1
|
Echeverria-Villalobos M, Guevara Y, Mitchell J, Ryskamp D, Conner J, Bush M, Periel L, Uribe A, Weaver TE. Potential perioperative cardiovascular outcomes in cannabis/cannabinoid users. A call for caution. Front Cardiovasc Med 2024; 11:1343549. [PMID: 38978789 PMCID: PMC11228818 DOI: 10.3389/fcvm.2024.1343549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 05/15/2024] [Indexed: 07/10/2024] Open
Abstract
Background Cannabis is one of the most widely used psychoactive substances. Its components act through several pathways, producing a myriad of side effects, of which cardiovascular events are the most life-threatening. However, only a limited number of studies address cannabis's perioperative impact on patients during noncardiac surgery. Methods Studies were identified by searching the PubMed, Medline, EMBASE, and Google Scholar databases using relevant keyword combinations pertinent to the topic. Results Current evidence shows that cannabis use may cause several cardiovascular events, including abnormalities in cardiac rhythm, myocardial infarction, heart failure, and cerebrovascular events. Additionally, cannabis interacts with anticoagulants and antiplatelet agents, decreasing their efficacy. Finally, the interplay of cannabis with inhalational and intravenous anesthetic agents may lead to adverse perioperative cardiovascular outcomes. Conclusions The use of cannabis can trigger cardiovascular events that may depend on factors such as the duration of consumption, the route of administration of the drug, and the dose consumed, which places these patients at risk of drug-drug interactions with anesthetic agents. However, large prospective randomized clinical trials are needed to further elucidate gaps in the body of knowledge regarding which patient population has a greater risk of perioperative complications after cannabis consumption.
Collapse
Affiliation(s)
| | - Yosira Guevara
- Department of Anesthesiology, St Elizabeth’s Medical Center, Brighton, MA, United States
| | - Justin Mitchell
- Department of Anesthesiology & Perioperative Medicine, UCLA Medical Center, Los Angeles, CA, United States
| | - David Ryskamp
- Department of Anesthesiology, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Joshua Conner
- Department of Anesthesiology, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Margo Bush
- University of Toledo, College of Medicine and Life Sciences, Toledo, OH, United States
| | - Luis Periel
- Touro College of Osteopathic Medicine, New York, NW, United States
| | - Alberto Uribe
- Department of Anesthesiology, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Tristan E. Weaver
- Department of Anesthesiology, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| |
Collapse
|
2
|
Mohammadpour-Asl S, Roshan-Milani S, Fard AA, Golchin A. Hormetic effects of a cannabinoid system component, 2-arachidonoyl glycerol, on cell viability and expression profile of growth factors in cultured mouse Sertoli cells: Friend or foe of male fertility? Reprod Toxicol 2024; 125:108575. [PMID: 38462211 DOI: 10.1016/j.reprotox.2024.108575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 03/05/2024] [Accepted: 03/08/2024] [Indexed: 03/12/2024]
Abstract
The generally undesired effects of exocannabinoids on male reproduction include alterations in testicular cell proliferation and function, as well as apoptosis induction. However, this paradigm has been challenged by the ability of endocannabinoids to regulate reproductive function. The present study addresses these paradoxical facts by investigating the effects of the endocannabinoid 2-arachidonoyl glycerol (2-AG) on mouse Sertoli cells' survival and apoptosis, with a mechanistic insight into Sertoli cell-based growth factors' production. The Mus musculus Sertoli cell line (TM4) was exposed to different concentrations of 2-AG, and cell viability was evaluated using MTT assay. Growth factors' gene and protein expressions were analyzed through RT-PCR and western blotting. 2-AG concentration dependently increased TM4 viability, with a slight increase starting at 0.0001 µM, a peak of 190% of the control level at 1 µM, and a decrease at 3 µM. Moreover, 2-AG paradoxically altered mRNA expression of caspase-3 and growth factors. Caspase-3 mRNA expression was down-regulated, and growth factors mRNA and protein expression were up-regulated when using a low concentration of 2-AG (1 μM). Opposite effects were observed by a higher concentration of 2-AG (3 μM). These paradoxical effects of 2-AG can be explained through the concept of hormesis. The results indicate the pivotal role of 2-AG in mediating Sertoli cell viability and apoptosis, at least in part, through altering growth factors secretion. Furthermore, they suggest the involvement of endocannabinoids in Sertoli cell-based physiological and pathological conditions and reflect the ability of abnormally elevated 2-AG to mimic the actions of exocannabinoids in reproductive dysfunction.
Collapse
Affiliation(s)
- Shadi Mohammadpour-Asl
- Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran; Department of Physiology, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Shiva Roshan-Milani
- Neurophysiology Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran.
| | - Amin Abdollahzade Fard
- Nephrology and Kidney Transplant Research Center, Clinical Research Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Ali Golchin
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran; Department of Clinical Biochemistry and Applied Cell Sciences, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| |
Collapse
|
3
|
Marchetti B, Bilel S, Tirri M, Corli G, Roda E, Locatelli CA, Cavarretta E, De-Giorgio F, Marti M. Acute Cardiovascular and Cardiorespiratory Effects of JWH-018 in Awake and Freely Moving Mice: Mechanism of Action and Possible Antidotal Interventions? Int J Mol Sci 2023; 24:7515. [PMID: 37108687 PMCID: PMC10142259 DOI: 10.3390/ijms24087515] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 04/14/2023] [Accepted: 04/17/2023] [Indexed: 04/29/2023] Open
Abstract
JWH-018 is the most known compound among synthetic cannabinoids (SCs) used for their psychoactive effects. SCs-based products are responsible for several intoxications in humans. Cardiac toxicity is among the main side effects observed in emergency departments: SCs intake induces harmful effects such as hypertension, tachycardia, chest pain, arrhythmias, myocardial infarction, breathing impairment, and dyspnea. This study aims to investigate how cardio-respiratory and vascular JWH-018 (6 mg/kg) responses can be modulated by antidotes already in clinical use. The tested antidotes are amiodarone (5 mg/kg), atropine (5 mg/kg), nifedipine (1 mg/kg), and propranolol (2 mg/kg). The detection of heart rate, breath rate, arterial oxygen saturation (SpO2), and pulse distention are provided by a non-invasive apparatus (Mouse Ox Plus) in awake and freely moving CD-1 male mice. Tachyarrhythmia events are also evaluated. Results show that while all tested antidotes reduce tachycardia and tachyarrhythmic events and improve breathing functions, only atropine completely reverts the heart rate and pulse distension. These data may suggest that cardiorespiratory mechanisms of JWH-018-induced tachyarrhythmia involve sympathetic, cholinergic, and ion channel modulation. Current findings also provide valuable impetus to identify potential antidotal intervention to support physicians in the treatment of intoxicated patients in emergency clinical settings.
Collapse
Affiliation(s)
- Beatrice Marchetti
- Department of Translational Medicine, Section of Legal Medicine and LTTA Center, University of Ferrara, 44121 Ferrara, Italy; (B.M.); (S.B.); (M.T.); (G.C.)
| | - Sabrine Bilel
- Department of Translational Medicine, Section of Legal Medicine and LTTA Center, University of Ferrara, 44121 Ferrara, Italy; (B.M.); (S.B.); (M.T.); (G.C.)
| | - Micaela Tirri
- Department of Translational Medicine, Section of Legal Medicine and LTTA Center, University of Ferrara, 44121 Ferrara, Italy; (B.M.); (S.B.); (M.T.); (G.C.)
| | - Giorgia Corli
- Department of Translational Medicine, Section of Legal Medicine and LTTA Center, University of Ferrara, 44121 Ferrara, Italy; (B.M.); (S.B.); (M.T.); (G.C.)
| | - Elisa Roda
- Laboratory of Clinical & Experimental Toxicology, Pavia Poison Centre, National Toxicology Information Centre, Toxicology Unit, Istituti Clinici Scientifici Maugeri IRCCS Pavia, 27100 Pavia, Italy; (E.R.); (C.A.L.)
| | - Carlo Alessandro Locatelli
- Laboratory of Clinical & Experimental Toxicology, Pavia Poison Centre, National Toxicology Information Centre, Toxicology Unit, Istituti Clinici Scientifici Maugeri IRCCS Pavia, 27100 Pavia, Italy; (E.R.); (C.A.L.)
| | - Elena Cavarretta
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 00185 Roma, Italy;
- Mediterrranea Cardiocentro, 80122 Napoli, Italy
| | - Fabio De-Giorgio
- Section of Legal Medicine, Department of Health Care Surveillance and Bioetics, Università Cattolica del Sacro Cuore, 00168 Rome, Italy;
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Matteo Marti
- Department of Translational Medicine, Section of Legal Medicine and LTTA Center, University of Ferrara, 44121 Ferrara, Italy; (B.M.); (S.B.); (M.T.); (G.C.)
