1
|
Adamopoulou E, Dimitriadis K, Kyriakoulis K, Pyrpyris N, Beneki E, Fragkoulis C, Konstantinidis D, Aznaouridis K, Tsioufis K. Defining "Vulnerable" in coronary artery disease: predisposing factors and preventive measures. Cardiovasc Pathol 2025; 77:107736. [PMID: 40228760 DOI: 10.1016/j.carpath.2025.107736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 03/16/2025] [Accepted: 04/10/2025] [Indexed: 04/16/2025] Open
Abstract
The likelihood of a plaque to cause an acute coronary syndrome (ACS) depends on several factors, both lesion- and patient-related. One of the most investigated and established contributing factors is the presence of high-risk or "vulnerable plaque" characteristics, which have been correlated with increased incidence of major adverse cardiovascular events (MACE). The recognition, however, that a significant percentage of vulnerable plaques do not result in causing clinical events has led the scientific community towards the more multifaceted concept of "vulnerable patients". Incorporating the morphological features of an atherosclerotic plaque into its hemodynamic surroundings can better predict the chance of its disruption, as altered fluid dynamics play a significant role in plaque destabilization. The advances in coronary imaging and the field of computational fluid dynamics (CFD) can contribute to develop more accurate lesion- and patient-related ACS prediction models that take into account both the morphology of a plaque and the forces applied upon it. The aim of this review is to provide the latest data regarding the aforementioned predictive factors as well as relevant preventive measures.
Collapse
Affiliation(s)
- Eleni Adamopoulou
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Hippokration General Hospital, 115 27, Athens, Greece
| | - Kyriakos Dimitriadis
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Hippokration General Hospital, 115 27, Athens, Greece.
| | - Konstantinos Kyriakoulis
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Hippokration General Hospital, 115 27, Athens, Greece
| | - Nikolaos Pyrpyris
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Hippokration General Hospital, 115 27, Athens, Greece
| | - Eirini Beneki
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Hippokration General Hospital, 115 27, Athens, Greece
| | - Christos Fragkoulis
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Hippokration General Hospital, 115 27, Athens, Greece
| | - Dimitris Konstantinidis
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Hippokration General Hospital, 115 27, Athens, Greece
| | - Konstantinos Aznaouridis
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Hippokration General Hospital, 115 27, Athens, Greece
| | - Konstantinos Tsioufis
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Hippokration General Hospital, 115 27, Athens, Greece
| |
Collapse
|
2
|
Hattori R, Kajiki M, Fujimoto T, Amano T, Kenny GP, Watanabe K, Nishiyasu T, Fujii N. TRPA1 channels modulate cutaneous vasodilation during exercise in the heat in young adults when NOS is inhibited. Am J Physiol Regul Integr Comp Physiol 2025; 328:R319-R328. [PMID: 39925087 DOI: 10.1152/ajpregu.00269.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 01/06/2025] [Accepted: 02/02/2025] [Indexed: 02/11/2025]
Abstract
Nitric oxide synthase (NOS) is an important mediator of cutaneous vasodilation during exercise-heat stress. We recently reported that pharmacological activation of transient receptor potential ankyrin 1 (TRPA1) channel mediates cutaneous vasodilation via NOS-dependent mechanisms under nonheat stress-resting conditions. Here, we hypothesized that TRPA1 channel activation would contribute to cutaneous vasodilation during exercise in the heat via NOS-dependent mechanisms. To assess this response, we first conducted TRPA1 channel antagonist verification substudy (10 young adults and 5 women) wherein 1 mM ASP7663 (TRPA1 channel agonist) increased cutaneous vascular conductance (CVC; cutaneous blood flow divided by mean arterial pressure) and this response was blocked by ∼50% with 100 μM HC030031, a known TRPA1 channel antagonist. Subsequently, 12 young adults (5 women) completed two bouts of 30-min moderate-intensity cycling (45% of their predetermined peak oxygen uptake) in the heat (35°C). During the first exercise, CVC was evaluated at four dorsal forearm skin sites perfused with a 5% DMSO, whereas in the second bout, all sites were treated with either 1) a 5% DMSO (control), 2) 100 µM HC030031, 3) 20 mM l-NAME, a nonselective NOS inhibitor, or 4) combination of both. No between-site differences in CVC were measured during the first exercise (P > 0.182). During the second exercise, HC030031 alone had no effect on CVC relative to the control (all P > 0.104). Both l-NAME and HC030031 + l-NAME reduced CVC (all P < 0.001), with the combined treatment showing a greater reduction (all P < 0.001). We showed that TRPA1 channels mediate cutaneous vasodilation during exercise-heat stress only when NOS is inhibited.NEW & NOTEWORTHY We demonstrated that the administration of TRPA1 channel antagonist HC030031 only appears to attenuate cutaneous vasodilation during exercise in the heat when nitric oxide synthase (NOS) is inhibited. TRPA1 channels may function as a "backup system" to maintain cutaneous vasodilation when NOS-dependent vasodilation is compromised during exercise in the heat.
Collapse
Affiliation(s)
- Rei Hattori
- Institute of Health and Sport Sciences, University of Tsukuba, Ibaraki, Japan
| | - Masanobu Kajiki
- Institute of Health and Sport Sciences, University of Tsukuba, Ibaraki, Japan
| | - Tomomi Fujimoto
- Department of Health and Sports, Niigata University of Health and Welfare, Niigata, Japan
| | - Tatsuro Amano
- Laboratory for Exercise and Environmental Physiology, Faculty of Education, Niigata University, Niigata, Japan
| | - Glen P Kenny
- Human and Environmental Physiology Research Unit, University of Ottawa, Ottawa, Ontario, Canada
| | - Koichi Watanabe
- Institute of Health and Sport Sciences, University of Tsukuba, Ibaraki, Japan
| | - Takeshi Nishiyasu
- Institute of Health and Sport Sciences, University of Tsukuba, Ibaraki, Japan
- Advanced Research Initiative for Human High Performance (ARIHHP), University of Tsukuba, Ibaraki, Japan
| | - Naoto Fujii
- Institute of Health and Sport Sciences, University of Tsukuba, Ibaraki, Japan
- Advanced Research Initiative for Human High Performance (ARIHHP), University of Tsukuba, Ibaraki, Japan
| |
Collapse
|
3
|
Nakamura K, Akagi S, Ejiri K, Taya S, Saito Y, Kuroda K, Takaya Y, Toh N, Nakayama R, Katanosaka Y, Yuasa S. Pathophysiology of Group 3 Pulmonary Hypertension Associated with Lung Diseases and/or Hypoxia. Int J Mol Sci 2025; 26:835. [PMID: 39859549 PMCID: PMC11765551 DOI: 10.3390/ijms26020835] [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: 11/18/2024] [Revised: 01/07/2025] [Accepted: 01/14/2025] [Indexed: 01/27/2025] Open
Abstract
Pulmonary hypertension associated with lung diseases and/or hypoxia is classified as group 3 in the clinical classification of pulmonary hypertension. The efficacy of existing selective pulmonary vasodilators for group 3 pulmonary hypertension is still unknown, and it is currently associated with a poor prognosis. The mechanisms by which pulmonary hypertension occurs include hypoxic pulmonary vasoconstriction, pulmonary vascular remodeling, a decrease in pulmonary vascular beds, endothelial dysfunction, endothelial-to-mesenchymal transition, mitochondrial dysfunction, oxidative stress, hypoxia-inducible factors (HIFs), inflammation, microRNA, and genetic predisposition. Among these, hypoxic pulmonary vasoconstriction and subsequent pulmonary vascular remodeling are characteristic factors involving the pulmonary vasculature and are the focus of this review. Several factors have been reported to mediate vascular remodeling induced by hypoxic pulmonary vasoconstriction, such as HIF-1α and mechanosensors, including TRP channels. New therapies that target novel molecules, such as mechanoreceptors, to inhibit vascular remodeling are awaited.
Collapse
Affiliation(s)
- Kazufumi Nakamura
- Department of Cardiovascular Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (S.A.); (K.E.); (S.T.); (Y.S.); (K.K.); (Y.T.); (N.T.); (R.N.); (S.Y.)
- Center for Advanced Heart Failure, Okayama University Hospital, Okayama 700-8558, Japan
| | - Satoshi Akagi
- Department of Cardiovascular Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (S.A.); (K.E.); (S.T.); (Y.S.); (K.K.); (Y.T.); (N.T.); (R.N.); (S.Y.)
| | - Kentaro Ejiri
- Department of Cardiovascular Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (S.A.); (K.E.); (S.T.); (Y.S.); (K.K.); (Y.T.); (N.T.); (R.N.); (S.Y.)
| | - Satoshi Taya
- Department of Cardiovascular Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (S.A.); (K.E.); (S.T.); (Y.S.); (K.K.); (Y.T.); (N.T.); (R.N.); (S.Y.)
| | - Yukihiro Saito
- Department of Cardiovascular Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (S.A.); (K.E.); (S.T.); (Y.S.); (K.K.); (Y.T.); (N.T.); (R.N.); (S.Y.)
| | - Kazuhiro Kuroda
- Department of Cardiovascular Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (S.A.); (K.E.); (S.T.); (Y.S.); (K.K.); (Y.T.); (N.T.); (R.N.); (S.Y.)
| | - Yoichi Takaya
- Department of Cardiovascular Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (S.A.); (K.E.); (S.T.); (Y.S.); (K.K.); (Y.T.); (N.T.); (R.N.); (S.Y.)
| | - Norihisa Toh
- Department of Cardiovascular Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (S.A.); (K.E.); (S.T.); (Y.S.); (K.K.); (Y.T.); (N.T.); (R.N.); (S.Y.)
| | - Rie Nakayama
- Department of Cardiovascular Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (S.A.); (K.E.); (S.T.); (Y.S.); (K.K.); (Y.T.); (N.T.); (R.N.); (S.Y.)
| | - Yuki Katanosaka
- Department of Pharmacy, Kinjo Gakuin University, Nagoya 463-8521, Japan;
- Graduate School of Pharmaceutical Sciences, Kinjo Gakuin University, Nagoya 463-8521, Japan
| | - Shinsuke Yuasa
- Department of Cardiovascular Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (S.A.); (K.E.); (S.T.); (Y.S.); (K.K.); (Y.T.); (N.T.); (R.N.); (S.Y.)
| |
Collapse
|
4
|
Hansen CE, Hollaus D, Kamermans A, de Vries HE. Tension at the gate: sensing mechanical forces at the blood-brain barrier in health and disease. J Neuroinflammation 2024; 21:325. [PMID: 39696463 PMCID: PMC11657007 DOI: 10.1186/s12974-024-03321-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 12/07/2024] [Indexed: 12/20/2024] Open
Abstract
Microvascular brain endothelial cells tightly limit the entry of blood components and peripheral cells into the brain by forming the blood-brain barrier (BBB). The BBB is regulated by a cascade of mechanical and chemical signals including shear stress and elasticity of the adjacent endothelial basement membrane (BM). During physiological aging, but especially in neurological diseases including multiple sclerosis (MS), stroke, small vessel disease, and Alzheimer's disease (AD), the BBB is exposed to inflammation, rigidity changes of the BM, and disturbed cerebral blood flow (CBF). These altered forces lead to increased vascular permeability, reduced endothelial reactivity to vasoactive mediators, and promote leukocyte transmigration. Whereas the molecular players involved in leukocyte infiltration have been described in detail, the importance of mechanical signalling throughout this process has only recently been recognized. Here, we review relevant features of mechanical forces acting on the BBB under healthy and pathological conditions, as well as the endothelial mechanosensory elements detecting and responding to altered forces. We demonstrate the underlying complexity by focussing on the family of transient receptor potential (TRP) ion channels. A better understanding of these processes will provide insights into the pathogenesis of several neurological disorders and new potential leads for treatment.
Collapse
Affiliation(s)
- Cathrin E Hansen
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands
- MS Center Amsterdam, Amsterdam UMC Location VU Medical Center, Amsterdam, The Netherlands
| | - David Hollaus
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
| | - Alwin Kamermans
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands
| | - Helga E de Vries
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands.
- Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands.
- MS Center Amsterdam, Amsterdam UMC Location VU Medical Center, Amsterdam, The Netherlands.
| |
Collapse
|
5
|
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
|
6
|
Sherman J, Bortz E, Antonio ES, Tseng HA, Raiff L, Han X. Ultrasound pulse repetition frequency preferentially activates different neuron populations independent of cell type. J Neural Eng 2024; 21:056008. [PMID: 39178904 PMCID: PMC11381926 DOI: 10.1088/1741-2552/ad731c] [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: 03/25/2024] [Revised: 06/18/2024] [Accepted: 08/23/2024] [Indexed: 08/26/2024]
Abstract
Objective. Transcranial ultrasound (US) stimulation serves as an external input to a neuron, and thus the evoked response relies on neurons' intrinsic properties. Neural activity is limited to a couple hundred hertz and often exhibits preference to input frequencies. Accordingly, US pulsed at specific physiologic pulse repetition frequencies (PRFs) may selectively engage neurons with the corresponding input frequency preference. However, most US parametric studies examine the effects of supraphysiologic PRFs. It remains unclear whether pulsing US at different physiologic PRFs could activate distinct neurons in the awake mammalian brain.Approach. We recorded cellular calcium responses of individual motor cortex neurons to US pulsed at PRFs of 10, 40, and 140 Hz in awake mice. We compared the evoked responses across these PRFs in the same neurons. To further understand the cell-type dependent effects, we categorized the recorded neurons as parvalbumin positive fast spiking interneurons or putative excitatory neurons and analyzed single-cell mechanosensitive channel expression in mice and humans using the Allen Brain Institute's RNA-sequencing databases.Main results. We discovered that many neurons were preferentially activated by only one PRF and different PRFs selectively engaged distinct neuronal populations. US-evoked cellular calcium responses exhibited the same characteristics as those naturally occurring during spiking, suggesting that US increases intrinsic neuronal activity. Furthermore, evoked responses were similar between fast-spiking inhibitory neurons and putative excitatory neurons. Thus, variation in individual neuron's cellular properties dominates US-evoked response heterogeneity, consistent with our observed cell-type independent expression patterns of mechanosensitive channels across individual neurons in mice and humans. Finally, US transiently increased network synchrony without producing prolonged over-synchronization that could be detrimental to neural circuit functions.Significance. These results highlight the feasibility of activating distinct neuronal subgroups by varying PRF and the potential to improve neuromodulation effects by combining physiologic PRFs.
