XV, 16132 Genoa, Italy
| | - Karim J Brandt
- Division of Cardiology, Foundation for Medical Researches, Department of Internal Medicine, University of Geneva, Av. de la Roseraie 64, CH 1211, Geneva 4, Switzerland
| | - François Mach
- Division of Cardiology, Foundation for Medical Researches, Department of Internal Medicine, University of Geneva, Av. de la Roseraie 64, CH 1211, Geneva 4, Switzerland
| |
Collapse
|
8
|
Karpińska O, Baranowska-Kuczko M, Malinowska B, Kloza M, Kusaczuk M, Gęgotek A, Golec P, Kasacka I, Kozłowska H. Mechanisms of l-alpha-lysophosphatidylinositol-induced relaxation in human pulmonary arteries. Life Sci 2017; 192:38-45. [PMID: 29155298 DOI: 10.1016/j.lfs.2017.11.020] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Revised: 11/11/2017] [Accepted: 11/15/2017] [Indexed: 01/13/2023]
Abstract
AIMS l-Alpha-lysophosphatidylinositol (LPI) is an endogenous agonist of G protein-coupled receptor 55 (GPR55) which relaxes mesenteric arteries on activation. The aim of the present study was to determine the influence and underlying mechanisms of LPI-induced relaxation in human pulmonary arteries (hPAs). MAIN METHODS Functional studies were performed in isolated hPAs using organ bath technique. The expression of GPR55 in hPAs and bronchioles was determined by real-time qPCR, Western blot analysis, and immunohistochemistry. KEY FINDINGS LPI induced a concentration-dependent vasorelaxation in endothelium-intact hPAs. This effect was attenuated by the GPR55 antagonist CID16020046, the peroxisome proliferator-activated receptor-γ (PPARγ) antagonist GW9662, the putative endothelial cannabinoid receptor (CBe) antagonist O-1918 and the inhibitor of nitric oxide (NO) synthase (L-NAME). In addition, vasorelaxation was also attenuated by the presence of a high KCl concentration, selective blockers of small (KCa2.3; UCL1684), intermediate (KCa3.1; TRAM-34) and large conductance (KCa1.1; iberiotoxin) calcium-activated potassium channels and by endothelium denudation. However, vasorelaxation was not attenuated by the cannabinoid CB1 receptor antagonist AM251 or by the cyclooxygenase inhibitor indomethacin. SIGNIFICANCE The study showed that the LPI-induced vasorelaxation was endothelium-dependent and mediated by GPR55, PPARγ and CBe receptors, occurred in a NO- and calcium-activated potassium channel-dependent manner in isolated hPAs. LPI seems to possess positive, hypotensive properties in pulmonary vascular bed.
Collapse
Affiliation(s)
- Olga Karpińska
- Department of Experimental Physiology and Pathophysiology, Medical University of Białystok, Mickiewicza Str. 2A, 15-089 Białystok, Poland.
| | - Marta Baranowska-Kuczko
- Department of Experimental Physiology and Pathophysiology, Medical University of Białystok, Mickiewicza Str. 2A, 15-089 Białystok, Poland
| | - Barbara Malinowska
- Department of Experimental Physiology and Pathophysiology, Medical University of Białystok, Mickiewicza Str. 2A, 15-089 Białystok, Poland
| | - Monika Kloza
- Department of Experimental Physiology and Pathophysiology, Medical University of Białystok, Mickiewicza Str. 2A, 15-089 Białystok, Poland
| | - Magdalena Kusaczuk
- Department of Pharmaceutical Biochemistry, Medical University of Białystok, Mickiewicza Str. 2A, 15-089 Białystok, Poland
| | - Agnieszka Gęgotek
- Department of Inorganic and Analytical Chemistry, Medical University of Białystok, Mickiewicza Str. 2D, 15-222 Białystok, Poland
| | - Paweł Golec
- Department of Thoracic Surgery, Medical University of Białystok, M. Skłodowskiej-Curie Str. 24A, 15-276 Białystok, Poland
| | - Irena Kasacka
- Department of Histology and Cytophysiology, Medical University of Białystok, Mickiewicza Str. 2C, 15-222 Białystok, Poland
| | - Hanna Kozłowska
- Department of Experimental Physiology and Pathophysiology, Medical University of Białystok, Mickiewicza Str. 2A, 15-089 Białystok, Poland
| |
Collapse
|
9
|
Baker D, Pryce G, Visintin C, Sisay S, Bondarenko AI, Vanessa Ho WS, Jackson SJ, Williams TE, Al-Izki S, Sevastou I, Okuyama M, Graier WF, Stevenson LA, Tanner C, Ross R, Pertwee RG, Henstridge CM, Irving AJ, Schulman J, Powell K, Baker MD, Giovannoni G, Selwood DL. Big conductance calcium-activated potassium channel openers control spasticity without sedation. Br J Pharmacol 2017; 174:2662-2681. [PMID: 28677901 PMCID: PMC5522996 DOI: 10.1111/bph.13889] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 04/27/2017] [Accepted: 05/17/2017] [Indexed: 12/19/2022] Open
Abstract
Background and Purpose Our initial aim was to generate cannabinoid agents that control spasticity, occurring as a consequence of multiple sclerosis (MS), whilst avoiding the sedative side effects associated with cannabis. VSN16R was synthesized as an anandamide (endocannabinoid) analogue in an anti‐metabolite approach to identify drugs that target spasticity. Experimental Approach Following the initial chemistry, a variety of biochemical, pharmacological and electrophysiological approaches, using isolated cells, tissue‐based assays and in vivo animal models, were used to demonstrate the activity, efficacy, pharmacokinetics and mechanism of action of VSN16R. Toxicological and safety studies were performed in animals and humans. Key Results VSN16R had nanomolar activity in tissue‐based, functional assays and dose‐dependently inhibited spasticity in a mouse experimental encephalomyelitis model of MS. This effect occurred with over 1000‐fold therapeutic window, without affecting normal muscle tone. Efficacy was achieved at plasma levels that are feasible and safe in humans. VSN16R did not bind to known CB1/CB2/GPPR55 cannabinoid‐related receptors in receptor‐based assays but acted on a vascular cannabinoid target. This was identified as the major neuronal form of the big conductance, calcium‐activated potassium (BKCa) channel. Drug‐induced opening of neuronal BKCa channels induced membrane hyperpolarization, limiting excessive neural‐excitability and controlling spasticity. Conclusions and Implications We identified the neuronal form of the BKCa channel as the target for VSN16R and demonstrated that its activation alleviates neuronal excitability and spasticity in an experimental model of MS, revealing a novel mechanism to control spasticity. VSN16R is a potential, safe and selective ligand for controlling neural hyper‐excitability in spasticity.