- Collaborative Center for the Italian National Early Warning System, Department of Anti-Drug Policies, 00186 Rome, Italy
| |
Collapse
|
4
|
Hiniesto-Iñigo I, Castro-Gonzalez LM, Corradi V, Skarsfeldt MA, Yazdi S, Lundholm S, Nikesjö J, Noskov SY, Bentzen BH, Tieleman DP, Liin SI. Endocannabinoids enhance hK V7.1/KCNE1 channel function and shorten the cardiac action potential and QT interval. EBioMedicine 2023; 89:104459. [PMID: 36796231 PMCID: PMC9958262 DOI: 10.1016/j.ebiom.2023.104459] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 01/11/2023] [Accepted: 01/18/2023] [Indexed: 02/15/2023] Open
Abstract
BACKGROUND Genotype-positive patients who suffer from the cardiac channelopathy Long QT Syndrome (LQTS) may display a spectrum of clinical phenotypes, with often unknown causes. Therefore, there is a need to identify factors influencing disease severity to move towards an individualized clinical management of LQTS. One possible factor influencing the disease phenotype is the endocannabinoid system, which has emerged as a modulator of cardiovascular function. In this study, we aim to elucidate whether endocannabinoids target the cardiac voltage-gated potassium channel KV7.1/KCNE1, which is the most frequently mutated ion channel in LQTS. METHODS We used two-electrode voltage clamp, molecular dynamics simulations and the E4031 drug-induced LQT2 model of ex-vivo guinea pig hearts. FINDINGS We found a set of endocannabinoids that facilitate channel activation, seen as a shifted voltage-dependence of channel opening and increased overall current amplitude and conductance. We propose that negatively charged endocannabinoids interact with known lipid binding sites at positively charged amino acids on the channel, providing structural insights into why only specific endocannabinoids modulate KV7.1/KCNE1. Using the endocannabinoid ARA-S as a prototype, we show that the effect is not dependent on the KCNE1 subunit or the phosphorylation state of the channel. In guinea pig hearts, ARA-S was found to reverse the E4031-prolonged action potential duration and QT interval. INTERPRETATION We consider the endocannabinoids as an interesting class of hKV7.1/KCNE1 channel modulators with putative protective effects in LQTS contexts. FUNDING ERC (No. 850622), Canadian Institutes of Health Research, Canada Research Chairs and Compute Canada, Swedish National Infrastructure for Computing.
Collapse
Affiliation(s)
- Irene Hiniesto-Iñigo
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Laura M Castro-Gonzalez
- Centre for Molecular Simulation and Department of Biological Sciences, University of Calgary, Calgary, AB, Canada
| | - Valentina Corradi
- Centre for Molecular Simulation and Department of Biological Sciences, University of Calgary, Calgary, AB, Canada
| | - Mark A Skarsfeldt
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Samira Yazdi
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Siri Lundholm
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Johan Nikesjö
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Sergei Yu Noskov
- Centre for Molecular Simulation and Department of Biological Sciences, University of Calgary, Calgary, AB, Canada
| | - Bo Hjorth Bentzen
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - D Peter Tieleman
- Centre for Molecular Simulation and Department of Biological Sciences, University of Calgary, Calgary, AB, Canada
| | - Sara I Liin
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden.
| |
Collapse
|
5
|
The Old and the New: Cardiovascular and Respiratory Alterations Induced by Acute JWH-018 Administration Compared to Δ 9-THC-A Preclinical Study in Mice. Int J Mol Sci 2023; 24:ijms24021631. [PMID: 36675144 PMCID: PMC9865969 DOI: 10.3390/ijms24021631] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 01/04/2023] [Accepted: 01/10/2023] [Indexed: 01/19/2023] Open
Abstract
Several new psychoactive substances (NPS) are responsible for intoxication involving the cardiovascular and respiratory systems. Among NPS, synthetic cannabinoids (SCs) provoked side effects in humans characterized by tachycardia, arrhythmias, hypertension, breathing difficulty, apnoea, myocardial infarction, and cardiac arrest. Therefore, the present study investigated the cardio-respiratory (MouseOx Plus; EMKA electrocardiogram (ECG) and plethysmography TUNNEL systems) and vascular (BP-2000 systems) effects induced by 1-naphthalenyl (1-pentyl-1H-indol-3-yl)-methanone (JWH-018; 0.3-3-6 mg/kg) and Δ9-tetrahydrocannabinol (Δ9-THC; 0.3-3-6 mg/kg), administered in awake CD-1 male mice. The results showed that higher doses of JWH-018 (3-6 mg/kg) induced deep and long-lasting bradycardia, alternated with bradyarrhythmia, spaced out by sudden episodes of tachyarrhythmias (6 mg/kg), and characterized by ECG electrical parameters changes, sustained bradypnea, and systolic and transient diastolic hypertension. Otherwise, Δ9-THC provoked delayed bradycardia (minor intensity tachyarrhythmias episodes) and bradypnea, also causing a transient and mild hypertensive effect at the tested dose range. These effects were prevented by both treatment with selective CB1 (AM 251, 6 mg/kg) and CB2 (AM 630, 6 mg/kg) receptor antagonists and with the mixture of the antagonists AM 251 and AM 630, even if in a different manner. Cardio-respiratory and vascular symptoms could be induced by peripheral and central CB1 and CB2 receptors stimulation, which could lead to both sympathetic and parasympathetic systems activation. These findings may represent a starting point for necessary future studies aimed at exploring the proper antidotal therapy to be used in SCs-intoxicated patient management.
Collapse
|
6
|
Vallés AS, Barrantes FJ. Interactions between the Nicotinic and Endocannabinoid Receptors at the Plasma Membrane. MEMBRANES 2022; 12:812. [PMID: 36005727 PMCID: PMC9414690 DOI: 10.3390/membranes12080812] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 08/08/2022] [Accepted: 08/18/2022] [Indexed: 06/15/2023]
Abstract
Compartmentalization, together with transbilayer and lateral asymmetries, provide the structural foundation for functional specializations at the cell surface, including the active role of the lipid microenvironment in the modulation of membrane-bound proteins. The chemical synapse, the site where neurotransmitter-coded signals are decoded by neurotransmitter receptors, adds another layer of complexity to the plasma membrane architectural intricacy, mainly due to the need to accommodate a sizeable number of molecules in a minute subcellular compartment with dimensions barely reaching the micrometer. In this review, we discuss how nature has developed suitable adjustments to accommodate different types of membrane-bound receptors and scaffolding proteins via membrane microdomains, and how this "effort-sharing" mechanism has evolved to optimize crosstalk, separation, or coupling, where/when appropriate. We focus on a fast ligand-gated neurotransmitter receptor, the nicotinic acetylcholine receptor, and a second-messenger G-protein coupled receptor, the cannabinoid receptor, as a paradigmatic example.
Collapse
Affiliation(s)
- Ana Sofía Vallés
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (UNS-CONICET), Bahía Blanca 8000, Argentina
| | - Francisco J. Barrantes
- Laboratory of Molecular Neurobiology, Institute of Biomedical Research (BIOMED), UCA-CONICET, Av. Alicia Moreau de Justo 1600, Buenos Aires C1107AFF, Argentina
| |
Collapse
|
7
|
Lin YF. Potassium channels as molecular targets of endocannabinoids. Channels (Austin) 2021; 15:408-423. [PMID: 34282702 PMCID: PMC8293965 DOI: 10.1080/19336950.2021.1910461] [Citation(s) in RCA: 2] [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: 01/15/2021] [Revised: 03/24/2021] [Accepted: 03/24/2021] [Indexed: 10/25/2022] Open
Abstract
Endocannabinoids are a group of endogenous mediators derived from membrane lipids, which are implicated in a wide variety of physiological functions such as blood pressure regulation, immunity, pain, memory, reward, perception, reproduction, and sleep. N-Arachidonoylethanolamine (anandamide; AEA) and 2-arachidonoylglycerol (2-AG) represent two major endocannabinoids in the human body and they exert many of their cellular and organ system effects by activating the Gi/o protein-coupled, cannabinoid type 1 (CB1) and type 2 (CB2) receptors. However, not all effects of cannabinoids are ascribable to their interaction with CB1 and CB2 receptors; indeed, macromolecules like other types of receptors, ion channels, transcription factors, enzymes, transporters, and cellular structure have been suggested to mediate the functional effects of cannabinoids. Among the proposed molecular targets of endocannabinoids, potassium channels constitute an intriguing group, because these channels not only are crucial in shaping action potentials and controlling the membrane potential and cell excitability, thereby regulating a wide array of physiological processes, but also serve as potential therapeutic targets for the treatment of cancer and metabolic, neurological and cardiovascular disorders. This review sought to survey evidence pertaining to the CB1 and CB2 receptor-independent actions of endocannabinoids on ion channels, with an emphasis on AEA and potassium channels. To better understand the functional roles as well as potential medicinal uses of cannabinoids in human health and disease, further mechanistic studies to delineate interactions between various types of cannabinoids and ion channels, including members in the potassium channel superfamily, are warranted.