Collapse
Affiliation(s)
- Jack Sherman
- Department of Biomedical Engineering, Boston University, Boston, MA, United States of America
- Department of Pharmacology and Experimental Therapeutics, Boston University, Boston, MA, United States of America
| | - Emma Bortz
- Department of Biomedical Engineering, Boston University, Boston, MA, United States of America
| | - Erynne San Antonio
- Department of Biomedical Engineering, Boston University, Boston, MA, United States of America
| | - Hua-an Tseng
- Department of Biomedical Engineering, Boston University, Boston, MA, United States of America
| | - Laura Raiff
- Department of Biomedical Engineering, Boston University, Boston, MA, United States of America
| | - Xue Han
- Department of Biomedical Engineering, Boston University, Boston, MA, United States of America
| |
Collapse
|
7
|
Li B, Zhao A, Tian T, Yang X. Mechanobiological insight into brain diseases based on mechanosensitive channels: Common mechanisms and clinical potential. CNS Neurosci Ther 2024; 30:e14809. [PMID: 38923822 PMCID: PMC11197048 DOI: 10.1111/cns.14809] [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: 02/28/2024] [Revised: 05/15/2024] [Accepted: 06/02/2024] [Indexed: 06/28/2024] Open
Abstract
BACKGROUND As physical signals, mechanical cues regulate the neural cells in the brain. The mechanosensitive channels (MSCs) perceive the mechanical cues and transduce them by permeating specific ions or molecules across the plasma membrane, and finally trigger a series of intracellular bioelectrical and biochemical signals. Emerging evidence supports that wide-distributed, high-expressed MSCs like Piezo1 play important roles in several neurophysiological processes and neurological disorders. AIMS To systematically conclude the functions of MSCs in the brain and provide a novel mechanobiological perspective for brain diseases. METHOD We summarized the mechanical cues and MSCs detected in the brain and the research progress on the functional roles of MSCs in physiological conditions. We then concluded the pathological activation and downstream pathways triggered by MSCs in two categories of brain diseases, neurodegenerative diseases and place-occupying damages. Finally, we outlined the methods for manipulating MSCs and discussed their medical potential with some crucial outstanding issues. RESULTS The MSCs present underlying common mechanisms in different brain diseases by acting as the "transportation hubs" to transduce the distinct signal patterns: the upstream mechanical cues and the downstream intracellular pathways. Manipulating the MSCs is feasible to alter the complicated downstream processes, providing them promising targets for clinical treatment. CONCLUSIONS Recent research on MSCs provides a novel insight into brain diseases. The common mechanisms mediated by MSCs inspire a wide range of therapeutic potentials targeted on MSCs in different brain diseases.
Collapse
Affiliation(s)
- Bolong Li
- Shenzhen Key Laboratory of Translational Research for Brain Diseases, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced TechnologyChinese Academy of SciencesShenzhenGuangdongChina
- College of Life SciencesUniversity of Chinese Academy of ScienceBeijingChina
| | - An‐ran Zhao
- Shenzhen Key Laboratory of Translational Research for Brain Diseases, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced TechnologyChinese Academy of SciencesShenzhenGuangdongChina
- College of Life SciencesUniversity of Chinese Academy of ScienceBeijingChina
- Faculty of Life and Health SciencesShenzhen University of Advanced TechnologyShenzhenGuangdongChina
| | - Tian Tian
- Shenzhen Key Laboratory of Translational Research for Brain Diseases, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced TechnologyChinese Academy of SciencesShenzhenGuangdongChina
- Faculty of Life and Health SciencesShenzhen University of Advanced TechnologyShenzhenGuangdongChina
| | - Xin Yang
- Shenzhen Key Laboratory of Translational Research for Brain Diseases, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced TechnologyChinese Academy of SciencesShenzhenGuangdongChina
- Faculty of Life and Health SciencesShenzhen University of Advanced TechnologyShenzhenGuangdongChina
| |
Collapse
|
8
|
Xie L, Huang B, Zhao X, Zhu N. Exploring the mechanisms underlying effects of bisphenol a on cardiovascular disease by network toxicology and molecular docking. Heliyon 2024; 10:e31473. [PMID: 38813174 PMCID: PMC11133888 DOI: 10.1016/j.heliyon.2024.e31473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 05/15/2024] [Accepted: 05/16/2024] [Indexed: 05/31/2024] Open
Abstract
Background Globally, cardiovascular disease (CVD) has emerged as a leading cause of mortality. Bisphenol A (BPA), recognized as one of the most prevalent and widely distributed endocrine-disrupting chemicals (EDCs), has been consistently linked to the progression of CVD. This research centers on unraveling the molecular mechanisms responsible for the toxic effects of BPA exposure on CVD. Key targets and pathways involved in action of BPA on CVD were investigated by network toxicology. Binding abilities of BPA to core targets were evaluated by molecular docking. Methods and results Based on information retrieved from ChEMBL, DrugBank, and OMIM databases, a total of 27 potential targets were found to be associated with the influence of BPA on CVD. Furthermore, the STRING and Cytoscape software were employed to identify three central genes-ESR1, PPARG, and PTGS2-and to construct both the protein-protein interaction network and an interaction diagram of potential targets. Gene ontology (GO) and KEGG (Kyoto Encyclopedia of Genes and Genomes, KEGG) pathway enrichment analyses via WebGestalt revealed key biological processes (BP), cellular components (CC), molecular functions (MF), and pathways, such as the calcium signaling pathway, inflammatory mediator regulation of TRP channels, gap junction, adrenergic signaling in cardiomyocytes, cGMP-PKG signaling pathway, and cAMP signaling pathway, predominantly involved in BPA-induced CVD toxicity. By using molecular docking investigations, it proved that BPA binds to ESR1, PPARG, and PTGS2 steadily and strongly. Conclusion This study not only establishes a theoretical framework for understanding the molecular toxicity mechanism of BPA in cardiovascular disease (CVD) but also introduces an innovative network toxicology approach to methodically investigate the influence of environmental contaminants on CVD. This methodology sets the stage for drug discovery efforts targeting CVD linked to exposure to endocrine-disrupting chemicals (EDCs).
Collapse
Affiliation(s)
- Lina Xie
- Department of Neurosurgery, The Wenzhou Third Clinical Institute Affiliated to Wenzhou Medical University, The Third Affiliated Hospital of Shanghai University, Wenzhou People's Hospital, China
| | - Bingwu Huang
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, China
| | - Xuyong Zhao
- Department of Cardiology, The Wenzhou Third Clinical Institute Affiliated to Wenzhou Medical University, The Third Affiliated Hospital of Shanghai University, Wenzhou People's Hospital, China
| | - Ning Zhu
- Department of Cardiology, The Wenzhou Third Clinical Institute Affiliated to Wenzhou Medical University, The Third Affiliated Hospital of Shanghai University, Wenzhou People's Hospital, China
| |
Collapse
|
9
|
Sherman J, Bortz E, Antonio ES, Tseng HA, Raiff L, Han X. Ultrasound pulse repetition frequency preferentially activates different neuron populations independent of cell type. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.25.586645. [PMID: 38585918 PMCID: PMC10996595 DOI: 10.1101/2024.03.25.586645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Transcranial ultrasound activates mechanosensitive cellular signaling and modulates neural dynamics. Given that intrinsic neuronal activity is limited to a couple hundred hertz and often exhibits frequency preference, we examined whether pulsing ultrasound at physiologic pulse repetition frequencies (PRFs) could selectively influence neuronal activity in the mammalian brain. We performed calcium imaging of individual motor cortex neurons, while delivering 0.35 MHz ultrasound at PRFs of 10, 40, and 140 Hz in awake mice. We found that most neurons were preferentially activated by only one of the three PRFs, highlighting unique cellular effects of physiologic PRFs. Further, ultrasound evoked responses were similar between excitatory neurons and parvalbumin positive interneurons regardless of PRFs, indicating that individual cell sensitivity dominates ultrasound-evoked effects, consistent with the heterogeneous mechanosensitive channel expression we found across single neurons in mice and humans. These results highlight the feasibility of tuning ultrasound neuromodulation effects through varying PRFs.
Collapse
|
10
|
Li MR, Luo XJ, Peng J. Role of sonic hedgehog signaling pathway in the regulation of ion channels: focus on its association with cardio-cerebrovascular diseases. J Physiol Biochem 2023; 79:719-730. [PMID: 37676576 DOI: 10.1007/s13105-023-00982-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 08/25/2023] [Indexed: 09/08/2023]
Abstract
Sonic hedgehog (SHH) signaling is vital for cell differentiation and proliferation during embryonic development, yet its role in cardiac, cerebral, and vascular pathophysiology is under debate. Recent studies have demonstrated that several compounds of SHH signaling regulate ion channels, which in turn affect the behavior of target cells. Some of these ion channels are involved in the cardio-cerebrovascular system. Here, we first reviewed the SHH signaling cascades, then its interaction with ion channels, and their impact on cardio-cerebrovascular diseases. Considering the complex cross talk of SHH signaling with other pathways that also affect ion channels and their potential impact on the cardio-cerebrovascular system, we highlight the necessity of thoroughly studying the effect of SHH signaling on ion homeostasis, which could serve as a novel mechanism for cardio-cerebrovascular diseases. Activation of SHH signaling influence ion channels activity, which in turn influence ion homeostasis, membrane potential, and electrophysiology, could serve as a novel strategy for cardio-cerebrovascular diseases.
Collapse
Affiliation(s)
- Ming-Rui Li
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| | - Xiu-Ju Luo
- Department of Laboratory Medicine, The Third Xiangya Hospital of Central South University, Changsha, 410013, China.
| | - Jun Peng
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China.
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China.
| |
Collapse
|
11
|
Rahaman SG, Mahanty M, Mukherjee P, Dutta B, Rahaman SO. Mechanosensing and Mechanosignal Transduction in Atherosclerosis. Curr Atheroscler Rep 2023; 25:711-721. [PMID: 37615786 DOI: 10.1007/s11883-023-01139-6] [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] [Accepted: 08/16/2023] [Indexed: 08/25/2023]
Abstract
PURPOSE OF REVIEW This review aims to summarize the latest findings on mechanosensing in atherosclerosis, elucidating the molecular mechanisms, cellular players, and potential therapeutic targets. RECENT FINDINGS Atherosclerosis, a chronic inflammatory disease characterized by the buildup of lipid-laden plaque within arterial walls, is a major contributor to cardiovascular disease-related mortality and morbidity. Interestingly, atherosclerosis predominantly occurs in arterial areas with curves and branches. In these regions, endothelial cells encounter irregular blood flow with distinctive low-intensity fluctuating shear stress. On the other hand, straight sections of arteries, subjected to a consistent flow and related high-intensity, one-way shear stress, are relatively safeguarded against atherosclerosis due to shear-dependent, disease-preventing endothelial cell reactions. In recent years, researchers have been investigating the role of mechanosensing in the development and progression of atherosclerosis. At the core of mechanosensing is the ability of various cells to sense and respond to biomechanical forces in their environment. In the context of atherosclerosis, endothelial cells, smooth muscle cells, and immune cells are subjected to various mechanical or physical stimuli, including shear stress, cyclic strain, and matrix stiffness. These mechanical cues play a crucial role in regulating cellular behavior and contribute to the pathophysiology of atherosclerosis. Accumulating evidence suggests that various mechanical or physical cues play a critical role in the development and promotion of atherosclerosis.
Collapse
Affiliation(s)
- Suneha G Rahaman
- University of Maryland, Department of Nutrition and Food Science, College Park, MD, 20742, USA
| | - Manisha Mahanty
- University of Maryland, Department of Nutrition and Food Science, College Park, MD, 20742, USA
| | - Pritha Mukherjee
- University of Maryland, Department of Nutrition and Food Science, College Park, MD, 20742, USA
| | - Bidisha Dutta
- University of Maryland, Department of Nutrition and Food Science, College Park, MD, 20742, USA
| | - Shaik O Rahaman
- University of Maryland, Department of Nutrition and Food Science, College Park, MD, 20742, USA.
| |
Collapse
|
12
|
Xu S, Wang F, Mai P, Peng Y, Shu X, Nie R, Zhang H. Mechanism Analysis of Vascular Calcification Based on Fluid Dynamics. Diagnostics (Basel) 2023; 13:2632. [PMID: 37627891 PMCID: PMC10453151 DOI: 10.3390/diagnostics13162632] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 08/05/2023] [Accepted: 08/07/2023] [Indexed: 08/27/2023] Open
Abstract
Vascular calcification is the abnormal deposition of calcium phosphate complexes in blood vessels, which is regarded as the pathological basis of multiple cardiovascular diseases. The flowing blood exerts a frictional force called shear stress on the vascular wall. Blood vessels have different hydrodynamic properties due to discrepancies in geometric and mechanical properties. The disturbance of the blood flow in the bending area and the branch point of the arterial tree produces a shear stress lower than the physiological magnitude of the laminar shear stress, which can induce the occurrence of vascular calcification. Endothelial cells sense the fluid dynamics of blood and transmit electrical and chemical signals to the full-thickness of blood vessels. Through crosstalk with endothelial cells, smooth muscle cells trigger osteogenic transformation, involved in mediating vascular intima and media calcification. In addition, based on the detection of fluid dynamics parameters, emerging imaging technologies such as 4D Flow MRI and computational fluid dynamics have greatly improved the early diagnosis ability of cardiovascular diseases, showing extremely high clinical application prospects.
