1
|
Incontro S, Musella ML, Sammari M, Di Scala C, Fantini J, Debanne D. Lipids shape brain function through ion channel and receptor modulations: physiological mechanisms and clinical perspectives. Physiol Rev 2025; 105:137-207. [PMID: 38990068 DOI: 10.1152/physrev.00004.2024] [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: 01/16/2024] [Revised: 05/28/2024] [Accepted: 07/01/2024] [Indexed: 07/12/2024] Open
Abstract
Lipids represent the most abundant molecular type in the brain, with a fat content of ∼60% of the dry brain weight in humans. Despite this fact, little attention has been paid to circumscribe the dynamic role of lipids in brain function and disease. Membrane lipids such as cholesterol, phosphoinositide, sphingolipids, arachidonic acid, and endocannabinoids finely regulate both synaptic receptors and ion channels that ensure critical neural functions. After a brief introduction on brain lipids and their respective properties, we review here their role in regulating synaptic function and ion channel activity, action potential propagation, neuronal development, and functional plasticity and their contribution in the development of neurological and neuropsychiatric diseases. We also provide possible directions for future research on lipid function in brain plasticity and diseases.
Collapse
Affiliation(s)
| | | | - Malika Sammari
- UNIS, INSERM, Aix-Marseille Université, Marseille, France
| | | | | | | |
Collapse
|
2
|
Gentile F, Orlando G, Montuoro S, Ferrari Chen YF, Macefield V, Passino C, Giannoni A, Emdin M. Treating heart failure by targeting the vagus nerve. Heart Fail Rev 2024; 29:1201-1215. [PMID: 39117958 PMCID: PMC11455679 DOI: 10.1007/s10741-024-10430-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/28/2024] [Indexed: 08/10/2024]
Abstract
Increased sympathetic and reduced parasympathetic nerve activity is associated with disease progression and poor outcomes in patients with chronic heart failure. The demonstration that markers of autonomic imbalance and vagal dysfunction, such as reduced heart rate variability and baroreflex sensitivity, hold prognostic value in patients with chronic heart failure despite modern therapies encourages the research for neuromodulation strategies targeting the vagus nerve. However, the approaches tested so far have yielded inconclusive results. This review aims to summarize the current knowledge about the role of the parasympathetic nervous system in chronic heart failure, describing the pathophysiological background, the methods of assessment, and the rationale, limits, and future perspectives of parasympathetic stimulation either by drugs or bioelectronic devices.
Collapse
Affiliation(s)
- Francesco Gentile
- Health Science Interdisciplinary Center, Scuola Superiore Sant'Anna, Piazza Martiri Della Libertà 33, 56127, Pisa, Italy.
- Cardiology and Cardiovascular Medicine Division, Fondazione Monasterio, Via G. Moruzzi 1, 56124, Pisa, Italy.
| | - Giulia Orlando
- Health Science Interdisciplinary Center, Scuola Superiore Sant'Anna, Piazza Martiri Della Libertà 33, 56127, Pisa, Italy
| | - Sabrina Montuoro
- Health Science Interdisciplinary Center, Scuola Superiore Sant'Anna, Piazza Martiri Della Libertà 33, 56127, Pisa, Italy
| | - Yu Fu Ferrari Chen
- Cardiology and Cardiovascular Medicine Division, Fondazione Monasterio, Via G. Moruzzi 1, 56124, Pisa, Italy
| | | | - Claudio Passino
- Health Science Interdisciplinary Center, Scuola Superiore Sant'Anna, Piazza Martiri Della Libertà 33, 56127, Pisa, Italy
- Cardiology and Cardiovascular Medicine Division, Fondazione Monasterio, Via G. Moruzzi 1, 56124, Pisa, Italy
| | - Alberto Giannoni
- Health Science Interdisciplinary Center, Scuola Superiore Sant'Anna, Piazza Martiri Della Libertà 33, 56127, Pisa, Italy
- Cardiology and Cardiovascular Medicine Division, Fondazione Monasterio, Via G. Moruzzi 1, 56124, Pisa, Italy
| | - Michele Emdin
- Health Science Interdisciplinary Center, Scuola Superiore Sant'Anna, Piazza Martiri Della Libertà 33, 56127, Pisa, Italy
- Cardiology and Cardiovascular Medicine Division, Fondazione Monasterio, Via G. Moruzzi 1, 56124, Pisa, Italy
| |
Collapse
|
3
|
Vakalopoulos A, Basting D, Brechmann M, Teller H, Boultadakis Arapinis M, Straub A, Mittendorf J, Meininghaus M, Müller T, Nowak-Reppel K, Schäfer M, Wittwer M, Kullmann M, Terjung C, Lang D, Poethko T, Marquardt T, Freudenberger T, Mondritzki T, Hüser J, Heckmann M, Tinel H. Discovery of BAY 2413555, First Selective Positive Allosteric Modulator of the M2 Receptor to Restore Cardiac Autonomic Balance. J Med Chem 2024. [PMID: 39463278 DOI: 10.1021/acs.jmedchem.4c01590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Autonomic disbalance, i.e., sympathetic overactivation and parasympathetic withdrawal, is a causal driver of disease progression in heart failure. While sympatholytic drugs are established treatments, no drug therapy restoring vagal control of cardiac function is available. We report here the HTS-based discovery of a novel class of 1,8-naphthyridin-4(1H)-one carboxamides acting as positive allosteric modulators (PAMs) of the M2 muscarinic acetylcholine receptor (M2R). M2R is the main postsynaptic myocyte receptor regulating heart rate, electrical conduction, and contractile strength. Extensive optimization of the screening hit in terms of potency, permeation, metabolic stability, and solubility ultimately resulted in the discovery of the first-in-class clinical candidate BAY 2413555 (27). With an overall technical profile compatible with once-daily oral administration in a phase 1 study, no apparent effects on blood pressure, and a mechanism that largely preserves autonomic regulatory capacity, BAY 2413555 could be the tool to finally study the restoration of autonomic balance.
Collapse
Affiliation(s)
- Alexandros Vakalopoulos
- Bayer AG, Pharmaceuticals, Research and Development, Aprather Weg 18a, Wuppertal 42113, Germany
| | - Daniel Basting
- Bayer AG, Pharmaceuticals, Research and Development, Aprather Weg 18a, Wuppertal 42113, Germany
| | - Markus Brechmann
- Bayer AG, Pharmaceuticals, Research and Development, Aprather Weg 18a, Wuppertal 42113, Germany
- CSL Innovation GmbH, Emil-von-Behring-Str. 76, Marburg 35041, Germany
| | - Henrik Teller
- Bayer AG, Pharmaceuticals, Research and Development, Aprather Weg 18a, Wuppertal 42113, Germany
- micromod Partikeltechnologie GmbH, Schillingallee 68, Rostock 18057, Germany
| | | | - Alexander Straub
- Bayer AG, Pharmaceuticals, Research and Development, Aprather Weg 18a, Wuppertal 42113, Germany
| | - Joachim Mittendorf
- Bayer AG, Pharmaceuticals, Research and Development, Aprather Weg 18a, Wuppertal 42113, Germany
| | - Mark Meininghaus
- Bayer AG, Pharmaceuticals, Research and Development, Aprather Weg 18a, Wuppertal 42113, Germany
| | - Thomas Müller
- Bayer AG, Pharmaceuticals, Research and Development, Aprather Weg 18a, Wuppertal 42113, Germany
| | - Katrin Nowak-Reppel
- Bayer AG, Pharmaceuticals, Research and Development, Aprather Weg 18a, Wuppertal 42113, Germany
| | - Martina Schäfer
- Bayer AG, Pharmaceuticals, Research and Development, Aprather Weg 18a, Wuppertal 42113, Germany
- Nuvisan ICB GmbH, Müllerstrasse 178, Berlin 13353, Germany
| | - Matthias Wittwer
- Bayer AG, Pharmaceuticals, Research and Development, Aprather Weg 18a, Wuppertal 42113, Germany
- F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel 4070, Switzerland
| | - Maximilian Kullmann
- Bayer AG, Pharmaceuticals, Research and Development, Aprather Weg 18a, Wuppertal 42113, Germany
- BioNTech SE, An der Goldgrube 12, Mainz 55131, Germany
| | - Carsten Terjung
- Bayer AG, Pharmaceuticals, Research and Development, Aprather Weg 18a, Wuppertal 42113, Germany
| | - Dieter Lang
- Bayer AG, Pharmaceuticals, Research and Development, Aprather Weg 18a, Wuppertal 42113, Germany
| | - Thorsten Poethko
- Bayer AG, Pharmaceuticals, Research and Development, Aprather Weg 18a, Wuppertal 42113, Germany
| | - Tobias Marquardt
- Bayer AG, Pharmaceuticals, Research and Development, Aprather Weg 18a, Wuppertal 42113, Germany
| | - Till Freudenberger
- Bayer AG, Pharmaceuticals, Research and Development, Aprather Weg 18a, Wuppertal 42113, Germany
| | - Thomas Mondritzki
- Bayer AG, Pharmaceuticals, Research and Development, Aprather Weg 18a, Wuppertal 42113, Germany
- University of Witten/Herdecke, Witten 58455, Germany
| | - Jörg Hüser
- Bayer AG, Pharmaceuticals, Research and Development, Aprather Weg 18a, Wuppertal 42113, Germany
- Axxam S.p.A., Openzone - Via Meucci 3, Bresso, Milan 20091, Italy
| | - Michael Heckmann
- Bayer AG, Pharmaceuticals, Research and Development, Aprather Weg 18a, Wuppertal 42113, Germany
| | - Hanna Tinel
- Bayer AG, Pharmaceuticals, Research and Development, Aprather Weg 18a, Wuppertal 42113, Germany
| |
Collapse
|
4
|
Dias P, Salam R, Moravcová M, Saadat S, Pourová J, Vopršalová M, Jirkovský E, Tebbens JD, Mladěnka P. 3-methoxycatechol causes vasodilation likely via K V channels: ex vivo, in silico docking and in vivo study. Vascul Pharmacol 2024; 156:107418. [PMID: 39159736 DOI: 10.1016/j.vph.2024.107418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 08/09/2024] [Accepted: 08/16/2024] [Indexed: 08/21/2024]
Abstract
Substituted catechols include both natural and synthetic compounds found in the environment and foods. Some of them are flavonoid metabolites formed by the gut microbiota which are absorbed afterwards. Our previous findings showed that one of these metabolites, 4-methylcatechol, exerts potent vasorelaxant effects in rats. In the current study, we aimed at testing of its 22 structural congeners in order to find the most potent structure and to investigate the mechanism of action. 3-methoxycatechol (3-MOC), 4-ethylcatechol, 3,5-dichlorocatechol, 4-tert-butylcatechol, 4,5-dichlorocatechol, 3-fluorocatechol, 3-isopropylcatechol, 3-methylcatechol and the parent 4-methylcatechol exhibited high vasodilatory activities on isolated rat aortic rings with EC50s ranging from ∼10 to 24 μM. Some significant sex-differences were found. The most potent compound, 3-MOC, relaxed also resistant mesenteric artery but not porcine coronary artery, and decreased arterial blood pressure in both male and female spontaneously hypertensive rats in vivo without affecting heart rate. It potentiated the vasodilation mediated by cAMP and cGMP, but did not impact L-type Ca2+-channels. By using two inhibitors, activation of voltage-gated potassium channels (KV) was found to be involved in the mechanism of action. This was corroborated by docking analysis of 3-MOC with the KV7.4 channel. None of the most active catechols decreased the viability of the A-10 rat embryonic thoracic aorta smooth muscle cell line. Our findings showed that various catechols can relax vascular smooth muscles and hence could provide templates for developing new antihypertensive vasodilator agents without affecting coronary circulation.
Collapse
MESH Headings
- Animals
- Vasodilation/drug effects
- Male
- Catechols/pharmacology
- Catechols/chemistry
- Molecular Docking Simulation
- Vasodilator Agents/pharmacology
- Vasodilator Agents/chemistry
- Female
- Rats, Inbred SHR
- Mesenteric Arteries/drug effects
- Mesenteric Arteries/metabolism
- Potassium Channels, Voltage-Gated/metabolism
- Potassium Channels, Voltage-Gated/antagonists & inhibitors
- Potassium Channels, Voltage-Gated/drug effects
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Swine
- Dose-Response Relationship, Drug
- Aorta, Thoracic/drug effects
- Aorta, Thoracic/metabolism
- Hypertension/drug therapy
- Hypertension/physiopathology
- Hypertension/metabolism
- Arterial Pressure/drug effects
- Coronary Vessels/drug effects
- Coronary Vessels/metabolism
- Rats
- Sex Factors
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Disease Models, Animal
- Structure-Activity Relationship
- Cyclic GMP/metabolism
Collapse
Affiliation(s)
- Patrícia Dias
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Charles University, Akademika Heyrovského 1203, 500 05 Hradec Kralove, Czech Republic; The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Wexner Medical Center, Columbus, Ohio, USA; Division of Outcomes and Translational Sciences, College of Pharmacy, The Ohio State University, Columbus, Ohio, USA.
| | - Rudy Salam
- Department of Biophysics and Physical Chemistry, Faculty of Pharmacy, Charles University, Akademika Heyrovského 1203, 500 05 Hradec Kralove, Czech Republic.
| | - Monika Moravcová
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Charles University, Akademika Heyrovského 1203, 500 05 Hradec Kralove, Czech Republic.
| | - Saina Saadat
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Charles University, Akademika Heyrovského 1203, 500 05 Hradec Kralove, Czech Republic.
| | - Jana Pourová
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Charles University, Akademika Heyrovského 1203, 500 05 Hradec Kralove, Czech Republic.
| | - Marie Vopršalová
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Charles University, Akademika Heyrovského 1203, 500 05 Hradec Kralove, Czech Republic.
| | - Eduard Jirkovský
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Charles University, Akademika Heyrovského 1203, 500 05 Hradec Kralove, Czech Republic.
| | - Jurjen Duintjer Tebbens
- Department of Biophysics and Physical Chemistry, Faculty of Pharmacy, Charles University, Akademika Heyrovského 1203, 500 05 Hradec Kralove, Czech Republic.
| | - Přemysl Mladěnka
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Charles University, Akademika Heyrovského 1203, 500 05 Hradec Kralove, Czech Republic.
| |
Collapse
|
5
|
Hong X, Yuan L, Zhao X, Shan Y, Qin T, Li J, Zha J. Embryonic Exposure to Organophosphate Flame Retardants (OPFRs) Differentially Induces Cardiotoxicity in Rare Minnow ( Gobiocypris rarus). ENVIRONMENTAL SCIENCE & TECHNOLOGY 2024; 58:13648-13657. [PMID: 39069658 DOI: 10.1021/acs.est.4c01927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Organophosphorus flame retardants (OPFRs) such as triphenyl phosphate (TPHP) and tris(1,3-dichloro-2-propyl) phosphate (TDCIPP) were reported to impair cardiac function in fish. However, limited information is available regarding their cardiotoxic mechanisms. Using rare minnow (Gobiocypris rarus) as a model, we found that both TPHP and TDCIPP exposures decreased heart rate at 96 h postfertilization (hpf) in embryos. Atropine (an mAChR antagonist) can significantly attenuate the bradycardia caused by TPHP, but only marginally attenuated in TDCIPP treatment, suggesting that TDCIPP-induced bradycardia is independent of mAChR. Unlike TDCIPP, although TPHP-induced bradycardia could be reversed by transferring larvae to a clean medium, the inhibitory effect of AChE activity persisted compared to 96 hpf, indicating the existence of other bradycardia regulatory mechanisms. Transcriptome profiling revealed cardiotoxicity-related pathways in treatments at 24 and 72 hpf in embryos/larvae. Similar transcriptional alterations were also confirmed in the hearts of adult fish. Further studies verified that TPHP and TDCIPP can interfere with Na+/Ca2+ transport and lead to disorders of cardiac excitation-contraction coupling in larvae. Our findings provide useful clues for unveiling the differential cardiotoxic mechanisms of OPFRs and identifying abnormal Na+/Ca2+ transport as one of a select few known factors sufficient to impair fish cardiac function.