Collapse
Affiliation(s)
- Yu-Fung Lin
- Department of Physiology and Membrane Biology, University of California Davis, Davis, CA, USA
- Department of Anesthesiology and Pain Medicine, University of California Davis, Davis, CA, USA
| |
Collapse
|
8
|
Topal L, Naveed M, Orvos P, Pászti B, Prorok J, Bajtel Á, Kiss T, Csupor-Löffler B, Csupor D, Baczkó I, Varró A, Virág L, Jost N. The electrophysiological effects of cannabidiol on action potentials and transmembrane potassium currents in rabbit and dog cardiac ventricular preparations. Arch Toxicol 2021; 95:2497-2505. [PMID: 34031697 PMCID: PMC8241752 DOI: 10.1007/s00204-021-03086-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 05/17/2021] [Indexed: 12/24/2022]
Abstract
Cannabis use is associated with known cardiovascular side effects such as cardiac arrhythmias or even sudden cardiac death. The mechanisms behind these adverse effects are unknown. The aim of the present work was to study the cellular cardiac electrophysiological effects of cannabidiol (CBD) on action potentials and several transmembrane potassium currents, such as the rapid (IKr) and slow (IKs) delayed rectifier, the transient outward (Ito) and inward rectifier (IK1) potassium currents in rabbit and dog cardiac preparations. CBD increased action potential duration (APD) significantly in both rabbit (from 211.7 ± 11.2. to 224.6 ± 11.4 ms, n = 8) and dog (from 215.2 ± 9.0 to 231.7 ± 4.7 ms, n = 6) ventricular papillary muscle at 5 µM concentration. CBD decreased IKr, IKs and Ito (only in dog) significantly with corresponding estimated EC50 values of 4.9, 3.1 and 5 µM, respectively, without changing IK1. Although the EC50 value of CBD was found to be higher than literary Cmax values after CBD smoking and oral intake, our results raise the possibility that potassium channel inhibition by lengthening cardiac repolarization might have a role in the possible proarrhythmic side effects of cannabinoids in situations where CBD metabolism and/or the repolarization reserve is impaired.
Collapse
Affiliation(s)
- Leila Topal
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Dóm tér 12, 6720, Szeged, Hungary
| | - Muhammad Naveed
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Dóm tér 12, 6720, Szeged, Hungary
| | - Péter Orvos
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Dóm tér 12, 6720, Szeged, Hungary
| | - Bence Pászti
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Dóm tér 12, 6720, Szeged, Hungary
| | - János Prorok
- ELKH-SZTE Research Group for Cardiovascular Pharmacology, Eötvös Loránd Research Network, Szeged, Hungary
| | - Ákos Bajtel
- Department of Pharmacognosy, Faculty of Pharmacy, University of Szeged, Szeged, Hungary
| | - Tivadar Kiss
- Department of Pharmacognosy, Faculty of Pharmacy, University of Szeged, Szeged, Hungary
| | | | - Dezső Csupor
- Department of Pharmacognosy, Faculty of Pharmacy, University of Szeged, Szeged, Hungary
- Institute for Translational Medicine, Medical School, University of Pécs, Pécs, Hungary
| | - István Baczkó
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Dóm tér 12, 6720, Szeged, Hungary
- Department of Pharmacology and Pharmacotherapy, Interdisciplinary Excellence Centre, University of Szeged, 6720, Szeged, Hungary
| | - András Varró
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Dóm tér 12, 6720, Szeged, Hungary
- ELKH-SZTE Research Group for Cardiovascular Pharmacology, Eötvös Loránd Research Network, Szeged, Hungary
- Department of Pharmacology and Pharmacotherapy, Interdisciplinary Excellence Centre, University of Szeged, 6720, Szeged, Hungary
| | - László Virág
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Dóm tér 12, 6720, Szeged, Hungary
- Department of Pharmacology and Pharmacotherapy, Interdisciplinary Excellence Centre, University of Szeged, 6720, Szeged, Hungary
| | - Norbert Jost
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Dóm tér 12, 6720, Szeged, Hungary.
- ELKH-SZTE Research Group for Cardiovascular Pharmacology, Eötvös Loránd Research Network, Szeged, Hungary.
| |
Collapse
|
9
|
Radaelli D, Manfredi A, Zanon M, Fattorini P, Scopetti M, Neri M, Frisoni P, D’Errico S. Synthetic Cannabinoids and Cathinones Cardiotoxicity: Facts and Perspectives. Curr Neuropharmacol 2021; 19:2038-2048. [PMID: 33845747 PMCID: PMC9185792 DOI: 10.2174/1570159x19666210412101929] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 03/06/2021] [Accepted: 03/18/2021] [Indexed: 11/23/2022] Open
Abstract
New psychoactive substances (NPS) constitute a group of psychotropic substances, designed to mimic the effects of traditional substances like cannabis, cocaine, MDMA, khat, which was not regulated by the 1961 United Nations Convention on Narcotics or the 1971 United Nations Convention on Psychotropic Substances. Illegal laboratories responsible for their production regularly developed new substances and placed them on the market to replace the ones that have been banned; for this reason, during the last decade this class of substances has represented a great challenge for the public health and forensic toxicologists. The spectrum of side effects caused by the intake of these drugs of abuse is very wide since they act on different systems with various mechanisms of action. To date most studies have focused on the neurotoxic effects, very few works focus on cardiotoxicity. Specifically, both synthetic cannabinoids and synthetic cathinones appear to be involved in different cardiac events, including myocardial infarction and sudden cardiac death due to fatal arrhythmias. Synthetic cannabinoids and cathinones cardiotoxicity are mainly mediated through activation of the CB1 receptor present on cardiomyocyte and involved with reactive oxygen species production, ATP depletion and cell death. Concerns with the adrenergic over-stimulation induced by this class of substances and increasing oxidative stress are mainly reported. In this systematic review we aim to summarize the data from all the works analyzing the possible mechanisms through which synthetic cannabinoids and synthetic cathinones damage the myocardial tissue.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Stefano D’Errico
- Address correspondence to this author at the Department of Medicine, Surgery and Health, University of Trieste, Italy; E-mail:
| |
Collapse
|
10
|
Orvos P, Pászti B, Topal L, Gazdag P, Prorok J, Polyák A, Kiss T, Tóth-Molnár E, Csupor-Löffler B, Bajtel Á, Varró A, Hohmann J, Virág L, Csupor D. The electrophysiological effect of cannabidiol on hERG current and in guinea-pig and rabbit cardiac preparations. Sci Rep 2020; 10:16079. [PMID: 32999428 PMCID: PMC7528081 DOI: 10.1038/s41598-020-73165-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 09/10/2020] [Indexed: 12/28/2022] Open
Abstract
Cannabis use is associated with cardiovascular adverse effects ranging from arrhythmias to sudden cardiac death. The exact mechanism of action behind these activities is unknown. The aim of our work was to study the effect of cannabidiol (CBD), tetrahydrocannabinol and 11-nor-9-carboxy-tetrahydrocannabinol on cellular cardiac electrophysiological properties including ECG parameters, action potentials, hERG and IKr ion channels in HEK cell line and in rabbit and guinea pig cardiac preparations. CBD increased action potential duration in rabbit and guinea pig right ventricular papillary muscle at lower concentrations (1 µM, 2.5 µM and 5 µM) but did not significantly change it at 10 µM. CBD at high concentration (10 µM) decreased inward late sodium and L-type calcium currents as well. CBD inhibited hERG potassium channels with an IC50 value of 2.07 µM at room temperature and delayed rectifier potassium current with 6.5 µM at 37 °C, respectively. The frequency corrected QT interval (QTc) was significantly lengthened in anaesthetized guinea pig without significantly changing other ECG parameters. Although the IC50 value of CBD was higher than literary Cmax values after CBD smoking and oral intake, our results raise the possibility that hERG and potassium channel inhibition might have a role in the possible proarrhythmic adverse effects of cannabinoids in situations where metabolism of CBD impaired and/or the repolarization reserve is weakened.
Collapse
Affiliation(s)
- Péter Orvos
- Department of Ophthalmology, Faculty of Medicine, Albert Szent-Györgyi Clinical Center, University of Szeged, Szeged, Hungary
| | - Bence Pászti
- Department of Pharmacology and Pharmacotherapy, University of Szeged, Szeged, Hungary
| | - Leila Topal
- Department of Pharmacology and Pharmacotherapy, University of Szeged, Szeged, Hungary
| | - Péter Gazdag
- Department of Pharmacology and Pharmacotherapy, University of Szeged, Szeged, Hungary
| | - János Prorok
- MTA-SZTE Research Group for Cardiovascular Pharmacology, Hungarian Academy of Sciences, Szeged, Hungary
| | - Alexandra Polyák
- Department of Pharmacology and Pharmacotherapy, University of Szeged, Szeged, Hungary
- MTA-SZTE Research Group for Cardiovascular Pharmacology, Hungarian Academy of Sciences, Szeged, Hungary
| | - Tivadar Kiss
- Department of Pharmacognosy, Faculty of Pharmacy, University of Szeged, Eötvös u. 6, Szeged, 672, Hungary
| | - Edit Tóth-Molnár
- Department of Ophthalmology, Faculty of Medicine, Albert Szent-Györgyi Clinical Center, University of Szeged, Szeged, Hungary
| | | | - Ákos Bajtel
- Department of Pharmacognosy, Faculty of Pharmacy, University of Szeged, Eötvös u. 6, Szeged, 672, Hungary
| | - András Varró
- Department of Pharmacology and Pharmacotherapy, University of Szeged, Szeged, Hungary
- MTA-SZTE Research Group for Cardiovascular Pharmacology, Hungarian Academy of Sciences, Szeged, Hungary
- Department of Pharmacology and Pharmacotherapy, Interdisciplinary Excellence Centre, University of Szeged, Szeged, 6720, Hungary
| | - Judit Hohmann
- Department of Pharmacognosy, Faculty of Pharmacy, University of Szeged, Eötvös u. 6, Szeged, 672, Hungary
| | - László Virág
- Department of Pharmacology and Pharmacotherapy, University of Szeged, Szeged, Hungary
- Department of Pharmacology and Pharmacotherapy, Interdisciplinary Excellence Centre, University of Szeged, Szeged, 6720, Hungary
| | - Dezső Csupor
- Department of Pharmacognosy, Faculty of Pharmacy, University of Szeged, Eötvös u. 6, Szeged, 672, Hungary.
| |
Collapse
|
11
|
Richards JR. Mechanisms for the Risk of Acute Coronary Syndrome and Arrhythmia Associated With Phytogenic and Synthetic Cannabinoid Use. J Cardiovasc Pharmacol Ther 2020; 25:508-522. [PMID: 32588641 DOI: 10.1177/1074248420935743] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Phytogenic cannabinoids from Cannabis sativa and synthetic cannabinoids are commonly used substances for their recreational and medicinal properties. There are increasing reports of cardiotoxicity in close temporal association with cannabinoid use in patients with structurally normal hearts and absence of coronary arterial disease. Associated adverse events include myocardial ischemia, conduction abnormalities, arrhythmias, and sudden death. This review details the effects of phytogenic and synthetic cannabinoids on diverse receptors based on evidence from in vitro, human, and animal studies to establish a molecular basis for these deleterious clinical effects. The synergism between endocannabinoid dysregulation, cannabinoid receptor, and noncannabinoid receptor binding, and impact on cellular ion flux and coronary microvascular circulation is delineated. Pharmacogenetic factors placing certain patients at higher risk for cardiotoxicity are also correlated with the diverse effects of cannabinoids.
