1
|
Wang M, Preckel B, Zuurbier CJ, Weber NC. Effects of SGLT2 inhibitors on ion channels in heart failure: focus on the endothelium. Basic Res Cardiol 2025:10.1007/s00395-025-01115-y. [PMID: 40366385 DOI: 10.1007/s00395-025-01115-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 05/06/2025] [Accepted: 05/09/2025] [Indexed: 05/15/2025]
Abstract
Heart failure (HF) is a life-threatening cardiovascular disease associated with high mortality, diminished quality of life, and a significant economic burden on both patients and society. The pathogenesis of HF is closely related to the endothelium, where endothelial ion channels play an important role in regulating intracellular Ca2+ signals. These ion channels are essential to maintain vascular function, including endothelium-dependent vascular tone, inflammation response, and oxidative stress. Sodium-glucose cotransporter 2 inhibitors (SGLT2i) have shown promising cardiovascular benefits in HF patients, reducing mortality risk and hospitalization in several large clinical trials. Clinical and preclinical studies indicate that the cardioprotective effects of SGLT2i in HF are mediated by endothelial nitric oxide (NO) pathways, as well as by reducing inflammation and reactive oxygen species in cardiac endothelial cells. Additionally, SGLT2i may confer endothelial protection by lowering intracellular Ca2+ level through the inhibition of sodium-hydrogen exchanger 1 (NHE1) and sodium-calcium exchanger (NCX) in endothelial cells. In this review, we discuss present knowledge regarding the expression and role of Ca2+-related ion channels in endothelial cells in HF, focusing on the effects of SGLT2i on endothelial NHE1, NCX as well as on vascular tone.
Collapse
Affiliation(s)
- Mengnan Wang
- Department of Anesthesiology - Laboratory of Experimental Intensive Care and Anesthesiology-L.E.I.C.A, Amsterdam University Medical Centers, Amsterdam Cardiovascular Science, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Benedikt Preckel
- Department of Anesthesiology - Laboratory of Experimental Intensive Care and Anesthesiology-L.E.I.C.A, Amsterdam University Medical Centers, Amsterdam Cardiovascular Science, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Coert J Zuurbier
- Department of Anesthesiology - Laboratory of Experimental Intensive Care and Anesthesiology-L.E.I.C.A, Amsterdam University Medical Centers, Amsterdam Cardiovascular Science, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Nina C Weber
- Department of Anesthesiology - Laboratory of Experimental Intensive Care and Anesthesiology-L.E.I.C.A, Amsterdam University Medical Centers, Amsterdam Cardiovascular Science, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands.
| |
Collapse
|
2
|
Knoepp F, Abid S, Houssaini A, Lipskaia L, Gökyildirim MY, Born E, Marcos E, Arhatte M, Glogowska E, Vienney N, Günther A, Kraut S, Breitenborn-Mueller I, Quanz K, Fenner-Nau D, Derumeaux G, Weissmann N, Honoré E, Adnot S. Piezo1 in PASMCs: Critical for Hypoxia-Induced Pulmonary Hypertension Development. Circ Res 2025; 136:1031-1048. [PMID: 40181773 PMCID: PMC12036789 DOI: 10.1161/circresaha.124.325475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 02/25/2025] [Accepted: 03/25/2025] [Indexed: 04/05/2025]
Abstract
BACKGROUND Pulmonary hypertension (PH) is a life-threatening and progressive yet incurable disease. The hallmarks of PH comprise (1) sustained contraction and (2) excessive proliferation of pulmonary arterial smooth muscle cells (PASMCs). A major stimulus to which PASMCs are exposed during PH development is altered mechanical stress, originating from increased blood pressure, changes in blood flow velocity, and a progressive stiffening of pulmonary arteries. Mechanosensitive ion channels, including Piezo1 (Piezo-type mechanosensitive ion channel component-1), perceive such mechanical stimuli and translate them into a variety of cellular responses, including contractility or proliferation. Thus, the objective of the present study was to elucidate the specific role of Piezo1 in PASMCs for PH development and progression. METHODS The cell-type specific function of Piezo1 in PH was assessed in (1) PASMCs and lung tissues from patients with PH and (2) 2 mouse strains characterized by smooth muscle cell-specific, conditional Piezo1 knockout. Taking advantage of these strains, the smooth muscle cell-specific role of Piezo1 in PH development and progression was assessed in isolated, perfused, and ventilated mouse lungs, wire myography, and proliferation assays. Finally, in vivo function of smooth muscle cell-specific Piezo1 knockout was evaluated upon induction of chronic hypoxia-induced PH in these mice with insights into pulmonary vascular cell senescence. RESULTS Compared with healthy controls, PASMCs from patients with PH featured an elevated Piezo1 expression and increased proliferative phenotype. Smooth muscle cell-specific Piezo1 deletion, as confirmed via quantitative real-time polymerase chain reaction and patch clamp recordings, prevented the hypoxia-induced increase in PASMC proliferation in mice. Moreover, Piezo1 knockout reduced hypoxic pulmonary vasoconstriction in isolated, perfused, and ventilated mouse lungs, endothelial-denuded pulmonary arteries, and hemodynamic measurements in vivo. Consequently, Piezo1-deficient mice were considerably protected against chronic hypoxia-induced PH development with ameliorated right heart hypertrophy and improved hemodynamic function. In addition, distal pulmonary capillaries were preserved in the Piezo1-knockout mice, associated with a lower number of senescent endothelial cells. CONCLUSIONS This study provides evidence that Piezo1 expressed in PASMCs is critically involved in the pathogenesis of PH by controlling pulmonary vascular tone, arterial remodeling, and associated lung capillary rarefaction due to endothelial cell senescence.
Collapse
MESH Headings
- Animals
- Ion Channels/genetics
- Ion Channels/metabolism
- Ion Channels/deficiency
- Pulmonary Artery/metabolism
- Pulmonary Artery/pathology
- Pulmonary Artery/physiopathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Hypertension, Pulmonary/metabolism
- Hypertension, Pulmonary/etiology
- Hypertension, Pulmonary/pathology
- Hypertension, Pulmonary/physiopathology
- Hypertension, Pulmonary/genetics
- Hypoxia/complications
- Hypoxia/metabolism
- Mice, Knockout
- Humans
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Mice
- Cell Proliferation
- Male
- Cells, Cultured
- Mice, Inbred C57BL
- Female
- Vascular Remodeling
- Disease Models, Animal
Collapse
Affiliation(s)
- Fenja Knoepp
- Excellence Cluster Cardio-Pulmonary Institute, Universities of Giessen and Marburg Lung Center, Member of the German Center for Lung Research, Justus Liebig University (F.K., M.Y.G., A.G., S.K., I.B.-M., K.Q., D.F.-N., N.W.)
| | - Shariq Abid
- Département de Physiologie-Explorations Fonctionnelles and FHU SENEC Hôpital Henri Mondor, French National Institute of Health and Medical Research Unit 955, AP-HP, Créteil, France (S. Abid, A.H., L.L., E.B., E.M., N.V., G.D., S. Adnot)
- Medical Research Center, Liaquat University of Medical and Health Sciences, Pakistan (S. Abid)
| | - Amal Houssaini
- Département de Physiologie-Explorations Fonctionnelles and FHU SENEC Hôpital Henri Mondor, French National Institute of Health and Medical Research Unit 955, AP-HP, Créteil, France (S. Abid, A.H., L.L., E.B., E.M., N.V., G.D., S. Adnot)
- Institute for Lung Health, Justus Liebig University, Giessen, Germany (A.H., M.Y.G., S. Adnot)
| | - Larissa Lipskaia
- Département de Physiologie-Explorations Fonctionnelles and FHU SENEC Hôpital Henri Mondor, French National Institute of Health and Medical Research Unit 955, AP-HP, Créteil, France (S. Abid, A.H., L.L., E.B., E.M., N.V., G.D., S. Adnot)
| | - Mira Yasemin Gökyildirim
- Excellence Cluster Cardio-Pulmonary Institute, Universities of Giessen and Marburg Lung Center, Member of the German Center for Lung Research, Justus Liebig University (F.K., M.Y.G., A.G., S.K., I.B.-M., K.Q., D.F.-N., N.W.)
- Institute for Lung Health, Justus Liebig University, Giessen, Germany (A.H., M.Y.G., S. Adnot)
| | - Emmanuelle Born
- Département de Physiologie-Explorations Fonctionnelles and FHU SENEC Hôpital Henri Mondor, French National Institute of Health and Medical Research Unit 955, AP-HP, Créteil, France (S. Abid, A.H., L.L., E.B., E.M., N.V., G.D., S. Adnot)
| | - Elisabeth Marcos
- Département de Physiologie-Explorations Fonctionnelles and FHU SENEC Hôpital Henri Mondor, French National Institute of Health and Medical Research Unit 955, AP-HP, Créteil, France (S. Abid, A.H., L.L., E.B., E.M., N.V., G.D., S. Adnot)
| | - Malika Arhatte
- Centre National de la Recherche Scientifique, Institut national de la santé et de la recherche médicale, Institut de Pharmacologie Moléculaire et Cellulaire, Université Côte d’Azur, Valbonne, France (M.A., E.G., E.H.)
| | - Edyta Glogowska
- Centre National de la Recherche Scientifique, Institut national de la santé et de la recherche médicale, Institut de Pharmacologie Moléculaire et Cellulaire, Université Côte d’Azur, Valbonne, France (M.A., E.G., E.H.)
| | - Nora Vienney
- Département de Physiologie-Explorations Fonctionnelles and FHU SENEC Hôpital Henri Mondor, French National Institute of Health and Medical Research Unit 955, AP-HP, Créteil, France (S. Abid, A.H., L.L., E.B., E.M., N.V., G.D., S. Adnot)
| | - Andreas Günther
- Excellence Cluster Cardio-Pulmonary Institute, Universities of Giessen and Marburg Lung Center, Member of the German Center for Lung Research, Justus Liebig University (F.K., M.Y.G., A.G., S.K., I.B.-M., K.Q., D.F.-N., N.W.)
| | - Simone Kraut
- Excellence Cluster Cardio-Pulmonary Institute, Universities of Giessen and Marburg Lung Center, Member of the German Center for Lung Research, Justus Liebig University (F.K., M.Y.G., A.G., S.K., I.B.-M., K.Q., D.F.-N., N.W.)
| | - Ingrid Breitenborn-Mueller
- Excellence Cluster Cardio-Pulmonary Institute, Universities of Giessen and Marburg Lung Center, Member of the German Center for Lung Research, Justus Liebig University (F.K., M.Y.G., A.G., S.K., I.B.-M., K.Q., D.F.-N., N.W.)
| | - Karin Quanz
- Excellence Cluster Cardio-Pulmonary Institute, Universities of Giessen and Marburg Lung Center, Member of the German Center for Lung Research, Justus Liebig University (F.K., M.Y.G., A.G., S.K., I.B.-M., K.Q., D.F.-N., N.W.)
| | - Dagmar Fenner-Nau
- Excellence Cluster Cardio-Pulmonary Institute, Universities of Giessen and Marburg Lung Center, Member of the German Center for Lung Research, Justus Liebig University (F.K., M.Y.G., A.G., S.K., I.B.-M., K.Q., D.F.-N., N.W.)
| | - Geneviève Derumeaux
- Département de Physiologie-Explorations Fonctionnelles and FHU SENEC Hôpital Henri Mondor, French National Institute of Health and Medical Research Unit 955, AP-HP, Créteil, France (S. Abid, A.H., L.L., E.B., E.M., N.V., G.D., S. Adnot)
| | - Norbert Weissmann
- Excellence Cluster Cardio-Pulmonary Institute, Universities of Giessen and Marburg Lung Center, Member of the German Center for Lung Research, Justus Liebig University (F.K., M.Y.G., A.G., S.K., I.B.-M., K.Q., D.F.-N., N.W.)
| | - Eric Honoré
- Centre National de la Recherche Scientifique, Institut national de la santé et de la recherche médicale, Institut de Pharmacologie Moléculaire et Cellulaire, Université Côte d’Azur, Valbonne, France (M.A., E.G., E.H.)
| | - Serge Adnot
- Département de Physiologie-Explorations Fonctionnelles and FHU SENEC Hôpital Henri Mondor, French National Institute of Health and Medical Research Unit 955, AP-HP, Créteil, France (S. Abid, A.H., L.L., E.B., E.M., N.V., G.D., S. Adnot)
- Institute for Lung Health, Justus Liebig University, Giessen, Germany (A.H., M.Y.G., S. Adnot)
| |
Collapse
|
3
|
Singh J, Ruhoff AM, Ashok D, Wise SG, Waterhouse A. Engineering advanced in vitro models of endothelial dysfunction. Trends Biotechnol 2025:S0167-7799(25)00089-7. [PMID: 40187930 DOI: 10.1016/j.tibtech.2025.03.004] [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: 08/29/2024] [Revised: 01/30/2025] [Accepted: 03/07/2025] [Indexed: 04/07/2025]
Abstract
Endothelial dysfunction is an important initiator of cardiovascular disease, the leading cause of death globally, and often manifests in arterial regions with disturbed blood flow. Experimental model advances have crucially helped unravel physiological mechanisms. While in vivo models provide a dynamic environment, they often fail to mimic human physiology precisely and face significant ethical barriers. Advanced in vitro models, including organs-on-chips and bioreactors, combine human cells and blood flow to accurately replicate endothelial dysfunction. Newer models have enhanced scalability and accuracy, with organs-on-chips commonly outperforming standard preclinical methods. Importantly, recent endothelial dysfunction discoveries leverage dynamic models to identify and evaluate clinically promising therapeutics. Here, we examine these developments and explore opportunities to develop next-generation in vitro models of endothelial dysfunction.
Collapse
Affiliation(s)
- Jasneil Singh
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, 2006, Australia; The Charles Perkins Centre, The University of Sydney, Sydney, NSW, 2006, Australia; The University of Sydney Nano Institute, The University of Sydney, Sydney, NSW 2006, Australia
| | - Alexander M Ruhoff
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, 2006, Australia; The Charles Perkins Centre, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Deepu Ashok
- The Charles Perkins Centre, The University of Sydney, Sydney, NSW, 2006, Australia; The University of Sydney Nano Institute, The University of Sydney, Sydney, NSW 2006, Australia; School of Biomedical Engineering, Faculty of Engineering and IT, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Steven G Wise
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, 2006, Australia; The Charles Perkins Centre, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Anna Waterhouse
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, 2006, Australia; The Charles Perkins Centre, The University of Sydney, Sydney, NSW, 2006, Australia; The University of Sydney Nano Institute, The University of Sydney, Sydney, NSW 2006, Australia.
| |
Collapse
|
4
|
Wei F, Lin Z, Lu W, Luo H, Feng H, Liu S, Zhang C, Zheng Y, Chen J, Mo S, Wang C, Zhang Z, Feng W, Zhu J, Yang Q, Du M, Kong W, Liu A, Lai J, Li X, Wu X, Lai N, Chen Y, Yang K, Wang J. Deficiency of Endothelial Piezo2 Impairs Pulmonary Vascular Angiogenesis and Predisposes Pulmonary Hypertension. Hypertension 2025; 82:583-597. [PMID: 39758000 DOI: 10.1161/hypertensionaha.124.22948] [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: 02/25/2024] [Accepted: 12/24/2024] [Indexed: 01/07/2025]
Abstract
BACKGROUND Mechanosensitive Piezo1 (Piezo Type Mechanosensitive Ion Channel Component 1) channel plays a key role in pulmonary hypertension (PH). However, the role of Piezo2 in PH remains unclear. METHODS Endothelial cell (EC)-specific Piezo2 knockout (Piezo2flox/flox, Tek-Cre+; Piezo2EC-/-) rats and primarily cultured pulmonary microvascular ECs were used to determine the role of Piezo2 in PH. RESULTS Data analysis of publicly accessible single-cell RNA-sequencing data sets uncovered significant downregulation of Piezo2 in lung ECs from patients with idiopathic pulmonary arterial hypertension, which was verified in the lungs/ECs from PH rat models induced by hypoxia or monocrotaline. Comparing to wild-type rats, Piezo2EC-/- rats exhibited exacerbated PH in both hypoxia-induced PH and monocrotaline-induced PH, characterized by the worsened hemodynamical and histological changes. Piezo2EC-/- rats showed dramatic loss of pulmonary microvessels, in association with the decreased intracellular free calcium concentration ([Ca2+]i) and downregulation of VEGFR2 (vascular endothelial growth factor receptor 2) and phosphorylated SRF (serum response factor) in pulmonary microvascular ECs. Knockout of Piezo2 or treatment with a calcium chelator, EDTA, impaired the ability of tube formation and migration in pulmonary microvascular ECs, which was restored by supplementation of extra calcium. A safflower oil diet rich in linoleic acid, which can enhance the stability and function of Piezo2, effectively alleviated PH development in a hypoxia-induced PH rat model. CONCLUSIONS This study demonstrates that EC-specific knockout of Piezo2 exacerbates PH pathogenesis, at least partially, through the suppression of [Ca2+]i/phosphorylated SRF/VEGFR2 signaling axis in pulmonary vascular ECs. Targeted activation of Piezo2 could be a novel effective strategy for the treatment of PH.