Collapse
Affiliation(s)
- Xiangsheng Hong
- National Engineering Research Center of Industrial Wastewater Detoxication and Resource Recovery, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
| | - Lilai Yuan
- Fishery Resource and Environment Research Center, Chinese Academy of Fishery Sciences, Beijing 100141, China
| | - Xu Zhao
- State Environmental Protection Key Laboratory of Environmental Pollution Health Risk Assessment, South China Institute of Environmental Sciences, Ministry of Environmental Protection of the People's Republic of China, Guangzhou 510655, China
| | - Yuan Shan
- National Fisheries Technology Extension Center and China Society of Fisheries, Beijing 100125, China
| | - Tianlong Qin
- Aquatic Technology Promotion Guidance Center for Wuhan, 821 Development Avenue, Jiangan District, Wuhan 430014, China
| | - Jiasu Li
- National Engineering Research Center of Industrial Wastewater Detoxication and Resource Recovery, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
| | - Jinmiao Zha
- National Engineering Research Center of Industrial Wastewater Detoxication and Resource Recovery, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
6
|
Gergs U, Wackerhagen S, Fuhrmann T, Schäfer I, Neumann J. Further investigations on the influence of protein phosphatases on the signaling of muscarinic receptors in the atria of mouse hearts. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:5731-5743. [PMID: 38308688 PMCID: PMC11329414 DOI: 10.1007/s00210-024-02973-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 01/21/2024] [Indexed: 02/05/2024]
Abstract
The vagal regulation of cardiac function involves acetylcholine (ACh) receptor activation followed by negative chronotropic and negative as well as positive inotropic effects. The resulting signaling pathways may include Gi/o protein-coupled reduction in adenylyl cyclase (AC) activity, direct Gi/o protein-coupled activation of ACh-activated potassium current (IKACh), inhibition of L-type calcium ion channels, and/or the activation of protein phosphatases. Here, we studied the role of the protein phosphatases 1 (PP1) and 2A (PP2A) for muscarinic receptor signaling in isolated atrial preparations of transgenic mice with cardiomyocyte-specific overexpression of either the catalytic subunit of PP2A (PP2A-TG) or the inhibitor-2 (I2) of PP1 (I2-TG) or in double transgenic mice overexpressing both PP2A and I2 (DT). In mouse left atrial preparations, carbachol (CCh), cumulatively applied (1 nM-10 µM), exerted at low concentrations a negative inotropic effect followed by a positive inotropic effect at higher concentrations. This biphasic effect was noted with CCh alone as well as when CCh was added after β-adrenergic pre-stimulation with isoprenaline (1 µM). Whereas the response to stimulation of β-adrenoceptors or adenosine receptors (used as controls) was changed in PP2A-TG, the response to CCh was unaffected in atrial preparations from all transgenic models studied here. Therefore, the present data tentatively indicate that neither PP2A nor PP1, but possibly other protein phosphatases, is involved in the muscarinic receptor-induced inotropic and chronotropic effects in the mouse heart.
Collapse
Affiliation(s)
- Ulrich Gergs
- Institut Für Pharmakologie Und Toxikologie, Medizinische Fakultät, Martin-Luther-Universität Halle-Wittenberg, D-06097, Magdeburger Str. 4, 06112, Halle, Germany.
| | - Silke Wackerhagen
- Institut Für Pharmakologie Und Toxikologie, Medizinische Fakultät, Martin-Luther-Universität Halle-Wittenberg, D-06097, Magdeburger Str. 4, 06112, Halle, Germany
| | - Tobias Fuhrmann
- Institut Für Pharmakologie Und Toxikologie, Medizinische Fakultät, Martin-Luther-Universität Halle-Wittenberg, D-06097, Magdeburger Str. 4, 06112, Halle, Germany
| | - Inka Schäfer
- Institut Für Pharmakologie Und Toxikologie, Medizinische Fakultät, Martin-Luther-Universität Halle-Wittenberg, D-06097, Magdeburger Str. 4, 06112, Halle, Germany
| | - Joachim Neumann
- Institut Für Pharmakologie Und Toxikologie, Medizinische Fakultät, Martin-Luther-Universität Halle-Wittenberg, D-06097, Magdeburger Str. 4, 06112, Halle, Germany
| |
Collapse
|
7
|
Liu X, Yu Y, Zhang H, Zhang M, Liu Y. The Role of Muscarinic Acetylcholine Receptor M 3 in Cardiovascular Diseases. Int J Mol Sci 2024; 25:7560. [PMID: 39062802 PMCID: PMC11277046 DOI: 10.3390/ijms25147560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 07/04/2024] [Accepted: 07/05/2024] [Indexed: 07/28/2024] Open
Abstract
The muscarinic acetylcholine receptor M3 (M3-mAChR) is involved in various physiological and pathological processes. Owing to specific cardioprotective effects, M3-mAChR is an ideal diagnostic and therapeutic biomarker for cardiovascular diseases (CVDs). Growing evidence has linked M3-mAChR to the development of multiple CVDs, in which it plays a role in cardiac protection such as anti-arrhythmia, anti-hypertrophy, and anti-fibrosis. This review summarizes M3-mAChR's expression patterns, functions, and underlying mechanisms of action in CVDs, especially in ischemia/reperfusion injury, cardiac hypertrophy, and heart failure, opening up a new research direction for the treatment of CVDs.
Collapse
Affiliation(s)
- Xinxing Liu
- Hainan Academy of Medical Sciences, School of Pharmacy, Hainan Medical University, Haikou 571199, China; (X.L.); (Y.Y.); (H.Z.)
| | - Yi Yu
- Hainan Academy of Medical Sciences, School of Pharmacy, Hainan Medical University, Haikou 571199, China; (X.L.); (Y.Y.); (H.Z.)
| | - Haiying Zhang
- Hainan Academy of Medical Sciences, School of Pharmacy, Hainan Medical University, Haikou 571199, China; (X.L.); (Y.Y.); (H.Z.)
| | - Min Zhang
- Hainan Academy of Medical Sciences, School of Pharmacy, Hainan Medical University, Haikou 571199, China; (X.L.); (Y.Y.); (H.Z.)
| | - Yan Liu
- Hainan Academy of Medical Sciences, School of Pharmacy, Hainan Medical University, Haikou 571199, China; (X.L.); (Y.Y.); (H.Z.)
- Engineering Research Center of Tropical Medicine Innovation and Transformation of Ministry of Education, Hainan Academy of Medical Sciences, Hainan Medical University, Haikou 571199, China
- International Joint Research Center of Human–Machine Intelligent Collaborative for Tumor Precision Diagnosis and Treatment of Hainan Province, Hainan Academy of Medical Sciences, Hainan Medical University, Haikou 571199, China
- Hainan Provincial Key Laboratory of Research and Development on Tropical Herbs, Hainan Academy of Medical Sciences, Hainan Medical University, Haikou 571199, China
| |
Collapse
|
8
|
Dolejší E, Janoušková A, Jakubík J. Muscarinic Receptors in Cardioprotection and Vascular Tone Regulation. Physiol Res 2024; 73:S389-S400. [PMID: 38634650 PMCID: PMC11412339 DOI: 10.33549/physiolres.935270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024] Open
Abstract
Muscarinic acetylcholine receptors are metabotropic G-protein coupled receptors. Muscarinic receptors in the cardiovascular system play a central role in its regulation. Particularly M2 receptors slow down the heart rate by reducing the impulse conductivity through the atrioventricular node. In general, activation of muscarinic receptors has sedative effects on the cardiovascular system, including vasodilation, negative chronotropic and inotropic effects on the heart, and cardioprotective effects, including antifibrillatory effects. First, we review the signaling of individual subtypes of muscarinic receptors and their involvement in the physiology and pathology of the cardiovascular system. Then we review age and disease-related changes in signaling via muscarinic receptors in the cardiovascular system. Finally, we review molecular mechanisms involved in cardioprotection mediated by muscarinic receptors leading to negative chronotropic and inotropic and antifibrillatory effects on heart and vasodilation, like activation of acetylcholine-gated inward-rectifier K+-currents and endothelium-dependent and -independent vasodilation. We relate this knowledge with well-established cardioprotective treatments by vagal stimulation and muscarinic agonists. It is well known that estrogen exerts cardioprotective effects against atherosclerosis and ischemia-reperfusion injury. Recently, some sex hormones and neurosteroids have been shown to allosterically modulate muscarinic receptors. Thus, we outline possible treatment by steroid-based positive allosteric modulators of acetylcholine as a novel pharmacotherapeutic tactic. Keywords: Muscarinic receptors, Muscarinic agonists, Allosteric modulation, Cardiovascular system, Cardioprotection, Steroids.
Collapse
Affiliation(s)
- E Dolejší
- Laboratory of Neurochemistry, Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic.
| | | | | |
Collapse
|
9
|
Schwarz R, Hofmann B, Gergs U, Neumann J. Cantharidin and sodium fluoride attenuate the negative inotropic effects of carbachol in the isolated human atrium. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:2183-2202. [PMID: 37801145 PMCID: PMC10933163 DOI: 10.1007/s00210-023-02747-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 09/23/2023] [Indexed: 10/07/2023]
Abstract
Carbachol, an agonist at muscarinic receptors, exerts a negative inotropic effect in human atrium. Carbachol can activate protein phosphatases (PP1 or PP2A). We hypothesized that cantharidin or sodium fluoride, inhibitors of PP1 and PP2A, may attenuate a negative inotropic effect of carbachol. During bypass-surgery trabeculae carneae of human atrial preparations (HAP) were obtained. These trabeculae were mounted in organ baths and electrically stimulated (1 Hz). Force of contraction was measured under isometric conditions. For comparison, we studied isolated electrically stimulated left atrial preparations (LA) from mice. Cantharidin (100 µM) and sodium fluoride (3 mM) increased force of contraction in LA (n = 5-8, p < 0.05) by 113% ± 24.5% and by 100% ± 38.2% and in HAP (n = 13-15, p < 0.05) by 625% ± 169% and by 196% ± 23.5%, respectively. Carbachol (1 µM) alone exerted a rapid transient maximum negative inotropic effect in LA (n = 6) and HAP (n = 14) to 46.9% ± 3.63% and 19.4% ± 3.74%, respectively (p < 0.05). These negative inotropic effects were smaller in LA (n = 4-6) and HAP (n = 9-12) pretreated with 100 µM cantharidin and amounted to 58.0% ± 2.27% and 59.2% ± 6.19% or 3 mM sodium fluoride to 63.7% ± 9.84% and 46.3% ± 5.69%, (p < 0.05). We suggest that carbachol, at least in part, exerts a negative inotropic effect in the human atrium by stimulating the enzymatic activity of PP1 and/or PP2A.
Collapse
Affiliation(s)
- Rebecca Schwarz
- Institute for Pharmacology and Toxicology, Medical Faculty, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Britt Hofmann
- Department of Cardiac Surgery, Mid-German Heart Center, University Hospital Halle, Halle (Saale), Germany
| | - Ulrich Gergs
- Institute for Pharmacology and Toxicology, Medical Faculty, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Joachim Neumann
- Institute for Pharmacology and Toxicology, Medical Faculty, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany.
| |
Collapse
|
10
|
Li SY, Shi WJ, Ma DD, Zhang JG, Lu ZJ, Long XB, Liu X, Huang CS, Ying GG. Effects of New Psychoactive Substance Esketamine on Behaviors and Transcription of Genes in Dopamine and GABA Pathways in Zebrafish Larvae. BULLETIN OF ENVIRONMENTAL CONTAMINATION AND TOXICOLOGY 2024; 112:51. [PMID: 38556558 DOI: 10.1007/s00128-024-03883-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 03/07/2024] [Indexed: 04/02/2024]
Abstract
Esketamine (ESK) is the S-enantiomer of ketamine racemate (a new psychoactive substance) that can result in illusions, and alter hearing, vision, and proprioception in human and mouse. Up to now, the neurotoxicity caused by ESK at environmental level in fish is still unclear. This work studied the effects of ESK on behaviors and transcriptions of genes in dopamine and GABA pathways in zebrafish larvae at ranging from 12.4 ng L- 1 to 11141.1 ng L- 1 for 7 days post fertilization (dpf). The results showed that ESK at 12.4 ng L- 1 significantly reduced the touch response of the larvae at 48 hpf. ESK at 12.4 ng L- 1 also reduced the time and distance of larvae swimming at the outer zone during light period, which implied that ESK might potentially decrease the anxiety level of larvae. In addition, ESK increased the transcription of th, ddc, drd1a, drd3 and drd4a in dopamine pathway. Similarly, ESK raised the transcription of slc6a1b, slc6a13 and slc12a2 in GABA pathway. This study suggested that ESK could affect the heart rate and behaviors accompanying with transcriptional alterations of genes in DA and GABA pathways at early-staged zebrafish, which resulted in neurotoxicity in zebrafish larvae.
Collapse
Affiliation(s)
- Si-Ying Li
- Guangdong Provincial Key Laboratory of Chemical Pollution and Environmental Safety & MOE Key Laboratory of Theoretical Chemistry of Environment, SCNU Environmental Research Institute, South China Normal University, Guangzhou, 510006, China
- School of Environment, South China Normal University, University Town, Guangzhou, 510006, China
| | - Wen-Jun Shi
- Guangdong Provincial Key Laboratory of Chemical Pollution and Environmental Safety & MOE Key Laboratory of Theoretical Chemistry of Environment, SCNU Environmental Research Institute, South China Normal University, Guangzhou, 510006, China.
- School of Environment, South China Normal University, University Town, Guangzhou, 510006, China.
| | - Dong-Dong Ma
- Guangdong Provincial Key Laboratory of Chemical Pollution and Environmental Safety & MOE Key Laboratory of Theoretical Chemistry of Environment, SCNU Environmental Research Institute, South China Normal University, Guangzhou, 510006, China
- School of Environment, South China Normal University, University Town, Guangzhou, 510006, China
| | - Jin-Ge Zhang
- Guangdong Provincial Key Laboratory of Chemical Pollution and Environmental Safety & MOE Key Laboratory of Theoretical Chemistry of Environment, SCNU Environmental Research Institute, South China Normal University, Guangzhou, 510006, China
- School of Environment, South China Normal University, University Town, Guangzhou, 510006, China
| | - Zhi-Jie Lu
- Guangdong Provincial Key Laboratory of Chemical Pollution and Environmental Safety & MOE Key Laboratory of Theoretical Chemistry of Environment, SCNU Environmental Research Institute, South China Normal University, Guangzhou, 510006, China
- School of Environment, South China Normal University, University Town, Guangzhou, 510006, China
| | - Xiao-Bing Long
- Guangdong Provincial Key Laboratory of Chemical Pollution and Environmental Safety & MOE Key Laboratory of Theoretical Chemistry of Environment, SCNU Environmental Research Institute, South China Normal University, Guangzhou, 510006, China
- School of Environment, South China Normal University, University Town, Guangzhou, 510006, China
| | - Xin Liu
- Anti-Drug Technology Center of Guangdong Province and National Anti-Drug Laboratory Guangdong Regional Center, Guangdong Provincial Key Laboratory of Psychoactive Substances Monitoring and Safety, Guangzhou, 510230, China
| | - Chu-Shu Huang
- Anti-Drug Technology Center of Guangdong Province and National Anti-Drug Laboratory Guangdong Regional Center, Guangdong Provincial Key Laboratory of Psychoactive Substances Monitoring and Safety, Guangzhou, 510230, China
| | - Guang-Guo Ying
- Guangdong Provincial Key Laboratory of Chemical Pollution and Environmental Safety & MOE Key Laboratory of Theoretical Chemistry of Environment, SCNU Environmental Research Institute, South China Normal University, Guangzhou, 510006, China.
- School of Environment, South China Normal University, University Town, Guangzhou, 510006, China.
| |
Collapse
|
11
|
Shahim B, Xu H, Haugaa K, Zetterberg H, Jurga J, Religa D, Eriksdotter M. Cholinesterase inhibitors are associated with reduced mortality in patients with Alzheimer's disease and previous myocardial infarction. EUROPEAN HEART JOURNAL. CARDIOVASCULAR PHARMACOTHERAPY 2024; 10:128-136. [PMID: 38224338 DOI: 10.1093/ehjcvp/pvad102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 11/13/2023] [Accepted: 12/18/2023] [Indexed: 01/16/2024]
Abstract
BACKGROUND Cholinesterase inhibitors (ChEIs) are the first-line symptomatic pharmacologic treatment for patients with mild-to-moderate Alzheimer's disease (AD). Although the target organ for this group of drugs is the brain, inhibition of the enzyme may affect cardiac function through vagotonic and anti-inflammatory effects. OBJECTIVE To assess the impact of ChEIs on outcomes in patients with AD who have experienced myocardial infarction (MI) prior to the AD diagnosis. METHODS Patients who had experienced MI before they were diagnosed with AD or Alzheimer's mixed dementia between 2008 and 2018 were identified from the Swedish Dementia Registry (SveDem, www.svedem.se), which was linked to the National Patient Registry to obtain data on MI and mortality. Cox proportional hazards regression model among a propensity score-matched dataset was performed to assess the association between ChEI treatment and clinical outcomes. RESULTS Of 3198 patients with previous MI and a diagnosis of AD or mixed dementia, 1705 (53%) were on treatment with ChEIs. Patients treated with ChEIs were more likely to be younger and have a better overall cardiovascular (CV) risk profile. The incidence rate of all-cause death (per 1000 patient-years) in the propensity-matched cohort of 1016 ChEI users and 1016 non-users was 168.6 in patients on treatment with ChEIs compared with 190.7 in patients not on treatment with ChEIs. In this propensity-matched cohort, treatment with ChEIs was associated with a significantly lower risk of all-cause death (adjusted hazard ratio 0.81, 95% confidence interval 0.71-0.92) and a greater reduction with higher doses of ChEIs. While in the unadjusted analysis, ChEIs were associated with a lower risk of both CV and non-CV death, only the association with non-CV death remained significant after accounting for baseline differences. CONCLUSION Treatment with ChEIs was associated with a significantly reduced risk of all-cause death, driven by lower rates of non-CV death in a nationwide cohort of patients with previous MI and a diagnosis of AD or mixed dementia. These associations were greater with higher ChEI doses. CONDENSED ABSTRACT We assessed the association between cholinesterase inhibitors (ChEIs) and clinical outcomes in a nationwide cohort of patients with previous myocardial infarction (MI) and a diagnosis of Alzheimer's disease (AD) or mixed dementi. In propensity-matched analysis, treatment with ChEIs was associated with a 19% reduction in all-cause death driven by non-cardiovascular death. The reduction in all-cause death was greater with the higher doses of ChEIs.
