1
|
Taylor JL, Baudel MMA, Nieves-Cintron M, Navedo MF. Vascular Function and Ion Channels in Alzheimer's Disease. Microcirculation 2024; 31:e12881. [PMID: 39190776 PMCID: PMC11498901 DOI: 10.1111/micc.12881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 08/06/2024] [Indexed: 08/29/2024]
Abstract
This review paper explores the critical role of vascular ion channels in the regulation of cerebral artery function and examines the impact of Alzheimer's disease (AD) on these processes. Vascular ion channels are fundamental in controlling vascular tone, blood flow, and endothelial function in cerebral arteries. Dysfunction of these channels can lead to impaired cerebral autoregulation, contributing to cerebrovascular pathologies. AD, characterized by the accumulation of amyloid beta (Aβ) plaques and neurofibrillary tangles, has been increasingly linked to vascular abnormalities, including altered vascular ion channel activity. Here, we briefly review the role of vascular ion channels in cerebral blood flow control and neurovascular coupling. We then examine the vascular defects in AD, the current understanding of how AD pathology affects vascular ion channel function, and how these changes may lead to compromised cerebral blood flow and neurodegenerative processes. Finally, we provide future perspectives and conclusions. Understanding this topic is important as ion channels may be potential therapeutic targets for improving cerebrovascular health and mitigating AD progression.
Collapse
Affiliation(s)
- Jade L. Taylor
- Department of Pharmacology, University of California Davis, Davis CA, 95616, USA
| | | | | | - Manuel F. Navedo
- Department of Pharmacology, University of California Davis, Davis CA, 95616, USA
| |
Collapse
|
2
|
Huang F, Zhang F, Huang L, Zhu X, Huang C, Li N, Da Q, Huang Y, Yang H, Wang H, Zhao L, Lin Q, Chen Z, Xu J, Liu J, Ren M, Wang Y, Han Z, Ouyang K. Inositol 1,4,5-Trisphosphate Receptors Regulate Vascular Smooth Muscle Cell Proliferation and Neointima Formation in Mice. J Am Heart Assoc 2024; 13:e034203. [PMID: 39023067 DOI: 10.1161/jaha.124.034203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 06/24/2024] [Indexed: 07/20/2024]
Abstract
BACKGROUND Vascular smooth muscle cell (VSMC) proliferation is involved in many types of arterial diseases, including neointima hyperplasia, in which Ca2+ has been recognized as a key player. However, the physiological role of Ca2+ release via inositol 1,4,5-trisphosphate receptors (IP3Rs) from endoplasmic reticulum in regulating VSMC proliferation has not been well determined. METHODS AND RESULTS Both in vitro cell culture models and in vivo mouse models were generated to investigate the role of IP3Rs in regulating VSMC proliferation. Expression of all 3 IP3R subtypes was increased in cultured VSMCs upon platelet-derived growth factor-BB and FBS stimulation as well as in the left carotid artery undergoing intimal thickening after vascular occlusion. Genetic ablation of all 3 IP3R subtypes abolished endoplasmic reticulum Ca2+ release in cultured VSMCs, significantly reduced cell proliferation induced by platelet-derived growth factor-BB and FBS stimulation, and also decreased cell migration of VSMCs. Furthermore, smooth muscle-specific deletion of all IP3R subtypes in adult mice dramatically attenuated neointima formation induced by left carotid artery ligation, accompanied by significant decreases in cell proliferation and matrix metalloproteinase-9 expression in injured vessels. Mechanistically, IP3R-mediated Ca2+ release may activate cAMP response element-binding protein, a key player in controlling VSMC proliferation, via Ca2+/calmodulin-dependent protein kinase II and Akt. Loss of IP3Rs suppressed cAMP response element-binding protein phosphorylation at Ser133 in both cultured VSMCs and injured vessels, whereas application of Ca2+ permeable ionophore, ionomycin, can reverse cAMP response element-binding protein phosphorylation in IP3R triple knockout VSMCs. CONCLUSIONS Our results demonstrated an essential role of IP3R-mediated Ca2+ release from endoplasmic reticulum in regulating cAMP response element-binding protein activation, VSMC proliferation, and neointima formation in mouse arteries.
Collapse
MESH Headings
- Animals
- Male
- Mice
- Becaplermin/pharmacology
- Becaplermin/metabolism
- Calcium/metabolism
- Calcium Signaling
- Carotid Artery Injuries/pathology
- Carotid Artery Injuries/metabolism
- Carotid Artery Injuries/genetics
- Cell Movement
- Cell Proliferation
- Cells, Cultured
- Cyclic AMP Response Element-Binding Protein/metabolism
- Cyclic AMP Response Element-Binding Protein/genetics
- Disease Models, Animal
- Endoplasmic Reticulum/metabolism
- Endoplasmic Reticulum/pathology
- Inositol 1,4,5-Trisphosphate Receptors/metabolism
- Inositol 1,4,5-Trisphosphate Receptors/genetics
- Mice, Inbred C57BL
- Mice, Knockout
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Neointima/pathology
- Phosphorylation
- Proto-Oncogene Proteins c-akt/metabolism
Collapse
Affiliation(s)
- Fang Huang
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School Peking University Shenzhen China
| | - Fei Zhang
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School Peking University Shenzhen China
| | - Lei Huang
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School Peking University Shenzhen China
| | - Xiangbin Zhu
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School Peking University Shenzhen China
| | - Can Huang
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School Peking University Shenzhen China
| | - Na Li
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School Peking University Shenzhen China
| | - Qingen Da
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School Peking University Shenzhen China
| | - Yu Huang
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School Peking University Shenzhen China
| | - Huihua Yang
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School Peking University Shenzhen China
| | - Hong Wang
- Central Laboratory Peking University Shenzhen Hospital Shenzhen China
| | - Lingyun Zhao
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School Peking University Shenzhen China
| | - Qingsong Lin
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School Peking University Shenzhen China
| | - Zee Chen
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School Peking University Shenzhen China
| | - Junjie Xu
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School Peking University Shenzhen China
| | - Jie Liu
- Department of Pathophysiology, School of Medicine Shenzhen University Shenzhen China
| | - Mingming Ren
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School Peking University Shenzhen China
| | - Yan Wang
- Department of Cardiology Qingdao Municipal Hospital Qingdao China
| | - Zhen Han
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School Peking University Shenzhen China
| | - Kunfu Ouyang
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School Peking University Shenzhen China
| |
Collapse
|
3
|
Arreola J. WNK kinase, ion channels and arachidonic acid metabolites choreographically execute endothelium-dependent vasodilation. Cell Calcium 2024; 121:102904. [PMID: 38728790 DOI: 10.1016/j.ceca.2024.102904] [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: 05/02/2024] [Accepted: 05/04/2024] [Indexed: 05/12/2024]
Abstract
The smooth muscle-walled blood vessels control blood pressure. The vessel lumen is lined by an endothelial cell (ECs) layer, interconnected to the surrounding smooth muscle cells (SMCs) by myoendothelial gap junctions. Gap junctions also maintain homo-cellular ECs-ECs and SMCs-SMCs connections. This gap junction network nearly equalises both cells' membrane potential and cytosolic ionic composition, whether in resting or stimulated conditions. When acetylcholine (ACh) activates ECs M3 receptors, a complex signalling cascade involving second messengers and ion channels is triggered to induce vasodilation.
Collapse
Affiliation(s)
- Jorge Arreola
- Physics Institute, Universidad Autónoma de San Luis Potosí, Av. Parque Chapultepec 1570, Privadas del Pedregal, 78295 San Luis Potosí, S.L.P, México.
| |
Collapse
|
4
|
Peixoto-Neves D, Jaggar JH. Physiological functions and pathological involvement of ion channel trafficking in the vasculature. J Physiol 2024; 602:3275-3296. [PMID: 37818949 PMCID: PMC11006830 DOI: 10.1113/jp285007] [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: 08/09/2023] [Accepted: 09/28/2023] [Indexed: 10/13/2023] Open
Abstract
A variety of ion channels regulate membrane potential and calcium influx in arterial smooth muscle and endothelial cells to modify vascular functions, including contractility. The current (I) generated by a population of ion channels is equally dependent upon their number (N), open probability (Po) and single channel current (i), such that I = N.PO.i. A conventional view had been that ion channels traffic to the plasma membrane in a passive manner, resulting in a static surface population. It was also considered that channels assemble with auxiliary subunits prior to anterograde trafficking of the multimeric complex to the plasma membrane. Recent studies have demonstrated that physiological stimuli can regulate the surface abundance (N) of several different ion channels in arterial smooth muscle and endothelial cells to control arterial contractility. Physiological stimuli can also regulate the number of auxiliary subunits present in the plasma membrane to modify the biophysical properties, regulatory mechanisms and physiological functions of some ion channels. Furthermore, ion channel trafficking becomes dysfunctional in the vasculature during hypertension, which negatively impacts the regulation of contractility. The temporal kinetics of ion channel and auxiliary subunit trafficking can also vary depending on the signalling mechanisms and proteins involved. This review will summarize recent work that has uncovered the mechanisms, functions and pathological modifications of ion channel trafficking in arterial smooth muscle and endothelial cells.
Collapse
Affiliation(s)
| | - Jonathan H. Jaggar
- Department of Physiology, University of Tennessee Health Science Center, Memphis TN 38139
| |
Collapse
|
5
|
Schulz ME, Akerstrom VL, Song K, Broyhill SE, Li M, Lambert MD, Goldberg TB, Kataru RP, Shin J, Braun SE, Norton CE, Czepielewski RS, Mehrara BJ, Domeier TL, Zawieja SD, Castorena-Gonzalez JA. TRPV4-Expressing Tissue-Resident Macrophages Regulate the Function of Collecting Lymphatic Vessels via Thromboxane A2 Receptors in Lymphatic Muscle Cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.21.595189. [PMID: 38826322 PMCID: PMC11142127 DOI: 10.1101/2024.05.21.595189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Rationale TRPV4 channels are critical regulators of blood vascular function and have been shown to be dysregulated in many disease conditions in association with inflammation and tissue fibrosis. These are key features in the pathophysiology of lymphatic system diseases, including lymphedema and lipedema; however, the role of TRPV4 channels in the lymphatic system remains largely unexplored. TRPV4 channels are calcium permeable, non-selective cation channels that are activated by diverse stimuli, including shear stress, stretch, temperature, and cell metabolites, which may regulate lymphatic contractile function. Objective To characterize the expression of TRPV4 channels in collecting lymphatic vessels and to determine the extent to which these channels regulate the contractile function of lymphatics. Methods and Results Pressure myography on intact, isolated, and cannulated lymphatic vessels showed that pharmacological activation of TRPV4 channels with GSK1016790A (GSK101) led to contractile dysregulation. The response to GSK101 was multiphasic and included, 1) initial robust constriction that was sustained for ≥1 minute and in some instances remained for ≥4 minutes; and 2) subsequent vasodilation and partial or complete inhibition of lymphatic contractions associated with release of nitric oxide. The functional response to activation of TRPV4 channels displayed differences across lymphatics from four anatomical regions, but these differences were consistent across different species (mouse, rat, and non-human primate). Importantly, similar responses were observed following activation of TRPV4 channels in arterioles. The initial and sustained constriction was prevented with the COX inhibitor, indomethacin. We generated a controlled and spatially defined single-cell RNA sequencing (scRNAseq) dataset from intact and microdissected collecting lymphatic vessels. Our data uncovered a subset of macrophages displaying the highest expression of Trpv4 compared to other cell types within and surrounding the lymphatic vessel wall. These macrophages displayed a transcriptomic profile consistent with that of tissue-resident macrophages (TRMs), including differential expression of Lyve1 , Cd163 , Folr2 , Mrc1 , Ccl8 , Apoe , Cd209f , Cd209d , and Cd209g ; and at least half of these macrophages also expressed Timd4. This subset of macrophages also highly expressed Txa2s , which encodes the thromboxane A2 (TXA2) synthase. Inhibition of TXA2 receptors (TXA2Rs) prevented TRPV4-mediated contractile dysregulation. TXA2R activation on LMCs caused an increase in mobilization of calcium from intracellular stores through Ip3 receptors which promoted store operated calcium entry and vasoconstriction. Conclusions Clinical studies have linked cancer-related lymphedema with an increased infiltration of macrophages. While these macrophages have known anti-inflammatory and pro-lymphangiogenic roles, as well as promote tissue repair, our results point to detrimental effects to the pumping capacity of collecting lymphatic vessels mediated by activation of TRPV4 channels in macrophages. Pharmacological targeting of TRPV4 channels in LYVE1-expressing macrophages or pharmacological targeting of TXA2Rs may offer novel therapeutic strategies to improve lymphatic pumping function and lymph transport in lymphedema.