Collapse
Affiliation(s)
- Feng Wei
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital (F.W., Z.L., W.L., H.L., H.F., S.L., C.Z., Y.Z., S.M., C.W., Z.Z., W.F., J.Z., Q.Y., M.D., W.K., A.L., J.L., X.L., X.W., N.L., Y.C., K.Y., J.W.), Guangzhou Medical University, China
| | - Ziying Lin
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital (F.W., Z.L., W.L., H.L., H.F., S.L., C.Z., Y.Z., S.M., C.W., Z.Z., W.F., J.Z., Q.Y., M.D., W.K., A.L., J.L., X.L., X.W., N.L., Y.C., K.Y., J.W.), Guangzhou Medical University, China
| | - Wenju Lu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital (F.W., Z.L., W.L., H.L., H.F., S.L., C.Z., Y.Z., S.M., C.W., Z.Z., W.F., J.Z., Q.Y., M.D., W.K., A.L., J.L., X.L., X.W., N.L., Y.C., K.Y., J.W.), Guangzhou Medical University, China
| | - Haiyun Luo
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital (F.W., Z.L., W.L., H.L., H.F., S.L., C.Z., Y.Z., S.M., C.W., Z.Z., W.F., J.Z., Q.Y., M.D., W.K., A.L., J.L., X.L., X.W., N.L., Y.C., K.Y., J.W.), Guangzhou Medical University, China
| | - Huazhuo Feng
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital (F.W., Z.L., W.L., H.L., H.F., S.L., C.Z., Y.Z., S.M., C.W., Z.Z., W.F., J.Z., Q.Y., M.D., W.K., A.L., J.L., X.L., X.W., N.L., Y.C., K.Y., J.W.), Guangzhou Medical University, China
| | - Shiyun Liu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital (F.W., Z.L., W.L., H.L., H.F., S.L., C.Z., Y.Z., S.M., C.W., Z.Z., W.F., J.Z., Q.Y., M.D., W.K., A.L., J.L., X.L., X.W., N.L., Y.C., K.Y., J.W.), Guangzhou Medical University, China
| | - Chenting Zhang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital (F.W., Z.L., W.L., H.L., H.F., S.L., C.Z., Y.Z., S.M., C.W., Z.Z., W.F., J.Z., Q.Y., M.D., W.K., A.L., J.L., X.L., X.W., N.L., Y.C., K.Y., J.W.), Guangzhou Medical University, China
| | - Yulin Zheng
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital (F.W., Z.L., W.L., H.L., H.F., S.L., C.Z., Y.Z., S.M., C.W., Z.Z., W.F., J.Z., Q.Y., M.D., W.K., A.L., J.L., X.L., X.W., N.L., Y.C., K.Y., J.W.), Guangzhou Medical University, China
| | - Jiyuan Chen
- Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China (J.C.)
| | - Shaocong Mo
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital (F.W., Z.L., W.L., H.L., H.F., S.L., C.Z., Y.Z., S.M., C.W., Z.Z., W.F., J.Z., Q.Y., M.D., W.K., A.L., J.L., X.L., X.W., N.L., Y.C., K.Y., J.W.), Guangzhou Medical University, China
| | - Chen Wang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital (F.W., Z.L., W.L., H.L., H.F., S.L., C.Z., Y.Z., S.M., C.W., Z.Z., W.F., J.Z., Q.Y., M.D., W.K., A.L., J.L., X.L., X.W., N.L., Y.C., K.Y., J.W.), Guangzhou Medical University, China
| | - Zizhou Zhang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital (F.W., Z.L., W.L., H.L., H.F., S.L., C.Z., Y.Z., S.M., C.W., Z.Z., W.F., J.Z., Q.Y., M.D., W.K., A.L., J.L., X.L., X.W., N.L., Y.C., K.Y., J.W.), Guangzhou Medical University, China
| | - Wei Feng
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital (F.W., Z.L., W.L., H.L., H.F., S.L., C.Z., Y.Z., S.M., C.W., Z.Z., W.F., J.Z., Q.Y., M.D., W.K., A.L., J.L., X.L., X.W., N.L., Y.C., K.Y., J.W.), Guangzhou Medical University, China
| | - Junqi Zhu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital (F.W., Z.L., W.L., H.L., H.F., S.L., C.Z., Y.Z., S.M., C.W., Z.Z., W.F., J.Z., Q.Y., M.D., W.K., A.L., J.L., X.L., X.W., N.L., Y.C., K.Y., J.W.), Guangzhou Medical University, China
| | - Qifeng Yang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital (F.W., Z.L., W.L., H.L., H.F., S.L., C.Z., Y.Z., S.M., C.W., Z.Z., W.F., J.Z., Q.Y., M.D., W.K., A.L., J.L., X.L., X.W., N.L., Y.C., K.Y., J.W.), Guangzhou Medical University, China
- Guangzhou National Laboratory, Guangzhou International Bio Island, Guangdong, China (Q.Y., J.W.)
| | - Min Du
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital (F.W., Z.L., W.L., H.L., H.F., S.L., C.Z., Y.Z., S.M., C.W., Z.Z., W.F., J.Z., Q.Y., M.D., W.K., A.L., J.L., X.L., X.W., N.L., Y.C., K.Y., J.W.), Guangzhou Medical University, China
- GMU-GIBH Joint School of Life Sciences (M.D.), Guangzhou Medical University, China
| | - Weiguo Kong
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital (F.W., Z.L., W.L., H.L., H.F., S.L., C.Z., Y.Z., S.M., C.W., Z.Z., W.F., J.Z., Q.Y., M.D., W.K., A.L., J.L., X.L., X.W., N.L., Y.C., K.Y., J.W.), Guangzhou Medical University, China
| | - Aofeng Liu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital (F.W., Z.L., W.L., H.L., H.F., S.L., C.Z., Y.Z., S.M., C.W., Z.Z., W.F., J.Z., Q.Y., M.D., W.K., A.L., J.L., X.L., X.W., N.L., Y.C., K.Y., J.W.), Guangzhou Medical University, China
| | - Jiaxuan Lai
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital (F.W., Z.L., W.L., H.L., H.F., S.L., C.Z., Y.Z., S.M., C.W., Z.Z., W.F., J.Z., Q.Y., M.D., W.K., A.L., J.L., X.L., X.W., N.L., Y.C., K.Y., J.W.), Guangzhou Medical University, China
| | - Xiang Li
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital (F.W., Z.L., W.L., H.L., H.F., S.L., C.Z., Y.Z., S.M., C.W., Z.Z., W.F., J.Z., Q.Y., M.D., W.K., A.L., J.L., X.L., X.W., N.L., Y.C., K.Y., J.W.), Guangzhou Medical University, China
- Center for Inflammation Science and Systems Medicine, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL (X.L.)
| | - Xuefen Wu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital (F.W., Z.L., W.L., H.L., H.F., S.L., C.Z., Y.Z., S.M., C.W., Z.Z., W.F., J.Z., Q.Y., M.D., W.K., A.L., J.L., X.L., X.W., N.L., Y.C., K.Y., J.W.), Guangzhou Medical University, China
| | - Ning Lai
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital (F.W., Z.L., W.L., H.L., H.F., S.L., C.Z., Y.Z., S.M., C.W., Z.Z., W.F., J.Z., Q.Y., M.D., W.K., A.L., J.L., X.L., X.W., N.L., Y.C., K.Y., J.W.), Guangzhou Medical University, China
| | - Yuqin Chen
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital (F.W., Z.L., W.L., H.L., H.F., S.L., C.Z., Y.Z., S.M., C.W., Z.Z., W.F., J.Z., Q.Y., M.D., W.K., A.L., J.L., X.L., X.W., N.L., Y.C., K.Y., J.W.), Guangzhou Medical University, China
| | - Kai Yang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital (F.W., Z.L., W.L., H.L., H.F., S.L., C.Z., Y.Z., S.M., C.W., Z.Z., W.F., J.Z., Q.Y., M.D., W.K., A.L., J.L., X.L., X.W., N.L., Y.C., K.Y., J.W.), Guangzhou Medical University, China
| | - Jian Wang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital (F.W., Z.L., W.L., H.L., H.F., S.L., C.Z., Y.Z., S.M., C.W., Z.Z., W.F., J.Z., Q.Y., M.D., W.K., A.L., J.L., X.L., X.W., N.L., Y.C., K.Y., J.W.), Guangzhou Medical University, China
- Guangzhou National Laboratory, Guangzhou International Bio Island, Guangdong, China (Q.Y., J.W.)
| |
Collapse
|
5
|
Walsh D, Kostyunina DS, Blake A, Boylan J, McLoughlin P. Shear stress-induced restoration of pulmonary microvascular endothelial barrier function following ischemia reperfusion injury requires VEGFR2 signaling. Am J Physiol Lung Cell Mol Physiol 2025; 328:L389-L404. [PMID: 39701597 DOI: 10.1152/ajplung.00200.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 12/03/2024] [Accepted: 12/11/2024] [Indexed: 12/21/2024] Open
Abstract
Normal shear stress produced by blood flow is sensed by the vascular endothelium and required for maintenance of the homeostatic functions of the endothelium in systemic conduit and resistance vessels. Many critical illnesses are characterized by periods of abnormally reduced or absent shear stress in the lung (e.g., hemorrhagic shock, embolism, ischemia reperfusion injury, and lung transplantation) and are complicated by pulmonary edema following reperfusion due to microvascular leak. The role of shear stress in regulating the pulmonary microvascular endothelial barrier in the intact vascular bed has not been previously examined. We tested the hypothesis that, in lungs injured by a period of ischemia and reperfusion (IRI), reduced shear stress contributes to increased pulmonary microvascular endothelial barrier permeability and edema formation. Furthermore, we examined the role of vascular endothelial-derived growth factor receptor 2 (VEGFR2) as a mechanosensor mediating the endothelial response to this altered shear stress. Following IRI, we perfused isolated ventilated mouse lungs with a low viscosity solution (LVS) or a higher, physiological viscosity solution (PVS) at constant flow to produce differing endothelial shear stresses in the intact microcirculation. Lungs perfused with LVS developed pulmonary edema due to increased endothelial permeability whereas those perfused with PVS were protected from edema formation by reduced endothelial permeability. This effect of PVS required normal VEGFR2 mechanoreceptor function. These data show for the first time that shear stress has an important role in restoring endothelial barrier function in the intact pulmonary microcirculation following injury and have important implications for the treatment of pulmonary edema in critically ill patients.NEW & NOTEWORTHY Critical illnesses are frequently complicated by noncardiogenic pulmonary edema. Many such illnesses include periods of reduced blood flow, often accompanied by hemodilution, which together reduce endothelial shear stress. We report that in ischemia-reperfusion injury reduced shear stress contributes to increased permeability of the in situ pulmonary microvascular endothelium and worsens alveolar edema. Restoring shear stress toward normal reduces endothelial permeability and edema formation, an effect that requires the normal mechanoreceptor function of VEGFR2.
Collapse
Affiliation(s)
- Don Walsh
- School of Medicine and Conway Institute, University College Dublin, Dublin, Ireland
| | - Daria S Kostyunina
- School of Medicine and Conway Institute, University College Dublin, Dublin, Ireland
| | - Aoife Blake
- School of Medicine and Conway Institute, University College Dublin, Dublin, Ireland
| | - John Boylan
- Department of Anaesthesia and Intensive Care, St. Vincent's University Hospital, Dublin, Ireland
| | - Paul McLoughlin
- School of Medicine and Conway Institute, University College Dublin, Dublin, Ireland
| |
Collapse
|
6
|
Li H, Li X, Sun Y, Zhi Z, Song L, Li M, Feng Y, Zhang Z, Liu Y, Chen Y, Zhao F, Zhu T. The Role of Ion Channels in Pulmonary Hypertension: A Review. Pulm Circ 2025; 15:e70050. [PMID: 39958971 PMCID: PMC11830494 DOI: 10.1002/pul2.70050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 08/16/2024] [Accepted: 01/31/2025] [Indexed: 02/18/2025] Open
Abstract
Pulmonary hypertension (PH) constitutes a critical challenge in cardiopulmonary medicine with a pathogenesis that is multifaceted and intricate. Ion channels, crucial determinants of cellular electrochemical gradient modulation, have emerged as significant participants in the pathophysiological progression of PH. These channels, abundant on the membranes of pulmonary artery smooth muscle cells (PASMCs) and pulmonary artery endothelial cells (PAECs), pivotally navigate the nuanced interplay of cell proliferation, migration, and endothelial function, each vital to the pulmonary vascular remodeling (PVR) hallmark of PH. Our review delves into the mechanistic insights of potassium, calcium, magnesium, zinc, and chloride ion channels in relation to their involvement in PH. It not only emphasizes the notable advances and discoveries that cast these ion channels as underlying factors in the etiology and exacerbation of PH but also highlights their potential as innovative therapeutic targets.
Collapse
Affiliation(s)
- Han‐Fei Li
- College of PharmacyXinxiang Medical UniversityXinxiangChina
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug InterventionXinxiangChina
| | - Xin‐Yao Li
- College of PharmacyXinxiang Medical UniversityXinxiangChina
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug InterventionXinxiangChina
| | - Yu‐Qing Sun
- College of PharmacyXinxiang Medical UniversityXinxiangChina
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug InterventionXinxiangChina
| | - Ze‐Ying Zhi
- College of PharmacyXinxiang Medical UniversityXinxiangChina
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug InterventionXinxiangChina
| | - Liao‐Fan Song
- College of PharmacyXinxiang Medical UniversityXinxiangChina
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug InterventionXinxiangChina
| | - Meng Li
- College of PharmacyXinxiang Medical UniversityXinxiangChina
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug InterventionXinxiangChina
| | - Yi‐Ming Feng
- College of PharmacyXinxiang Medical UniversityXinxiangChina
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug InterventionXinxiangChina
| | - Zhi‐Hao Zhang
- College of PharmacyXinxiang Medical UniversityXinxiangChina
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug InterventionXinxiangChina
| | - Yan‐Feng Liu
- College of PharmacyXinxiang Medical UniversityXinxiangChina
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug InterventionXinxiangChina
| | - Yu‐Jing Chen
- College of PharmacyXinxiang Medical UniversityXinxiangChina
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug InterventionXinxiangChina
| | - Fan‐Rong Zhao
- College of PharmacyXinxiang Medical UniversityXinxiangChina
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug InterventionXinxiangChina
| | - Tian‐Tian Zhu
- College of PharmacyXinxiang Medical UniversityXinxiangChina
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug InterventionXinxiangChina
- Department of PharmacyThe First Affiliated Hospital of Xinxiang Medical UniversityXinxiangChina
| |
Collapse
|
7
|
Cubero-Sarabia M, Kapetanaki AM, Vassalli M. Biophysical assays to test cellular mechanosensing: moving towards high throughput. Biophys Rev 2024; 16:875-882. [PMID: 39830126 PMCID: PMC11735701 DOI: 10.1007/s12551-024-01263-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 12/04/2024] [Indexed: 01/22/2025] Open
Abstract
Mechanosensitivity is the ability of cells to sense and respond to mechanical stimuli. In order to do this, cells are endowed with different components that allow them to react to a broad range of stimuli, such as compression or shear forces, pressure, and vibrations. This sensing process, mechanosensing, is involved in fundamental physiological mechanisms, such as stem cell differentiation and migration, but it is also central to the development of pathogenic states. Here, we review the approaches that have been proposed to quantify mechanosensation in living cells, with a specific focus on methodologies that enable higher experimental throughput. This aspect is crucial to fully understand the nuances of mechanosensation and how it impacts the physiology and pathology of living systems. We will discuss traditional methods for studying mechanosensing at the level of single cells, with particular attention to the activation of the mechanosensitive ion channel piezo1. Moreover, we will present recent attempts to push the analysis towards higher throughput.