Collapse
Affiliation(s)
- Shuwan Xu
- Department of Cardiology, The Eighth Affiliated Hospital of Sun Yat-Sen University, Shenzhen 518033, China; (S.X.); (F.W.); (P.M.)
| | - Feng Wang
- Department of Cardiology, The Eighth Affiliated Hospital of Sun Yat-Sen University, Shenzhen 518033, China; (S.X.); (F.W.); (P.M.)
| | - Peibiao Mai
- Department of Cardiology, The Eighth Affiliated Hospital of Sun Yat-Sen University, Shenzhen 518033, China; (S.X.); (F.W.); (P.M.)
| | - Yanren Peng
- Department of Cardiology, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, Guangzhou 510120, China; (Y.P.); (X.S.)
| | - Xiaorong Shu
- Department of Cardiology, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, Guangzhou 510120, China; (Y.P.); (X.S.)
| | - Ruqiong Nie
- Department of Cardiology, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, Guangzhou 510120, China; (Y.P.); (X.S.)
| | - Huanji Zhang
- Department of Cardiology, The Eighth Affiliated Hospital of Sun Yat-Sen University, Shenzhen 518033, China; (S.X.); (F.W.); (P.M.)
| |
Collapse
|
13
|
Herrera-Pérez S, Lamas JA. TREK channels in Mechanotransduction: a Focus on the Cardiovascular System. Front Cardiovasc Med 2023; 10:1180242. [PMID: 37288256 PMCID: PMC10242076 DOI: 10.3389/fcvm.2023.1180242] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 04/26/2023] [Indexed: 06/09/2023] Open
Abstract
Mechano-electric feedback is one of the most important subsystems operating in the cardiovascular system, but the underlying molecular mechanism remains rather unknown. Several proteins have been proposed to explain the molecular mechanism of mechano-transduction. Transient receptor potential (TRP) and Piezo channels appear to be the most important candidates to constitute the molecular mechanism behind of the inward current in response to a mechanical stimulus. However, the inhibitory/regulatory processes involving potassium channels that operate on the cardiac system are less well known. TWIK-Related potassium (TREK) channels have emerged as strong candidates due to their capacity for the regulation of the flow of potassium in response to mechanical stimuli. Current data strongly suggest that TREK channels play a role as mechano-transducers in different components of the cardiovascular system, not only at central (heart) but also at peripheral (vascular) level. In this context, this review summarizes and highlights the main existing evidence connecting this important subfamily of potassium channels with the cardiac mechano-transduction process, discussing molecular and biophysical aspects of such a connection.
Collapse
Affiliation(s)
- Salvador Herrera-Pérez
- Laboratory of Neuroscience, CINBIO, University of Vigo, Vigo, Spain
- Laboratory of Neuroscience, Galicia Sur Health Research Institute (IISGS), Vigo, Spain
| | - José Antonio Lamas
- Laboratory of Neuroscience, CINBIO, University of Vigo, Vigo, Spain
- Laboratory of Neuroscience, Galicia Sur Health Research Institute (IISGS), Vigo, Spain
| |
Collapse
|
14
|
Xiao T, Sun M, Zhao C, Kang J. TRPV1: A promising therapeutic target for skin aging and inflammatory skin diseases. Front Pharmacol 2023; 14:1037925. [PMID: 36874007 PMCID: PMC9975512 DOI: 10.3389/fphar.2023.1037925] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 01/20/2023] [Indexed: 02/17/2023] Open
Abstract
TRPV1 is a non-selective channel receptor widely expressed in skin tissues, including keratinocytes, peripheral sensory nerve fibers and immune cells. It is activated by a variety of exogenous or endogenous inflammatory mediators, triggering neuropeptide release and neurogenic inflammatory response. Previous studies have shown that TRPV1 is closely related to the occurrence and/or development of skin aging and various chronic inflammatory skin diseases, such as psoriasis, atopic dermatitis, rosacea, herpes zoster, allergic contact dermatitis and prurigo nodularis. This review summarizes the structure of the TRPV1 channel and discusses the expression of TRPV1 in the skin as well as its role of TRPV1 in skin aging and inflammatory skin diseases.
Collapse
Affiliation(s)
- Tengfei Xiao
- Department of Clinical Laboratory, The Sixth Affiliated Hospital of Nantong University, Yancheng Third People's Hospital, Yancheng, Jiangsu, China
| | - Mingzhong Sun
- Department of Clinical Laboratory, The Sixth Affiliated Hospital of Nantong University, Yancheng Third People's Hospital, Yancheng, Jiangsu, China
| | - Chuanxiang Zhao
- Institute of Medical Genetics and Reproductive Immunity, School of Medical Science and Laboratory Medicine, Jiangsu College of Nursing, Huai'an, Jiangsu, China
| | - Jingjing Kang
- Department of Clinical Laboratory, Affiliated Hospital of Nanjing University Medical School, Yancheng First People's Hospital, Yancheng, Jiangsu, China
| |
Collapse
|
15
|
Gwanyanya A, Mubagwa K. Emerging role of transient receptor potential (TRP) ion channels in cardiac fibroblast pathophysiology. Front Physiol 2022; 13:968393. [PMID: 36277180 PMCID: PMC9583832 DOI: 10.3389/fphys.2022.968393] [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: 06/13/2022] [Accepted: 09/20/2022] [Indexed: 11/13/2022] Open
Abstract
Cardiac fibroblasts make up a major proportion of non-excitable cells in the heart and contribute to the cardiac structural integrity and maintenance of the extracellular matrix. During myocardial injury, fibroblasts can be activated to trans-differentiate into myofibroblasts, which secrete extracellular matrix components as part of healing, but may also induce cardiac fibrosis and pathological cardiac structural and electrical remodeling. The mechanisms regulating such cellular processes still require clarification, but the identification of transient receptor potential (TRP) channels in cardiac fibroblasts could provide further insights into the fibroblast-related pathophysiology. TRP proteins belong to a diverse superfamily, with subgroups such as the canonical (TRPC), vanilloid (TRPV), melastatin (TRPM), ankyrin (TRPA), polycystin (TRPP), and mucolipin (TRPML). Several TRP proteins form non-selective channels that are permeable to cations like Na+ and Ca2+ and are activated by various chemical and physical stimuli. This review highlights the role of TRP channels in cardiac fibroblasts and the possible underlying signaling mechanisms. Changes in the expression or activity of TRPs such as TRPCs, TRPVs, TRPMs, and TRPA channels modulate cardiac fibroblasts and myofibroblasts, especially under pathological conditions. Such TRPs contribute to cardiac fibroblast proliferation and differentiation as well as to disease conditions such as cardiac fibrosis, atrial fibrillation, and fibroblast metal toxicity. Thus, TRP channels in fibroblasts represent potential drug targets in cardiac disease.
Collapse
Affiliation(s)
- Asfree Gwanyanya
- Department of Human Biology, University of Cape Town, Cape Town, South Africa
- *Correspondence: Asfree Gwanyanya,
| | - Kanigula Mubagwa
- Department of Cardiovascular Sciences, K U Leuven, Leuven, Belgium
- Department of Basic Sciences, Faculty of Medicine, Université Catholique de Bukavu, Bukavu, Democratic Republic of Congo
| |
Collapse
|
16
|
Li S, Xiao L, Sun Y, Hu S, Hu D. A TRPV1 common missense variant affected the prognosis of ischemic cardiomyopathy. Medicine (Baltimore) 2022; 101:e29892. [PMID: 35905222 PMCID: PMC9333512 DOI: 10.1097/md.0000000000029892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
The purpose was to identify the Transient receptor potential (TRP) superfamily gene variants associate with the prognosis of ischemic cardiomyopathy (ICM). A whole-exome sequencing study involving 252 ICM and 252 healthy controls participants enrolled from March 2003 to November 2017. Optimal sequence kernel association test and Cox regression dominant was conducted to identify the cause genes of TRP with ICM and association of common SNPs with prognosis of ICM. Rs224534 was verified in the replication population. Besides, the expression of TRPV1 was detectable in human failed heart ventricular tissues. The TRPs was not associated with the risk of ICM (P > .05). Rs224534 was significantly associated with the prognosis of ICM (Hazard ratio, 2.27, 95%CI: 1.31-3.94; P = 3.7 × 10-3), in the replication cohort, (hazard ratio 1.47, 95%CI: 1.04-2.07; P = 2.9 × 10-2), and in combined cohort hazard ratio 1.62 (95%CI: 1.21-2.18; P = 1.1 × 10-3). The common SNP of TRPV1 (rs224534) is associated with the prognosis of ICM, and homozygote rs224534-AA showed an unfavorable prognosis of ICM in the dominant model tested. Genotyping the variant may benefit to further progress judgment of ICM.
Collapse
Affiliation(s)
- Shiyang Li
- Division of Cardiology, Panzhihua Central Hospital, Panzhihua, China
- *Correspondence: Panzhihua Central Hospital, 34# Yi kang Ave., Panzhihua 617000, China (e-mail: )
| | - Lei Xiao
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Huazhong University of Science and Technology, Hubei Province, Wuhan, China
| | - Yang Sun
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Huazhong University of Science and Technology, Hubei Province, Wuhan, China
| | - Senlin Hu
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Huazhong University of Science and Technology, Hubei Province, Wuhan, China
| | - Dong Hu
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Huazhong University of Science and Technology, Hubei Province, Wuhan, China
| |
Collapse
|
17
|
Chen Y, Su Y, Wang F. The Piezo1 ion channel in glaucoma: a new perspective on mechanical stress. Hum Cell 2022; 35:1307-1322. [PMID: 35767143 DOI: 10.1007/s13577-022-00738-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Accepted: 06/13/2022] [Indexed: 11/26/2022]
Abstract
Glaucomatous optic nerve damage caused by pathological intraocular pressure elevation is irreversible, and its course is often difficult to control. This group of eye diseases is closely related to biomechanics, and the correlation between glaucoma pathogenesis and mechanical stimulation has been studied in recent decades. The nonselective cation channel Piezo1, the most important known mechanical stress sensor, is a transmembrane protein widely expressed in various cell types. Piezo1 has been detected throughout the eye, and the close relationship between Piezo1 and glaucoma is being confirmed. Pathological changes in glaucoma occur in both the anterior and posterior segments of the eye, and it is of great interest for researchers to determine whether Piezo1 plays a role in these changes and how it functions. The elucidation of the mechanisms of Piezo1 action in nonocular tissues and the reported roles of similar mechanically activated ion channels in glaucoma will provide an appropriate basis for further investigation. From a new perspective, this review provides a detailed description of the current progress in elucidating the role of Piezo1 in glaucoma, including relevant questions and assumptions, the remaining challenging research directions and mechanism-related therapeutic potential.
Collapse
Affiliation(s)
- Yidan Chen
- Department of Ophthalmology, Fourth Affiliated Hospital, Harbin Medical University, Yiyuan Road, Harbin, 150001, China
| | - Ying Su
- Eye Hospital, First Affiliated Hospital, Harbin Medical University, Yiman Road, Harbin, 150007, China.
| | - Feng Wang
- Department of Ophthalmology, Fourth Affiliated Hospital, Harbin Medical University, Yiyuan Road, Harbin, 150001, China.
| |
Collapse
|
18
|
Streiff ME, Sachse FB. Effects of Sarcolemmal Background Ca2+ Entry and Sarcoplasmic Ca2+ Leak Currents on Electrophysiology and Ca2+ Transients in Human Ventricular Cardiomyocytes: A Computational Comparison. Front Physiol 2022; 13:916278. [PMID: 35784869 PMCID: PMC9243544 DOI: 10.3389/fphys.2022.916278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Accepted: 05/27/2022] [Indexed: 11/30/2022] Open
Abstract
The intricate regulation of the compartmental Ca2+ concentrations in cardiomyocytes is critical for electrophysiology, excitation-contraction coupling, and other signaling pathways. Research into the complex signaling pathways is motivated by cardiac pathologies including arrhythmia and maladaptive myocyte remodeling, which result from Ca2+ dysregulation. Of interest to this investigation are two types of Ca2+ currents in cardiomyocytes: 1) background Ca2+ entry, i.e., Ca2+ transport across the sarcolemma from the extracellular space into the cytosol, and 2) Ca2+ leak from the sarcoplasmic reticulum (SR) across the SR membrane into the cytosol. Candidates for the ion channels underlying background Ca2+ entry and SR Ca2+ leak channels include members of the mechano-modulated transient receptor potential (TRP) family. We used a mathematical model of a human ventricular myocyte to analyze the individual contributions of background Ca2+ entry and SR Ca2+ leak to the modulation of Ca2+ transients and SR Ca2+ load at rest and during action potentials. Background Ca2+ entry exhibited a positive relationship with both [Ca2+]i and [Ca2+]SR. Modulating SR Ca2+ leak had opposite effects of background Ca2+ entry. Effects of SR Ca2+ leak on Ca2+ were particularly pronounced at lower pacing frequency. In contrast to the pronounced effects of background and leak Ca2+ currents on Ca2+ concentrations, the effects on cellular electrophysiology were marginal. Our studies provide quantitative insights into the differential modulation of compartmental Ca2+ concentrations by the background and leak Ca2+ currents. Furthermore, our studies support the hypothesis that TRP channels play a role in strain-modulation of cardiac contractility. In summary, our investigations shed light on the physiological effects of the background and leak Ca2+ currents and their contribution to the development of disease caused by Ca2+ dysregulation.
Collapse
Affiliation(s)
- Molly E. Streiff
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, United States
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, United States
| | - Frank B. Sachse
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, United States
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, United States
- *Correspondence: Frank B. Sachse,
| |
Collapse
|
19
|
Brandt MM, Cheng C, Merkus D, Duncker DJ, Sorop O. Mechanobiology of Microvascular Function and Structure in Health and Disease: Focus on the Coronary Circulation. Front Physiol 2022; 12:771960. [PMID: 35002759 PMCID: PMC8733629 DOI: 10.3389/fphys.2021.771960] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 11/11/2021] [Indexed: 12/19/2022] Open
Abstract
The coronary microvasculature plays a key role in regulating the tight coupling between myocardial perfusion and myocardial oxygen demand across a wide range of cardiac activity. Short-term regulation of coronary blood flow in response to metabolic stimuli is achieved via adjustment of vascular diameter in different segments of the microvasculature in conjunction with mechanical forces eliciting myogenic and flow-mediated vasodilation. In contrast, chronic adjustments in flow regulation also involve microvascular structural modifications, termed remodeling. Vascular remodeling encompasses changes in microvascular diameter and/or density being largely modulated by mechanical forces acting on the endothelium and vascular smooth muscle cells. Whereas in recent years, substantial knowledge has been gathered regarding the molecular mechanisms controlling microvascular tone and how these are altered in various diseases, the structural adaptations in response to pathologic situations are less well understood. In this article, we review the factors involved in coronary microvascular functional and structural alterations in obstructive and non-obstructive coronary artery disease and the molecular mechanisms involved therein with a focus on mechanobiology. Cardiovascular risk factors including metabolic dysregulation, hypercholesterolemia, hypertension and aging have been shown to induce microvascular (endothelial) dysfunction and vascular remodeling. Additionally, alterations in biomechanical forces produced by a coronary artery stenosis are associated with microvascular functional and structural alterations. Future studies should be directed at further unraveling the mechanisms underlying the coronary microvascular functional and structural alterations in disease; a deeper understanding of these mechanisms is critical for the identification of potential new targets for the treatment of ischemic heart disease.