Collapse
Affiliation(s)
- Bahira Shahim
- Heart, Vascular and Neuro Theme, Karolinska University Hospital, 17177 Stockholm, Sweden
- Department of Medicine Solna, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Hong Xu
- Department of Neurobiology, Care Sciences and Society, Division of Clinical Geriatrics, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Kristina Haugaa
- Heart, Vascular and Neuro Theme, Karolinska University Hospital, 17177 Stockholm, Sweden
- Department of Medicine Solna, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Henrik Zetterberg
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, 41345 Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, 41345 Mölndal, Sweden
- UK Dementia Research Institute at UCL, London, UK
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong SAR, China
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Juliane Jurga
- Heart, Vascular and Neuro Theme, Karolinska University Hospital, 17177 Stockholm, Sweden
- Department of Medicine Solna, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Dorota Religa
- Department of Neurobiology, Care Sciences and Society, Division of Clinical Geriatrics, Karolinska Institutet, 17177 Stockholm, Sweden
- Theme Inflammation and Aging, Karolinska University Hospital, Huddinge, Sweden
| | - Maria Eriksdotter
- Department of Neurobiology, Care Sciences and Society, Division of Clinical Geriatrics, Karolinska Institutet, 17177 Stockholm, Sweden
- Theme Inflammation and Aging, Karolinska University Hospital, Huddinge, Sweden
| |
Collapse
|
12
|
Kang H, Kim J, Park CH, Jeong B, So I. Direct modulation of TRPC ion channels by Gα proteins. Front Physiol 2024; 15:1362987. [PMID: 38384797 PMCID: PMC10880550 DOI: 10.3389/fphys.2024.1362987] [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: 12/29/2023] [Accepted: 01/26/2024] [Indexed: 02/23/2024] Open
Abstract
GPCR-Gi protein pathways are involved in the regulation of vagus muscarinic pathway under physiological conditions and are closely associated with the regulation of internal visceral organs. The muscarinic receptor-operated cationic channel is important in GPCR-Gi protein signal transduction as it decreases heart rate and increases GI rhythm frequency. In the SA node of the heart, acetylcholine binds to the M2 receptor and the released Gβγ activates GIRK (I(K,ACh)) channel, inducing a negative chronotropic action. In gastric smooth muscle, there are two muscarinic acetylcholine receptor (mAChR) subtypes, M2 and M3. M2 receptor activates the muscarinic receptor-operated nonselective cationic current (mIcat, NSCC(ACh)) and induces positive chronotropic effect. Meanwhile, M3 receptor induces hydrolysis of PIP2 and releases DAG and IP3. This IP3 increases intracellular Ca2+ and then leads to contraction of GI smooth muscles. The activation of mIcat is inhibited by anti-Gi/o protein antibodies in GI smooth muscle, indicating the involvement of Gαi/o protein in the activation of mIcat. TRPC4 channel is a molecular candidate for mIcat and can be directly activated by constitutively active Gαi QL proteins. TRPC4 and TRPC5 belong to the same subfamily and both are activated by Gi/o proteins. Initial studies suggested that the binding sites for G protein exist at the rib helix or the CIRB domain of TRPC4/5 channels. However, recent cryo-EM structure showed that IYY58-60 amino acids at ARD of TRPC5 binds with Gi3 protein. Considering the expression of TRPC4/5 in the brain, the direct G protein activation on TRPC4/5 is important in terms of neurophysiology. TRPC4/5 channels are also suggested as a coincidence detector for Gi and Gq pathway as Gq pathway increases intracellular Ca2+ and the increased Ca2+ facilitates the activation of TRPC4/5 channels. More complicated situation would occur when GIRK, KCNQ2/3 (IM) and TRPC4/5 channels are co-activated by stimulation of muscarinic receptors at the acetylcholine-releasing nerve terminals. This review highlights the effects of GPCR-Gi protein pathway, including dopamine, μ-opioid, serotonin, glutamate, GABA, on various oragns, and it emphasizes the importance of considering TRPC4/5 channels as crucial players in the field of neuroscience.
Collapse
Affiliation(s)
- Hana Kang
- Department of Physiology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jinhyeong Kim
- Department of Physiology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Christine Haewon Park
- Department of Physiology, University of California, San Francisco, San Francisco, CA, United States
| | - Byeongseok Jeong
- Department of Physiology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Insuk So
- Department of Physiology, Seoul National University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
13
|
An X, Cho H. Increased GIRK channel activity prevents arrhythmia in mice with heart failure by enhancing ventricular repolarization. Sci Rep 2023; 13:22479. [PMID: 38110503 PMCID: PMC10728207 DOI: 10.1038/s41598-023-50088-2] [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: 06/26/2023] [Accepted: 12/15/2023] [Indexed: 12/20/2023] Open
Abstract
Ventricular arrhythmia causing sudden cardiac death is the leading mode of death in patients with heart failure. Yet, the mechanisms that prevent ventricular arrhythmias in heart failure are not well characterized. Using a mouse model of heart failure created by transverse aorta constriction, we show that GIRK channel, an important regulator of cardiac action potentials, is constitutively active in failing ventricles in contrast to normal cells. Evidence is presented indicating that the tonic activation of M2 muscarinic acetylcholine receptors by endogenously released acetylcholine contributes to the constitutive GIRK activity. This constitutive GIRK activity prevents the action potential prolongation in heart failure ventricles. Consistently, GIRK channel blockade with tertiapin-Q induces QT interval prolongation and increases the incidence of arrhythmia in heart failure, but not in control mice. These results suggest that constitutive GIRK channels comprise a key mechanism to protect against arrhythmia by providing repolarizing currents in heart failure ventricles.
Collapse
Affiliation(s)
- Xue An
- Department of Physiology, Sungkyunkwan University School of Medicine, Suwon, 16419, Korea
- Department of Critical Care Medicine, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Hana Cho
- Department of Physiology, Sungkyunkwan University School of Medicine, Suwon, 16419, Korea.
| |
Collapse
|
14
|
Nissen SD, Saljic A, Carstensen H, Braunstein TH, Hesselkilde EM, Kjeldsen ST, Hopster-Iversen C, D’Souza A, Jespersen T, Buhl R. Muscarinic acetylcholine receptors M 2 are upregulated in the atrioventricular nodal tract in horses with a high burden of second-degree atrioventricular block. Front Cardiovasc Med 2023; 10:1102164. [PMID: 38034369 PMCID: PMC10687567 DOI: 10.3389/fcvm.2023.1102164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 10/30/2023] [Indexed: 12/02/2023] Open
Abstract
Background Second-degree atrioventricular (AV) block at rest is very common in horses. The underlying molecular mechanisms are unexplored, but commonly attributed to high vagal tone. Aim To assess whether AV block in horses is due to altered expression of the effectors of vagal signalling in the AV node, with specific emphasis on the muscarinic acetylcholine receptor (M2) and the G protein-gated inwardly rectifying K+ (GIRK4) channel that mediates the cardiac IK,ACh current. Method Eighteen horses with a low burden of second-degree AV block (median 8 block per 20 h, IQR: 32 per 20 h) were assigned to the control group, while 17 horses with a high burden of second-degree AV block (median: 408 block per 20 h, IQR: 1,436 per 20 h) were assigned to the AV block group. Radiotelemetry ECG recordings were performed to assess PR interval and incidence of second-degree AV block episodes at baseline and on pharmacological blockade of the autonomic nervous system (ANS). Wenckebach cycle length was measured by intracardiac pacing (n = 16). Furthermore, the expression levels of the M2 receptor and the GIRK4 subunit of the IKACh channel were quantified in biopsies from the right atrium, the AV node and right ventricle using immunohistochemistry and machine learning-based automated segmentation analysis (n = 9 + 9). Results The AV block group had a significantly longer PR interval (mean ± SD, 0.40 ± 0.05 s; p < 0.001) and a longer Wenckebach cycle length (mean ± SD, 995 ± 86 ms; p = 0.007) at baseline. After blocking the ANS, all second-degree AV block episodes were abolished, and the difference in PR interval disappered (p = 0.80). The AV block group had significantly higher expression of the M2 receptor (p = 0.02), but not the GIRK4 (p = 0.25) in the AV node compared to the control group. Both M2 and GIRK4 were highly expressed in the AV node and less expressed in the atria and the ventricles. Conclusion Here, we demonstrate the involvement of the m2R-IK,ACh pathway in underlying second-degree AV block in horses. The high expression level of the M2 receptor may be responsible for the high burden of second-degree AV blocks seen in some horses.
Collapse
Affiliation(s)
- Sarah Dalgas Nissen
- Laboratory of Cardiac Physiology, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Arnela Saljic
- Laboratory of Cardiac Physiology, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Institute of Pharmacology, West German Heart and Vascular Centre, University Duisburg-Essen, Essen, Germany
| | - Helena Carstensen
- Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Taastrup, Denmark
| | - Thomas Hartig Braunstein
- Core Facility for Integrated Microscopy, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Eva Melis Hesselkilde
- Laboratory of Cardiac Physiology, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Sofie Troest Kjeldsen
- Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Taastrup, Denmark
| | - Charlotte Hopster-Iversen
- Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Taastrup, Denmark
| | - Alicia D’Souza
- Division of Cardiovascular Sciences, University of Manchester, 3.30 Core Technology, Manchester, United Kingdom
| | - Thomas Jespersen
- Laboratory of Cardiac Physiology, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Rikke Buhl
- Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Taastrup, Denmark
| |
Collapse
|
15
|
Huang F, Mariani N, Pariante CM, Borsini A. From dried bear bile to molecular investigation of differential effects of bile acids in ex vivo and in vitro models of myocardial dysfunction: Relevance for neuroinflammation. Brain Behav Immun Health 2023; 32:100674. [PMID: 37593199 PMCID: PMC10430170 DOI: 10.1016/j.bbih.2023.100674] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 07/27/2023] [Indexed: 08/19/2023] Open
Abstract
Bile acids have been known to have both beneficial and detrimental effects on heart function, and as a consequence this can affect the brain. Inflammation is a key factor linking the heart and the brain, bile acids can reduce inflammation in the heart and, as a consequence, neuroinflammation, which may be due to the activation of different peripheral and central cellular and molecular mechanisms. Herein, we compile data published so far and summarise evidence demonstrating the effects of bile acids on myocardial cell viability and function, and its related mechanisms, in ex vivo and in vitro studies conducted in homeostatic state or in models of cardiovascular diseases. Studies show that ursodeoxycholic acid (UDCA) and tauroursodeoxycholic acid (TUDCA) do not affect the viability or contraction of cardiomyocytes in homeostatic state, and while UDCA has the capability to prevent the effect of hypoxia on reduced cell viability and beating rate, TUDCA can protect endoplasmic reticulum (ER) stress-induced apoptosis and cardiac contractile dysfunction. In contrast, deoxycholic acid (DCA) decreases contraction rate in homeostatic state, but it also prevents hypoxia-induced inflammation and oxidative stress, whereas lithocholic acid (LCA) can rescue doxazosin-induced apoptosis. Moreover, glycodeoxycholic acid (GDCA), cholic acid (CA), chenodeoxycholic acid (CDCA), glycocholic acid (GCA), taurocholic acid (TCA), taurochenodeoxycholic acid (TCDCA) and taurodeoxycholic acid (TDCA) decrease contraction, whereas CDCA decreases cell viability in homeostatic conditions. The mechanisms underlying the aforementioned contrasting effects involve a differential regulation of the TGR5, M2R and FXR receptors, as well as the cAMP signalling pathway. Overall, this review confirms the therapeutic potential of certain types of bile acids: UDCA, TUDCA, and potentially LCA, in cardiovascular diseases. By reducing inflammation in the heart, bile acids can improve heart-brain communication and promote overall health. Additional investigations are required to better elucidate mechanisms of action and more personalized clinical therapeutic doses.
Collapse
Affiliation(s)
- Fei Huang
- Stress, Psychiatry and Immunology Laboratory, Department of Psychological Medicine, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, UK
- Shanghai Key Laboratory of Compound Chinese Medicines, Shanghai R&D Centre for Standardization of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, PR China
| | - Nicole Mariani
- Stress, Psychiatry and Immunology Laboratory, Department of Psychological Medicine, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, UK
| | - Carmine M. Pariante
- Stress, Psychiatry and Immunology Laboratory, Department of Psychological Medicine, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, UK
| | - Alessandra Borsini
- Stress, Psychiatry and Immunology Laboratory, Department of Psychological Medicine, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, UK
| |
Collapse
|
16
|
Hiremath K, Dodakallanavar J, Sampat GH, Patil VS, Harish DR, Chavan R, Hegde HV, Roy S. Three finger toxins of elapids: structure, function, clinical applications and its inhibitors. Mol Divers 2023:10.1007/s11030-023-10734-3. [PMID: 37749455 DOI: 10.1007/s11030-023-10734-3] [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: 06/13/2023] [Accepted: 09/18/2023] [Indexed: 09/27/2023]
Abstract
The WHO lists snakebite as a "neglected tropical disease". In tropical and subtropical areas, envenoming is an important public health issue. This review article describes the structure, function, chemical composition, natural inhibitors, and clinical applications of Elapids' Three Finger Toxins (3FTX) using scientific research data. The primary venomous substance belonging to Elapidae is 3FTX, that targets nAChR. Three parallel β-sheets combine to create 3FTX, which has four or five disulfide bonds. The three primary types of 3FTX are short-chain, long-chain, and nonconventional 3FTX. The functions of 3FTX depend on the specific toxin subtype and the target receptor or ion channel. The well-known effect of 3FTX is probably neurotoxicity because of the severe consequences of muscular paralysis and respiratory failure in snakebite victims. 3FTX have also been studied for their potential clinical applications. α-bungarotoxin has been used as a molecular probe to study the structure and function of nAChRs (Nicotinic Acetylcholine Receptors). Acid-sensing ion channel (ASIC) isoforms 1a and 1b are inhibited by Mambalgins, derived from Black mamba venom, which hinders their function and provide an analgesic effect. α- Cobra toxin is a neurotoxin purified from Chinese cobra (Naja atra) binds to nAChR at the neuronal junction and causes an analgesic effect for moderate to severe pain. Some of the plants and their compounds have been shown to inhibit the activity of 3FTX, and their mechanisms of action are discussed.
Collapse
Affiliation(s)
- Kashinath Hiremath
- ICMR-National Institute of Traditional Medicine, Belagavi, Karnataka, 590010, India
- KLE College of Pharmacy, Belagavi, KLE Academy of Higher Education and Research, Belagavi, Karnataka, 590010, India
| | - Jagadeesh Dodakallanavar
- ICMR-National Institute of Traditional Medicine, Belagavi, Karnataka, 590010, India
- KLE College of Pharmacy, Belagavi, KLE Academy of Higher Education and Research, Belagavi, Karnataka, 590010, India
| | - Ganesh H Sampat
- ICMR-National Institute of Traditional Medicine, Belagavi, Karnataka, 590010, India
- KLE College of Pharmacy, Belagavi, KLE Academy of Higher Education and Research, Belagavi, Karnataka, 590010, India
| | - Vishal S Patil
- ICMR-National Institute of Traditional Medicine, Belagavi, Karnataka, 590010, India
- KLE College of Pharmacy, Belagavi, KLE Academy of Higher Education and Research, Belagavi, Karnataka, 590010, India
| | - Darasaguppe R Harish
- ICMR-National Institute of Traditional Medicine, Belagavi, Karnataka, 590010, India.
| | - Rajashekar Chavan
- KLE College of Pharmacy, Belagavi, KLE Academy of Higher Education and Research, Belagavi, Karnataka, 590010, India.
| | - Harsha V Hegde
- ICMR-National Institute of Traditional Medicine, Belagavi, Karnataka, 590010, India
| | - Subarna Roy
- ICMR-National Institute of Traditional Medicine, Belagavi, Karnataka, 590010, India
| |
Collapse
|
17
|
Elia A, Fossati S. Autonomic nervous system and cardiac neuro-signaling pathway modulation in cardiovascular disorders and Alzheimer's disease. Front Physiol 2023; 14:1060666. [PMID: 36798942 PMCID: PMC9926972 DOI: 10.3389/fphys.2023.1060666] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 01/19/2023] [Indexed: 01/31/2023] Open
Abstract
The heart is a functional syncytium controlled by a delicate and sophisticated balance ensured by the tight coordination of its several cell subpopulations. Accordingly, cardiomyocytes together with the surrounding microenvironment participate in the heart tissue homeostasis. In the right atrium, the sinoatrial nodal cells regulate the cardiac impulse propagation through cardiomyocytes, thus ensuring the maintenance of the electric network in the heart tissue. Notably, the central nervous system (CNS) modulates the cardiac rhythm through the two limbs of the autonomic nervous system (ANS): the parasympathetic and sympathetic compartments. The autonomic nervous system exerts non-voluntary effects on different peripheral organs. The main neuromodulator of the Sympathetic Nervous System (SNS) is norepinephrine, while the principal neurotransmitter of the Parasympathetic Nervous System (PNS) is acetylcholine. Through these two main neurohormones, the ANS can gradually regulate cardiac, vascular, visceral, and glandular functions by turning on one of its two branches (adrenergic and/or cholinergic), which exert opposite effects on targeted organs. Besides these neuromodulators, the cardiac nervous system is ruled by specific neuropeptides (neurotrophic factors) that help to preserve innervation homeostasis through the myocardial layers (from epicardium to endocardium). Interestingly, the dysregulation of this neuro-signaling pathway may expose the cardiac tissue to severe disorders of different etiology and nature. Specifically, a maladaptive remodeling of the cardiac nervous system may culminate in a progressive loss of neurotrophins, thus leading to severe myocardial denervation, as observed in different cardiometabolic and neurodegenerative diseases (myocardial infarction, heart failure, Alzheimer's disease). This review analyzes the current knowledge on the pathophysiological processes involved in cardiac nervous system impairment from the perspectives of both cardiac disorders and a widely diffused and devastating neurodegenerative disorder, Alzheimer's disease, proposing a relationship between neurodegeneration, loss of neurotrophic factors, and cardiac nervous system impairment. This overview is conducive to a more comprehensive understanding of the process of cardiac neuro-signaling dysfunction, while bringing to light potential therapeutic scenarios to correct or delay the adverse cardiovascular remodeling, thus improving the cardiac prognosis and quality of life in patients with heart or neurodegenerative disorders.