Collapse
Affiliation(s)
| | | | - Massimo Vassalli
- James Watt School of Engineering, University of Glasgow, Glasgow, UK
| |
Collapse
|
8
|
Kinsella JA, Debant M, Parsonage G, Morley LC, Bajarwan M, Revill C, Foster R, Beech DJ. Pharmacology of PIEZO1 channels. Br J Pharmacol 2024; 181:4714-4732. [PMID: 39402010 DOI: 10.1111/bph.17351] [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: 02/06/2024] [Revised: 06/15/2024] [Accepted: 08/25/2024] [Indexed: 11/05/2024] Open
Abstract
PIEZO1 is a eukaryotic membrane protein that assembles as trimers to form calcium-permeable, non-selective cation channels with exquisite capabilities for mechanical force sensing and transduction of force into effect in diverse cell types that include blood cells, endothelial cells, epithelial cells, fibroblasts and stem cells and diverse systems that include bone, lymphatics and muscle. The channel has wide-ranging roles and is considered as a target for novel therapeutics in ailments spanning cancers and cardiovascular, dental, gastrointestinal, hepatobiliary, infectious, musculoskeletal, nervous system, ocular, pregnancy, renal, respiratory and urological disorders. The identification of PIEZO1 modulators is in its infancy but useful experimental tools emerged for activating, and to a lesser extent inhibiting, the channels. Elementary structure-activity relationships are known for the Yoda series of small molecule agonists, which show the potential for diverse physicochemical and pharmacological properties. Intriguing effects of Yoda1 include the stimulated removal of excess cerebrospinal fluid. Despite PIEZO1's broad expression, opportunities are suggested for selective positive or negative modulation without intolerable adverse effects. Here we provide a focused, non-systematic, narrative review of progress with this pharmacology and discuss potential future directions for research in the area.
Collapse
Affiliation(s)
- Jacob A Kinsella
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, UK
- School of Chemistry, University of Leeds, Leeds, UK
| | - Marjolaine Debant
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, UK
| | - Gregory Parsonage
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, UK
| | - Lara C Morley
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, UK
| | - Muath Bajarwan
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, UK
| | | | | | - David J Beech
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, UK
| |
Collapse
|
9
|
Mierke CT. Mechanosensory entities and functionality of endothelial cells. Front Cell Dev Biol 2024; 12:1446452. [PMID: 39507419 PMCID: PMC11538060 DOI: 10.3389/fcell.2024.1446452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Accepted: 10/04/2024] [Indexed: 11/08/2024] Open
Abstract
The endothelial cells of the blood circulation are exposed to hemodynamic forces, such as cyclic strain, hydrostatic forces, and shear stress caused by the blood fluid's frictional force. Endothelial cells perceive mechanical forces via mechanosensors and thus elicit physiological reactions such as alterations in vessel width. The mechanosensors considered comprise ion channels, structures linked to the plasma membrane, cytoskeletal spectrin scaffold, mechanoreceptors, and junctional proteins. This review focuses on endothelial mechanosensors and how they alter the vascular functions of endothelial cells. The current state of knowledge on the dysregulation of endothelial mechanosensitivity in disease is briefly presented. The interplay in mechanical perception between endothelial cells and vascular smooth muscle cells is briefly outlined. Finally, future research avenues are highlighted, which are necessary to overcome existing limitations.
Collapse
|
10
|
Abstract
Mechanical forces influence different cell types in our bodies. Among the earliest forces experienced in mammals is blood movement in the vascular system. Blood flow starts at the embryonic stage and ceases when the heart stops. Blood flow exposes endothelial cells (ECs) that line all blood vessels to hemodynamic forces. ECs detect these mechanical forces (mechanosensing) through mechanosensors, thus triggering physiological responses such as changes in vascular diameter. In this review, we focus on endothelial mechanosensing and on how different ion channels, receptors, and membrane structures detect forces and mediate intricate mechanotransduction responses. We further highlight that these responses often reflect collaborative efforts involving several mechanosensors and mechanotransducers. We close with a consideration of current knowledge regarding the dysregulation of endothelial mechanosensing during disease. Because hemodynamic disruptions are hallmarks of cardiovascular disease, studying endothelial mechanosensing holds great promise for advancing our understanding of vascular physiology and pathophysiology.
Collapse
Affiliation(s)
- Xin Rui Lim
- Department of Pharmacology, Larner College of Medicine and Vermont Center for Cardiovascular and Brain Health, University of Vermont, Burlington, Vermont, USA;
| | - Osama F Harraz
- Department of Pharmacology, Larner College of Medicine and Vermont Center for Cardiovascular and Brain Health, University of Vermont, Burlington, Vermont, USA;
| |
Collapse
|
11
|
Beverley KM, Levitan I. Cholesterol regulation of mechanosensitive ion channels. Front Cell Dev Biol 2024; 12:1352259. [PMID: 38333595 PMCID: PMC10850386 DOI: 10.3389/fcell.2024.1352259] [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: 12/07/2023] [Accepted: 01/17/2024] [Indexed: 02/10/2024] Open
Abstract
The purpose of this review is to evaluate the role of cholesterol in regulating mechanosensitive ion channels. Ion channels discussed in this review are sensitive to two types of mechanical signals, fluid shear stress and/or membrane stretch. Cholesterol regulates the channels primarily in two ways: 1) indirectly through localizing the channels into cholesterol-rich membrane domains where they interact with accessory proteins and/or 2) direct binding of cholesterol to the channel at specified putative binding sites. Cholesterol may also regulate channel function via changes of the biophysical properties of the membrane bilayer. Changes in cholesterol affect both mechanosensitivity and basal channel function. We focus on four mechanosensitive ion channels in this review Piezo, Kir2, TRPV4, and VRAC channels. Piezo channels were shown to be regulated by auxiliary proteins that enhance channel function in high cholesterol domains. The direct binding mechanism was shown in Kir2.1 and TRPV4 where cholesterol inhibits channel function. Finally, cholesterol regulation of VRAC was attributed to changes in the physical properties of lipid bilayer. Additional studies should be performed to determine the physiological implications of these sterol effects in complex cellular environments.
Collapse
Affiliation(s)
- Katie M. Beverley
- Division of Pulmonary, Critical Care, Sleep, and Allergy, Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Irena Levitan
- Division of Pulmonary, Critical Care, Sleep, and Allergy, Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| |
Collapse
|
12
|
Zheng M, Borkar NA, Yao Y, Ye X, Vogel ER, Pabelick CM, Prakash YS. Mechanosensitive channels in lung disease. Front Physiol 2023; 14:1302631. [PMID: 38033335 PMCID: PMC10684786 DOI: 10.3389/fphys.2023.1302631] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 11/06/2023] [Indexed: 12/02/2023] Open
Abstract
Mechanosensitive channels (MS channels) are membrane proteins capable of responding to mechanical stress over a wide dynamic range of external mechanical stimuli. In recent years, it has been found that MS channels play an important role as "sentinels" in the process of cell sensing and response to extracellular and intracellular force signals. There is growing appreciation for mechanical activation of ion channels and their subsequent initiation of downstream signaling pathways. Members of the transient receptor potential (TRP) superfamily and Piezo channels are broadly expressed in human tissues and contribute to multiple cellular functions. Both TRP and Piezo channels are thought to play key roles in physiological homeostasis and pathophysiology of disease states including in the lung. Here, we review the current state of knowledge on the expression, regulation, and function of TRP and Piezo channels in the context of the adult lung across the age spectrum, and in lung diseases such as asthma, COPD and pulmonary fibrosis where mechanical forces likely play varied roles in the structural and functional changes characteristic of these diseases. Understanding of TRP and Piezo in the lung can provide insights into new targets for treatment of pulmonary disease.
Collapse
Affiliation(s)
- Mengning Zheng
- Department of Respiratory and Critical Care Medicine, Guizhou Province People’s Hospital, Guiyang, Guizhou, China
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, United States
| | - Niyati A. Borkar
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, United States
| | - Yang Yao
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, United States
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi’an Medical University, Xi’an, Shaanxi, China
| | - Xianwei Ye
- Department of Respiratory and Critical Care Medicine, Guizhou Province People’s Hospital, Guiyang, Guizhou, China
| | - Elizabeth R. Vogel
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, United States
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, United States
| | - Christina M. Pabelick
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, United States
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, United States
| | - Y. S. Prakash
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, United States
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
13
|
Abstract
All cells in the body are exposed to physical force in the form of tension, compression, gravity, shear stress, or pressure. Cells convert these mechanical cues into intracellular biochemical signals; this process is an inherent property of all cells and is essential for numerous cellular functions. A cell's ability to respond to force largely depends on the array of mechanical ion channels expressed on the cell surface. Altered mechanosensing impairs conscious senses, such as touch and hearing, and unconscious senses, like blood pressure regulation and gastrointestinal (GI) activity. The GI tract's ability to sense pressure changes and mechanical force is essential for regulating motility, but it also underlies pain originating in the GI tract. Recent identification of the mechanically activated ion channels Piezo1 and Piezo2 in the gut and the effects of abnormal ion channel regulation on cellular function indicate that these channels may play a pathogenic role in disease. Here, we discuss our current understanding of mechanically activated Piezo channels in the pathogenesis of pancreatic and GI diseases, including pancreatitis, diabetes mellitus, irritable bowel syndrome, GI tumors, and inflammatory bowel disease. We also describe how Piezo channels could be important targets for treating GI diseases.
Collapse
|
14
|
Zong B, Yu F, Zhang X, Pang Y, Zhao W, Sun P, Li L. Mechanosensitive Piezo1 channel in physiology and pathophysiology of the central nervous system. Ageing Res Rev 2023; 90:102026. [PMID: 37532007 DOI: 10.1016/j.arr.2023.102026] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/29/2023] [Accepted: 07/29/2023] [Indexed: 08/04/2023]
Abstract
Since the discovery of the mechanosensitive Piezo1 channel in 2010, there has been a significant amount of research conducted to explore its regulatory role in the physiology and pathology of various organ systems. Recently, a growing body of compelling evidence has emerged linking the activity of the mechanosensitive Piezo1 channel to health and disease of the central nervous system. However, the exact mechanisms underlying these associations remain inadequately comprehended. This review systematically summarizes the current research on the mechanosensitive Piezo1 channel and its implications for central nervous system mechanobiology, retrospects the results demonstrating the regulatory role of the mechanosensitive Piezo1 channel on various cell types within the central nervous system, including neural stem cells, neurons, oligodendrocytes, microglia, astrocytes, and brain endothelial cells. Furthermore, the review discusses the current understanding of the involvement of the Piezo1 channel in central nervous system disorders, such as Alzheimer's disease, multiple sclerosis, glaucoma, stroke, and glioma.
Collapse
Affiliation(s)
- Boyi Zong
- College of Physical Education and Health, East China Normal University, Shanghai 200241, China; Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai 200241, China
| | - Fengzhi Yu
- School of Exercise and Health, Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai 200438, China
| | - Xiaoyou Zhang
- College of Physical Education and Health, East China Normal University, Shanghai 200241, China; Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai 200241, China
| | - Yige Pang
- Department of Neurosurgery, Zibo Central Hospital, Zibo 255000, Shandong, China
| | - Wenrui Zhao
- College of Physical Education and Health Sciences, Zhejiang Normal University, Jinhua 321004, Zhejiang, China
| | - Peng Sun
- College of Physical Education and Health, East China Normal University, Shanghai 200241, China; Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai 200241, China
| | - Lin Li
- College of Physical Education and Health, East China Normal University, Shanghai 200241, China; Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai 200241, China.
| |
Collapse
|
15
|
Di X, Gao X, Peng L, Ai J, Jin X, Qi S, Li H, Wang K, Luo D. Cellular mechanotransduction in health and diseases: from molecular mechanism to therapeutic targets. Signal Transduct Target Ther 2023; 8:282. [PMID: 37518181 PMCID: PMC10387486 DOI: 10.1038/s41392-023-01501-9] [Citation(s) in RCA: 134] [Impact Index Per Article: 67.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 05/10/2023] [Accepted: 05/11/2023] [Indexed: 08/01/2023] Open
Abstract
Cellular mechanotransduction, a critical regulator of numerous biological processes, is the conversion from mechanical signals to biochemical signals regarding cell activities and metabolism. Typical mechanical cues in organisms include hydrostatic pressure, fluid shear stress, tensile force, extracellular matrix stiffness or tissue elasticity, and extracellular fluid viscosity. Mechanotransduction has been expected to trigger multiple biological processes, such as embryonic development, tissue repair and regeneration. However, prolonged excessive mechanical stimulation can result in pathological processes, such as multi-organ fibrosis, tumorigenesis, and cancer immunotherapy resistance. Although the associations between mechanical cues and normal tissue homeostasis or diseases have been identified, the regulatory mechanisms among different mechanical cues are not yet comprehensively illustrated, and no effective therapies are currently available targeting mechanical cue-related signaling. This review systematically summarizes the characteristics and regulatory mechanisms of typical mechanical cues in normal conditions and diseases with the updated evidence. The key effectors responding to mechanical stimulations are listed, such as Piezo channels, integrins, Yes-associated protein (YAP) /transcriptional coactivator with PDZ-binding motif (TAZ), and transient receptor potential vanilloid 4 (TRPV4). We also reviewed the key signaling pathways, therapeutic targets and cutting-edge clinical applications of diseases related to mechanical cues.
Collapse
Affiliation(s)
- Xingpeng Di
- Department of Urology and Institute of Urology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Xiaoshuai Gao
- Department of Urology and Institute of Urology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Liao Peng
- Department of Urology and Institute of Urology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Jianzhong Ai
- Department of Urology and Institute of Urology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Xi Jin
- Department of Urology and Institute of Urology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Shiqian Qi
- Department of Urology and Institute of Urology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, P.R. China
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Hong Li
- Department of Urology and Institute of Urology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Kunjie Wang
- Department of Urology and Institute of Urology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, P.R. China.
| | - Deyi Luo
- Department of Urology and Institute of Urology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, P.R. China.
| |
Collapse
|
16
|
Barnett SD, Asif H, Buxton ILO. Novel identification and modulation of the mechanosensitive Piezo1 channel in human myometrium. J Physiol 2023; 601:1675-1690. [PMID: 35941750 PMCID: PMC9905381 DOI: 10.1113/jp283299] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 06/14/2022] [Indexed: 11/08/2022] Open
Abstract
Approximately 10% of US births deliver preterm before 37 weeks of completed gestation. Premature infants are at risk for life-long debilitating morbidities and death, and spontaneous preterm labour explains 50% of preterm births. In all cases existing treatments are ineffective, and none are FDA approved. The mechanisms that initiate preterm labour are not well understood but may result from dysfunctional regulation of quiescence mechanisms. Human pregnancy is accompanied by large increases in blood flow, and the uterus must enlarge by orders of magnitude to accommodate the growing fetus. This mechanical strain suggests that stretch-activated channels may constitute a mechanism to explain gestational quiescence. Here we identify for the first time that Piezo1, a mechanosensitive cation channel, is present in the uterine smooth muscle and microvascular endothelium of pregnant myometrium. Piezo is downregulated during preterm labour, and stimulation of myometrial Piezo1 in an organ bath with the agonist Yoda1 relaxes the tissue in a dose-dependent fashion. Further, stimulation of Piezo1 while inhibiting protein kinase A, AKT, or endothelial nitric oxide synthase mutes the negative inotropic effects of Piezo1 activation, intimating that actions on the myocyte and endothelial nitric oxide signalling contribute to Piezo1-mediated contractile dynamics. Taken together, these data highlight the importance of stretch-activated channels in pregnancy maintenance and parturition, and identify Piezo1 as a tocolytic target of interest. KEY POINTS: Spontaneous preterm labour is a serious obstetric dilemma without a known cause or effective treatments. Piezo1 is a stretch-activated channel important to muscle contractile dynamics. Piezo1 is present in the myometrium and is dysregulated in women who experience preterm labour. Activation of Piezo1 by the agonist Yoda1 relaxes the myometrium in a dose-dependent fashion, indicating that Piezo1 modulation may have therapeutic benefits to treat preterm labour.