Collapse
Affiliation(s)
- David Baker
- Neuroimmunology Unit, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK.,Department of Neuroinflammation, UCL Institute of Neurology, University College London, London, UK
| | - Gareth Pryce
- Neuroimmunology Unit, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK.,Department of Neuroinflammation, UCL Institute of Neurology, University College London, London, UK
| | - Cristina Visintin
- Department of Neuroinflammation, UCL Institute of Neurology, University College London, London, UK.,Department of Medicinal Chemistry, UCL Wolfson Institute for Biomedical Research, University College London, London, UK
| | - Sofia Sisay
- Neuroimmunology Unit, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Alexander I Bondarenko
- Institute of Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria.,A.A. Bogomoletz Institute of Physiology, Kiev, Ukraine
| | - W S Vanessa Ho
- Vascular Biology Research Centre. St. George's, University of London, London, UK
| | - Samuel J Jackson
- Neuroimmunology Unit, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Thomas E Williams
- Neuroimmunology Unit, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Sarah Al-Izki
- Neuroimmunology Unit, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Ioanna Sevastou
- Department of Medicinal Chemistry, UCL Wolfson Institute for Biomedical Research, University College London, London, UK
| | - Masahiro Okuyama
- Department of Medicinal Chemistry, UCL Wolfson Institute for Biomedical Research, University College London, London, UK
| | - Wolfgang F Graier
- Institute of Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Lesley A Stevenson
- Vascular Biology Research Centre. St. George's, University of London, London, UK
| | - Carolyn Tanner
- Department of Biomedical Sciences, Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
| | - Ruth Ross
- Department of Biomedical Sciences, Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
| | - Roger G Pertwee
- Department of Biomedical Sciences, Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
| | - Christopher M Henstridge
- Neurosciences Institute, Division of Pathology and Neuroscience, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | - Andrew J Irving
- Neurosciences Institute, Division of Pathology and Neuroscience, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | - Jesse Schulman
- Canbex Therapeutics Ltd, London BioScience Innovation Centre, London, UK
| | - Keith Powell
- Canbex Therapeutics Ltd, London BioScience Innovation Centre, London, UK
| | - Mark D Baker
- Neuroimmunology Unit, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Gavin Giovannoni
- Neuroimmunology Unit, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK.,Department of Neuroinflammation, UCL Institute of Neurology, University College London, London, UK
| | - David L Selwood
- Department of Medicinal Chemistry, UCL Wolfson Institute for Biomedical Research, University College London, London, UK
| |
Collapse
|
10
|
Direct activation of Ca 2+ and voltage-gated potassium channels of large conductance by anandamide in endothelial cells does not support the presence of endothelial atypical cannabinoid receptor. Eur J Pharmacol 2017; 805:14-24. [PMID: 28327344 DOI: 10.1016/j.ejphar.2017.03.038] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 03/15/2017] [Accepted: 03/17/2017] [Indexed: 11/23/2022]
Abstract
Endocannabinoid anandamide induces endothelium-dependent relaxation commonly attributed to stimulation of the G-protein coupled endothelial anandamide receptor. The study addressed the receptor-independent effect of anandamide on large conductance Ca2+-dependent K+ channels expressed in endothelial cell line EA.hy926. Under resting conditions, 10µM anandamide did not significantly influence the resting membrane potential. In a Ca2+-free solution the cells were depolarized by ~10mV. Further administration of 10µM anandamide hyperpolarized the cells by ~8mV. In voltage-clamp mode, anandamide elicited the outwardly rectifying whole-cell current sensitive to paxilline but insensitive to GDPβS, a G-protein inhibitor. Administration of 70µM Mn2+, an agent used to promote integrin clustering, reversibly stimulated whole-cell current, but failed to further facilitate the anandamide-stimulated current. In an inside-out configuration, anandamide (0.1-30µM) facilitated single BKCa channel activity in a concentration-dependent manner within a physiological Ca2+ range and a wide range of voltages, mainly by reducing mean closed time. The effect is essentially eliminated following chelation of Ca2+ from the cytosolic face and pre-exposure to cholesterol-reducing agent methyl-β-cyclodextrin. O-1918 (3µM), a cannabidiol analog used as a selective antagonist of endothelial anandamide receptor, reduced BKCa channel activity in inside-out patches. These results do not support the existence of endothelial cannabinoid receptor and indicate that anandamide acts as a direct BKCa opener. The action does not require cell integrity or integrins and is caused by direct modification of BKCa channel activity.
Collapse
|
11
|
Bondarenko AI, Montecucco F, Panasiuk O, Sagach V, Sidoryak N, Brandt KJ, Mach F. GPR55 agonist lysophosphatidylinositol and lysophosphatidylcholine inhibit endothelial cell hyperpolarization via GPR-independent suppression of Na +-Ca 2+ exchanger and endoplasmic reticulum Ca 2+ refilling. Vascul Pharmacol 2017; 89:39-48. [PMID: 28064014 DOI: 10.1016/j.vph.2017.01.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 12/21/2016] [Accepted: 01/03/2017] [Indexed: 11/18/2022]
Abstract
Lysophosphatidylinositol (LPI) and lysophosphatidylcholine (LPC) are lipid signaling molecules that induce endothelium-dependent vasodilation. In addition, LPC suppresses acetylcholine (Ach)-induced responses. We aimed to determine the influence of LPC and LPI on hyperpolarizing responses in vitro and in situ endothelial cells (EC) and identify the underlying mechanisms. Using patch-clamp method, we show that LPI and LPC inhibit EC hyperpolarization to histamine and suppress Na+/Ca2+ exchanged (NCX) currents in a concentration-dependent manner. The inhibition is non-mode-specific and unaffected by intracellular GDPβS infusion and tempol, a superoxide dismutase mimetic. In excised mouse aorta, LPI strongly inhibits the sustained and the peak endothelial hyperpolarization induced by Ach, but not by SKA-31, an opener of Ca2+-dependent K+ channels of intermediate and small conductance. The hyperpolarizing responses to consecutive histamine applications are strongly reduced by NCX inhibition. In a Ca2+-re-addition protocol, bepridil, a NCX inhibitor, and KB-R7943, a blocker of reversed NCX, inhibit the hyperpolarizing responses to Ca2+-re-addition following Ca2+ stores depletion. These finding indicate that LPC and LPI inhibit endothelial hyperpolarization to Ach and histamine independently of G-protein coupled receptors and superoxide anions. Reversed NCX is critical for ER Ca2+ refilling in EC. The inhibition of NCX by LPI and LPC underlies diminished endothelium-dependent responses and endothelial dysfunction accompanied by increased levels of these lipids in the blood.
Collapse
Affiliation(s)
- Alexander I Bondarenko
- Circulatory Physiology Department, Bogomoletz Institute of Physiology NAS of Ukraine, Bogomoletz Str.4, 01024 Kiev, Ukraine; Medical University of Graz, Institute of Molecular Biology and Biochemistry, Graz 8010, Austria.