Collapse
|
6
|
Ta HQ, Kuppusamy M, Sonkusare SK, Roeser ME, Laubach VE. The endothelium: gatekeeper to lung ischemia-reperfusion injury. Respir Res 2024; 25:172. [PMID: 38637760 PMCID: PMC11027545 DOI: 10.1186/s12931-024-02776-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 03/14/2024] [Indexed: 04/20/2024] Open
Abstract
The success of lung transplantation is limited by the high rate of primary graft dysfunction due to ischemia-reperfusion injury (IRI). Lung IRI is characterized by a robust inflammatory response, lung dysfunction, endothelial barrier disruption, oxidative stress, vascular permeability, edema, and neutrophil infiltration. These events are dependent on the health of the endothelium, which is a primary target of IRI that results in pulmonary endothelial barrier dysfunction. Over the past 10 years, research has focused more on the endothelium, which is beginning to unravel the multi-factorial pathogenesis and immunologic mechanisms underlying IRI. Many important proteins, receptors, and signaling pathways that are involved in the pathogenesis of endothelial dysfunction after IR are starting to be identified and targeted as prospective therapies for lung IRI. In this review, we highlight the more significant mediators of IRI-induced endothelial dysfunction discovered over the past decade including the extracellular glycocalyx, endothelial ion channels, purinergic receptors, kinases, and integrins. While there are no definitive clinical therapies currently available to prevent lung IRI, we will discuss potential clinical strategies for targeting the endothelium for the treatment or prevention of IRI. The accruing evidence on the essential role the endothelium plays in lung IRI suggests that promising endothelial-directed treatments may be approaching the clinic soon. The application of therapies targeting the pulmonary endothelium may help to halt this rapid and potentially fatal injury.
Collapse
Affiliation(s)
- Huy Q Ta
- Department of Surgery, University of Virginia, P. O. Box 801359, Charlottesville, VA, 22908, USA
| | - Maniselvan Kuppusamy
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, 22908, USA
| | - Swapnil K Sonkusare
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, 22908, USA
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA, 22908, USA
| | - Mark E Roeser
- Department of Surgery, University of Virginia, P. O. Box 801359, Charlottesville, VA, 22908, USA
| | - Victor E Laubach
- Department of Surgery, University of Virginia, P. O. Box 801359, Charlottesville, VA, 22908, USA.
| |
Collapse
|
7
|
Liu W, Zhang W, Wang C, Song J, Li K, Zhang X, Wu X, Guo H. TRPV4 antagonist suppresses retinal ganglion cell apoptosis by regulating the activation of CaMKII and TNF-α expression in a chronic ocular hypertension rat model. Int Immunopharmacol 2024; 130:111811. [PMID: 38457929 DOI: 10.1016/j.intimp.2024.111811] [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: 01/13/2024] [Revised: 02/24/2024] [Accepted: 03/04/2024] [Indexed: 03/10/2024]
Abstract
Glaucoma is characterized by a progressive loss of retinal ganglion cells (RGCs), leading to irreversible visual function impairment. Sustained increase in intraocular pressure represents a major risk factor for glaucoma, yet the underlying mechanisms of RGC apoptosis induced by intraocular pressure remains unclear. This study aims to investigate the role of TRPV4 in RGC apoptosis in a rat model of chronic ocular hypertension (COH) and the underlying molecular mechanism. In the COH rat models, we evaluated the visual function, retinal pathological changes and RGC apoptosis. TRPV4 expression and downstream signaling molecules were also detected. We found that RGC density decreased and RGC apoptosis was induced in COH eyes compared with control eyes. TRPV4 expression increased significantly in response to elevated IOP. TRPV4 inhibition by the TRPV4 antagonist HC-067047 (HC-067) suppressed RGC apoptosis and protected visual function. HC-067 treatment upregulated the phosphorylation of CaMKII in both control and COH eyes. Finally, HC-067 treatment suppressed the production of TNF-α induced by ocular hypertension. The TRPV4 antagonist HC-067 might suppress RGC apoptosis by regulating the activation of CaMKII and inhibiting the production of TNF-α in the COH model. This indicated that TRPV4 antagonists may be a potential and novel therapeutic strategy for glaucoma.
Collapse
Affiliation(s)
- Wenhui Liu
- Department of Ophthalmology, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Wenzhe Zhang
- Department of Ophthalmology, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Chen Wang
- Department of Ophthalmology, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Jiarun Song
- Department of Ophthalmology, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Kaiyue Li
- Department of Nuclear Medicine, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Xia Zhang
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Xinyi Wu
- Department of Ophthalmology, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China.
| | - Hui Guo
- Department of Ophthalmology, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China.
| |
Collapse
|
8
|
Asulin M, Gorodetzer N, Fridman R, Shelly Ben-Shushan R, Cohen Z, Beyer AM, Chuyun D, Gutterman DD, Szuchman-Sapir A. 5,6-diHETE lactone (EPA-L) mediates hypertensive microvascular dilation by activating the endothelial GPR-PLC-IP 3 signaling pathway. Biochem Biophys Res Commun 2024; 700:149585. [PMID: 38290177 DOI: 10.1016/j.bbrc.2024.149585] [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: 12/20/2023] [Revised: 01/24/2024] [Accepted: 01/25/2024] [Indexed: 02/01/2024]
Abstract
Endothelial microvascular dysfunction affects multi-organ pathologic processes that contribute to increased vascular tone and is at the base of impaired metabolic and cardiovascular diseases. The vascular dilation impaired by nitric oxide (NO) deficiency in such dysfunctional endothelium is often balanced by endothelial-derived hyperpolarizing factors (EDHFs), which play a critical role in managing vascular tone. Our latest research has uncovered a new group of lactone oxylipins produced in the polyunsaturated fatty acids (PUFAs) CYP450 epoxygenase pathway, significantly affecting vascular dilation. The lactone oxylipin, derived from arachidonic acid (5,6-diHET lactone, AA-L), has been previously shown to facilitate vasodilation dependent on the endothelium in isolated human microvessels. The administration of the lactone oxylipin derived from eicosapentaenoic acid (5,6-diHETE lactone, EPA-L) to hypertensive rats demonstrated a significant decrease in blood pressure and improvement in the relaxation of microvessels. However, the molecular signaling processes that underlie these observations were not fully understood. The current study delineates the molecular pathways through which EPA-L promotes endothelium-dependent vascular dilation. In microvessels from hypertensive individuals, it was found that EPA-L mediates endothelium-dependent vasodilation while the signaling pathway was not dependent on NO. In vitro studies on human endothelial cells showed that the hyperpolarization mediated by EPA-L relies on G-protein-coupled receptor (GPR)-phospholipase C (PLC)-IP3 signaling that further activates calcium-dependent potassium flux. The pathway was confirmed using a range of inhibitors and cells overexpressing GPR40, where a specific antagonist reduced the calcium levels and outward currents induced by EPA-L. The downstream AKT and endothelial NO synthase (eNOS) phosphorylations were non-significant. These findings show that the GPR-PLC-IP3 pathway is a key mediator in the EPA-L-triggered vasodilation of arterioles. Therefore, EPA-L is identified as a significant lactone-based PUFA metabolite that contributes to endothelial and vascular health.
Collapse
Affiliation(s)
- Meitar Asulin
- Laboratory of Vascular Signaling, MIGAL-Galilee Research Institute, Kiryat Shmona, Israel; Tel-Hai College, Upper Galilee, Israel
| | - Nadav Gorodetzer
- Laboratory of Vascular Signaling, MIGAL-Galilee Research Institute, Kiryat Shmona, Israel; Tel-Hai College, Upper Galilee, Israel
| | - Rotem Fridman
- Laboratory of Vascular Signaling, MIGAL-Galilee Research Institute, Kiryat Shmona, Israel
| | | | - Zohar Cohen
- Laboratory of Vascular Signaling, MIGAL-Galilee Research Institute, Kiryat Shmona, Israel; Tel-Hai College, Upper Galilee, Israel
| | - Andreas M Beyer
- Department of Medicine and Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, USA; Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | | | - David D Gutterman
- Department of Medicine and Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Andrea Szuchman-Sapir
- Laboratory of Vascular Signaling, MIGAL-Galilee Research Institute, Kiryat Shmona, Israel; Tel-Hai College, Upper Galilee, Israel.
| |
Collapse
|
9
|
Rios FJ, Sarafian RD, Camargo LL, Montezano AC, Touyz RM. Recent Advances in Understanding the Mechanistic Role of Transient Receptor Potential Ion Channels in Patients With Hypertension. Can J Cardiol 2023; 39:1859-1873. [PMID: 37865227 DOI: 10.1016/j.cjca.2023.10.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 10/17/2023] [Accepted: 10/17/2023] [Indexed: 10/23/2023] Open
Abstract
The transient receptor potential (TRP) channel superfamily is a group of nonselective cation channels that function as cellular sensors for a wide range of physical, chemical, and environmental stimuli. According to sequence homology, TRP channels are categorized into 6 subfamilies: TRP canonical, TRP vanilloid, TRP melastatin, TRP ankyrin, TRP mucolipin, and TRP polycystin. They are widely expressed in different cell types and tissues and have essential roles in various physiological and pathological processes by regulating the concentration of ions (Ca2+, Mg2+, Na+, and K+) and influencing intracellular signalling pathways. Human data and experimental models indicate the importance of TRP channels in vascular homeostasis and hypertension. Furthermore, TRP channels have emerged as key players in oxidative stress and inflammation, important in the pathophysiology of cardiovascular diseases, including hypertension. In this review, we present an overview of the TRP channels with a focus on their role in hypertension. In particular, we highlight mechanisms activated by TRP channels in vascular smooth muscle and endothelial cells and discuss their contribution to processes underlying vascular dysfunction in hypertension.