Collapse
Affiliation(s)
- John R Richards
- Department of Emergency Medicine, 70083University of California Davis Medical Center, Sacramento, California, CA, USA
| |
Collapse
|
12
|
Ozturk HM, Yetkin E, Ozturk S. Synthetic Cannabinoids and Cardiac Arrhythmia Risk: Review of the Literature. Cardiovasc Toxicol 2020; 19:191-197. [PMID: 31030341 DOI: 10.1007/s12012-019-09522-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Synthetic cannabinoids (SCBs) are widely used recreational substances especially among adults. Although they have been considered as safe during the marketing process, our knowledge about their adverse effects has evolved since years. SCBs are associated with various cardiac events including acute myocardial infarction and sudden cardiac death. There is also growing evidence that SCBs are associated with cardiac arrhythmia development both in acute and chronic exposure. SCBs have been shown to be associated with both supraventricular and ventricular arrhythmias. However, the exact mechanism of the SCB related arrhythmia remains unknown. Understanding the exact association and possible mechanisms may help us to identify high risk patients at an early stage and to develop treatment modalities to prevent or reverse the arrhythmic effects of SCBs.
Collapse
Affiliation(s)
| | - Ertan Yetkin
- Department of Cardiology, Istinye University Liv Hospital, Istanbul, Turkey
| | - Selcuk Ozturk
- Cardiology Clinic, Ankara Education and Research Hospital, Ankara, Turkey.
| |
Collapse
|
13
|
Cheng N, Ren S, Yang JF, Liu XM, Li XT. Carvedilol blockage of delayed rectifier Kv2.1 channels and its molecular basis. Eur J Pharmacol 2019; 855:50-55. [PMID: 31063774 DOI: 10.1016/j.ejphar.2019.05.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 04/30/2019] [Accepted: 05/03/2019] [Indexed: 11/19/2022]
Abstract
Previous studies indicated that one of the action targets of carvedilol is the voltage-gated potassium (Kv) channel, which has a fundamental role in the control of electrical properties in excitable cells. It is not clear whether this compound exerts any actions specifically on delayed rectifier Kv2.1 channels. The present study is conducted on Kv2.1 currents heterologously expressed in HEK293 cells to characterize the block by carvedilol in detail, identifying molecular determinants and providing biophysical insights of the block. Macroscopic Kv2.1 currents obtained by whole-cell recording were substantially inhibited after addition of carvedilol with an IC50 value of 5.1 μM. This drug also led to a largely hyperpolarizing shift (30 mV) of the inactivation curve, which may contribute to the blocking action due to more inactivation of Kv2.1 currents occurred in depolarization potentials. Mutations at Y380 (a putative TEA binding site) and K356 (a K+ binding site) in the outer vestibule of Kv2.1 channels significantly eliminated the inhibitory effects of carvedilol and prevented the leftward shift of inactivation. Moreover, mutations at above positions modulated the effects of carvedilol on the deactivation but not activation kinetics of Kv2.1 channels. Collected data indicate that carvedilol can exert a blocking effect on the closed-state of Kv2.1 channels, and specific residues within the S5-P and P-S6 linkers in the outer vestibule may play crucial roles in carvedilol-induced blocking for Kv2.1 channels.
Collapse
Affiliation(s)
- Neng Cheng
- College of Life Science, South-Central University for Nationalities, Wuhan, 430074, China
| | - Sheng Ren
- College of Life Science, South-Central University for Nationalities, Wuhan, 430074, China
| | - Jin-Feng Yang
- College of Life Science, South-Central University for Nationalities, Wuhan, 430074, China
| | - Xiang-Ming Liu
- GongQing Institute of Science and Technology, Gongqing City, 332020, China
| | - Xian-Tao Li
- College of Life Science, South-Central University for Nationalities, Wuhan, 430074, China.
| |
Collapse
|
14
|
Yang JF, Cheng N, Ren S, Liu XM, Li XT. Characterization and molecular basis for the block of Kv1.3 channels induced by carvedilol in HEK293 cells. Eur J Pharmacol 2018; 834:206-212. [PMID: 30016664 DOI: 10.1016/j.ejphar.2018.07.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 07/04/2018] [Accepted: 07/13/2018] [Indexed: 10/28/2022]
Abstract
Carvedilol is a non-selective β-adrenoreceptor antagonist and exhibits a wide range of biological activities. The voltage-gated K+ (Kv) channel is one of the target ion channels of this compound. The rapidly activating Kv1.3 channel is expressed in several different tissues and plays an important role in the regulation of physiological functions, including cell proliferation and apoptosis. However, little is known about the possible action of carvedilol on Kv1.3 currents. Using the whole-cell configuration of the patch-clamp technique, we have revealed that exposure to carvedilol produced a concentration-dependent blocking of Kv1.3 channels heterologously expressed in HEK293 cells, with an IC50 value of 9.7 μM. This chemical decelerated the deactivation tail current of Kv1.3 currents, resulting in a tail crossover phenomenon. In addition, carvedilol generated a markedly hyperpolarizing shift (20 mV) of the inactivation curve, but failed to affect the activation curve. Mutagenesis experiments of Kv1.3 channels identified G427 and H451, two related sites of TEA block, as important residues for carvedilol-mediated blocking. The present results suggest that carvedilol acts directly on Kv1.3 currents by inducing closed- and open-channel block and helps to elucidate the mechanisms of action of this compound on Kv channels.
Collapse
Affiliation(s)
- Jin-Feng Yang
- College of Life Science, South-Central University for Nationalities, Wuhan 430074, China
| | - Neng Cheng
- College of Life Science, South-Central University for Nationalities, Wuhan 430074, China
| | - Sheng Ren
- College of Life Science, South-Central University for Nationalities, Wuhan 430074, China
| | - Xiang-Ming Liu
- GongQing Institute of Science and Technology, Gongqing City 332020, China
| | - Xian-Tao Li
- College of Life Science, South-Central University for Nationalities, Wuhan 430074, China.
| |
Collapse
|
15
|
Hansen HS, Vana V. Non-endocannabinoid N-acylethanolamines and 2-monoacylglycerols in the intestine. Br J Pharmacol 2018; 176:1443-1454. [PMID: 29473944 DOI: 10.1111/bph.14175] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 01/23/2018] [Accepted: 02/05/2018] [Indexed: 12/11/2022] Open
Abstract
This review focuses on recent findings of the physiological and pharmacological role of non-endocannabinoid N-acylethanolamines (NAEs) and 2-monoacylglycerols (2-MAGs) in the intestine and their involvement in the gut-brain signalling. Dietary fat suppresses food intake, and much research concerns the known gut peptides, for example, glucagon-like peptide-1 (GLP-1) and cholecystokinin (CCK). NAEs and 2-MAGs represent another class of local gut signals most probably involved in the regulation of food intake. We discuss the putative biosynthetic pathways and targets of NAEs in the intestine as well as their anorectic role and changes in intestinal levels depending on the dietary status. NAEs can activate the transcription factor PPARα, but studies to evaluate the role of endogenous NAEs are generally lacking. Finally, we review the role of diet-derived 2-MAGs in the secretion of anorectic gut peptides via activation of GPR119. Both PPARα and GPR119 have potential as pharmacological targets for the treatment of obesity and the former for treatment of intestinal inflammation. LINKED ARTICLES: This article is part of a themed section on 8th European Workshop on Cannabinoid Research. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.10/issuetoc.
Collapse
Affiliation(s)
- Harald S Hansen
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Vasiliki Vana
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
16
|
Matsui A, Alvarez VA. Undercover Power of Endocannabinoids: Postsynaptic Ion-Channel Modulator. Neuron 2017; 93:1243-1244. [PMID: 28334600 DOI: 10.1016/j.neuron.2017.03.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
In this issue of Neuron, Gantz and Bean (2017) show that the endocannabinoid 2-arachidonoyl glycerol (2-AG) can directly alter the properties of native ion-channel Kv4.3 and accelerate the pacemaker activity of rodent dopamine neurons. These findings are one of the first demonstrations of postsynaptic, cell-autonomous actions of endocannabinoids in the mammalian brain.