Collapse
Affiliation(s)
- Scott D Barnett
- Department of Pharmacology, Center for Molecular Medicine, Reno School of Medicine, University of Nevada, Reno, NV, USA
| | - Hazik Asif
- Department of Pharmacology, Center for Molecular Medicine, Reno School of Medicine, University of Nevada, Reno, NV, USA
| | - Iain L O Buxton
- Department of Pharmacology, Center for Molecular Medicine, Reno School of Medicine, University of Nevada, Reno, NV, USA
| |
Collapse
|
17
|
Davis MJ, Earley S, Li YS, Chien S. Vascular mechanotransduction. Physiol Rev 2023; 103:1247-1421. [PMID: 36603156 PMCID: PMC9942936 DOI: 10.1152/physrev.00053.2021] [Citation(s) in RCA: 90] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 09/26/2022] [Accepted: 10/04/2022] [Indexed: 01/07/2023] Open
Abstract
This review aims to survey the current state of mechanotransduction in vascular smooth muscle cells (VSMCs) and endothelial cells (ECs), including their sensing of mechanical stimuli and transduction of mechanical signals that result in the acute functional modulation and longer-term transcriptomic and epigenetic regulation of blood vessels. The mechanosensors discussed include ion channels, plasma membrane-associated structures and receptors, and junction proteins. The mechanosignaling pathways presented include the cytoskeleton, integrins, extracellular matrix, and intracellular signaling molecules. These are followed by discussions on mechanical regulation of transcriptome and epigenetics, relevance of mechanotransduction to health and disease, and interactions between VSMCs and ECs. Throughout this review, we offer suggestions for specific topics that require further understanding. In the closing section on conclusions and perspectives, we summarize what is known and point out the need to treat the vasculature as a system, including not only VSMCs and ECs but also the extracellular matrix and other types of cells such as resident macrophages and pericytes, so that we can fully understand the physiology and pathophysiology of the blood vessel as a whole, thus enhancing the comprehension, diagnosis, treatment, and prevention of vascular diseases.
Collapse
Affiliation(s)
- Michael J Davis
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
| | - Scott Earley
- Department of Pharmacology, University of Nevada, Reno, Nevada
| | - Yi-Shuan Li
- Department of Bioengineering, University of California, San Diego, California
- Institute of Engineering in Medicine, University of California, San Diego, California
| | - Shu Chien
- Department of Bioengineering, University of California, San Diego, California
- Institute of Engineering in Medicine, University of California, San Diego, California
- Department of Medicine, University of California, San Diego, California
| |
Collapse
|
18
|
Qiu X, Deng Z, Wang M, Feng Y, Bi L, Li L. Piezo protein determines stem cell fate by transmitting mechanical signals. Hum Cell 2023; 36:540-553. [PMID: 36580272 DOI: 10.1007/s13577-022-00853-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 12/21/2022] [Indexed: 12/30/2022]
Abstract
Piezo ion channel is a mechanosensitive protein on the cell membrane, which contains Piezo1 and Piezo2. Piezo channels are activated by mechanical forces, including stretch, matrix stiffness, static pressure, and shear stress. Piezo channels transmit mechanical signals that cause different downstream responses in the differentiation process, including integrin signaling pathway, ERK1/2 MAPK signaling pathway, Notch signaling, and WNT signaling pathway. In the fate of stem cell differentiation, scientists found differences in Piezo channel expression and found that Piezo channel expression is related to developmental diseases. Here, we briefly review the structure and function of Piezo channels and the relationship between Piezo and mechanical signals, discussing the current understanding of the role of Piezo channels in stem cell fate and associated molecules and developmental diseases. Ultimately, we believe this review will help identify the association between Piezo channels and stem cell fate.
Collapse
Affiliation(s)
- Xiaolei Qiu
- Department of Vascular Surgery, China-Japan Union Hospital of Jilin University, Changchun, 130033, China
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China
| | - Zhuoyue Deng
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China
| | - Meijing Wang
- Department of Pathology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Yuqi Feng
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China
| | - Lintao Bi
- Department of Hematology and Oncology, China-Japan Union Hospital of Jilin University, Changchun, 130033, China.
| | - Lisha Li
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China
| |
Collapse
|
19
|
Dela Justina V, de Freitas RA, Arishe OO, Giachini FR, Webb RC, Priviero F. Piezo1 activation induces relaxation of the pudendal artery and corpus cavernosum. Front Physiol 2023; 14:998951. [PMID: 36846322 PMCID: PMC9950814 DOI: 10.3389/fphys.2023.998951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 01/30/2023] [Indexed: 02/12/2023] Open
Abstract
Piezo1 channel is a sensor for shear-stress in the vasculature. Piezo1 activation induces vasodilation, and its deficiency contributes to vascular disorders, such as hypertension. In this study, we aimed to determine whether Piezo1 channel has a functional role in the dilation of pudendal arteries and corpus cavernosum (CC). For this, male Wistar rats were used, and the relaxation of the pudendal artery and CC was obtained using the Piezo1 activator, Yoda1, in the presence and absence of Dooku (Yoda1 antagonist), GsMTx4 (non-selective mechanosensory channel inhibitor) and L-NAME (nitric oxide synthase inhibitor). In the CC, Yoda1 was also tested in the presence of indomethacin (non-selective COX inhibitor) and tetraethylammonium (TEA, non-selective potassium channel inhibitor). The expression of Piezo1 was confirmed by Western blotting. Our data show that Piezo1 activation leads to the relaxation of the pudendal artery and CC as the chemical activator of Piezo1, Yoda1, relaxed the pudendal artery (47%) and CC (41%). This response was impaired by L-NAME and abolished by Dooku and GsMTx4 in the pudendal artery only. Indomethacin and TEA did not affect the relaxation induced by Yoda1 in the CC. Limited tools to explore this channel prevent further investigation of its underlying mechanisms of action. In conclusion, our data demonstrate that Piezo1 is expressed and induced the relaxation of the pudendal artery and CC. Further studies are necessary to determine its role in penile erection and if erectile dysfunction is associated with Piezo1 deficiency.
Collapse
Affiliation(s)
- Vanessa Dela Justina
- Graduate Program in Biological Sciences, Federal University of Goias, Goiânia, Goias, Brazil
| | - Raiany Alves de Freitas
- Graduate Program in Biological Sciences, Federal University of Goias, Goiânia, Goias, Brazil
| | - Olufunke O. Arishe
- Department of Cell Biology and Anatomy—School of Medicine, University of South Carolina, Columbia, SC, United States,Cardiovascular Translational Research Center—School of Medicine, University of South Carolina, Columbia, SC, United States
| | - Fernanda R. Giachini
- Graduate Program in Biological Sciences, Federal University of Goias, Goiânia, Goias, Brazil,Institute of Biological Sciences and Health, Federal University of Mato Grosso, Barra do Garças, Mato Grosso, Brazil
| | - R. Clinton Webb
- Department of Cell Biology and Anatomy—School of Medicine, University of South Carolina, Columbia, SC, United States,Cardiovascular Translational Research Center—School of Medicine, University of South Carolina, Columbia, SC, United States
| | - Fernanda Priviero
- Department of Cell Biology and Anatomy—School of Medicine, University of South Carolina, Columbia, SC, United States,Cardiovascular Translational Research Center—School of Medicine, University of South Carolina, Columbia, SC, United States,*Correspondence: Fernanda Priviero,
| |
Collapse
|
20
|
Tian S, Cai Z, Sen P, van Uden D, van de Kamp E, Thuillet R, Tu L, Guignabert C, Boomars K, Van der Heiden K, Brandt MM, Merkus D. Loss of lung microvascular endothelial Piezo2 expression impairs NO synthesis, induces EndMT, and is associated with pulmonary hypertension. Am J Physiol Heart Circ Physiol 2022; 323:H958-H974. [PMID: 36149769 DOI: 10.1152/ajpheart.00220.2022] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Mechanical forces are translated into biochemical stimuli by mechanotransduction channels, such as the mechanically activated cation channel Piezo2. Lung Piezo2 expression has recently been shown to be restricted to endothelial cells. Hence, we aimed to investigate the role of Piezo2 in regulation of pulmonary vascular function and structure, as well as its contribution to development of pulmonary arterial hypertension (PAH). The expression of Piezo2 was significantly reduced in pulmonary microvascular endothelial cells (MVECs) from patients with PAH, in lung tissue from mice with a Bmpr2+/R899X knock-in mutation commonly found in patients with pulmonary hypertension, and in lung tissue of monocrotaline (MCT) and sugen-hypoxia-induced PH (SuHx) PAH rat models, as well as from a swine model with pulmonary vein banding. In MVECs, Piezo2 expression was reduced in response to abnormal shear stress, hypoxia, and TGFβ stimulation. Functional studies in MVECs exposed to shear stress illustrated that siRNA-mediated Piezo2 knockdown impaired endothelial alignment, calcium influx, phosphorylation of AKT, and nitric oxide production. In addition, siPiezo2 reduced the expression of the endothelial marker PECAM-1 and increased the expression of vascular smooth muscle markers ACTA2, SM22a, and calponin. Thus, Piezo2 acts as a mechanotransduction channel in pulmonary MVECs, stimulating shear-induced production of nitric oxide and is essentially involved in preventing endothelial to mesenchymal transition. Its blunted expression in pulmonary hypertension could impair the vasodilator capacity and stimulate vascular remodeling, indicating that Piezo2 might be an interesting therapeutic target to attenuate progression of the disease.NEW & NOTEWORTHY The mechanosensory ion channel Piezo2 is exclusively expressed in lung microvascular endothelial cells (MVECs). Patient MVECs as well as animal models of pulmonary (arterial) hypertension showed lower expression of Piezo2 in the lung. Mechanistically, Piezo2 is required for calcium influx and NO production in response to shear stress, whereas stimuli known to induce endothelial to mesenchymal transition (EndMT) reduce Piezo2 expression in MVECs, and Piezo2 knockdown induces a gene and protein expression pattern consistent with EndMT.
Collapse
Affiliation(s)
- Siyu Tian
- Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Zongye Cai
- Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Payel Sen
- Walter Brendel Center of Experimental Medicine, University Clinic Munich, Munich, Germany.,German Center for Cardiovascular Research, Partner Site Munich, Munich Heart Alliance, Munich, Germany
| | - Denise van Uden
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Esther van de Kamp
- Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Raphael Thuillet
- INSERM UMR_S 999, Hôpital Marie Lannelongue, Le Plessis-Robinson, France.,School of Medicine, Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Ly Tu
- INSERM UMR_S 999, Hôpital Marie Lannelongue, Le Plessis-Robinson, France.,School of Medicine, Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Christophe Guignabert
- INSERM UMR_S 999, Hôpital Marie Lannelongue, Le Plessis-Robinson, France.,School of Medicine, Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Karin Boomars
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Kim Van der Heiden
- Biomedical Engineering, Department of Cardiology, Thoraxcenter, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Maarten M Brandt
- Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Daphne Merkus
- Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus University Medical Center, Rotterdam, The Netherlands.,Walter Brendel Center of Experimental Medicine, University Clinic Munich, Munich, Germany.,German Center for Cardiovascular Research, Partner Site Munich, Munich Heart Alliance, Munich, Germany
| |
Collapse
|
21
|
Zhao T, Parmisano S, Soroureddin Z, Zhao M, Yung L, Thistlethwaite PA, Makino A, Yuan JXJ. Mechanosensitive cation currents through TRPC6 and Piezo1 channels in human pulmonary arterial endothelial cells. Am J Physiol Cell Physiol 2022; 323:C959-C973. [PMID: 35968892 PMCID: PMC9485000 DOI: 10.1152/ajpcell.00313.2022] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 08/09/2022] [Accepted: 08/09/2022] [Indexed: 11/22/2022]
Abstract
Mechanosensitive cation channels and Ca2+ influx through these channels play an important role in the regulation of endothelial cell functions. Transient receptor potential canonical channel 6 (TRPC6) is a diacylglycerol-sensitive nonselective cation channel that forms receptor-operated Ca2+ channels in a variety of cell types. Piezo1 is a mechanosensitive cation channel activated by membrane stretch and shear stress in lung endothelial cells. In this study, we report that TRPC6 and Piezo1 channels both contribute to membrane stretch-mediated cation currents and Ca2+ influx or increase in cytosolic-free Ca2+ concentration ([Ca2+]cyt) in human pulmonary arterial endothelial cells (PAECs). The membrane stretch-mediated cation currents and increase in [Ca2+]cyt in human PAECs were significantly decreased by GsMTX4, a blocker of Piezo1 channels, and by BI-749327, a selective blocker of TRPC6 channels. Extracellular application of 1-oleoyl-2-acetyl-sn-glycerol (OAG), a membrane permeable analog of diacylglycerol, rapidly induced whole cell cation currents and increased [Ca2+]cyt in human PAECs and human embryonic kidney (HEK)-cells transiently transfected with the human TRPC6 gene. Furthermore, membrane stretch with hypo-osmotic or hypotonic solution enhances the cation currents in TRPC6-transfected HEK cells. In HEK cells transfected with the Piezo1 gene, however, OAG had little effect on the cation currents, but membrane stretch significantly enhanced the cation currents. These data indicate that, while both TRPC6 and Piezo1 are involved in generating mechanosensitive cation currents and increases in [Ca2+]cyt in human PAECs undergoing mechanical stimulation, only TRPC6 (but not Piezo1) is sensitive to the second messenger diacylglycerol. Selective blockers of these channels may help develop novel therapies for mechanotransduction-associated pulmonary vascular remodeling in patients with pulmonary arterial hypertension.
Collapse
Affiliation(s)
- Tengteng Zhao
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, California
| | - Sophia Parmisano
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, California
| | - Zahra Soroureddin
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, California
| | - Manjia Zhao
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, California
| | - Lauren Yung
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, California
| | - Patricia A Thistlethwaite
- Division of Cardiothoracic Surgery, Department of Surgery, University of California, San Diego, California
| | - Ayako Makino
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, California
| | - Jason X-J Yuan
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, California
| |
Collapse
|
22
|
Xu H, He Y, Hong T, Bi C, Li J, Xia M. Piezo1 in vascular remodeling of atherosclerosis and pulmonary arterial hypertension: A potential therapeutic target. Front Cardiovasc Med 2022; 9:1021540. [PMID: 36247424 PMCID: PMC9557227 DOI: 10.3389/fcvm.2022.1021540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 09/12/2022] [Indexed: 11/13/2022] Open
Abstract
Vascular remodeling (VR) is a structural and functional change of blood vessels to adapt to the changes of internal and external environment. It is one of the common pathological features of many vascular proliferative diseases. The process of VR is mainly manifested in the changes of vascular wall structure and function, including intimal hyperplasia, thickening or thinning of media, fibrosis of adventitia, etc. These changes are also the pathological basis of aging and various cardiovascular diseases. Mechanical force is the basis of cardiovascular biomechanics, and the newly discovered mechanical sensitive ion channel Piezo1 is widely distributed in the whole cardiovascular system. Studies have confirmed that Piezo1, a mechanically sensitive ion channel, plays an important role in cardiovascular remodeling diseases. This article reviews the molecular mechanism of Piezo1 in atherosclerosis, hypertension and pulmonary hypertension, in order to provide a theoretical basis for the further study of vascular remodeling.