Collapse
Affiliation(s)
- Maarten M Brandt
- Division of Experimental Cardiology, Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Caroline Cheng
- Division of Experimental Cardiology, Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands.,Division of Internal Medicine and Dermatology, Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, Netherlands
| | - Daphne Merkus
- Division of Experimental Cardiology, Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands.,Walter Brendel Center of Experimental Medicine (WBex), LMU Munich, Munich, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart Alliance (MHA), Munich, Germany
| | - Dirk J Duncker
- Division of Experimental Cardiology, Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Oana Sorop
- Division of Experimental Cardiology, Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| |
Collapse
|
20
|
Miller M, Koch SE, Veteto A, Domeier T, Rubinstein J. Role of Known Transient Receptor Potential Vanilloid Channels in Modulating Cardiac Mechanobiology. Front Physiol 2021; 12:734113. [PMID: 34867442 PMCID: PMC8637880 DOI: 10.3389/fphys.2021.734113] [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: 06/30/2021] [Accepted: 08/27/2021] [Indexed: 12/17/2022] Open
Abstract
The transient receptor potential (TRP) channels have been described in almost every mammalian cell type. Several members of the Vanilloid (TRPV) subtype have been found to play important roles in modulating cardiac structure and function through Ca2+ handling in response to systemic and local mechanobiological cues. In this review, we will consider the most studied TRPV channels in the cardiovascular field; transient receptor potential vanilloid 1 as a modulator of cardiac hypertrophy; transient receptor potential vanilloid 2 as a structural and functional protein; transient receptor potential vanilloid 3 in the development of hypertrophy and myocardial fibrosis; and transient receptor potential vanilloid 4 in its roles modulating the fibrotic and functional responses of the heart to pressure overload. Lastly, we will also review the potential overlapping roles of these channels with other TRP proteins as well as the advances in translational and clinical arenas associated with TRPV channels.
Collapse
Affiliation(s)
- Michael Miller
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, KY, United States
| | - Sheryl E Koch
- Department of Internal Medicine, Division of Cardiovascular Health and Disease, College of Medicine, University of Cincinnati Medical Center, Cincinnati, OH, United States
| | - Adam Veteto
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, KY, United States.,IonOptix, LLC, Westwood, MA, United States
| | - Timothy Domeier
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, KY, United States
| | - Jack Rubinstein
- Department of Internal Medicine, Division of Cardiovascular Health and Disease, College of Medicine, University of Cincinnati Medical Center, Cincinnati, OH, United States.,Division of Cardiovascular Medicine, Cincinnati Veterans Affairs Medical Center, Cincinnati, OH, United States
| |
Collapse
|
21
|
Bektur Aykanat NE, Şahin E, Kaçar S, Bağcı R, Karakaya Ş, Burukoğlu Dönmez D, Şahintürk V. Cardiac hypertrophy caused by hyperthyroidism in rats: the role of ATF-6 and TRPC1 channels. Can J Physiol Pharmacol 2021; 99:1226-1233. [PMID: 34283935 DOI: 10.1139/cjpp-2021-0260] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Hyperthyroidism influences the development of cardiac hypertrophy. Transient receptor potential canonical channels (TRPCs) and endoplasmic reticulum (ER) stress are regarded as critical pathways in cardiac hypertrophy. Hence, we aimed to identify the TRPCs associated with ER stress in hyperthyroidism-induced cardiac hypertrophy. Twenty adult Wistar albino male rats were used in the study. The control group was fed with standard food and tap water. The group with hyperthyroidism was also fed with standard rat food, along with tap water that contained 12 mg/L of thyroxine (T4) for 4 weeks. At the end of the fourth week, the serum-free triiodothyronine (T3), T4, and thyroid-stimulating hormone (TSH) levels of the groups were measured. The left ventricle of each rat was used for histochemistry, immunohistochemistry, Western blot, total antioxidant capacity (TAC), and total oxidant status (TOS) analysis. As per our results, activating transcription factor 6 (ATF-6), inositol-requiring kinase 1 (IRE-1), and TRPC1, which play a significant role in cardiac hypertrophy caused by hyperthyroidism, showed increased activation. Moreover, TOS and serum-free T3 levels increased, while TAC and TSH levels decreased. With the help of the literature review in our study, we could, for the first time, indicate that the increased activation of ATF-6, IRE-1, and TRPC1-induced deterioration of the Ca2+ ion balance leads to hypertrophy in hyperthyroidism due to heart failure.
Collapse
Affiliation(s)
| | - Erhan Şahin
- Department of Histology and Embryology, Eskişehir Osmangazi University Faculty of Medicine, Eskişehir, Turkey
| | - Sedat Kaçar
- Department of Histology and Embryology, Eskişehir Osmangazi University Faculty of Medicine, Eskişehir, Turkey
| | - Rıdvan Bağcı
- Adana City Training and Research Hospital, Adana, Turkey
| | - Şerife Karakaya
- Department of Histology and Embryology, Eskişehir Osmangazi University Faculty of Medicine, Eskişehir, Turkey
| | - Dilek Burukoğlu Dönmez
- Department of Histology and Embryology, Eskişehir Osmangazi University Faculty of Medicine, Eskişehir, Turkey
| | - Varol Şahintürk
- Department of Histology and Embryology, Eskişehir Osmangazi University Faculty of Medicine, Eskişehir, Turkey
| |
Collapse
|
22
|
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
|
23
|
Turner D, Kang C, Mesirca P, Hong J, Mangoni ME, Glukhov AV, Sah R. Electrophysiological and Molecular Mechanisms of Sinoatrial Node Mechanosensitivity. Front Cardiovasc Med 2021; 8:662410. [PMID: 34434970 PMCID: PMC8382116 DOI: 10.3389/fcvm.2021.662410] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 06/24/2021] [Indexed: 01/01/2023] Open
Abstract
The understanding of the electrophysiological mechanisms that underlie mechanosensitivity of the sinoatrial node (SAN), the primary pacemaker of the heart, has been evolving over the past century. The heart is constantly exposed to a dynamic mechanical environment; as such, the SAN has numerous canonical and emerging mechanosensitive ion channels and signaling pathways that govern its ability to respond to both fast (within second or on beat-to-beat manner) and slow (minutes) timescales. This review summarizes the effects of mechanical loading on the SAN activity and reviews putative candidates, including fast mechanoactivated channels (Piezo, TREK, and BK) and slow mechanoresponsive ion channels [including volume-regulated chloride channels and transient receptor potential (TRP)], as well as the components of mechanochemical signal transduction, which may contribute to SAN mechanosensitivity. Furthermore, we examine the structural foundation for both mechano-electrical and mechanochemical signal transduction and discuss the role of specialized membrane nanodomains, namely, caveolae, in mechanical regulation of both membrane and calcium clock components of the so-called coupled-clock pacemaker system responsible for SAN automaticity. Finally, we emphasize how these mechanically activated changes contribute to the pathophysiology of SAN dysfunction and discuss controversial areas necessitating future investigations. Though the exact mechanisms of SAN mechanosensitivity are currently unknown, identification of such components, their impact into SAN pacemaking, and pathological remodeling may provide new therapeutic targets for the treatment of SAN dysfunction and associated rhythm abnormalities.
Collapse
Affiliation(s)
- Daniel Turner
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, United States
| | - Chen Kang
- Cardiovascular Division, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, United States
| | - Pietro Mesirca
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Juan Hong
- Cardiovascular Division, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, United States
| | - Matteo E Mangoni
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Alexey V Glukhov
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, United States
| | - Rajan Sah
- Cardiovascular Division, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, United States
| |
Collapse
|
24
|
TRPV1 activation and internalization is part of the LPS-induced inflammation in human iPSC-derived cardiomyocytes. Sci Rep 2021; 11:14689. [PMID: 34282193 PMCID: PMC8289830 DOI: 10.1038/s41598-021-93958-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 06/28/2021] [Indexed: 12/18/2022] Open
Abstract
The non-selective cation channel transient receptor potential vanilloid 1 (TRPV1) is expressed throughout the cardiovascular system. Recent evidence shows a role for TRPV1 in inflammatory processes. The role of TRPV1 for myocardial inflammation has not been established yet. Human induced pluripotent stem cell (iPSC)-derived cardiomyocytes (hiPSC-CM) from 4 healthy donors were incubated with lipopolysaccharides (LPS, 6 h), TRPV1 agonist capsaicin (CAP, 20 min) or the antagonist capsazepine (CPZ, 20 min). TRPV1 expression was studied by PCR and western blotting. TRPV1 internalization was analyzed by immunofluorescence. Interleukin-6 (IL-6) secretion and phosphorylation of JNK, p38 and ERK were determined by ELISA. TRPV1-associated ion channel current was measured by patch clamp. TRPV1-mRNA and -protein were expressed in hiPSC-CM. TRPV1 was localized in the plasma membrane. LPS significantly increased secretion of IL-6 by 2.3-fold, which was prevented by pre-incubation with CPZ. LPS induced TRPV1 internalization. Phosphorylation levels of ERK, p38 or JNK were not altered by TRPV1 stimulation or inhibition. LPS and IL-6 significantly lowered TRPV1-mediated ion channel current. TRPV1 mediates the LPS-induced inflammation in cardiomyocytes, associated with changes of cellular electrophysiology. LPS-induced inflammation results in TRPV1 internalization. Further studies have to examine the underlying pathways and the clinical relevance of these findings.
Collapse
|
25
|
Yan X, Zhang S, Zhao H, Liu P, Huang H, Niu W, Wang W, Zhang C. ASIC2 Synergizes with TRPV1 in the Mechano-Electrical Transduction of Arterial Baroreceptors. Neurosci Bull 2021; 37:1381-1396. [PMID: 34215968 DOI: 10.1007/s12264-021-00737-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 03/09/2021] [Indexed: 11/24/2022] Open
Abstract
Mechanosensitive ion channels (MSCs) are key molecules in the mechano-electrical transduction of arterial baroreceptors. Among them, acid-sensing ion channel 2 (ASIC2) and transient receptor potential vanilloid subfamily member 1 (TRPV1) have been studied extensively and documented to play important roles. In this study, experiments using aortic arch-aortic nerve preparations isolated from rats revealed that both ASIC2 and TRPV1 are functionally necessary, as blocking either abrogated nearly all pressure-dependent neural discharge. However, whether ASIC2 and TRPV1 work in coordination remained unclear. So we carried out cell-attached patch-clamp recordings in HEK293T cells co-expressing ASIC2 and TRPV1 and found that inhibition of ASIC2 completely blocked stretch-activated currents while inhibition of TRPV1 only partially blocked these currents. Immunofluorescence staining of aortic arch-aortic adventitia from rats showed that ASIC2 and TRPV1 are co-localized in the aortic nerve endings, and co-immunoprecipitation assays confirmed that the two proteins form a compact complex in HEK293T cells and in baroreceptors. Moreover, protein modeling analysis, exogenous co-immunoprecipitation assays, and biotin pull-down assays indicated that ASIC2 and TRPV1 interact directly. In summary, our research suggests that ASIC2 and TRPV1 form a compact complex and function synergistically in the mechano-electrical transduction of arterial baroreceptors. The model of synergism between MSCs may have important biological significance beyond ASIC2 and TRPV1.
Collapse
Affiliation(s)
- Xiaodong Yan
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Sitao Zhang
- Department of Orthopedics, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Haiyan Zhao
- Yanjing Medical College, Capital Medical University, Beijing, 101300, China
| | - Ping Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Haixia Huang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Weizhen Niu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Wei Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China. .,Beijing Laboratory for Cardiovascular Precision Medicine, Capital Medical University, Beijing, 100069, China.
