1
|
Guo Y, Cheng D, Yu ZY, Schiatti T, Chan AY, Hill AP, Peyronnet R, Feneley MP, Cox CD, Martinac B. Functional coupling between Piezo1 and TRPM4 influences the electrical activity of HL-1 atrial myocytes. J Physiol 2024; 602:4363-4386. [PMID: 38098265 DOI: 10.1113/jp284474] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 11/30/2023] [Indexed: 09/25/2024] Open
Abstract
The transient receptor potential melastatin 4 (TRPM4) channel contributes extensively to cardiac electrical activity, especially cardiomyocyte action potential formation. Mechanical stretch can induce changes in heart rate and rhythm, and the mechanosensitive channel Piezo1 is expressed in many cell types within the myocardium. Our previous study showed that TRPM4 and Piezo1 are closely co-localized in the t-tubules of ventricular cardiomyocytes and contribute to the Ca2+-dependent signalling cascade that underlies hypertrophy in response to mechanical pressure overload. However, there was no direct evidence showing that Piezo1 activation was related to TRPM4 activation in situ. In the present study, we employed the HL-1 mouse atrial myocyte-like cell line as an in vitro model to investigate whether Piezo1-TRPM4 coupling can affect action potential properties. We used the small molecule Piezo1 agonist, Yoda1, as a surrogate for mechanical stretch to activate Piezo1 and detected the action potential changes in HL-1 cells using FluoVolt, a fluorescent voltage sensitive dye. Our results demonstrate that Yoda1-induced activation of Piezo1 changes the action potential frequency in HL-1 cells. This change in action potential frequency is reduced by Piezo1 knockdown using small intefering RNA. Importantly knockdown or pharmacological inhibition of TRPM4 significantly affected the degree to which Yoda1-evoked Piezo1 activation influenced action potential frequency. Thus, the present study provides in vitro evidence of a functional coupling between Piezo1 and TRPM4 in a cardiomyocyte-like cell line. The coupling of a mechanosensitive Ca2+ permeable channel and a Ca2+-activated TRP channel probably represents a ubiquitous model for the role of TRP channels in mechanosensory transduction. KEY POINTS: The transient receptor potential melastatin 4 (TRPM4) and Piezo1 channels have been confirmed to contribute to the Ca2+-dependent signalling cascade that underlies cardiac hypertrophy in response to mechanical pressure overload. However, there was no direct evidence showing that Piezo1 activation was related to TRPM4 activation in situ. We employed the HL-1 mouse atrial myocyte-like cell line as an in vitro model to investigate the effect of Piezo1-TRPM4 coupling on cardiac electrical properties. The results show that both pharmacological and genetic inhibition of TRPM4 significantly affected the degree to which Piezo1 activation influenced action potential frequency in HL-1 cells. Our findings provide in vitro evidence of a functional coupling between Piezo1 and TRPM4 in a cardiomyocyte-like cell line. The coupling of a mechanosensitive Ca2+ permeable channel and a Ca2+-activated TRP channel probably represents a ubiquitous model for the role of TRP channels in mechanosensory transduction in various (patho)physiological processes.
Collapse
Affiliation(s)
- Yang Guo
- Victor Chang Cardiac Research Institute, Sydney, NSW, Australia
- Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Delfine Cheng
- Victor Chang Cardiac Research Institute, Sydney, NSW, Australia
- Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Ze-Yan Yu
- Victor Chang Cardiac Research Institute, Sydney, NSW, Australia
- Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Teresa Schiatti
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg-Bad Krozingen, and Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Andrea Y Chan
- Victor Chang Cardiac Research Institute, Sydney, NSW, Australia
| | - Adam P Hill
- Victor Chang Cardiac Research Institute, Sydney, NSW, Australia
- Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Rémi Peyronnet
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg-Bad Krozingen, and Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Michael P Feneley
- Victor Chang Cardiac Research Institute, Sydney, NSW, Australia
- Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
- Department of Cardiology, St Vincent's Hospital, Sydney, NSW, Australia
| | - Charles D Cox
- Victor Chang Cardiac Research Institute, Sydney, NSW, Australia
- Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Boris Martinac
- Victor Chang Cardiac Research Institute, Sydney, NSW, Australia
- Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
2
|
Naya NM, Kelly J, Hogwood A, Abbate A, Toldo S. Therapeutic potential of cannabidiol (CBD) in the treatment of cardiovascular diseases. Expert Opin Investig Drugs 2024; 33:699-712. [PMID: 38703078 DOI: 10.1080/13543784.2024.2351513] [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: 02/16/2024] [Accepted: 05/01/2024] [Indexed: 05/06/2024]
Abstract
INTRODUCTION Cannabidiol (CBD) is the primary non-psychoactive chemical derived from Cannabis Sativa, and its growing popularity is due to its potential therapeutic properties while avoiding the psychotropic effects of other phytocannabinoids, such as tetrahydrocannabinol (THC). Numerous pre-clinical studies in cellular and animal models and human clinical trials have demonstrated a positive impact of CBD on physiological and pathological processes. Recently, the FDA approved its use for the treatment of seizures, and clinical trials to test the efficacy of CBD in myocarditis and pericarditis are ongoing. AREAS COVERED We herein reviewed the current literature on the reported effects of CBD in the cardiovascular system, highlighting the physiological effects and the outcomes of using CBD as a therapeutic tool in pathological conditions to address this significant global health concern. EXPERT OPINION The comprehensive examination of the literature emphasizes the potential of CBD as a therapeutic option for treating cardiovascular diseases through its anti-inflammatory, vasodilatory, anti-fibrotic, and antioxidant properties in different conditions such as diabetic cardiomyopathy, myocarditis, doxorubicin-induced cardiotoxicity, and ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Nadia Martinez Naya
- Robert M. Berne Cardiovascular Research Center, Division of Cardiovascular Medicine, School of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Jazmin Kelly
- Robert M. Berne Cardiovascular Research Center, Division of Cardiovascular Medicine, School of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Austin Hogwood
- Robert M. Berne Cardiovascular Research Center, Division of Cardiovascular Medicine, School of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Antonio Abbate
- Robert M. Berne Cardiovascular Research Center, Division of Cardiovascular Medicine, School of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Stefano Toldo
- Robert M. Berne Cardiovascular Research Center, Division of Cardiovascular Medicine, School of Medicine, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
3
|
Liu H, Fu M, Zhang Y, You Q, Wang L. Small molecules targeting canonical transient receptor potential channels: an update. Drug Discov Today 2024; 29:103951. [PMID: 38514041 DOI: 10.1016/j.drudis.2024.103951] [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: 10/25/2023] [Revised: 03/06/2024] [Accepted: 03/14/2024] [Indexed: 03/23/2024]
Abstract
Transient receptor potential canonical (TRPC) channels belong to an important class of non-selective cation channels. This channel family consists of multiple members that widely participate in various physiological and pathological processes. Previous studies have uncovered the intricate regulation of these channels, as well as the spatial arrangement of TRPCs and the binding sites for various small molecule compounds. Multiple small molecules have been identified as selective agonists or inhibitors targeting different subtypes of TRPC, including potential preclinical drug candidates. This review covers recent advancements in the understanding of TRPC regulation and structure and the discovery of TRPC small molecules over the past few years, with the aim of facilitating research on TRPCs and small-molecule drug discovery.
Collapse
Affiliation(s)
- Hua Liu
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Min Fu
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Yifan Zhang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Qidong You
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China.
| | - Lei Wang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
4
|
Yu C, Li X, Ma J, Liang S, Zhao Y, Li Q, Zhang R. Spatiotemporal modulation of nitric oxide and Notch signaling by hemodynamic-responsive Trpv4 is essential for ventricle regeneration. Cell Mol Life Sci 2024; 81:60. [PMID: 38279064 PMCID: PMC10817848 DOI: 10.1007/s00018-023-05092-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 12/08/2023] [Accepted: 12/13/2023] [Indexed: 01/28/2024]
Abstract
Zebrafish have a remarkable ability to regenerate injured hearts. Altered hemodynamic forces after larval ventricle ablation activate the endocardial Klf2a-Notch signaling cascade to direct zebrafish cardiac regeneration. However, how the heart perceives blood flow changes and initiates signaling pathways promoting regeneration is not fully understood. The present study demonstrated that the mechanosensitive channel Trpv4 sensed the altered hemodynamic forces in injured hearts and its expression was regulated by blood flow. In addition to mediating the endocardial Klf2a-Notch signal cascade around the atrioventricular canal (AVC), we discovered that Trpv4 regulated nitric oxide (NO) signaling in the bulbus arteriosus (BA). Further experiments indicated that Notch signaling primarily acted at the early stage of regeneration, and the major role of NO signaling was at the late stage and through TGF-β pathway. Overall, our findings revealed that mechanosensitive channels perceived the changes in hemodynamics after ventricle injury, and provide novel insights into the temporal and spatial coordination of multiple signaling pathways regulating heart regeneration.
Collapse
Affiliation(s)
- Chunxiao Yu
- TaiKang Medical School, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Xueyu Li
- School of Life Sciences, Fudan University, Shanghai, 200433, China
| | - Jinmin Ma
- Medical Frontier Innovation Research Center, The First Hospital of Lanzhou University, The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, China
| | - Shuzhang Liang
- School of Life Sciences, Fudan University, Shanghai, 200433, China
| | - Yan Zhao
- TaiKang Medical School, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Qi Li
- TaiKang Medical School, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Ruilin Zhang
- TaiKang Medical School, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China.
- Institute of Myocardial Injury and Repair, Wuhan University, Wuhan, 430071, China.
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China.
| |
Collapse
|
5
|
Huang J, Korsunsky A, Yazdani M, Chen J. Targeting TRP channels: recent advances in structure, ligand binding, and molecular mechanisms. Front Mol Neurosci 2024; 16:1334370. [PMID: 38273937 PMCID: PMC10808746 DOI: 10.3389/fnmol.2023.1334370] [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: 11/07/2023] [Accepted: 12/26/2023] [Indexed: 01/27/2024] Open
Abstract
Transient receptor potential (TRP) channels are a large and diverse family of transmembrane ion channels that are widely expressed, have important physiological roles, and are associated with many human diseases. These proteins are actively pursued as promising drug targets, benefitting greatly from advances in structural and mechanistic studies of TRP channels. At the same time, the complex, polymodal activation and regulation of TRP channels have presented formidable challenges. In this short review, we summarize recent progresses toward understanding the structural basis of TRP channel function, as well as potential ligand binding sites that could be targeted for therapeutics. A particular focus is on the current understanding of the molecular mechanisms of TRP channel activation and regulation, where many fundamental questions remain unanswered. We believe that a deeper understanding of the functional mechanisms of TRP channels will be critical and likely transformative toward developing successful therapeutic strategies targeting these exciting proteins. This endeavor will require concerted efforts from computation, structural biology, medicinal chemistry, electrophysiology, pharmacology, drug safety and clinical studies.
Collapse
Affiliation(s)
- Jian Huang
- Department of Chemistry, University of Massachusetts, Amherst, MA, United States
| | - Aron Korsunsky
- Department of Chemistry, University of Massachusetts, Amherst, MA, United States
| | - Mahdieh Yazdani
- Modeling and Informatics, Merck & Co., Inc., West Point, PA, United States
| | - Jianhan Chen
- Department of Chemistry, University of Massachusetts, Amherst, MA, United States
| |
Collapse
|
6
|
Khan SU, Khan SU, Suleman M, Khan MU, Alsuhaibani AM, Refat MS, Hussain T, Ud Din MA, Saeed S. The Multifunctional TRPC6 Protein: Significance in the Field of Cardiovascular Studies. Curr Probl Cardiol 2024; 49:102112. [PMID: 37774899 DOI: 10.1016/j.cpcardiol.2023.102112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 09/22/2023] [Indexed: 10/01/2023]
Abstract
Cardiovascular disease is the leading cause of death, medical complications, and healthcare costs. Although recent advances have been in treating cardiovascular disorders linked with a reduced ejection fraction, acutely decompensate cardiac failure remains a significant medical problem. The transient receptor potential cation channel (TRPC6) family responds to neurohormonal and mechanical stress, playing critical roles in cardiovascular diseases. Therefore, TRP C6 channels have great promise as therapeutic targets. Numerous studies have investigated the roles of TRP C6 channels in pain neurons, highlighting their significance in cardiovascular research. The TRPC6 protein exhibits a broad distribution in various organs and tissues, including the brain, nerves, heart, blood vessels, lungs, kidneys, gastrointestinal tract, and other bodily structures. Its activation can be triggered by alterations in osmotic pressure, mechanical stimulation, and diacylglycerol. Consequently, TRPC6 plays a significant role in the pathophysiological mechanisms underlying diverse diseases within living organisms. A recent study has indicated a strong correlation between the disorder known as TRPC6 and the development of cardiovascular diseases. Consequently, investigations into the association between TRPC6 and cardiovascular diseases have gained significant attention in the scientific community. This review explores the most recent developments in the recognition and characterization of TRPC6. Additionally, it considers the field's prospects while examining how TRPC6 might be altered and its clinical applications.