Collapse
Affiliation(s)
- Han Xu
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yu He
- Cardiovascular Surgery Department, The First Affiliated Hospital of Xi'an Jiaotong University, Xian, China
| | - Tianying Hong
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Cong Bi
- Department of Vascular Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Jing Li
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
- Jing Li
| | - Mingfeng Xia
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
- *Correspondence: Mingfeng Xia
| |
Collapse
|
23
|
Piezo1 Channel Activation Reverses Pulmonary Artery Vasoconstriction in an Early Rat Model of Pulmonary Hypertension: The Role of Ca2+ Influx and Akt-eNOS Pathway. Cells 2022; 11:cells11152349. [PMID: 35954193 PMCID: PMC9367624 DOI: 10.3390/cells11152349] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 07/18/2022] [Accepted: 07/26/2022] [Indexed: 02/05/2023] Open
Abstract
In intrapulmonary arteries (IPAs), mechanical forces due to blood flow control vessel tone, and these forces change during pulmonary hypertension (PH). Piezo1, a stretch-activated calcium channel, is a sensor of mechanical stress present in both endothelial cells (ECs) and smooth muscle cells (SMCs). The present study investigated the role of Piezo1 on IPA in the chronic hypoxia model of PH. Rats were raised in chronically hypoxic conditions for 1 (1W-CH, early stage) or 3 weeks (3W-CH, late-stage) of PH or in normoxic conditions (Nx). Immunofluorescence labeling and patch-clamping revealed the presence of Piezo1 in both ECs and SMCs. The Piezo1 agonist, Yoda1, induced an IPA contraction in Nx and 3W-CH. Conversely, Yoda1 induced an endothelial nitric oxide (eNOS) dependent relaxation in 1W-CH. In ECs, the Yoda1-mediated intracellular calcium concentration ([Ca2+]i) increase was greater in 1W-CH as compared to Nx. Yoda1 induced an EC hyperpolarization in 1W-CH. The eNOS levels were increased in 1W-CH IPA compared to Nx or 3W-CH PH and Yoda1 activated phosphorylation of Akt (Ser473) and eNOS (Ser1177). Thus, we demonstrated that endothelial Piezo1 contributes to intrapulmonary vascular relaxation by controlling endothelial [Ca2+]i, endothelial-dependent hyperpolarization, and Akt-eNOS pathway activation in the early stage of PH.
Collapse
|
24
|
Xiong H, Yang J, Guo J, Ma A, Wang B, Kang Y. Mechanosensitive Piezo channels mediate the physiological and pathophysiological changes in the respiratory system. Respir Res 2022; 23:196. [PMID: 35906615 PMCID: PMC9338466 DOI: 10.1186/s12931-022-02122-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 07/22/2022] [Indexed: 02/08/2023] Open
Abstract
Mechanosensitive Piezo ion channels were first reported in 2010 in a mouse neuroblastoma cell line, opening up a new field for studying the composition and function of eukaryotic mechanically activated channels. During the past decade, Piezo ion channels were identified in many species, such as bacteria, Drosophila, and mammals. In mammals, basic life activities, such as the sense of touch, proprioception, hearing, vascular development, and blood pressure regulation, depend on the activation of Piezo ion channels. Cumulative evidence suggests that Piezo ion channels play a major role in lung vascular development and function and diseases like pneumonia, pulmonary hypertension, apnea, and other lung-related diseases. In this review, we focused on studies that reported specific functions of Piezos in tissues and emphasized the physiological and pathological effects of their absence or functional mutations on the respiratory system.
Collapse
Affiliation(s)
- Huaiyu Xiong
- Department of Critical Care Medicine, West China Hospital of Sichuan University, No. 17, Section 3, Renmin South Road, Wuhou District, Chengdu, 610000, Sichuan, China
| | - Jing Yang
- Department of Critical Care Medicine, West China Hospital of Sichuan University, No. 17, Section 3, Renmin South Road, Wuhou District, Chengdu, 610000, Sichuan, China
| | - Jun Guo
- Department of Critical Care Medicine, West China Hospital of Sichuan University, No. 17, Section 3, Renmin South Road, Wuhou District, Chengdu, 610000, Sichuan, China
| | - Aijia Ma
- Department of Critical Care Medicine, West China Hospital of Sichuan University, No. 17, Section 3, Renmin South Road, Wuhou District, Chengdu, 610000, Sichuan, China
| | - Bo Wang
- Department of Critical Care Medicine, West China Hospital of Sichuan University, No. 17, Section 3, Renmin South Road, Wuhou District, Chengdu, 610000, Sichuan, China.
| | - Yan Kang
- Department of Critical Care Medicine, West China Hospital of Sichuan University, No. 17, Section 3, Renmin South Road, Wuhou District, Chengdu, 610000, Sichuan, China.
| |
Collapse
|
25
|
Zheng Q, Zou Y, Teng P, Chen Z, Wu Y, Dai X, Li X, Hu Z, Wu S, Xu Y, Zou W, Song H, Ma L. Mechanosensitive Channel PIEZO1 Senses Shear Force to Induce KLF2/4 Expression via CaMKII/MEKK3/ERK5 Axis in Endothelial Cells. Cells 2022; 11:cells11142191. [PMID: 35883633 PMCID: PMC9317998 DOI: 10.3390/cells11142191] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 07/04/2022] [Accepted: 07/11/2022] [Indexed: 01/27/2023] Open
Abstract
Shear stress exerted by the blood stream modulates endothelial functions through altering gene expression. KLF2 and KLF4, the mechanosensitive transcription factors, are promoted by laminar flow to maintain endothelial homeostasis. However, how the expression of KLF2/4 is regulated by shear stress is poorly understood. Here, we showed that the activation of PIEZO1 upregulates the expression of KLF2/4 in endothelial cells. Mice with endothelial-specific deletion of Piezo1 exhibit reduced KLF2/4 expression in thoracic aorta and pulmonary vascular endothelial cells. Mechanistically, shear stress activates PIEZO1, which results in a calcium influx and subsequently activation of CaMKII. CaMKII interacts with and activates MEKK3 to promote MEKK3/MEK5/ERK5 signaling and ultimately induce the transcription of KLF2/4. Our data provide the molecular insight into how endothelial cells sense and convert mechanical stimuli into a biological response to promote KLF2/4 expression for the maintenance of endothelial function and homeostasis.
Collapse
Affiliation(s)
- Qi Zheng
- Department of Cardiovascular Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China; (Q.Z.); (P.T.); (Z.C.); (X.D.); (S.W.)
| | - Yonggang Zou
- The MOE Key Laboratory of Biosystems Homeostasis & Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang 310058, China; (Y.Z.); (Y.W.); (X.L.); (Z.H.); (Y.X.)
| | - Peng Teng
- Department of Cardiovascular Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China; (Q.Z.); (P.T.); (Z.C.); (X.D.); (S.W.)
| | - Zhenghua Chen
- Department of Cardiovascular Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China; (Q.Z.); (P.T.); (Z.C.); (X.D.); (S.W.)
| | - Yuefeng Wu
- The MOE Key Laboratory of Biosystems Homeostasis & Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang 310058, China; (Y.Z.); (Y.W.); (X.L.); (Z.H.); (Y.X.)
| | - Xiaoyi Dai
- Department of Cardiovascular Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China; (Q.Z.); (P.T.); (Z.C.); (X.D.); (S.W.)
| | - Xiya Li
- The MOE Key Laboratory of Biosystems Homeostasis & Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang 310058, China; (Y.Z.); (Y.W.); (X.L.); (Z.H.); (Y.X.)
| | - Zonghao Hu
- The MOE Key Laboratory of Biosystems Homeostasis & Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang 310058, China; (Y.Z.); (Y.W.); (X.L.); (Z.H.); (Y.X.)
| | - Shengjun Wu
- Department of Cardiovascular Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China; (Q.Z.); (P.T.); (Z.C.); (X.D.); (S.W.)
| | - Yanhua Xu
- The MOE Key Laboratory of Biosystems Homeostasis & Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang 310058, China; (Y.Z.); (Y.W.); (X.L.); (Z.H.); (Y.X.)
| | - Weiguo Zou
- CAS Center for Excellence in Molecular Cell Sciences, State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Shanghai 200031, China
- Correspondence: (W.Z.); (H.S.); (L.M.)
| | - Hai Song
- The MOE Key Laboratory of Biosystems Homeostasis & Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang 310058, China; (Y.Z.); (Y.W.); (X.L.); (Z.H.); (Y.X.)
- Correspondence: (W.Z.); (H.S.); (L.M.)
| | - Liang Ma
- Department of Cardiovascular Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China; (Q.Z.); (P.T.); (Z.C.); (X.D.); (S.W.)
- Correspondence: (W.Z.); (H.S.); (L.M.)
| |
Collapse
|
26
|
Suki B, Bates JH, Bartolák-Suki E. Remodeling of the Aged and Emphysematous Lungs: Roles of Microenvironmental Cues. Compr Physiol 2022; 12:3559-3574. [PMID: 35766835 PMCID: PMC11470990 DOI: 10.1002/cphy.c210033] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Aging is a slow process that affects all organs, and the lung is no exception. At the alveolar level, aging increases the airspace size with thicker and stiffer septal walls and straighter and thickened collagen and elastic fibers. This creates a microenvironment that interferes with the ability of cells in the parenchyma to maintain normal homeostasis and respond to injury. These changes also make the lung more susceptible to disease such as emphysema. Emphysema is characterized by slow but progressive remodeling of the deep alveolar regions that leads to airspace enlargement and increased but disorganized elastin and collagen deposition. This remodeling has been attributed to ongoing inflammation that involves inflammatory cells and the cytokines they produce. Cellular senescence, another consequence of aging, weakens the ability of cells to properly respond to injury, something that also occurs in emphysema. These factors conspire to make alveolar walls more prone to mechanical failure, which can set emphysema in motion by driving inflammation through immune stimulation by protein fragments. Both aging and emphysema are influenced by microenvironmental conditions such as local inflammation, chemical makeup, tissue stiffness, and mechanical stresses. Although aging and emphysema are not equivalent, they have the potential to influence each other in synergistic ways; aging sets up the conditions for emphysema to develop, while emphysema may accelerate cellular senescence and thus aging itself. This article focuses on the similarities and differences between the remodeled microenvironment of the aging and emphysematous lung, with special emphasis on the alveolar septal wall. © 2022 American Physiological Society. Compr Physiol 12:3559-3574, 2022.
Collapse
Affiliation(s)
- Béla Suki
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts
| | - Jason H.T. Bates
- Depatment of Medicine, University of Vermont Larner College of Medicine, Burlington, Vermont
| | | |
Collapse
|
27
|
Gao DD, Huang JH, Ding N, Deng WJ, Li PL, Mai YN, Wu JR, Hu M. Mechanosensitive Piezo1 channel in rat epididymal epithelial cells promotes transepithelial K+ secretion. Cell Calcium 2022; 104:102571. [DOI: 10.1016/j.ceca.2022.102571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 03/09/2022] [Accepted: 03/10/2022] [Indexed: 11/28/2022]
|
28
|
Chen J, Rodriguez M, Miao J, Liao J, Jain PP, Zhao M, Zhao T, Babicheva A, Wang Z, Parmisano S, Powers R, Matti M, Paquin C, Soroureddin Z, Shyy JYJ, Thistlethwaite PA, Makino A, Wang J, Yuan JXJ. Mechanosensitive channel Piezo1 is required for pulmonary artery smooth muscle cell proliferation. Am J Physiol Lung Cell Mol Physiol 2022; 322:L737-L760. [PMID: 35318857 PMCID: PMC9076422 DOI: 10.1152/ajplung.00447.2021] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 03/10/2022] [Accepted: 03/17/2022] [Indexed: 01/10/2023] Open
Abstract
Concentric pulmonary vascular wall thickening due partially to increased pulmonary artery (PA) smooth muscle cell (PASMC) proliferation contributes to elevating pulmonary vascular resistance (PVR) in patients with pulmonary hypertension (PH). Although pulmonary vasoconstriction may be an early contributor to increasing PVR, the transition of contractile PASMCs to proliferative PASMCs may play an important role in the development and progression of pulmonary vascular remodeling in PH. A rise in cytosolic Ca2+ concentration ([Ca2+]cyt) is a trigger for PASMC contraction and proliferation. Here, we report that upregulation of Piezo1, a mechanosensitive cation channel, is involved in the contractile-to-proliferative phenotypic transition of PASMCs and potential development of pulmonary vascular remodeling. By comparing freshly isolated PA (contractile PASMCs) and primary cultured PASMCs (from the same rat) in a growth medium (proliferative PASMCs), we found that Piezo1, Notch2/3, and CaSR protein levels were significantly higher in proliferative PASMCs than in contractile PASMCs. Upregulated Piezo1 was associated with an increase in expression of PCNA, a marker for cell proliferation, whereas downregulation (with siRNA) or inhibition (with GsMTx4) of Piezo1 attenuated PASMC proliferation. Furthermore, Piezo1 in the remodeled PA from rats with experimental PH was upregulated compared with PA from control rats. These data indicate that PASMC contractile-to-proliferative phenotypic transition is associated with the transition or adaptation of membrane channels and receptors. Upregulated Piezo1 may play a critical role in PASMC phenotypic transition and PASMC proliferation. Upregulation of Piezo1 in proliferative PASMCs may likely be required to provide sufficient Ca2+ to assure nuclear/cell division and PASMC proliferation, contributing to the development and progression of pulmonary vascular remodeling in PH.
Collapse
Affiliation(s)
- Jiyuan Chen
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of California, San Diego, La Jolla, California
- State Key Laboratory of Respiratory Disease and First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Marisela Rodriguez
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of California, San Diego, La Jolla, California
| | - Jinrui Miao
- State Key Laboratory of Respiratory Disease and First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jing Liao
- State Key Laboratory of Respiratory Disease and First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Pritesh P Jain
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of California, San Diego, La Jolla, California
| | - Manjia Zhao
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of California, San Diego, La Jolla, California
| | - Tengteng Zhao
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of California, San Diego, La Jolla, California
| | - Aleksandra Babicheva
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of California, San Diego, La Jolla, California
| | - Ziyi Wang
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of California, San Diego, La Jolla, California
- State Key Laboratory of Respiratory Disease and First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Sophia Parmisano
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of California, San Diego, La Jolla, California
| | - Ryan Powers
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of California, San Diego, La Jolla, California
| | - Moreen Matti
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of California, San Diego, La Jolla, California
| | - Cole Paquin
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of California, San Diego, La Jolla, California
| | - Zahra Soroureddin
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of California, San Diego, La Jolla, California
| | - John Y-J Shyy
- Division of Cardiovascular Medicine, Department of Medicine, University of California, San Diego, La Jolla, California
| | - Patricia A Thistlethwaite
- Division of Cardiothoracic Surgery, Department of Surgery, University of California, San Diego, La Jolla, California
| | - Ayako Makino
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, La Jolla, California
| | - Jian Wang
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of California, San Diego, La Jolla, California
- State Key Laboratory of Respiratory Disease and First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jason X-J Yuan
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of California, San Diego, La Jolla, California
| |
Collapse
|
29
|
Role of Ion Channel Remodeling in Endothelial Dysfunction Induced by Pulmonary Arterial Hypertension. Biomolecules 2022; 12:biom12040484. [PMID: 35454073 PMCID: PMC9031742 DOI: 10.3390/biom12040484] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/14/2022] [Accepted: 03/15/2022] [Indexed: 12/12/2022] Open
Abstract
Endothelial dysfunction is a key player in advancing vascular pathology in pulmonary arterial hypertension (PAH), a disease essentially characterized by intense remodeling of the pulmonary vasculature, vasoconstriction, endothelial dysfunction, inflammation, oxidative stress, and thrombosis in situ. These vascular features culminate in an increase in pulmonary vascular resistance, subsequent right heart failure, and premature death. Over the past years, there has been a great development in our understanding of pulmonary endothelial biology related to the genetic and molecular mechanisms that modulate the endothelial response to direct or indirect injury and how their dysregulation can promote PAH pathogenesis. Ion channels are key regulators of vasoconstriction and proliferative/apoptotic phenotypes; however, they are poorly studied at the endothelial level. The current review will describe and categorize different expression, functions, regulation, and remodeling of endothelial ion channels (K+, Ca2+, Na+, and Cl− channels) in PAH. We will focus on the potential pathogenic role of ion channel deregulation in the onset and progression of endothelial dysfunction during the development of PAH and its potential therapeutic role.