Collapse
Affiliation(s)
- Francisco J Rios
- Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada.
| | - Raquel D Sarafian
- Institute of Biosciences, Department of Genetics and Evolutionary Biology, University of Sao Paulo, Sao Paulo, Brazil
| | - Livia L Camargo
- Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Augusto C Montezano
- Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Rhian M Touyz
- Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada; Department of Medicine, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
10
|
Matsumoto T, Taguchi K, Kobayashi T. Role of TRPV4 on vascular tone regulation in pathophysiological states. Eur J Pharmacol 2023; 959:176104. [PMID: 37802278 DOI: 10.1016/j.ejphar.2023.176104] [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: 08/18/2023] [Revised: 09/20/2023] [Accepted: 10/04/2023] [Indexed: 10/08/2023]
Abstract
Vascular tone regulation is a key event in controlling blood flow in the body. Endothelial cells (ECs) and vascular smooth muscle cells (VSMCs) help regulate the vascular tone. Abnormal vascular responsiveness to various stimuli, including constrictors and dilators, has been observed in pathophysiological states although EC and VSMC coordinate to maintain the exquisite balance between contraction and relaxation in vasculatures. Thus, investigating the mechanisms underlying vascular tone abnormality is very important in maintaining vascular health and treating vasculopathy. Increased intracellular free Ca2+ concentration ([Ca2+]i) is one of the major triggers initiating each EC and VSMC response. Transient receptor potential vanilloid family member 4 (TRPV4) is a Ca2+-permeable non-selective ion channel, which is activated by several stimuli, and is presented in both ECs and VSMCs. Therefore, TRPV4 plays an important role in vascular responses. Emerging evidence indicates the role of TRPV4 on the functions of ECs and VSMCs in various pathophysiological states, including hypertension, diabetes, and obesity. This review focused on the link between TRPV4 and the functions of ECs/VSMCs, particularly its role in vascular tone and responsiveness to vasoactive substances.
Collapse
Affiliation(s)
- Takayuki Matsumoto
- Department of Pharmaceutical Education and Research, Pharmaceutical Education and Research Center, Hoshi University, Shinagawa-ku, Tokyo, 142-8501, Japan.
| | - Kumiko Taguchi
- Department of Physiology and Morphology, Institute of Medicinal Chemistry, Hoshi University, Shinagawa-ku, Tokyo, 142-8501, Japan
| | - Tsuneo Kobayashi
- Department of Physiology and Morphology, Institute of Medicinal Chemistry, Hoshi University, Shinagawa-ku, Tokyo, 142-8501, Japan
| |
Collapse
|
11
|
Behringer EJ. Impact of aging on vascular ion channels: perspectives and knowledge gaps across major organ systems. Am J Physiol Heart Circ Physiol 2023; 325:H1012-H1038. [PMID: 37624095 PMCID: PMC10908410 DOI: 10.1152/ajpheart.00288.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 08/21/2023] [Accepted: 08/21/2023] [Indexed: 08/26/2023]
Abstract
Individuals aged ≥65 yr will comprise ∼20% of the global population by 2030. Cardiovascular disease remains the leading cause of death in the world with age-related endothelial "dysfunction" as a key risk factor. As an organ in and of itself, vascular endothelium courses throughout the mammalian body to coordinate blood flow to all other organs and tissues (e.g., brain, heart, lung, skeletal muscle, gut, kidney, skin) in accord with metabolic demand. In turn, emerging evidence demonstrates that vascular aging and its comorbidities (e.g., neurodegeneration, diabetes, hypertension, kidney disease, heart failure, and cancer) are "channelopathies" in large part. With an emphasis on distinct functional traits and common arrangements across major organs systems, the present literature review encompasses regulation of vascular ion channels that underlie blood flow control throughout the body. The regulation of myoendothelial coupling and local versus conducted signaling are discussed with new perspectives for aging and the development of chronic diseases. Although equipped with an awareness of knowledge gaps in the vascular aging field, a section has been included to encompass general feasibility, role of biological sex, and additional conceptual and experimental considerations (e.g., cell regression and proliferation, gene profile analyses). The ultimate goal is for the reader to see and understand major points of deterioration in vascular function while gaining the ability to think of potential mechanistic and therapeutic strategies to sustain organ perfusion and whole body health with aging.
Collapse
Affiliation(s)
- Erik J Behringer
- Basic Sciences, Loma Linda University, Loma Linda, California, United States
| |
Collapse
|
12
|
Shemarova I. The Dysfunction of Ca 2+ Channels in Hereditary and Chronic Human Heart Diseases and Experimental Animal Models. Int J Mol Sci 2023; 24:15682. [PMID: 37958665 PMCID: PMC10650855 DOI: 10.3390/ijms242115682] [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: 09/11/2023] [Revised: 10/20/2023] [Accepted: 10/23/2023] [Indexed: 11/15/2023] Open
Abstract
Chronic heart diseases, such as coronary heart disease, heart failure, secondary arterial hypertension, and dilated and hypertrophic cardiomyopathies, are widespread and have a fairly high incidence of mortality and disability. Most of these diseases are characterized by cardiac arrhythmias, conduction, and contractility disorders. Additionally, interruption of the electrical activity of the heart, the appearance of extensive ectopic foci, and heart failure are all symptoms of a number of severe hereditary diseases. The molecular mechanisms leading to the development of heart diseases are associated with impaired permeability and excitability of cell membranes and are mainly caused by the dysfunction of cardiac Ca2+ channels. Over the past 50 years, more than 100 varieties of ion channels have been found in the cardiovascular cells. The relationship between the activity of these channels and cardiac pathology, as well as the general cellular biological function, has been intensively studied on several cell types and experimental animal models in vivo and in situ. In this review, I discuss the origin of genetic Ca2+ channelopathies of L- and T-type voltage-gated calcium channels in humans and the role of the non-genetic dysfunctions of Ca2+ channels of various types: L-, R-, and T-type voltage-gated calcium channels, RyR2, including Ca2+ permeable nonselective cation hyperpolarization-activated cyclic nucleotide-gated (HCN), and transient receptor potential (TRP) channels, in the development of cardiac pathology in humans, as well as various aspects of promising experimental studies of the dysfunctions of these channels performed on animal models or in vitro.
Collapse
Affiliation(s)
- Irina Shemarova
- I.M. Sechenov Institute of Evolutionary Physiology and Biochemistry of the Russian Academy of Sciences, 194223 Saint-Petersburg, Russia
| |
Collapse
|
13
|
Wen X, Peng Y, Peng Y, Zhu Y, Yu F, Geng L, Zhou T, Wang X, Feng L, Meng Q. Aortic smooth muscle TRPV4 channels regulate vasoconstriction in high salt-induced hypertension. Hypertens Res 2023; 46:2356-2367. [PMID: 37532951 DOI: 10.1038/s41440-023-01363-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 06/21/2023] [Accepted: 06/23/2023] [Indexed: 08/04/2023]
Abstract
Recent studies have focused on the contribution of vascular endothelial transient receptor potential vanilloid 4 (TRPV4) channels to hypertension. However, in hypertension, TRPV4 channels in vascular smooth muscle remain unexplored. In the present study, we performed wire myograph experiments in isolated aortas from endothelial cell specific TRPV4 channel knockout (TRPV4EC-/-) mice to demonstrate that GSK1016790A (a specific TRPV4 channel agonist) triggered aortic smooth muscle-dependent contractions from mice on a normal-salt diet, and the contractions were enhanced in high-salt diet (HSD) mice. Intracellular Ca2+ concentration ([Ca2+]i) and Ca2+ imaging assays showed that TRPV4-induced [Ca2+]i was significantly higher in aortic smooth muscle cells (ASMCs) from HSD-induced hypertensive mice, and application of an inositol trisphosphate receptor (IP3R) inhibitor markedly attenuated TRPV4-induced [Ca2+]i. IP3R2 expression was enhanced in ASMCs from HSD-induced hypertensive mice and the contractile response induced by TRPV4 was inhibited by the IP3R inhibitor. Whole-transcriptome analysis by RNA-seq and western blot assays revealed the involvement of interferon regulatory factor 7 (IRF7) in TRPV4-IRF7-IP3R2 signaling in HSD-induced hypertension. These results suggested that TRPV4 channels regulate smooth muscle-dependent contractions in high salt-induced hypertension, and this contraction involves increased [Ca2+]i, IP3R2, and IRF7 activity. Our study revealed a considerable effect of TRPV4 channels in smooth muscle-dependent contraction in mice during high-salt induced hypertension.
Collapse
Affiliation(s)
- Xin Wen
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214000, China
| | - Yidi Peng
- School of Pharmaceutical Sciences, Jiangnan University, Wuxi, 214000, China
| | - Yuefeng Peng
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214000, China
| | - Yuzhong Zhu
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214000, China
| | - Fan Yu
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214000, China
| | - Li Geng
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214000, China
| | - Tingting Zhou
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214000, China
| | - Xianfeng Wang
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214000, China
| | - Lei Feng
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214000, China
| | - Qingyou Meng
- Department of Vascular Surgery, General Surgery Clinical Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
14
|
Arrowsmith S. Multiple pregnancies, the myometrium and the role of mechanical factors in the timing of labour. Curr Res Physiol 2023; 6:100105. [PMID: 38107788 PMCID: PMC10724211 DOI: 10.1016/j.crphys.2023.100105] [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: 05/08/2023] [Revised: 07/17/2023] [Accepted: 08/23/2023] [Indexed: 12/19/2023] Open
Abstract
Multiple pregnancy remains a relatively common occurrence, but it is associated with increased risks of adverse outcomes for the mother and her babies and presents unique challenges to healthcare providers. This review will briefly discuss multiple pregnancies, their aetiology and their problems, including preterm birth, before reviewing the processes leading to normal labour onset and how they may be different in a multiple pregnancy. The mechanisms by which mechanical factors i.e., uterine distension or 'stretch' contribute to uterine excitability and the timing of labour onset will be the major focus, and how over distention may pre-dispose multiple pregnancies to preterm birth. This includes current thinking around the role of mechano (stretch) sensitive ion channels in the myometrium and changes to other important regulators of excitability and contraction which have been identified from studies using in vitro and in vivo models of uterine stretch. Physiological stimuli arising from the fetus(es) and placenta(s) will also be discussed. In reviewing what we know about the myometrium in multiple pregnancy in humans, the focus will be on twin pregnancy as it is the most common type of multiple pregnancy and has been the most studied.