Collapse
Affiliation(s)
- Aya Matsui
- Laboratory on Neurobiology of Compulsive Behaviors, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Veronica A Alvarez
- Laboratory on Neurobiology of Compulsive Behaviors, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland 20892, USA.
| |
Collapse
|
17
|
Bondarenko AI, Panasiuk O, Okhai I, Montecucco F, Brandt KJ, Mach F. 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 PMCID: PMC6520242 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] [MESH Headings] [Grants] [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
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, 8010 Graz, Austria
| | - Olga Panasiuk
- Circulatory Physiology Department, Bogomoletz Institute of Physiology NAS of Ukraine, Bogomoletz Str, 4, Kiev 01024, Ukraine
| | - Iryna Okhai
- 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; IRCCS AOU San Martino - IST, Genoa, 10 Largo Benzi, 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
| | - Francois 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
|
18
|
Medeiros D, Silva-Gonçalves LDC, da Silva AMB, Dos Santos Cabrera MP, Arcisio-Miranda M. Membrane-mediated action of the endocannabinoid anandamide on membrane proteins: implications for understanding the receptor-independent mechanism. Sci Rep 2017; 7:41362. [PMID: 28128290 PMCID: PMC5269673 DOI: 10.1038/srep41362] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Accepted: 12/20/2016] [Indexed: 12/31/2022] Open
Abstract
Endocannabinoids are amphiphilic molecules that play crucial neurophysiological functions acting as lipid messengers. Antagonists and knockdown of the classical CB1 and CB2 cannabinoid receptors do not completely abolish many endocannabinoid activities, supporting the idea of a mechanism independent of receptors whose mode of action remains unclear. Here we combine gramicidin A (gA) single channel recordings and membrane capacitance measurements to investigate the lipid bilayer-modifying activity of endocannabinoids. Single channel recordings show that the incorporation of endocannabinoids into lipid bilayers reduces the free energy necessary for gramicidin channels to transit from the monomeric to the dimeric conformation. Membrane capacitance demonstrates that the endocannabinoid anandamide has limited effects on the overall structure of the lipid bilayers. Our results associated with the theory of membrane elastic deformation reveal that the action of endocannabinoids on membrane proteins can involve local adjustments of the lipid/protein hydrophobic interface. The current findings shed new light on the receptor-independent mode of action of endocannabinoids on membrane proteins, with important implications towards their neurobiological function.
Collapse
Affiliation(s)
- Djalma Medeiros
- Laboratório de Neurobiologia Estrutural e Funcional (LaNEF), Departamento de Biofísica, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brasil.,Curso de Filosofia, Faculdade de São Bento, São Paulo, SP, Brasil
| | - Laíz da Costa Silva-Gonçalves
- Laboratório de Neurobiologia Estrutural e Funcional (LaNEF), Departamento de Biofísica, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brasil
| | - Annielle Mendes Brito da Silva
- Laboratório de Neurobiologia Estrutural e Funcional (LaNEF), Departamento de Biofísica, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brasil
| | | | - Manoel Arcisio-Miranda
- Laboratório de Neurobiologia Estrutural e Funcional (LaNEF), Departamento de Biofísica, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brasil
| |
Collapse
|
19
|
Wu W, Sanguinetti MC. Molecular Basis of Cardiac Delayed Rectifier Potassium Channel Function and Pharmacology. Card Electrophysiol Clin 2016; 8:275-84. [PMID: 27261821 DOI: 10.1016/j.ccep.2016.01.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Human cardiomyocytes express 3 distinct types of delayed rectifier potassium channels. Human ether-a-go-go-related gene (hERG) channels conduct the rapidly activating current IKr; KCNQ1/KCNE1 channels conduct the slowly activating current IKs; and Kv1.5 channels conduct an ultrarapid activating current IKur. Here the authors provide a general overview of the mechanistic and structural basis of ion selectivity, gating, and pharmacology of the 3 types of cardiac delayed rectifier potassium ion channels. Most blockers bind to S6 residues that line the central cavity of the channel, whereas activators interact with the channel at 4 symmetric binding sites outside the cavity.
Collapse
Affiliation(s)
- Wei Wu
- Department of Medicine, Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, 95 South 2000 East, Salt Lake City, UT 84112, USA
| | - Michael C Sanguinetti
- Department of Medicine, Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, 95 South 2000 East, Salt Lake City, UT 84112, USA.
| |
Collapse
|
20
|
Abstract
The endocannabinoid system (ECS) comprises a complex of receptors, enzymes, and endogenous agonists that are widely distributed in the central nervous system of mammals and participates in a considerable number of neuromodulatory functions, including neurotransmission, immunological control, and cell signaling. In turn, the kynurenine pathway (KP) is the most relevant metabolic route for tryptophan degradation to form the metabolic precursor NAD(+). Recent studies demonstrate that the control exerted by the pharmacological manipulation of the ECS on the glutamatergic system in the brain may offer key information not only on the development of psychiatric disorders like psychosis and schizophrenia-like symptoms, but it also may constitute a solid basis for the development of therapeutic strategies to combat excitotoxic events occurring in neurological disorders like Huntington's disease (HD). Part of the evidence pointing to the last approach is based on experimental protocols demonstrating the efficacy of cannabinoids to prevent the deleterious actions of the endogenous neurotoxin and KP metabolite quinolinic acid (QUIN). These findings intuitively raise the question about what is the precise role of the ECS in tryptophan metabolism through KP and vice versa. In this chapter, we will review basic concepts on the physiology of both the ECS and the KP to finally describe those recent findings combining the components of these two systems and hypothesize the future course that the research in this emerging field will take in the next years.
Collapse
|
21
|
Expression and Function of the Endocannabinoid System in the Retina and the Visual Brain. Neural Plast 2015; 2016:9247057. [PMID: 26839718 PMCID: PMC4709729 DOI: 10.1155/2016/9247057] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Revised: 09/24/2015] [Accepted: 09/27/2015] [Indexed: 12/16/2022] Open
Abstract
Endocannabinoids are important retrograde modulators of synaptic transmission throughout the nervous system. Cannabinoid receptors are seven transmembrane G-protein coupled receptors favoring Gi/o protein. They are known to play an important role in various processes, including metabolic regulation, craving, pain, anxiety, and immune function. In the last decade, there has been a growing interest for endocannabinoids in the retina and their role in visual processing. The purpose of this review is to characterize the expression and physiological functions of the endocannabinoid system in the visual system, from the retina to the primary visual cortex, with a main interest regarding the retina, which is the best-described area in this system so far. It will show that the endocannabinoid system is widely present in the retina, mostly in the through pathway where it can modulate neurotransmitter release and ion channel activity, although some evidence also indicates possible mechanisms via amacrine, horizontal, and Müller cells. The presence of multiple endocannabinoid ligands, synthesizing and catabolizing enzymes, and receptors highlights various pharmacological targets for novel therapeutic application to retinal diseases.
Collapse
|
22
|
Bai J, Ding W, Kojima A, Seto T, Matsuura H. Putative binding sites for arachidonic acid on the human cardiac Kv 1.5 channel. Br J Pharmacol 2015; 172:5281-92. [PMID: 26292661 PMCID: PMC5341216 DOI: 10.1111/bph.13314] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Revised: 08/01/2015] [Accepted: 08/18/2015] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND AND PURPOSE In human heart, the Kv 1.5 channel contributes to repolarization of atrial action potentials. This study examined the electrophysiological and molecular mechanisms underlying arachidonic acid (AA)-induced inhibition of the human Kv 1.5 (hKv 1.5) channel. EXPERIMENTAL APPROACH Site-directed mutagenesis was conducted to mutate amino acids that reside within the pore domain of the hKv 1.5 channel. Whole-cell patch-clamp method was used to record membrane currents through wild type and mutant hKv 1.5 channels heterologously expressed in CHO cells. Computer docking simulation was conducted to predict the putative binding site(s) of AA in an open-state model of the Kv 1.5 channel. KEY RESULTS The hKv 1.5 current was minimally affected at the onset of depolarization but was progressively reduced during depolarization by the presence of AA, suggesting that AA acts as an open-channel blocker. AA itself affected the channel at extracellular sites independently of its metabolites and signalling pathways. The blocking effect of AA was attenuated at pH 8.0 but not at pH 6.4. The blocking action of AA developed rather rapidly by co-expression of Kv β1.3. The AA-induced block was significantly attenuated in H463C, T480A, R487V, I502A, I508A, V512A and V516A, but not in T462C, A501V and L510A mutants of the hKv 1.5 channel. Docking simulation predicted that H463, T480, R487, I508, V512 and V516 are potentially accessible for interaction with AA. CONCLUSIONS AND IMPLICATIONS AA itself interacts with multiple amino acids located in the pore domain of the hKv 1.5 channel. These findings may provide useful information for future development of selective blockers of hKv 1.5 channels.