| | - Chen Zhang
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|
26
|
de Coulon E, Dellenbach C, Rohr S. Advancing mechanobiology by performing whole-cell patch clamp recording on mechanosensitive cells subjected simultaneously to dynamic stretch events. iScience 2021; 24:102041. [PMID: 33532717 PMCID: PMC7822953 DOI: 10.1016/j.isci.2021.102041] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 12/03/2020] [Accepted: 01/04/2021] [Indexed: 11/05/2022] Open
Abstract
A comprehensive understanding of mechano-electrical coupling requires continuous intracellular electrical recordings being performed on cells undergoing simultaneously in vivo like strain events. Here, we introduce a linear strain single-cell electrophysiology (LSSE) system that meets these requirements by delivering highly reproducible unidirectional strain events with magnitudes up to 12% and strain rates exceeding 200%s−1 to adherent cells kept simultaneously in whole-cell patch-clamp recording configuration. Proof-of-concept measurements with NIH3T3 cells demonstrate that stable recording conditions are maintained over tens of strain cycles at maximal amplitudes and strain rates thereby permitting a full electrophysiological characterization of mechanically activated ion currents. Because mechano-electrical responses to predefined strain patterns can be investigated using any adherent wild-type or genetically modified cell type of interest, the LSSE system offers the perspective of providing advanced insights into mechanosensitive ion channel function that can finally be compared quantitatively among different types of channels and cells. The methodology presented enables investigations of adherent mechanosensitive cells Whole-cell patch-clamp recording is performed while cells are dynamically stretched Continuous recording of sequences of physiological mechanical stimuli is practicable Experiments with NIH3T3 cells reveal a robust atypical mechanosensitive current
Collapse
Affiliation(s)
- Etienne de Coulon
- Department of Physiology, University of Bern, Bühlplatz 5, Bern, CH-3012, Switzerland
| | - Christian Dellenbach
- Department of Physiology, University of Bern, Bühlplatz 5, Bern, CH-3012, Switzerland
| | - Stephan Rohr
- Department of Physiology, University of Bern, Bühlplatz 5, Bern, CH-3012, Switzerland
| |
Collapse
|
27
|
Yadav S, Ta HT, Nguyen N. Mechanobiology in cardiology: Micro‐ and nanotechnologies to probe mechanosignaling. VIEW 2021. [DOI: 10.1002/viw.20200080] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Affiliation(s)
- Sharda Yadav
- Queensland Micro‐ and Nanotechnology Centre Griffith University Nathan Queensland Australia
| | - Hang T. Ta
- Queensland Micro‐ and Nanotechnology Centre Griffith University Nathan Queensland Australia
- School of Environment and Science Griffith University Nathan Queensland Australia
| | - Nam‐Trung Nguyen
- Queensland Micro‐ and Nanotechnology Centre Griffith University Nathan Queensland Australia
| |
Collapse
|
28
|
Zhu W, Hou F, Fang J, Bahrani Fard MR, Liu Y, Ren S, Wu S, Qi Y, Sui S, Read AT, Sherwood JM, Zou W, Yu H, Zhang J, Overby DR, Wang N, Ethier CR, Wang K. The role of Piezo1 in conventional aqueous humor outflow dynamics. iScience 2021; 24:102042. [PMID: 33532718 PMCID: PMC7829208 DOI: 10.1016/j.isci.2021.102042] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 12/16/2020] [Accepted: 01/05/2021] [Indexed: 12/16/2022] Open
Abstract
Controlling intraocular pressure (IOP) remains the mainstay of glaucoma therapy. The trabecular meshwork (TM), the key tissue responsible for aqueous humor (AH) outflow and IOP maintenance, is very sensitive to mechanical forces. However, it is not understood whether Piezo channels, very sensitive mechanosensors, functionally influence AH outflow. Here, we characterize the role of Piezo1 in conventional AH outflow. Immunostaining and western blot analysis showed that Piezo1 is widely expressed by TM. Patch-clamp recordings in TM cells confirmed the activation of Piezo1-derived mechanosensitive currents. Importantly, the antagonist GsMTx4 for mechanosensitive channels significantly decreased steady-state facility, yet activation of Piezo1 by the specific agonist Yoda1 did not lead to a facility change. Furthermore, GsMTx4, but not Yoda1, caused a significant increase in ocular compliance, a measure of the eye's transient response to IOP perturbation. Our findings demonstrate a potential role for Piezo1 in conventional outflow, likely under pathological and rapid transient conditions. Piezo1 is functionally expressed in the TM, the most important tissue controlling IOP Suppression of mechanosensitive channel leads to a significant decrease in facility Our data suggest a role for Piezo in pathological situations and rapid IOP transients
Collapse
Affiliation(s)
- Wei Zhu
- Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao University Medical College, 38 Dengzhou Road, Qingdao 266021, Shandong, China.,Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Beihang University & Capital Medical University, Beijing 100730, China
| | - Fei Hou
- Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao University Medical College, 38 Dengzhou Road, Qingdao 266021, Shandong, China.,Department of Pharmacy, Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Jingwang Fang
- Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao University Medical College, 38 Dengzhou Road, Qingdao 266021, Shandong, China
| | - Mohammad Reza Bahrani Fard
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta 30332, GA, USA
| | - Yani Liu
- Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao University Medical College, 38 Dengzhou Road, Qingdao 266021, Shandong, China
| | - Shouyan Ren
- Department of Pharmacy, Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Shen Wu
- Beijing Institute of Ophthalmology, Beijing Tongren Hospital Eye Center, Beijing 100730 China
| | - Yunkun Qi
- Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao University Medical College, 38 Dengzhou Road, Qingdao 266021, Shandong, China
| | - Shangru Sui
- Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao University Medical College, 38 Dengzhou Road, Qingdao 266021, Shandong, China
| | - A Thomas Read
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta 30332, GA, USA
| | | | - Wei Zou
- School of Mechatronic Engineering, Jiangsu Normal University, Xuzhou 221116, China
| | - Hongxia Yu
- Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao University Medical College, 38 Dengzhou Road, Qingdao 266021, Shandong, China
| | - Jingxue Zhang
- Beijing Institute of Ophthalmology, Beijing Tongren Hospital Eye Center, Beijing 100730 China
| | - Darryl R Overby
- Department of Bioengineering, Imperial College London, London, UK
| | - Ningli Wang
- Beijing Institute of Ophthalmology, Beijing Tongren Hospital Eye Center, Beijing 100730 China
| | - C Ross Ethier
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta 30332, GA, USA.,Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, 30332, GA, United States
| | - KeWei Wang
- Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao University Medical College, 38 Dengzhou Road, Qingdao 266021, Shandong, China.,Institute of Innovative Drugs, Qingdao University, 38 Dengzhou Road, Qingdao 266021, Shandong, China
| |
Collapse
|
29
|
Campbell T, Shenton FC, Lucking E, Pyner S, Jones JFX. Electrophysiological characterisation of atrial volume receptors using ex vivo models of isolated rat cardiac atria. Exp Physiol 2020; 105:2190-2206. [PMID: 33372723 DOI: 10.1113/ep088972] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 10/12/2020] [Indexed: 11/08/2022]
Abstract
NEW FINDINGS What is the central question of this study? What ex vivo preparation of the rat's cavoatrial junction is efficient for characterising atrial mechanoreceptors? What is the main finding and its importance? Of four different ex vivo preparations, static pressure, flow, open and euthermic, the optimal preparation was the euthermic one and involved direct recording from the right cardiac vagal branch with a Langendorff style perfusion at 37°C. Type A receptors were most common, and appeared insensitive to stretch and sensitive to atrial contraction. Type B and intermediate receptors were not isolated at 20°C but were observed closer to 37°C. The findings may suggest that type A and B receptors utilise different molecular transduction mechanisms. ABSTRACT Atrial volume receptors are a family of afferent neurons whose mechanically sensitive endings terminate in the atria, particularly at the cavoatrial junctions. These mechanosensors form the afferent limb of an atrial volume receptor reflex that regulates plasma volume. The prevailing functional classification of atrial receptors arose as a result of in vivo recordings in the cat and dog and were classified as type A, B or intermediate according to the timing of peak discharge during the cardiac cycle. In contrast, there have been far fewer studies of the common small laboratory mammals such as the rat. Using several ex vivo rat cavoatrial preparations, a total of 30 successful single cavoatrial mechanosensory recordings were obtained. These experiments show that the rat possesses type A, B and intermediate atrial mechanoreceptors as described for larger mammals. Recording these cavoatrial receptors proved challenging from the main vagus, but direct recording from the cardiac vagal branch greatly increased the yield of mechanically sensitive single units. In contrast to type A units, type B atrial mechanoreceptor activity was never observed at room temperature but required elevation of temperature to a more physiological range in order to be detected. The adequate stimulus for these receptors remains unclear; however, type A atrial receptors appear insensitive to direct atrial stretch when applied using a programmable positioner. The findings may suggest that type A and type B atrial receptors utilise different molecular transduction mechanisms.
Collapse
Affiliation(s)
- Thomas Campbell
- Discipline of Anatomy, School of Medicine, University College Dublin, Dublin, Ireland
| | | | - Eric Lucking
- Discipline of Anatomy, School of Medicine, University College Dublin, Dublin, Ireland
| | - Susan Pyner
- Department of Biosciences, Durham University, Durham, UK
| | | |
Collapse
|
30
|
Freeberg MAT, Perelas A, Rebman JK, Phipps RP, Thatcher TH, Sime PJ. Mechanical Feed-Forward Loops Contribute to Idiopathic Pulmonary Fibrosis. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 191:18-25. [PMID: 33031756 PMCID: PMC7768346 DOI: 10.1016/j.ajpath.2020.09.008] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 08/25/2020] [Accepted: 09/08/2020] [Indexed: 12/11/2022]
Abstract
Idiopathic pulmonary fibrosis is a progressive scarring disease characterized by extracellular matrix accumulation and altered mechanical properties of lung tissue. Recent studies support the hypothesis that these compositional and mechanical changes create a progressive feed-forward loop in which enhanced matrix deposition and tissue stiffening contribute to fibroblast and myofibroblast differentiation and activation, which further perpetuates matrix production and stiffening. The biomechanical properties of tissues are sensed and responded to by mechanotransduction pathways that facilitate sensing of changes in mechanical cues by tissue resident cells and convert the mechanical signals into downstream biochemical signals. Although our understanding of mechanotransduction pathways associated with pulmonary fibrosis remains incomplete, recent progress has allowed us to begin to elucidate the specific mechanisms supporting fibrotic feed-forward loops. The mechanosensors discussed here include integrins, Piezo channels, transient receptor potential channels, and nonselective ion channels. Also discussed are downstream transcription factors, including myocardin-related transcription factor and Yes-associated protein/transcriptional coactivator with PDZ-binding motif. This review describes mechanosensors and mechanotransduction pathways associated with fibrosis progression and highlights promising therapeutic insights.
Collapse
Affiliation(s)
- Margaret A T Freeberg
- Division of Pulmonary Disease and Critical Care Medicine, Virginia Commonwealth University, Richmond, Virginia
| | - Apostolos Perelas
- Division of Pulmonary Disease and Critical Care Medicine, Virginia Commonwealth University, Richmond, Virginia
| | - Jane K Rebman
- Division of Pulmonary Disease and Critical Care Medicine, Virginia Commonwealth University, Richmond, Virginia
| | | | - Thomas H Thatcher
- Division of Pulmonary Disease and Critical Care Medicine, Virginia Commonwealth University, Richmond, Virginia
| | - Patricia J Sime
- Division of Pulmonary Disease and Critical Care Medicine, Virginia Commonwealth University, Richmond, Virginia.
| |
Collapse
|
31
|
Shamsaldeen YA, Lione LA, Benham CD. Dysregulation of TRPV4, eNOS and caveolin-1 contribute to endothelial dysfunction in the streptozotocin rat model of diabetes. Eur J Pharmacol 2020; 888:173441. [PMID: 32810492 DOI: 10.1016/j.ejphar.2020.173441] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 07/28/2020] [Accepted: 07/29/2020] [Indexed: 12/30/2022]
Abstract
Endothelial dysfunction is a common complication in diabetes in which endothelium-dependent vasorelaxation is impaired. The aim of this study was to examine the involvement of the TRPV4 ion channel in type 1 diabetic endothelial dysfunction and the possible association of endothelial dysfunction with reduced expression of TRPV4, endothelial nitric oxide synthase (eNOS) and caveolin-1. Male Wistar rats (350-450 g) were injected with 65 mg/kg i.p. streptozotocin (STZ) or vehicle. Endothelial function was investigated in aortic rings and mesenteric arteries using organ bath and myograph, respectively. TRPV4 function was studied with fura-2 calcium imaging in endothelial cells cultured from aortas from control and STZ treated rats. TRPV4, caveolin-1 and eNOS expression was investigated in these cells using immunohistochemistry. STZ-treated diabetic rats showed significant endothelial dysfunction characterised by impaired muscarinic-induced vasorelaxation (aortic rings: STZ-diabetics: Emax = 29.6 ± 9.3%; control: Emax = 77.2 ± 2.5% P˂0.001), as well as significant impairment in TRPV4-induced vasorelaxation (aortic rings, 4αPDD STZ-diabetics: Emax = 56.0 ± 5.5%; control: Emax = 81.1 ± 2.1% P˂0.001). Furthermore, STZ-diabetic primary aortic endothelial cells showed a significant reduction in TRPV4-induced intracellular calcium elevation (P˂0.05) compared with the control group. This was associated with significantly lower expression of TRPV4, caveolin-1 and eNOS and this was reversed by insulin treatment of the endothelial cultures from STZ -diabetic rats. Taken together, these data are consistent with the hypothesis that signalling through TRPV4, caveolin-1, and eNOS is downregulated in STZ-diabetic aortic endothelial cells and restored by insulin treatment.
Collapse
Affiliation(s)
- Yousif A Shamsaldeen
- School of Life and Medical Sciences, University of Hertfordshire, College Lane, Hatfield, Hertfordshire, AL10 9AB, UK; Department of Pharmacy, Kuwait Hospital, Sabah Alsalem, 44001, Kuwait.
| | - Lisa A Lione
- School of Life and Medical Sciences, University of Hertfordshire, College Lane, Hatfield, Hertfordshire, AL10 9AB, UK
| | - Christopher D Benham
- School of Life and Medical Sciences, University of Hertfordshire, College Lane, Hatfield, Hertfordshire, AL10 9AB, UK
| |
Collapse
|
32
|
Gao S, Kaudimba KK, Guo S, Zhang S, Liu T, Chen P, Wang R. Transient Receptor Potential Ankyrin Type-1 Channels as a Potential Target for the Treatment of Cardiovascular Diseases. Front Physiol 2020; 11:836. [PMID: 32903613 PMCID: PMC7438729 DOI: 10.3389/fphys.2020.00836] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Accepted: 06/22/2020] [Indexed: 12/15/2022] Open
Abstract
Cardiovascular disease is one of the chronic conditions with the highest mortality rate in the world. Underlying conditions such as hypertension, metabolic disorders, and habits like smoking are contributors to the manifestation of cardiovascular diseases. The treatment of cardiovascular diseases is inseparable from the development of drugs. Consequently, this has led to many researchers to focus on the search for effective drug targets. The transient receptor potential channel Ankyrin 1 (TRPA1) subtype is a non-selective cation channel, which belongs to the transient receptor potential (TRP) ion channel. Previous studies have shown that members of the TRP family contribute significantly to cardiovascular disease. However, many researchers have not explored the role of TRPA1 as a potential target for the treatment of cardiovascular diseases. Furthermore, recent studies revealed that TRPA1 is commonly expressed in the vascular endothelium. The endothelium is linked to the causes of some cardiovascular diseases, such as atherosclerosis, myocardial fibrosis, heart failure, and arrhythmia. The activation of TRPA1 has a positive effect on atherosclerosis, but it has a negative effect on other cardiovascular diseases such as myocardial fibrosis and heart failure. This review introduces the structural and functional characteristics of TRPA1 and its importance on vascular physiology and common cardiovascular diseases. Moreover, this review summarizes some evidence that TRPA1 is correlated to cardiovascular disease risk factors.