Collapse
Affiliation(s)
- Sarah Arrowsmith
- Department of Life Sciences, Manchester Metropolitan University, John Dalton Building, Chester Street, Manchester, M1 5GD, UK
| |
Collapse
|
15
|
Chaigne S, Barbeau S, Ducret T, Guinamard R, Benoist D. Pathophysiological Roles of the TRPV4 Channel in the Heart. Cells 2023; 12:1654. [PMID: 37371124 DOI: 10.3390/cells12121654] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 06/10/2023] [Accepted: 06/14/2023] [Indexed: 06/29/2023] Open
Abstract
The transient receptor potential vanilloid 4 (TRPV4) channel is a non-selective cation channel that is mostly permeable to calcium (Ca2+), which participates in intracellular Ca2+ handling in cardiac cells. It is widely expressed through the body and is activated by a large spectrum of physicochemical stimuli, conferring it a role in a variety of sensorial and physiological functions. Within the cardiovascular system, TRPV4 expression is reported in cardiomyocytes, endothelial cells (ECs) and smooth muscle cells (SMCs), where it modulates mitochondrial activity, Ca2+ homeostasis, cardiomyocytes electrical activity and contractility, cardiac embryonic development and fibroblast proliferation, as well as vascular permeability, dilatation and constriction. On the other hand, TRPV4 channels participate in several cardiac pathological processes such as the development of cardiac fibrosis, hypertrophy, ischemia-reperfusion injuries, heart failure, myocardial infarction and arrhythmia. In this manuscript, we provide an overview of TRPV4 channel implications in cardiac physiology and discuss the potential of the TRPV4 channel as a therapeutic target against cardiovascular diseases.
Collapse
Affiliation(s)
- Sébastien Chaigne
- IHU LIRYC Electrophysiology and Heart Modeling Institute, Fondation Bordeaux Université, 33600 Bordeaux, France
- Centre de Recherche Cardio-Thoracique de Bordeaux, INSERM U1045, University of Bordeaux, 33600 Pessac, France
- Electrophysiology and Ablation Unit, Bordeaux University Hospital, 33604 Pessac, France
| | - Solène Barbeau
- Centre de Recherche Cardio-Thoracique de Bordeaux, INSERM U1045, University of Bordeaux, 33600 Pessac, France
| | - Thomas Ducret
- Centre de Recherche Cardio-Thoracique de Bordeaux, INSERM U1045, University of Bordeaux, 33600 Pessac, France
| | - Romain Guinamard
- UR4650, Physiopathologie et Stratégies d'Imagerie du Remodelage Cardiovasculaire, GIP Cyceron, Université de Caen Normandie, 14032 Caen, France
| | - David Benoist
- IHU LIRYC Electrophysiology and Heart Modeling Institute, Fondation Bordeaux Université, 33600 Bordeaux, France
- Centre de Recherche Cardio-Thoracique de Bordeaux, INSERM U1045, University of Bordeaux, 33600 Pessac, France
| |
Collapse
|
16
|
Kumar M, Zaman MK, Das S, Goyary D, Pathak MP, Chattopadhyay P. Transient Receptor Potential Vanilloid (TRPV4) channel inhibition: A novel promising approach for the treatment of lung diseases. Biomed Pharmacother 2023; 163:114861. [PMID: 37178575 DOI: 10.1016/j.biopha.2023.114861] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 05/03/2023] [Accepted: 05/06/2023] [Indexed: 05/15/2023] Open
Abstract
Research on transient receptor potential vanilloid-4 (TRPV4) can provide a promising potential therapeutic target in the development of novel medicines for lung disorders. TRPV4 expresses in lung tissue and plays an important role in the maintenance of respiratory homeostatic function. TRPV4 is upregulated in life-threatening respiratory diseases like pulmonary hypertension, asthma, cystic fibrosis, and chronic obstructive pulmonary diseases. TRPV4 is linked to several proteins that have physiological functions and are sensitive to a wide variety of stimuli, such as mechanical stimulation, changes in temperature, and hypotonicity, and responds to a variety of proteins and lipid mediators, including anandamide (AA), the arachidonic acid metabolite, 5,6-epoxyeicosatrienoic acid (5,6-EET), a plant dimeric diterpenoid called bisandrographolide A (BAA), and the phorbol ester 4-alpha-phorbol-12,13-didecanoate (4α-PDD). This study focused on relevant research evidence of TRPV4 in lung disorders and its agonist and antagonist effects. TRPV4 can be a possible target of discovered molecules that exerts high therapeutic potential in the treatment of respiratory diseases by inhibiting TRPV4.
Collapse
Affiliation(s)
- Mohit Kumar
- Division of Pharmaceutical Technology, Defence Research Laboratory, Tezpur, Assam 784001, India; Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, Assam 786004, India
| | - Md Kamaruz Zaman
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, Assam 786004, India
| | - Sanghita Das
- Division of Pharmaceutical Technology, Defence Research Laboratory, Tezpur, Assam 784001, India; Pharmaceutical & Fine Chemical Division, Department of Chemical Technology, University of Calcutta, Kolkata, West Bengal 700073, India
| | - Danswrang Goyary
- Division of Pharmaceutical Technology, Defence Research Laboratory, Tezpur, Assam 784001, India
| | - Manash Pratim Pathak
- Faculty of Pharmaceutical Science, Assam down town University, Guwahati, Assam 781026, India.
| | - Pronobesh Chattopadhyay
- Division of Pharmaceutical Technology, Defence Research Laboratory, Tezpur, Assam 784001, India.
| |
Collapse
|
17
|
Dryn DO, Melnyk MI, Melanaphy D, Kizub IV, Johnson CD, Zholos AV. Bidirectional TRP/L Type Ca 2+ Channel/RyR/BK Ca Molecular and Functional Signaloplex in Vascular Smooth Muscles. Biomolecules 2023; 13:biom13050759. [PMID: 37238629 DOI: 10.3390/biom13050759] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 04/24/2023] [Accepted: 04/25/2023] [Indexed: 05/28/2023] Open
Abstract
TRP channels are expressed both in vascular myocytes and endothelial cells, but knowledge of their operational mechanisms in vascular tissue is particularly limited. Here, we show for the first time the biphasic contractile reaction with relaxation followed by a contraction in response to TRPV4 agonist, GSK1016790A, in a rat pulmonary artery preconstricted with phenylephrine. Similar responses were observed both with and without endothelium, and these were abolished by the TRPV4 selective blocker, HC067047, confirming the specific role of TRPV4 in vascular myocytes. Using selective blockers of BKCa and L-type voltage-gated Ca2+ channels (CaL), we found that the relaxation phase was inducted by BKCa activation generating STOCs, while subsequent slowly developing TRPV4-mediated depolarisation activated CaL, producing the second contraction phase. These results are compared to TRPM8 activation using menthol in rat tail artery. Activation of both types of TRP channels produces highly similar changes in membrane potential, namely slow depolarisation with concurrent brief hyperpolarisations due to STOCs. We thus propose a general concept of bidirectional TRP-CaL-RyR-BKCa molecular and functional signaloplex in vascular smooth muscles. Accordingly, both TRPV4 and TRPM8 channels enhance local Ca2+ signals producing STOCs via TRP-RyR-BKCa coupling while simultaneously globally engaging BKCa and CaL channels by altering membrane potential.
Collapse
Affiliation(s)
- Dariia O Dryn
- O.O. Bogomoletz Institute of Physiology, National Academy of Sciences of Ukraine, 01024 Kyiv, Ukraine
| | - Mariia I Melnyk
- O.O. Bogomoletz Institute of Physiology, National Academy of Sciences of Ukraine, 01024 Kyiv, Ukraine
- ESC "Institute of Biology and Medicine", Taras Shevchenko National University of Kyiv, 01601 Kyiv, Ukraine
| | - Donal Melanaphy
- Centre for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast BT9 7BL, UK
| | - Igor V Kizub
- Division of Endocrinology and Metabolism, Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Christopher D Johnson
- Centre for Biomedical Sciences Education, Queen's University Belfast, Whitla Medical Building, Belfast BT9 7BL, UK
| | - Alexander V Zholos
- ESC "Institute of Biology and Medicine", Taras Shevchenko National University of Kyiv, 01601 Kyiv, Ukraine
| |
Collapse
|
18
|
Davis MJ, Earley S, Li YS, Chien S. Vascular mechanotransduction. Physiol Rev 2023; 103:1247-1421. [PMID: 36603156 PMCID: PMC9942936 DOI: 10.1152/physrev.00053.2021] [Citation(s) in RCA: 53] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 09/26/2022] [Accepted: 10/04/2022] [Indexed: 01/07/2023] Open
Abstract
This review aims to survey the current state of mechanotransduction in vascular smooth muscle cells (VSMCs) and endothelial cells (ECs), including their sensing of mechanical stimuli and transduction of mechanical signals that result in the acute functional modulation and longer-term transcriptomic and epigenetic regulation of blood vessels. The mechanosensors discussed include ion channels, plasma membrane-associated structures and receptors, and junction proteins. The mechanosignaling pathways presented include the cytoskeleton, integrins, extracellular matrix, and intracellular signaling molecules. These are followed by discussions on mechanical regulation of transcriptome and epigenetics, relevance of mechanotransduction to health and disease, and interactions between VSMCs and ECs. Throughout this review, we offer suggestions for specific topics that require further understanding. In the closing section on conclusions and perspectives, we summarize what is known and point out the need to treat the vasculature as a system, including not only VSMCs and ECs but also the extracellular matrix and other types of cells such as resident macrophages and pericytes, so that we can fully understand the physiology and pathophysiology of the blood vessel as a whole, thus enhancing the comprehension, diagnosis, treatment, and prevention of vascular diseases.
Collapse
Affiliation(s)
- Michael J Davis
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
| | - Scott Earley
- Department of Pharmacology, University of Nevada, Reno, Nevada
| | - Yi-Shuan Li
- Department of Bioengineering, University of California, San Diego, California
- Institute of Engineering in Medicine, University of California, San Diego, California
| | - Shu Chien
- Department of Bioengineering, University of California, San Diego, California
- Institute of Engineering in Medicine, University of California, San Diego, California
- Department of Medicine, University of California, San Diego, California
| |
Collapse
|
19
|
Benítez-Angeles M, Juárez-González E, Vergara-Jaque A, Llorente I, Rangel-Yescas G, Thébault SC, Hiriart M, Islas LD, Rosenbaum T. Unconventional interactions of the TRPV4 ion channel with beta-adrenergic receptor ligands. Life Sci Alliance 2023; 6:6/3/e202201704. [PMID: 36549871 PMCID: PMC9780703 DOI: 10.26508/lsa.202201704] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 12/05/2022] [Accepted: 12/08/2022] [Indexed: 12/24/2022] Open
Abstract
The transient receptor potential vanilloid 4 (TRPV4) ion channel is present in different tissues including those of the airways. This channel is activated in response to stimuli such as changes in temperature, hypoosmotic conditions, mechanical stress, and chemicals from plants, lipids, and others. TRPV4's overactivity and/or dysfunction has been associated with several diseases, such as skeletal dysplasias, neuromuscular disorders, and lung pathologies such as asthma and cardiogenic lung edema and COVID-19-related respiratory malfunction. TRPV4 antagonists and blockers have been described; nonetheless, the mechanisms involved in achieving inhibition of the channel remain scarce, and the search for safe use of these molecules in humans continues. Here, we show that the widely used bronchodilator salbutamol and other ligands of β-adrenergic receptors inhibit TRPV4's activation. We also demonstrate that inhibition of TRPV4 by salbutamol is achieved through interaction with two residues located in the outer region of the pore and that salbutamol leads to channel closing, consistent with an allosteric mechanism. Our study provides molecular insights into the mechanisms that regulate the activity of this physiopathologically important ion channel.