| | - Fabrizio Montecucco
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy; IRCCS AOU San Martino - IST Istituto Nazionale per la Ricerca sul Cancro, Largo Benzi 10, 16132 Genoa, Italy
| | - Olga Panasiuk
- Circulatory Physiology Department, Bogomoletz Institute of Physiology NAS of Ukraine, Bogomoletz Str.4, 01024 Kiev, Ukraine
| | - Vadim Sagach
- Circulatory Physiology Department, Bogomoletz Institute of Physiology NAS of Ukraine, Bogomoletz Str.4, 01024 Kiev, Ukraine
| | - Nataliya Sidoryak
- Department of Physiology of Human and Animals, Melitopol State Pedagogical University, Ukraine
| | - Karim J Brandt
- Division of Cardiology, Foundation for Medical Researches, Department of Internal Medicine, University of Geneva, Av. de la Roseraie 64, CH-1211 Geneva 4, Switzerland
| | - François Mach
- Division of Cardiology, Foundation for Medical Researches, Department of Internal Medicine, University of Geneva, Av. de la Roseraie 64, CH-1211 Geneva 4, Switzerland
| |
Collapse
|
12
|
Liu B, Song S, Ruz-Maldonado I, Pingitore A, Huang GC, Baker D, Jones PM, Persaud SJ. GPR55-dependent stimulation of insulin secretion from isolated mouse and human islets of Langerhans. Diabetes Obes Metab 2016; 18:1263-1273. [PMID: 27561953 DOI: 10.1111/dom.12780] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2016] [Revised: 08/10/2016] [Accepted: 08/22/2016] [Indexed: 01/15/2023]
Abstract
AIMS The novel cannabinoid receptor GPR55 is expressed by rodent islets and it has been implicated in β-cell function in response to a range of ligands. This study evaluated the effects of GPR55 ligands on intracellular calcium ([Ca2+ ]i ) levels and insulin secretion from islets isolated from GPR55 knockout (GPR55 -/- ) mice, age-matched wildtype (WT) mice and human pancreas. MATERIALS AND METHODS GPR55 expression was determined by Western blotting and fluorescent immunohistochemistry. Changes in [Ca2+ ]i were measured by Fura-2 microfluorimetry. Dynamic insulin secretion was quantified by radioimmunoassay following perifusion of isolated islets. RhoA activity was monitored using a Rho binding domain pull down assay. RESULTS Western blotting indicated that MIN6 β-cells, mouse and human islets express GPR55 and its localization on human β-cells was demonstrated by fluorescent immunohistochemistry. The pharmacological GPR55 agonist O-1602 (10 μM) significantly stimulated [Ca2+ ]i and insulin secretion from WT mouse islets and these stimulatory effects were abolished in islets isolated from GPR55 -/- mice. In contrast, while the putative endogenous GPR55 agonist lysophosphatidylinositol (LPI, 5 µM) and the GPR55 antagonist cannabidiol (CBD, 1 µM) also elevated [Ca2+ ]i and insulin secretion, these effects were sustained in islets from GPR55 -/- mice. Stimulatory effects of O-1602 on [Ca2+ ]i and insulin secretion were also observed in experiments using human islets, but O-1602 did not activate RhoA in MIN6 β-cells. CONCLUSIONS Our results therefore suggest that GPR55 plays an important role in the regulation of mouse and human islet physiology, but LPI and CBD exert stimulatory effects on islet function by a GPR55-independent pathway(s).
Collapse
Affiliation(s)
- Bo Liu
- Division of Diabetes and Nutritional Sciences, Diabetes Research Group, King's College London, London, UK
| | - Shuang Song
- Division of Diabetes and Nutritional Sciences, Diabetes Research Group, King's College London, London, UK
| | - Inmaculada Ruz-Maldonado
- Division of Diabetes and Nutritional Sciences, Diabetes Research Group, King's College London, London, UK
| | - Attilio Pingitore
- Division of Diabetes and Nutritional Sciences, Diabetes Research Group, King's College London, London, UK
| | - Guo C Huang
- Division of Diabetes and Nutritional Sciences, Diabetes Research Group, King's College London, London, UK
| | - David Baker
- Centre for Neuroscience and Trauma, Barts and The London School of Medicine and Dentistry, London, UK
| | - Peter M Jones
- Division of Diabetes and Nutritional Sciences, Diabetes Research Group, King's College London, London, UK
| | - Shanta J Persaud
- Division of Diabetes and Nutritional Sciences, Diabetes Research Group, King's College London, London, UK
| |
Collapse
|
13
|
AL SULEIMANI YM, HILEY CR. Characterization of Calcium Signals Provoked by Lysophosphatidylinositol in Human Microvascular Endothelial Cells. Physiol Res 2016; 65:53-62. [DOI: 10.33549/physiolres.932962] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
The lipid molecule, lysophosphatidylinositol (LPI), is hypothesised to form part of a novel lipid signalling system that involves the G protein-coupled receptor GPR55 and distinct intracellular signalling cascades in endothelial cells. This work aimed to study the possible mechanisms involved in LPI-evoked cytosolic Ca2+ mobilization in human brain microvascular endothelial cells. Changes in intracellular Ca2+ concentrations were measured using cell population Ca2+ assay. LPI evoked biphasic elevation of intracellular calcium concentration, a rapid phase and a sustained phase. The rapid phase was attenuated by the inhibitor of PLC (U 73122), inhibitor of IP3 receptors, 2-APB and the depletor of endoplasmic reticulum Ca2+ store, thapsigargin. The sustained phase, on the other hand, was enhanced by U 73122 and abolished by the RhoA kinase inhibitor, Y-27632. In conclusion, the Ca2+ signal evoked by LPI is characterised by a rapid phase of Ca2+ release from the endoplasmic reticulum, and requires activation of the PLC-IP3 signalling pathway. The sustained phase mainly depends on RhoA kinase activation. LPI acts as novel lipid signalling molecule in endothelial cells, and elevation of cytosolic Ca2+ triggered by it may present an important intracellular message required in gene expression and controlling of vascular tone.
Collapse
Affiliation(s)
- Y. M. AL SULEIMANI
- Department of Pharmacology and Clinical Pharmacy, College of Medicine and Health Sciences, Sultan Qaboos University, Sultanate of Oman
| | | |
Collapse
|
14
|
Hofmann NA, Yang J, Trauger SA, Nakayama H, Huang L, Strunk D, Moses MA, Klagsbrun M, Bischoff J, Graier WF. The GPR 55 agonist, L-α-lysophosphatidylinositol, mediates ovarian carcinoma cell-induced angiogenesis. Br J Pharmacol 2015; 172:4107-18. [PMID: 25989290 PMCID: PMC4543616 DOI: 10.1111/bph.13196] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Revised: 04/10/2015] [Accepted: 05/11/2015] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND AND PURPOSE Highly vascularized ovarian carcinoma secretes the putative endocannabinoid and GPR55 agonist, L-α-lysophosphatidylinositol (LPI), into the circulation. We aimed to assess the involvement of this agonist and its receptor in ovarian cancer angiogenesis. EXPERIMENTAL APPROACH Secretion of LPI by three ovarian cancer cell lines (OVCAR-3, OVCAR-5 and COV-362) was tested by mass spectrometry. Involvement of cancer cell-derived LPI on angiogenesis was tested in the in vivo chicken chorioallantoic membrane (CAM) assay along with the assessment of the effect of LPI on proliferation, network formation, and migration of neonatal and adult human endothelial colony-forming cells (ECFCs). Engagement of GPR55 was verified by using its pharmacological inhibitor CID16020046 and diminution of GPR55 expression by four different target-specific siRNAs. To study underlying signal transduction, Western blot analysis was performed. KEY RESULTS Ovarian carcinoma cell-derived LPI stimulated angiogenesis in the CAM assay. Applied LPI stimulated proliferation, network formation, and migration of neonatal ECFCs in vitro and angiogenesis in the in vivo CAM. The pharmacological GPR55 inhibitor CID16020046 inhibited LPI-stimulated ECFC proliferation, network formation and migration in vitro as well as ovarian carcinoma cell- and LPI-induced angiogenesis in vivo. Four target-specific siRNAs against GPR55 prevented these effects of LPI on angiogenesis. These pro-angiogenic effects of LPI were transduced by GPR55-dependent phosphorylation of ERK1/2 and p38 kinase. CONCLUSIONS AND IMPLICATIONS We conclude that inhibiting the pro-angiogenic LPI/GPR55 pathway appears a promising target against angiogenesis in ovarian carcinoma.