Collapse
Affiliation(s)
- Jia‐Yu Bai
- Department of PhysiologyShiga University of Medical ScienceOtsuJapan
| | - Wei‐Guang Ding
- Department of PhysiologyShiga University of Medical ScienceOtsuJapan
| | - Akiko Kojima
- Department of AnesthesiologyShiga University of Medical ScienceOtsuJapan
| | - Tomoyoshi Seto
- Department of AnesthesiologyShiga University of Medical ScienceOtsuJapan
| | - Hiroshi Matsuura
- Department of PhysiologyShiga University of Medical ScienceOtsuJapan
| |
Collapse
|
23
|
Al Kury LT, Voitychuk OI, Yang KHS, Thayyullathil FT, Doroshenko P, Ramez AM, Shuba YM, Galadari S, Howarth FC, Oz M. Effects of the endogenous cannabinoid anandamide on voltage-dependent sodium and calcium channels in rat ventricular myocytes. Br J Pharmacol 2015; 171:3485-98. [PMID: 24758718 DOI: 10.1111/bph.12734] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Revised: 02/17/2014] [Accepted: 03/14/2014] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND AND PURPOSE The endocannabinoid anandamide (N-arachidonoyl ethanolamide; AEA) exerts negative inotropic and antiarrhythmic effects in ventricular myocytes. EXPERIMENTAL APPROACH Whole-cell patch-clamp technique and radioligand-binding methods were used to analyse the effects of anandamide in rat ventricular myocytes. KEY RESULTS In the presence of 1-10 μM AEA, suppression of both Na(+) and L-type Ca(2+) channels was observed. Inhibition of Na(+) channels was voltage and Pertussis toxin (PTX) - independent. Radioligand-binding studies indicated that specific binding of [(3) H] batrachotoxin (BTX) to ventricular muscle membranes was also inhibited significantly by 10 μM metAEA, a non-metabolized AEA analogue, with a marked decrease in Bmax values but no change in Kd . Further studies on L-type Ca(2+) channels indicated that AEA potently inhibited these channels (IC50 0.1 μM) in a voltage- and PTX-independent manner. AEA inhibited maximal amplitudes without affecting the kinetics of Ba(2+) currents. MetAEA also inhibited Na(+) and L-type Ca(2+) currents. Radioligand studies indicated that specific binding of [(3) H]isradipine, was inhibited significantly by metAEA. (10 μM), changing Bmax but not Kd . CONCLUSION AND IMPLICATIONS Results indicate that AEA inhibited the function of voltage-dependent Na(+) and L-type Ca(2+) channels in rat ventricular myocytes, independent of CB1 and CB2 receptor activation.
Collapse
Affiliation(s)
- Lina T Al Kury
- Laboratory of Functional Lipidomics, Department of Pharmacology, UAE University, Al Ain, UAE
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
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
|
25
|
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
|
26
|
Kleberg K, Hassing HA, Hansen HS. Classical endocannabinoid-like compounds and their regulation by nutrients. Biofactors 2014; 40:363-72. [PMID: 24677570 DOI: 10.1002/biof.1158] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2014] [Accepted: 01/07/2014] [Indexed: 11/08/2022]
Abstract
Endocannabinoid-like compounds are structurally related to the true endocannabinoids but do not contain highly unsaturated fatty acids, and they do not bind the cannabinoid receptors. The classical endocannabinoid-like compounds include N-acylethanolamines and 2-monoacylglycerols, and their structural resemblance to the endocannabinoids makes them players in the endocannabinoid system, where they can interfere with the actions of the true endocannabinoids, because they in several cases engage the same synthesizing and degrading enzymes. In addition they have pharmacological actions of their own, which are particularly interesting in a nutritional and metabolic context. Exogenously supplied oleoylethanolamide, palmitoylethanolamide, and linoleoylethanolamide have anorexic effects, and the endogenous formation of these N-acylethanolamines in the small intestine may serve an important role in regulating food intake, through signaling via PPARα and the vagus nerve to the brain appetite center. A chronic high-fat diet will decrease intestinal levels of these anorectic N-acylethanolamines and this may contribute to the hyperphagic effect of high-fat diet; 2-monoacylglycerols mediate endocrine responses in the small intestine; probably trough activation of GPR119 on enteroendocrine cells, and diet-derived 2-monoacylglycerols, for example, 2-oleoylglycerol and 2-palmitoylglycerol might be important for intestinal fat sensing. Whether these 2-monoacylglycerols have signaling functions in other tissues is unclear at present.
Collapse
Affiliation(s)
- Karen Kleberg
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | | | | |
Collapse
|
27
|
Palmitoylethanolamide in CNS health and disease. Pharmacol Res 2014; 86:32-41. [PMID: 24844438 DOI: 10.1016/j.phrs.2014.05.006] [Citation(s) in RCA: 113] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Revised: 05/07/2014] [Accepted: 05/08/2014] [Indexed: 01/08/2023]
Abstract
The existence of acylethanolamides (AEs) in the mammalian brain has been known for decades. Among AEs, palmitoylethanolamide (PEA) is abundant in the central nervous system (CNS) and conspicuously produced by neurons and glial cells. Antihyperalgesic and neuroprotective properties of PEA have been mainly related to the reduction of neuronal firing and to control of inflammation. Growing evidence suggest that PEA may be neuroprotective during CNS neurodegenerative diseases. Advances in the understanding of the physiology and pharmacology of PEA have potentiated its interest as useful biological tool for disease management. Several rapid non-genomic and delayed genomic mechanisms of action have been identified for PEA as peroxisome proliferator-activated receptor (PPAR)-α dependent. First, an early molecular control, through Ca(+2)-activated intermediate- and/or big-conductance K(+) channels opening, drives to rapid neuronal hyperpolarization. This is reinforced by the increase of the inward Cl(-) currents due to the modulation of the gamma aminobutyric acid A receptor and by the desensitization of the transient receptor potential channel type V1. Moreover, the gene transcription-mediated mechanism sustains the long-term anti-inflammatory effects, by reducing pro-inflammatory enzyme expression and increasing neurosteroid synthesis. Overall, the integration of these different modes of action allows PEA to exert an immediate and prolonged efficacious control in neuron signaling either on inflammatory process or neuronal excitability, maintaining cellular homeostasis. In this review, we will discuss the effect of PEA on metabolism, behavior, inflammation and pain perception, related to the control of central functions and the emerging evidence demonstrating its therapeutic efficacy in several neurodegenerative diseases.
Collapse
|
28
|
Hansen HS. Role of anorectic N-acylethanolamines in intestinal physiology and satiety control with respect to dietary fat. Pharmacol Res 2014; 86:18-25. [PMID: 24681513 DOI: 10.1016/j.phrs.2014.03.006] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Revised: 03/17/2014] [Accepted: 03/18/2014] [Indexed: 02/06/2023]
Abstract
Anandamide is a well-known agonist for the cannabinoid receptors. Along with endogenous anandamide other non-endocannabinoid N-acylethanolamines are also formed, apparently in higher amounts. These include mainly oleoylethanolamide (OEA), palmitoyelethanolamide (PEA) and linoleoylethanolamide (LEA), and they have biological activity by themselves being anorectic and anti-inflammatory. It appears that the major effect of dietary fat on the level of these molecules is in the gastrointestinal system, where OEA, PEA and LEA in the enterocytes may function as homeostatic signals, which are decreased by prolonged consumption of a high-fat diet. These lipid amides appear to mediate their signaling activity via activation of PPARα in the enterocyte followed by activation of afferent vagal fibers leading to the brain. Through this mechanism OEA, PEA and LEA may both reduce the consumption of a meal as well as increase the reward value of the food. Thus, they may function as homeostatic intestinal signals involving hedonic aspects that contribute to the regulation of the amounts of dietary fat to be ingested.
Collapse
Affiliation(s)
- Harald S Hansen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark.
| |
Collapse
|
29
|
Andrag E, Curtis MJ. Feasibility of targeting ischaemia-related ventricular arrhythmias by mimicry of endogenous protection by endocannabinoids. Br J Pharmacol 2014; 169:1840-8. [PMID: 23713981 DOI: 10.1111/bph.12252] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2013] [Revised: 05/03/2013] [Accepted: 05/15/2013] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND AND PURPOSE The hypothesis that endocannabinoids protect hearts against ventricular fibrillation (VF) induced by myocardial ischaemia and reperfusion was examined, and the concept that cannabinoids may represent a new class of anti-VF drug was tested. EXPERIMENTAL APPROACH In rat isolated hearts (Langendorff perfusion), VF evoked by reperfusion after 60 min regional ischaemia is known to be exacerbated by inhibitors of endogenous protectants such as nitric oxide. This preparation was used to assay the effects of cannabinoid agonists and antagonists, and the protocols were varied to examine mechanisms. KEY RESULTS Reperfusion-induced VF was not facilitated by relatively selective CB1 (1 μM AM251) or CB2 (1 μM AM630) antagonists. VF evoked during early (30 min) acute ischaemia was also unaffected. However, AM251 significantly increased the incidence of VF and the duration of VF episodes occurring during the later stage of acute ischaemia (30-60 min). AM630 had no such effects. In a separate study, cannabinoid perfusion (anandamide or 2-arachidonoylglycerol, both 0.01-1 μM) failed to reduce VF incidence concentration-dependently during 30 min ischaemia. In all these studies, changes in ancillary variables (QT, PR, heart rate) were unrelated to changes in VF. CONCLUSIONS AND IMPLICATIONS Endocannabinoids are not endogenous anti-VF mediators during reperfusion, but may have a weak protective effect during the late stages of ischaemia, mediated via CB1 agonism. This does not suggest endocannabinoids are important endogenous protectants in these settings, or that CB1 (or CB2) receptors are useful novel targets for developing drugs for VF.