Collapse
Affiliation(s)
- Safir Ullah Khan
- Hefei National Laboratory for Physical Sciences at the Microscale, School of Life Sciences, University of Science and Technology of China, Hefei, China.
| | - Shahid Ullah Khan
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City and Southwest University, College of Agronomy and Biotechnology, Southwest University, Chongqing, China; Department of Biochemistry, Women Medical and Dental College, Khyber Medical University, Abbottabad, Pakistan.
| | - Muhammad Suleman
- Center for Biotechnology and Microbiology, University of Swat, Swat, Pakistan
| | - Munir Ullah Khan
- Department of Polymer Science and Engineering, MOE Key Laboratory of Macromolecular Synthesis and Functionalization, International Research Center for X Polymers, Zhejiang University, Hangzhou, China
| | - Amnah Mohammed Alsuhaibani
- Department of Physical Sport Science, College of Education, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Moamen S Refat
- Department of Chemistry, College of Science, Taif University, Taif, Saudi Arabia
| | - Talib Hussain
- Women Dental College, Khyber Medical University, Abbottabad, Pakistan
| | - Muhammad Azhar Ud Din
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, P.R. China
| | - Sumbul Saeed
- School of Environment and Science, Griffith University, Nathan, QLD, Australia
| |
Collapse
|
7
|
Paris D, Palomba L, Albertini MC, Tramice A, Motta L, Giammattei E, Ambrosino P, Maniscalco M, Motta A. The biomarkers' landscape of post-COVID-19 patients can suggest selective clinical interventions. Sci Rep 2023; 13:22496. [PMID: 38110483 PMCID: PMC10728085 DOI: 10.1038/s41598-023-49601-4] [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: 12/15/2022] [Accepted: 12/10/2023] [Indexed: 12/20/2023] Open
Abstract
In COVID-19 clinical symptoms can persist even after negativization also in individuals who have had mild or moderate disease. We here investigated the biomarkers that define the post-COVID-19 clinical state analyzing the exhaled breath condensate (EBC) of 38 post COVID-19 patients and 38 sex and age-matched healthy controls via nuclear magnetic resonance (NMR)-based metabolomics. Predicted gene-modulated microRNAs (miRNAs) related to COVID-19 were quantified from EBC of 10 patients and 10 controls. Finally, clinical parameters from all post-COVID-19 patients were correlated with metabolomic data. Post-COVID-19 patients and controls showed different metabolic phenotype ("metabotype"). From the metabolites, by using enrichment analysis we identified miRNAs that resulted up-regulated (hsa-miR146a-5p) and down-regulated (hsa-miR-126-3p and hsa-miR-223-3p) in post-COVID-19. Taken together, our multiomics data indicate that post-COVID-19 patients before rehabilitation are characterized by persistent inflammation, dysregulation of liver, endovascular thrombotic and pulmonary processes, and physical impairment, which should be the primary clinical targets to contrast the post-acute sequelae of COVID-19.
Collapse
Affiliation(s)
- Debora Paris
- Institute of Biomolecular Chemistry, National Research Council, 80078, Pozzuoli (Naples), Italy
| | - Letizia Palomba
- Department of Biomolecular Sciences, "Carlo Bo" University, 61029, Urbino, Italy
| | | | - Annabella Tramice
- Institute of Biomolecular Chemistry, National Research Council, 80078, Pozzuoli (Naples), Italy
| | - Lorenzo Motta
- Neuroradiology Unit, Ospedale Santa Maria Della Misericordia, 45100, Rovigo, Italy
- IRCCS Istituto Delle Scienze Neurologiche (Padiglione G), via Altura 3, 40139, Bologna, Italy
| | - Eleonora Giammattei
- Department of Biomolecular Sciences, "Carlo Bo" University, 61029, Urbino, Italy
| | - Pasquale Ambrosino
- Directorate of Telese Terme Institute, Istituti Clinici Scientifici Maugeri IRCCS, 82037, Telese Terme (Benevento), Italy
| | - Mauro Maniscalco
- Pulmonary Rehabilitation Unit of the Telese Terme Institute, Istituti Clinici Scientifici Maugeri IRCCS, 82037, Telese Terme (Benevento), Italy.
- Department of Clinical Medicine and Surgery, Section of Respiratory Disease, University of Naples Federico II, 80131, Naples, Italy.
| | - Andrea Motta
- Institute of Biomolecular Chemistry, National Research Council, 80078, Pozzuoli (Naples), Italy.
| |
Collapse
|
8
|
Chakraborty P, Azam MA, Massé S, Lai PF, Rose RA, Ibarra Moreno CA, Riazi S, Nanthakumar K. Uncoupling cytosolic calcium from membrane voltage by transient receptor potential melastatin 4 channel (TRPM4) modulation: A novel strategy to treat ventricular arrhythmias. Heart Rhythm O2 2023; 4:725-732. [PMID: 38034891 PMCID: PMC10685170 DOI: 10.1016/j.hroo.2023.10.001] [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] [Indexed: 12/02/2023] Open
Abstract
The current antiarrhythmic paradigm is mainly centered around modulating membrane voltage. However, abnormal cytosolic calcium (Ca2+) signaling, which plays an important role in driving membrane voltage, has not been targeted for therapeutic purposes in arrhythmogenesis. There is clear evidence for bidirectional coupling between membrane voltage and intracellular Ca2+. Cytosolic Ca2+ regulates membrane voltage through Ca2+-sensitive membrane currents. As a component of Ca2+-sensitive currents, Ca2+-activated nonspecific cationic current through the TRPM4 (transient receptor potential melastatin 4) channel plays a significant role in Ca2+-driven changes in membrane electrophysiology. In myopathic and ischemic ventricles, upregulation and/or enhanced activity of this current is associated with the generation of afterdepolarization (both early and delayed), reduction of repolarization reserve, and increased propensity to ventricular arrhythmias. In this review, we describe a novel concept for the management of ventricular arrhythmias in the remodeled ventricle based on mechanistic concepts from experimental studies, by uncoupling the Ca2+-induced changes in membrane voltage by inhibition of this TRPM4-mediated current.
Collapse
Affiliation(s)
- Praloy Chakraborty
- Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
| | - Mohammed Ali Azam
- Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
| | - Stéphane Massé
- Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
| | - Patrick F.H. Lai
- Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
| | - Robert A. Rose
- Libin Cardiovascular Institute of Alberta, University of Calgary, Calgary, Alberta, Canada
| | - Carlos A. Ibarra Moreno
- Malignant Hyperthermia Investigation Unit, Department of Anesthesiology and Pain Medicine, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
| | - Sheila Riazi
- Malignant Hyperthermia Investigation Unit, Department of Anesthesiology and Pain Medicine, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
| | - Kumaraswamy Nanthakumar
- Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
| |
Collapse
|
9
|
Streiff ME, Corbin AC, Ahmad AA, Hunter C, Sachse FB. TRPC1 channels underlie stretch-modulated sarcoplasmic reticulum calcium leak in cardiomyocytes. Front Physiol 2022; 13:1056657. [PMID: 36620209 PMCID: PMC9817106 DOI: 10.3389/fphys.2022.1056657] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 12/12/2022] [Indexed: 12/25/2022] Open
Abstract
Transient receptor potential canonical 1 (TRPC1) channels are Ca2+-permeable ion channels expressed in cardiomyocytes. An involvement of TRPC1 channels in cardiac diseases is widely established. However, the physiological role of TRPC1 channels and the mechanisms through which they contribute to disease development are still under investigation. Our prior work suggested that TRPC1 forms Ca2+ leak channels located in the sarcoplasmic reticulum (SR) membrane. Prior studies suggested that TRPC1 channels in the cell membrane are mechanosensitive, but this was not yet investigated in cardiomyocytes or for SR localized TRPC1 channels. We applied adenoviral transfection to overexpress or suppress TRPC1 expression in neonatal rat ventricular myocytes (NRVMs). Transfections were evaluated with RT-qPCR, western blot, and fluorescent imaging. Single-molecule localization microscopy revealed high colocalization of exogenously expressed TRPC1 and the sarco/endoplasmic reticulum Ca2+ ATPase (SERCA2). To test our hypothesis that TRPC1 channels contribute to mechanosensitive Ca2+ SR leak, we directly measured SR Ca2+ concentration ([Ca2+]SR) using adenoviral transfection with a novel ratiometric genetically encoded SR-targeting Ca2+ sensor. We performed fluorescence imaging to quantitatively assess [Ca2+]SR and leak through TRPC1 channels of NRVMs cultured on stretchable silicone membranes. [Ca2+]SR was increased in cells with suppressed TRPC1 expression vs. control and Transient receptor potential canonical 1-overexpressing cells. We also detected a significant reduction in [Ca2+]SR in cells with Transient receptor potential canonical 1 overexpression when 10% uniaxial stretch was applied. These findings indicate that TRPC1 channels underlie the mechanosensitive modulation of [Ca2+]SR. Our findings are critical for understanding the physiological role of TRPC1 channels and support the development of pharmacological therapies for cardiac diseases.
Collapse
Affiliation(s)
- Molly E. Streiff
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, United States
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, United States
| | - Andrea C. Corbin
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, United States
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, United States
| | - Azmi A. Ahmad
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, United States
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, United States
| | - Chris Hunter
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, United States
| | - Frank B. Sachse
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, United States
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, United States
| |
Collapse
|
10
|
Szabó L, Balogh N, Tóth A, Angyal Á, Gönczi M, Csiki DM, Tóth C, Balatoni I, Jeney V, Csernoch L, Dienes B. The mechanosensitive Piezo1 channels contribute to the arterial medial calcification. Front Physiol 2022; 13:1037230. [PMID: 36439266 PMCID: PMC9685409 DOI: 10.3389/fphys.2022.1037230] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 10/20/2022] [Indexed: 07/27/2023] Open
Abstract
Vascular calcification (VC) is associated with a number of cardiovascular diseases, as well as chronic kidney disease. The role of smooth muscle cells (SMC) has already been widely explored in VC, as has the role of intracellular Ca2+ in regulating SMC function. Increased intracellular calcium concentration ([Ca2+]i) in vascular SMC has been proposed to stimulate VC. However, the contribution of the non-selective Piezo1 mechanosensitive cation channels to the elevation of [Ca2+]i, and consequently to the process of VC has never been examined. In this work the essential contribution of Piezo1 channels to arterial medial calcification is demonstrated. The presence of Piezo1 was proved on human aortic smooth muscle samples using immunohistochemistry. Quantitative PCR and Western blot analysis confirmed the expression of the channel on the human aortic smooth muscle cell line (HAoSMC). Functional measurements were done on HAoSMC under control and calcifying condition. Calcification was induced by supplementing the growth medium with inorganic phosphate (1.5 mmol/L, pH 7.4) and calcium (CaCl2, 0.6 mmol/L) for 7 days. Measurement of [Ca2+]i using fluorescent Fura-2 dye upon stimulation of Piezo1 channels (either by hypoosmolarity, or Yoda1) demonstrated significantly higher calcium transients in calcified as compared to control HAoSMCs. The expression of mechanosensitive Piezo1 channel is augmented in calcified arterial SMCs leading to a higher calcium influx upon stimulation. Activation of the channel by Yoda1 (10 μmol/L) enhanced calcification of HAoSMCs, while Dooku1, which antagonizes the effect of Yoda1, reduced this amplification. Application of Dooku1 alone inhibited the calcification. Knockdown of Piezo1 by siRNA suppressed the calcification evoked by Yoda1 under calcifying conditions. Our results demonstrate the pivotal role of Piezo1 channels in arterial medial calcification.
Collapse
Affiliation(s)
- László Szabó
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- Doctoral School of Molecular Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- ELKH-DE Cell Physiology Research Group, University of Debrecen, Debrecen, Hungary
| | - Norbert Balogh
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Andrea Tóth
- MTA-DE Lendület Vascular Pathophysiology Research Group, Research Centre for Molecular Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- Doctoral School of Molecular Cell and Immune Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Ágnes Angyal
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Mónika Gönczi
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- ELKH-DE Cell Physiology Research Group, University of Debrecen, Debrecen, Hungary
| | - Dávid Máté Csiki
- MTA-DE Lendület Vascular Pathophysiology Research Group, Research Centre for Molecular Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- Doctoral School of Molecular Cell and Immune Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Csaba Tóth
- Department of Surgery, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | | | - Viktória Jeney
- MTA-DE Lendület Vascular Pathophysiology Research Group, Research Centre for Molecular Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - László Csernoch
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- ELKH-DE Cell Physiology Research Group, University of Debrecen, Debrecen, Hungary
| | - Beatrix Dienes
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
11
|
Sabourin J, Beauvais A, Luo R, Montani D, Benitah JP, Masson B, Antigny F. The SOCE Machinery: An Unbalanced Knowledge between Left and Right Ventricular Pathophysiology. Cells 2022; 11:cells11203282. [PMID: 36291148 PMCID: PMC9600889 DOI: 10.3390/cells11203282] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 10/09/2022] [Accepted: 10/15/2022] [Indexed: 11/16/2022] Open
Abstract
Right ventricular failure (RVF) is the most important prognostic factor for morbidity and mortality in pulmonary arterial hypertension (PAH) or pulmonary hypertension (PH) caused by left heart diseases. However, right ventricle (RV) remodeling is understudied and not targeted by specific therapies. This can be partly explained by the lack of basic knowledge of RV remodeling. Since the physiology and hemodynamic function of the RV differ from those of the left ventricle (LV), the mechanisms of LV dysfunction cannot be generalized to that of the RV, albeit a knowledge of these being helpful to understanding RV remodeling and dysfunction. Store-operated Ca2+ entry (SOCE) has recently emerged to participate in the LV cardiomyocyte Ca2+ homeostasis and as a critical player in Ca2+ mishandling in a pathological context. In this paper, we highlight the current knowledge on the SOCE contribution to the LV and RV dysfunctions, as SOCE molecules are present in both compartments. he relative lack of studies on RV dysfunction indicates the necessity of further investigations, a significant challenge over the coming years.