Collapse
Affiliation(s)
- Nicole A Hofmann
- Institute for Molecular Biology and Biochemistry, Medical University GrazGraz, Austria
- Vascular Biology Program, Boston Children's HospitalBoston, MA, USA
- Department of Surgery, Harvard Medical SchoolBoston, MA, USA
| | - Jiang Yang
- Vascular Biology Program, Boston Children's HospitalBoston, MA, USA
- Department of Surgery, Harvard Medical SchoolBoston, MA, USA
| | - Sunia A Trauger
- FAS Small Molecule Mass Spectrometry Facility, Harvard UniversityBoston, MA, USA
| | - Hironao Nakayama
- Vascular Biology Program, Boston Children's HospitalBoston, MA, USA
- Department of Surgery, Harvard Medical SchoolBoston, MA, USA
| | - Lan Huang
- Vascular Biology Program, Boston Children's HospitalBoston, MA, USA
- Department of Surgery, Harvard Medical SchoolBoston, MA, USA
| | - Dirk Strunk
- Experimental and Clinical Cell Therapy Institute, Paracelsus Medical UniversitySalzburg, Austria
| | - Marsha A Moses
- Vascular Biology Program, Boston Children's HospitalBoston, MA, USA
- Department of Surgery, Harvard Medical SchoolBoston, MA, USA
| | - Michael Klagsbrun
- Vascular Biology Program, Boston Children's HospitalBoston, MA, USA
- Department of Surgery, Harvard Medical SchoolBoston, MA, USA
| | - Joyce Bischoff
- Department of Surgery, Harvard Medical SchoolBoston, MA, USA
- Department of Pathology, Harvard Medical SchoolBoston, MA, USA
| | - Wolfgang F Graier
- Institute for Molecular Biology and Biochemistry, Medical University GrazGraz, Austria
| |
Collapse
|
15
|
AlSuleimani YM, Hiley CR. The GPR55 agonist lysophosphatidylinositol relaxes rat mesenteric resistance artery and induces Ca(2+) release in rat mesenteric artery endothelial cells. Br J Pharmacol 2015; 172:3043-57. [PMID: 25652040 DOI: 10.1111/bph.13107] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2014] [Revised: 01/11/2015] [Accepted: 02/02/2015] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND AND PURPOSE Lysophosphatidylinositol (LPI), a lipid signalling molecule, activates GPR55 and elevates intracellular Ca(2+). Here, we examine the actions of LPI in the rat resistance mesenteric artery and Ca(2+) responses in endothelial cells isolated from the artery. EXPERIMENTAL APPROACH Vascular responses were studied using wire myographs. Single-cell fluorescence imaging was performed using a MetaFluor system. Hypotensive effects of LPI were assessed using a Biopac system. KEY RESULTS In isolated arteries, LPI-induced vasorelaxation was concentration- and endothelium-dependent and inhibited by CID 16020046, a GPR55 antagonist. The CB1 receptor antagonist AM 251 had no effect, whereas rimonabant and O-1918 significantly potentiated LPI responses. Vasorelaxation was reduced by charybdotoxin and iberiotoxin, alone or combined. LPI decreased systemic arterial pressure. GPR55 is expressed in rat mesenteric artery. LPI caused biphasic elevations of endothelial cell intracellular Ca(2+). Pretreatment with thapsigargin or 2-aminoethoxydiphenyl borate abolished both phases. The PLC inhibitor U73122 attenuated the initial phase and enhanced the second phase, whereas the Rho-associated kinase inhibitor Y-27632 abolished the late phase but not the early phase. CONCLUSIONS AND IMPLICATIONS LPI is an endothelium-dependent vasodilator in the rat small mesenteric artery and a hypotensive agent. The vascular response involves activation of Ca(2+)-sensitive K(+) channels and is not mediated by CB1 receptors, but unexpectedly enhanced by antagonists of the 'endothelial anandamide' receptor. In endothelial cells, LPI utilizes PLC-IP3 and perhaps ROCK-RhoA pathways to elevate intracellular Ca(2+). Overall, these findings support an endothelial site of action for LPI and suggest a possible role for GPR55 in vasculature.
Collapse
Affiliation(s)
- Y M AlSuleimani
- Department of Pharmacology, University of Cambridge, Cambridge, UK.,Department of Pharmacology and Clinical Pharmacy, College of Medicine and Health Sciences, Sultan Qaboos University, Alkoudh, Sultanate of Oman
| | - C R Hiley
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| |
Collapse
|
16
|
Liu B, Song S, Jones PM, Persaud SJ. GPR55: from orphan to metabolic regulator? Pharmacol Ther 2015; 145:35-42. [PMID: 24972076 DOI: 10.1016/j.pharmthera.2014.06.007] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Accepted: 06/18/2014] [Indexed: 12/26/2022]
Abstract
GPR55 belongs to the class A family of G-protein coupled receptor (GPCRs) and its activity is regulated by a range of synthetic and endogenous cannabinoids, and by lipid-derived ligands. Cannabinoids are known to be important in controlling appetite and metabolic balance, and it is now emerging that GPR55 may have a role to play in energy homeostasis through the regulation of food intake, fuel storage in adipocytes, gut motility and insulin secretion. This review summarises our current knowledge of expression and function of GPR55 in tissues involved in metabolic regulation, the signalling cascades through which GPR55 is reported to act to exert its effects, and it comments on the difficulties in reaching firm conclusions when using GPR55 ligands of poor specificity. Understanding the role of GPR55 in energy homeostasis may provide a novel target for therapeutic intervention in obesity and type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Bo Liu
- Diabetes Research Group, Division of Diabetes & Nutritional Sciences, King's College London, London SE1 1UL, UK
| | - Shuang Song
- Diabetes Research Group, Division of Diabetes & Nutritional Sciences, King's College London, London SE1 1UL, UK
| | - Peter M Jones
- Diabetes Research Group, Division of Diabetes & Nutritional Sciences, King's College London, London SE1 1UL, UK
| | - Shanta J Persaud
- Diabetes Research Group, Division of Diabetes & Nutritional Sciences, King's College London, London SE1 1UL, UK.
| |
Collapse
|
17
|
Bondarenko AI. Endothelial atypical cannabinoid receptor: do we have enough evidence? Br J Pharmacol 2014; 171:5573-88. [PMID: 25073723 PMCID: PMC4290703 DOI: 10.1111/bph.12866] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2014] [Revised: 07/14/2014] [Accepted: 07/24/2014] [Indexed: 12/16/2022] Open
Abstract
Cannabinoids and their synthetic analogues affect a broad range of physiological functions, including cardiovascular variables. Although direct evidence is still missing, the relaxation of a vast range of vascular beds induced by cannabinoids is believed to involve a still unidentified non-CB1 , non-CB2 Gi/o protein-coupled receptor located on endothelial cells, the so called endothelial cannabinoid receptor (eCB receptor). Evidence for the presence of an eCB receptor comes mainly from vascular relaxation studies, which commonly employ pertussis toxin as an indicator for GPCR-mediated signalling. In addition, a pharmacological approach is widely used to attribute the relaxation to eCB receptors. Recent findings have indicated a number of GPCR-independent targets for both agonists and antagonists of the presumed eCB receptor, warranting further investigations and cautious interpretation of the vascular relaxation studies. This review will provide a brief historical overview on the proposed novel eCB receptor, drawing attention to the discrepancies between the studies on the pharmacological profile of the eCB receptor and highlighting the Gi/o protein-independent actions of the eCB receptor inhibitors widely used as selective compounds. As the eCB receptor represents an attractive pharmacological target for a number of cardiovascular abnormalities, defining its molecular identity and the extent of its regulation of vascular function will have important implications for drug discovery. This review highlights the need to re-evaluate this subject in a thoughtful and rigorous fashion. More studies are needed to differentiate Gi/o protein-dependent endothelial cannabinoid signalling from that involving the classical CB1 and CB2 receptors as well as its relevance for pathophysiological conditions.