Collapse
Affiliation(s)
- Song Gao
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | | | - Shanshan Guo
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Shuang Zhang
- School of Kinesiology, Shanghai University of Sport, Shanghai, China.,Institute of Sport Science, Harbin Sport University, Harbin, China
| | - Tiemin Liu
- School of Kinesiology, Shanghai University of Sport, Shanghai, China.,State Key Laboratory of Genetic Engineering, Institute of Metabolism and Integrative Biology, Human Phenome Institute, Department of Endocrinology and Metabolism, and School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Peijie Chen
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Ru Wang
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| |
Collapse
|
33
|
Wang J, He Y, Yang G, Li N, Li M, Zhang M. Transient receptor potential canonical 1 channel mediates the mechanical stress‑induced epithelial‑mesenchymal transition of human bronchial epithelial (16HBE) cells. Int J Mol Med 2020; 46:320-330. [PMID: 32319532 PMCID: PMC7255483 DOI: 10.3892/ijmm.2020.4568] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2019] [Accepted: 03/17/2020] [Indexed: 01/16/2023] Open
Abstract
Airway remodeling is a central event in the pathology of chronic obstructive pulmonary disease (COPD) that leads to airway narrowing and subsequently, to increased mechanical pressure. High mechanical pressure can exacerbate airway remodeling. Thus, a treatment regimen aimed at disrupting this high‑pressure airway remodeling vicious cycle may improve the prognosis of patients with COPD. Recent studies have demonstrated that mechanical stress induces lung epithelial‑mesenchymal transition (EMT), which is commonly present in airway epithelial cells of patients with COPD. As TRPC1 functions as a mechanosensitive channel that mediates non‑selective cation entry in response to increased membrane stretch, the present study investigated the role of TRPC1 in the occurrence of EMT induced by mechanical stress. In the present study, the expression of TRPC1 in the bronchial epithelium was examined in vivo by immunohistochemistry. In vitro, human bronchial epithelial (16HBE) cells were subjected to mechanical stretching for up to 48 h, and TRPC1 expression was then examined by RT‑qPCR and western blot analysis. In addition, TRPC1 receptor function was assessed by Ca2+ imaging and siRNA transfection. EMT was identified using immunofluorescence, western blot analysis and RT‑qPCR. It was found that TRPC1 expression was upregulated in patients with COPD and in 16HBE cells subjected to mechanical stretch. The mechanical stress‑induced activation of TRPC1 in 16HBE cells increased the intracellular calcium concentration and subsequently decreased the expression of cytokeratin 8 and E‑cadherin, and increased the expression of α‑smooth muscle actin, indicating the occurrence of EMT. On the whole, the findings of the present study demonstrate that TRPC1 plays a key role in the occurrence of EMT in human lung epithelial cells in response to mechanical stretch; thus, this protein may serve as a novel therapeutic target for progressive airway remodeling in COPD.
Collapse
Affiliation(s)
- Jing Wang
- Department of Respiratory Medicine, The Second Clinical Hospital of Chongqing Medical University, Chongqing 400010
| | - Ye He
- Department of Geriatrics, Sichuan Provincial People's Hospital, Sichuan Academy of Medical Science, Chengdu, Sichuan 610072
| | - Gang Yang
- Department of Neurosurgery, The First Clinical Hospital of Chongqing Medical University, Chongqing 400016
| | - Na Li
- Division of Nephrology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong 518107, P.R. China
| | - Minchao Li
- Department of Respiratory Medicine, The Second Clinical Hospital of Chongqing Medical University, Chongqing 400010
| | - Min Zhang
- Department of Geriatrics, Sichuan Provincial People's Hospital, Sichuan Academy of Medical Science, Chengdu, Sichuan 610072
| |
Collapse
|
34
|
Ai X, Wu Y, Lu W, Zhang X, Zhao L, Tu P, Wang K, Jiang Y. A Precise Microfluidic Assay in Single-Cell Profile for Screening of Transient Receptor Potential Channel Modulators. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:2000111. [PMID: 32537418 PMCID: PMC7284206 DOI: 10.1002/advs.202000111] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 03/03/2020] [Accepted: 03/08/2020] [Indexed: 05/14/2023]
Abstract
Transient receptor potential (TRP) channels are emerging drug targets, and TRP channel modulators possess therapeutic potential for many indications. However, there is a lack of intellectual and robust screening assays against TRP channels utilizing the least amount of compounds. Here, a precise microfluidic assay in single-cell profile is developed for the screening of TRP channel modulators. The geometrically optimized microchip is designed for both trapping single cells and utilizing passive pumping for sequential media replacement with low shear stress. The microfluidic chip exhibits superior performance in screening, repeatable compound administration, and improved reproducibility. Using this screening platform, the false-positive and negative rate of the commonly used Ca2+ imaging is reduced from 76.2% to 4.8% and four coumarin derivatives isolated from Murraya species that inhibit TRP channels are identified. One coumarin derivative B-304 reverses TRPA1-mediated inflammatory pain in vivo. Taken together, the data demonstrate that the established microfluidic assay in single-cell profile could be used for the screening of TRP channel modulators that may have therapeutic potential for the channelopathies.
Collapse
Affiliation(s)
- Xiaoni Ai
- State Key Laboratory of Natural and Biomimetic DrugsSchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
| | - Yang Wu
- State Key Laboratory of Natural and Biomimetic DrugsSchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
- Department of PharmacologyQingdao University School of Pharmacy38 Dengzhou RoadQingdao266021China
| | - Wenbo Lu
- State Key Laboratory of Natural and Biomimetic DrugsSchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
| | - Xinran Zhang
- State Key Laboratory of Natural and Biomimetic DrugsSchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
- Department of PharmacologyQingdao University School of Pharmacy38 Dengzhou RoadQingdao266021China
| | - Lin Zhao
- State Key Laboratory of Natural and Biomimetic DrugsSchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
| | - Pengfei Tu
- State Key Laboratory of Natural and Biomimetic DrugsSchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
| | - KeWei Wang
- Department of PharmacologyQingdao University School of Pharmacy38 Dengzhou RoadQingdao266021China
| | - Yong Jiang
- State Key Laboratory of Natural and Biomimetic DrugsSchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
| |
Collapse
|
35
|
Quinn TA, Kohl P. Cardiac Mechano-Electric Coupling: Acute Effects of Mechanical Stimulation on Heart Rate and Rhythm. Physiol Rev 2020; 101:37-92. [PMID: 32380895 DOI: 10.1152/physrev.00036.2019] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The heart is vital for biological function in almost all chordates, including humans. It beats continually throughout our life, supplying the body with oxygen and nutrients while removing waste products. If it stops, so does life. The heartbeat involves precise coordination of the activity of billions of individual cells, as well as their swift and well-coordinated adaption to changes in physiological demand. Much of the vital control of cardiac function occurs at the level of individual cardiac muscle cells, including acute beat-by-beat feedback from the local mechanical environment to electrical activity (as opposed to longer term changes in gene expression and functional or structural remodeling). This process is known as mechano-electric coupling (MEC). In the current review, we present evidence for, and implications of, MEC in health and disease in human; summarize our understanding of MEC effects gained from whole animal, organ, tissue, and cell studies; identify potential molecular mediators of MEC responses; and demonstrate the power of computational modeling in developing a more comprehensive understanding of ‟what makes the heart tick.ˮ.
Collapse
Affiliation(s)
- T Alexander Quinn
- Department of Physiology and Biophysics and School of Biomedical Engineering, Dalhousie University, Halifax, Nova Scotia, Canada; Institute for Experimental Cardiovascular Medicine, University Heart Centre Freiburg/Bad Krozingen, Medical Faculty of the University of Freiburg, Freiburg, Germany; and CIBSS-Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany
| | - Peter Kohl
- Department of Physiology and Biophysics and School of Biomedical Engineering, Dalhousie University, Halifax, Nova Scotia, Canada; Institute for Experimental Cardiovascular Medicine, University Heart Centre Freiburg/Bad Krozingen, Medical Faculty of the University of Freiburg, Freiburg, Germany; and CIBSS-Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany
| |
Collapse
|
36
|
Severino P, D’Amato A, Pucci M, Infusino F, Birtolo LI, Mariani MV, Lavalle C, Maestrini V, Mancone M, Fedele F. Ischemic Heart Disease and Heart Failure: Role of Coronary Ion Channels. Int J Mol Sci 2020; 21:E3167. [PMID: 32365863 PMCID: PMC7246492 DOI: 10.3390/ijms21093167] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 04/28/2020] [Accepted: 04/28/2020] [Indexed: 01/09/2023] Open
Abstract
Heart failure is a complex syndrome responsible for high rates of death and hospitalization. Ischemic heart disease is one of the most frequent causes of heart failure and it is normally attributed to coronary artery disease, defined by the presence of one or more obstructive plaques, which determine a reduced coronary blood flow, causing myocardial ischemia and consequent heart failure. However, coronary obstruction is only an element of a complex pathophysiological process that leads to myocardial ischemia. In the literature, attention paid to the role of microcirculation, in the pathophysiology of ischemic heart disease and heart failure, is growing. Coronary microvascular dysfunction determines an inability of coronary circulation to satisfy myocardial metabolic demands, due to the imbalance of coronary blood flow regulatory mechanisms, including ion channels, leading to the development of hypoxia, fibrosis and tissue death, which may determine a loss of myocardial function, even beyond the presence of atherosclerotic epicardial plaques. For this reason, ion channels may represent the link among coronary microvascular dysfunction, ischemic heart disease and consequent heart failure.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Francesco Fedele
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico, 155-00161 Rome, Italy; (P.S.); (A.D.); (M.P.); (F.I.); (L.I.B.); (M.V.M.); (C.L.); (V.M.); (M.M.)
| |
Collapse
|
37
|
Wang J, Zhao M, Jia P, Liu FF, Chen K, Meng FY, Hong JH, Zhang T, Jin XH, Shi J. The analgesic action of larixyl acetate, a potent TRPC6 inhibitor, in rat neuropathic pain model induced by spared nerve injury. J Neuroinflammation 2020; 17:118. [PMID: 32299452 PMCID: PMC7164269 DOI: 10.1186/s12974-020-01767-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Accepted: 03/05/2020] [Indexed: 01/01/2023] Open
Abstract
Background Neuropathic pain is a debilitating status that is insusceptible to the existing analgesics. It is important to explore the underlying pathophysiological changes and search for new pharmacological approaches. Transient receptor potential canonical 6 (TRPC6) is a mechanosensitive channel that is expressed by dorsal root ganglia and glial cells. It has been demonstrated that this channel in dorsal root ganglia plays essential roles in the formation of mechanical hyperalgesia in neuropathic pain. Recent pharmacological screening suggests that larixyl acetate (LA), a main constituent of larch resin, is able to selectively inhibit TRPC6 function. But whether LA is effective in treating neuropathic pain remains unknown. We investigated the efficacy of LA in rat neuropathic pain model, examined its effects on central neuroinflammation, and explored the possible molecular mechanisms by targeting the spinal dorsal horn. Methods Spared nerve injury (SNI) was conducted in Sprague-Dawley rats. Mechanical hypersensitivity and cold allodynia before and after single and multiple i.t. applications of LA at the dose of 3, 10, and 30 μM were evaluated by von Frey filament and acetone tests, respectively. Western blot, immunohistochemical, and immunocytochemical stainings were employed to examine the level and expression feature of ionized calcium-binding adaptor molecule 1 (Iba-1), glial fibrillary acidic protein (GFAP), TRPC6, and phosphorylated p38 kinase. The changes of cytokine concentrations, including that of TNF-α, IL-1β, IL-6, and IL-10, were also assessed by multiplex analysis. TRPC6 antisense strategy was finally adopted to investigate the action mechanisms of LA. Results Single application of LA on day 5 post injury caused dose-dependent inhibition of mechanical allodynia with the ED50 value of 13.43 μM. Multiple applications of LA at 30 μM not only enhanced the analgesic efficacy but also elongated the effective duration without obvious influences on animal locomotor activities. Single and multiple administrations of LA at 30 μM played similar but weaker inhibitory effects on cold allodynia. In addition to behavioral improvements, multiple applications of LA for 6 days dose-dependently inhibited the upregulation of Iba-1, TNF-α, IL-1β, and IL-6, whereas had no obvious effects on the levels of GFAP and IL-10. Combined Western blot and immunostaining assays revealed that the expression of TRPC6 was significantly increased in both spinal dorsal horn after nerve injury and the cultured microglia challenged by LPS, which was however suppressed by the addition of LA at 30 μM or 10 μM, respectively. Further knockdown of TRPC6 with antisense oligodeoxynucleotide produced prominent analgesic effects in rats with SNI, accompanied by the reduced phosphorylation level of p38 in the microglia. Conclusions These data demonstrate that i.t. applied LA exhibits analgesic and anti-inflammatory action in neuropathic pain. The action of LA involves the suppression of TRPC6 and p38 signaling in the microglia. LA may be thus a promising pharmacological candidate for the treatment of intractable chronic pain.
Collapse
Affiliation(s)
- Jing Wang
- Department of Human Anatomy, Histology and Embryology & K. K. Leung Brain Research Centre, Preclinical School of Medicine, The Fourth Military Medical University, Xi'an, 710032, China.,Student Brigade, Preclinical School of Medicine, The Fourth Military Medical University, Xi'an, 710032, China
| | - Ming Zhao
- Department of Human Anatomy, Histology and Embryology & K. K. Leung Brain Research Centre, Preclinical School of Medicine, The Fourth Military Medical University, Xi'an, 710032, China.,Student Brigade, Preclinical School of Medicine, The Fourth Military Medical University, Xi'an, 710032, China
| | - Peng Jia
- Department of Human Anatomy, Histology and Embryology & K. K. Leung Brain Research Centre, Preclinical School of Medicine, The Fourth Military Medical University, Xi'an, 710032, China.,Student Brigade, Preclinical School of Medicine, The Fourth Military Medical University, Xi'an, 710032, China
| | - Fang-Fang Liu
- Department of Neurobiology, Preclinical School of Medicine, The Fourth Military Medical University, Xi'an, 710032, China
| | - Kun Chen
- Department of Human Anatomy, Histology and Embryology & K. K. Leung Brain Research Centre, Preclinical School of Medicine, The Fourth Military Medical University, Xi'an, 710032, China
| | - Fei-Yang Meng
- Department of Human Anatomy, Histology and Embryology & K. K. Leung Brain Research Centre, Preclinical School of Medicine, The Fourth Military Medical University, Xi'an, 710032, China.,Student Brigade, Preclinical School of Medicine, The Fourth Military Medical University, Xi'an, 710032, China
| | - Jiang-Hao Hong
- Department of Human Anatomy, Histology and Embryology & K. K. Leung Brain Research Centre, Preclinical School of Medicine, The Fourth Military Medical University, Xi'an, 710032, China.,Student Brigade, Preclinical School of Medicine, The Fourth Military Medical University, Xi'an, 710032, China
| | - Ting Zhang
- Department of Human Anatomy, Histology and Embryology & K. K. Leung Brain Research Centre, Preclinical School of Medicine, The Fourth Military Medical University, Xi'an, 710032, China
| | - Xiao-Hang Jin
- Department of Human Anatomy, Histology and Embryology & K. K. Leung Brain Research Centre, Preclinical School of Medicine, The Fourth Military Medical University, Xi'an, 710032, China. .,Department of Basic Medical Morphology, Medical College, Xijing University, Xi' an, 710123, China.
| | - Juan Shi
- Department of Human Anatomy, Histology and Embryology & K. K. Leung Brain Research Centre, Preclinical School of Medicine, The Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
38
|
Andrei SR, Ghosh M, Sinharoy P, Damron DS. Stimulation of TRPA1 attenuates ischemia-induced cardiomyocyte cell death through an eNOS-mediated mechanism. Channels (Austin) 2020; 13:192-206. [PMID: 31161862 PMCID: PMC6557600 DOI: 10.1080/19336950.2019.1623591] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The functional expression of transient receptor potential cation channel of the ankyrin-1 subtype (TRPA1) has recently been identified in adult mouse cardiac tissue where stimulation of this ion channel leads to increases in adult mouse ventricular cardiomyocyte (CM) contractile function via a Ca2+-Calmodulin-dependent kinase (CaMKII) pathway. However, the extent to which TRPA1 induces nitric oxide (NO) production in CMs, and whether this signaling cascade mediates physiological or pathophysiological events in cardiac tissue remains elusive. Freshly isolated CMs from wild-type (WT) or TRPA1 knockout (TRPA1-/-) mouse hearts were treated with AITC (100 µM) and prepared for immunoblot, NO detection or ischemia protocols. Our findings demonstrate that TRPA1 stimulation with AITC results in phosphorylation of protein kinase B (Akt) and endothelial NOS (eNOS) concomitantly with NO production in a concentration- and time-dependent manner. Additionally, we found that TRPA1 induced increases in CM [Ca2+]i and contractility occur independently of Akt and eNOS activation mechanisms. Further analysis revealed that the presence and activation of TRPA1 promotes CM survival and viability following ischemic insult via a mechanism partially dependent upon eNOS. Therefore, activation of the TRPA1/Akt/eNOS pathway attenuates ischemia-induced CM cell death.