Collapse
Affiliation(s)
- Miguel Benítez-Angeles
- Departamento de Neurociencia Cognitiva, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México (UNAM), México, México
| | - Emmanuel Juárez-González
- Departamento de Neurociencia Cognitiva, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México (UNAM), México, México
| | - Ariela Vergara-Jaque
- Center for Bioinformatics, Simulation and Modeling, Faculty of Engineering, Universidad de Talca, Talca, Chile.,Millennium Nucleus of Ion Channel-Associated Diseases, Santiago, Chile
| | - Itzel Llorente
- Departamento de Neurociencia Cognitiva, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México (UNAM), México, México
| | | | | | - Marcia Hiriart
- Departamento de Neurociencia Cognitiva, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México (UNAM), México, México
| | - León D Islas
- Departamento de Fisiología, Facultad de Medicina, UNAM, México, México
| | - Tamara Rosenbaum
- Departamento de Neurociencia Cognitiva, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México (UNAM), México, México
| |
Collapse
|
20
|
Ca 2+-Activated K + Channels and the Regulation of the Uteroplacental Circulation. Int J Mol Sci 2023; 24:ijms24021349. [PMID: 36674858 PMCID: PMC9867535 DOI: 10.3390/ijms24021349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 01/06/2023] [Accepted: 01/08/2023] [Indexed: 01/13/2023] Open
Abstract
Adequate uteroplacental blood supply is essential for the development and growth of the placenta and fetus during pregnancy. Aberrant uteroplacental perfusion is associated with pregnancy complications such as preeclampsia, fetal growth restriction (FGR), and gestational diabetes. The regulation of uteroplacental blood flow is thus vital to the well-being of the mother and fetus. Ca2+-activated K+ (KCa) channels of small, intermediate, and large conductance participate in setting and regulating the resting membrane potential of vascular smooth muscle cells (VSMCs) and endothelial cells (ECs) and play a critical role in controlling vascular tone and blood pressure. KCa channels are important mediators of estrogen/pregnancy-induced adaptive changes in the uteroplacental circulation. Activation of the channels hyperpolarizes uteroplacental VSMCs/ECs, leading to attenuated vascular tone, blunted vasopressor responses, and increased uteroplacental blood flow. However, the regulation of uteroplacental vascular function by KCa channels is compromised in pregnancy complications. This review intends to provide a comprehensive overview of roles of KCa channels in the regulation of the uteroplacental circulation under physiological and pathophysiological conditions.
Collapse
|
21
|
Negri S, Sanford M, Shi H, Tarantini S. The role of endothelial TRP channels in age-related vascular cognitive impairment and dementia. Front Aging Neurosci 2023; 15:1149820. [PMID: 37020858 PMCID: PMC10067599 DOI: 10.3389/fnagi.2023.1149820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 02/28/2023] [Indexed: 04/07/2023] Open
Abstract
Transient receptor potential (TRP) proteins are part of a superfamily of polymodal cation channels that can be activated by mechanical, physical, and chemical stimuli. In the vascular endothelium, TRP channels regulate two fundamental parameters: the membrane potential and the intracellular Ca2+ concentration [(Ca2+)i]. TRP channels are widely expressed in the cerebrovascular endothelium, and are emerging as important mediators of several brain microvascular functions (e.g., neurovascular coupling, endothelial function, and blood-brain barrier permeability), which become impaired with aging. Aging is the most significant risk factor for vascular cognitive impairment (VCI), and the number of individuals affected by VCI is expected to exponentially increase in the coming decades. Yet, there are currently no preventative or therapeutic treatments available against the development and progression of VCI. In this review, we discuss the involvement of endothelial TRP channels in diverse physiological processes in the brain as well as in the pathogenesis of age-related VCI to explore future potential neuroprotective strategies.
Collapse
Affiliation(s)
- Sharon Negri
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging/Center for Geroscience and Healthy Brain Aging, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
| | - Madison Sanford
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging/Center for Geroscience and Healthy Brain Aging, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
| | - Helen Shi
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging/Center for Geroscience and Healthy Brain Aging, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Stefano Tarantini
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging/Center for Geroscience and Healthy Brain Aging, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- *Correspondence: Stefano Tarantini,
| |
Collapse
|
22
|
Caires R, Garrud TAC, Romero LO, Fernández-Peña C, Vásquez V, Jaggar JH, Cordero-Morales JF. Genetic- and diet-induced ω-3 fatty acid enrichment enhances TRPV4-mediated vasodilation in mice. Cell Rep 2022; 40:111306. [PMID: 36070688 PMCID: PMC9498980 DOI: 10.1016/j.celrep.2022.111306] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 07/05/2022] [Accepted: 08/11/2022] [Indexed: 11/19/2022] Open
Abstract
TRPV4 channel activation in endothelial cells leads to vasodilation, while impairment of TRPV4 activity is implicated in vascular dysfunction. Strategies that increase TRPV4 activity could enhance vasodilation and ameliorate vascular disorders. Here, we show that supplementation with eicosapentaenoic acid (EPA), an ω-3 polyunsaturated fatty acid known to have beneficial cardiovascular effects, increases TRPV4 activity in human endothelial cells of various vascular beds. Mice carrying the C. elegans FAT-1 enzyme, which converts ω-6 to ω-3 polyunsaturated fatty acids, display higher EPA content and increased TRPV4-mediated vasodilation in mesenteric arteries. Likewise, mice fed an EPA-enriched diet exhibit enhanced and prolonged TRPV4-dependent vasodilation in an endothelial cell-specific manner. We also show that EPA supplementation reduces TRPV4 desensitization, which contributes to the prolonged vasodilation. Neutralization of positive charges in the TRPV4 N terminus impairs the effect of EPA on channel desensitization. These findings highlight the beneficial effects of manipulating fatty acid content to enhance TRPV4-mediated vasodilation.
Collapse
Affiliation(s)
- Rebeca Caires
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Tessa A C Garrud
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Luis O Romero
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA; Integrated Biomedical Sciences Graduate Program, College of Graduate Health Sciences, Memphis, TN 38163, USA
| | - Carlos Fernández-Peña
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Valeria Vásquez
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Jonathan H Jaggar
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Julio F Cordero-Morales
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| |
Collapse
|
23
|
Kazandzhieva K, Mammadova-Bach E, Dietrich A, Gudermann T, Braun A. TRP channel function in platelets and megakaryocytes: basic mechanisms and pathophysiological impact. Pharmacol Ther 2022; 237:108164. [PMID: 35247518 DOI: 10.1016/j.pharmthera.2022.108164] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 01/29/2022] [Accepted: 02/28/2022] [Indexed: 12/30/2022]
Abstract
Transient receptor potential (TRP) proteins form a superfamily of cation channels that are expressed in a wide range of tissues and cell types. During the last years, great progress has been made in understanding the molecular complexity and the functions of TRP channels in diverse cellular processes, including cell proliferation, migration, adhesion and activation. The diversity of functions depends on multiple regulatory mechanisms by which TRP channels regulate Ca2+ entry mechanisms and intracellular Ca2+ dynamics, either through membrane depolarization involving cation influx or store- and receptor-operated mechanisms. Abnormal function or expression of TRP channels results in vascular pathologies, including hypertension, ischemic stroke and inflammatory disorders through effects on vascular cells, including the components of blood vessels and platelets. Moreover, some TRP family members also regulate megakaryopoiesis and platelet production, indicating a complex role of TRP channels in pathophysiological conditions. In this review, we describe potential roles of TRP channels in megakaryocytes and platelets, as well as their contribution to diseases such as thrombocytopenia, thrombosis and stroke. We also critically discuss the potential of TRP channels as possible targets for disease prevention and treatment.
Collapse
Affiliation(s)
- Kalina Kazandzhieva
- Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilians-University, Munich, Germany
| | - Elmina Mammadova-Bach
- Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilians-University, Munich, Germany; Division of Nephrology, Department of Medicine IV, Ludwig-Maximilians-University Hospital, Munich, Germany
| | - Alexander Dietrich
- Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilians-University, Munich, Germany; German Center for Lung Research (DZL), Munich, Germany
| | - Thomas Gudermann
- Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilians-University, Munich, Germany; German Center for Lung Research (DZL), Munich, Germany.
| | - Attila Braun
- Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilians-University, Munich, Germany.
| |
Collapse
|
24
|
Pereira da Silva EA, Martín-Aragón Baudel M, Navedo MF, Nieves-Cintrón M. Ion channel molecular complexes in vascular smooth muscle. Front Physiol 2022; 13:999369. [PMID: 36091375 PMCID: PMC9459047 DOI: 10.3389/fphys.2022.999369] [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: 07/21/2022] [Accepted: 08/02/2022] [Indexed: 11/30/2022] Open
Abstract
Ion channels that influence membrane potential and intracellular calcium concentration control vascular smooth muscle excitability. Voltage-gated calcium channels (VGCC), transient receptor potential (TRP) channels, voltage (KV), and Ca2+-activated K+ (BK) channels are key regulators of vascular smooth muscle excitability and contractility. These channels are regulated by various signaling cues, including protein kinases and phosphatases. The effects of these ubiquitous signaling molecules often depend on the formation of macromolecular complexes that provide a platform for targeting and compartmentalizing signaling events to specific substrates. This manuscript summarizes our current understanding of specific molecular complexes involving VGCC, TRP, and KV and BK channels and their contribution to regulating vascular physiology.