Collapse
Affiliation(s)
- Alexander I Bondarenko
- Circulatory Physiology Department, O.O.Bogomoletz Institute of PhysiologyKiev, Ukraine
- Institute of Molecular Biology and Biochemistry, Medical University of GrazGraz, Austria
| |
Collapse
|
18
|
Bondarenko AI, Drachuk K, Panasiuk O, Sagach V, Deak AT, Malli R, Graier WF. N-Arachidonoyl glycine suppresses Na⁺/Ca²⁺ exchanger-mediated Ca²⁺ entry into endothelial cells and activates BK(Ca) channels independently of GPCRs. Br J Pharmacol 2014; 169:933-48. [PMID: 23517055 PMCID: PMC3687672 DOI: 10.1111/bph.12180] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2012] [Revised: 01/14/2013] [Accepted: 02/20/2013] [Indexed: 02/03/2023] Open
Abstract
Background and Purpose N-arachidonoyl glycine (NAGly) is a lipoamino acid with vasorelaxant properties. We aimed to explore the mechanisms of NAGly's action on unstimulated and agonist-stimulated endothelial cells. Experimental Approach The effects of NAGly on endothelial electrical signalling were studied in combination with vascular reactivity. Key Results In EA.hy926 cells, the sustained hyperpolarization to histamine was inhibited by the non-selective Na+/Ca2+ exchanger (NCX) inhibitor bepridil and by an inhibitor of reversed mode NCX, KB-R7943. In cells dialysed with Cs+-based Na+-containing solution, the outwardly rectifying current with typical characteristics of NCX was augmented following histamine exposure, further increased upon external Na+ withdrawal and inhibited by bepridil. NAGly (0.3–30 μM) suppressed NCX currents in a URB597- and guanosine 5′-O-(2-thiodiphosphate) (GDPβS)-insensitive manner, [Ca2+]i elevation evoked by Na+ removal and the hyperpolarization to histamine. In rat aorta, NAGly opposed the endothelial hyperpolarization and relaxation response to ACh. In unstimulated EA.hy926 cells, NAGly potentiated the whole-cell current attributable to large-conductance Ca2+-activated K+ (BKCa) channels in a GDPβS-insensitive, paxilline-sensitive manner and produced a sustained hyperpolarization. In cell-free inside-out patches, NAGly stimulated single BKCa channel activity. Conclusion and Implications Our data showed that NCX is a Ca2+ entry pathway in endothelial cells and that NAGly is a potent G-protein-independent modulator of endothelial electrical signalling and has a dual effect on endothelial electrical responses. In agonist pre-stimulated cells, NAGly opposes hyperpolarization and relaxation via inhibition of NCX-mediated Ca2+ entry, while in unstimulated cells, it promotes hyperpolarization via receptor-independent activation of BKCa channels.
Collapse
Affiliation(s)
- Alexander I Bondarenko
- Institute of Molecular Biology and Biochemistry, Center of Molecular Medicine, Medical University of Graz, Graz, Austria.
| | | | | | | | | | | | | |
Collapse
|
19
|
Dopico AM, Bukiya AN. Lipid regulation of BK channel function. Front Physiol 2014; 5:312. [PMID: 25202277 PMCID: PMC4141547 DOI: 10.3389/fphys.2014.00312] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Accepted: 07/31/2014] [Indexed: 01/11/2023] Open
Abstract
This mini-review focuses on lipid modulation of BK (MaxiK, BKCa) current by a direct interaction between lipid and the BK subunits and/or their immediate lipid environment. Direct lipid-BK protein interactions have been proposed for fatty and epoxyeicosatrienoic acids, phosphoinositides and cholesterol, evidence for such action being less clear for other lipids. BK α (slo1) subunits are sufficient to support current perturbation by fatty and epoxyeicosatrienoic acids, glycerophospholipids and cholesterol, while distinct BK β subunits seem necessary for current modulation by most steroids. Subunit domains or amino acids that participate in lipid action have been identified in a few cases: hslo1 Y318, cerebral artery smooth muscle (cbv1) R334,K335,K336, cbv1 seven cytosolic CRAC domains, slo1 STREX and β1 T169,L172,L173 for docosahexaenoic acid, PIP2, cholesterol, sulfatides, and cholane steroids, respectively. Whether these protein motifs directly bind lipids or rather transmit the energy of lipid binding to other areas and trigger protein conformation change remains unresolved. The impact of direct lipid-BK interaction on physiology is briefly discussed.
Collapse
Affiliation(s)
- Alex M Dopico
- Department of Pharmacology, The University of Tennessee Health Science Center Memphis, TN, USA
| | - Anna N Bukiya
- Department of Pharmacology, The University of Tennessee Health Science Center Memphis, TN, USA
| |
Collapse
|
20
|
Marichal-Cancino BA, Manrique-Maldonado G, Altamirano-Espinoza AH, Ruiz-Salinas I, González-Hernández A, MaassenVanDenBrink A, Villalón CM. Analysis of anandamide- and lysophosphatidylinositol-induced inhibition of the vasopressor responses produced by sympathetic stimulation or noradrenaline in pithed rats. Eur J Pharmacol 2013; 721:168-77. [DOI: 10.1016/j.ejphar.2013.09.039] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2013] [Revised: 08/09/2013] [Accepted: 09/08/2013] [Indexed: 12/28/2022]
|
21
|
Yu J, Deliu E, Zhang XQ, Hoffman NE, Carter RL, Grisanti LA, Brailoiu GC, Madesh M, Cheung JY, Force T, Abood ME, Koch WJ, Tilley DG, Brailoiu E. Differential activation of cultured neonatal cardiomyocytes by plasmalemmal versus intracellular G protein-coupled receptor 55. J Biol Chem 2013; 288:22481-92. [PMID: 23814062 DOI: 10.1074/jbc.m113.456178] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The L-α-lysophosphatidylinositol (LPI)-sensitive receptor GPR55 is coupled to Ca(2+) signaling. Low levels of GPR55 expression in the heart have been reported. Similar to other G protein-coupled receptors involved in cardiac function, GPR55 may be expressed both at the sarcolemma and intracellularly. Thus, to explore the role of GPR55 in cardiomyocytes, we used calcium and voltage imaging and extracellular administration or intracellular microinjection of GPR55 ligands. We provide the first evidence that, in cultured neonatal ventricular myocytes, LPI triggers distinct signaling pathways via GPR55, depending on receptor localization. GPR55 activation at the sarcolemma elicits, on one hand, Ca(2+) entry via L-type Ca(2+) channels and, on the other, inositol 1,4,5-trisphosphate-dependent Ca(2+) release. The latter signal is further amplified by Ca(2+)-induced Ca(2+) release via ryanodine receptors. Conversely, activation of GPR55 at the membrane of intracellular organelles promotes Ca(2+) release from acidic-like Ca(2+) stores via the endolysosomal NAADP-sensitive two-pore channels. This response is similarly enhanced by Ca(2+)-induced Ca(2+) release via ryanodine receptors. Extracellularly applied LPI produces Ca(2+)-independent membrane depolarization, whereas the Ca(2+) signal induced by intracellular microinjection of LPI converges to hyperpolarization of the sarcolemma. Collectively, our findings point to GPR55 as a novel G protein-coupled receptor regulating cardiac function at two cellular sites. This work may serve as a platform for future studies exploring the potential of GPR55 as a therapeutic target in cardiac disorders.