Collapse
|
18
|
Refisch A, Komatsuzaki S, Ungelenk M, Chung HY, Schumann A, Schilling SS, Jantzen W, Schröder S, Mühleisen TW, Nöthen MM, Hübner CA, Bär KJ. Associations of common genetic risk variants of the muscarinic acetylcholine receptor M2 with cardiac autonomic dysfunction in patients with schizophrenia. World J Biol Psychiatry 2023; 24:1-11. [PMID: 35172679 DOI: 10.1080/15622975.2022.2043561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
OBJECTIVES Decreased vagal modulation, which has consistently been observed in schizophrenic patients, might contribute to increased cardiac mortality in schizophrenia. Previously, associations between CHRM2 (Cholinergic Receptor Muscarinic 2) and cardiac autonomic features have been reported. Here, we tested for possible associations between these polymorphisms and heart rate variability in patients with schizophrenia. METHODS A total of three single nucleotide polymorphisms (SNPs) in CHRM2 (rs73158705 A>G, rs8191992 T>A and rs2350782 T>C) that achieved significance (p < 5 * 10-8) in genome-wide association studies for cardiac autonomic features were genotyped in 88 drug-naïve patients, 61 patients receiving antipsychotic medication and 144 healthy controls. Genotypes were analysed for associations with parameters of heart rate variability and complexity, in each diagnostic group. RESULTS We observed a significantly altered heart rate variability in unmedicated patients with identified genetic risk status in rs73158705 A>G, rs8191992 T>A and rs2350782 T>C as compared to genotype non-risk status. In patients receiving antipsychotic medication and healthy controls, these associations were not observed. DISCUSSION We report novel candidate genetic associations with cardiac autonomic dysfunction in schizophrenia, but larger cohorts are required for replication.
Collapse
Affiliation(s)
- Alexander Refisch
- Department of Psychiatry and Psychotherapy, Jena University Hospital, Jena, Germany.,Department of Psychosomatic Medicine and Psychotherapy, Lab for Autonomic Neuroscience, Imaging and Cognition (LANIC)1, Jena University Hospital, Jena, Germany
| | - Shoko Komatsuzaki
- Institute of Human Genetics, Jena University Hospital, Jena, Germany
| | - Martin Ungelenk
- Institute of Human Genetics, Jena University Hospital, Jena, Germany
| | - Ha-Yeun Chung
- Department of Neurology, Section Translational Neuroimmunology, Jena University Hospital, Jena, Germany
| | - Andy Schumann
- Department of Psychosomatic Medicine and Psychotherapy, Lab for Autonomic Neuroscience, Imaging and Cognition (LANIC)1, Jena University Hospital, Jena, Germany
| | - Susann S Schilling
- Department of Psychosomatic Medicine and Psychotherapy, Lab for Autonomic Neuroscience, Imaging and Cognition (LANIC)1, Jena University Hospital, Jena, Germany
| | - Wibke Jantzen
- Department of Psychosomatic Medicine and Psychotherapy, Lab for Autonomic Neuroscience, Imaging and Cognition (LANIC)1, Jena University Hospital, Jena, Germany
| | - Sabine Schröder
- Department of Psychosomatic Medicine and Psychotherapy, Lab for Autonomic Neuroscience, Imaging and Cognition (LANIC)1, Jena University Hospital, Jena, Germany
| | - Thomas W Mühleisen
- Institute of Neuroscience and Medicine (INM-1), Research Center Juelich, Juelich, Germany.,Medical Faculty, Cécile and Oskar Vogt Institute of Brain Research, Heinrich Heine University Düsseldorf, Düsseldorf, Germany.,Department of Biomedicine, Human Genomics Research Group, University of Basel, Basel, Switzerland
| | - Markus M Nöthen
- Institute of Human Genetics, University of Bonn, Bonn, Germany.,Department of Genomics, Life and Brain Center, University of Bonn, Bonn, Germany
| | | | - Karl-Jürgen Bär
- Department of Psychosomatic Medicine and Psychotherapy, Lab for Autonomic Neuroscience, Imaging and Cognition (LANIC)1, Jena University Hospital, Jena, Germany
| |
Collapse
|
19
|
Carbone AM, Del Calvo G, Nagliya D, Sharma K, Lymperopoulos A. Autonomic Nervous System Regulation of Epicardial Adipose Tissue: Potential Roles for Regulator of G Protein Signaling-4. Curr Issues Mol Biol 2022; 44:6093-6103. [PMID: 36547076 PMCID: PMC9776453 DOI: 10.3390/cimb44120415] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 11/23/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022] Open
Abstract
The epicardial adipose tissue (EAT) or epicardial fat is a visceral fat depot in the heart that contains intrinsic adrenergic and cholinergic nerves, through which it interacts with the cardiac sympathetic (adrenergic) and parasympathetic (cholinergic) nervous systems. These EAT nerves represent a significant source of several adipokines and other bioactive molecules, including norepinephrine, epinephrine, and free fatty acids. The production of these molecules is biologically relevant for the heart, since abnormalities in EAT secretion are implicated in the development of pathological conditions, including coronary atherosclerosis, atrial fibrillation, and heart failure. Sympathetic hyperactivity and parasympathetic (cholinergic) derangement are associated with EAT dysfunction, leading to a variety of adverse cardiac conditions, such as heart failure, diastolic dysfunction, atrial fibrillation, etc.; therefore, several studies have focused on exploring the autonomic regulation of EAT as it pertains to heart disease pathogenesis and progression. In addition, Regulator of G protein Signaling (RGS)-4 is a protein with significant regulatory roles in both adrenergic and muscarinic receptor signaling in the heart. In this review, we provide an overview of the autonomic regulation of EAT, with a specific focus on cardiac RGS4 and the potential roles this protein plays in this regulation.
Collapse
|
20
|
Moen JM, Morrell CH, Matt MG, Ahmet I, Tagirova S, Davoodi M, Petr M, Charles S, de Cabo R, Yaniv Y, Lakatta EG. Emergence of heartbeat frailty in advanced age I: perspectives from life-long EKG recordings in adult mice. GeroScience 2022; 44:2801-2830. [PMID: 35759167 PMCID: PMC9768068 DOI: 10.1007/s11357-022-00605-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 06/06/2022] [Indexed: 01/07/2023] Open
Abstract
The combined influences of sinoatrial nodal (SAN) pacemaker cell automaticity and its response to autonomic input determine the heart's beating interval variability and mean beating rate. To determine the intrinsic SAN and autonomic signatures buried within EKG RR interval time series change in advanced age, we measured RR interval variability before and during double autonomic blockade at 3-month intervals from 6 months of age until the end of life in long-lived (those that achieved the total cohort median life span of 24 months and beyond) C57/BL6 mice. Prior to 21 months of age, time-dependent changes in intrinsic RR interval variability and mean RR interval were relatively minor. Between 21 and 30 months of age, however, marked changes emerged in intrinsic SAN RR interval variability signatures, pointing to a reduction in the kinetics of pacemaker clock mechanisms, leading to reduced synchronization of molecular functions within and among SAN cells. This loss of high-frequency signal processing within intrinsic SAN signatures resulted in a marked increase in the mean intrinsic RR interval. The impact of autonomic signatures on RR interval variability were net sympathetic and partially compensated for the reduced kinetics of the intrinsic SAN RR interval variability signatures, and partially, but not completely, shifted the EKG RR time series intervals to a more youthful pattern. Cross-sectional analyses of other subsets of C57/BL6 ages indicated that at or beyond the median life span of our longitudinal cohort, noncardiac, constitutional, whole-body frailty was increased, energetic efficiency was reduced, and the respiratory exchange ratio increased. We interpret the progressive reduction in kinetics in intrinsic SAN RR interval variability signatures in this context of whole-body frailty beyond 21 months of age to be a manifestation of "heartbeat frailty."
Collapse
Affiliation(s)
- Jack M Moen
- Laboratory of Cardiovascular Science, National Institute On Aging, National Institutes of Health, Baltimore, MD, USA
- Department of Cellular and Molecular Pharmacology, UCSF, San Francisco, CA, USA
| | - Christopher H Morrell
- Laboratory of Cardiovascular Science, National Institute On Aging, National Institutes of Health, Baltimore, MD, USA
| | - Michael G Matt
- Laboratory of Cardiovascular Science, National Institute On Aging, National Institutes of Health, Baltimore, MD, USA
- Pediatric Residency Program, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Ismayil Ahmet
- Laboratory of Cardiovascular Science, National Institute On Aging, National Institutes of Health, Baltimore, MD, USA
| | - Syevda Tagirova
- Laboratory of Cardiovascular Science, National Institute On Aging, National Institutes of Health, Baltimore, MD, USA
| | - Moran Davoodi
- Biomedical Engineering Faculty, Technion-IIT, Haifa, Israel
| | - Michael Petr
- Laboratory of Experimental Gerontology Intramural Research Program, National Institute On Aging, National Institutes of Health, Baltimore, MD, USA
- Center for Healthy Aging, University of Copenhagen, Copenhagen, Denmark
| | - Shaquille Charles
- Laboratory of Cardiovascular Science, National Institute On Aging, National Institutes of Health, Baltimore, MD, USA
| | - Rafael de Cabo
- Laboratory of Experimental Gerontology Intramural Research Program, National Institute On Aging, National Institutes of Health, Baltimore, MD, USA
| | - Yael Yaniv
- Biomedical Engineering Faculty, Technion-IIT, Haifa, Israel
| | - Edward G Lakatta
- Laboratory of Cardiovascular Science, National Institute On Aging, National Institutes of Health, Baltimore, MD, USA.
| |
Collapse
|
21
|
Verkerk AO, Doszpod IJ, Mengarelli I, Magyar T, Polyák A, Pászti B, Efimov IR, Wilders R, Koncz I. Acetylcholine Reduces L-Type Calcium Current without Major Changes in Repolarization of Canine and Human Purkinje and Ventricular Tissue. Biomedicines 2022; 10:biomedicines10112987. [PMID: 36428555 PMCID: PMC9687254 DOI: 10.3390/biomedicines10112987] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/11/2022] [Accepted: 11/17/2022] [Indexed: 11/24/2022] Open
Abstract
Vagal nerve stimulation (VNS) holds a strong basis as a potentially effective treatment modality for chronic heart failure, which explains why a multicenter VNS study in heart failure with reduced ejection fraction is ongoing. However, more detailed information is required on the effect of acetylcholine (ACh) on repolarization in Purkinje and ventricular cardiac preparations to identify the advantages, risks, and underlying cellular mechanisms of VNS. Here, we studied the effect of ACh on the action potential (AP) of canine Purkinje fibers (PFs) and several human ventricular preparations. In addition, we characterized the effects of ACh on the L-type Ca2+ current (ICaL) and AP of human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) and performed computer simulations to explain the observed effects. Using microelectrode recordings, we found a small but significant AP prolongation in canine PFs. In the human myocardium, ACh slightly prolonged the AP in the midmyocardium but resulted in minor AP shortening in subepicardial tissue. Perforated patch-clamp experiments on hiPSC-CMs demonstrated that 5 µM ACh caused an ≈15% decrease in ICaL density without changes in gating properties. Using dynamic clamp, we found that under blocked K+ currents, 5 µM ACh resulted in an ≈23% decrease in AP duration at 90% of repolarization in hiPSC-CMs. Computer simulations using the O'Hara-Rudy human ventricular cell model revealed that the overall effect of ACh on AP duration is a tight interplay between the ACh-induced reduction in ICaL and ACh-induced changes in K+ currents. In conclusion, ACh results in minor changes in AP repolarization and duration of canine PFs and human ventricular myocardium due to the concomitant inhibition of inward ICaL and outward K+ currents, which limits changes in net repolarizing current and thus prevents major changes in AP repolarization.
Collapse
Affiliation(s)
- Arie O. Verkerk
- Department of Experimental Cardiology, Heart Center, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Illés J. Doszpod
- Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, 6721 Szeged, Hungary
| | - Isabella Mengarelli
- Department of Experimental Cardiology, Heart Center, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Tibor Magyar
- Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, 6721 Szeged, Hungary
| | - Alexandra Polyák
- Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, 6721 Szeged, Hungary
| | - Bence Pászti
- Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, 6721 Szeged, Hungary
| | - Igor R. Efimov
- Department of Biomedical Engineering, The George Washington University, Washington, DC 20052, USA
- Department of Biomedical Engineering, Northwestern University, Chicago, IL 60611, USA
- Department of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Ronald Wilders
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
- Correspondence: (R.W.); (I.K.)
| | - István Koncz
- Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, 6721 Szeged, Hungary
- Department of Biomedical Engineering, The George Washington University, Washington, DC 20052, USA
- Correspondence: (R.W.); (I.K.)
| |
Collapse
|
22
|
Yeruva S, Körber L, Hiermaier M, Egu DT, Kempf E, Waschke J. Cholinergic signaling impairs cardiomyocyte cohesion. Acta Physiol (Oxf) 2022; 236:e13881. [PMID: 36039679 DOI: 10.1111/apha.13881] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 08/23/2022] [Accepted: 08/24/2022] [Indexed: 01/29/2023]
Abstract
AIM Cardiac autonomic nervous system (ANS) dysregulation is a hallmark of several cardiovascular diseases. Adrenergic signaling enhanced cardiomyocyte cohesion via PKA-mediated plakoglobin phosphorylation at serine 665, referred to as positive adhesiotropy. This study investigated cholinergic regulation of cardiomyocyte cohesion using muscarinic receptor agonist carbachol (CCH). METHODS Dissociation assays, Western blot analysis, immunostaining, atomic force microscopy (AFM), immunoprecipitation, transmission electron microscopy (TEM), triton assays, and siRNA knockdown of genes were performed in either HL-1 cells or plakoglobin (PG) wild type (Jup+/+ ) and knockout (Jup-/- ) mice, which served as a model for arrhythmogenic cardiomyopathy. RESULTS In HL-1 cells grown in norepinephrine (NE)-containing medium for baseline adrenergic stimulation, and murine cardiac slice cultures from Jup+/+ and Jup-/- mice CCH treatment impaired cardiomyocyte cohesion. Immunostainings and AFM experiments revealed that CCH reduced desmoglein 2 (DSG2) localization and binding at cell borders. Furthermore, CCH reduced intercalated disc plaque thickness in both Jup+/+ and Jup-/- mice, evidenced by TEM analysis. Immunoprecipitation experiments in HL-1 cells revealed no changes in DSG2 interaction with desmoplakin (DP), plakophilin 2 (PKP2), PG, and desmin (DES) after CCH treatment. However, knockdown of any of the above proteins abolished CCH-mediated loss of cardiomyocyte cohesion. Furthermore, in HL-1 cells, CCH inhibited adrenergic-stimulated ERK phosphorylation but not PG phosphorylation at serine 665. In addition, CCH activated the AKT/GSK-3β axis in the presence of NE. CONCLUSION Our results demonstrate that cholinergic signaling antagonizes the positive effect of adrenergic signaling on cardiomyocyte cohesion and thus causes negative adhesiotropy independent of PG phosphorylation.