Collapse
Affiliation(s)
- Jessica Sabourin
- Signalisation et Physiopathologie Cardiovasculaire, Inserm, Université Paris-Saclay, UMR-S 1180, 91400 Orsay, France
- Correspondence: (J.S.); (F.A.); Tel.: +(33)-180-006-302 (J.S.); +(33)-140-942-299 (F.A.)
| | - Antoine Beauvais
- Faculté de Médecine, Université Paris-Saclay, 94270 Le Kremlin-Bicêtre, France
- Hypertension Pulmonaire: Physiopathologie et Innovation Thérapeutique, Hôpital Marie Lannelongue, Université Paris-Saclay, Inserm, UMR-S 999, 92350 Le Plessis-Robinson, France
- Service de Pneumologie et Soins Intensifs Respiratoires, Centre de Référence de l’Hypertension Pulmonaire, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Bicêtre, 94270 Le Kremlin-Bicêtre, France
| | - Rui Luo
- Signalisation et Physiopathologie Cardiovasculaire, Inserm, Université Paris-Saclay, UMR-S 1180, 91400 Orsay, France
| | - David Montani
- Faculté de Médecine, Université Paris-Saclay, 94270 Le Kremlin-Bicêtre, France
- Hypertension Pulmonaire: Physiopathologie et Innovation Thérapeutique, Hôpital Marie Lannelongue, Université Paris-Saclay, Inserm, UMR-S 999, 92350 Le Plessis-Robinson, France
- Service de Pneumologie et Soins Intensifs Respiratoires, Centre de Référence de l’Hypertension Pulmonaire, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Bicêtre, 94270 Le Kremlin-Bicêtre, France
| | - Jean-Pierre Benitah
- Signalisation et Physiopathologie Cardiovasculaire, Inserm, Université Paris-Saclay, UMR-S 1180, 91400 Orsay, France
| | - Bastien Masson
- Faculté de Médecine, Université Paris-Saclay, 94270 Le Kremlin-Bicêtre, France
- Hypertension Pulmonaire: Physiopathologie et Innovation Thérapeutique, Hôpital Marie Lannelongue, Université Paris-Saclay, Inserm, UMR-S 999, 92350 Le Plessis-Robinson, France
- Service de Pneumologie et Soins Intensifs Respiratoires, Centre de Référence de l’Hypertension Pulmonaire, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Bicêtre, 94270 Le Kremlin-Bicêtre, France
| | - Fabrice Antigny
- Faculté de Médecine, Université Paris-Saclay, 94270 Le Kremlin-Bicêtre, France
- Hypertension Pulmonaire: Physiopathologie et Innovation Thérapeutique, Hôpital Marie Lannelongue, Université Paris-Saclay, Inserm, UMR-S 999, 92350 Le Plessis-Robinson, France
- Service de Pneumologie et Soins Intensifs Respiratoires, Centre de Référence de l’Hypertension Pulmonaire, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Bicêtre, 94270 Le Kremlin-Bicêtre, France
- Correspondence: (J.S.); (F.A.); Tel.: +(33)-180-006-302 (J.S.); +(33)-140-942-299 (F.A.)
| |
Collapse
|
12
|
Wei Y, Cai J, Zhu R, Xu K, Li H, Li J. Function and therapeutic potential of transient receptor potential ankyrin 1 in fibrosis. Front Pharmacol 2022; 13:1014041. [PMID: 36278189 PMCID: PMC9582847 DOI: 10.3389/fphar.2022.1014041] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 09/26/2022] [Indexed: 11/18/2022] Open
Abstract
The transient receptor potential (TRP) protein superfamily is a special group of cation channels expressed in different cell types and signaling pathways. In this review, we focus on TRPA1 (transient receptor potential ankyrin 1), an ion channel in this family that exists in the cell membrane and shows a different function from other TRP channels. TRPA1 usually has a special activation effect that can induce cation ions, especially calcium ions, to flow into activated cells. In this paper, we review the role of TRPA1 in fibroblasts. To clarify the relationship between fibroblasts and TRPA1, we have also paid special attention to the interactions between TRPA1 and inflammatory factors leading to fibroblast activation. TRPA1 has different functions in the fibrosis process in different organs, and there have also been interesting discussions of the mechanism of TRPA1 in fibroblasts. Therefore, this review aims to describe the function of TRP channels in controlling fibrosis through fibroblasts in different organ inflammatory and immune-mediated diseases. We attempt to prove that TRPA1 is a target for fibrosis. In fact, some clinical trials have already proven that TRPA1 is a potential adjuvant therapy for treating fibrosis.
Collapse
Affiliation(s)
- Yicheng Wei
- Third Affiliated Hospital of Shanghai University/Wenzhou People’s Hospital, Wenzhou, China
- Shanghai Putuo Central School of Clinical Medicine, Anhui Medical University, Hefei, Anhui, China
- Interventional Cancer Institute of Chinese Integrative Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jialuo Cai
- Interventional Cancer Institute of Chinese Integrative Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Ruiqiu Zhu
- Shanghai Putuo Central School of Clinical Medicine, Anhui Medical University, Hefei, Anhui, China
- Interventional Cancer Institute of Chinese Integrative Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ke Xu
- Musculoskeletal Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, China
- Wenzhou Institute of Shanghai University, Wenzhou, China
- *Correspondence: Ke Xu, , ; Hongchang Li, ; Jianxin Li,
| | - Hongchang Li
- Department of General Surgery, Institute of Fudan–Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, China
- *Correspondence: Ke Xu, , ; Hongchang Li, ; Jianxin Li,
| | - Jianxin Li
- Third Affiliated Hospital of Shanghai University/Wenzhou People’s Hospital, Wenzhou, China
- *Correspondence: Ke Xu, , ; Hongchang Li, ; Jianxin Li,
| |
Collapse
|
13
|
Ding K, Gui Y, Hou X, Ye L, Wang L. Transient Receptor Potential Channels, Natriuretic Peptides, and Angiotensin Receptor-Neprilysin Inhibitors in Patients With Heart Failure. Front Cardiovasc Med 2022; 9:904881. [PMID: 35722101 PMCID: PMC9204593 DOI: 10.3389/fcvm.2022.904881] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Accepted: 04/26/2022] [Indexed: 12/11/2022] Open
Abstract
Heart failure (HF) remains the leading cause of death, morbidity, and medical expenses worldwide. Treatments for HF with reduced ejection fraction have progressed in recent years; however, acute decompensated heart failure remains difficult to treat. The transient receptor potential (TRP) channel family plays roles in various cardiovascular diseases, responding to neurohormonal and mechanical load stimulation. Thus, TRP channels are promising targets for drug discovery, and many studies have evaluated the roles of TRP channels expressed on pain neurons. The natriuretic peptide (NP) family of proteins regulates blood volume, natriuresis, and vasodilation and can antagonize the renin-angiotensin-aldosterone system and participate in the pathogenesis of major cardiovascular diseases, such as HF, coronary atherosclerotic heart disease, and left ventricular hypertrophy. NPs are degraded by neprilysin, and the blood level of NPs has predictive value in the diagnosis and prognostic stratification of HF. In this review, we discuss the relationships between typical TRP family channels (e.g., transient receptor potential cation channel subfamily V member 1 andTRPV1, transient receptor potential cation channel subfamily C member 6) and the NP system (e.g., atrial NP, B-type NP, and C-type NP) and their respective roles in HF. We also discuss novel drugs introduced for the treatment of HF.
Collapse
Affiliation(s)
- Kun Ding
- Bengbu Medical College, Bengbu, China
- Zhejiang Provincial People’s Hospital, Hangzhou, China
| | - Yang Gui
- Bengbu Medical College, Bengbu, China
- Zhejiang Provincial People’s Hospital, Hangzhou, China
| | - Xu Hou
- Bengbu Medical College, Bengbu, China
- Zhejiang Provincial People’s Hospital, Hangzhou, China
| | - Lifang Ye
- Zhejiang Provincial People’s Hospital, Hangzhou, China
| | - Lihong Wang
- Zhejiang Provincial People’s Hospital, Hangzhou, China
| |
Collapse
|
14
|
Fallah HP, Ahuja E, Lin H, Qi J, He Q, Gao S, An H, Zhang J, Xie Y, Liang D. A Review on the Role of TRP Channels and Their Potential as Drug Targets_An Insight Into the TRP Channel Drug Discovery Methodologies. Front Pharmacol 2022; 13:914499. [PMID: 35685622 PMCID: PMC9170958 DOI: 10.3389/fphar.2022.914499] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 04/27/2022] [Indexed: 01/13/2023] Open
Abstract
Transient receptor potential (TRP) proteins are a large group of ion channels that control many physiological functions in our body. These channels are considered potential therapeutic drug targets for various diseases such as neurological disorders, cancers, cardiovascular disease, and many more. The Nobel Prize in Physiology/Medicine in the year 2021 was awarded to two scientists for the discovery of TRP and PIEZO ion channels. Improving our knowledge of technologies for their study is essential. In the present study, we reviewed the role of TRP channel types in the control of normal physiological functions as well as disease conditions. Also, we discussed the current and novel technologies that can be used to study these channels successfully. As such, Flux assays for detecting ionic flux through ion channels are among the core and widely used tools for screening drug compounds. Technologies based on these assays are available in fully automated high throughput set-ups and help detect changes in radiolabeled or non-radiolabeled ionic flux. Aurora's Ion Channel Reader (ICR), which works based on label-free technology of flux assay, offers sensitive, accurate, and reproducible measurements to perform drug ranking matching with patch-clamp (gold standard) data. The non-radiolabeled trace-based flux assay coupled with the ICR detects changes in various ion types, including potassium, calcium, sodium, and chloride channels, by using appropriate tracer ions. This technology is now considered one of the very successful approaches for analyzing ion channel activity in modern drug discovery. It could be a successful approach for studying various ion channels and transporters, including the different members of the TRP family of ion channels.
Collapse
Affiliation(s)
| | - Ekta Ahuja
- Aurora Biomed Inc., Vancouver, BC, Canada
| | | | - Jinlong Qi
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, China
| | - Qian He
- Aurora Discovery Inc., Foshan, China
| | - Shan Gao
- Aurora Discovery Inc., Foshan, China
| | | | | | | | - Dong Liang
- Aurora Biomed Inc., Vancouver, BC, Canada
- Aurora Discovery Inc., Foshan, China
- Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| |
Collapse
|
15
|
Jacobs T, Abdinghoff J, Tschernig T. Protein detection and localization of the non-selective cation channel TRPC6 in the human heart. Eur J Pharmacol 2022; 924:174972. [PMID: 35483666 DOI: 10.1016/j.ejphar.2022.174972] [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: 02/09/2022] [Revised: 03/28/2022] [Accepted: 04/19/2022] [Indexed: 11/03/2022]
Abstract
Due to longer lifespans in societies in industrialized countries, cardiovascular diseases are becoming increasingly important for medical research. It has already been shown that the cell membrane-bound, non-selective TRPC6 ion channel is important in the pathogenesis of heart diseases. Among other things, it is permeable to calcium ion, which plays a critical role in cardiac contraction and relaxation. The TRPC6 ion channel is a promising therapeutic target in the treatment of cardiovascular diseases. A deeper understanding of the physiological and pathophysiological role as well as the localization of TRPC6 in human cardiac tissue is the basis for new drug development. Although the TRPC6 channel has been detected in animal studies, at the mRNA level in humans, and sparse TRPC6 protein has been detected in humans, there are no systematic studies of TRPC6 protein detection in the human heart. For the first time, TRPC6 ion channel protein was detected histologically in human heart tissue from body donors in different structures, localizations, and histological layers - particularly in cardiomyocytes and intramuscular arterioles - by immunohistochemistry, just as TRPC6 expression has already been shown in animal models of the heart by other research groups. In the sense of the translational concept, this indicates a possible transferability of research results from animal models to humans.
Collapse
Affiliation(s)
- Tobias Jacobs
- Institute of Anatomy and Cell Biology, Saarland University, Medical Campus, Homburg, Saar, Germany
| | - Jan Abdinghoff
- Institute of Anatomy and Cell Biology, Saarland University, Medical Campus, Homburg, Saar, Germany
| | - Thomas Tschernig
- Institute of Anatomy and Cell Biology, Saarland University, Medical Campus, Homburg, Saar, Germany.
| |
Collapse
|
16
|
Murakami M, Murakami AM, Nemoto T, Ohba T, Yonekura M, Toyama Y, Tomita H, Matsuzaki Y, Sawamura D, Hirota K, Itagaki S, Asada Y, Miyoshi I. Enhanced β-adrenergic response in mice with dominant-negative expression of the PKD2L1 channel. PLoS One 2022; 17:e0261668. [PMID: 35051185 PMCID: PMC8775249 DOI: 10.1371/journal.pone.0261668] [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: 03/18/2021] [Accepted: 12/08/2021] [Indexed: 11/19/2022] Open
Abstract
Polycystic kidney disease (PKD) is the most common genetic cause of kidney failure in humans. Among the various PKD-related molecules, PKD2L1 forms cation channels, but its physiological importance is obscure. In the present study, we established a transgenic mouse line by overexpressing the dominant-negative form of the mouse PKD2L1 gene (i.e., lacking the pore-forming domain). The resulting PKD2L1del-Tg mice exhibited supraventricular premature contraction, as well as enhanced sensitivity to β-adrenergic stimulation and unstable R-R intervals in electrocardiography. During spontaneous atrial contraction, PKD2L1del-Tg atria showed enhanced sensitivity to isoproterenol, norepinephrine, and epinephrine. Action potential recording revealed a shortened action potential duration in PKD2L1del-Tg atria in response to isoproterenol. These findings indicated increased adrenergic sensitivity in PKD2L1del-Tg mice, suggesting that PKD2L1 is involved in sympathetic regulation.