Collapse
Affiliation(s)
- Justine Yu
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania 19140, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Deak AT, Groschner LN, Alam MR, Seles E, Bondarenko AI, Graier WF, Malli R. The endocannabinoid N-arachidonoyl glycine (NAGly) inhibits store-operated Ca2+ entry by preventing STIM1-Orai1 interaction. J Cell Sci 2012; 126:879-88. [PMID: 23239024 DOI: 10.1242/jcs.118075] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The endocannabiniod anandamide (AEA) and its derivate N-arachidonoyl glycine (NAGly) have a broad spectrum of physiological effects, which are induced by both binding to receptors and receptor-independent modulations of ion channels and transporters. The impact of AEA and NAGly on store-operated Ca(2+) entry (SOCE), a ubiquitous Ca(2+) entry pathway regulating many cellular functions, is unknown. Here we show that NAGly, but not AEA reversibly hinders SOCE in a time- and concentration-dependent manner. The inhibitory effect of NAGly on SOCE was found in the human endothelial cell line EA.hy926, the rat pancreatic β-cell line INS-1 832/13, and the rat basophilic leukemia cell line RBL-2H3. NAGly diminished SOCE independently from the mode of Ca(2+) depletion of the endoplasmic reticulum, whereas it had no effect on Ca(2+) entry through L-type voltage-gated Ca(2+) channels. Enhanced Ca(2+) entry was effectively hampered by NAGly in cells overexpressing the key molecular constituents of SOCE, stromal interacting molecule 1 (STIM1) and the pore-forming subunit of SOCE channels, Orai1. Fluorescence microscopy revealed that NAGly did not affect STIM1 oligomerization, STIM1 clustering, or the colocalization of STIM1 with Orai1, which were induced by Ca(2+) depletion of the endoplasmic reticulum. In contrast, independently from its slow depolarizing effect on mitochondria, NAGly instantly and strongly diminished the interaction of STIM1 with Orai1, indicating that NAGly inhibits SOCE primarily by uncoupling STIM1 from Orai1. In summary, our findings revealed the STIM1-Orai1-mediated SOCE machinery as a molecular target of NAGly, which might have many implications in cell physiology.
Collapse
Affiliation(s)
- Andras T Deak
- Institute of Molecular Biology and Biochemistry, Center of Molecular Medicine, Medical University Graz, Graz, Austria
| | | | | | | | | | | | | |
Collapse
|
23
|
Grzelczyk A, Gendaszewska-Darmach E. Novel bioactive glycerol-based lysophospholipids: new data -- new insight into their function. Biochimie 2012; 95:667-79. [PMID: 23089136 DOI: 10.1016/j.biochi.2012.10.009] [Citation(s) in RCA: 125] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2012] [Accepted: 10/11/2012] [Indexed: 11/28/2022]
Abstract
Based on the results of research conducted over last two decades, lysophospholipids (LPLs) were observed to be not only structural components of cellular membranes but also biologically active molecules influencing a broad variety of processes such as carcinogenesis, neurogenesis, immunity, vascular development or regulation of metabolic diseases. With a growing interest in the involvement of extracellular lysophospholipids in both normal physiology and pathology, it has become evident that those small molecules may have therapeutic potential. While lysophosphatidic acid (LPA) and sphingosine-1-phosphate (S1P) have been studied in detail, other LPLs such as lysophosphatidylglycerol (LPG), lysophosphatidylserine (LPS), lysophosphatidylinositol (LPI), lysophosphatidylethanolamine (LPE) or even lysophosphatidylcholine (LPC) have not been elucidated to such a high degree. Although information concerning the latter LPLs is sparse as compared to LPA and S1P, within the last couple of years much progress has been made. Recently published data suggest that these compounds may regulate fundamental cellular activities by modulating multiple molecular targets, e.g. by binding to specific receptors and/or altering the structure and fluidity of lipid rafts. Therefore, the present review is devoted to novel bioactive glycerol-based lysophospholipids and recent findings concerning their functions and possible signaling pathways regulating physiological and pathological processes.
Collapse
Affiliation(s)
- Anna Grzelczyk
- Institute of Technical Biochemistry, Faculty of Biotechnology and Food Sciences, Lodz University of Technology, Stefanowskiego 4/10, 90-924 Lodz, Poland
| | | |
Collapse
|
24
|
Zhao P, Abood ME. GPR55 and GPR35 and their relationship to cannabinoid and lysophospholipid receptors. Life Sci 2012; 92:453-7. [PMID: 22820167 DOI: 10.1016/j.lfs.2012.06.039] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2012] [Revised: 06/25/2012] [Accepted: 06/27/2012] [Indexed: 01/23/2023]
Abstract
This review presents a summary of what is known about the G-protein coupled receptors GPR35 and GPR55 and their potential characterization as lysophospholipid or cannabinoid receptors, respectively. Both GPR35 and GPR55 have been implicated as important targets in pain and cancer, and additional diseases as well. While kynurenic acid was suggested to be an endogenous ligand for GPR35, so was 2-arachidonoyl lysophosphatidic acid (LPA). Similarly, GPR55 has been suggested to be a cannabinoid receptor, but is quite clearly also a receptor for lysophosphatidylinositol. Interestingly, 2-arachidonyl glycerol (2-AG), an endogenous ligand for cannabinoid receptors, can be metabolized to 2-arachidonoyl LPA through the action of a monoacylglycerol kinase; the reverse reaction has also been demonstrated. Thus, it appears that mutual interconversion is possible between 2-arachidonoyl LPA and 2-AG within a cell, though the direction of the reaction may be site-dependent. The GPR55 natural ligand, 2-arachidonoyl LPI, can be degraded either to 2-AG by phospholipase C or to 2-arachidonoyl LPA by phospholipase D. Thus, GPR35, GPR55 and CB receptors are linked together through their natural ligand conversions. Additional agonists and antagonists have been identified for both GPR35 and GPR55, which will facilitate the future study of these receptors with respect to their physiological function. Potential therapeutic targets include pain, cancer, metabolic diseases and drug addiction.