Collapse
|
30
|
Lin C, Zheng X, Lin S, Zhang Y, Wu J, Li Y. Mechanotransduction Regulates the Interplays Between Alveolar Epithelial and Vascular Endothelial Cells in Lung. Front Physiol 2022; 13:818394. [PMID: 35250619 PMCID: PMC8895143 DOI: 10.3389/fphys.2022.818394] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 01/28/2022] [Indexed: 12/22/2022] Open
Abstract
Mechanical stress plays a critical role among development, functional maturation, and pathogenesis of pulmonary tissues, especially for the alveolar epithelial cells and vascular endothelial cells located in the microenvironment established with vascular network and bronchial-alveolar network. Alveolar epithelial cells are mainly loaded by cyclic strain and air pressure tension. While vascular endothelial cells are exposed to shear stress and cyclic strain. Currently, the emerging evidences demonstrated that non-physiological mechanical forces would lead to several pulmonary diseases, including pulmonary hypertension, fibrosis, and ventilation induced lung injury. Furthermore, a series of intracellular signaling had been identified to be involved in mechanotransduction and participated in regulating the physiological homeostasis and pathophysiological process. Besides, the communications between alveolar epithelium and vascular endothelium under non-physiological stress contribute to the remodeling of the pulmonary micro-environment in collaboration, including hypoxia induced injuries, endothelial permeability impairment, extracellular matrix stiffness elevation, metabolic alternation, and inflammation activation. In this review, we aim to summarize the current understandings of mechanotransduction on the relation between mechanical forces acting on the lung and biological response in mechanical overloading related diseases. We also would like to emphasize the interplays between alveolar epithelium and vascular endothelium, providing new insights into pulmonary diseases pathogenesis, and potential targets for therapy.
Collapse
Affiliation(s)
- Chuyang Lin
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Xiaolan Zheng
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Sha Lin
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Yue Zhang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Jinlin Wu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Yifei Li
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
31
|
Barbeau S, Porto Ribeiro T, Gilbert G, Cardouat G, Baudrimont I, Freund-Michel V, Guibert C, Marthan R, Vacher P, Quignard JF, Ducret T. Implication des canaux mécanosensibles Piezo1 et TRPV4 dans l’hypertension pulmonaire. Rev Mal Respir 2022; 39:79-83. [DOI: 10.1016/j.rmr.2022.01.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 01/05/2022] [Indexed: 11/28/2022]
|
32
|
Wang Z, Chen J, Babicheva A, Jain PP, Rodriguez M, Ayon RJ, Ravellette KS, Wu L, Balistrieri F, Tang H, Wu X, Zhao T, Black SM, Desai AA, Garcia JGN, Sun X, Shyy JYJ, Valdez-Jasso D, Thistlethwaite PA, Makino A, Wang J, Yuan JXJ. Endothelial upregulation of mechanosensitive channel Piezo1 in pulmonary hypertension. Am J Physiol Cell Physiol 2021; 321:C1010-C1027. [PMID: 34669509 PMCID: PMC8714987 DOI: 10.1152/ajpcell.00147.2021] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 09/22/2021] [Accepted: 10/12/2021] [Indexed: 12/16/2022]
Abstract
Piezo is a mechanosensitive cation channel responsible for stretch-mediated Ca2+ and Na+ influx in multiple types of cells. Little is known about the functional role of Piezo1 in the lung vasculature and its potential pathogenic role in pulmonary arterial hypertension (PAH). Pulmonary arterial endothelial cells (PAECs) are constantly under mechanic stretch and shear stress that are sufficient to activate Piezo channels. Here, we report that Piezo1 is significantly upregulated in PAECs from patients with idiopathic PAH and animals with experimental pulmonary hypertension (PH) compared with normal controls. Membrane stretch by decreasing extracellular osmotic pressure or by cyclic stretch (18% CS) increases Ca2+-dependent phosphorylation (p) of AKT and ERK, and subsequently upregulates expression of Notch ligands, Jagged1/2 (Jag-1 and Jag-2), and Delta like-4 (DLL4) in PAECs. siRNA-mediated downregulation of Piezo1 significantly inhibited the stretch-mediated pAKT increase and Jag-1 upregulation, whereas downregulation of AKT by siRNA markedly attenuated the stretch-mediated Jag-1 upregulation in human PAECs. Furthermore, the mRNA and protein expression level of Piezo1 in the isolated pulmonary artery, which mainly contains pulmonary arterial smooth muscle cells (PASMCs), from animals with severe PH was also significantly higher than that from control animals. Intraperitoneal injection of a Piezo1 channel blocker, GsMTx4, ameliorated experimental PH in mice. Taken together, our study suggests that membrane stretch-mediated Ca2+ influx through Piezo1 is an important trigger for pAKT-mediated upregulation of Jag-1 in PAECs. Upregulation of the mechanosensitive channel Piezo1 and the resultant increase in the Notch ligands (Jag-1/2 and DLL4) in PAECs may play a critical pathogenic role in the development of pulmonary vascular remodeling in PAH and PH.
Collapse
Affiliation(s)
- Ziyi Wang
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, California
- Departments of Medicine and Physiology, The University of Arizona College of Medicine, Tucson, Arizona
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jiyuan Chen
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, California
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Aleksandra Babicheva
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, California
| | - Pritesh P Jain
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, California
| | - Marisela Rodriguez
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, California
- Departments of Medicine and Physiology, The University of Arizona College of Medicine, Tucson, Arizona
| | - Ramon J Ayon
- Departments of Medicine and Physiology, The University of Arizona College of Medicine, Tucson, Arizona
| | - Keeley S Ravellette
- Departments of Medicine and Physiology, The University of Arizona College of Medicine, Tucson, Arizona
| | - Linda Wu
- Departments of Medicine and Physiology, The University of Arizona College of Medicine, Tucson, Arizona
| | - Francesca Balistrieri
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, California
| | - Haiyang Tang
- Departments of Medicine and Physiology, The University of Arizona College of Medicine, Tucson, Arizona
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiaomin Wu
- Departments of Medicine and Physiology, The University of Arizona College of Medicine, Tucson, Arizona
| | - Tengteng Zhao
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, California
| | - Stephen M Black
- Departments of Medicine and Physiology, The University of Arizona College of Medicine, Tucson, Arizona
| | - Ankit A Desai
- Departments of Medicine and Physiology, The University of Arizona College of Medicine, Tucson, Arizona
- Department of Medicine, Indiana University, Indianapolis, Indiana
| | - Joe G N Garcia
- Departments of Medicine and Physiology, The University of Arizona College of Medicine, Tucson, Arizona
| | - Xin Sun
- Department of Pediatrics, University of California, San Diego, La Jolla, California
| | - John Y-J Shyy
- Division of Cardiovascular Medicine, Department of Medicine, University of California, San Diego, La Jolla, California
| | - Daniela Valdez-Jasso
- Department of Bioengineering, University of California, San Diego, La Jolla, California
| | | | - Ayako Makino
- Division of Endocrinology and Metabolism, University of California, San Diego, La Jolla, California
- Departments of Medicine and Physiology, The University of Arizona College of Medicine, Tucson, Arizona
| | - Jian Wang
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, California
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jason X-J Yuan
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, California
- Departments of Medicine and Physiology, The University of Arizona College of Medicine, Tucson, Arizona
| |
Collapse
|
33
|
Favre J, Vessieres E, Guihot AL, Proux C, Grimaud L, Rivron J, Garcia MC, Réthoré L, Zahreddine R, Davezac M, Fébrissy C, Adlanmerini M, Loufrani L, Procaccio V, Foidart JM, Flouriot G, Lenfant F, Fontaine C, Arnal JF, Henrion D. Membrane estrogen receptor alpha (ERα) participates in flow-mediated dilation in a ligand-independent manner. eLife 2021; 10:68695. [PMID: 34842136 PMCID: PMC8676342 DOI: 10.7554/elife.68695] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 11/26/2021] [Indexed: 12/13/2022] Open
Abstract
Estrogen receptor alpha (ERα) activation by estrogens prevents atheroma through its nuclear action, whereas plasma membrane-located ERα accelerates endothelial healing. The genetic deficiency of ERα was associated with a reduction in flow-mediated dilation (FMD) in one man. Here, we evaluated ex vivo the role of ERα on FMD of resistance arteries. FMD, but not agonist (acetylcholine, insulin)-mediated dilation, was reduced in male and female mice lacking ERα (Esr1-/- mice) compared to wild-type mice and was not dependent on the presence of estrogens. In C451A-ERα mice lacking membrane ERα, not in mice lacking AF2-dependent nuclear ERα actions, FMD was reduced, and restored by antioxidant treatments. Compared to wild-type mice, isolated perfused kidneys of C451A-ERα mice revealed a decreased flow-mediated nitrate production and an increased H2O2 production. Thus, endothelial membrane ERα promotes NO bioavailability through inhibition of oxidative stress and thereby participates in FMD in a ligand-independent manner.
Collapse
Affiliation(s)
- Julie Favre
- Angers University, MITOVASC, CNRS UMR 6015, INSERM U1083, Angers, France
| | - Emilie Vessieres
- Angers University, MITOVASC, CNRS UMR 6015, INSERM U1083, Angers, France.,CARFI facility, Angers University, Angers, France
| | - Anne-Laure Guihot
- Angers University, MITOVASC, CNRS UMR 6015, INSERM U1083, Angers, France.,CARFI facility, Angers University, Angers, France
| | - Coralyne Proux
- Angers University, MITOVASC, CNRS UMR 6015, INSERM U1083, Angers, France.,CARFI facility, Angers University, Angers, France
| | - Linda Grimaud
- Angers University, MITOVASC, CNRS UMR 6015, INSERM U1083, Angers, France
| | - Jordan Rivron
- Angers University, MITOVASC, CNRS UMR 6015, INSERM U1083, Angers, France.,CARFI facility, Angers University, Angers, France
| | - Manuela Cl Garcia
- Angers University, MITOVASC, CNRS UMR 6015, INSERM U1083, Angers, France.,CARFI facility, Angers University, Angers, France
| | - Léa Réthoré
- Angers University, MITOVASC, CNRS UMR 6015, INSERM U1083, Angers, France
| | - Rana Zahreddine
- INSERM U1297, Paul Sabatier University (Toulouse III) , University Hospital (UHC) of Toulouse, Toulouse, France
| | - Morgane Davezac
- INSERM U1297, Paul Sabatier University (Toulouse III) , University Hospital (UHC) of Toulouse, Toulouse, France
| | - Chanaelle Fébrissy
- INSERM U1297, Paul Sabatier University (Toulouse III) , University Hospital (UHC) of Toulouse, Toulouse, France
| | - Marine Adlanmerini
- INSERM U1297, Paul Sabatier University (Toulouse III) , University Hospital (UHC) of Toulouse, Toulouse, France
| | - Laurent Loufrani
- Angers University, MITOVASC, CNRS UMR 6015, INSERM U1083, Angers, France.,University Hospital (CHU) of Angers, Angers, France
| | - Vincent Procaccio
- Angers University, MITOVASC, CNRS UMR 6015, INSERM U1083, Angers, France.,University Hospital (CHU) of Angers, Angers, France
| | - Jean-Michel Foidart
- Groupe Interdisciplinaire de Génoprotéomique Appliquée, Université de Liège, Liège, Belgium
| | - Gilles Flouriot
- INSERM U1085, IRSET (Institut de Recherche en Santé, Environnement et Travail), University of Rennes, Rennes, France
| | - Françoise Lenfant
- INSERM U1297, Paul Sabatier University (Toulouse III) , University Hospital (UHC) of Toulouse, Toulouse, France
| | - Coralie Fontaine
- INSERM U1297, Paul Sabatier University (Toulouse III) , University Hospital (UHC) of Toulouse, Toulouse, France
| | - Jean-François Arnal
- INSERM U1297, Paul Sabatier University (Toulouse III) , University Hospital (UHC) of Toulouse, Toulouse, France
| | - Daniel Henrion
- Angers University, MITOVASC, CNRS UMR 6015, INSERM U1083, Angers, France.,CARFI facility, Angers University, Angers, France.,University Hospital (CHU) of Angers, Angers, France
| |
Collapse
|
34
|
Qin L, He T, Chen S, Yang D, Yi W, Cao H, Xiao G. Roles of mechanosensitive channel Piezo1/2 proteins in skeleton and other tissues. Bone Res 2021; 9:44. [PMID: 34667178 PMCID: PMC8526690 DOI: 10.1038/s41413-021-00168-8] [Citation(s) in RCA: 95] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 06/16/2021] [Accepted: 07/08/2021] [Indexed: 12/12/2022] Open
Abstract
Mechanotransduction is a fundamental ability that allows living organisms to receive and respond to physical signals from both the external and internal environments. The mechanotransduction process requires a range of special proteins termed mechanotransducers to convert mechanical forces into biochemical signals in cells. The Piezo proteins are mechanically activated nonselective cation channels and the largest plasma membrane ion channels reported thus far. The regulation of two family members, Piezo1 and Piezo2, has been reported to have essential functions in mechanosensation and transduction in different organs and tissues. Recently, the predominant contributions of the Piezo family were reported to occur in the skeletal system, especially in bone development and mechano-stimulated bone homeostasis. Here we review current studies focused on the tissue-specific functions of Piezo1 and Piezo2 in various backgrounds with special highlights on their importance in regulating skeletal cell mechanotransduction. In this review, we emphasize the diverse functions of Piezo1 and Piezo2 and related signaling pathways in osteoblast lineage cells and chondrocytes. We also summarize our current understanding of Piezo channel structures and the key findings about PIEZO gene mutations in human diseases.
Collapse
Affiliation(s)
- Lei Qin
- Department of Orthopedics, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, Guangdong, China
- Department of Biochemistry, School of Medicine, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Tailin He
- Department of Biochemistry, School of Medicine, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Sheng Chen
- Department of Biochemistry, School of Medicine, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, Shenzhen, Guangdong, China
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Dazhi Yang
- Department of Orthopedics, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Weihong Yi
- Department of Orthopedics, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, Guangdong, China.
| | - Huiling Cao
- Department of Biochemistry, School of Medicine, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, Shenzhen, Guangdong, China.
| | - Guozhi Xiao
- Department of Biochemistry, School of Medicine, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, Shenzhen, Guangdong, China.
| |
Collapse
|
35
|
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
|
36
|
Huang W, Liu H, Pan Y, Yang H, Lin J, Zhang H. Mechanical stretching of the pulmonary vein mediates pulmonary hypertension due to left heart disease by regulating SAC/MAPK pathway and the expression of IL-6 and TNF-α. J Cardiothorac Surg 2021; 16:127. [PMID: 33971931 PMCID: PMC8107413 DOI: 10.1186/s13019-021-01471-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Accepted: 04/05/2021] [Indexed: 01/09/2023] Open
Abstract
Background This study aimed to explore whether the mechanical stretching-induced expression of interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α) in pulmonary veins occurred through the stretch-activated channel (SAC)/ mitogen-activated protein kinases (MAPKs) pathway. Methods Sixty male Sprague-Dawley rats were divided into three sham groups and seven model groups. A metal clip was placed on the ascending aorta in the model group to establish PH-LHD rat model. The sham group received a similar operation without ascending aorta clamped. On day 25, pulmonary vein was given mechanical stretching with 0 g, 2.0 g tension in two model groups and two sham groups. Another four model groups were given 2.0 g tension after MAPKs pathway inhibitors soaked. The last sham group and model group rats’ pulmonary veins, pulmonary artery and lung tissues were obtained on day 35. Pulmonary vein, pulmonary artery and lung tissue were evaluated by echocardiography, HE staining, immunohistochemistry and western blotting respectively. Results On day 25, left heart weight, right ventricular pressure (35.339 cmH2O) and left atrial pressure (13.657 cmH2O) were increased in model group than those in sham group. Echocardiography showed left heart failure in the PH-LHD group (Interventrieular septum dimension 1.716 mm, left ventricular internal end diastolic dimension 4.888 mm, left ventricular posterior wall thickness in diastole 1.749 mm, ejection fraction 76.917%). But there was no difference in lung tissue between the sham group and PH-LHD group as showed by HE staining. Our results showed that the expression of IL-6 and TNF-α was highly expressed in PH-LHD rats’ serum and pulmonary vein, which were further increased after 2.0 g tension was given and were decreased after SAC/MAPKs inhibitors treatment. Meanwhile, on day 25, immunohistochemistry analysis showed the expression of IL-6 and TNF-α was higher in the PH-LHD rats’ pulmonary vein than that in pulmonary artery and lung tissue, and these expressions in pulmonary vein of PH-LHD group were also higher than that in sham group. However, on day 35, IL-6 and TNF-α were all increased in the pulmonary veins, arteries and lung tissues. Besides, our results uncovered that SAC/MAPKs pathway were upregulating in PH-LHD rats’ pulmonary vein. Conclusion In conclusion, pulmonary vein mechanical stretching exacerbated PH-LHD possibly through the SAC/MAPKs pathway and upregulating expression of IL-6 and TNF-α.