Collapse
Affiliation(s)
- Manabu Murakami
- Department of Pharmacology, Faculty of Medicine, University of Miyazaki, Miyazaki, Miyazaki, Japan
| | - Agnieszka M. Murakami
- Department of Pharmacology, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Takayuki Nemoto
- Department of Pharmacology, Faculty of Medicine, University of Miyazaki, Miyazaki, Miyazaki, Japan
| | - Takayoshi Ohba
- Department of Cell Physiology, Akita University School of Medicine, Akita, Akita, Japan
| | - Manabu Yonekura
- Department of Cardiology and Nephrology, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Yuichi Toyama
- Department of Cardiology and Nephrology, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Hirofumi Tomita
- Department of Cardiology and Nephrology, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Yasushi Matsuzaki
- Department of Dermatology, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Daisuke Sawamura
- Department of Dermatology, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Kazuyoshi Hirota
- Department of Anesthesiology, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Shirou Itagaki
- Collaboration Center for Community and Industry, Sapporo Medical University, Sapporo, Hokkaido, Japan
| | - Yujiro Asada
- Division of Pathophysiology, Department of Pathology, Faculty of Medicine, University of Miyazaki, Miyazaki, Miyazaki, Japan
| | - Ichiro Miyoshi
- Department of Laboratory Animal Medicine, Tohoku University School of Medicine, Sendai, Miyagi, Japan
| |
Collapse
|
17
|
TRPV4-dependent signaling mechanisms in systemic and pulmonary vasculature. CURRENT TOPICS IN MEMBRANES 2022; 89:1-41. [DOI: 10.1016/bs.ctm.2022.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
18
|
Onyali VC, Domeier TL. Cardiac TRPV4 channels. CURRENT TOPICS IN MEMBRANES 2022; 89:63-74. [DOI: 10.1016/bs.ctm.2022.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
19
|
Pharmacological Modulation and (Patho)Physiological Roles of TRPM4 Channel-Part 2: TRPM4 in Health and Disease. Pharmaceuticals (Basel) 2021; 15:ph15010040. [PMID: 35056097 PMCID: PMC8779181 DOI: 10.3390/ph15010040] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 12/21/2021] [Accepted: 12/22/2021] [Indexed: 02/06/2023] Open
Abstract
Transient receptor potential melastatin 4 (TRPM4) is a unique member of the TRPM protein family and, similarly to TRPM5, is Ca2+ sensitive and permeable for monovalent but not divalent cations. It is widely expressed in many organs and is involved in several functions; it regulates membrane potential and Ca2+ homeostasis in both excitable and non-excitable cells. This part of the review discusses the currently available knowledge about the physiological and pathophysiological roles of TRPM4 in various tissues. These include the physiological functions of TRPM4 in the cells of the Langerhans islets of the pancreas, in various immune functions, in the regulation of vascular tone, in respiratory and other neuronal activities, in chemosensation, and in renal and cardiac physiology. TRPM4 contributes to pathological conditions such as overactive bladder, endothelial dysfunction, various types of malignant diseases and central nervous system conditions including stroke and injuries as well as in cardiac conditions such as arrhythmias, hypertrophy, and ischemia-reperfusion injuries. TRPM4 claims more and more attention and is likely to be the topic of research in the future.
Collapse
|
20
|
Husti Z, Varró A, Baczkó I. Arrhythmogenic Remodeling in the Failing Heart. Cells 2021; 10:cells10113203. [PMID: 34831426 PMCID: PMC8623396 DOI: 10.3390/cells10113203] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 11/05/2021] [Accepted: 11/11/2021] [Indexed: 12/12/2022] Open
Abstract
Chronic heart failure is a clinical syndrome with multiple etiologies, associated with significant morbidity and mortality. Cardiac arrhythmias, including ventricular tachyarrhythmias and atrial fibrillation, are common in heart failure. A number of cardiac diseases including heart failure alter the expression and regulation of ion channels and transporters leading to arrhythmogenic electrical remodeling. Myocardial hypertrophy, fibrosis and scar formation are key elements of arrhythmogenic structural remodeling in heart failure. In this article, the mechanisms responsible for increased arrhythmia susceptibility as well as the underlying changes in ion channel, transporter expression and function as well as alterations in calcium handling in heart failure are discussed. Understanding the mechanisms of arrhythmogenic remodeling is key to improving arrhythmia management and the prevention of sudden cardiac death in patients with heart failure.
Collapse
Affiliation(s)
- Zoltán Husti
- Department of Pharmacology and Pharmacotherapy, University of Szeged, 6720 Szeged, Hungary; (Z.H.); (A.V.)
- Department of Pharmacology and Pharmacotherapy, Interdisciplinary Excellence Centre, University of Szeged, 6720 Szeged, Hungary
| | - András Varró
- Department of Pharmacology and Pharmacotherapy, University of Szeged, 6720 Szeged, Hungary; (Z.H.); (A.V.)
- Department of Pharmacology and Pharmacotherapy, Interdisciplinary Excellence Centre, University of Szeged, 6720 Szeged, Hungary
- ELKH-SZTE Research Group for Cardiovascular Pharmacology, Eötvös Loránd Research Network, 6720 Szeged, Hungary
| | - István Baczkó
- Department of Pharmacology and Pharmacotherapy, University of Szeged, 6720 Szeged, Hungary; (Z.H.); (A.V.)
- Department of Pharmacology and Pharmacotherapy, Interdisciplinary Excellence Centre, University of Szeged, 6720 Szeged, Hungary
- Correspondence:
| |
Collapse
|
21
|
Barbeau S, Gilbert G, Cardouat G, Baudrimont I, Freund-Michel V, Guibert C, Marthan R, Vacher P, Quignard JF, Ducret T. Mechanosensitivity in Pulmonary Circulation: Pathophysiological Relevance of Stretch-Activated Channels in Pulmonary Hypertension. Biomolecules 2021; 11:biom11091389. [PMID: 34572602 PMCID: PMC8470538 DOI: 10.3390/biom11091389] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 09/17/2021] [Accepted: 09/19/2021] [Indexed: 01/03/2023] Open
Abstract
A variety of cell types in pulmonary arteries (endothelial cells, fibroblasts, and smooth muscle cells) are continuously exposed to mechanical stimulations such as shear stress and pulsatile blood pressure, which are altered under conditions of pulmonary hypertension (PH). Most functions of such vascular cells (e.g., contraction, migration, proliferation, production of extracellular matrix proteins, etc.) depend on a key event, i.e., the increase in intracellular calcium concentration ([Ca2+]i) which results from an influx of extracellular Ca2+ and/or a release of intracellular stored Ca2+. Calcium entry from the extracellular space is a major step in the elevation of [Ca2+]i, involving a variety of plasmalemmal Ca2+ channels including the superfamily of stretch-activated channels (SAC). A common characteristic of SAC is that their gating depends on membrane stretch. In general, SAC are non-selective Ca2+-permeable cation channels, including proteins of the TRP (Transient Receptor Potential) and Piezo channel superfamily. As membrane mechano-transducers, SAC convert physical forces into biological signals and hence into a cell response. Consequently, SAC play a major role in pulmonary arterial calcium homeostasis and, thus, appear as potential novel drug targets for a better management of PH.
Collapse
Affiliation(s)
- Solène Barbeau
- Centre de Recherche Cardio-Thoracique de Bordeaux, Univ. Bordeaux, U1045, F-33600 Pessac, France; (S.B.); (G.C.); (I.B.); (V.F.-M.); (C.G.); (R.M.); (P.V.); (J.-F.Q.)
- INSERM, Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, F-33600 Pessac, France
| | - Guillaume Gilbert
- ORPHY, UFR Sciences et Techniques, University of Brest, EA 4324, F-29238 Brest, France;
| | - Guillaume Cardouat
- Centre de Recherche Cardio-Thoracique de Bordeaux, Univ. Bordeaux, U1045, F-33600 Pessac, France; (S.B.); (G.C.); (I.B.); (V.F.-M.); (C.G.); (R.M.); (P.V.); (J.-F.Q.)
- INSERM, Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, F-33600 Pessac, France
| | - Isabelle Baudrimont
- Centre de Recherche Cardio-Thoracique de Bordeaux, Univ. Bordeaux, U1045, F-33600 Pessac, France; (S.B.); (G.C.); (I.B.); (V.F.-M.); (C.G.); (R.M.); (P.V.); (J.-F.Q.)
- INSERM, Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, F-33600 Pessac, France
| | - Véronique Freund-Michel
- Centre de Recherche Cardio-Thoracique de Bordeaux, Univ. Bordeaux, U1045, F-33600 Pessac, France; (S.B.); (G.C.); (I.B.); (V.F.-M.); (C.G.); (R.M.); (P.V.); (J.-F.Q.)
- INSERM, Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, F-33600 Pessac, France
| | - Christelle Guibert
- Centre de Recherche Cardio-Thoracique de Bordeaux, Univ. Bordeaux, U1045, F-33600 Pessac, France; (S.B.); (G.C.); (I.B.); (V.F.-M.); (C.G.); (R.M.); (P.V.); (J.-F.Q.)
- INSERM, Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, F-33600 Pessac, France
| | - Roger Marthan
- Centre de Recherche Cardio-Thoracique de Bordeaux, Univ. Bordeaux, U1045, F-33600 Pessac, France; (S.B.); (G.C.); (I.B.); (V.F.-M.); (C.G.); (R.M.); (P.V.); (J.-F.Q.)
- INSERM, Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, F-33600 Pessac, France
| | - Pierre Vacher
- Centre de Recherche Cardio-Thoracique de Bordeaux, Univ. Bordeaux, U1045, F-33600 Pessac, France; (S.B.); (G.C.); (I.B.); (V.F.-M.); (C.G.); (R.M.); (P.V.); (J.-F.Q.)
- INSERM, Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, F-33600 Pessac, France
| | - Jean-François Quignard
- Centre de Recherche Cardio-Thoracique de Bordeaux, Univ. Bordeaux, U1045, F-33600 Pessac, France; (S.B.); (G.C.); (I.B.); (V.F.-M.); (C.G.); (R.M.); (P.V.); (J.-F.Q.)
- INSERM, Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, F-33600 Pessac, France
| | - Thomas Ducret
- Centre de Recherche Cardio-Thoracique de Bordeaux, Univ. Bordeaux, U1045, F-33600 Pessac, France; (S.B.); (G.C.); (I.B.); (V.F.-M.); (C.G.); (R.M.); (P.V.); (J.-F.Q.)
- INSERM, Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, F-33600 Pessac, France
- Correspondence:
| |
Collapse
|
22
|
Wen J, Bo T, Zhao Z, Wang D. Role of transient receptor potential vanilloid-1 in behavioral thermoregulation of the Mongolian gerbil Meriones unguiculatus. Integr Zool 2021; 17:608-618. [PMID: 34498418 DOI: 10.1111/1749-4877.12587] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Ambient temperature considerably affects the physiology and behavior of mammals. Thermosensory and thermoregulatory abilities play an important role in the response to changing ambient temperature in endotherms. However, the molecular mechanisms of behavioral thermoregulation remain poorly understood. Transient receptor potential vanilloid-1 (TRPV1) is activated by changes in ambient temperature and is involved in acute thermoregulation. Here, we aimed to determine whether TRPV1 is involved in behavioral thermoregulation in wild rodents by conducting 2 experiments. In the first, 42 adult Mongolian gerbils (Meriones unguiculatus; 14 per treatment) were randomly assigned to 3 housing temperatures (4, 23, and 36°C) for 4 weeks. In the second, 20 gerbils (10 per treatment) were randomly injected with capsaicin (TRPV1 agonist) or AMG517 (TRPV1 antagonist). The results showed a significant decrease in food intake and non-shivering thermogenesis in the gerbils housed at 36°C. Additionally, there was a significant increase in the preference of gerbils housed at 4°C to low temperatures. The expression of TRPV1 protein in the brown adipose tissue (BAT) and liver was significantly positively correlated with that of protein kinase A (PKA). The expression of TRPV1 and PKA proteins in the BAT was positively correlated with the temperature preference of the gerbils. The gerbils injected with capsaicin preferred significantly lower temperatures than the control group gerbils. These findings suggest that TRPV1 and PKA are involved in behavioral thermoregulation in Mongolian gerbils.
Collapse
Affiliation(s)
- Jing Wen
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,School of Life and Environmental Sciences, Wenzhou University, Wenzhou, China
| | - Tingbei Bo
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, China
| | - Zhijun Zhao
- School of Life and Environmental Sciences, Wenzhou University, Wenzhou, China
| | - Dehua Wang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, China.,School of Life Sciences, Shandong University, Qingdao, China
| |
Collapse
|
23
|
Guo Y, Yu ZY, Wu J, Gong H, Kesteven S, Iismaa SE, Chan AY, Holman S, Pinto S, Pironet A, Cox CD, Graham RM, Vennekens R, Feneley MP, Martinac B. The Ca 2+-activated cation channel TRPM4 is a positive regulator of pressure overload-induced cardiac hypertrophy. eLife 2021; 10:66582. [PMID: 34190686 PMCID: PMC8245133 DOI: 10.7554/elife.66582] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 06/20/2021] [Indexed: 01/19/2023] Open
Abstract
Pathological left ventricular hypertrophy (LVH) occurs in response to pressure overload and remains the single most important clinical predictor of cardiac mortality. The molecular pathways in the induction of pressure overload LVH are potential targets for therapeutic intervention. Current treatments aim to remove the pressure overload stimulus for LVH, but do not completely reverse adverse cardiac remodelling. Although numerous molecular signalling steps in the induction of LVH have been identified, the initial step by which mechanical stretch associated with cardiac pressure overload is converted into a chemical signal that initiates hypertrophic signalling remains unresolved. In this study, we show that selective deletion of transient receptor potential melastatin 4 (TRPM4) channels in mouse cardiomyocytes results in an approximately 50% reduction in the LVH induced by transverse aortic constriction. Our results suggest that TRPM4 channel is an important component of the mechanosensory signalling pathway that induces LVH in response to pressure overload and represents a potential novel therapeutic target for the prevention of pathological LVH.