Collapse
Affiliation(s)
- Sunil Yeruva
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilian-University (LMU) Munich, Munich, Germany
| | - Lars Körber
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilian-University (LMU) Munich, Munich, Germany
| | - Matthias Hiermaier
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilian-University (LMU) Munich, Munich, Germany
| | - Desalegn T Egu
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilian-University (LMU) Munich, Munich, Germany
| | - Ellen Kempf
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilian-University (LMU) Munich, Munich, Germany
| | - Jens Waschke
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilian-University (LMU) Munich, Munich, Germany
| |
Collapse
|
23
|
Myslivecek J. Multitargeting nature of muscarinic orthosteric agonists and antagonists. Front Physiol 2022; 13:974160. [PMID: 36148314 PMCID: PMC9486310 DOI: 10.3389/fphys.2022.974160] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 08/01/2022] [Indexed: 11/16/2022] Open
Abstract
Muscarinic receptors (mAChRs) are typical members of the G protein-coupled receptor (GPCR) family and exist in five subtypes from M1 to M5. Muscarinic receptor subtypes do not sufficiently differ in affinity to orthosteric antagonists or agonists; therefore, the analysis of receptor subtypes is complicated, and misinterpretations can occur. Usually, when researchers mainly specialized in CNS and peripheral functions aim to study mAChR involvement in behavior, learning, spinal locomotor networks, biological rhythms, cardiovascular physiology, bronchoconstriction, gastrointestinal tract functions, schizophrenia, and Parkinson's disease, they use orthosteric ligands and they do not use allosteric ligands. Moreover, they usually rely on manufacturers' claims that could be misleading. This review aimed to call the attention of researchers not deeply focused on mAChR pharmacology to this fact. Importantly, limited selective binding is not only a property of mAChRs but is a general attribute of most neurotransmitter receptors. In this review, we want to give an overview of the most common off-targets for established mAChR ligands. In this context, an important point is a mention the tremendous knowledge gap on off-targets for novel compounds compared to very well-established ligands. Therefore, we will summarize reported affinities and give an outline of strategies to investigate the subtype's function, thereby avoiding ambiguous results. Despite that, the multitargeting nature of drugs acting also on mAChR could be an advantage when treating such diseases as schizophrenia. Antipsychotics are a perfect example of a multitargeting advantage in treatment. A promising strategy is the use of allosteric ligands, although some of these ligands have also been shown to exhibit limited selectivity. Another new direction in the development of muscarinic selective ligands is functionally selective and biased agonists. The possible selective ligands, usually allosteric, will also be listed. To overcome the limited selectivity of orthosteric ligands, the recommended process is to carefully examine the presence of respective subtypes in specific tissues via knockout studies, carefully apply "specific" agonists/antagonists at appropriate concentrations and then calculate the probability of a specific subtype involvement in specific functions. This could help interested researchers aiming to study the central nervous system functions mediated by the muscarinic receptor.
Collapse
Affiliation(s)
- Jaromir Myslivecek
- Institute of Physiology, 1 Faculty of Medicine, Charles University, Prague, Czechia
| |
Collapse
|
24
|
Prathumsap N, Ongnok B, Khuanjing T, Arinno A, Maneechote C, Apaijai N, Chunchai T, Arunsak B, Shinlapawittayatorn K, Chattipakorn SC, Chattipakorn N. Acetylcholine receptor agonists provide cardioprotection in doxorubicin-induced cardiotoxicity via modulating muscarinic M 2 and α7 nicotinic receptor expression. Transl Res 2022; 243:33-51. [PMID: 34920165 DOI: 10.1016/j.trsl.2021.12.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 11/17/2021] [Accepted: 12/13/2021] [Indexed: 10/19/2022]
Abstract
The balance between cardiac sympathetic and parasympathetic activities has been intricately linked to mitochondrial function, cellular oxidative status, and immunomodulation in healthy and diseased myocardium. Cardiac autonomic neuropathy, along with the associated mitochondrial and cellular dysfunction, is an important pathophysiological feature of doxorubicin-induced cardiotoxicity (DIC). We tested the hypothesis that autonomic modulation by activation of acetylcholine receptors (AChR) effectively attenuates DIC. Rats were divided into control (0.9% sodium chloride solution) and doxorubicin groups (DOX, 3 mg/kg/d, 6 doses). Rats in the DOX group were equally subdivided into 4 interventional groups and treated for 30 days: vehicle, α7 nicotinic receptor agonist (PNU: PNU-282987, 3 mg/kg/d), muscarinic receptor agonist (BET: bethanechol, 12 mg/kg/d), and combined α7nAChR and mAChR agonists group (COM). Cardiac biochemical and functional analyses were done. The results show that AChR agonists protected the heart against DIC via improving mitochondrial and cardiac function, which was accompanied by reducing mitochondrial oxidative damage, apoptosis, and inflammation. Strikingly, PNU and BET exerted cardioprotection through different molecular pathways. PNU-mediated α7nAChR activation promoted mitochondrial fusion via upregulation of Mfn1-2 and attenuated DOX-induced autophagy. Contrarily, activation of mAChR by BET attenuated mitochondrial fission and mitophagy. The in vitro experiments confirmed the cytoprotective effects of AChR activation in DOX-treated H9c2 cells without compromising the anticancer effect of DOX in cancer cells. In conclusion, α7nAChR and mAChR agonists exerted cardioprotection against DIC via rebalancing autonomic function, improving mitochondrial function, reducing oxidative stress, and decreased cardiomyocyte apoptosis and inflammation, leading to improved cardiac function.
Collapse
Affiliation(s)
- Nanthip Prathumsap
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Center of Excellence in Cardiac Electrophysiology, Chiang Mai University, Chiang Mai, Thailand
| | - Benjamin Ongnok
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Center of Excellence in Cardiac Electrophysiology, Chiang Mai University, Chiang Mai, Thailand
| | - Thawatchai Khuanjing
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Center of Excellence in Cardiac Electrophysiology, Chiang Mai University, Chiang Mai, Thailand
| | - Apiwan Arinno
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Center of Excellence in Cardiac Electrophysiology, Chiang Mai University, Chiang Mai, Thailand
| | - Chayodom Maneechote
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Center of Excellence in Cardiac Electrophysiology, Chiang Mai University, Chiang Mai, Thailand
| | - Nattayaporn Apaijai
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Center of Excellence in Cardiac Electrophysiology, Chiang Mai University, Chiang Mai, Thailand
| | - Titikorn Chunchai
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Center of Excellence in Cardiac Electrophysiology, Chiang Mai University, Chiang Mai, Thailand
| | - Busarin Arunsak
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Center of Excellence in Cardiac Electrophysiology, Chiang Mai University, Chiang Mai, Thailand
| | - Krekwit Shinlapawittayatorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Center of Excellence in Cardiac Electrophysiology, Chiang Mai University, Chiang Mai, Thailand
| | - Siriporn C Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Center of Excellence in Cardiac Electrophysiology, Chiang Mai University, Chiang Mai, Thailand; Department of Oral Biology and Diagnostic Science, Faculty of Dentistry, Chiang Mai University, Chiang Mai, Thailand
| | - Nipon Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Center of Excellence in Cardiac Electrophysiology, Chiang Mai University, Chiang Mai, Thailand.
| |
Collapse
|
25
|
Muscarinic receptor activation reduces force and arrhythmias in human atria independent of IK,ACh. J Cardiovasc Pharmacol 2022; 79:678-686. [PMID: 35170489 DOI: 10.1097/fjc.0000000000001237] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 01/15/2022] [Indexed: 11/27/2022]
Abstract
ABSTRACT In human hearts, muscarinic receptors (M-R) are expressed in ventricular and atrial tissue, but the acetylcholine-activated potassium current (IK,ACh) is expressed mainly in the atrium. M-R activation decreases force and increases electrical stability in human atrium, but the impact of IK,ACh to both effects remains unclear. We employed a new selective blocker of IK,ACh to elaborate the contribution of IK,ACh to M-R activation-mediated effects in human atrium.Force and action potentials were measured in rat atria and in human right atrial trabeculae. Cumulative concentration-effect curves for norepinephrine-induced force and arrhythmias were measured in the presence of either carbachol (CCh;1µM) or CCh together with the IK,ACh -blocker XAF-1407 (1 µM) or in time-matched controls. To investigate the vulnerability to arrhythmias we performed some experiments also in the presence of cilostamide (0.3µM) and rolipram (1µM), inhibiting PDE3 and PDE4.In rat atria and human right atrial trabeculae, CCh shortened the action potential duration persistently. However, the direct negative inotropy of CCh was only transient in human, but stable in rat atria. In both rat and human atria, the negative inotropic effect was insensitive to blockage of IK,ACh by XAF-1407. In the presence of cilostamide and rolipram about 40% of trabeculae developed arrhythmias when exposed to norepinephrine. CCh prevented these concentration-dependent norepinephrine-induced arrhythmias, again insensitive to XAF-1407. Maximum catecholamine-induced force was not depressed by CCh.In human atrium, both the direct and the indirect negative inotropic effect of CCh are independent of IK,ACh. The same applies to the CCh-mediated suppression of norepinephrine/PDE-inhibition-induced arrhythmias.
Collapse
|
26
|
Koncz I, Verkerk AO, Nicastro M, Wilders R, Árpádffy-Lovas T, Magyar T, Tóth N, Nagy N, Madrid M, Lin Z, Efimov IR. Acetylcholine Reduces IKr and Prolongs Action Potentials in Human Ventricular Cardiomyocytes. Biomedicines 2022; 10:biomedicines10020244. [PMID: 35203454 PMCID: PMC8869322 DOI: 10.3390/biomedicines10020244] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 01/14/2022] [Accepted: 01/19/2022] [Indexed: 02/07/2023] Open
Abstract
Vagal nerve stimulation (VNS) has a meaningful basis as a potentially effective treatment for heart failure with reduced ejection fraction. There is an ongoing VNS randomized study, and four studies are completed. However, relatively little is known about the effect of acetylcholine (ACh) on repolarization in human ventricular cardiomyocytes, as well as the effect of ACh on the rapid component of the delayed rectifier K+ current (IKr). Here, we investigated the effect of ACh on the action potential parameters in human ventricular preparations and on IKr in human induced pluripotent stem-cell-derived cardiomyocytes (hiPSC-CMs). Using standard microelectrode technique, we demonstrated that ACh (5 µM) significantly increased the action potential duration in human left ventricular myocardial slices. ACh (5 µM) also prolonged repolarization in a human Purkinje fiber and a papillary muscle. Optical mapping revealed that ACh increased the action potential duration in human left ventricular myocardial slices and that the effect was dose-dependent. Perforated patch clamp experiments demonstrated action potential prolongation and a significant decrease in IKr by ACh (5 µM) in hiPSC-CMs. Computer simulations of the electrical activity of a human ventricular cardiomyocyte showed an increase in action potential duration upon implementation of the experimentally observed ACh-induced changes in the fully activated conductance and steady-state activation of IKr. Our findings support the hypothesis that ACh can influence the repolarization in human ventricular cardiomyocytes by at least changes in IKr.
Collapse
Affiliation(s)
- István Koncz
- Department of Biomedical Engineering, The George Washington University, Washington, DC 20052, USA; (I.K.); (M.M.); (Z.L.)
- Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, 6721 Szeged, Hungary; (T.Á.-L.); (T.M.); (N.T.); (N.N.)
| | - Arie O. Verkerk
- Heart Center, Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (A.O.V.); (M.N.)
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands;
| | - Michele Nicastro
- Heart Center, Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (A.O.V.); (M.N.)
| | - Ronald Wilders
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands;
| | - Tamás Árpádffy-Lovas
- Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, 6721 Szeged, Hungary; (T.Á.-L.); (T.M.); (N.T.); (N.N.)
| | - Tibor Magyar
- Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, 6721 Szeged, Hungary; (T.Á.-L.); (T.M.); (N.T.); (N.N.)
| | - Noémi Tóth
- Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, 6721 Szeged, Hungary; (T.Á.-L.); (T.M.); (N.T.); (N.N.)
| | - Norbert Nagy
- Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, 6721 Szeged, Hungary; (T.Á.-L.); (T.M.); (N.T.); (N.N.)
- ELKH-SZTE Research Group of Cardiovascular Pharmacology, 6721 Szeged, Hungary
| | - Micah Madrid
- Department of Biomedical Engineering, The George Washington University, Washington, DC 20052, USA; (I.K.); (M.M.); (Z.L.)
| | - Zexu Lin
- Department of Biomedical Engineering, The George Washington University, Washington, DC 20052, USA; (I.K.); (M.M.); (Z.L.)
| | - Igor R. Efimov
- Department of Biomedical Engineering, The George Washington University, Washington, DC 20052, USA; (I.K.); (M.M.); (Z.L.)
- Querrey Simpson Institute for Bioelectronics, Northwestern University, Chicago, IL 60611, USA
- Correspondence: ; Tel.: +1-202-294-8182
| |
Collapse
|
27
|
López-Serrano AL, Zamora-Cárdenas R, Aréchiga-Figueroa IA, Salazar-Fajardo PD, Ferrer T, Alamilla J, Sánchez-Chapula JA, Navarro-Polanco RA, Moreno-Galindo EG. Differential voltage-dependent modulation of the ACh-gated K+ current by adenosine and acetylcholine. PLoS One 2022; 17:e0261960. [PMID: 35030226 PMCID: PMC8759768 DOI: 10.1371/journal.pone.0261960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 12/14/2021] [Indexed: 11/18/2022] Open
Abstract
Inhibitory regulation of the heart is determined by both cholinergic M2 receptors (M2R) and adenosine A1 receptors (A1R) that activate the same signaling pathway, the ACh-gated inward rectifier K+ (KACh) channels via Gi/o proteins. Previously, we have shown that the agonist-specific voltage sensitivity of M2R underlies several voltage-dependent features of IKACh, including the ‘relaxation’ property, which is characterized by a gradual increase or decrease of the current when cardiomyocytes are stepped to hyperpolarized or depolarized voltages, respectively. However, it is unknown whether membrane potential also affects A1R and how this could impact IKACh. Upon recording whole-cell currents of guinea-pig cardiomyocytes, we found that stimulation of the A1R-Gi/o-IKACh pathway with adenosine only caused a very slight voltage dependence in concentration-response relationships (~1.2-fold EC50 increase with depolarization) that was not manifested in the relative affinity, as estimated by the current deactivation kinetics (τ = 4074 ± 214 ms at -100 mV and τ = 4331 ± 341 ms at +30 mV; P = 0.31). Moreover, IKACh did not exhibit relaxation. Contrarily, activation of the M2R-Gi/o-IKACh pathway with acetylcholine induced the typical relaxation of the current, which correlated with the clear voltage-dependent effect observed in the concentration-response curves (~2.8-fold EC50 increase with depolarization) and in the IKACh deactivation kinetics (τ = 1762 ± 119 ms at -100 mV and τ = 1503 ± 160 ms at +30 mV; P = 0.01). Our findings further substantiate the hypothesis of the agonist-specific voltage dependence of GPCRs and that the IKACh relaxation is consequence of this property.
Collapse
Affiliation(s)
- Ana Laura López-Serrano
- Centro Universitario de Investigaciones Biomédicas, Universidad de Colima, Colima, Col., Mexico
| | - Rodrigo Zamora-Cárdenas
- Centro Universitario de Investigaciones Biomédicas, Universidad de Colima, Colima, Col., Mexico
| | - Iván A. Aréchiga-Figueroa
- CONACYT, Facultad de Medicina, Universidad Autónoma de San Luis Potosí, San Luis Potosí, S.L.P., Mexico
| | | | - Tania Ferrer
- Centro Universitario de Investigaciones Biomédicas, Universidad de Colima, Colima, Col., Mexico
| | - Javier Alamilla
- CONACYT, Centro Universitario de Investigaciones Biomédicas, Universidad de Colima, Colima, Col., Mexico
| | - José A. Sánchez-Chapula
- Centro Universitario de Investigaciones Biomédicas, Universidad de Colima, Colima, Col., Mexico
| | | | - Eloy G. Moreno-Galindo
- Centro Universitario de Investigaciones Biomédicas, Universidad de Colima, Colima, Col., Mexico
- * E-mail:
| |
Collapse
|
28
|
Dias P, Pourová J, Vopršalová M, Nejmanová I, Mladěnka P. 3-Hydroxyphenylacetic Acid: A Blood Pressure-Reducing Flavonoid Metabolite. Nutrients 2022; 14:nu14020328. [PMID: 35057508 PMCID: PMC8781193 DOI: 10.3390/nu14020328] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/06/2022] [Accepted: 01/11/2022] [Indexed: 12/11/2022] Open
Abstract
Regular intake of polyphenol-rich food has been associated with a wide variety of beneficial health effects, including the prevention of cardiovascular diseases. However, the parent flavonoids have mostly low bioavailability and, hence, their metabolites have been hypothesized to be bioactive. One of these metabolites, 3-hydroxyphenylacetic acid (3-HPAA), formed by the gut microbiota, was previously reported to exert vasorelaxant effects ex vivo. The aim of this study was to shed more light on this effect in vivo, and to elucidate the mechanism of action. 3-HPAA gave rise to a dose-dependent decrease in arterial blood pressure when administered i.v. both as a bolus and infusion to spontaneously hypertensive rats. In contrast, no significant changes in heart rate were observed. In ex vivo experiments, where porcine hearts from a slaughterhouse were used to decrease the need for laboratory animals, 3-HPAA relaxed precontracted porcine coronary artery segments via a mechanism partially dependent on endothelium integrity. This relaxation was significantly impaired after endothelial nitric oxide synthase inhibition. In contrast, the blockade of SKCa or IKCa channels, or muscarinic receptors, did not affect 3-HPAA relaxation. Similarly, no effects of 3-HPAA on cyclooxygenase nor L-type calcium channels were observed. Thus, 3-HPAA decreases blood pressure in vivo via vessel relaxation, and this mechanism might be based on the release of nitric oxide by the endothelial layer.