Collapse
Affiliation(s)
- Wenhui Huang
- Department of Cardiovascular Surgery, Union Hospital, Fujian Medical University, Fuzhou, 350001, Fujian Province, People's Republic of China.,Anesthesiology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350004, Fujian Province, People's Republic of China
| | - Hongjin Liu
- Department of Cardiovascular Surgery, Union Hospital, Fujian Medical University, Fuzhou, 350001, Fujian Province, People's Republic of China
| | - Yichao Pan
- Department of Cardiovascular Surgery, Union Hospital, Fujian Medical University, Fuzhou, 350001, Fujian Province, People's Republic of China
| | - Hongwei Yang
- Department of Cardiovascular Surgery, Union Hospital, Fujian Medical University, Fuzhou, 350001, Fujian Province, People's Republic of China
| | - Jing Lin
- Department of Cardiovascular Surgery, Union Hospital, Fujian Medical University, Fuzhou, 350001, Fujian Province, People's Republic of China
| | - Hui Zhang
- Department of Intensive Care Unit, Union Hospital, Fujian Medical University, Fuzhou, 350004, Fujian Province, People's Republic of China.
| |
Collapse
|
37
|
Liao J, Lu W, Chen Y, Duan X, Zhang C, Luo X, Lin Z, Chen J, Liu S, Yan H, Chen Y, Feng H, Zhou D, Chen X, Zhang Z, Yang Q, Liu X, Tang H, Li J, Makino A, Yuan JXJ, Zhong N, Yang K, Wang J. Upregulation of Piezo1 (Piezo Type Mechanosensitive Ion Channel Component 1) Enhances the Intracellular Free Calcium in Pulmonary Arterial Smooth Muscle Cells From Idiopathic Pulmonary Arterial Hypertension Patients. Hypertension 2021; 77:1974-1989. [PMID: 33813851 DOI: 10.1161/hypertensionaha.120.16629] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Jing Liao
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, China (J. Liao, W.L., Yuqin Chen, C.Z., X. Luo, Z.L., J.C., S.L., H.Y., Yilin Chen, H.F., D.Z., X.C., Z.Z., Q.Y., X. Liu, H.T., N.Z., K.Y., J.W.)
| | - Wenju Lu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, China (J. Liao, W.L., Yuqin Chen, C.Z., X. Luo, Z.L., J.C., S.L., H.Y., Yilin Chen, H.F., D.Z., X.C., Z.Z., Q.Y., X. Liu, H.T., N.Z., K.Y., J.W.)
| | - Yuqin Chen
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, China (J. Liao, W.L., Yuqin Chen, C.Z., X. Luo, Z.L., J.C., S.L., H.Y., Yilin Chen, H.F., D.Z., X.C., Z.Z., Q.Y., X. Liu, H.T., N.Z., K.Y., J.W.)
| | - Xin Duan
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (X.D.)
| | - Chenting Zhang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, China (J. Liao, W.L., Yuqin Chen, C.Z., X. Luo, Z.L., J.C., S.L., H.Y., Yilin Chen, H.F., D.Z., X.C., Z.Z., Q.Y., X. Liu, H.T., N.Z., K.Y., J.W.)
| | - Xiaoyun Luo
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, China (J. Liao, W.L., Yuqin Chen, C.Z., X. Luo, Z.L., J.C., S.L., H.Y., Yilin Chen, H.F., D.Z., X.C., Z.Z., Q.Y., X. Liu, H.T., N.Z., K.Y., J.W.)
| | - Ziying Lin
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, China (J. Liao, W.L., Yuqin Chen, C.Z., X. Luo, Z.L., J.C., S.L., H.Y., Yilin Chen, H.F., D.Z., X.C., Z.Z., Q.Y., X. Liu, H.T., N.Z., K.Y., J.W.)
| | - Jiyuan Chen
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, China (J. Liao, W.L., Yuqin Chen, C.Z., X. Luo, Z.L., J.C., S.L., H.Y., Yilin Chen, H.F., D.Z., X.C., Z.Z., Q.Y., X. Liu, H.T., N.Z., K.Y., J.W.).,Department of Medicine, University of California, San Diego, La Jolla (J.C., A.M., J.X.-J.Y., J.W.)
| | - Shiyun Liu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, China (J. Liao, W.L., Yuqin Chen, C.Z., X. Luo, Z.L., J.C., S.L., H.Y., Yilin Chen, H.F., D.Z., X.C., Z.Z., Q.Y., X. Liu, H.T., N.Z., K.Y., J.W.)
| | - Han Yan
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, China (J. Liao, W.L., Yuqin Chen, C.Z., X. Luo, Z.L., J.C., S.L., H.Y., Yilin Chen, H.F., D.Z., X.C., Z.Z., Q.Y., X. Liu, H.T., N.Z., K.Y., J.W.)
| | - Yilin Chen
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, China (J. Liao, W.L., Yuqin Chen, C.Z., X. Luo, Z.L., J.C., S.L., H.Y., Yilin Chen, H.F., D.Z., X.C., Z.Z., Q.Y., X. Liu, H.T., N.Z., K.Y., J.W.)
| | - Huazhuo Feng
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, China (J. Liao, W.L., Yuqin Chen, C.Z., X. Luo, Z.L., J.C., S.L., H.Y., Yilin Chen, H.F., D.Z., X.C., Z.Z., Q.Y., X. Liu, H.T., N.Z., K.Y., J.W.)
| | - Dansha Zhou
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, China (J. Liao, W.L., Yuqin Chen, C.Z., X. Luo, Z.L., J.C., S.L., H.Y., Yilin Chen, H.F., D.Z., X.C., Z.Z., Q.Y., X. Liu, H.T., N.Z., K.Y., J.W.)
| | - Xu Chen
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, China (J. Liao, W.L., Yuqin Chen, C.Z., X. Luo, Z.L., J.C., S.L., H.Y., Yilin Chen, H.F., D.Z., X.C., Z.Z., Q.Y., X. Liu, H.T., N.Z., K.Y., J.W.)
| | - Zizhou Zhang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, China (J. Liao, W.L., Yuqin Chen, C.Z., X. Luo, Z.L., J.C., S.L., H.Y., Yilin Chen, H.F., D.Z., X.C., Z.Z., Q.Y., X. Liu, H.T., N.Z., K.Y., J.W.)
| | - Qifeng Yang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, China (J. Liao, W.L., Yuqin Chen, C.Z., X. Luo, Z.L., J.C., S.L., H.Y., Yilin Chen, H.F., D.Z., X.C., Z.Z., Q.Y., X. Liu, H.T., N.Z., K.Y., J.W.)
| | - Xinyi Liu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, China (J. Liao, W.L., Yuqin Chen, C.Z., X. Luo, Z.L., J.C., S.L., H.Y., Yilin Chen, H.F., D.Z., X.C., Z.Z., Q.Y., X. Liu, H.T., N.Z., K.Y., J.W.)
| | - Haiyang Tang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, China (J. Liao, W.L., Yuqin Chen, C.Z., X. Luo, Z.L., J.C., S.L., H.Y., Yilin Chen, H.F., D.Z., X.C., Z.Z., Q.Y., X. Liu, H.T., N.Z., K.Y., J.W.)
| | - Jing Li
- Lingnan Medical Research Center, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, China (J. Li)
| | - Ayako Makino
- Department of Medicine, University of California, San Diego, La Jolla (J.C., A.M., J.X.-J.Y., J.W.)
| | - Jason X-J Yuan
- Department of Medicine, University of California, San Diego, La Jolla (J.C., A.M., J.X.-J.Y., J.W.)
| | - Nanshan Zhong
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, China (J. Liao, W.L., Yuqin Chen, C.Z., X. Luo, Z.L., J.C., S.L., H.Y., Yilin Chen, H.F., D.Z., X.C., Z.Z., Q.Y., X. Liu, H.T., N.Z., K.Y., J.W.)
| | - Kai Yang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, China (J. Liao, W.L., Yuqin Chen, C.Z., X. Luo, Z.L., J.C., S.L., H.Y., Yilin Chen, H.F., D.Z., X.C., Z.Z., Q.Y., X. Liu, H.T., N.Z., K.Y., J.W.)
| | - Jian Wang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, China (J. Liao, W.L., Yuqin Chen, C.Z., X. Luo, Z.L., J.C., S.L., H.Y., Yilin Chen, H.F., D.Z., X.C., Z.Z., Q.Y., X. Liu, H.T., N.Z., K.Y., J.W.).,Department of Medicine, University of California, San Diego, La Jolla (J.C., A.M., J.X.-J.Y., J.W.)
| |
Collapse
|
38
|
Ottolini M, Sonkusare SK. The Calcium Signaling Mechanisms in Arterial Smooth Muscle and Endothelial Cells. Compr Physiol 2021; 11:1831-1869. [PMID: 33792900 PMCID: PMC10388069 DOI: 10.1002/cphy.c200030] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The contractile state of resistance arteries and arterioles is a crucial determinant of blood pressure and blood flow. Physiological regulation of arterial contractility requires constant communication between endothelial and smooth muscle cells. Various Ca2+ signals and Ca2+ -sensitive targets ensure dynamic control of intercellular communications in the vascular wall. The functional effect of a Ca2+ signal on arterial contractility depends on the type of Ca2+ -sensitive target engaged by that signal. Recent studies using advanced imaging methods have identified the spatiotemporal signatures of individual Ca2+ signals that control arterial and arteriolar contractility. Broadly speaking, intracellular Ca2+ is increased by ion channels and transporters on the plasma membrane and endoplasmic reticular membrane. Physiological roles for many vascular Ca2+ signals have already been confirmed, while further investigation is needed for other Ca2+ signals. This article focuses on endothelial and smooth muscle Ca2+ signaling mechanisms in resistance arteries and arterioles. We discuss the Ca2+ entry pathways at the plasma membrane, Ca2+ release signals from the intracellular stores, the functional and physiological relevance of Ca2+ signals, and their regulatory mechanisms. Finally, we describe the contribution of abnormal endothelial and smooth muscle Ca2+ signals to the pathogenesis of vascular disorders. © 2021 American Physiological Society. Compr Physiol 11:1831-1869, 2021.
Collapse
Affiliation(s)
- Matteo Ottolini
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia, USA
| | - Swapnil K Sonkusare
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia, USA.,Department of Molecular Physiology & Biological Physics, University of Virginia, Charlottesville, Virginia, USA.,Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
39
|
Dolgorukova A, Isaeva JE, Verbitskaya E, Lyubashina OA, Giniatullin RА, Sokolov AY. Differential effects of the Piezo1 agonist Yoda1 in the trigeminovascular system: An electrophysiological and intravital microscopy study in rats. Exp Neurol 2021; 339:113634. [PMID: 33549548 DOI: 10.1016/j.expneurol.2021.113634] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 01/18/2021] [Accepted: 02/01/2021] [Indexed: 01/15/2023]
Abstract
Migraine is associated with the activation and sensitisation of the trigeminovascular system and is often accompanied by mechanical hyperalgesia and allodynia. The mechanisms of mechanotransduction during a migraine attack are yet unknown. We have proposed that the ion channel Piezo1 may be involved, since it is expressed in endothelial cells as well as in trigeminal ganglion neurons, and thus, may contribute to the activation of both the vascular and neuronal component of the trigeminovascular system. We took advantage of extracellular recordings from the trigeminocervical complex - a key relay centre in the migraine pain pathway, to directly assess the impact of the differently applied Piezo1 agonist Yoda1 on the sensory processing at the spinal level. At a low dose, Yoda1 slightly facilitated the ongoing firing of central trigeminovascular neurons, however, at a high dose, this substance contributed to the suppression of their activity. Using intravital microscopy, we have revealed that Yoda1 at high dose can also induce the dilation of meningeal arteries innervated by trigeminal afferents. Collectively, here we have identified both neuronal and vascular modulation via selective activation of mechanosensitive Piezo1 channels, which provide new evidence in favour of the Piezo1 role in migraine pathogenesis. We propose several mechanisms that may underlie the revealed effects of Yoda1.
Collapse
Affiliation(s)
- Antonina Dolgorukova
- Valdman Institute of Pharmacology, Pavlov First Saint Petersburg State Medical University, Saint Petersburg 197022, Russia.
| | - Julia E Isaeva
- Valdman Institute of Pharmacology, Pavlov First Saint Petersburg State Medical University, Saint Petersburg 197022, Russia
| | - Elena Verbitskaya
- Valdman Institute of Pharmacology, Pavlov First Saint Petersburg State Medical University, Saint Petersburg 197022, Russia
| | - Olga A Lyubashina
- Valdman Institute of Pharmacology, Pavlov First Saint Petersburg State Medical University, Saint Petersburg 197022, Russia; Laboratory of Cortico-Visceral Physiology, Pavlov Institute of Physiology of the Russian Academy of Sciences, Saint Petersburg 199034, Russia
| | - Rashid А Giniatullin
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio 70211, Finland
| | - Alexey Y Sokolov
- Valdman Institute of Pharmacology, Pavlov First Saint Petersburg State Medical University, Saint Petersburg 197022, Russia; Laboratory of Cortico-Visceral Physiology, Pavlov Institute of Physiology of the Russian Academy of Sciences, Saint Petersburg 199034, Russia
| |
Collapse
|
40
|
A New Hope in Spinal Degenerative Diseases: Piezo1. BIOMED RESEARCH INTERNATIONAL 2021; 2021:6645193. [PMID: 33575334 PMCID: PMC7857891 DOI: 10.1155/2021/6645193] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 01/09/2021] [Indexed: 12/19/2022]
Abstract
As a newly discovered mechanosensitive ion channel protein, the piezo1 protein participates in the transmission of mechanical signals on the cell membrane and plays a vital role in mammalian biomechanics. Piezo1 has attracted widespread attention since it was discovered in 2010. In recent years, studies on piezo1 have gradually increased and deepened. In addition to the discovery that piezo1 is expressed in the respiratory, cardiovascular, gastrointestinal, and urinary systems, it is also stably expressed in cells such as mesenchymal stem cells (MSCs), osteoblasts, osteoclasts, chondrocytes, and nucleus pulposus cells that constitute vertebral bodies and intervertebral discs. They can all receive external mechanical stimulation through the piezo1 protein channel to affect cell proliferation, differentiation, migration, and apoptosis to promote the occurrence and development of lumbar degenerative diseases. Through reviewing the relevant literature of piezo1 in the abovementioned cells, this paper discusses the effect of piezo1 protein expression under mechanical stress stimuli on spinal degenerative disease, providing the molecular basis for the pathological mechanism of spinal degenerative disease and also a new basis, ideas, and methods for the prevention and treatment of this degenerative disease.