Collapse
Affiliation(s)
- Yang Guo
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Sydney, Australia.,Cardiac Physiology and Transplantation Division, Victor Chang Cardiac Research Institute, Sydney, Australia.,St Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - Ze-Yan Yu
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Sydney, Australia.,Cardiac Physiology and Transplantation Division, Victor Chang Cardiac Research Institute, Sydney, Australia.,St Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - Jianxin Wu
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Sydney, Australia
| | - Hutao Gong
- Cardiac Physiology and Transplantation Division, Victor Chang Cardiac Research Institute, Sydney, Australia
| | - Scott Kesteven
- Cardiac Physiology and Transplantation Division, Victor Chang Cardiac Research Institute, Sydney, Australia.,St Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - Siiri E Iismaa
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Sydney, Australia.,St Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - Andrea Y Chan
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Sydney, Australia
| | - Sara Holman
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Sydney, Australia
| | - Silvia Pinto
- Laboratory of Ion Channel Research, Department of Molecular and Cellular Medicine, Katholieke Universiteit Leuven, Leuven, Belgium.,TRP Research Platform Leuven (TRPLe), Katholieke Universiteit Leuven, Leuven, Belgium
| | - Andy Pironet
- Laboratory of Ion Channel Research, Department of Molecular and Cellular Medicine, Katholieke Universiteit Leuven, Leuven, Belgium.,TRP Research Platform Leuven (TRPLe), Katholieke Universiteit Leuven, Leuven, Belgium
| | - Charles D Cox
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Sydney, Australia.,St Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - Robert M Graham
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Sydney, Australia.,St Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - Rudi Vennekens
- Laboratory of Ion Channel Research, Department of Molecular and Cellular Medicine, Katholieke Universiteit Leuven, Leuven, Belgium.,TRP Research Platform Leuven (TRPLe), Katholieke Universiteit Leuven, Leuven, Belgium
| | - Michael P Feneley
- Cardiac Physiology and Transplantation Division, Victor Chang Cardiac Research Institute, Sydney, Australia.,St Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, Australia.,Department of Cardiology, St Vincent's Hospital, Sydney, Australia
| | - Boris Martinac
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Sydney, Australia.,St Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, Australia
| |
Collapse
|
24
|
Jaffal SM, Abbas MA. TRP channels in COVID-19 disease: Potential targets for prevention and treatment. Chem Biol Interact 2021; 345:109567. [PMID: 34166652 PMCID: PMC8217345 DOI: 10.1016/j.cbi.2021.109567] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 06/15/2021] [Accepted: 06/21/2021] [Indexed: 01/05/2023]
Abstract
Coronavirus disease 2019 [COVID-19] is a global health threat caused by severe acute respiratory syndrome coronavirus 2 [SARS-CoV2] that requires two proteins for entry: angiotensin-converting enzyme 2 [ACE2] and -membrane protease serine 2 [TMPRSS2]. Many patients complain from pneumonia, cough, fever, and gastrointestinal (GI) problems. Notably, different TRP channels are expressed in various tissues infected by SARS-CoV-2. TRP channels are cation channels that show a common architecture with high permeability to calcium [Ca2+] in most sub-families. Literature review shed light on the possible role of TRP channels in COVID-19 disease. TRP channels may take part in inflammation, pain, fever, anosmia, ageusia, respiratory, cardiovascular, GI and neurological complications related to COVID-19. Also, TRP channels could be the targets for many active compounds that showed effectiveness against SARS-CoV-2. Desensitization or blocking TRP channels by antibodies, aptamers, small molecules or venoms can be an option for COVID-19 prevention and future treatment. This review provides insights into the involvement of TRP channels in different symptoms and mechanisms of SARS-CoV-2 , potential treatments targeting these channels and highlights missing gaps in literature.
Collapse
Affiliation(s)
- Sahar M Jaffal
- Department of Biological Sciences, Faculty of Science, The University of Jordan, 11942, Amman, Jordan.
| | - Manal A Abbas
- Department of Medical Laboratory Sciences, Faculty of Allied Medical Sciences, Al-Ahliyya Amman University, 19328, Amman, Jordan; Pharmacological and Diagnostic Research Center, Al-Ahliyya Amman University, 19328, Amman, Jordan
| |
Collapse
|
25
|
Canonical transient receptor potential channels and their modulators: biology, pharmacology and therapeutic potentials. Arch Pharm Res 2021; 44:354-377. [PMID: 33763843 PMCID: PMC7989688 DOI: 10.1007/s12272-021-01319-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Accepted: 03/14/2021] [Indexed: 12/17/2022]
Abstract
Canonical transient receptor potential channels (TRPCs) are nonselective, high calcium permeability cationic channels. The TRPCs family includes TRPC1, TRPC2, TRPC3, TRPC4, TRPC5, TRPC6, and TRPC7. These channels are widely expressed in the cardiovascular and nervous systems and exist in many other human tissues and cell types, playing several crucial roles in the human physiological and pathological processes. Hence, the emergence of TRPCs modulators can help investigate these channels’ applications in health and disease. It is worth noting that the TRPCs subfamilies have structural and functional similarities, which presents a significant difficulty in screening and discovering of TRPCs modulators. In the past few years, only a limited number of selective modulators of TRPCs were detected; thus, additional research on more potent and more selective TRPCs modulators is needed. The present review focuses on the striking desired therapeutic effects of TRPCs modulators, which provides intel on the structural modification of TRPCs modulators and further pharmacological research. Importantly, TRPCs modulators can significantly facilitate future studies of TRPCs and TRPCs related diseases.
Collapse
|
26
|
Beddek K, Raffin F, Borgel D, Saller F, Riccobono D, Bobe R, Boittin F. TRPV4 channel activation induces the transition of venous and arterial endothelial cells toward a pro-inflammatory phenotype. Physiol Rep 2021; 9:e14613. [PMID: 33512067 PMCID: PMC7845413 DOI: 10.14814/phy2.14613] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 09/20/2020] [Accepted: 09/22/2020] [Indexed: 11/24/2022] Open
Abstract
The Transient Receptor Potential Vanilloid 4 (TRPV4) of endothelial cells contributes to many important functions including the regulation of Ca2+ homeostasis, cell volume, endothelial barrier permeability, and smooth muscle tone. However, its role in the transition of endothelial cells toward a pro-inflammatory phenotype has not been studied so far. Using both arterial and venous endothelial cells, we first show that the pharmacological activation of TRPV4 channels with GSK1016790A, a potent TRPV4 agonist, triggers robust and sustained Ca2+ increases, which are blocked by both TRPV4 antagonists HC067047 and RN9893. TRPV4 activation also triggers the actin cytoskeleton and adherens junction (VE-Cadherin) rearrangement in both arterial and venous endothelial cells and leads to rapid decreases of trans-endothelial electrical resistance. In addition to its effect on endothelial barrier integrity, TRPV4 activation selectively increases ICAM-1 surface expression in arterial and venous endothelial cells, due to the stimulation of ICAM-1 gene expression through the NF-κB transcription factor. TRPV4 channel activation also induced apoptosis of venous and arterial endothelial cells, while TRPV4 blockade reduced apoptosis, even in the absence of TRPV4 activation. As altered barrier integrity, increased adhesion molecule expression and apoptosis are hallmarks of the pro-inflammatory state of endothelial cells, our results indicate that TRPV4 channel activity can induce the transition of both venous and arterial endothelial cells toward a pro-inflammatory phenotype.
Collapse
Affiliation(s)
- Kathia Beddek
- INSERM Unité Mixte de Recherche‐Santé 1176Université Paris‐SudUniversité Paris‐SaclayLe Kremlin‐BicêtreFrance
| | - Florent Raffin
- Département des Plateformesunité Analyses BiologiquesIRBA (Institut de Recherche Biomédicale des Armées)Brétigny‐sur‐OrgeFrance
| | - Delphine Borgel
- INSERM Unité Mixte de Recherche‐Santé 1176Université Paris‐SudUniversité Paris‐SaclayLe Kremlin‐BicêtreFrance
| | - François Saller
- INSERM Unité Mixte de Recherche‐Santé 1176Université Paris‐SudUniversité Paris‐SaclayLe Kremlin‐BicêtreFrance
| | - Diane Riccobono
- Département Effets Biologiques des Rayonnementsunité de RadiobiologieIRBA (Institut de Recherche Biomédicale des Armées)Brétigny‐sur‐OrgeFrance
| | - Régis Bobe
- INSERM Unité Mixte de Recherche‐Santé 1176Université Paris‐SudUniversité Paris‐SaclayLe Kremlin‐BicêtreFrance
| | - François‐Xavier Boittin
- Département Effets Biologiques des Rayonnementsunité de RadiobiologieIRBA (Institut de Recherche Biomédicale des Armées)Brétigny‐sur‐OrgeFrance
| |
Collapse
|
27
|
Shamsaldeen YA, Lione LA, Benham CD. Dysregulation of TRPV4, eNOS and caveolin-1 contribute to endothelial dysfunction in the streptozotocin rat model of diabetes. Eur J Pharmacol 2020; 888:173441. [PMID: 32810492 DOI: 10.1016/j.ejphar.2020.173441] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 07/28/2020] [Accepted: 07/29/2020] [Indexed: 12/30/2022]
Abstract
Endothelial dysfunction is a common complication in diabetes in which endothelium-dependent vasorelaxation is impaired. The aim of this study was to examine the involvement of the TRPV4 ion channel in type 1 diabetic endothelial dysfunction and the possible association of endothelial dysfunction with reduced expression of TRPV4, endothelial nitric oxide synthase (eNOS) and caveolin-1. Male Wistar rats (350-450 g) were injected with 65 mg/kg i.p. streptozotocin (STZ) or vehicle. Endothelial function was investigated in aortic rings and mesenteric arteries using organ bath and myograph, respectively. TRPV4 function was studied with fura-2 calcium imaging in endothelial cells cultured from aortas from control and STZ treated rats. TRPV4, caveolin-1 and eNOS expression was investigated in these cells using immunohistochemistry. STZ-treated diabetic rats showed significant endothelial dysfunction characterised by impaired muscarinic-induced vasorelaxation (aortic rings: STZ-diabetics: Emax = 29.6 ± 9.3%; control: Emax = 77.2 ± 2.5% P˂0.001), as well as significant impairment in TRPV4-induced vasorelaxation (aortic rings, 4αPDD STZ-diabetics: Emax = 56.0 ± 5.5%; control: Emax = 81.1 ± 2.1% P˂0.001). Furthermore, STZ-diabetic primary aortic endothelial cells showed a significant reduction in TRPV4-induced intracellular calcium elevation (P˂0.05) compared with the control group. This was associated with significantly lower expression of TRPV4, caveolin-1 and eNOS and this was reversed by insulin treatment of the endothelial cultures from STZ -diabetic rats. Taken together, these data are consistent with the hypothesis that signalling through TRPV4, caveolin-1, and eNOS is downregulated in STZ-diabetic aortic endothelial cells and restored by insulin treatment.
Collapse
Affiliation(s)
- Yousif A Shamsaldeen
- School of Life and Medical Sciences, University of Hertfordshire, College Lane, Hatfield, Hertfordshire, AL10 9AB, UK; Department of Pharmacy, Kuwait Hospital, Sabah Alsalem, 44001, Kuwait.
| | - Lisa A Lione
- School of Life and Medical Sciences, University of Hertfordshire, College Lane, Hatfield, Hertfordshire, AL10 9AB, UK
| | - Christopher D Benham
- School of Life and Medical Sciences, University of Hertfordshire, College Lane, Hatfield, Hertfordshire, AL10 9AB, UK
| |
Collapse
|
28
|
Genova T, Gaglioti D, Munaron L. Regulation of Vessel Permeability by TRP Channels. Front Physiol 2020; 11:421. [PMID: 32431625 PMCID: PMC7214926 DOI: 10.3389/fphys.2020.00421] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 04/07/2020] [Indexed: 12/13/2022] Open
Abstract
The vascular endothelium constitutes a semi-permeable barrier between blood and interstitial fluids. Since an augmented endothelial permeability is often associated to pathological states, understanding the molecular basis for its regulation is a crucial biomedical and clinical challenge. This review focuses on the processes controlling paracellular permeability that is the permeation of fluids between adjacent endothelial cells (ECs). Cytosolic calcium changes are often detected as early events preceding the alteration of the endothelial barrier (EB) function. For this reason, great interest has been devoted in the last decades to unveil the molecular mechanisms underlying calcium fluxes and their functional relationship with vessel permeability. Beyond the dicotomic classification between store-dependent and independent calcium entry at the plasma membrane level, the search for the molecular components of the related calcium-permeable channels revealed a difficult task for intrinsic and technical limitations. The contribution of redundant channel-forming proteins including members of TRP superfamily and Orai1, together with the very complex intracellular modulatory pathways, displays a huge variability among tissues and along the vascular tree. Moreover, calcium-independent events could significantly concur to the regulation of vascular permeability in an intricate and fascinating multifactorial framework.
Collapse
Affiliation(s)
- Tullio Genova
- Department of Life Sciences and Systems Biology, University of Turin, Turin, Italy
| | - Deborah Gaglioti
- Department of Life Sciences and Systems Biology, University of Turin, Turin, Italy
| | - Luca Munaron
- Department of Life Sciences and Systems Biology, University of Turin, Turin, Italy
| |
Collapse
|
29
|
Sutanto H, Laudy L, Clerx M, Dobrev D, Crijns HJ, Heijman J. Maastricht antiarrhythmic drug evaluator (MANTA): A computational tool for better understanding of antiarrhythmic drugs. Pharmacol Res 2019; 148:104444. [DOI: 10.1016/j.phrs.2019.104444] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 07/10/2019] [Accepted: 09/03/2019] [Indexed: 12/14/2022]
|
30
|
Smani T, Gallardo-Castillo I, Ávila-Médina J, Jimenez-Navarro MF, Ordoñez A, Hmadcha A. Impact of Diabetes on Cardiac and Vascular Disease: Role of Calcium Signaling. Curr Med Chem 2019; 26:4166-4177. [DOI: 10.2174/0929867324666170523140925] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Revised: 02/14/2017] [Accepted: 05/04/2017] [Indexed: 12/12/2022]
Abstract
The pathophysiology linking diabetes and cardiovascular disease (CVD) is
complex and multifactorial. The specific type of cardiomyopathy associated with diabetes,
known as diabetic cardiomyopathy (DCM), is recognized as asymptomatic progression
of structural and functional remodeling in the heart of diabetic patients in the absence
of coronary atherosclerosis and hypertension. In other words, the presence of heart disease
specifically in diabetic patients is also known as diabetic heart disease. This article
reviews the impact of diabetes in heart and vascular beds focusing on molecular mechanisms
involving the oxidative stress, the inflammation, the endothelium dysfunction and
the alteration of the homeostasis of calcium, among others mechanisms. Understanding
these mechanisms will help identify and treat CVD in patients with diabetes, as well as to
plan efficient strategies to mitigate DCM impact in those patients.