Collapse
|
25
|
Chen YL, Daneva Z, Kuppusamy M, Ottolini M, Baker TM, Klimentova E, Shah SA, Sokolowski JD, Park MS, Sonkusare SK. Novel Smooth Muscle Ca 2+-Signaling Nanodomains in Blood Pressure Regulation. Circulation 2022; 146:548-564. [PMID: 35758040 PMCID: PMC9378684 DOI: 10.1161/circulationaha.121.058607] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Ca2+ signals in smooth muscle cells (SMCs) contribute to vascular resistance and control blood pressure. Increased vascular resistance in hypertension has been attributed to impaired SMC Ca2+ signaling mechanisms. In this regard, transient receptor potential vanilloid 4 (TRPV4SMC) ion channels are a crucial Ca2+ entry pathway in SMCs. However, their role in blood pressure regulation has not been identified. METHODS We used SMC-specific TRPV4-/- (TRPV4SMC-/-) mice to assess the role of TRPV4SMC channels in blood pressure regulation. We determined the contribution of TRPV4SMC channels to the constrictor effect of α1 adrenergic receptor (α1AR) stimulation and elevated intraluminal pressure: 2 main physiologic stimuli that constrict resistance-sized arteries. The contribution of spatially separated TRPV4SMC channel subpopulations to elevated blood pressure in hypertension was evaluated in angiotensin II-infused mice and patients with hypertension. RESULTS We provide first evidence that TRPV4SMC channel activity elevates resting blood pressure in normal mice. α1AR stimulation activated TRPV4SMC channels through PKCα (protein kinase Cα) signaling, which contributed significantly to vasoconstriction and blood pressure elevation. Intraluminal pressure-induced TRPV4SMC channel activity opposed vasoconstriction through activation of Ca2+-sensitive K+ (BK) channels, indicating functionally opposite pools of TRPV4SMC channels. Superresolution imaging of SMCs revealed spatially separated α1AR:TRPV4 and TRPV4:BK nanodomains in SMCs. These data suggest that spatially separated α1AR-TRPV4SMC and intraluminal pressure-TRPV4SMC-BK channel signaling have opposite effects on blood pressure, with α1AR-TRPV4SMC signaling dominating under resting conditions. Furthermore, in patients with hypertension and a mouse model of hypertension, constrictor α1AR-PKCα-TRPV4 signaling was upregulated, whereas dilator pressure-TRPV4-BK channel signaling was disrupted, thereby increasing vasoconstriction and elevating blood pressure. CONCLUSIONS Our data identify novel smooth muscle Ca2+-signaling nanodomains that regulate blood pressure and demonstrate their impairment in hypertension.
Collapse
Affiliation(s)
- Yen-Lin Chen
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, 22908, USA
| | - Zdravka Daneva
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, 22908, USA
| | - Maniselvan Kuppusamy
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, 22908, USA
| | - Matteo Ottolini
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, 22908, USA
| | - Thomas M. Baker
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, 22908, USA
| | - Eliska Klimentova
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, 22908, USA
| | - Soham A. Shah
- Department of Neurosurgery, University of Virginia, Charlottesville, VA, 22908, USA
| | - Jennifer D. Sokolowski
- Department of Biomedical Engineering, University of Virginia, Charlottesville, United States, VA, 22908, USA
| | - Min S. Park
- Department of Biomedical Engineering, University of Virginia, Charlottesville, United States, VA, 22908, USA
| | - Swapnil K. Sonkusare
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, 22908, USA
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA, 22908, USA
| |
Collapse
|
26
|
Morin EE, Salbato S, Walker BR, Naik JS. Endothelial cell membrane cholesterol content regulates the contribution of TRPV4 channels in ACh-induced vasodilation in rat gracilis arteries. Microcirculation 2022; 29:e12774. [PMID: 35689491 PMCID: PMC10389065 DOI: 10.1111/micc.12774] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 05/26/2022] [Accepted: 06/06/2022] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Our previous work demonstrated that endothelial cell (EC) membrane cholesterol is reduced following 48 h of chronic hypoxia (CH). CH couples endothelial transient receptor potential subfamily V member 4 (TRPV4) channels to muscarinic receptor signaling through an endothelium-dependent hyperpolarization (EDH) pathway does not present in control animals. TRVPV4 channel activity has been shown to be regulated by membrane cholesterol. Hence, we hypothesize that acute manipulation of endothelial cell membrane cholesterol inversely determines the contribution of TRPV4 channels to endothelium-dependent vasodilation. METHODS Male Sprague-Dawley rats were exposed to ambient atmospheric (atm.) pressure or 48-h of hypoxia (0.5 atm). Vasodilation to acetylcholine (ACh) was determined using pressure myography in gracilis arteries. EC membrane cholesterol was depleted using methyl-β-cyclodextrin (MβCD) and supplemented with MβCD-cholesterol. RESULTS Inhibiting TRPV4 did not affect ACh-induced vasodilation in normoxic controls. However, TRPV4 inhibition reduced resting diameter in control arteries suggesting basal activity. TRPV4 contributes to ACh-induced vasodilation in these arteries when EC membrane cholesterol is depleted. Inhibiting TRPV4 attenuated ACh-induced vasodilation in arteries from CH animals that exhibit lower EC membrane cholesterol than normoxic controls. EC cholesterol repletion in arteries from CH animals abolished the contribution of TRPV4 to ACh-induced vasodilation. CONCLUSION Endothelial cell membrane cholesterol impedes the contribution of TRPV4 channels in EDH-mediated dilation. These results provide additional evidence for the importance of plasma membrane cholesterol content in regulating intracellular signaling and vascular function.
Collapse
Affiliation(s)
- Emily E Morin
- Department of Cell Biology and Physiology, University of New Mexico School of Medicine, Albuquerque, New Mexico, USA
| | - Sophia Salbato
- Department of Cell Biology and Physiology, University of New Mexico School of Medicine, Albuquerque, New Mexico, USA
| | - Benjimen R Walker
- Department of Cell Biology and Physiology, University of New Mexico School of Medicine, Albuquerque, New Mexico, USA
| | - Jay S Naik
- Department of Cell Biology and Physiology, University of New Mexico School of Medicine, Albuquerque, New Mexico, USA
| |
Collapse
|
27
|
Lansdell TA, Chambers LC, Dorrance AM. Endothelial Cells and the Cerebral Circulation. Compr Physiol 2022; 12:3449-3508. [PMID: 35766836 DOI: 10.1002/cphy.c210015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Endothelial cells form the innermost layer of all blood vessels and are the only vascular component that remains throughout all vascular segments. The cerebral vasculature has several unique properties not found in the peripheral circulation; this requires that the cerebral endothelium be considered as a unique entity. Cerebral endothelial cells perform several functions vital for brain health. The cerebral vasculature is responsible for protecting the brain from external threats carried in the blood. The endothelial cells are central to this requirement as they form the basis of the blood-brain barrier. The endothelium also regulates fibrinolysis, thrombosis, platelet activation, vascular permeability, metabolism, catabolism, inflammation, and white cell trafficking. Endothelial cells regulate the changes in vascular structure caused by angiogenesis and artery remodeling. Further, the endothelium contributes to vascular tone, allowing proper perfusion of the brain which has high energy demands and no energy stores. In this article, we discuss the basic anatomy and physiology of the cerebral endothelium. Where appropriate, we discuss the detrimental effects of high blood pressure on the cerebral endothelium and the contribution of cerebrovascular disease endothelial dysfunction and dementia. © 2022 American Physiological Society. Compr Physiol 12:3449-3508, 2022.
Collapse
Affiliation(s)
- Theresa A Lansdell
- Department of Pharmacology and Toxicology, College of Osteopathic Medicine, Michigan State University, East Lansing, MI, 48824, USA
| | - Laura C Chambers
- Department of Pharmacology and Toxicology, College of Osteopathic Medicine, Michigan State University, East Lansing, MI, 48824, USA
| | - Anne M Dorrance
- Department of Pharmacology and Toxicology, College of Osteopathic Medicine, Michigan State University, East Lansing, MI, 48824, USA
| |
Collapse
|
28
|
Malacarne PF, Bezzenberger J, Lopez M, Warwick T, Müller N, Brandes RP, Rezende F. Epoxyeicosatrienoic Acid and Prostanoid Crosstalk at the Receptor and Intracellular Signaling Levels to Maintain Vascular Tone. Int J Mol Sci 2022; 23:ijms23115939. [PMID: 35682616 PMCID: PMC9180422 DOI: 10.3390/ijms23115939] [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] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 05/18/2022] [Accepted: 05/23/2022] [Indexed: 11/16/2022] Open
Abstract
Epoxyeicosatrienoic acids (EETs) are signaling lipids produced by the cytochrome P450-(CYP450)-mediated epoxygenation of arachidonic acid. EETs have numerous biological effects on the vascular system, but aspects including their species specificity make their effects on vascular tone controversial. CYP450 enzymes require the 450-reductase (POR) for their activity. We set out to determine the contribution of endothelial CYP450 to murine vascular function using isolated aortic ring preparations from tamoxifen-inducible endothelial cell-specific POR knockout mice (ecPOR-/-). Constrictor responses to phenylephrine were similar between control (CTR) and ecPOR-/- mice. Contrastingly, sensitivity to the thromboxane receptor agonist U46619 and prostaglandin E2 (PGE2) was increased following the deletion of POR. Ex vivo incubation with a non-hydrolyzable EET (14,15-EE-8(Z)-E, EEZE) reversed the increased sensitivity to U46619 to the levels of CTR. EETs had no effect on vascular tone in phenylephrine-preconstricted vessels, but dilated vessels contracted with U46619 or PGE2. As U46619 acts through RhoA-dependent kinase, this system was analyzed. The deletion of POR affected the expression of genes in this pathway and the inhibition of Rho-GTPase with SAR407899 decreased sensitivity to U46619. These data suggest that EET and prostanoid crosstalk at the receptor level and that lack of EET production sensitizes vessels to vasoconstriction via the induction of the Rho kinase system.
Collapse
Affiliation(s)
- Pedro Felipe Malacarne
- Institute for Cardiovascular Physiology, Goethe-University, 60590 Frankfurt, Germany; (P.F.M.); (J.B.); (M.L.); (T.W.); (N.M.); (R.P.B.)
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhein-Main, 60590 Frankfurt, Germany
| | - Justus Bezzenberger
- Institute for Cardiovascular Physiology, Goethe-University, 60590 Frankfurt, Germany; (P.F.M.); (J.B.); (M.L.); (T.W.); (N.M.); (R.P.B.)
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhein-Main, 60590 Frankfurt, Germany
| | - Melina Lopez
- Institute for Cardiovascular Physiology, Goethe-University, 60590 Frankfurt, Germany; (P.F.M.); (J.B.); (M.L.); (T.W.); (N.M.); (R.P.B.)
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhein-Main, 60590 Frankfurt, Germany
| | - Timothy Warwick
- Institute for Cardiovascular Physiology, Goethe-University, 60590 Frankfurt, Germany; (P.F.M.); (J.B.); (M.L.); (T.W.); (N.M.); (R.P.B.)
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhein-Main, 60590 Frankfurt, Germany
| | - Niklas Müller
- Institute for Cardiovascular Physiology, Goethe-University, 60590 Frankfurt, Germany; (P.F.M.); (J.B.); (M.L.); (T.W.); (N.M.); (R.P.B.)
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhein-Main, 60590 Frankfurt, Germany
| | - Ralf P. Brandes
- Institute for Cardiovascular Physiology, Goethe-University, 60590 Frankfurt, Germany; (P.F.M.); (J.B.); (M.L.); (T.W.); (N.M.); (R.P.B.)
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhein-Main, 60590 Frankfurt, Germany
| | - Flávia Rezende
- Institute for Cardiovascular Physiology, Goethe-University, 60590 Frankfurt, Germany; (P.F.M.); (J.B.); (M.L.); (T.W.); (N.M.); (R.P.B.)