Collapse
Affiliation(s)
- Patrícia Dias
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Charles University, Akademika Heyrovskeho 1203, 500 05 Hradec Kralove, Czech Republic; (P.D.); (M.V.); (P.M.)
| | - Jana Pourová
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Charles University, Akademika Heyrovskeho 1203, 500 05 Hradec Kralove, Czech Republic; (P.D.); (M.V.); (P.M.)
- Correspondence: ; Tel.: +420-495-067-295; Fax: +420-495-067-170
| | - Marie Vopršalová
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Charles University, Akademika Heyrovskeho 1203, 500 05 Hradec Kralove, Czech Republic; (P.D.); (M.V.); (P.M.)
| | - Iveta Nejmanová
- Department of Biological and Medical Sciences, Faculty of Pharmacy, Charles University, Akademika Heyrovskeho 1203, 500 05 Hradec Kralove, Czech Republic;
| | - Přemysl Mladěnka
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Charles University, Akademika Heyrovskeho 1203, 500 05 Hradec Kralove, Czech Republic; (P.D.); (M.V.); (P.M.)
| |
Collapse
|
29
|
Tapilina SV, Ivanova AD, Filatova TS, Galenko-Yaroshevsky PA, Abramochkin DV. The role of M3 receptors in regulation of electrical activity deteriorates in the rat heart during ageing. Curr Res Physiol 2022; 5:1-7. [PMID: 34977599 PMCID: PMC8685909 DOI: 10.1016/j.crphys.2021.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 11/25/2021] [Accepted: 12/02/2021] [Indexed: 11/29/2022] Open
Abstract
Ageing is a complex process which affects all systems of the organism and therefore changes the environment where the heart is working. In this study we demonstrate the ageing-related changes in the mechanisms of parasympathetic regulation of mammalian heart. Electrophysiological effects produced by selective activation of M3-cholinoreceptors were compared in isolated cardiac preparations from young adult (4 months), adult (1 year) and ageing (2 years) rats using sharp glass microelectrode technique. M3-receptors were activated with muscarinic agonist pilocarpine (10-5M) in the presence of selective M2 antagonist AQ-RA741 (10-7M). In atrial and ventricular myocardium from young rats M3 stimulation induced shortening of action potentials(APs), while no significant effect was observed in both elder groups. The main mechanism of M3-induced AP shortening is inhibition of L-type Ca2+ current, estimated using whole-cell patch-clamp. It was negligible in atrial myocytes from ageing animals in comparison with young rats. The loss of sensitivity to stimulation of M3-receptors is due to decrease in M3 gene expression, shown by RT-PCR both in atrial and ventricular samples from ageing rats. Thus, in ageing rat heart M3-receptors are down-regulated and not involved in regulation of electrical activity. Stimulation of M3-receptors shortens action potentials (APs) in rat myocardium. This effect of M3-stimulation is diminished in 1 and 2-year old rats. Underlying M3-mediated inhibition of L-type Ca2+ current deteriorates in aged rats. These age-related changes are due to downregulation of M3 receptors.
Collapse
Affiliation(s)
- Svetlana V Tapilina
- Department of Human and Animal Physiology, Moscow State University, Leninskiye Gory 1, 12, Moscow, Russia
| | - Alexandra D Ivanova
- Department of Human and Animal Physiology, Moscow State University, Leninskiye Gory 1, 12, Moscow, Russia
| | - Tatiana S Filatova
- Department of Human and Animal Physiology, Moscow State University, Leninskiye Gory 1, 12, Moscow, Russia.,Department of Physiology, Pirogov Russian National Research Medical University, Ostrovityanova Str 1, Moscow, Russia.,Laboratory of Cardiac Electrophysiology, National Medical Research Center for Cardiology, Moscow, Russia
| | | | - Denis V Abramochkin
- Department of Human and Animal Physiology, Moscow State University, Leninskiye Gory 1, 12, Moscow, Russia
| |
Collapse
|
30
|
Ehlen JC, Forman CM, Ostrowski D, Ostrowski TD. Autonomic Dysfunction Impairs Baroreflex Function in an Alzheimer's Disease Animal Model. J Alzheimers Dis 2022; 90:1449-1464. [PMID: 36278348 PMCID: PMC9742304 DOI: 10.3233/jad-220496] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Alzheimer's disease (AD) patients frequently present with orthostatic hypotension. This inability to reflexively increase blood pressure on standing is a serious health concern and increases the risk of stroke and cardiovascular diseases. OBJECTIVE Since there are no clear mechanisms for orthostatic hypotension in human AD, the present study assessed the autonomic changes that could explain this comorbidity in an AD animal model. METHODS We used the established streptozotocin-induced rat model of AD (STZ-AD), which mimics many hallmark symptoms of sporadic AD in humans. Baroreflex responses were analyzed in anesthetized STZ-AD rats using femoral catheterization for blood pressure and heart rate, and autonomic activity was assessed using specific blockers and splanchnic sympathetic nerve recordings. Expression levels of autonomic receptors at the heart were examined using the western blot technique. RESULTS Baroreflex function in STZ-AD showed a blunted heart rate (HR) response to low blood pressure challenges, and the maximal sympathetic nerve activity was reduced. Conversely, HR responses to high blood pressure were similar to control, indicating no change in parasympathetic nerve activity. Under resting conditions, autonomic blockade demonstrated a baseline shift to increased sympathetic tone in STZ-AD. Protein expression levels of beta-1 adrenergic receptor and muscarinic acetylcholine receptor M2 in the heart were unchanged. CONCLUSION Our study provides the first data on the pathological influence of AD on baroreflex function, which primarily affected the sympathetic nervous system in STZ-AD. These results represent the first mechanisms that may correlate with the orthostatic hypotension in human AD.
Collapse
Affiliation(s)
- John C. Ehlen
- Department of Physiology, Kirksville College of Osteopathic Medicine, A.T. Still University of Health Sciences, Kirksville, MO, USA
| | | | | | - Tim D. Ostrowski
- Department of Physiology, Kirksville College of Osteopathic Medicine, A.T. Still University of Health Sciences, Kirksville, MO, USA
| |
Collapse
|
31
|
RGS3L allows for an M 2 muscarinic receptor-mediated RhoA-dependent inotropy in cardiomyocytes. Basic Res Cardiol 2022; 117:8. [PMID: 35230541 PMCID: PMC8888479 DOI: 10.1007/s00395-022-00915-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 01/27/2022] [Accepted: 01/28/2022] [Indexed: 01/31/2023]
Abstract
The role and outcome of the muscarinic M2 acetylcholine receptor (M2R) signaling in healthy and diseased cardiomyocytes is still a matter of debate. Here, we report that the long isoform of the regulator of G protein signaling 3 (RGS3L) functions as a switch in the muscarinic signaling, most likely of the M2R, in primary cardiomyocytes. High levels of RGS3L, as found in heart failure, redirect the Gi-mediated Rac1 activation into a Gi-mediated RhoA/ROCK activation. Functionally, this switch resulted in a reduced production of reactive oxygen species (- 50%) in cardiomyocytes and an inotropic response (+ 18%) in transduced engineered heart tissues. Importantly, we could show that an adeno-associated virus 9-mediated overexpression of RGS3L in rats in vivo, increased the contractility of ventricular strips by maximally about twofold. Mechanistically, we demonstrate that this switch is mediated by a complex formation of RGS3L with the GTPase-activating protein p190RhoGAP, which balances the activity of RhoA and Rac1 by altering its substrate preference in cardiomyocytes. Enhancement of this complex formation could open new possibilities in the regulation of the contractility of the diseased heart.
Collapse
|
32
|
Cavalcante GL, Brognara F, Oliveira LVDC, Lataro RM, Durand MDT, Oliveira AP, Nóbrega ACL, Salgado HC, Sabino JPJ. Benefits of pharmacological and electrical cholinergic stimulation in hypertension and heart failure. Acta Physiol (Oxf) 2021; 232:e13663. [PMID: 33884761 DOI: 10.1111/apha.13663] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 03/12/2021] [Accepted: 04/06/2021] [Indexed: 12/11/2022]
Abstract
Systemic arterial hypertension and heart failure are cardiovascular diseases that affect millions of individuals worldwide. They are characterized by a change in the autonomic nervous system balance, highlighted by an increase in sympathetic activity associated with a decrease in parasympathetic activity. Most therapeutic approaches seek to treat these diseases by medications that attenuate sympathetic activity. However, there is a growing number of studies demonstrating that the improvement of parasympathetic function, by means of pharmacological or electrical stimulation, can be an effective tool for the treatment of these cardiovascular diseases. Therefore, this review aims to describe the advances reported by experimental and clinical studies that addressed the potential of cholinergic stimulation to prevent autonomic and cardiovascular imbalance in hypertension and heart failure. Overall, the published data reviewed demonstrate that the use of central or peripheral acetylcholinesterase inhibitors is efficient to improve the autonomic imbalance and hemodynamic changes observed in heart failure and hypertension. Of note, the baroreflex and the vagus nerve activation have been shown to be safe and effective approaches to be used as an alternative treatment for these cardiovascular diseases. In conclusion, pharmacological and electrical stimulation of the parasympathetic nervous system has the potential to be used as a therapeutic tool for the treatment of hypertension and heart failure, deserving to be more explored in the clinical setting.
Collapse
Affiliation(s)
- Gisele L. Cavalcante
- Graduate Program in Pharmaceutical Sciences Department of Biophysics and Physiology Federal University of Piaui Teresina PI Brazil
- Department of Pharmacology Ribeirão Preto Medical School University of São Paulo Ribeirão Preto SP Brazil
| | - Fernanda Brognara
- Department of Physiology Ribeirão Preto Medical School University of São Paulo Ribeirão Preto SP Brazil
| | - Lucas Vaz de C. Oliveira
- Graduate Program in Pharmaceutical Sciences Department of Biophysics and Physiology Federal University of Piaui Teresina PI Brazil
| | - Renata M. Lataro
- Department of Physiological Sciences Center of Biological Sciences Federal University of Santa Catarina Florianópolis SP Brazil
| | | | - Aldeidia P. Oliveira
- Graduate Program in Pharmacology Department of Biophysics and Physiology Federal University of Piaui Teresina PI Brazil
| | | | - Helio C. Salgado
- Department of Physiology Ribeirão Preto Medical School University of São Paulo Ribeirão Preto SP Brazil
| | - João Paulo J. Sabino
- Graduate Program in Pharmaceutical Sciences Department of Biophysics and Physiology Federal University of Piaui Teresina PI Brazil
| |
Collapse
|
33
|
Walther LM, von Känel R, Heimgartner N, Zuccarella-Hackl C, Ehlert U, Wirtz PH. Altered Cardiovascular Reactivity to and Recovery from Cold Face Test-Induced Parasympathetic Stimulation in Essential Hypertension. J Clin Med 2021; 10:2714. [PMID: 34205387 PMCID: PMC8235104 DOI: 10.3390/jcm10122714] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 06/11/2021] [Accepted: 06/14/2021] [Indexed: 12/15/2022] Open
Abstract
Essential hypertension is associated with increased sympathetic and diminished parasympathetic activity as well as impaired reactivity to sympathetic stimulation. However, reactivity and recovery from parasympathetic stimulation in hypertension are unknown. We investigated reactivity and recovery to primarily parasympathetic stimulation by Cold Face Test (CFT) in essential hypertension. Moreover, we tested whether chronic stress modulates CFT-reactivity dependent on hypertension status. The CFT was conducted by applying a cold face-mask for 2 min in 24 unmedicated, otherwise healthy hypertensive men and in 24 normotensive controls. Systolic and diastolic blood pressure (BP) and heart rate (HR) were measured repeatedly. Chronic stress was assessed with the Trier-Inventory-for-Chronic-Stress-Screening-Scale. Hypertensives did not exhibit diastolic BP decreases after CFT-cessation (p = 0.59) as did normotensives (p = 0.002) and failed to show HR decreases in immediate response to CFT (p = 0.62) when compared to normotensives (p < 0.001). Systolic BP reactivity and recovery patterns did not differ between hypertensives and normotensives (p = 0.44). Chronic stress moderated HR (p = 0.045) but not BP CFT-reactivity (p's > 0.64) with chronically stressed normotensives showing similar HR reactivity as hypertensives. Our findings indicate impaired diastolic BP and HR reactivity to and recovery from CFT in hypertensives and a moderating effect of chronic stress on HR reactivity potentially reflecting reduced relaxation ability of the cardiovascular system.
Collapse
Affiliation(s)
- Lisa-Marie Walther
- Biological Work and Health Psychology, University of Konstanz, 78457 Konstanz, Germany;
- Centre for the Advanced Study of Collective Behaviour, University of Konstanz, 78457 Konstanz, Germany
| | - Roland von Känel
- Department of Consultation-Liaison Psychiatry and Psychosomatic Medicine, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland; (R.v.K.); (C.Z.-H.)
| | - Nadja Heimgartner
- Division of Clinical Psychology and Psychotherapy, University of Basel, 4055 Basel, Switzerland;
| | - Claudia Zuccarella-Hackl
- Department of Consultation-Liaison Psychiatry and Psychosomatic Medicine, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland; (R.v.K.); (C.Z.-H.)
| | - Ulrike Ehlert
- Department of Clinical Psychology and Psychotherapy, University of Zurich, 8050 Zurich, Switzerland;
| | - Petra H. Wirtz
- Biological Work and Health Psychology, University of Konstanz, 78457 Konstanz, Germany;
- Centre for the Advanced Study of Collective Behaviour, University of Konstanz, 78457 Konstanz, Germany
| |
Collapse
|
34
|
Schoeller C, Hoffmann S, Adolph S, Regenthal R, Abraham G. Expression of muscarinic acetylcholine receptors in turkey cardiac chambers. Res Vet Sci 2021; 136:602-608. [PMID: 33895569 DOI: 10.1016/j.rvsc.2021.04.016] [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: 05/03/2020] [Revised: 04/12/2021] [Accepted: 04/13/2021] [Indexed: 10/21/2022]
Abstract
The aim of the present study was to characterize the specific binding sites for [N-methyl-3H]-scopolamine ([3H]-NMS), a radioligand for labeling muscarinic acetylcholine receptors (mAChRs), in membranes of four heart chambers obtained from adult male British United Turkey (BUT) Big 6 ("meat-type") and Cröllwitzer ("wild-type") turkeys. MAChR subtypes were examined by inhibiting [3H]-NMS binding with subtype selective non-labelled receptor antagonists. In all left and right atria as well as left and right ventricles of both turkey breeds, the specific [3H]-NMS binding was saturable, reversible and of high affinity (KD range: 0.5-1.0 nM). The maximum receptor density (Bmax) was not significantly different between the four cardiac chambers of BUT Big 6 turkeys, but a significant difference was found between atria and ventricles of Cröllwitzer turkeys. Moreover, significant lower Bmax was found in the atria of Cröllwitzer turkeys than in the atria of BUT Big 6, while the ventricular Bmax was significantly higher. In all cardiac chambers, unlabeled mAChR antagonists competed for specific [3H]-NMS binding sites in a concentration-dependent manner, suggesting the presence of the M3 and M2 receptor subtypes, whereby the latter was the predominant subtype. The presence of the M1 subtype could not be excluded. In conclusion, there was a difference between BUT Big 6 ("meat-type") and Cröllwitzer ("wild-type") turkeys with regard to receptor density in heart chambers with dominant M2 and M3 receptor subtypes.
Collapse
Affiliation(s)
- Caroline Schoeller
- Institute of Pharmacology, Pharmacy and Toxicology, Faculty of Veterinary Medicine, University of Leipzig, An den Tierkliniken 15, D-04103 Leipzig, Germany
| | - Sandra Hoffmann
- Institute of Pharmacology, Pharmacy and Toxicology, Faculty of Veterinary Medicine, University of Leipzig, An den Tierkliniken 15, D-04103 Leipzig, Germany
| | - Stephanie Adolph
- Institute of Pharmacology, Pharmacy and Toxicology, Faculty of Veterinary Medicine, University of Leipzig, An den Tierkliniken 15, D-04103 Leipzig, Germany
| | - Ralf Regenthal
- Rudolf-Boehm-Institute of Pharmacology and Toxicology, Clinical Pharmacology, Faculty of Medicine, University of Leipzig, Härtelstr. 16 -18, D-04107 Leipzig, Germany.
| | - Getu Abraham
- Institute of Pharmacology, Pharmacy and Toxicology, Faculty of Veterinary Medicine, University of Leipzig, An den Tierkliniken 15, D-04103 Leipzig, Germany.
| |
Collapse
|
35
|
Ali O, Tolaymat M, Hu S, Xie G, Raufman JP. Overcoming Obstacles to Targeting Muscarinic Receptor Signaling in Colorectal Cancer. Int J Mol Sci 2021; 22:ijms22020716. [PMID: 33450835 PMCID: PMC7828259 DOI: 10.3390/ijms22020716] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 01/08/2021] [Accepted: 01/10/2021] [Indexed: 01/05/2023] Open
Abstract
Despite great advances in our understanding of the pathobiology of colorectal cancer and the genetic and environmental factors that mitigate its onset and progression, a paucity of effective treatments persists. The five-year survival for advanced, stage IV disease remains substantially less than 20%. This review examines a relatively untapped reservoir of potential therapies to target muscarinic receptor expression, activation, and signaling in colorectal cancer. Most colorectal cancers overexpress M3 muscarinic receptors (M3R), and both in vitro and in vivo studies have shown that activating these receptors stimulates cellular programs that result in colon cancer growth, survival, and spread. In vivo studies using mouse models of intestinal neoplasia have shown that using either genetic or pharmacological approaches to block M3R expression and activation, respectively, attenuates the development and progression of colon cancer. Moreover, both in vitro and in vivo studies have shown that blocking the activity of matrix metalloproteinases (MMPs) that are induced selectively by M3R activation, i.e., MMP1 and MMP7, also impedes colon cancer growth and progression. Nonetheless, the widespread expression of muscarinic receptors and MMPs and their importance for many cellular functions raises important concerns about off-target effects and the safety of employing similar strategies in humans. As we highlight in this review, highly selective approaches can overcome these obstacles and permit clinicians to exploit the reliance of colon cancer cells on muscarinic receptors and their downstream signal transduction pathways for therapeutic purposes.