Collapse
Affiliation(s)
- Tarik Smani
- Group of Cardiovascular Physiopathology, Institute of Biomedicine of Seville-IBiS, HUVR/University of Seville/CSIC, Seville, Spain
| | | | - Javier Ávila-Médina
- Group of Cardiovascular Physiopathology, Institute of Biomedicine of Seville-IBiS, HUVR/University of Seville/CSIC, Seville, Spain
| | - Manuel F. Jimenez-Navarro
- UGC del Corazon, Instituto de Biomedicina de Malaga (IBIMA), Hospital Clínico Universitario Virgen de la Victoria, Universidad de Malaga, Malaga, Spain
| | - Antonio Ordoñez
- Group of Cardiovascular Physiopathology, Institute of Biomedicine of Seville-IBiS, HUVR/University of Seville/CSIC, Seville, Spain
| | - Abdelkrim Hmadcha
- Department of Regeneration and Cell Therapy, Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), Unversity of Pablo de Olavide- University of Seville-CSIC, Seville, Spain
| |
Collapse
|
31
|
Glucocorticoid stimulation increases cardiac contractility by SGK1-dependent SOCE-activation in rat cardiac myocytes. PLoS One 2019; 14:e0222341. [PMID: 31498847 PMCID: PMC6733454 DOI: 10.1371/journal.pone.0222341] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Accepted: 08/27/2019] [Indexed: 01/28/2023] Open
Abstract
Aims Glucocorticoid (GC) stimulation has been shown to increase cardiac contractility by elevated intracellular [Ca] but the sources for Ca entry are unclear. This study aims to determine the role of store-operated Ca entry (SOCE) for GC-mediated inotropy. Methods and results Dexamethasone (Dex) pretreatment significantly increased cardiac contractile force ex vivo in Langendorff-perfused Sprague-Dawley rat hearts (2 mg/kg BW i.p. Dex 24 h prior to experiment). Moreover, Ca transient amplitude as well as fractional shortening were significantly enhanced in Fura-2-loaded isolated rat ventricular myocytes exposed to Dex (1 mg/mL Dex, 24 h). Interestingly, these Dex-dependent effects could be abolished in the presence of SOCE-inhibitors SKF-96356 (SKF, 2 μM) and BTP2 (5 μM). Ca transient kinetics (time to peak, decay time) were not affected by SOCE stimulation. Direct SOCE measurements revealed a negligible magnitude in untreated myocytes but a dramatic increase in SOCE upon Dex-pretreatment. Importantly, the Dex-dependent stimulation of SOCE could be blocked by inhibition of serum and glucocorticoid-regulated kinase 1 (SGK1) using EMD638683 (EMD, 50 μM). Dex preincubation also resulted in increased mRNA expression of proteins involved in SOCE (stromal interaction molecule 2, STIM2, and transient receptor potential cation channels 3/6, TRPC 3/6), which were also prevented in the presence of EMD. Conclusion Short-term GC-stimulation with Dex improves cardiac contractility by a SOCE-dependent mechanism, which appears to involve increased SGK1-dependent expression of the SOCE-related proteins. Since Ca transient kinetics were unaffected, SOCE appears to influence Ca cycling more by an integrated response across multiple cardiac cycles but not on a beat-to-beat basis.
Collapse
|
32
|
Cheng QY, Yang MC, Wu J, Jia XL, Xiao C, Lian T, Zhang SZ. Reduced cardiac ischemia/reperfusion injury by hypothermic reperfusion via activation of transient receptor potential M8 channel. Life Sci 2019; 232:116658. [PMID: 31310758 DOI: 10.1016/j.lfs.2019.116658] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 07/05/2019] [Accepted: 07/12/2019] [Indexed: 01/23/2023]
Abstract
AIMS To investigate the cardioprotective effects of hypothermic (25 °C) reperfusion on ischemia/reperfusion injury and the role of transient potential channel M8 (TRPM8) in this process. MAIN METHODS Western blot and real-time PCR were used to monitor the expression of TRPM8 in myocardium. Myocardial ischemia/reperfusion injury was induced by 30 min of global ischemia followed by 120 min of reperfusion in Langendorff-perfused hearts from Sprague-Dawley rats. The reperfusion was either normothermic (37 °C) or hypothermic (25 °C). Infarct size and left ventricular function were assessed, and lactate dehydrogenase (LDH), superoxide dismutase (SOD), and malondialdehyde (MDA) in the coronary effluent were measured spectrophotometrically, and cardiomyocyte apoptosis was detected by TUNEL assay. The expression of TRPM8, Bcl-2, Bax, cleaved capspase-3, RhoA, and ROCK2 was quantified. KEY FINDINGS TRPM8 protein and mRNA were expressed in rat myocardium. Hypothermic reperfusion decreased the infarct size, LDH activity, MDA content, apoptosis, and expression of Bax, cleaved caspase-3, RhoA, and ROCK2 compared with normothermic reperfusion. These effects were associated with improved recovery of left ventricular contractility, and were reduced by BCTC, a TRPM8 antagonist. Ischemia/reperfusion injury and the increased expression of Bax, caspase-3, RhoA, and ROCK2 induced by normothermic reperfusion were reduced by Icilin, a TRPM8 agonist. SIGNIFICANCE Hypothermic reperfusion at 25 °C has cardioprotective effects against ischemia/reperfusion injury via activation of TRPM8 to inhibit the oxidative stress-related RhoA/ROCK2 signal pathway.
Collapse
Affiliation(s)
- Quan-Yi Cheng
- Department of Physiology, Medical College of China Three Gorges University, 8 Daxue Road, Yichang 443002, China
| | - Meng-Cheng Yang
- Department of Physiology, Medical College of China Three Gorges University, 8 Daxue Road, Yichang 443002, China
| | - Jing Wu
- Department of Physiology, Medical College of China Three Gorges University, 8 Daxue Road, Yichang 443002, China
| | - Xiao-Li Jia
- Department of Physiology, Medical College of China Three Gorges University, 8 Daxue Road, Yichang 443002, China
| | - Chao Xiao
- Department of Physiology, Medical College of China Three Gorges University, 8 Daxue Road, Yichang 443002, China
| | - Ting Lian
- Department of Physiology, Medical College of China Three Gorges University, 8 Daxue Road, Yichang 443002, China
| | - Shi-Zhong Zhang
- Department of Physiology, Medical College of China Three Gorges University, 8 Daxue Road, Yichang 443002, China.
| |
Collapse
|
33
|
Zhang ZM, Wu XL, Zhang GY, Ma X, He DX. Functional food development: Insights from TRP channels. J Funct Foods 2019. [DOI: 10.1016/j.jff.2019.03.023] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
|
34
|
Falcón D, Galeano-Otero I, Calderón-Sánchez E, Del Toro R, Martín-Bórnez M, Rosado JA, Hmadcha A, Smani T. TRP Channels: Current Perspectives in the Adverse Cardiac Remodeling. Front Physiol 2019; 10:159. [PMID: 30881310 PMCID: PMC6406032 DOI: 10.3389/fphys.2019.00159] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Accepted: 02/08/2019] [Indexed: 12/22/2022] Open
Abstract
Calcium is an important second messenger required not only for the excitation-contraction coupling of the heart but also critical for the activation of cell signaling pathways involved in the adverse cardiac remodeling and consequently for the heart failure. Sustained neurohumoral activation, pressure-overload, or myocardial injury can cause pathologic hypertrophic growth of the heart followed by interstitial fibrosis. The consequent heart’s structural and molecular adaptation might elevate the risk of developing heart failure and malignant arrhythmia. Compelling evidences have demonstrated that Ca2+ entry through TRP channels might play pivotal roles in cardiac function and pathology. TRP proteins are classified into six subfamilies: TRPC (canonical), TRPV (vanilloid), TRPM (melastatin), TRPA (ankyrin), TRPML (mucolipin), and TRPP (polycystin), which are activated by numerous physical and/or chemical stimuli. TRP channels participate to the handling of the intracellular Ca2+ concentration in cardiac myocytes and are mediators of different cardiovascular alterations. This review provides an overview of the current knowledge of TRP proteins implication in the pathologic process of some frequent cardiac diseases associated with the adverse cardiac remodeling such as cardiac hypertrophy, fibrosis, and conduction alteration.
Collapse
Affiliation(s)
- Debora Falcón
- Department of Medical Physiology and Biophysics, Institute of Biomedicine of Seville, University of Seville, Sevilla, Spain
| | - Isabel Galeano-Otero
- Department of Medical Physiology and Biophysics, Institute of Biomedicine of Seville, University of Seville, Sevilla, Spain
| | - Eva Calderón-Sánchez
- Department of Medical Physiology and Biophysics, Institute of Biomedicine of Seville, University of Seville, Sevilla, Spain.,CIBERCV, Madrid, Spain
| | - Raquel Del Toro
- Department of Medical Physiology and Biophysics, Institute of Biomedicine of Seville, University of Seville, Sevilla, Spain.,CIBERCV, Madrid, Spain
| | - Marta Martín-Bórnez
- Department of Medical Physiology and Biophysics, Institute of Biomedicine of Seville, University of Seville, Sevilla, Spain
| | - Juan A Rosado
- Department of Physiology (Cell Physiology Research Group), University of Extremadura, Cáceres, Spain
| | - Abdelkrim Hmadcha
- Department of Generation and Cell Therapy, Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), University of Pablo de Olavide-University of Seville-CSIC, Sevilla, Spain.,CIBERDEM, Madrid, Spain
| | - Tarik Smani
- Department of Medical Physiology and Biophysics, Institute of Biomedicine of Seville, University of Seville, Sevilla, Spain.,CIBERCV, Madrid, Spain
| |
Collapse
|
35
|
Deteriorated gene expression of selected calcium transporters in streptozotocin-induced diabetic hearts of Wistar rats. EUROPEAN PHARMACEUTICAL JOURNAL 2018. [DOI: 10.2478/afpuc-2018-0005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
Aim: The aim is to identify the possible changes in the expression of genes, that regulate calcium homeostasis in cardiomyocytes in diabetes mellitus.
Methods: Male Wistar rats were randomized into two experimental protocols: short-term 5-days streptozotocin-induced diabetes protocol with 20 weeks old animals at the end of the protocol (total N = 20) and long-term 4-weeks protocol with 18 weeks of age at the end of the protocol (total N = 38). 50 mg/kg of streptozotocin (STZ) was administered in both protocols by a single intraperitoneal injection in 0,1M citrate buffer (pH = 4.5). Control group (CON) received only vehiculum. Gene expressions in samples of left heart ventricle were measured by RT-qPCR method.
Results: The expression of SERCA2a in short-term protocol was decreased. In long-term protocol, decreased SERCA2a, TRPC4 and TRPC6 mRNA levels were observed (*p < 0.05). SERCA2a and TRPC4 mRNA levels exhibited statistical monotonic correlation in STZ-treated group in long-term protocol.
Conclusions: In diabetes mellitus, the calcium homeostasis in cardiomyocytes is altered and there could be a relation between alteration of internal sarcoplasmatic stores and store-operated calcium entry.
Collapse
|
36
|
Novel Potentials of the DPP-4 Inhibitor Sitagliptin against Ischemia-Reperfusion (I/R) Injury in Rat Ex-Vivo Heart Model. Int J Mol Sci 2018; 19:ijms19103226. [PMID: 30340421 PMCID: PMC6213995 DOI: 10.3390/ijms19103226] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 10/12/2018] [Accepted: 10/15/2018] [Indexed: 02/06/2023] Open
Abstract
Dipeptidyl peptidase-4 (DPP-4) inhibitors are a class of oral anti-diabetic drugs, implicated in pleiotropic secondary cardioprotective effects. The aim of the study was to unveil the unknown and possible cardioprotective targets that can be exerted by sitagliptin (Sitg) against ischemia-reperfusion (I/R) injury. Male wistar rats received 2 weeks’ Sitg oral treatment of different doses (25, 50, 100, and 150 mg/kg/day), or saline as a Control. Hearts were then isolated and subjected to two different I/R injury protocols: 10 min perfusion, 45 min regional ischemia, and 120 min reperfusion for infarct size (IS) measurement, or: 10 min perfusion, 45 min regional ischemia and 10 min reperfusion for biochemical analysis: nitric oxide synthases (NOSs) and DPP-4 activity, glucagon-like peptide-1 (GLP-1), Calcium, transient receptor potential vanilloid (TRPV)-1 and calcitonin gene-related peptide (CGRP) levels, transient receptor potential canonical (TRPC)-1 and e-NOS protein expression. NOS inhibitor (l-NAME) and TRPV-1 inhibitor (Capsazepine) were utilized to confirm the implication of both signaling mechanisms in DPP-4 inhibition-induced at the level of IS. Findings show that Sitg (50 mg) resulted in significant decrease in IS and DPP-4 activity, and significant increase in GLP-1, NOS activity, e-NOS expression, TRPV-1 level and TRPC-1 expression, compared to controls. Results of CGRP are in line with TRPV-1, as a downstream regulatory effect. NOS system and transient receptor potential (TRP) channels can contribute to DPP-4 inhibition-mediated cardioprotection against I/R injury using Sitagliptin.