Collapse
MESH Headings
- Animals
- Cannabinoids/pharmacology
- Cannabinoids/therapeutic use
- Gene Expression
- Humans
- Ligands
- Receptors, Cannabinoid/drug effects
- Receptors, Cannabinoid/genetics
- Receptors, Cannabinoid/physiology
- Receptors, G-Protein-Coupled/biosynthesis
- Receptors, G-Protein-Coupled/drug effects
- Receptors, G-Protein-Coupled/genetics
- Receptors, G-Protein-Coupled/physiology
- Receptors, Lysophospholipid/drug effects
- Receptors, Lysophospholipid/genetics
- Receptors, Lysophospholipid/physiology
Collapse
Affiliation(s)
- Pingwei Zhao
- Center for Substance Abuse Research and Department of Anatomy and Cell Biology, Temple University School of Medicine, Philadelphia, PA, USA
| | | |
Collapse
|
25
|
Lysophosphatidylinositol signalling: New wine from an old bottle. Biochim Biophys Acta Mol Cell Biol Lipids 2012; 1821:694-705. [DOI: 10.1016/j.bbalip.2012.01.009] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2011] [Revised: 12/02/2011] [Accepted: 01/03/2012] [Indexed: 01/29/2023]
|
26
|
Sharir H, Console-Bram L, Mundy C, Popoff SN, Kapur A, Abood ME. The endocannabinoids anandamide and virodhamine modulate the activity of the candidate cannabinoid receptor GPR55. J Neuroimmune Pharmacol 2012; 7:856-65. [PMID: 22454039 DOI: 10.1007/s11481-012-9351-6] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2011] [Accepted: 02/21/2012] [Indexed: 01/28/2023]
Abstract
The role of cannabinoid receptors in inflammation has been the topic of many research endeavors. Despite this effort, to date the involvement of the endocannabinoid system (ECS) in inflammation remains obscure. The ambiguity of cannabinoid involvement may be explained by the existence of cannabinoid receptors, other than CB(1) and CB(2), or a consequence of interaction of endocannabinoids with other signaling systems. GPR55 has been proposed to be a cannabinoid receptor; however the interaction of the endocannabinoid system with GPR55 remains elusive. Consequently this study set about to examine the effects of the endocannabinoids, anandamide (AEA) and virodhamine, on GPR55 mediated signaling. Specifically, we assessed changes in β-arrestin2 (βarr2) distribution and GPR55 receptor internalization following activation by lysophosphatidylinositol (LPI), the synthetic cannabinoid ligand SR141716A, and new selective synthetic GPR55 agonists. Data obtained from the experiments presented herein demonstrate that AEA and virodhamine modulate agonist-mediated recruitment of βarr2. AEA and virodhamine act as partial agonists; enhancing the agonist effect at low concentrations and inhibiting it at high concentrations. Furthermore, both virodhamine and AEA significantly attenuated agonist-induced internalization of GPR55. These effects are attributed to the expression of GPR55, and not CB(1) and CB(2) receptors, as we have established negligible expression of CB(1) and CB(2) in these GPR55-transfected U2OS cells. The identification of select endocannabinoids as GPR55 modulators will aide in elucidating the function of GPR55 in the ECS.
Collapse
Affiliation(s)
- Haleli Sharir
- Department of Anatomy and Cell Biology, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | | | | | | | | | | |
Collapse
|
27
|
Moreno-Navarrete JM, Catalán V, Whyte L, Díaz-Arteaga A, Vázquez-Martínez R, Rotellar F, Guzmán R, Gómez-Ambrosi J, Pulido MR, Russell WR, Imbernón M, Ross RA, Malagón MM, Dieguez C, Fernández-Real JM, Frühbeck G, Nogueiras R. The L-α-lysophosphatidylinositol/GPR55 system and its potential role in human obesity. Diabetes 2012; 61:281-91. [PMID: 22179809 PMCID: PMC3266411 DOI: 10.2337/db11-0649] [Citation(s) in RCA: 123] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
GPR55 is a putative cannabinoid receptor, and l-α-lysophosphatidylinositol (LPI) is its only known endogenous ligand. We investigated 1) whether GPR55 is expressed in fat and liver; 2) the correlation of both GPR55 and LPI with several metabolic parameters; and 3) the actions of LPI on human adipocytes. We analyzed CB1, CB2, and GPR55 gene expression and circulating LPI levels in two independent cohorts of obese and lean subjects, with both normal or impaired glucose tolerance and type 2 diabetes. Ex vivo experiments were used to measure intracellular calcium and lipid accumulation. GPR55 levels were augmented in the adipose tissue of obese subjects and further so in obese patients with type 2 diabetes when compared with nonobese subjects. Visceral adipose tissue GPR55 correlated positively with weight, BMI, and percent fat mass, particularly in women. Hepatic GPR55 gene expression was similar in obese and type 2 diabetic subjects. Circulating LPI levels were increased in obese patients and correlated with fat percentage and BMI in women. LPI increased the expression of lipogenic genes in visceral adipose tissue explants and intracellular calcium in differentiated visceral adipocytes. These findings indicate that the LPI/GPR55 system is positively associated with obesity in humans.
Collapse
Affiliation(s)
- José María Moreno-Navarrete
- Department of Diabetes, Endocrinology, and Nutrition, Institut d'Investigació Biomédica de Girona Catalonia, Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición, Girona, Spain
| | - Victoria Catalán
- Metabolic Research Laboratory, Clínica Universidad de Navarra, University of Navarra, Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición, Pamplona, Spain
| | - Lauren Whyte
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, Scotland, U.K
| | - Adenis Díaz-Arteaga
- Department of Physiology, School of Medicine, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición, Santiago de Compostela, Spain
| | - Rafael Vázquez-Martínez
- Department of Cell Biology, Physiology, and Immunology, Instituto Maimónides de Investigaciones Biomédicas de Córdoba, Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición, Córdoba, Spain
| | - Fernando Rotellar
- Metabolic Research Laboratory, Clínica Universidad de Navarra, University of Navarra, Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición, Pamplona, Spain
| | - Rocío Guzmán
- Department of Cell Biology, Physiology, and Immunology, Instituto Maimónides de Investigaciones Biomédicas de Córdoba, Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición, Córdoba, Spain
| | - Javier Gómez-Ambrosi
- Metabolic Research Laboratory, Clínica Universidad de Navarra, University of Navarra, Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición, Pamplona, Spain
| | - Marina R. Pulido
- Department of Cell Biology, Physiology, and Immunology, Instituto Maimónides de Investigaciones Biomédicas de Córdoba, Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición, Córdoba, Spain
| | - Wendy R. Russell
- Rowett Institute of Nutrition and Health, University of Aberdeen, Aberdeen, Scotland, U.K
| | - Mónica Imbernón
- Department of Physiology, School of Medicine, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición, Santiago de Compostela, Spain
| | - Ruth A. Ross
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, Scotland, U.K
| | - María M. Malagón
- Department of Cell Biology, Physiology, and Immunology, Instituto Maimónides de Investigaciones Biomédicas de Córdoba, Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición, Córdoba, Spain
| | - Carlos Dieguez
- Department of Physiology, School of Medicine, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición, Santiago de Compostela, Spain
| | - José Manuel Fernández-Real
- Department of Diabetes, Endocrinology, and Nutrition, Institut d'Investigació Biomédica de Girona Catalonia, Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición, Girona, Spain
| | - Gema Frühbeck
- Metabolic Research Laboratory, Clínica Universidad de Navarra, University of Navarra, Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición, Pamplona, Spain
| | - Ruben Nogueiras
- Department of Physiology, School of Medicine, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición, Santiago de Compostela, Spain
- Corresponding author: Ruben Nogueiras,
| |
Collapse
|
28
|
Rojo ML, Rodriguez-Gaztelumendi A, Fowler CJ. Lysophosphatidylinositol stimulates [³⁵S]GTPγS binding in the rat prefrontal cortex and hippocampus. Neurochem Res 2012; 37:1037-42. [PMID: 22270908 DOI: 10.1007/s11064-012-0704-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2011] [Accepted: 01/06/2012] [Indexed: 12/19/2022]
Abstract
Lysophosphatidylinositol (LPI) is a biologically active lipid that produces a number of responses in cultured cells, and has been suggested to have neuroprotective properties in vivo. Some of the actions of LPI are mediated by G-protein coupled receptors, but it is not known whether G-protein coupled receptor-mediated responses can be seen in intact brain tissue. In consequence, in the present study, we investigated autoradiographically whether LPI increased the [(35)S]GTPγS binding level in brain tissue slices. In standard assay conditions, where as a positive control a robust response to cannabinoid receptor activation by the agonist ligand CP55,940 was seen, there was no increase in the autoradiographic density over basal produced by LPI. However, when the conditions were modified (incubation at 4°C rather than at 25°C, incubation time increased to 3 h, GDP concentration reduced from 2 to 0.1 mM), a significant increase in [(35)S]GTPγS autoradiographic density in response to 10 μM LPI was seen in the prefrontal cortex, hippocampus, and cortex at the level of the hippocampus, although the degree of increase was small and very variable. No significant increases were seen in the hypothalamus or cerebellum. It is concluded that LPI, in the right conditions, can activate a sufficient number of G-proteins in the rat prefrontal cortex and hippocampus to produce a response in the [(35)S]GTPγS autoradiographic assay of G-protein coupled receptor function.