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhein-Main, 60590 Frankfurt, Germany
- Correspondence: ; Tel.: +49-69-6301-6996; Fax: +49-69-6301-7668
| |
Collapse
|
29
|
Chen Y, Sonkusare SK. Mechanosensitive Angiotensin II Receptor Signaling in Pressure‐Induced Vasoconstriction. J Am Heart Assoc 2022; 11:e024740. [PMID: 35156384 PMCID: PMC9245830 DOI: 10.1161/jaha.121.024740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Yen‐Lin Chen
- Robert M. Berne Cardiovascular Research Center University of Virginia Charlottesville VA
- Department of Molecular Physiology and Biological Physics University of Virginia Charlottesville VA
| | - Swapnil K. Sonkusare
- Robert M. Berne Cardiovascular Research Center University of Virginia Charlottesville VA
- Department of Molecular Physiology and Biological Physics University of Virginia Charlottesville VA
| |
Collapse
|
30
|
Reid C, Romero M, Chang SB, Osman N, Puglisi JL, Wilson CG, Blood AB, Zhang L, Wilson SM. Long-Term Hypoxia Negatively Influences Ca2+ Signaling in Basilar Arterial Myocytes of Fetal and Adult Sheep. Front Physiol 2022; 12:760176. [PMID: 35115953 PMCID: PMC8804533 DOI: 10.3389/fphys.2021.760176] [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: 08/17/2021] [Accepted: 10/25/2021] [Indexed: 11/21/2022] Open
Abstract
Cerebral arterial vasoreactivity is vital to the regulation of cerebral blood flow. Depolarization of arterial myocytes elicits whole-cell Ca2+ oscillations as well as subcellular Ca2+ sparks due to activation of ryanodine receptors on the sarcoplasmic reticulum. Previous evidence illustrates that contraction of cerebral arteries from sheep and underlying Ca2+ signaling pathways are modified by age and that long-term hypoxia (LTH) causes aberrations in Ca2+ signaling pathways and downstream effectors impacting vasoregulation. We hypothesize that age and LTH affect the influence of membrane depolarization on whole-cell intracellular Ca2+ oscillations and sub-cellular Ca2+ spark activity in cerebral arteries. To test this hypothesis, we examined Ca2+ oscillatory and spark activities using confocal fluorescence imaging techniques of Fluo-4 loaded basilar arterial myocytes of low- and high-altitude term fetal (∼145 days of gestation) and adult sheep, where high-altitude pregnant and non-pregnant sheep were placed at 3,801 m for >100 days. Ca2+ oscillations and sparks were recorded using an in situ preparation evaluated in the absence or presence of 30 mM K+ (30K) to depolarize myocytes. Myocytes from adult animals tended to have a lower basal rate of whole-cell Ca2+ oscillatory activity and 30K increased the activity within cells. LTH decreased the ability of myocytes to respond to depolarization independent of age. These observations illustrate that both altitude and age play a role in affecting whole-cell and localized Ca2+ signaling, which are important to arterial vasoreactivity and cerebral blood flow.
Collapse
Affiliation(s)
- Casey Reid
- Lawrence D. Longo, MD Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, CA, United States
| | - Monica Romero
- Advanced Imaging and Microscopy Core, Loma Linda University School of Medicine, Loma Linda, CA, United States
| | - Stephanie B. Chang
- Lawrence D. Longo, MD Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, CA, United States
| | - Noah Osman
- Lawrence D. Longo, MD Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, CA, United States
| | - Jose L. Puglisi
- Department of Biostatistics, School of Medicine, California Northstate University, Elk Grove, CA, United States
| | - Christopher G. Wilson
- Lawrence D. Longo, MD Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, CA, United States
| | - Arlin B. Blood
- Lawrence D. Longo, MD Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, CA, United States
| | - Lubo Zhang
- Lawrence D. Longo, MD Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, CA, United States
| | - Sean M. Wilson
- Lawrence D. Longo, MD Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, CA, United States
- Advanced Imaging and Microscopy Core, Loma Linda University School of Medicine, Loma Linda, CA, United States
- *Correspondence: Sean M. Wilson,
| |
Collapse
|
31
|
Shah KR, Guan X, Yan J. Structural and Functional Coupling of Calcium-Activated BK Channels and Calcium-Permeable Channels Within Nanodomain Signaling Complexes. Front Physiol 2022; 12:796540. [PMID: 35095560 PMCID: PMC8795833 DOI: 10.3389/fphys.2021.796540] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Accepted: 12/28/2021] [Indexed: 11/13/2022] Open
Abstract
Biochemical and functional studies of ion channels have shown that many of these integral membrane proteins form macromolecular signaling complexes by physically associating with many other proteins. These macromolecular signaling complexes ensure specificity and proper rates of signal transduction. The large-conductance, Ca2+-activated K+ (BK) channel is dually activated by membrane depolarization and increases in intracellular free Ca2+ ([Ca2+]i). The activation of BK channels results in a large K+ efflux and, consequently, rapid membrane repolarization and closing of the voltage-dependent Ca2+-permeable channels to limit further increases in [Ca2+]i. Therefore, BK channel-mediated K+ signaling is a negative feedback regulator of both membrane potential and [Ca2+]i and plays important roles in many physiological processes and diseases. However, the BK channel formed by the pore-forming and voltage- and Ca2+-sensing α subunit alone requires high [Ca2+]i levels for channel activation under physiological voltage conditions. Thus, most native BK channels are believed to co-localize with Ca2+-permeable channels within nanodomains (a few tens of nanometers in distance) to detect high levels of [Ca2+]i around the open pores of Ca2+-permeable channels. Over the last two decades, advancement in research on the BK channel’s coupling with Ca2+-permeable channels including recent reports involving NMDA receptors demonstrate exemplary models of nanodomain structural and functional coupling among ion channels for efficient signal transduction and negative feedback regulation. We hereby review our current understanding regarding the structural and functional coupling of BK channels with different Ca2+-permeable channels.
Collapse
Affiliation(s)
- Kunal R. Shah
- Department of Anesthesiology & Perioperative Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Xin Guan
- Department of Anesthesiology & Perioperative Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Jiusheng Yan
- Department of Anesthesiology & Perioperative Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- Neuroscience Program, Graduate School of Biomedical Sciences, UT Health, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- Biochemistry and Cell Biology Program, Graduate School of Biomedical Sciences, UT Health, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- *Correspondence: Jiusheng Yan,
| |
Collapse
|
32
|
TRPV4-dependent signaling mechanisms in systemic and pulmonary vasculature. CURRENT TOPICS IN MEMBRANES 2022; 89:1-41. [DOI: 10.1016/bs.ctm.2022.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
33
|
De Silva TM, Sobey CG. Cerebral Vascular Biology in Health and Disease. Stroke 2022. [DOI: 10.1016/b978-0-323-69424-7.00001-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
34
|
The Emerging Pro-Algesic Profile of Transient Receptor Potential Vanilloid Type 4. Rev Physiol Biochem Pharmacol 2022; 186:57-93. [PMID: 36378366 DOI: 10.1007/112_2022_75] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Transient receptor potential vanilloid type 4 (TRPV4) channels are Ca2+-permeable non-selective cation channels which mediate a wide range of physiological functions and are activated and modulated by a diverse array of stimuli. One of this ion channel's least discussed functions is in relation to the generation and maintenance of certain pain sensations. However, in the two decades which have elapsed since the identification of this ion channel, considerable data has emerged concerning its function in mediating pain sensations. TRPV4 is a mediator of mechanical hyperalgesia in the various contexts in which a mechanical stimulus, comprising trauma (at the macro-level) or discrete extracellular pressure or stress (at the micro-level), results in pain. TRPV4 is also recognised as constituting an essential component in mediating inflammatory pain. It also plays a role in relation to many forms of neuropathic-type pain, where it functions in mediating mechanical allodynia and hyperalgesia.Here, we review the role of TRPV4 in mediating pain sensations.
Collapse
|
35
|
Abstract
The alveolo-capillary barrier is relatively impermeable, and facilitates gas exchange via the large alveolar surface in the lung. Disruption of alveolo-capillary barrier leads to accumulation of edema fluid in lung injury. Studies in animal models of various forms of lung injury provide evidence that TRPV4 channels play a critical role in disruption of the alveolo-capillary barrier and pathogenesis of lung injury. TRPV4 channels from capillary endothelial cells, alveolar epithelial cells, and immune cells have been implicated in the pathogenesis of lung injury. Recent studies in endothelium-specific TRPV4 knockout mice point to a central role for endothelial TRPV4 channels in lung injury. In this chapter, we review the findings on the pathological roles of endothelial TRPV4 channels in different forms of lung injury and future directions for further investigation.
Collapse
|
36
|
Dixon RE, Navedo MF, Binder MD, Santana LF. Mechanisms and Physiological Implications of Cooperative Gating of Ion Channels Clusters. Physiol Rev 2021; 102:1159-1210. [PMID: 34927454 DOI: 10.1152/physrev.00022.2021] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Ion channels play a central role in the regulation of nearly every cellular process. Dating back to the classic 1952 Hodgkin-Huxley model of the generation of the action potential, ion channels have always been thought of as independent agents. A myriad of recent experimental findings exploiting advances in electrophysiology, structural biology, and imaging techniques, however, have posed a serious challenge to this long-held axiom as several classes of ion channels appear to open and close in a coordinated, cooperative manner. Ion channel cooperativity ranges from variable-sized oligomeric cooperative gating in voltage-gated, dihydropyridine-sensitive Cav1.2 and Cav1.3 channels to obligatory dimeric assembly and gating of voltage-gated Nav1.5 channels. Potassium channels, transient receptor potential channels, hyperpolarization cyclic nucleotide-activated channels, ryanodine receptors (RyRs), and inositol trisphosphate receptors (IP3Rs) have also been shown to gate cooperatively. The implications of cooperative gating of these ion channels range from fine tuning excitation-contraction coupling in muscle cells to regulating cardiac function and vascular tone, to modulation of action potential and conduction velocity in neurons and cardiac cells, and to control of pace-making activity in the heart. In this review, we discuss the mechanisms leading to cooperative gating of ion channels, their physiological consequences and how alterations in cooperative gating of ion channels may induce a range of clinically significant pathologies.