Collapse
|
37
|
Wang Z, Xu Y, Wang M, Ye J, Liu J, Jiang H, Ye D, Wan J. TRPA1 inhibition ameliorates pressure overload-induced cardiac hypertrophy and fibrosis in mice. EBioMedicine 2018; 36:54-62. [PMID: 30297144 PMCID: PMC6197736 DOI: 10.1016/j.ebiom.2018.08.022] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 08/09/2018] [Accepted: 08/09/2018] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Recent evidence has indicated that the transient receptor potential ankyrin 1 (TRPA1) is expressed in the cardiovascular system and implicated in the development and progression of several cardiovascular diseases. However, the effects of TRPA1 on cardiac hypertrophy development remain unclear. The aim of this study was to determine the role of TRPA1 in cardiac hypertrophy and fibrosis development. METHODS C57BL/6J mice were subjected to transverse aortic constriction (TAC) and were orally treated with the TRPA1 selective inhibitors HC-030031 (HC) and TCS-5861528 (TCS). Morphological assessments, echocardiographic parameters, histological analyses and flow cytometry were used to evaluate cardiac hypertrophy and fibrosis. RESULTS Human and mouse hypertrophic hearts presented with noticeably increased TRPA1 protein levels. Inhibition of TRPA1 by HC and TCS attenuated cardiac hypertrophy and preserved cardiac function after chronic pressure overload, as evidenced by increased heart weight/body weight ratio, cardiomyocyte cross-sectional area and mRNA expression of hypertrophic markers, including ANP, BNP and β-MHC. Dramatic interstitial fibrosis was observed in the mice subjected to TAC surgery, and this was markedly attenuated in the HC and TCS treated mice. Mechanistically, the results revealed that TRPA1 inhibition ameliorated pressure overload-induced cardiac hypertrophy by negatively regulating Ca2+/calmodulin-dependent protein kinase II (CaMKII) and calcineurin signaling pathways. We also demonstrated that blocking TRPA1 decreased the proportion of M2 macrophages and reduced profibrotic cytokine levels, thereby improving cardiac fibrosis. CONCLUSIONS TRPA1 inhibition protected against cardiac hypertrophy and suppressed cardiac dysfunction via Ca2+-dependent signal pathways and inhibition of the M2 macrophages transition. These results suggest that TRPA1 may represent a potential therapeutic drug target for cardiac hypertrophy and fibrosis.
Collapse
Affiliation(s)
- Zhen Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China; Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| | - Yao Xu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China; Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| | - Menglong Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China; Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| | - Jing Ye
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China; Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| | - Jianfang Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China; Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| | - Huimin Jiang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China; Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| | - Di Ye
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China; Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| | - Jun Wan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China; Hubei Key Laboratory of Cardiology, Wuhan 430060, China.
| |
Collapse
|
38
|
Lu Z, Cui Y, Wei X, Gao P, Zhang H, Wei X, Li Q, Sun F, Yan Z, Zheng H, Yang G, Liu D, Zhu Z. Deficiency of PKD2L1 (TRPP3) Exacerbates Pathological Cardiac Hypertrophy by Augmenting NCX1-Mediated Mitochondrial Calcium Overload. Cell Rep 2018; 24:1639-1652. [DOI: 10.1016/j.celrep.2018.07.022] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 04/19/2018] [Accepted: 07/06/2018] [Indexed: 10/28/2022] Open
|
39
|
Barbiero S, Aimo A, Castiglione V, Giannoni A, Vergaro G, Passino C, Emdin M. Healthy hearts at hectic pace: From daily life stress to abnormal cardiomyocyte function and arrhythmias. Eur J Prev Cardiol 2018; 25:1419-1430. [PMID: 30052067 DOI: 10.1177/2047487318790614] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The hectic pace of contemporary life is a major source of acute and chronic stress, which may have a deleterious impact on body health . In the field of cardiovascular disease, acute emotional stress has been associated with coronary spasm and Takotsubo cardiomyopathy, whereas the manifestations of chronic stress have been overlooked, and most underlying pathophysiology remains to be elucidated. Chronic stress affects the neuronal circuitry composed of cortico-limbic structures and the nuclei regulating autonomic function, eliciting a sympatho-vagal imbalance, characterised by adrenergic activation and vagal withdrawal. Sympathetic terminals are connected to cardiomyocytes in a quasi-synaptic way, producing the so called 'neuro-cardiac junction'. During chronic stress, norepinephrine release is increased, leading to overstimulation of cardiomyocytes via β1-adrenergic receptors, influencing mainly calcium dynamics, and β2-adrenergic receptors, which control housekeeping functions. The circadian rhythm of cardiomyocytes is then impaired, with elongation of the catabolic ('light' phase) over the anabolic ('nocturnal') phase. This leads to a depletion of cell energy storage, and a decreased turnover of cell constituents. Even cell interactions are affected, as coupling between cardiomyocytes decreases while coupling between cardiomyocytes and fibroblasts increases. The ultimate results are changes in the shape and velocity of action potential, fibroblast activation and deposition of extracellular matrix. These alterations may predispose to arrhythmias and may favour the development of a stress-related cardiomyopathy. A better comprehension of this cascade of events may allow us to identify screening protocols and treatment strategies (meditation, yoga, physical activity, psychological assistance, β-blockers) to prevent or relieve ongoing cardiac damage.
Collapse
Affiliation(s)
- Silvia Barbiero
- 1 Institute of Life Sciences, Scuola Superiore Sant'Anna, Italy
| | - Alberto Aimo
- 1 Institute of Life Sciences, Scuola Superiore Sant'Anna, Italy.,2 Cardiology Division, University Hospital of Pisa, Italy
| | | | - Alberto Giannoni
- 1 Institute of Life Sciences, Scuola Superiore Sant'Anna, Italy.,3 Cardiology Division, Fondazione Toscana Gabriele Monasterio, Italy
| | - Giuseppe Vergaro
- 1 Institute of Life Sciences, Scuola Superiore Sant'Anna, Italy.,3 Cardiology Division, Fondazione Toscana Gabriele Monasterio, Italy
| | - Claudio Passino
- 1 Institute of Life Sciences, Scuola Superiore Sant'Anna, Italy.,3 Cardiology Division, Fondazione Toscana Gabriele Monasterio, Italy
| | - Michele Emdin
- 1 Institute of Life Sciences, Scuola Superiore Sant'Anna, Italy.,3 Cardiology Division, Fondazione Toscana Gabriele Monasterio, Italy
| |
Collapse
|
40
|
Zhang H, Liu H, Luo X, Wang Y, Liu Y, Jin H, Liu Z, Yang W, Yu P, Zhang L, Zhang L. Design, synthesis and biological activities of 2,3-dihydroquinazolin-4(1H)-one derivatives as TRPM2 inhibitors. Eur J Med Chem 2018; 152:235-252. [PMID: 29723786 DOI: 10.1016/j.ejmech.2018.04.045] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 04/06/2018] [Accepted: 04/21/2018] [Indexed: 12/26/2022]
Abstract
Transient receptor potential melastatin 2 (TRPM2), a Ca2+-permeable cationic channel, plays critical roles in insulin release, cytokine production, body temperature regulation and cell death as a reactive oxygen species (ROS) and temperature sensor. However, few TRPM2 inhibitors have been reported, especially TRP-subtype selective inhibitors, which hampers the investigation and validation of TRPM2 as a drug target. To discover novel TRPM2 inhibitors, 3D similarity-based virtual screening method was employed, by which 2,3-dihydroquinazolin-4(1H)-one derivative H1 was identified as a TRPM2 inhibitor. A series of novel 2,3-dihydroquinazolin-4(1H)-one derivatives were subsequently synthesized and characterized. Their inhibitory activities against the TRPM2 channel were evaluated by calcium imaging and electrophysiology approaches. Some of the compounds exhibited significant inhibitory activity, especially D9 which showed an IC50 of 3.7 μM against TRPM2 and did not affect the TRPM8 channel. The summarized structure-activity relationship (SAR) provides valuable insights for further development of specific TRPM2 targeted inhibitors.
Collapse
Affiliation(s)
- Han Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, PR China
| | - Huan Liu
- Department of Neurobiology, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, PR China
| | - Xiao Luo
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, PR China
| | - Yuxi Wang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, PR China
| | - Yuan Liu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, PR China
| | - Hongwei Jin
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, PR China
| | - Zhenming Liu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, PR China
| | - Wei Yang
- Department of Neurobiology, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, PR China
| | - Peilin Yu
- Department of Toxicology, School of Public Health, Zhejiang University, Hangzhou, Zhejiang, 310058, PR China.
| | - Liangren Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, PR China.
| | - Lihe Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, PR China
| |
Collapse
|
41
|
|
42
|
Er LK, Teng MS, Wu S, Hsu LA, Tzeng IS, Cheng CF, Chang HI, Chou HH, Ko YL. Combined effect of acid-sensing ion channel 3 and transient receptor potential vanilloid 1 gene polymorphisms on blood pressure variations in Taiwanese. Tzu Chi Med J 2018; 30:29-36. [PMID: 29643714 PMCID: PMC5883834 DOI: 10.4103/tcmj.tcmj_187_17] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 08/03/2017] [Accepted: 09/06/2017] [Indexed: 01/05/2023] Open
Abstract
OBJECTIVES Both acid-sensing ion channel acid-sensing ion channel 3 (ASIC3) and transient receptor potential vanilloid 1 (TRPV1) have been proposed to be involved in the pathophysiology of hypertension. Common colocalization of ASIC3 and TRPV1 channels in the same sensory neuron has been reported. We aimed to study the combined ASIC3 and TRPV1 gene polymorphisms in the risk of hypertension. MATERIALS AND METHODS To test the statistical association between genetic polymorphisms of the ASIC3 and TRPV1 genes and blood pressure (BP) variations in Taiwanese, 551 unrelated individuals (286 men and 265 women) having routine health examinations were recruited. The participants had no history of cardiovascular disease or use of medication for hypertension. RESULTS Six ASIC3 and four TRPV1 gene polymorphisms were genotyped, and only the ASIC3 rs2288646 polymorphism was associated with variations in BP in the participants. In subgroup analysis, we found participants carrying the combined ASIC3 rs2288646 AA or AG and TRPV1 rs8065080 CC genotypes (combined genotypes) had significantly higher systolic, mean and diastolic BP compared with the other subgroups (P = 0.009, 0.003, and 0.006, respectively, after Bonferroni correction). Interaction analysis also revealed significant gene-gene interaction in the systolic, mean, and diastolic BP in the ASIC3 and TRPV1 genotypes (interaction P = 0.006, 0.002, and 0.002, respectively). A trend of increasing frequencies of the combined genotype was observed in normotensive, prehypertensive, and hypertensive subgroups (P for trend = 0.001), as well as in those with higher systolic and diastolic BPs (P for trend = 9.13 × 10-4 and P for trend = 5.5 × 10-5, respectively). CONCLUSION Our data show a combined effect of ASIC3 and TRPV1 gene polymorphisms in BP variations in Taiwanese. These results suggest that the interaction between ASIC3 and TRPV1 is involved in BP regulation.
Collapse
Affiliation(s)
- Leay-Kiaw Er
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei, Taiwan
| | - Ming-Sheng Teng
- Department of Research, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei, Taiwan
| | - Semon Wu
- Department of Research, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei, Taiwan
- Department of Life Science, Chinese Culture University, Taipei, Taiwan
| | - Lung-An Hsu
- First Cardiovascular Division, Department of Internal Medicine, Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - I-Shiang Tzeng
- Department of Research, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei, Taiwan
| | - Ching-Feng Cheng
- Department of Pediatrics, Buddhist Tzu Chi General Hospital, Hualien, Taiwan
- School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Hsin-I Chang
- Division of Cardiology, Department of Internal Medicine and Cardiovascular Medical Center, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei, Taiwan
| | - Hsin-Hua Chou
- School of Medicine, Tzu Chi University, Hualien, Taiwan
- Division of Cardiology, Department of Internal Medicine and Cardiovascular Medical Center, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei, Taiwan
| | - Yu-Lin Ko
- Department of Research, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei, Taiwan
- School of Medicine, Tzu Chi University, Hualien, Taiwan
- Division of Cardiology, Department of Internal Medicine and Cardiovascular Medical Center, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei, Taiwan
| |
Collapse
|
43
|
Adding dimension to cellular mechanotransduction: Advances in biomedical engineering of multiaxial cell-stretch systems and their application to cardiovascular biomechanics and mechano-signaling. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2017. [DOI: 10.1016/j.pbiomolbio.2017.06.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
44
|
Liang M, Zhong W, Miao F, Wu H, Liu Y. Effects of losartan on vasomotor function and canonical transient receptor potential channels in the aortas of sinoaortic denervation rats. Clin Exp Hypertens 2017; 40:39-48. [PMID: 29072489 DOI: 10.1080/10641963.2017.1299746] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Increased short-term blood pressure variability (BPV) is strongly correlated with target organ damage. However, the molecular mechanisms underlying abnormal BPV-induced organ damage and effective therapeutic targets are poorly understood. The purpose of this study was to investigate the effects of losartan on vasomotor function and canonical transient receptor potential (TRPC) channels in the aortas of rats with arterial pressure lability induced by sinoaortic denervation (SAD). SAD was performed in male Sprague-Dawley rats at the age of 10 weeks. The experiment included sham-operated (Sham), SAD, and losartan-treated SAD (SAD+Los) groups. After 8 weeks of treatment, hemodynamic parameters were measured via catheterization, thoracic aortic vasomotor functions were evaluated using a physiological vascular ring tension recording system, and TRPC1 and 6 mRNA and protein expression levels in the endothelial cells (ECs) and smooth muscle cells (SMCs) of the thoracic aorta were determined via reverse transcription polymerase chain reaction (RT-PCR) and Western-blotting, respectively. Compared with Sham rats, SAD rats exhibited significantly increased BPV, enhanced norepinephrine-induced aortic contraction, and attenuated acetylcholine-induced aortic relaxation. Both the mRNA and the protein expression levels of TRPC1 and 6 were significantly downregulated in the ECs and upregulated in the SMCs of the thoracic aortas of SAD rats. Losartan treatment prevented these SAD-induced changes. In conclusion, losartan efficiently prevented vasomotor function impairment in SAD rats by reducing BPV and regulating TRPC1 and 6 expression levels.