Collapse
Affiliation(s)
- Osman Ali
- Department of Medicine, Division of Gastroenterology & Hepatology, University of Maryland School of Medicine, Baltimore, MA 21201, USA; (O.A.); (M.T.); (S.H.); (G.X.)
| | - Mazen Tolaymat
- Department of Medicine, Division of Gastroenterology & Hepatology, University of Maryland School of Medicine, Baltimore, MA 21201, USA; (O.A.); (M.T.); (S.H.); (G.X.)
| | - Shien Hu
- Department of Medicine, Division of Gastroenterology & Hepatology, University of Maryland School of Medicine, Baltimore, MA 21201, USA; (O.A.); (M.T.); (S.H.); (G.X.)
- Veterans Affairs Maryland Healthcare System, Baltimore, MA 21201, USA
| | - Guofeng Xie
- Department of Medicine, Division of Gastroenterology & Hepatology, University of Maryland School of Medicine, Baltimore, MA 21201, USA; (O.A.); (M.T.); (S.H.); (G.X.)
- Veterans Affairs Maryland Healthcare System, Baltimore, MA 21201, USA
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MA 21201, USA
| | - Jean-Pierre Raufman
- Department of Medicine, Division of Gastroenterology & Hepatology, University of Maryland School of Medicine, Baltimore, MA 21201, USA; (O.A.); (M.T.); (S.H.); (G.X.)
- Veterans Affairs Maryland Healthcare System, Baltimore, MA 21201, USA
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MA 21201, USA
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MA 21201, USA
- Correspondence: ; Tel.: +1-410-328-8728
| |
Collapse
|
36
|
Anderson A, Masuho I, Marron Fernandez de Velasco E, Nakano A, Birnbaumer L, Martemyanov KA, Wickman K. GPCR-dependent biasing of GIRK channel signaling dynamics by RGS6 in mouse sinoatrial nodal cells. Proc Natl Acad Sci U S A 2020; 117:14522-14531. [PMID: 32513692 PMCID: PMC7322085 DOI: 10.1073/pnas.2001270117] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
How G protein-coupled receptors (GPCRs) evoke specific biological outcomes while utilizing a limited array of G proteins and effectors is poorly understood, particularly in native cell systems. Here, we examined signaling evoked by muscarinic (M2R) and adenosine (A1R) receptor activation in the mouse sinoatrial node (SAN), the cardiac pacemaker. M2R and A1R activate a shared pool of cardiac G protein-gated inwardly rectifying K+ (GIRK) channels in SAN cells from adult mice, but A1R-GIRK responses are smaller and slower than M2R-GIRK responses. Recordings from mice lacking Regulator of G protein Signaling 6 (RGS6) revealed that RGS6 exerts a GPCR-dependent influence on GIRK-dependent signaling in SAN cells, suppressing M2R-GIRK coupling efficiency and kinetics and A1R-GIRK signaling amplitude. Fast kinetic bioluminescence resonance energy transfer assays in transfected HEK cells showed that RGS6 prefers Gαo over Gαi as a substrate for its catalytic activity and that M2R signals preferentially via Gαo, while A1R does not discriminate between inhibitory G protein isoforms. The impact of atrial/SAN-selective ablation of Gαo or Gαi2 was consistent with these findings. Gαi2 ablation had minimal impact on M2R-GIRK and A1R-GIRK signaling in SAN cells. In contrast, Gαo ablation decreased the amplitude and slowed the kinetics of M2R-GIRK responses, while enhancing the sensitivity and prolonging the deactivation rate of A1R-GIRK signaling. Collectively, our data show that differences in GPCR-G protein coupling preferences, and the Gαo substrate preference of RGS6, shape A1R- and M2R-GIRK signaling dynamics in mouse SAN cells.
Collapse
Affiliation(s)
- Allison Anderson
- Department of Pharmacology, University of Minnesota, Minneapolis, MN 55455
| | - Ikuo Masuho
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL 33458
| | | | - Atsushi Nakano
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, CA 90095
| | - Lutz Birnbaumer
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709
- Biomedical Research Institute, Catholic University of Argentina, C1107AAZ Buenos Aires, Argentina
| | | | - Kevin Wickman
- Department of Pharmacology, University of Minnesota, Minneapolis, MN 55455;
| |
Collapse
|
37
|
Kini RM, Koh CY. Snake venom three-finger toxins and their potential in drug development targeting cardiovascular diseases. Biochem Pharmacol 2020; 181:114105. [PMID: 32579959 DOI: 10.1016/j.bcp.2020.114105] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 06/13/2020] [Accepted: 06/17/2020] [Indexed: 12/15/2022]
Abstract
Cardiovascular diseases such as coronary and peripheral artery diseases, venous thrombosis, stroke, hypertension, and heart failure are enormous burden to health and economy globally. Snake venoms have been the sources of discovery of successful therapeutics targeting cardiovascular diseases. For example, the first-in-class angiotensin-converting enzyme inhibitor captopril was designed largely based on bradykinin-potentiating peptides from Bothrops jararaca venom. In the recent years, sensitive and high throughput approaches drive discovery and cataloging of new snake venom toxins. As one of the largest class of snake venom toxin, there are now>700 sequences of three-finger toxins (3FTxs) available, many of which are yet to be studied. While the function of 3FTxs are normally associated with neurotoxicity, increasingly more 3FTxs have been characterized to have pharmacological effects on cardiovascular systems. Here we focus on this family of snake venom toxins and their potential in developing therapeutics against cardiovascular diseases.
Collapse
Affiliation(s)
- R Manjunatha Kini
- Department of Biological Sciences, Faculty of Science, National University of Singapore, 117558, Singapore; Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117600, Singapore
| | - Cho Yeow Koh
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, 119228, Singapore.
| |
Collapse
|
38
|
Using Network Pharmacology to Explore Potential Treatment Mechanism for Coronary Heart Disease Using Chuanxiong and Jiangxiang Essential Oils in Jingzhi Guanxin Prescriptions. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 2019:7631365. [PMID: 31772600 PMCID: PMC6854988 DOI: 10.1155/2019/7631365] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 08/30/2019] [Accepted: 09/14/2019] [Indexed: 01/06/2023]
Abstract
Background To predict the active components and potential targets of traditional Chinese medicine and to determine the mechanism behind the curative effect of traditional Chinese medicine, a multitargeted method was used. Jingzhi Guanxin prescriptions expressed a high efficacy for coronary heart disease (CHD) patients of which essential oils from Chuanxiong and Jiangxiang were confirmed to be the most important effective substance. However, the interaction between the active components and the targets for the treatment of CHD has not been clearly explained in previous studies. Materials and Methods Genes associated with the disease and the treatment strategy were searched from the electronic database and analyzed by Cytoscape (version 3.2.1). Protein-protein interaction network diagram of CHD with Jiangxiang and Chuanxiong essential oils was constructed by Cytoscape. Pathway functional enrichment analysis was executed by clusterProfiler package in R platform. Results 121 ingredients of Chuanxiong and Jiangxiang essential oils were analyzed, and 393 target genes of the compositions and 912 CHD-related genes were retrieved. 15 coexpression genes were selected, including UGT1A1, DPP4, RXRA, ADH1A, RXRG, UGT1A3, PPARA, TRPC3, CYP1A1, ABCC2, AHR, and ADRA2A. The crucial pathways of occurrence and treatment molecular mechanism of CHD were analyzed, including retinoic acid metabolic process, flavonoid metabolic process, response to xenobiotic stimulus, cellular response to xenobiotic stimulus, cellular response to steroid hormone stimulus, retinoid binding, retinoic acid binding, and monocarboxylic acid binding. Finally, we elucidate the underlying role and mechanism behind these genes in the pathogenesis and treatment of CHD. Conclusions Generally speaking, the nodes in subnetwork affect the pathological process of CHD, thus indicating the mechanism of Jingzhi Guanxin prescriptions containing Chuanxiong and Jiangxiang essential oils in the treatment of CHD.
Collapse
|
39
|
Riefolo F, Matera C, Garrido-Charles A, Gomila AMJ, Sortino R, Agnetta L, Claro E, Masgrau R, Holzgrabe U, Batlle M, Decker M, Guasch E, Gorostiza P. Optical Control of Cardiac Function with a Photoswitchable Muscarinic Agonist. J Am Chem Soc 2019; 141:7628-7636. [DOI: 10.1021/jacs.9b03505] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Fabio Riefolo
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute for Science and Technology (BIST), Carrer de Baldiri Reixac 15-21, 08028 Barcelona, Spain
- Network Biomedical Research Center in Bioengineering, Biomaterials, and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
| | - Carlo Matera
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute for Science and Technology (BIST), Carrer de Baldiri Reixac 15-21, 08028 Barcelona, Spain
- Network Biomedical Research Center in Bioengineering, Biomaterials, and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
| | - Aida Garrido-Charles
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute for Science and Technology (BIST), Carrer de Baldiri Reixac 15-21, 08028 Barcelona, Spain
- Network Biomedical Research Center in Bioengineering, Biomaterials, and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
| | - Alexandre M. J. Gomila
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute for Science and Technology (BIST), Carrer de Baldiri Reixac 15-21, 08028 Barcelona, Spain
- Network Biomedical Research Center in Bioengineering, Biomaterials, and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
| | - Rosalba Sortino
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute for Science and Technology (BIST), Carrer de Baldiri Reixac 15-21, 08028 Barcelona, Spain
- Network Biomedical Research Center in Bioengineering, Biomaterials, and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
| | - Luca Agnetta
- Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy and Food Chemistry, Julius Maximilian University of Würzburg, Am Hubland, D-97074 Würzburg, Germany
| | - Enrique Claro
- Institut de Neurociències (INc), and Dept. Bioquímica i Biologia Molecular, Unitat de Bioquímica de Medicina, Universitat Autònoma de Barcelona (UAB), 08193 Bellaterra, Barcelona, Spain
| | - Roser Masgrau
- Institut de Neurociències (INc), and Dept. Bioquímica i Biologia Molecular, Unitat de Bioquímica de Medicina, Universitat Autònoma de Barcelona (UAB), 08193 Bellaterra, Barcelona, Spain
| | - Ulrike Holzgrabe
- Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy and Food Chemistry, Julius Maximilian University of Würzburg, Am Hubland, D-97074 Würzburg, Germany
| | - Montserrat Batlle
- Cardiovascular Institute, Hospital Clinic, University of Barcelona (UB), IDIBAPS, 08036 Barcelona, Spain
| | - Michael Decker
- Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy and Food Chemistry, Julius Maximilian University of Würzburg, Am Hubland, D-97074 Würzburg, Germany
| | - Eduard Guasch
- Cardiovascular Institute, Hospital Clinic, University of Barcelona (UB), IDIBAPS, 08036 Barcelona, Spain
- Network Biomedical Research Center in Cardiovascular Diseases (CIBER-CV), 28029 Madrid, Spain
| | - Pau Gorostiza
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute for Science and Technology (BIST), Carrer de Baldiri Reixac 15-21, 08028 Barcelona, Spain
- Network Biomedical Research Center in Bioengineering, Biomaterials, and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), 08010 Barcelona, Spain
| |
Collapse
|
40
|
Cowley AW. Chrm3 Gene and M3 Muscarinic Receptors Contribute to Salt-Sensitive Hypertension. Hypertension 2019; 72:588-591. [PMID: 30354773 DOI: 10.1161/hypertensionaha.118.11494] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Allen W Cowley
- From the Department of Physiology, Medical College of Wisconsin, Milwaukee
| |
Collapse
|
41
|
Boutagy NE, Feher A, Alkhalil I, Umoh N, Sinusas AJ. Molecular Imaging of the Heart. Compr Physiol 2019; 9:477-533. [PMID: 30873600 DOI: 10.1002/cphy.c180007] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Multimodality cardiovascular imaging is routinely used to assess cardiac function, structure, and physiological parameters to facilitate the diagnosis, characterization, and phenotyping of numerous cardiovascular diseases (CVD), as well as allows for risk stratification and guidance in medical therapy decision-making. Although useful, these imaging strategies are unable to assess the underlying cellular and molecular processes that modulate pathophysiological changes. Over the last decade, there have been great advancements in imaging instrumentation and technology that have been paralleled by breakthroughs in probe development and image analysis. These advancements have been merged with discoveries in cellular/molecular cardiovascular biology to burgeon the field of cardiovascular molecular imaging. Cardiovascular molecular imaging aims to noninvasively detect and characterize underlying disease processes to facilitate early diagnosis, improve prognostication, and guide targeted therapy across the continuum of CVD. The most-widely used approaches for preclinical and clinical molecular imaging include radiotracers that allow for high-sensitivity in vivo detection and quantification of molecular processes with single photon emission computed tomography and positron emission tomography. This review will describe multimodality molecular imaging instrumentation along with established and novel molecular imaging targets and probes. We will highlight how molecular imaging has provided valuable insights in determining the underlying fundamental biology of a wide variety of CVDs, including: myocardial infarction, cardiac arrhythmias, and nonischemic and ischemic heart failure with reduced and preserved ejection fraction. In addition, the potential of molecular imaging to assist in the characterization and risk stratification of systemic diseases, such as amyloidosis and sarcoidosis will be discussed. © 2019 American Physiological Society. Compr Physiol 9:477-533, 2019.
Collapse
Affiliation(s)
- Nabil E Boutagy
- Department of Medicine, Yale Translational Research Imaging Center, Yale University School of Medicine, Section of Cardiovascular Medicine, New Haven, Connecticut, USA
| | - Attila Feher
- Department of Medicine, Yale Translational Research Imaging Center, Yale University School of Medicine, Section of Cardiovascular Medicine, New Haven, Connecticut, USA
| | - Imran Alkhalil
- Department of Medicine, Yale Translational Research Imaging Center, Yale University School of Medicine, Section of Cardiovascular Medicine, New Haven, Connecticut, USA
| | - Nsini Umoh
- Department of Medicine, Yale Translational Research Imaging Center, Yale University School of Medicine, Section of Cardiovascular Medicine, New Haven, Connecticut, USA
| | - Albert J Sinusas
- Department of Medicine, Yale Translational Research Imaging Center, Yale University School of Medicine, Section of Cardiovascular Medicine, New Haven, Connecticut, USA.,Yale University School of Medicine, Department of Radiology and Biomedical Imaging, New Haven, Connecticut, USA
| |
Collapse
|
42
|
Ivanova AD, Tapilina SV, Kuz'min VS. Role of Muscarinic M1, M2, and M3 Receptors in the Regulation of Electrical Activity of Myocardial Tissue of Caval Veins during the Early Postnatal Ontogeny. Bull Exp Biol Med 2019; 166:421-425. [PMID: 30783837 DOI: 10.1007/s10517-019-04364-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Indexed: 01/29/2023]
Abstract
We studied the influence of blockers of muscarinic M1, M2, and M3 receptors on the effect of acetylcholine in the myocardial tissue of caval veins in rats at the early stage of ontogeny. The experiments were performed on isolated preparations of the right superior vena cava working under their own rhythm. Action potentials were recorded using the standard microelectrode technique. Acetylcholine (1 μM) suppressed automatic activity in the superior vena cava myocardium. Preliminary perfusion of the preparation with non-selective blocker atropine (1 μM) completely abolished the effect of acetylcholine, treatment with M2 receptor blocker AQ-RA 741 (1 μM) led to partial suppression of the effect of acetylcholine. Blockers of M1 and M3 receptors pirenzepine (1 μM) and 4DAMP (0.1 μM) did not suppress the effect of acetylcholine. Thus, the effect of acetylcholine is predominantly realized via M2 receptors, but M3 receptors can also partially mediate its effect in the superior vena cava myocardium in rats at the early stages of ontogeny.