Collapse
Affiliation(s)
- Maria Luisa Rojo
- Department of Pharmacology and Clinical Neuroscience, Umeå University, 901 87 Umeå, Sweden
| | | | | |
Collapse
|
29
|
Henstridge CM, Balenga NAB, Kargl J, Andradas C, Brown AJ, Irving A, Sanchez C, Waldhoer M. Minireview: recent developments in the physiology and pathology of the lysophosphatidylinositol-sensitive receptor GPR55. Mol Endocrinol 2011; 25:1835-48. [PMID: 21964594 PMCID: PMC5417173 DOI: 10.1210/me.2011-1197] [Citation(s) in RCA: 123] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2011] [Accepted: 09/01/2011] [Indexed: 11/19/2022] Open
Abstract
Emerging data suggest that off-target cannabinoid effects may be mediated via novel seven-transmembrane spanning/G protein-coupled receptors. Due to its cannabinoid sensitivity, the G protein-coupled receptor 55 (GPR55) was recently proposed as a candidate; however, GPR55 is phylogenetically distinct from the traditional cannabinoid receptors, and the conflicting pharmacology, signaling, and functional data have prevented its classification as a novel cannabinoid receptor. Indeed, the most consistent and potent agonist to date is the noncannabinoid lysophospholipid, lysophosphatidylinositol. Here we present new human GPR55 mRNA expression data, providing supportive evidence of GPR55 expression in a vast array of tissues and cell types. Moreover, we summarize major recent developments in GPR55 research and aim to update the reader in the rapidly expanding fields of GPR55 pharmacology, physiology, and pathology.
Collapse
|
30
|
|
31
|
Waldeck-Weiermair M, Jean-Quartier C, Rost R, Khan MJ, Vishnu N, Bondarenko AI, Imamura H, Malli R, Graier WF. Leucine zipper EF hand-containing transmembrane protein 1 (Letm1) and uncoupling proteins 2 and 3 (UCP2/3) contribute to two distinct mitochondrial Ca2+ uptake pathways. J Biol Chem 2011; 286:28444-55. [PMID: 21613221 PMCID: PMC3151087 DOI: 10.1074/jbc.m111.244517] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Cytosolic Ca2+ signals are transferred into mitochondria over a huge concentration range. In our recent work we described uncoupling proteins 2 and 3 (UCP2/3) to be fundamental for mitochondrial uptake of high Ca2+ domains in mitochondria-ER junctions. On the other hand, the leucine zipper EF hand-containing transmembrane protein 1 (Letm1) was identified as a mitochondrial Ca2+/H+ antiporter that achieved mitochondrial Ca2+ sequestration at small Ca2+ increases. Thus, the contributions of Letm1 and UCP2/3 to mitochondrial Ca2+ uptake were compared in endothelial cells. Knock-down of Letm1 did not affect the UCP2/3-dependent mitochondrial uptake of intracellularly released Ca2+ but strongly diminished the transfer of entering Ca2+ into mitochondria, subsequently, resulting in a reduction of store-operated Ca2+ entry (SOCE). Knock-down of Letm1 and UCP2/3 did neither impact on cellular ATP levels nor the membrane potential. The enhanced mitochondrial Ca2+ signals in cells overexpressing UCP2/3 rescued SOCE upon Letm1 knock-down. In digitonin-permeabilized cells, Letm1 exclusively contributed to mitochondrial Ca2+ uptake at low Ca2+ conditions. Neither the Letm1- nor the UCP2/3-dependent mitochondrial Ca2+ uptake was affected by a knock-down of mRNA levels of mitochondrial calcium uptake 1 (MICU1), a protein that triggers mitochondrial Ca2+ uptake in HeLa cells. Our data indicate that Letm1 and UCP2/3 independently contribute to two distinct, mitochondrial Ca2+ uptake pathways in intact endothelial cells.
Collapse
Affiliation(s)
- Markus Waldeck-Weiermair
- Institute of Molecular Biology and Biochemistry, Molecular and Cellular Physiology Research Unit, Center of Molecular Medicine, Medical University Graz, Harrachgasse 21/III, 8010 Graz, Austria
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
The GPR55 agonist lysophosphatidylinositol directly activates intermediate-conductance Ca2+ -activated K+ channels. Pflugers Arch 2011; 462:245-55. [PMID: 21603896 PMCID: PMC3132407 DOI: 10.1007/s00424-011-0977-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2011] [Revised: 04/27/2011] [Accepted: 05/10/2011] [Indexed: 12/16/2022]
Abstract
Lysophosphatidylinositol (LPI) was recently shown to act both as an extracellular mediator binding to G protein-coupled receptor 55 (GPR55) and as an intracellular messenger directly affecting a number of ion channels including large-conductance Ca(2+) and voltage-gated potassium (BK(Ca)) channels. Here, we explored the effect of LPI on intermediate-conductance Ca(2+)-activated K(+) (IK(Ca)) channels using excised inside-out patches from endothelial cells. The functional expression of IK(Ca) was confirmed by the charybdotoxin- and TRAM-34-sensitive hyperpolarization to histamine and ATP. Moreover, the presence of single IK(Ca) channels with a slope conductance of 39 pS in symmetric K(+) gradient was directly confirmed in inside-out patches. When cytosolically applied in the range of concentrations of 0.3-10 μM, which are well below the herein determined critical micelle concentration of approximately 30 μM, LPI potentiated the IK(Ca) single-channel activity in a concentration-dependent manner, while single-channel current amplitude was not affected. In the whole-cell configuration, LPI in the pipette was found to facilitate membrane hyperpolarization in response to low (0.5 μM) histamine concentrations in a TRAM-34-sensitive manner. These results demonstrate a so far not-described receptor-independent effect of LPI on the IK(Ca) single-channel activity of endothelial cells, thus, highlighting LPI as a potent intracellular messenger capable of modulating electrical responses in the vasculature.
Collapse
|