Collapse
Affiliation(s)
- Spencer R Andrei
- a Department of Medicine , Vanderbilt University Medical Center , Nashville , TN , USA
| | - Monica Ghosh
- b Department of Biomedical Sciences , Kent State University , Kent , OH , USA
| | - Pritam Sinharoy
- c Department of Biopharmaceutical Development , Medimmune LLC , Gaithersburg , MD , USA
| | - Derek S Damron
- b Department of Biomedical Sciences , Kent State University , Kent , OH , USA
| |
Collapse
|
39
|
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
|
40
|
Barabas P, Augustine J, Fernández JA, McGeown JG, McGahon MK, Curtis TM. Ion channels and myogenic activity in retinal arterioles. CURRENT TOPICS IN MEMBRANES 2020; 85:187-226. [PMID: 32402639 DOI: 10.1016/bs.ctm.2020.01.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Retinal pressure autoregulation is an important mechanism that protects the retina by stabilizing retinal blood flow during changes in arterial or intraocular pressure. Similar to other vascular beds, retinal pressure autoregulation is thought to be mediated largely through the myogenic response of small arteries and arterioles which constrict when transmural pressure increases or dilate when it decreases. Over recent years, we and others have investigated the signaling pathways underlying the myogenic response in retinal arterioles, with particular emphasis on the involvement of different ion channels expressed in the smooth muscle layer of these vessels. Here, we review and extend previous work on the expression and spatial distribution of the plasma membrane and sarcoplasmic reticulum ion channels present in retinal vascular smooth muscle cells (VSMCs) and discuss their contribution to pressure-induced myogenic tone in retinal arterioles. This includes new data demonstrating that several key players and modulators of the myogenic response show distinctively heterogeneous expression along the length of the retinal arteriolar network, suggesting differences in myogenic signaling between larger and smaller pre-capillary arterioles. Our immunohistochemical investigations have also highlighted the presence of actin-containing microstructures called myobridges that connect the retinal VSMCs to one another. Although further work is still needed, studies to date investigating myogenic mechanisms in the retina have contributed to a better understanding of how blood flow is regulated in this tissue. They also provide a basis to direct future research into retinal diseases where blood flow changes contribute to the pathology.
Collapse
Affiliation(s)
- Peter Barabas
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University of Belfast, Belfast, United Kingdom
| | - Josy Augustine
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University of Belfast, Belfast, United Kingdom
| | - José A Fernández
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University of Belfast, Belfast, United Kingdom
| | - J Graham McGeown
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University of Belfast, Belfast, United Kingdom
| | - Mary K McGahon
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University of Belfast, Belfast, United Kingdom
| | - Tim M Curtis
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University of Belfast, Belfast, United Kingdom.
| |
Collapse
|
41
|
Kaur S, Shen X, Power A, Ward ML. Stretch modulation of cardiac contractility: importance of myocyte calcium during the slow force response. Biophys Rev 2020; 12:135-142. [PMID: 31939110 DOI: 10.1007/s12551-020-00615-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 01/07/2020] [Indexed: 12/11/2022] Open
Abstract
The mechanical response of the heart to myocardial stretch has been understood since the work of muscle physiologists more than 100 years ago, whereby an increase in ventricular chamber filling during diastole increases the subsequent force of contraction. The stretch-induced increase in contraction is biphasic. There is an abrupt increase in the force that coincides with the stretch (the rapid response), which is then followed by a slower response that develops over several minutes (the slow force response, or SFR). The SFR is associated with a progressive increase in the magnitude of the Ca2+ transient, the event that initiates myocyte cross-bridge cycling and force development. However, the mechanisms underlying the stretch-dependent increase in the Ca2+ transient are still debated. This review outlines recent literature on the SFR and summarizes the different stretch-activated Ca2+ entry pathways. The SFR might result from a combination of several different cellular mechanisms initiated in response to activation of different cellular stretch sensors.
Collapse
Affiliation(s)
- Sarbjot Kaur
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Xin Shen
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,K.G.Jebsen Center for Cardiac Research, Oslo, Norway
| | - Amelia Power
- Department of Physiology, University of Otago, Dunedin, New Zealand
| | - Marie-Louise Ward
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand.
| |
Collapse
|
42
|
Yu S, Huang S, Ding Y, Wang W, Wang A, Lu Y. Transient receptor potential ion-channel subfamily V member 4: a potential target for cancer treatment. Cell Death Dis 2019; 10:497. [PMID: 31235786 PMCID: PMC6591233 DOI: 10.1038/s41419-019-1708-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Revised: 05/13/2019] [Accepted: 05/28/2019] [Indexed: 12/29/2022]
Abstract
The transient receptor potential ion-channel superfamily consists of nonselective cation channels located mostly on the plasma membranes of numerous animal cell types, which are closely related to sensory information transmission (e.g., vision, pain, and temperature perception), as well as regulation of intracellular Ca2+ balance and physiological activities of growth and development. Transient receptor potential ion channel subfamily V (TRPV) is one of the largest and most diverse subfamilies, including TRPV1-TRPV6 involved in the regulation of a variety of cellular functions. TRPV4 can be activated by various physical and chemical stimuli, such as heat, mechanical force, and phorbol ester derivatives participating in the maintenance of normal cellular functions. In recent years, the roles of TRPV4 in cell proliferation, differentiation, apoptosis, and migration have been extensively studied. Its abnormal expression has also been closely related to the onset and progression of multiple tumors, so TRPV4 may be a target for cancer diagnosis and treatment. In this review, we focused on the latest studies concerning the role of TRPV4 in tumorigenesis and the therapeutic potential. As evidenced by the effects on cancerogenesis, TRPV4 is a potential target for anticancer therapy.
Collapse
Affiliation(s)
- Suyun Yu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, P. R. China
| | - Shuai Huang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, P. R. China
| | - Yushi Ding
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, P. R. China
| | - Wei Wang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, P. R. China
| | - Aiyun Wang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, P. R. China
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing, P. R. China
| | - Yin Lu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, P. R. China.
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing, P. R. China.
| |
Collapse
|
43
|
Zhang K, Wu WY, Li G, Zhang YH, Sun Y, Qiu F, Yang Q, Xiao GS, Li GR, Wang Y. Regulation of the TRPC1 channel by endothelin-1 in human atrial myocytes. Heart Rhythm 2019; 16:1575-1583. [PMID: 30954598 DOI: 10.1016/j.hrthm.2019.04.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Indexed: 01/13/2023]
Abstract
BACKGROUND Our recent study demonstrated that the nonselective cation current mediated by the transient receptor potential canonical 1 (TRPC1) channel is activated by endothelin-1 (ET-1) in human atrial myocytes; however, the related signal molecules involved are unknown. OBJECTIVE The purpose of this study was to investigate how the TRPC1 channel is regulated by ET-1 and whether it is upregulated in human atria from patients with atrial fibrillation (AF). METHODS Whole-cell patch technique and molecular biology techniques were used in the study. RESULTS The ET-1-evoked TRPC1 current was inhibited by the ET-1 type A (ETA) receptor antagonist BQ123 and the ET-1 type B (ETB) receptor antagonist BQ788 as well as the protein kinase C inhibitor chelerythrine. ETA receptor-mediated TRPC1 channel activity was selectively inhibited by the phosphoinositide-3-kinase inhibitor wortmannin, while ETB receptor-mediated TRPC1 activity was inhibited by the phospholipase C inhibitor U73122. The messenger RNAs and proteins of the TRPC1 channel and ETA receptor, but not the ETB receptor, were significantly upregulated in atria from patients with AF. The basal TRPC1 current increased in AF myocytes, and the response to ET-1 was greater in AF myocytes than in sinus rhythm myocytes. ET-1 induced a delayed repolarization in 20% of AF myocytes. CONCLUSION These results demonstrate for the first time that TRPC1 activation by ET-1 is mediated by protein kinase C through the distinct phospholipids pathways phosphoinositide-3-kinase and phospholipase C and that the TRPC1 channel and ETA receptor are upregulated in AF atria, which are likely involved in atrial electrical remodeling in patients with AF.
Collapse
Affiliation(s)
- Kai Zhang
- Xiamen Cardiovascular Hospital, Medical School of Xiamen University, Xiamen, Fujian, China
| | - Wei-Yin Wu
- Xiamen Cardiovascular Hospital, Medical School of Xiamen University, Xiamen, Fujian, China
| | - Gang Li
- Xiamen Cardiovascular Hospital, Medical School of Xiamen University, Xiamen, Fujian, China
| | - Yan-Hui Zhang
- Xiamen Cardiovascular Hospital, Medical School of Xiamen University, Xiamen, Fujian, China
| | - Yong Sun
- Xiamen Cardiovascular Hospital, Medical School of Xiamen University, Xiamen, Fujian, China
| | - Feng Qiu
- Xiamen Cardiovascular Hospital, Medical School of Xiamen University, Xiamen, Fujian, China
| | - Qian Yang
- Xiamen Cardiovascular Hospital, Medical School of Xiamen University, Xiamen, Fujian, China
| | - Guo-Sheng Xiao
- Xiamen Cardiovascular Hospital, Medical School of Xiamen University, Xiamen, Fujian, China
| | - Gui-Rong Li
- Xiamen Cardiovascular Hospital, Medical School of Xiamen University, Xiamen, Fujian, China.
| | - Yan Wang
- Xiamen Cardiovascular Hospital, Medical School of Xiamen University, Xiamen, Fujian, China.
| |
Collapse
|
44
|
Choi D, Park E, Jung E, Cha B, Lee S, Yu J, Kim PM, Lee S, Hong YJ, Koh CJ, Cho CW, Wu Y, Li Jeon N, Wong AK, Shin L, Kumar SR, Bermejo-Moreno I, Srinivasan RS, Cho IT, Hong YK. Piezo1 incorporates mechanical force signals into the genetic program that governs lymphatic valve development and maintenance. JCI Insight 2019; 4:125068. [PMID: 30676326 DOI: 10.1172/jci.insight.125068] [Citation(s) in RCA: 106] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 01/17/2019] [Indexed: 01/05/2023] Open
Abstract
The lymphatic system plays crucial roles in tissue homeostasis, lipid absorption, and immune cell trafficking. Although lymphatic valves ensure unidirectional lymph flows, the flow itself controls lymphatic valve formation. Here, we demonstrate that a mechanically activated ion channel Piezo1 senses oscillating shear stress (OSS) and incorporates the signal into the genetic program controlling lymphatic valve development and maintenance. Time-controlled deletion of Piezo1 using a pan-endothelial Cre driver (Cdh5[PAC]-CreERT2) or lymphatic-specific Cre driver (Prox1-CreERT2) equally inhibited lymphatic valve formation in newborn mice. Furthermore, Piezo1 deletion in adult lymphatics caused substantial lymphatic valve degeneration. Piezo1 knockdown in cultured lymphatic endothelial cells (LECs) largely abrogated the OSS-induced upregulation of the lymphatic valve signature genes. Conversely, ectopic Piezo1 overexpression upregulated the lymphatic valve genes in the absence of OSS. Remarkably, activation of Piezo1 using chemical agonist Yoda1 not only accelerated lymphatic valve formation in animals, but also triggered upregulation of some lymphatic valve genes in cultured LECs without exposure to OSS. In summary, our studies together demonstrate that Piezo1 is the force sensor in the mechanotransduction pathway controlling lymphatic valve development and maintenance, and Piezo1 activation is a potentially novel therapeutic strategy for congenital and surgery-associated lymphedema.