Collapse
Affiliation(s)
- A D Ivanova
- Department of Human and Animal Physiology, M. V. Lomonosov Moscow State University, Ministry of Health of the Russian Federation, Moscow, Russia.
| | - S V Tapilina
- Department of Human and Animal Physiology, M. V. Lomonosov Moscow State University, Ministry of Health of the Russian Federation, Moscow, Russia
- Department of Physiology, N. I. Pirogov Russian National Research Medical University, Ministry of Health of the Russian Federation, Moscow, Russia
| | - V S Kuz'min
- Department of Human and Animal Physiology, M. V. Lomonosov Moscow State University, Ministry of Health of the Russian Federation, Moscow, Russia
- Department of Physiology, N. I. Pirogov Russian National Research Medical University, Ministry of Health of the Russian Federation, Moscow, Russia
| |
Collapse
|
43
|
Chenopodium ambrosioides induces an endothelium-dependent relaxation of rat isolated aorta. JOURNAL OF INTEGRATIVE MEDICINE-JIM 2019; 17:115-124. [PMID: 30738772 DOI: 10.1016/j.joim.2019.01.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 12/27/2018] [Indexed: 12/30/2022]
Abstract
OBJECTIVE This study aims to evaluate the vasodilatory effect of Chenopodium ambrosioides on the isolated rat aorta, and to explore its mechanism of action. METHODS The vasorelaxant effect and the mode of action of various extracts from the leaves of C. ambrosioides were evaluated on thoracic aortic rings isolated from Wistar rats. In addition, ethyl acetate and methanol fractions were analyzed, using thin-layer chromatography and high-performance liquid chromatography techniques, for their polyphenolic content. RESULTS The various active extracts of C. ambrosioides at four concentrations (10-3, 10-2, 10-1 and 1 mg/mL) relaxed the contraction elicited by phenylephrine, in a concentration-dependent manner. This effect seems to be endothelium-dependent, since the vasodilatory effect was entirely absent in denuded aortic rings. The vasorelaxant effect of the methanol fraction (MF) of C. ambrosioides at 1 mg/mL was also inhibited by atropine and tetraethylammonium. This effect remained unchanged by Nω-nitro-l-arginine methyl ester hydrochloride and glibenclamide. The preliminary phytochemical analysis showed that the leaves of C. ambrosioides are rich in phenolic and flavonoid derivatives. CONCLUSION These results suggest that the MF of C. ambrosioides produces an endothelium-dependent relaxation of the isolated rat aorta, which is thought to be mediated mainly through stimulation of the muscarinic receptors, and probably involving the opening of Ca2+-activated potassium channels.
Collapse
|
44
|
Calloe K. Doctoral Dissertation: The transient outward potassium current in healthy and diseased hearts. Acta Physiol (Oxf) 2019; 225 Suppl 717:e13225. [PMID: 30628199 DOI: 10.1111/apha.13225] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Revised: 11/20/2018] [Accepted: 11/21/2018] [Indexed: 12/13/2022]
Affiliation(s)
- Kirstine Calloe
- Section for Anatomy; Biochemistry and Physiology; Department for Veterinary and Animal Sciences; Faculty of Health and Medical Sciences; University of Copenhagen; Frederiksberg C Denmark
| |
Collapse
|
45
|
Hanafi NI, Mohamed AS, Sheikh Abdul Kadir SH, Othman MHD. Overview of Bile Acids Signaling and Perspective on the Signal of Ursodeoxycholic Acid, the Most Hydrophilic Bile Acid, in the Heart. Biomolecules 2018; 8:E159. [PMID: 30486474 PMCID: PMC6316857 DOI: 10.3390/biom8040159] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 11/15/2018] [Accepted: 11/15/2018] [Indexed: 12/12/2022] Open
Abstract
Bile acids (BA) are classically known as an important agent in lipid absorption and cholesterol metabolism. Nowadays, their role in glucose regulation and energy homeostasis are widely reported. BAs are involved in various cellular signaling pathways, such as protein kinase cascades, cyclic AMP (cAMP) synthesis, and calcium mobilization. They are ligands for several nuclear hormone receptors, including farnesoid X-receptor (FXR). Recently, BAs have been shown to bind to muscarinic receptor and Takeda G-protein-coupled receptor 5 (TGR5), both G-protein-coupled receptor (GPCR), independent of the nuclear hormone receptors. Moreover, BA signals have also been elucidated in other nonclassical BA pathways, such as sphingosine-1-posphate and BK (large conductance calcium- and voltage activated potassium) channels. Hydrophobic BAs have been proven to affect heart rate and its contraction. Elevated BAs are associated with arrhythmias in adults and fetal heart, and altered ratios of primary and secondary bile acid are reported in chronic heart failure patients. Meanwhile, in patients with liver cirrhosis, cardiac dysfunction has been strongly linked to the increase in serum bile acid concentrations. In contrast, the most hydrophilic BA, known as ursodeoxycholic acid (UDCA), has been found to be beneficial in improving peripheral blood flow in chronic heart failure patients and in protecting the heart against reperfusion injury. This review provides an overview of BA signaling, with the main emphasis on past and present perspectives on UDCA signals in the heart.
Collapse
Affiliation(s)
- Noorul Izzati Hanafi
- Institute of Medical Molecular Biotechnology, Faculty of Medicine, Universiti Teknologi MARA, Sungai Buloh 47000, Selangor, Malaysia.
| | - Anis Syamimi Mohamed
- Institute of Medical Molecular Biotechnology, Faculty of Medicine, Universiti Teknologi MARA, Sungai Buloh 47000, Selangor, Malaysia.
| | - Siti Hamimah Sheikh Abdul Kadir
- Institute of Medical Molecular Biotechnology, Faculty of Medicine, Universiti Teknologi MARA, Sungai Buloh 47000, Selangor, Malaysia.
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Universiti Teknologi MARA, Sungai Buloh 47000, Selangor, Malaysia.
| | - Mohd Hafiz Dzarfan Othman
- Advanced Membrane Technology Research Centre (AMTEC), Universiti Teknologi Malaysia, Johor Bharu 81310, Johor, Malaysia.
| |
Collapse
|
46
|
Saw EL, Kakinuma Y, Fronius M, Katare R. The non-neuronal cholinergic system in the heart: A comprehensive review. J Mol Cell Cardiol 2018; 125:129-139. [PMID: 30343172 DOI: 10.1016/j.yjmcc.2018.10.013] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 09/24/2018] [Accepted: 10/14/2018] [Indexed: 01/01/2023]
Abstract
The autonomic influences on the heart have a ying-yang nature, albeit oversimplified, the interplay between the sympathetic and parasympathetic system (known as the cholinergic system) is often complex and remain poorly understood. Recently, the heart has been recognized to consist of neuronal and non-neuronal cholinergic system (NNCS). The existence of cardiac NNCS has been confirmed by the presence of cholinergic markers in the cardiomyocytes, which are crucial for synthesis (choline acetyltransferase, ChAT), storage (vesicular acetylcholine transporter, VAChT), reuptake of choline for synthesis (high-affinity choline transporter, CHT1) and degradation (acetylcholinesterase, AChE) of acetylcholine (ACh). The non-neuronal ACh released from cardiomyocytes is believed to locally regulate some of the key physiological functions of the heart, such as regulation of heart rate, offsetting hypertrophic signals, maintenance of action potential propagation as well as modulation of cardiac energy metabolism via the muscarinic ACh receptor in an auto/paracrine manner. Apart from this, several studies have also provided evidence for the beneficial role of ACh released from cardiomyocytes against cardiovascular diseases such as sympathetic hyperactivity-induced cardiac remodeling and dysfunction as well as myocardial infarction, confirming the important role of NNCS in disease prevention. In this review, we aim to provide a fundamental overview of cardiac NNCS, and information about its physiological role, regulatory factors as well as its cardioprotective effects. Finally, we propose the different approaches to target cardiac NNCS as an adjunctive treatment to specifically address the withdrawal of neuronal cholinergic system in cardiovascular disease such as heart failure.
Collapse
Affiliation(s)
- Eng Leng Saw
- Department of Physiology-HeartOtago, School of Biomedical Sciences, University of Otago, New Zealand
| | - Yoshihiko Kakinuma
- Department of Physiology (Bioregulatory Science), Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| | - Martin Fronius
- Department of Physiology-HeartOtago, School of Biomedical Sciences, University of Otago, New Zealand.
| | - Rajesh Katare
- Department of Physiology-HeartOtago, School of Biomedical Sciences, University of Otago, New Zealand.
| |
Collapse
|
47
|
Lee SW, Anderson A, Guzman PA, Nakano A, Tolkacheva EG, Wickman K. Atrial GIRK Channels Mediate the Effects of Vagus Nerve Stimulation on Heart Rate Dynamics and Arrhythmogenesis. Front Physiol 2018; 9:943. [PMID: 30072916 PMCID: PMC6060443 DOI: 10.3389/fphys.2018.00943] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 06/27/2018] [Indexed: 01/09/2023] Open
Abstract
Diminished parasympathetic influence is central to the pathogenesis of cardiovascular diseases, including heart failure and hypertension. Stimulation of the vagus nerve has shown promise in treating cardiovascular disease, prompting renewed interest in understanding the signaling pathway(s) that mediate the vagal influence on cardiac physiology. Here, we evaluated the contribution of G protein-gated inwardly rectifying K+ (GIRK/Kir3) channels to the effect of vagus nerve stimulation (VNS) on heart rate (HR), HR variability (HRV), and arrhythmogenesis in anesthetized mice. As parasympathetic fibers innervate both atria and ventricle, and GIRK channels contribute to the cholinergic impact on atrial and ventricular myocytes, we collected in vivo electrocardiogram recordings from mice lacking either atrial or ventricular GIRK channels, during VNS. VNS decreased HR and increased HRV in control mice, in a muscarinic receptor-dependent manner. This effect was preserved in mice lacking ventricular GIRK channels, but was nearly completely absent in mice lacking GIRK channels in the atria. In addition, atrial-specific ablation of GIRK channels conferred resistance to arrhythmic episodes induced by VNS. These data indicate that atrial GIRK channels are the primary mediators of the impact of VNS on HR, HRV, and arrhythmogenesis in the anesthetized mouse.
Collapse
Affiliation(s)
- Steven W. Lee
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, United States
| | - Allison Anderson
- Department of Pharmacology, University of Minnesota, Minneapolis, MN, United States
| | - Pilar A. Guzman
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, United States
| | - Atsushi Nakano
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, Los Angeles, CA, United States
| | - Elena G. Tolkacheva
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, United States
| | - Kevin Wickman
- Department of Pharmacology, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
48
|
Bile acids and their respective conjugates elicit different responses in neonatal cardiomyocytes: role of Gi protein, muscarinic receptors and TGR5. Sci Rep 2018; 8:7110. [PMID: 29740092 PMCID: PMC5940781 DOI: 10.1038/s41598-018-25569-4] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 04/18/2018] [Indexed: 12/27/2022] Open
Abstract
Bile acids are recognised as bioactive signalling molecules. While they are known to influence arrhythmia susceptibility in cholestasis, there is limited knowledge about the underlying mechanisms. To delineate mechanisms underlying fetal heart rhythm disturbances in cholestatic pregnancy, we used FRET microscopy to monitor cAMP release and contraction measurements in isolated rodent neonatal cardiomyocytes. The unconjugated bile acids CDCA, DCA and UDCA and, to a lesser extent, CA were found to be relatively potent agonists for the GPBAR1 (TGR5) receptor and elicit cAMP release, whereas all glyco- and tauro- conjugated bile acids are weak agonists. The bile acid-induced cAMP production does not lead to an increase in contraction rate, and seems to be mediated by the RI isoform of adenylate cyclase, unlike adrenaline-dependent release which is mediated by the RII isoform. In contrast, bile acids elicited slowing of neonatal cardiomyocyte contraction indicating that other signalling pathways are involved. The conjugated bile acids were found to be partial agonists of the muscarinic M2, but not sphingosin-1-phosphate-2, receptors, and act partially through the Gi pathway. Furthermore, the contraction slowing effect of unconjugated bile acids may also relate to cytotoxicity at higher concentrations.
Collapse
|
49
|
Gurgul S, Buyukakilli B, Komur M, Okuyaz C, Balli E, Ozcan T. Does Levetiracetam Administration Prevent Cardiac Damage in Adulthood Rats Following Neonatal Hypoxia/Ischemia-Induced Brain Injury? ACTA ACUST UNITED AC 2018; 54:medicina54020012. [PMID: 30344243 PMCID: PMC6037241 DOI: 10.3390/medicina54020012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 04/04/2018] [Accepted: 04/06/2018] [Indexed: 12/12/2022]
Abstract
Cardiovascular abnormalities are widespread when a newborn is exposed to a hypoxic-ischemic injury in the neonatal period. Although the neuroprotective effects of levetiracetam (LEV) have been reported after hypoxia, the cardioprotective effects of LEV have not been documented. Therefore, we aimed to investigate whether levetiracetam (LEV) has a protective effect on cardiac-contractility and ultrastructure of heart muscle in rats exposed to hypoxia-ischemia (HI) during the neonatal period. A total of 49 seven-day-old rat pups were separated into four groups. For HI induction, a combination of right common carotid artery ligation with 8% oxygen in seven-day-old rat pups for 2 h was performed for saline, LEV100, and LEV200 groups. Just after hypoxia, LEV100 and LEV200 groups were administered with 100 mg/kg and 200 mg/kg of LEV, respectively. The arteries of rats in the control group were only detected; no ligation or hypoxia was performed. At the end of the 16th week after HI, cardiac mechanograms were recorded, and samples of tissue were explored by electronmicroscopy.While ventricular contractility in the control group was similar to LEV100, there were significant decreases in both saline and LEV200 groups (p < 0.05). Although ventricular contractile duration of the control and saline groups was found to be similar, durations in the LEV100 and LEV200 groups were significantly higher (p < 0.05). After HI, mitochondrial damage and ultrastructural deteriorative alterations in ventricles and atriums of the LEV-administered groups were significantly less severe than the saline group. The present study showed that neonatal HI caused long-term cardiac dysfunction and ultrastructural deteriorations in cardiac muscles. LEV administration just after HI might possess some protective effects against myocardial damage and contractility.
Collapse
Affiliation(s)
- Serkan Gurgul
- Department of Biophysics, Faculty of Medicine, Gaziantep University, TR-27310 Gaziantep, Turkey.
| | - Belgin Buyukakilli
- Department of Biophysics, Faculty of Medicine, Mersin University, TR-33343 Mersin, Turkey.
| | - Mustafa Komur
- Department of Child Health and Disease, Faculty of Medicine, Mersin University, TR-33343 Mersin, Turkey.
| | - Cetin Okuyaz
- Department of Child Health and Disease, Faculty of Medicine, Mersin University, TR-33343 Mersin, Turkey.
| | - Ebru Balli
- Department of Histology and Embryology, Faculty of Medicine, Mersin University, TR-33343 Mersin, Turkey.
| | - Tuba Ozcan
- Department of Histology and Embryology, Faculty of Medicine, K. Sütcü Imam University, TR-46040 Kahramanmaraş, Turkey.
| |
Collapse
|
50
|
Rosa GM, Baccino D, Valbusa A, Scala C, Barra F, Brunelli C, Ferrero S. Cardiovascular effects of antimuscarinic agents and beta3-adrenergic receptor agonist for the treatment of overactive bladder. Expert Opin Drug Saf 2018. [PMID: 29542337 DOI: 10.1080/14740338.2018.1453496] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
INTRODUCTION Overactive bladder (OAB) syndrome is common in the general population, particularly in elderly patients. Antimuscarinic drugs (AMs) are considered the mainstay pharmaceutical treatment of OAB whereas β3-adrenoceptor agonists, such as mirabegron, represent a good alternative. Owing to the important role of muscarinic and β3 receptors in cardiovascular (CV) tissue and to the fact that OAB patients often have CV comorbidities, the safety-profile of these drugs constitute an important challenge. AREAS COVERED The aim of this review is to evaluate the CV effects of AMs and mirabegron in OAB. A systematic literature search from inception until December 2017 was performed on PubMed and Medline. EXPERT OPINION AMs are generally considered to have good CV safety profile but, however, they may cause undesirable adverse events, such as dry mouth, constipation. CV AEs are rare but noteworthy, the most common CV consequences related to the use of these drugs are constituted by an increase in HR and QT interval. Mirabegron has similar efficacy and tolerability to AMs but causes less adverse events, with either modest hypertension and modest increase in HR (<5 bpm) being the most commonly reported.
Collapse
Affiliation(s)
- Gian Marco Rosa
- a Department of Internal Medicine, Cardiology , Ospedale Policlinico San Martino , Genoa , Italy
| | - Danilo Baccino
- a Department of Internal Medicine, Cardiology , Ospedale Policlinico San Martino , Genoa , Italy
| | - Alberto Valbusa
- a Department of Internal Medicine, Cardiology , Ospedale Policlinico San Martino , Genoa , Italy
| | - Carolina Scala
- b Academic Unit of Obstetrics and Gynecology , Ospedale Policlinico San Martino , Genoa , Italy.,c Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DiNOGMI) , University of Genoa , Genoa , Italy
| | - Fabio Barra
- b Academic Unit of Obstetrics and Gynecology , Ospedale Policlinico San Martino , Genoa , Italy.,c Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DiNOGMI) , University of Genoa , Genoa , Italy
| | - Claudio Brunelli
- a Department of Internal Medicine, Cardiology , Ospedale Policlinico San Martino , Genoa , Italy
| | - Simone Ferrero
- b Academic Unit of Obstetrics and Gynecology , Ospedale Policlinico San Martino , Genoa , Italy.,c Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DiNOGMI) , University of Genoa , Genoa , Italy
| |
Collapse
|