Collapse
Affiliation(s)
- Ellen Andrag
- Cardiovascular Division, King's College London, London, UK
| | | |
Collapse
|
30
|
Effects of cannabinoid receptor type 2 on endogenous myocardial regeneration by activating cardiac progenitor cells in mouse infarcted heart. SCIENCE CHINA-LIFE SCIENCES 2014; 57:201-8. [PMID: 24430557 DOI: 10.1007/s11427-013-4604-z] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Accepted: 09/20/2013] [Indexed: 01/26/2023]
Abstract
Cannabinoid receptor type 2 (CB2) activation is recently reported to promote proliferation of some types of resident stem cells (e.g., hematopoietic stem/progenitor cell or neural progenitor cell). Resident cardiac progenitor cell (CPC) activation and proliferation are crucial for endogenous cardiac regeneration and cardiac repair after myocardial infarction (MI). This study aims to explore the role and possible mechanisms of CB2 receptor activation in enhancing myocardial repair. Our results revealed that CB2 receptor agonist AM1241 can significantly increase CPCs by c-kit and Runx1 staining in ischemic myocardium as well as improve cardiomyocyte proliferation. AM1241 also decreased serum levels of MDA, TNF-α and IL-6 after MI. In addition, AM1241 can ameliorate left ventricular ejection fraction and fractional shortening, and reduce fibrosis. Moreover, AM1241 treatment markedly increased p-Akt and HO-1 expression, and promoted Nrf-2 nuclear translocation. However, PI3K inhibitor wortmannin eliminated these cardioprotective roles of AM1241. In conclusion, AM1241 could induce myocardial regeneration and improve cardiac function, which might be associated with PI3K/Akt/Nrf2 signaling pathway activation. Our findings may provide a promising strategy for cardiac endogenous regeneration after MI.
Collapse
|
31
|
Keppel Hesselink JM, Kopsky DJ, Witkamp RF. Palmitoylethanolamide (PEA)—‘Promiscuous’ anti-inflammatory and analgesic molecule at the interface between nutrition and pharma. PHARMANUTRITION 2014. [DOI: 10.1016/j.phanu.2013.11.127] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
32
|
Zhang CQ, Wu HJ, Wang SY, Yin S, Lu XJ, Miao Y, Wang XH, Yang XL, Wang Z. Suppression of outward K⁺ currents by WIN55212-2 in rat retinal ganglion cells is independent of CB1/CB2 receptors. Neuroscience 2013; 253:183-93. [PMID: 24013008 DOI: 10.1016/j.neuroscience.2013.08.056] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2013] [Revised: 08/25/2013] [Accepted: 08/26/2013] [Indexed: 10/26/2022]
Abstract
Cannabinoid CB1 receptor (CB1R) signaling system is extensively distributed in the vertebrate retina. Activation of CB1Rs regulates a variety of functions of retinal neurons through modulating different ion channels. In the present work we studied effects of this receptor signaling on K(+) channels in retinal ganglion cells by patch-clamp techniques. The CB1R agonist WIN55212-2 (WIN) suppressed outward K(+) currents in acutely isolated rat retinal ganglion cells in a dose-dependent manner, with an IC50 of 4.7 μM. We further showed that WIN mainly suppressed the tetraethylammonium (TEA)-sensitive K(+) current component. While CB1Rs were expressed in rat retinal ganglion cells, the WIN effect on K(+) currents was not blocked by either AM251/SR141716, specific CB1R antagonists, or AM630, a selective CB2R antagonist. Consistently, cAMP-protein kinase A (PKA) and mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK) signaling pathways were unlikely involved in the WIN-induced suppression of the K(+) currents because both PKA inhibitors H-89/Rp-cAMP and MAPK/ERK1/2 inhibitor U0126 failed to block the WIN effects. WIN-induced suppression of the K(+) currents was not observed when WIN was intracellularly applied. Furthermore, an endogenous ligand of the cannabinoid receptor anandamide, the specific CB1R agonist ACEA and the selective CB2R agonist CB65 also suppressed the K(+) currents, and the effects were not blocked by AM251/SR141716 or AM630 respectively. All these results suggest that the WIN-induced suppression of the outward K(+) currents in rat retinal ganglion cells, thereby regulating the cell excitability, were not through CB1R/CB2R signaling pathways.
Collapse
Affiliation(s)
- C-Q Zhang
- Institutes of Brain Science, Institute of Neurobiology and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200032, China
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Smithers N, Bolivar JH, Lee AG, East JM. Characterizing the fatty acid binding site in the cavity of potassium channel KcsA. Biochemistry 2012; 51:7996-8002. [PMID: 22971149 PMCID: PMC3466778 DOI: 10.1021/bi3009196] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
![]()
We show that interactions of fatty acids with the central
cavity of potassium channel KcsA can be characterized using the fluorescence
probe 11-dansylaminoundecanoic acid (Dauda). The fluorescence emission
spectrum of Dauda bound to KcsA in bilayers of dioleoylphosphatidylcholine
contains three components, which can be attributed to KcsA-bound and
lipid-bound Dauda together with unbound Dauda. The binding of Dauda
to KcsA was characterized by a dissociation constant of 0.47 ±
0.10 μM with 0.94 ± 0.06 binding site per KcsA tetramer.
Displacement of KcsA-bound Dauda by the tetrabutylammonium (TBA) ion
confirmed that the Dauda binding site was in the central cavity of
KcsA. Dissociation constants for a range of fatty acids were determined
by displacement of Dauda: binding of fatty acids increased in strength
with an increasing chain length from C14 to C20 but then decreased
in strength from C20 to C22. Increasing the number of double bonds
in the chain from one to four had little effect on binding, dissociation
constants for oleic acid and arachidonic acid, for example, being
2.9 ± 0.2 and 3.0 ± 0.4 μM, respectively. Binding
of TBA to KcsA was very slow, whereas binding of Dauda was fast, suggesting
that TBA can enter the cavity only through an open channel whereas
Dauda can bind to the closed channel, presumably entering the cavity
via the lipid bilayer.
Collapse
Affiliation(s)
- Natalie Smithers
- Centre for Biological Sciences, Life Sciences Building, University of Southampton , Southampton SO17 1BJ, UK
| | | | | | | |
Collapse
|
34
|
Patrón electrocardiográfico de Brugada inducido por cannabis. Rev Esp Cardiol 2012; 65:856-8. [DOI: 10.1016/j.recesp.2012.01.013] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2011] [Accepted: 01/01/2012] [Indexed: 11/18/2022]
|
35
|
Voitychuk OI, Asmolkova VS, Gula NM, Sotkis GV, Galadari S, Howarth FC, Oz M, Shuba YM. Modulation of excitability, membrane currents and survival of cardiac myocytes by N-acylethanolamines. Biochim Biophys Acta Mol Cell Biol Lipids 2012; 1821:1167-76. [DOI: 10.1016/j.bbalip.2012.05.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2011] [Revised: 04/17/2012] [Accepted: 05/11/2012] [Indexed: 12/21/2022]
|
36
|
Romero-Puche AJ, Trigueros-Ruiz N, Cerdán-Sánchez MC, Pérez-Lorente F, Roldán D, Vicente-Vera T. Brugada Electrocardiogram Pattern Induced by Cannabis. ACTA ACUST UNITED AC 2012. [DOI: 10.1016/j.rec.2012.01.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
37
|
Li Q, Ma HJ, Song SL, Shi M, Ma HJ, Li DP, Zhang Y. Effects of anandamide on potassium channels in rat ventricular myocytes: a suppression of Ito and augmentation of KATP channels. Am J Physiol Cell Physiol 2012; 302:C924-30. [DOI: 10.1152/ajpcell.00228.2011] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Anandamide is an endocannabinoid that has antiarrhythmic effects through inhibition of L-type Ca2+ channels in cardiomyocytes. In this study, we investigated the electrophysiological effects of anandamide on K+ channels in rat ventricular myocytes. Whole cell patch-clamp technique was used to record K+ currents, including transient outward potassium current ( Ito), steady-state outward potassium current ( Iss), inward rectifier potassium current ( IK1), and ATP-sensitive potassium current ( IKATP) in isolated rat cardiac ventricular myocytes. Anandamide decreased Ito while increasing IKATP in a concentration-dependent manner but had no effect on Iss and IK1 in isolated ventricular myocytes. Furthermore, anandamide shifted steady-state inactivation curve of Ito to the left and shifted the recovery curve of Ito to the right. However, neither cannabinoid 1 (CB1) receptor antagonist AM251 nor CB2 receptor antagonist AM630 eliminated the inhibitory effect of anandamide on Ito. In addition, blockade of CB2 receptors, but not CB1 receptors, eliminated the augmentation effect of anandamide on IKATP. These data suggest that anandamide suppresses Ito through a non-CB1 and non-CB2 receptor-mediated pathway while augmenting IKATP through CB2 receptors in ventricular myocytes.