Collapse
Affiliation(s)
- Dongwon Choi
- Department of Surgery, and.,Department of Biochemistry and Molecular Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, UCLA, Los Angeles, California, USA
| | - Eunkyung Park
- Department of Surgery, and.,Department of Biochemistry and Molecular Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, UCLA, Los Angeles, California, USA
| | - Eunson Jung
- Department of Surgery, and.,Department of Biochemistry and Molecular Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, UCLA, Los Angeles, California, USA
| | - Boksik Cha
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Somin Lee
- Department of Mechanical and Aerospace Engineering, Seoul National University, Seoul, South Korea
| | - James Yu
- Department of Mechanical and Aerospace Engineering, Seoul National University, Seoul, South Korea
| | - Paul M Kim
- Department of Surgery, and.,Department of Biochemistry and Molecular Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, UCLA, Los Angeles, California, USA
| | - Sunju Lee
- Department of Surgery, and.,Department of Biochemistry and Molecular Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, UCLA, Los Angeles, California, USA
| | - Yeo Jin Hong
- Department of Surgery, and.,Department of Biochemistry and Molecular Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, UCLA, Los Angeles, California, USA
| | - Chester J Koh
- Division of Pediatric Urology, Texas Children's Hospital, Baylor Collexge of Medicine, Houston, Texas, USA
| | - Chang-Won Cho
- Department of Surgery, and.,Traditional Food Research Group, Korea Food Research Institute, Wanju-gun, Jeollabuk-do, South Korea
| | - Yifan Wu
- Department of Surgery, and.,Department of Biochemistry and Molecular Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, UCLA, Los Angeles, California, USA
| | - Noo Li Jeon
- Department of Mechanical and Aerospace Engineering, Seoul National University, Seoul, South Korea
| | | | | | | | - Ivan Bermejo-Moreno
- Department of Aerospace and Mechanical Engineering, University of Southern California, Los Angeles, California, USA
| | - R Sathish Srinivasan
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | | | - Young-Kwon Hong
- Department of Surgery, and.,Department of Biochemistry and Molecular Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, UCLA, Los Angeles, California, USA
| |
Collapse
|
45
|
Inoue R, Kurahara LH, Hiraishi K. TRP channels in cardiac and intestinal fibrosis. Semin Cell Dev Biol 2018; 94:40-49. [PMID: 30445149 DOI: 10.1016/j.semcdb.2018.11.002] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 11/05/2018] [Accepted: 11/09/2018] [Indexed: 02/06/2023]
Abstract
It is now widely accepted that advanced fibrosis underlies many chronic inflammatory disorders and is the main cause of morbidity and mortality of the modern world. The pathogenic mechanism of advanced fibrosis involves diverse and intricate interplays between numerous extracellular and intracellular signaling molecules, among which the non-trivial roles of a stress-responsive Ca2+/Na+-permeable cation channel superfamily, the transient receptor potential (TRP) protein, are receiving growing attention. Available evidence suggests that several TRP channels such as TRPC3, TRPC6, TRPV1, TRPV3, TRPV4, TRPA1, TRPM6 and TRPM7 may play central roles in the progression and/or prevention of fibroproliferative disorders in vital visceral organs such as lung, heart, liver, kidney, and bowel as well as brain, blood vessels and skin, and may contribute to both acute and chronic inflammatory processes involved therein. This short paper overviews the current knowledge accumulated in this rapidly growing field, with particular focus on cardiac and intestinal fibrosis, which are tightly associated with the pathogenesis of atrial fibrillation and inflammatory bowel diseases such as Crohn's disease.
Collapse
Affiliation(s)
- Ryuji Inoue
- Department of Physiology, Fukuoka University School of medicine, Nanakuma 7-451, Jonan-ku, Fukuoka 814-0180, Japan.
| | - Lin-Hai Kurahara
- Department of Physiology, Fukuoka University School of medicine, Nanakuma 7-451, Jonan-ku, Fukuoka 814-0180, Japan
| | - Keizo Hiraishi
- Department of Physiology, Fukuoka University School of medicine, Nanakuma 7-451, Jonan-ku, Fukuoka 814-0180, Japan
| |
Collapse
|
46
|
Johnson DM, Antoons G. Arrhythmogenic Mechanisms in Heart Failure: Linking β-Adrenergic Stimulation, Stretch, and Calcium. Front Physiol 2018; 9:1453. [PMID: 30374311 PMCID: PMC6196916 DOI: 10.3389/fphys.2018.01453] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 09/25/2018] [Indexed: 12/22/2022] Open
Abstract
Heart failure (HF) is associated with elevated sympathetic tone and mechanical load. Both systems activate signaling transduction pathways that increase cardiac output, but eventually become part of the disease process itself leading to further worsening of cardiac function. These alterations can adversely contribute to electrical instability, at least in part due to the modulation of Ca2+ handling at the level of the single cardiac myocyte. The major aim of this review is to provide a definitive overview of the links and cross talk between β-adrenergic stimulation, mechanical load, and arrhythmogenesis in the setting of HF. We will initially review the role of Ca2+ in the induction of both early and delayed afterdepolarizations, the role that β-adrenergic stimulation plays in the initiation of these and how the propensity for these may be altered in HF. We will then go onto reviewing the current data with regards to the link between mechanical load and afterdepolarizations, the associated mechano-sensitivity of the ryanodine receptor and other stretch activated channels that may be associated with HF-associated arrhythmias. Furthermore, we will discuss how alterations in local Ca2+ microdomains during the remodeling process associated the HF may contribute to the increased disposition for β-adrenergic or stretch induced arrhythmogenic triggers. Finally, the potential mechanisms linking β-adrenergic stimulation and mechanical stretch will be clarified, with the aim of finding common modalities of arrhythmogenesis that could be targeted by novel therapeutic agents in the setting of HF.
Collapse
Affiliation(s)
- Daniel M Johnson
- Department of Cardiothoracic Surgery, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, Netherlands
| | - Gudrun Antoons
- Department of Physiology, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, Netherlands
| |
Collapse
|
47
|
Zhang Q, Qi H, Cao Y, Shi P, Song C, Ba L, Chen Y, Gao J, Li S, Li B, Sun H. Activation of transient receptor potential vanilloid 3 channel (TRPV3) aggravated pathological cardiac hypertrophy via calcineurin/NFATc3 pathway in rats. J Cell Mol Med 2018; 22:6055-6067. [PMID: 30299584 PMCID: PMC6237578 DOI: 10.1111/jcmm.13880] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 07/13/2018] [Indexed: 12/23/2022] Open
Abstract
Cardiac hypertrophy is a compensatory response to mechanical stimuli and neurohormonal factors, ultimately progresses to heart failure. The proteins of some transient receptor potential (TRP) channels, Ca2+‐permeable nonselective cation channel, are highly expressed in cardiomyocytes, and associated with the occurrence of cardiac hypertrophy. Transient receptor potential vanilloid 3 (TRPV3) is a member of TRP, however, the functional role of TRPV3 in cardiac hypertrophy remains unclear. TRPV3 was elevated in pathological cardiac hypertrophy, but not in swimming exercise‐induced physiological cardiac hypertrophy in rats. TRPV3 expression was also increased in Ang II–induced cardiomyocyte hypertrophy in vitro, which was remarkably increased by carvacrol (a nonselective TRPV channel agonist), and reduced by ruthenium red (a nonselective TRPV channel antagonist). Interestingly, we found that activated TRPV3 in Ang II–induced cardiomyocyte hypertrophy was accompanied with increasing intracellular calcium concentration, promoting calcineurin, and phosphorylated CaMKII protein expression, and enhancing NFATc3 nuclear translocation. However, blocking or knockdown of TRPV3 could inhibit the expressions of calcineurin, phosphorylated CaMKII and NFATc3 protein by Western blot. In conclusion, the activation of TRPV3 aggravated pathological cardiac hypertrophy through calcineurin/NFATc3 signalling pathway and correlated with the protein expression levels of calcineurin, phosphorylated CaMKII and NFATc3, revealing that TRPV3 might be a potential therapeutic target for cardiac hypertrophy.
Collapse
Affiliation(s)
- Qianhui Zhang
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, Heilongjiang, China
| | - Hanping Qi
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, Heilongjiang, China
| | - Yonggang Cao
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, Heilongjiang, China
| | - Pilong Shi
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, Heilongjiang, China
| | - Chao Song
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, Heilongjiang, China
| | - Lina Ba
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, Heilongjiang, China
| | - Yunping Chen
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, Heilongjiang, China
| | - Jingquan Gao
- Department of Nursing, Harbin Medical University-Daqing, Daqing, Heilongjiang, China
| | - Shuzhi Li
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, Heilongjiang, China
| | - Baiyan Li
- Department of Pharmacology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Hongli Sun
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, Heilongjiang, China
| |
Collapse
|
48
|
Randhawa PK, Jaggi AS. Exploring the putative role of TRPV 1 -dependent CGRP release in remote hind preconditioning-induced cardioprotection. Cardiovasc Ther 2018; 35. [PMID: 28599085 DOI: 10.1111/1755-5922.12276] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Revised: 05/18/2017] [Accepted: 06/03/2017] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Remote ischemic preconditioning (RIPC) is a phenomenon whereby transient nonlethal ischemia and reperfusion episodes confer protection against prolonged ischemia reperfusion-induced injury. However, the underlying intracellular signaling has not been extensively explored. OBJECTIVE This study aimed to inspect the putative involvement of TRPV1 -dependent CGRP release in mediating remote hind limb preconditioning-induced cardioprotection. METHODS In this study, remote hind limb preconditioning stimulus was delivered (four consecutive episodes of 5 minutes of ischemia reperfusion) using a blood pressure cuff tied at the inguinal level of the rat. The isolated rat hearts were perfused on the Langendorff's system and were subjected to 30-minutes global ischemia and 120-minutes reperfusion. Prolonged ischemia and subsequent reperfusion led to myocardial injury that was evaluated in terms of infarct size, LDH release, CK release, LVDP, +dp/dtmax , -dp/dtmin , and coronary flow rate. The pharmacological agents used in this study included capsaicin as TRPV1 channel activator, sumatriptan and CGRP8-37 as CGRP blockers. RESULTS Remote hind limb and capsaicin preconditioning (10 mg/kg-1 ) significantly reduced the infarct size, LDH release, CK release and significantly improved LVDP, +dp/dtmax , -dp/dtmin , and coronary flow rate. However, remote hind limb and capsaicin preconditioning-induced cardioprotective effects were remarkably reduced in the presence of sumatriptan (8 mg/kg-1 ) and CGRP8-37 (1 mg/kg-1 ). CONCLUSION This indicates that remote hind limb preconditioning stimulus probably activates TRPV1 channels which subsequently induces CGRP release to produce cardioprotective effects.
Collapse
Affiliation(s)
- Puneet Kaur Randhawa
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University Patiala, Patiala, India
| | - Amteshwar Singh Jaggi
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University Patiala, Patiala, India
| |
Collapse
|
49
|
Sáez PJ, Sáez JC, Lennon-Duménil AM, Vargas P. Role of calcium permeable channels in dendritic cell migration. Curr Opin Immunol 2018; 52:74-80. [PMID: 29715579 DOI: 10.1016/j.coi.2018.04.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 04/03/2018] [Accepted: 04/05/2018] [Indexed: 12/22/2022]
Abstract
Calcium ion (Ca2+) is an essential second messenger involved in multiple cellular and subcellular processes. Ca2+ can be released and sensed globally or locally within cells, providing complex signals of variable amplitudes and time-scales. The key function of Ca2+ in the regulation of acto-myosin contractility has provided a simple explanation for its role in the regulation of immune cell migration. However, many questions remain, including the identity of the Ca2+ stores, channels and upstream signals involved in this process. Here, we focus on dendritic cells (DCs), because their immune sentinel function heavily relies on their capacity to migrate within tissues and later on between tissues and lymphoid organs. Deciphering the mechanisms by which cytoplasmic Ca2+ regulate DC migration should shed light on their role in initiating and tuning immune responses.
Collapse
Affiliation(s)
- Pablo J Sáez
- Institut Curie, PSL Research University, CNRS, UMR144, F-75005, France; Instittut Pierre-Gilles de Gennes, PSL Research University, F-75005, France.
| | - Juan C Sáez
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica, Santiago 6513677, Chile; Instituto Milenio, Centro Interdisciplinario de Neurociencias de Valparaíso, Valparaíso 2360103, Chile
| | | | - Pablo Vargas
- Institut Curie, PSL Research University, CNRS, UMR144, F-75005, France; Instittut Pierre-Gilles de Gennes, PSL Research University, F-75005, France.
| |
Collapse
|
50
|
Yamaguchi Y, Iribe G, Kaneko T, Takahashi K, Numaga-Tomita T, Nishida M, Birnbaumer L, Naruse K. TRPC3 participates in angiotensin II type 1 receptor-dependent stress-induced slow increase in intracellular Ca 2+ concentration in mouse cardiomyocytes. J Physiol Sci 2018; 68:153-164. [PMID: 28105583 PMCID: PMC10718017 DOI: 10.1007/s12576-016-0519-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Accepted: 12/26/2016] [Indexed: 11/26/2022]
Abstract
When a cardiac muscle is held in a stretched position, its [Ca2+] transient increases slowly over several minutes in a process known as stress-induced slow increase in intracellular Ca2+ concentration ([Ca2+]i) (SSC). Transient receptor potential canonical (TRPC) 3 forms a non-selective cation channel regulated by the angiotensin II type 1 receptor (AT1R). In this study, we investigated the role of TRPC3 in the SSC. Isolated mouse ventricular myocytes were electrically stimulated and subjected to sustained stretch. An AT1R blocker, a phospholipase C inhibitor, and a TRPC3 inhibitor suppressed the SSC. These inhibitors also abolished the observed SSC-like slow increase in [Ca2+]i induced by angiotensin II, instead of stretch. Furthermore, the SSC was not observed in TRPC3 knockout mice. Simulation and immunohistochemical studies suggest that sarcolemmal TRPC3 is responsible for the SSC. These results indicate that sarcolemmal TRPC3, regulated by AT1R, causes the SSC.
Collapse
Affiliation(s)
- Yohei Yamaguchi
- Department of Cardiovascular Physiology, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, 700-8558, Japan
| | - Gentaro Iribe
- Department of Cardiovascular Physiology, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, 700-8558, Japan.
| | - Toshiyuki Kaneko
- Department of Physiology, Asahikawa Medical University, Asahikawa, Hokkaido, 078-8510, Japan
| | - Ken Takahashi
- Department of Cardiovascular Physiology, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, 700-8558, Japan
| | - Takuro Numaga-Tomita
- Division of Cardiocirculatory Signaling, Okazaki Institute for Integrative Bioscience (National Institute for Physiological Sciences), National Institutes of Natural Sciences, Okazaki, Aichi, 444-8787, Japan
| | - Motohiro Nishida
- Division of Cardiocirculatory Signaling, Okazaki Institute for Integrative Bioscience (National Institute for Physiological Sciences), National Institutes of Natural Sciences, Okazaki, Aichi, 444-8787, Japan
| | - Lutz Birnbaumer
- Neurobiology Laboratory, National Institute of Environmental Health Science, Research Triangle Park, NC, 27709, USA
| | - Keiji Naruse
- Department of Cardiovascular Physiology, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, 700-8558, Japan
| |
Collapse
|