Collapse
Affiliation(s)
- Minlie Liang
- a Department of Cardiology , Zhujiang Hospital Affiliated to Southern Medical University , Guangzhou , Guangdong Province , P.R. China.,b Department of Cardiology , The First Hospital of Nanping , Nanping , Fujian Province , P.R. China
| | - Wenliang Zhong
- b Department of Cardiology , The First Hospital of Nanping , Nanping , Fujian Province , P.R. China
| | - Fei Miao
- a Department of Cardiology , Zhujiang Hospital Affiliated to Southern Medical University , Guangzhou , Guangdong Province , P.R. China
| | - Hongchao Wu
- a Department of Cardiology , Zhujiang Hospital Affiliated to Southern Medical University , Guangzhou , Guangdong Province , P.R. China
| | - Yingfeng Liu
- a Department of Cardiology , Zhujiang Hospital Affiliated to Southern Medical University , Guangzhou , Guangdong Province , P.R. China
| |
Collapse
|
45
|
Sinharoy P, Bratz IN, Sinha S, Showalter LE, Andrei SR, Damron DS. TRPA1 and TRPV1 contribute to propofol-mediated antagonism of U46619-induced constriction in murine coronary arteries. PLoS One 2017; 12:e0180106. [PMID: 28644897 PMCID: PMC5482493 DOI: 10.1371/journal.pone.0180106] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 06/09/2017] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Transient receptor potential (TRP) ion channels have emerged as key components contributing to vasoreactivity. Propofol, an anesthetic is associated with adverse side effects including hypotension and acute pain upon infusion. Our objective was to determine the extent to which TRPA1 and/or TRPV1 ion channels are involved in mediating propofol-induced vasorelaxation of mouse coronary arterioles in vitro and elucidate the potential cellular signal transduction pathway by which this occurs. METHODS Hearts were excised from anesthetized mice and coronary arterioles were dissected from control C57Bl/6J, TRPA1-/-, TRPV1-/- and double-knockout mice (TRPAV-/-). Isolated microvessels were cannulated and secured in a temperature-controlled chamber and allowed to equilibrate for 1 hr. Vasoreactivity studies were performed in microvessels pre-constricted with U46619 to assess the dose-dependent relaxation effects of propofol on coronary microvascular tone. RESULTS Propofol-induced relaxation was unaffected in vessels obtained from TRPV1-/- mice, markedly attenuated in pre-constricted vessels obtained from TRPA1-/- mice and abolished in vessels obtained from TRPAV-/- mice. Furthermore, NOS inhibition with L-NAME or endothelium denuding abolished the proporfol-induced depressor response in pre-constricted vessels obtained from all mice. In the absence of L-NAME, BKCa inhibition with penitrem A markedly attenuated propofol-mediated relaxation in vessels obtained from wild-type mice and to a lesser extent in vessels obtained from TRPV1-/-, mice with no effect in vessels obtained from TRPA1-/- or TRPAV-/- mice. CONCLUSIONS TRPA1 and TRPV1 appear to contribute to the propofol-mediated antagonism of U46619-induced constriction in murine coronary microvessels that involves activation of NOS and BKCa.
Collapse
Affiliation(s)
- Pritam Sinharoy
- Department of Anesthesia, Perioperative and Pain Medicine, Stanford School of Medicine, Stanford, California, United States of America
| | - Ian N. Bratz
- Department of Integrative Medical Sciences, Northeast Ohio Medical College, Rootstown, Ohio, United States of America
| | - Sayantani Sinha
- Department of Surgery, Division of Orthopedic Surgery, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, United States of America
| | - Loral E. Showalter
- Department of Biological Sciences, Kent State University, Kent, Ohio, United States of America
| | - Spencer R. Andrei
- Department of Biological Sciences, Kent State University, Kent, Ohio, United States of America
| | - Derek S. Damron
- Department of Biological Sciences, Kent State University, Kent, Ohio, United States of America
| |
Collapse
|
46
|
Alonso-Carbajo L, Kecskes M, Jacobs G, Pironet A, Syam N, Talavera K, Vennekens R. Muscling in on TRP channels in vascular smooth muscle cells and cardiomyocytes. Cell Calcium 2017; 66:48-61. [PMID: 28807149 DOI: 10.1016/j.ceca.2017.06.004] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 06/08/2017] [Accepted: 06/08/2017] [Indexed: 02/07/2023]
Abstract
The human TRP protein family comprises a family of 27 cation channels with diverse permeation and gating properties. The common theme is that they are very important regulators of intracellular Ca2+ signaling in diverse cell types, either by providing a Ca2+ influx pathway, or by depolarising the membrane potential, which on one hand triggers the activation of voltage-gated Ca2+ channels, and on the other limits the driving force for Ca2+ entry. Here we focus on the role of these TRP channels in vascular smooth muscle and cardiac striated muscle. We give an overview of highlights from the recent literature, and highlight the important and diverse roles of TRP channels in the pathophysiology of the cardiovascular system. The discovery of the superfamily of Transient Receptor Potential (TRP) channels has significantly enhanced our knowledge of multiple signal transduction mechanisms in cardiac muscle and vascular smooth muscle cells (VSMC). In recent years, multiple studies have provided evidence for the involvement of these channels, not only in the regulation of contraction, but also in cell proliferation and remodeling in pathological conditions. The mammalian family of TRP cation channels is composed by 28 genes which can be divided into 6 subfamilies groups based on sequence similarity: TRPC (Canonical), TRPM (Melastatin), TRPML (Mucolipins), TRPV (Vanilloid), TRPP (Policystin) and TRPA (Ankyrin-rich protein). Functional TRP channels are believed to form four-unit complexes in the plasma, each of them expressed with six transmembrane domain and intracellular N and C termini. Here we review the current knowledge on the expression of TRP channels in both muscle types, and discuss their functional properties and role in physiological and pathophysiological processes.
Collapse
Affiliation(s)
- Lucía Alonso-Carbajo
- Laboratory of Ion Channel Research, TRP Research Platform Leuven (TRPLe), Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium
| | - Miklos Kecskes
- Laboratory of Ion Channel Research, TRP Research Platform Leuven (TRPLe), Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium
| | - Griet Jacobs
- Laboratory of Ion Channel Research, TRP Research Platform Leuven (TRPLe), Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium
| | - Andy Pironet
- Laboratory of Ion Channel Research, TRP Research Platform Leuven (TRPLe), Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium
| | - Ninda Syam
- Laboratory of Ion Channel Research, TRP Research Platform Leuven (TRPLe), Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium
| | - Karel Talavera
- Laboratory of Ion Channel Research, TRP Research Platform Leuven (TRPLe), Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium.
| | - Rudi Vennekens
- Laboratory of Ion Channel Research, TRP Research Platform Leuven (TRPLe), Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium.
| |
Collapse
|
47
|
Leyva-López N, Gutiérrez-Grijalva EP, Vazquez-Olivo G, Heredia JB. Essential Oils of Oregano: Biological Activity beyond Their Antimicrobial Properties. Molecules 2017; 22:E989. [PMID: 28613267 PMCID: PMC6152729 DOI: 10.3390/molecules22060989] [Citation(s) in RCA: 192] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Revised: 06/08/2017] [Accepted: 06/10/2017] [Indexed: 02/07/2023] Open
Abstract
Essential oils of oregano are widely recognized for their antimicrobial activity, as well as their antiviral and antifungal properties. Nevertheless, recent investigations have demonstrated that these compounds are also potent antioxidant, anti-inflammatory, antidiabetic and cancer suppressor agents. These properties of oregano essential oils are of potential interest to the food, cosmetic and pharmaceutical industries. The aim of this manuscript is to review the latest evidence regarding essential oils of oregano and their beneficial effects on health.
Collapse
Affiliation(s)
- Nayely Leyva-López
- Centro de Investigación en Alimentación y Desarrollo A.C., Carretera a El Dorado km 5.5 Col. El Diez C.P., Culiacán, Sinaloa 80129, Mexico.
| | - Erick P Gutiérrez-Grijalva
- Centro de Investigación en Alimentación y Desarrollo A.C., Carretera a El Dorado km 5.5 Col. El Diez C.P., Culiacán, Sinaloa 80129, Mexico.
| | - Gabriela Vazquez-Olivo
- Centro de Investigación en Alimentación y Desarrollo A.C., Carretera a El Dorado km 5.5 Col. El Diez C.P., Culiacán, Sinaloa 80129, Mexico.
| | - J Basilio Heredia
- Centro de Investigación en Alimentación y Desarrollo A.C., Carretera a El Dorado km 5.5 Col. El Diez C.P., Culiacán, Sinaloa 80129, Mexico.
| |
Collapse
|
48
|
Machida T, Onoguchi A, Iizuka K, Ishibashi S, Yutani M, Hirafuji M. Effect of Docosahexaenoic Acid on Voltage-Independent Ca 2+ Entry Pathways in Cultured Vascular Smooth Muscle Cells Stimulated with 5-Hydroxytryptamine. Biol Pharm Bull 2017; 40:916-921. [DOI: 10.1248/bpb.b16-00788] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Takuji Machida
- Department of Pharmacological Sciences, School of Pharmaceutical Sciences, Health Sciences University of Hokkaido
| | - Akina Onoguchi
- Department of Pharmacological Sciences, School of Pharmaceutical Sciences, Health Sciences University of Hokkaido
| | - Kenji Iizuka
- Department of Pharmacological Sciences, School of Pharmaceutical Sciences, Health Sciences University of Hokkaido
| | - Sayuri Ishibashi
- Department of Pharmacological Sciences, School of Pharmaceutical Sciences, Health Sciences University of Hokkaido
| | - Mikiko Yutani
- Department of Pharmacological Sciences, School of Pharmaceutical Sciences, Health Sciences University of Hokkaido
| | - Masahiko Hirafuji
- Department of Pharmacological Sciences, School of Pharmaceutical Sciences, Health Sciences University of Hokkaido
| |
Collapse
|
49
|
Ma R, Chaudhari S, Li W. Canonical Transient Receptor Potential 6 Channel: A New Target of Reactive Oxygen Species in Renal Physiology and Pathology. Antioxid Redox Signal 2016; 25:732-748. [PMID: 26937558 PMCID: PMC5079416 DOI: 10.1089/ars.2016.6661] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 02/06/2016] [Indexed: 02/07/2023]
Abstract
SIGNIFICANCE Regulation of Ca2+ signaling cascade by reactive oxygen species (ROS) is becoming increasingly evident and this regulation represents a key mechanism for control of many fundamental cellular functions. Canonical transient receptor potential (TRPC) 6, a member of Ca2+-conductive channel in the TRPC family, is widely expressed in kidney cells, including glomerular mesangial cells, podocytes, tubular epithelial cells, and vascular myocytes in renal microvasculature. Both overproduction of ROS and dysfunction of TRPC6 channel are involved in renal injury in animal models and human subjects. Although regulation of TRPC channel function by ROS has been well described in other tissues and cell types, such as vascular smooth muscle, this important cell regulatory mechanism has not been fully reviewed in kidney cells. Recent Advances: Accumulating evidence has shown that TRPC6 is a redox-sensitive channel, and modulation of TRPC6 Ca2+ signaling by altering TRPC6 protein expression or TRPC6 channel activity in kidney cells is a downstream mechanism by which ROS induce renal damage. CRITICAL ISSUES This review highlights how recent studies analyzing function and expression of TRPC6 channels in the kidney and their response to ROS improve our mechanistic understanding of oxidative stress-related kidney diseases. FUTURE DIRECTIONS Although it is evident that ROS regulate TRPC6-mediated Ca2+ signaling in several types of kidney cells, further study is needed to identify the underlying molecular mechanism. We hope that the newly identified ROS/TRPC6 pathway will pave the way to new, promising therapeutic strategies to target kidney diseases such as diabetic nephropathy. Antioxid. Redox Signal. 25, 732-748.
Collapse
Affiliation(s)
- Rong Ma
- Institute for Cardiovascular and Metabolic Diseases, University of North Texas Health Science Center, Fort Worth, Texas
| | - Sarika Chaudhari
- Institute for Cardiovascular and Metabolic Diseases, University of North Texas Health Science Center, Fort Worth, Texas
| | - Weizu Li
- Department of Pharmacology, Anhui Medical University, Hefei, People's Republic of China
| |
Collapse
|
50
|
Schwingshackl A. The role of stretch-activated ion channels in acute respiratory distress syndrome: finally a new target? Am J Physiol Lung Cell Mol Physiol 2016; 311:L639-52. [PMID: 27521425 DOI: 10.1152/ajplung.00458.2015] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 08/05/2016] [Indexed: 02/06/2023] Open
Abstract
Mechanical ventilation (MV) and oxygen therapy (hyperoxia; HO) comprise the cornerstones of life-saving interventions for patients with acute respiratory distress syndrome (ARDS). Unfortunately, the side effects of MV and HO include exacerbation of lung injury by barotrauma, volutrauma, and propagation of lung inflammation. Despite significant improvements in ventilator technologies and a heightened awareness of oxygen toxicity, besides low tidal volume ventilation few if any medical interventions have improved ARDS outcomes over the past two decades. We are lacking a comprehensive understanding of mechanotransduction processes in the healthy lung and know little about the interactions between simultaneously activated stretch-, HO-, and cytokine-induced signaling cascades in ARDS. Nevertheless, as we are unraveling these mechanisms we are gathering increasing evidence for the importance of stretch-activated ion channels (SACs) in the activation of lung-resident and inflammatory cells. In addition to the discovery of new SAC families in the lung, e.g., two-pore domain potassium channels, we are increasingly assigning mechanosensing properties to already known Na(+), Ca(2+), K(+), and Cl(-) channels. Better insights into the mechanotransduction mechanisms of SACs will improve our understanding of the pathways leading to ventilator-induced lung injury and lead to much needed novel therapeutic approaches against ARDS by specifically targeting SACs. This review 1) summarizes the reasons why the time has come to seriously consider SACs as new therapeutic targets against ARDS, 2) critically analyzes the physiological and experimental factors that currently limit our knowledge about SACs, and 3) outlines the most important questions future research studies need to address.
Collapse
|