Collapse
Affiliation(s)
- Qian Li
- Department of Physiology, Hebei Medical University, Shijiazhuang, China
| | - Hui-Jie Ma
- Department of Physiology, Hebei Medical University, Shijiazhuang, China
| | - Sheng-Li Song
- Department of Internal Medicine Renal Division, First Hospital of Baoding City, Baoding, China; and
| | - Min Shi
- Department of Physiology, Hebei Medical University, Shijiazhuang, China
| | - Hui-Juan Ma
- Department of Physiology, Hebei Medical University, Shijiazhuang, China
| | - De-Pei Li
- Department of Critical Care, The University of Texas MD Anderson Cancer Center, Houston Texas
| | - Yi Zhang
- Department of Physiology, Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
38
|
Chinchilla A, Daimi H, Lozano-Velasco E, Dominguez JN, Caballero R, Delpón E, Tamargo J, Cinca J, Hove-Madsen L, Aranega AE, Franco D. PITX2 insufficiency leads to atrial electrical and structural remodeling linked to arrhythmogenesis. CIRCULATION. CARDIOVASCULAR GENETICS 2011; 4:269-79. [PMID: 21511879 DOI: 10.1161/circgenetics.110.958116] [Citation(s) in RCA: 196] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND Pitx2 is a homeobox transcription factor that plays a pivotal role in early left/right determination during embryonic development. Pitx2 loss-of-function mouse mutants display early embryonic lethality with severe cardiac malformations, demonstrating the importance of Pitx2 during cardiogenesis. Recently, independent genome-wide association studies have provided new evidence for a putative role of PITX2 in the adult heart. These studies have independently reported several risk variants close to the PITX2 locus on chromosome 4q25 that are strongly associated with atrial fibrillation in humans. METHODS AND RESULTS We show for the first time that PITX2C expression is significantly decreased in human patients with sustained atrial fibrillation, thus providing a molecular link between PITX2 loss of function and atrial fibrillation. In addition, morphological, molecular, and electrophysiological characterization of chamber-specific Pitx2 conditional mouse mutants reveals that atrial but not ventricular chamber-specific deletion of Pitx2 results in differences in the action potential amplitude and resting membrane potential in the adult heart as well as ECG characteristics of atrioventricular block. Lack of Pitx2 in atrial myocardium impairs sodium channel and potassium channel expression, mediated in part by miRNA misexpression. CONCLUSIONS This study thus identifies Pitx2 as an upstream transcriptional regulator of atrial electric function, the insufficiency of which results in cellular and molecular changes leading to atrial electric and structural remodeling linked to arrhythmogenesis.
Collapse
Affiliation(s)
- Ana Chinchilla
- Department of Experimental Biology, University of Jaén, Jaén, Spain
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Ravens U, Wettwer E. Ultra-rapid delayed rectifier channels: molecular basis and therapeutic implications. Cardiovasc Res 2010; 89:776-85. [DOI: 10.1093/cvr/cvq398] [Citation(s) in RCA: 97] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
|
40
|
Pavón FJ, Serrano A, Romero-Cuevas M, Alonso M, Rodríguez de Fonseca F. Oleoylethanolamide: a new player in peripheral control of energy metabolism. Therapeutic implications. ACTA ACUST UNITED AC 2010. [DOI: 10.1016/j.ddmec.2011.02.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
41
|
Pertwee RG, Howlett AC, Abood ME, Alexander SPH, Di Marzo V, Elphick MR, Greasley PJ, Hansen HS, Kunos G, Mackie K, Mechoulam R, Ross RA. International Union of Basic and Clinical Pharmacology. LXXIX. Cannabinoid receptors and their ligands: beyond CB₁ and CB₂. Pharmacol Rev 2010; 62:588-631. [PMID: 21079038 PMCID: PMC2993256 DOI: 10.1124/pr.110.003004] [Citation(s) in RCA: 1220] [Impact Index Per Article: 81.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
There are at least two types of cannabinoid receptors (CB(1) and CB(2)). Ligands activating these G protein-coupled receptors (GPCRs) include the phytocannabinoid Δ(9)-tetrahydrocannabinol, numerous synthetic compounds, and endogenous compounds known as endocannabinoids. Cannabinoid receptor antagonists have also been developed. Some of these ligands activate or block one type of cannabinoid receptor more potently than the other type. This review summarizes current data indicating the extent to which cannabinoid receptor ligands undergo orthosteric or allosteric interactions with non-CB(1), non-CB(2) established GPCRs, deorphanized receptors such as GPR55, ligand-gated ion channels, transient receptor potential (TRP) channels, and other ion channels or peroxisome proliferator-activated nuclear receptors. From these data, it is clear that some ligands that interact similarly with CB(1) and/or CB(2) receptors are likely to display significantly different pharmacological profiles. The review also lists some criteria that any novel "CB(3)" cannabinoid receptor or channel should fulfil and concludes that these criteria are not currently met by any non-CB(1), non-CB(2) pharmacological receptor or channel. However, it does identify certain pharmacological targets that should be investigated further as potential CB(3) receptors or channels. These include TRP vanilloid 1, which possibly functions as an ionotropic cannabinoid receptor under physiological and/or pathological conditions, and some deorphanized GPCRs. Also discussed are 1) the ability of CB(1) receptors to form heteromeric complexes with certain other GPCRs, 2) phylogenetic relationships that exist between CB(1)/CB(2) receptors and other GPCRs, 3) evidence for the existence of several as-yet-uncharacterized non-CB(1), non-CB(2) cannabinoid receptors; and 4) current cannabinoid receptor nomenclature.
Collapse
MESH Headings
- Cannabinoid Receptor Agonists
- Cannabinoid Receptor Antagonists
- Cannabinoid Receptor Modulators/metabolism
- Cannabinoids/metabolism
- Humans
- Ligands
- Phylogeny
- Receptor, Cannabinoid, CB1/agonists
- Receptor, Cannabinoid, CB1/antagonists & inhibitors
- Receptor, Cannabinoid, CB1/metabolism
- Receptor, Cannabinoid, CB2/agonists
- Receptor, Cannabinoid, CB2/antagonists & inhibitors
- Receptor, Cannabinoid, CB2/metabolism
- Receptors, Cannabinoid/metabolism
- Terminology as Topic
Collapse
Affiliation(s)
- R G Pertwee
- School of Medical Sciences, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, Scotland, UK.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Moreno-Galindo EG, Barrio-Echavarría GF, Vásquez JC, Decher N, Sachse FB, Tristani-Firouzi M, Sánchez-Chapula JA, Navarro-Polanco RA. Molecular basis for a high-potency open-channel block of Kv1.5 channel by the endocannabinoid anandamide. Mol Pharmacol 2010; 77:751-8. [PMID: 20133392 DOI: 10.1124/mol.109.063008] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The endocannabinoid, N-arachidonoylethanolamine (anandamide; AEA) is known to interact with voltage-gated K(+) (Kv) channels in a cannabinoid receptor-independent manner. AEA modulates the functional properties of Kv channels, converting channels with slowly inactivating current into apparent fast inactivation. In this study, we characterize the mechanism of action and binding site for AEA on Kv1.5 channels expressed on HEK-293 cells using the patch-clamp techniques. AEA exhibited high-potency block (IC(50) approximately 200 nM) from the cytoplasmic membrane surface, consistent with open-channel block. Alanine-scanning mutagenesis revealed that AEA interacts with two crucial beta-branching amino acids, Val505 and Ile508 within the S6 domain. Both residues face toward the central cavity and constitute a motif that forms a hydrophobic ring around the ion conduction pathway. This hydrophobic ring motif may be a critical determinant of cannabinoid receptor-independent AEA modulation in other K(+) channel families.
Collapse
Affiliation(s)
- Eloy G Moreno-Galindo
- Centro Universitario de Investigaciones Biomédicas, Universidad de Colima, Colima, Mexico
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Hansen HS. Palmitoylethanolamide and other anandamide congeners. Proposed role in the diseased brain. Exp Neurol 2010; 224:48-55. [PMID: 20353771 DOI: 10.1016/j.expneurol.2010.03.022] [Citation(s) in RCA: 92] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2010] [Accepted: 03/25/2010] [Indexed: 10/19/2022]
Abstract
Acylethanolamides are formed in the brain "on demand" from membrane phospholipids called N-acylated phosphatidylethanolamines. The acylethanolamides are signaling molecules of lipid nature, and this lipofilicity suggests an autocrine function. The acylethanolamides include palmitoylethanolamide (PEA), oleoylethanolamide (OEA), stearoylethanolamide (SEA), and several other quantitative minor species including anandamide (= arachidonoylethanolamide). PEA and OEA can activate several different receptors and inhibit some ion channels, e.g., PPARalpha, vanilloid receptor, K(+) channels (Kv4.3, Kv1.5), and OEA can activate GPR119 and inhibit ceramidases. Targets for SEA are less clear, but it has some cannabimimetic actions in rats in vivo. All acylethanolamides accumulate during neuronal injury, and injected OEA has neuroprotective effects, and PEA has anti-inflammatory effects as studied in the peripheral system. Several of the pharmacological effects seem to be mediated via activation of PPARalpha. Recently, injected OEA has been found to consolidate memories in rats. Inhibitors of the acylethanolamide-degrading enzyme FAAH can increase levels of all acylethanolamides including annandamide, and some of the pharmacological effects caused by these inhibitors may be explained by increased cerebral levels of OEA and PEA, e.g., suppression of nicotine-induced activation of dopamine neurons. Furthermore, through activation of PPARalpha, OEA and PEA may stimulate neurosteroid synthesis, thereby modulating several biological functions mediated by GABA(A) receptors. The existence of acylethanolamides in the mammalian brain has been known for decades, but it is first within the last few years that the putative biological functions of the three most abundant acylethanolamides species are starting to emerge.
Collapse
Affiliation(s)
- Harald S Hansen
- Department of Pharmacology & Pharmacotherapy, Faculty of Pharmaceutical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark.
| |
Collapse
|
44
|
Answer to the "comment on functional consequences of Kv1.3 ion channel rearrangement into the immunological synapse" by Stefan Bittner et al. [Immunol. Lett. 125 (Aug 15 (2)) (2009) 156-157]. Immunol Lett 2010; 129:47-9. [PMID: 20079762 DOI: 10.1016/j.imlet.2009.12.026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2009] [Revised: 12/22/2009] [Accepted: 12/27/2009] [Indexed: 11/24/2022]
|