Collapse
|
41
|
Fang XZ, Zhou T, Xu JQ, Wang YX, Sun MM, He YJ, Pan SW, Xiong W, Peng ZK, Gao XH, Shang Y. Structure, kinetic properties and biological function of mechanosensitive Piezo channels. Cell Biosci 2021; 11:13. [PMID: 33422128 PMCID: PMC7796548 DOI: 10.1186/s13578-020-00522-z] [Citation(s) in RCA: 115] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 12/16/2020] [Indexed: 02/06/2023] Open
Abstract
Mechanotransduction couples mechanical stimulation with ion flux, which is critical for normal biological processes involved in neuronal cell development, pain sensation, and red blood cell volume regulation. Although they are key mechanotransducers, mechanosensitive ion channels in mammals have remained difficult to identify. In 2010, Coste and colleagues revealed a novel family of mechanically activated cation channels in eukaryotes, consisting of Piezo1 and Piezo2 channels. These have been proposed as the long-sought-after mechanosensitive cation channels in mammals. Piezo1 and Piezo2 exhibit a unique propeller-shaped architecture and have been implicated in mechanotransduction in various critical processes, including touch sensation, balance, and cardiovascular regulation. Furthermore, several mutations in Piezo channels have been shown to cause multiple hereditary human disorders, such as autosomal recessive congenital lymphatic dysplasia. Notably, mutations that cause dehydrated hereditary xerocytosis alter the rate of Piezo channel inactivation, indicating the critical role of their kinetics in normal physiology. Given the importance of Piezo channels in understanding the mechanotransduction process, this review focuses on their structural details, kinetic properties and potential function as mechanosensors. We also briefly review the hereditary diseases caused by mutations in Piezo genes, which is key for understanding the function of these proteins.
Collapse
Affiliation(s)
- Xiang-Zhi Fang
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ting Zhou
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ji-Qian Xu
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ya-Xin Wang
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Miao-Miao Sun
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ya-Jun He
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shang-Wen Pan
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Xiong
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhe-Kang Peng
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xue-Hui Gao
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - You Shang
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China. .,Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
42
|
Guo Y, Merten AL, Schöler U, Yu ZY, Cvetkovska J, Fatkin D, Feneley MP, Martinac B, Friedrich O. In vitro cell stretching technology (IsoStretcher) as an approach to unravel Piezo1-mediated cardiac mechanotransduction. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2021; 159:22-33. [PMID: 32763257 DOI: 10.1016/j.pbiomolbio.2020.07.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 07/09/2020] [Accepted: 07/13/2020] [Indexed: 12/12/2022]
Abstract
The transformation of electrical signals into mechanical action of the heart underlying blood circulation results in mechanical stimuli during active contraction or passive filling distention, which conversely modulate electrical signals. This feedback mechanism is known as cardiac mechano-electric coupling (MEC). The cardiac MEC involves complex activation of mechanical biosensors initiating short-term and long-term effects through Ca2+ signals in cardiomyocytes in acute and chronic pressure overload scenarios (e.g. cardiac hypertrophy). Although it is largely still unknown how mechanical forces alter cardiac function at the molecular level, mechanosensitive channels, including the recently discovered family of Piezo channels, have been thought to play a major role in the cardiac MEC and are also suspected to contribute to development of cardiac hypertrophy and heart failure. The earliest reports of mechanosensitive channel activity recognized that their gating could be controlled by membrane stretch. In this article, we provide an overview of the stretch devices, which have been employed for studies of the effects of mechanical stimuli on muscle and heart cells. We also describe novel experiments examining the activity of Piezo1 channels under multiaxial stretch applied using polydimethylsiloxane (PDMS) stretch chambers and IsoStretcher technology to achieve isotropic stretching stimulation to cultured HL-1 cardiac muscle cells which express an appreciable amount of Piezo1.
Collapse
Affiliation(s)
- Yang Guo
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia; Cardiac Physiology and Transplantation Division, Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia; Faculty of Medicine, University of New South Wales, Kensington, New South Wales, Australia
| | - Anna-Lena Merten
- Institute of Medical Biotechnology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Germany; School in Advanced Optical Technologies, Friedrich-Alexander-Universität Erlangen-Nürnberg, Germany; Muscle Research Center Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Ulrike Schöler
- Institute of Medical Biotechnology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Germany; School in Advanced Optical Technologies, Friedrich-Alexander-Universität Erlangen-Nürnberg, Germany; Muscle Research Center Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Ze-Yan Yu
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia; Cardiac Physiology and Transplantation Division, Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia; Faculty of Medicine, University of New South Wales, Kensington, New South Wales, Australia
| | - Jasmina Cvetkovska
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia
| | - Diane Fatkin
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia; Faculty of Medicine, University of New South Wales, Kensington, New South Wales, Australia; Department of Cardiology, St Vincent's Hospital, Darlinghurst, NSW, Australia
| | - Michael P Feneley
- Cardiac Physiology and Transplantation Division, Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia; Faculty of Medicine, University of New South Wales, Kensington, New South Wales, Australia; Department of Cardiology, St Vincent's Hospital, Darlinghurst, NSW, Australia
| | - Boris Martinac
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia; Faculty of Medicine, University of New South Wales, Kensington, New South Wales, Australia.
| | - Oliver Friedrich
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia; Institute of Medical Biotechnology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Germany; School in Advanced Optical Technologies, Friedrich-Alexander-Universität Erlangen-Nürnberg, Germany; Muscle Research Center Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
43
|
Richardson KJ, Kuck L, Simmonds MJ. Beyond oxygen transport: active role of erythrocytes in the regulation of blood flow. Am J Physiol Heart Circ Physiol 2020; 319:H866-H872. [PMID: 32857630 DOI: 10.1152/ajpheart.00441.2020] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
It was classically thought that the function of mammalian red blood cells (RBCs) was limited to serving as a vehicle for oxygen, given the cells' abundance of cytosolic hemoglobin. Over the past decades, however, accumulating evidence indicates that RBCs have the capacity to sense low-oxygen tensions in hypoxic tissues, and, subsequently, release signaling molecules that influence the distribution of blood flow. The precise mechanisms that facilitate RBC modulation of blood flow are still being elucidated, although recent evidence indicates involvement of 1) adenosine triphosphate, capable of binding to purinergic receptors located on the vascular wall before initiating nitric oxide (NO; a powerful vasodilator) production in endothelial cells, and/or 2) nonvascular NO, which is now known to have several modes of production within RBCs, including an enzymatic process via a unique isoform of NO synthase (i.e., RBC-NOS), which has potential effects on the vascular smooth muscle. The physical properties of RBCs, including their tendency to form three-dimensional structures in low shear flow (i.e., aggregation) and their capacity to elongate in high shear flow (i.e., deformability), are only recently being viewed as mechanotransductive processes, with profound effects on vascular reactivity and tissue perfusion. Recent developments in intracellular signaling in RBCs, and the subsequent effects on the mechanical properties of blood, and blood flow, thus present a vivid expansion on the classic perspective of these abundant cells.
Collapse
Affiliation(s)
- Kieran J Richardson
- Biorheology Research Laboratory, Griffith University, Gold Coast, Australia.,Menzies Health Institute Queensland, Griffith University, Gold Coast, Australia
| | - Lennart Kuck
- Biorheology Research Laboratory, Griffith University, Gold Coast, Australia.,Menzies Health Institute Queensland, Griffith University, Gold Coast, Australia
| | - Michael J Simmonds
- Biorheology Research Laboratory, Griffith University, Gold Coast, Australia.,Menzies Health Institute Queensland, Griffith University, Gold Coast, Australia
| |
Collapse
|
44
|
Goncharova EA, Chan SY, Ventetuolo CE, Weissmann N, Schermuly RT, Mullin CJ, Gladwin MT. Update in Pulmonary Vascular Diseases and Right Ventricular Dysfunction 2019. Am J Respir Crit Care Med 2020; 202:22-28. [PMID: 32311291 PMCID: PMC7328315 DOI: 10.1164/rccm.202003-0576up] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 04/20/2020] [Indexed: 12/15/2022] Open
Affiliation(s)
- Elena A. Goncharova
- Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute
- Division of Pulmonary, Allergy and Critical Care Medicine
| | - Stephen Y. Chan
- Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute
- Center for Pulmonary Vascular Biology and Medicine, and
- Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Medicine, Alpert Medical School, and
| | - Corey E. Ventetuolo
- Department of Medicine, Alpert Medical School, and
- Department of Health Services, Policy, and Practice, School of Public Health, Brown University, Providence, Rhode Island; and
| | - Norbert Weissmann
- Department of Internal Medicine, Justus-Liebig University Giessen, Giessen, Germany
| | - Ralph T. Schermuly
- Department of Internal Medicine, Justus-Liebig University Giessen, Giessen, Germany
| | | | - Mark T. Gladwin
- Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute
- Division of Pulmonary, Allergy and Critical Care Medicine
| |
Collapse
|
45
|
Xu S. Therapeutic potential of blood flow mimetic compounds in preventing endothelial dysfunction and atherosclerosis. Pharmacol Res 2020; 155:104737. [DOI: 10.1016/j.phrs.2020.104737] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 02/28/2020] [Accepted: 02/28/2020] [Indexed: 01/08/2023]
|
46
|
Affiliation(s)
- Rebecca L Heise
- Department of Biomedical EngineeringVirginia Commonwealth UniversityRichmond, Virginiaand
| | - Laszlo Farkas
- College of MedicineThe Ohio State UniversityColumbus, Ohio
| |
Collapse
|
47
|
It takes more than two to tango: mechanosignaling of the endothelial surface. Pflugers Arch 2020; 472:419-433. [PMID: 32239285 PMCID: PMC7165135 DOI: 10.1007/s00424-020-02369-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 03/13/2020] [Accepted: 03/18/2020] [Indexed: 02/07/2023]
Abstract
The endothelial surface is a highly flexible signaling hub which is able to sense the hemodynamic forces of the streaming blood. The subsequent mechanosignaling is basically mediated by specific structures, like the endothelial glycocalyx building the top surface layer of endothelial cells as well as mechanosensitive ion channels within the endothelial plasma membrane. The mechanical properties of the endothelial cell surface are characterized by the dynamics of cytoskeletal proteins and play a key role in the process of signal transmission from the outside (lumen of the blood vessel) to the interior of the cell. Thus, the cell mechanics directly interact with the function of mechanosensitive structures and ion channels. To precisely maintain the vascular tone, a coordinated functional interdependency between endothelial cells and vascular smooth muscle cells is necessary. This is given by the fact that mechanosensitive ion channels are expressed in both cell types and that signals are transmitted via autocrine/paracrine mechanisms from layer to layer. Thus, the outer layer of the endothelial cells can be seen as important functional mechanosensitive and reactive cellular compartment. This review aims to describe the known mechanosensitive structures of the vessel building a bridge between the important role of physiological mechanosignaling and the proper vascular function. Since mutations and dysfunction of mechanosensitive proteins are linked to vascular pathologies such as hypertension, they play a potent role in the field of channelopathies and mechanomedicine.
Collapse
|
48
|
Xiao B. Levering Mechanically Activated Piezo Channels for Potential Pharmacological Intervention. Annu Rev Pharmacol Toxicol 2020; 60:195-218. [DOI: 10.1146/annurev-pharmtox-010919-023703] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The mechanically activated Piezo channels, including Piezo1 and Piezo2 in mammals, function as key mechanotransducers for converting mechanical force into electrochemical signals. This review highlights key evidence for the potential of Piezo channel drug discovery. First, both mouse and human genetic studies have unequivocally demonstrated the prominent role of Piezo channels in various mammalian physiologies and pathophysiologies, validating their potential as novel therapeutic targets. Second, the cryo-electron microscopy structure of the 2,547-residue mouse Piezo1 trimer has been determined, providing a solid foundation for studying its structure-function relationship and drug action mechanisms and conducting virtual drug screening. Third, Piezo1 chemical activators, named Yoda1 and Jedi1/2, have been identified through high-throughput screening assays, demonstrating the drugability of Piezo channels. However, the pharmacology of Piezo channels is in its infancy. By establishing an integrated drug discovery platform, we may hopefully discover and develop a fleet of Jedi masters for battling Piezo-related human diseases.
Collapse
Affiliation(s)
- Bailong Xiao
- State Key Laboratory of Membrane Biology; Tsinghua-Peking Joint Center for Life Sciences; IDG/McGovern Institute for Brain Research; Beijing Advanced Innovation Center for Structural Biology; and School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
| |
Collapse
|
49
|
Douguet D, Patel A, Xu A, Vanhoutte PM, Honoré E. Piezo Ion Channels in Cardiovascular Mechanobiology. Trends Pharmacol Sci 2019; 40:956-970. [PMID: 31704174 DOI: 10.1016/j.tips.2019.10.002] [Citation(s) in RCA: 119] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 09/25/2019] [Accepted: 10/10/2019] [Indexed: 01/05/2023]
Abstract
Mechanotransduction has a key role in vascular development, physiology, and disease states. Piezo1 is a mechanosensitive (MS) nonselective cationic channel that occurs in endothelial and vascular smooth muscle cells. It is activated by shear stress associated with increases in local blood flow, as well as by cell membrane stretch upon elevation of blood pressure. Here, we briefly review the pharmacological modulators of Piezo and discuss current understanding of the role of Piezo1 in vascular mechanobiology and associated clinical disorders, such as atherosclerosis and hypertension. Ultimately, we believe that this research will help identify novel therapeutic strategies for the treatment of vascular diseases.
Collapse
Affiliation(s)
- Dominique Douguet
- Université Côte d'Azur, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Institut de Pharmacologie Moléculaire et Cellulaire, Labex ICST, Valbonne, France
| | - Amanda Patel
- Université Côte d'Azur, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Institut de Pharmacologie Moléculaire et Cellulaire, Labex ICST, Valbonne, France
| | - Aimin Xu
- State Key Laboratory of Biopharmaceutical Technologies, Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong, China
| | - Paul M Vanhoutte
- State Key Laboratory of Biopharmaceutical Technologies, Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong, China; Department of Cardiovascular and Renal Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Eric Honoré
- Université Côte d'Azur, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Institut de Pharmacologie Moléculaire et Cellulaire, Labex ICST, Valbonne, France.
| |
Collapse
|
50
|
Beech DJ, Kalli AC. Force Sensing by Piezo Channels in Cardiovascular Health and Disease. Arterioscler Thromb Vasc Biol 2019; 39:2228-2239. [PMID: 31533470 PMCID: PMC6818984 DOI: 10.1161/atvbaha.119.313348] [Citation(s) in RCA: 164] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 09/03/2019] [Indexed: 02/07/2023]
Abstract
Mechanical forces are fundamental in cardiovascular biology, and deciphering the mechanisms by which they act remains a testing frontier in cardiovascular research. Here, we raise awareness of 2 recently discovered proteins, Piezo1 and Piezo2, which assemble as transmembrane triskelions to combine exquisite force sensing with regulated calcium influx. There is emerging evidence for their importance in endothelial shear stress sensing and secretion, NO generation, vascular tone, angiogenesis, atherosclerosis, vascular permeability and remodeling, blood pressure regulation, insulin sensitivity, exercise performance, and baroreceptor reflex, and there are early suggestions of relevance to cardiac fibroblasts and myocytes. Human genetic analysis points to significance in lymphatic disease, anemia, varicose veins, and potentially heart failure, hypertension, aneurysms, and stroke. These channels appear to be versatile force sensors, used creatively to inform various force-sensing situations. We discuss emergent concepts and controversies and suggest that the potential for new important understanding is substantial.
Collapse
Affiliation(s)
- David J. Beech
- From the Department of Discovery and Translational Science, Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, England, United Kingdom
| | - Antreas C. Kalli
- From the Department of Discovery and Translational Science, Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, England, United Kingdom
| |
Collapse
|