Collapse
Affiliation(s)
- Rose Ellen Dixon
- Department of Physiology and Membrane Biology, University of California, Davis, CA, United States
| | - Manuel F Navedo
- Department of Pharmacology, University of California, Davis, CA, United States
| | - Marc D Binder
- Department of Physiology and Biophysics, University of Washington, Seattle, WA, United States
| | - L Fernando Santana
- Department of Physiology and Membrane Biology, University of California, Davis, CA, United States
| |
Collapse
|
37
|
Harraz OF, Jensen LJ. Vascular calcium signalling and ageing. J Physiol 2021; 599:5361-5377. [PMID: 34705288 PMCID: PMC9002240 DOI: 10.1113/jp280950] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 10/12/2021] [Indexed: 02/05/2023] Open
Abstract
Changes in cellular Ca2+ levels have major influences on vascular function and blood pressure regulation. Vascular smooth muscle cells (SMCs) and endothelial cells (ECs) orchestrate vascular activity in distinct ways, often involving highly specific fluctuations in Ca2+ signalling. Ageing is a major risk factor for cardiovascular diseases, but the impact of ageing per se on vascular Ca2+ signalling has received insufficient attention. We reviewed the literature for age-related changes in Ca2+ signalling in relation to vascular structure and function. Vascular tone dysregulation in several vascular beds has been linked to abnormal expression or activity of SMC voltage-gated Ca2+ channels, Ca2+ -activated K+ channels or TRPC6 channels. Some of these effects were linked to altered caveolae density, microRNA expression or 20-HETE abundance. Intracellular store Ca2+ handling was suppressed in ageing mainly via reduced expression of intracellular Ca2+ release channels, and Ca2+ reuptake or efflux pumps. An increase in mitochondrial Ca2+ uptake, leading to oxidative stress, could also play a role in SMC hypercontractility and structural remodelling in ageing. In ECs, ageing entailed diverse effects on spontaneous and evoked Ca2+ transients, as well as structural changes at the EC-SMC interface. The concerted effects of altered Ca2+ signalling on myogenic tone, endothelium-dependent vasodilatation, and vascular structure are likely to contribute to blood pressure dysregulation and blood flow distribution deficits in critical organs. With the increase in the world's ageing population, future studies should be directed at solving specific ageing-induced Ca2+ signalling deficits to combat the imminent accelerated vascular ageing and increased risk of cardiovascular diseases.
Collapse
Affiliation(s)
- Osama F. Harraz
- Department of Pharmacology, Larner College of Medicine, University of Vermont, Burlington, Vermont, USA,Vermont Center for Cardiovascular and Brain Health, University of Vermont, Burlington, Vermont, USA
| | - Lars Jørn Jensen
- Pathobiological Sciences, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg C, Denmark
| |
Collapse
|
38
|
Kuppusamy M, Ottolini M, Sonkusare SK. Role of TRP ion channels in cerebral circulation and neurovascular communication. Neurosci Lett 2021; 765:136258. [PMID: 34560190 PMCID: PMC8572163 DOI: 10.1016/j.neulet.2021.136258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 08/26/2021] [Accepted: 08/28/2021] [Indexed: 11/17/2022]
Abstract
The dynamic regulation of blood flow is essential for meeting the high metabolic demands of the brain and maintaining brain function. Cerebral blood flow is regulated primarily by 1) the intrinsic mechanisms that determine vascular contractility and 2) signals from neurons and astrocytes that alter vascular contractility. Stimuli from neurons and astrocytes can also initiate a signaling cascade in the brain capillary endothelium to increase regional blood flow. Recent studies provide evidence that TRP channels in endothelial cells, smooth muscle cells, neurons, astrocytes, and perivascular nerves control cerebrovascular contractility and cerebral blood flow. TRP channels exert their functional effects either through cell membrane depolarization or by serving as a Ca2+ influx pathway. Endothelial cells and astrocytes also maintain the integrity of the blood-brain barrier. Both endothelial cells and astrocytes express TRP channels, and an increase in endothelial TRP channel activity has been linked with a disrupted endothelial barrier function. Therefore, TRP channels can play a potentially important role in regulating blood-brain barrier integrity. Here, we review the regulation of cerebrovascular contractility by TRP channels under healthy and disease conditions and their potential roles in maintaining blood-brain barrier function.
Collapse
Affiliation(s)
- Maniselvan Kuppusamy
- Robert M. Berne Cardiovascular Research Center, University of Virginia-School of Medicine, Charlottesville, VA, USA
| | - Matteo Ottolini
- Robert M. Berne Cardiovascular Research Center, University of Virginia-School of Medicine, Charlottesville, VA, USA; Department of Pharmacology, University of Virginia-School of Medicine, Charlottesville, VA, USA
| | - Swapnil K Sonkusare
- Robert M. Berne Cardiovascular Research Center, University of Virginia-School of Medicine, Charlottesville, VA, USA; Department of Molecular Physiology and Biological Physics, University of Virginia-School of Medicine, Charlottesville, VA, USA.
| |
Collapse
|
39
|
Li Q, Cheng Y, Zhang S, Sun X, Wu J. TRPV4-induced Müller cell gliosis and TNF-α elevation-mediated retinal ganglion cell apoptosis in glaucomatous rats via JAK2/STAT3/NF-κB pathway. J Neuroinflammation 2021; 18:271. [PMID: 34789280 PMCID: PMC8596927 DOI: 10.1186/s12974-021-02315-8] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 11/01/2021] [Indexed: 01/22/2023] Open
Abstract
Background Glaucoma, the leading cause of irreversible blindness worldwide, is a type of retinal disease characterized by the selective death of retinal ganglion cells (RGCs). However, the pathogenesis of glaucoma has not been fully elucidated. Transient receptor potential vanilloid 4 (TRPV4) is a pressure-sensitive and calcium-permeable cation channel. TRPV4 is widely distributed in the retina and its sustained activation leads to RGC death; indicating that TRPV4 may be a possible target for glaucoma treatment. Here, we investigated the effects of TRPV4 on RGC apoptosis in a rat model of chronic ocular hypertension (COH), then examined the mechanism underlying these effects. Methods The COH model was established by injection of micro-magnetic beads into the anterior chamber of adult male rats. The expression levels of TRPV4, glial fibrillary acidic protein, and inflammatory factors were assessed by immunohistochemistry and immunoblotting. RGC apoptosis and visual dysfunction were evaluated by TUNEL assay and photopic negative response. Functional expression of TRPV4 was examined by electrophysiology and calcium imaging. Real-time polymerase chain reaction and immunoblotting were employed to investigate the molecular mechanism underlying the effects of TRPV4 on tumor necrosis factor-α (TNF-α) release. Results We found that TRPV4 played an essential role in glaucoma, such that high levels of TRPV4 expression were associated with elevated intraocular pressure. Furthermore, TRPV4 activation was involved in glaucoma-induced RGC apoptosis and RGC-related reductions in visual function. Mechanistic investigation demonstrated that TRPV4 activation led to enhanced Müller cell gliosis and TNF-α release via the JAK2/STAT3/NF-kB pathway, while TRPV4 inhibition could reverse these effects. Finally, TRPV4 activation could lead to elevated expression of TNF receptor 1 in RGCs, while inhibition of TNF-α could reduce TRPV4-mediated RGC apoptosis. Conclusions TRPV4 activation induces Müller cell gliosis and TNF-α elevation via the JAK2/STAT3/NF-κB pathway, which may exacerbate RGC apoptosis in glaucoma; these results suggest that TRPV4 can serve as a therapeutic target in glaucoma treatment. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-021-02315-8.
Collapse
Affiliation(s)
- Qian Li
- Eye Institute, Eye and ENT Hospital, College of Medicine, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Visual Impairment and Restoration, Science and Technology Commission of Shanghai Municipality, Shanghai, China.,NHC Key Laboratory of Myopia, Fudan University, Shanghai, China.,Key Laboratory of Myopia, Chinese Academy of Medical Sciences, National Health Commission, #83 Fenyang Road, 200031, Shanghai, China
| | - Yun Cheng
- Eye Institute, Eye and ENT Hospital, College of Medicine, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Visual Impairment and Restoration, Science and Technology Commission of Shanghai Municipality, Shanghai, China.,NHC Key Laboratory of Myopia, Fudan University, Shanghai, China.,Key Laboratory of Myopia, Chinese Academy of Medical Sciences, National Health Commission, #83 Fenyang Road, 200031, Shanghai, China
| | - Shenghai Zhang
- Shanghai Key Laboratory of Visual Impairment and Restoration, Science and Technology Commission of Shanghai Municipality, Shanghai, China.,NHC Key Laboratory of Myopia, Fudan University, Shanghai, China.,Key Laboratory of Myopia, Chinese Academy of Medical Sciences, National Health Commission, #83 Fenyang Road, 200031, Shanghai, China
| | - Xinghuai Sun
- Eye Institute, Eye and ENT Hospital, College of Medicine, Fudan University, Shanghai, China. .,Shanghai Key Laboratory of Visual Impairment and Restoration, Science and Technology Commission of Shanghai Municipality, Shanghai, China. .,NHC Key Laboratory of Myopia, Fudan University, Shanghai, China. .,Key Laboratory of Myopia, Chinese Academy of Medical Sciences, National Health Commission, #83 Fenyang Road, 200031, Shanghai, China.
| | - Jihong Wu
- Eye Institute, Eye and ENT Hospital, College of Medicine, Fudan University, Shanghai, China. .,Shanghai Key Laboratory of Visual Impairment and Restoration, Science and Technology Commission of Shanghai Municipality, Shanghai, China. .,NHC Key Laboratory of Myopia, Fudan University, Shanghai, China. .,Key Laboratory of Myopia, Chinese Academy of Medical Sciences, National Health Commission, #83 Fenyang Road, 200031, Shanghai, China.
| |
Collapse
|
40
|
Lu T, Lee HC. Coronary Large Conductance Ca 2+-Activated K + Channel Dysfunction in Diabetes Mellitus. Front Physiol 2021; 12:750618. [PMID: 34744789 PMCID: PMC8567020 DOI: 10.3389/fphys.2021.750618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 09/14/2021] [Indexed: 11/24/2022] Open
Abstract
Diabetes mellitus (DM) is an independent risk of macrovascular and microvascular complications, while cardiovascular diseases remain a leading cause of death in both men and women with diabetes. Large conductance Ca2+-activated K+ (BK) channels are abundantly expressed in arteries and are the key ionic determinant of vascular tone and organ perfusion. It is well established that the downregulation of vascular BK channel function with reduced BK channel protein expression and altered intrinsic BK channel biophysical properties is associated with diabetic vasculopathy. Recent efforts also showed that diabetes-associated changes in signaling pathways and transcriptional factors contribute to the downregulation of BK channel expression. This manuscript will review our current understandings on the molecular, physiological, and biophysical mechanisms that underlie coronary BK channelopathy in diabetes mellitus.
Collapse
Affiliation(s)
- Tong Lu
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
| | - Hon-Chi Lee
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
41
|
Gebremendhin D, Lindemer B, Weihrauch D, Harder DR, Lohr NL. Electromagnetic energy (670 nm) stimulates vasodilation through activation of the large conductance potassium channel (BKCa). PLoS One 2021; 16:e0257896. [PMID: 34610026 PMCID: PMC8491904 DOI: 10.1371/journal.pone.0257896] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 09/13/2021] [Indexed: 11/18/2022] Open
Abstract
INTRODUCTION Peripheral artery disease (PAD) is a highly morbid condition in which impaired blood flow to the limbs leads to pain and tissue loss. Previously we identified 670 nm electromagnetic energy (R/NIR) to increase nitric oxide levels in cells and tissue. NO elicits relaxation of smooth muscle (SMC) by stimulating potassium efflux and membrane hyperpolarization. The actions of energy on ion channel activity have yet to be explored. Here we hypothesized R/NIR stimulates vasodilation through activation of potassium channels in SMC. METHODS Femoral arteries or facial arteries from C57Bl/6 and Slo1-/- mice were isolated, pressurized to 60 mmHg, pre-constricted with U46619, and irradiated twice with energy R/NIR (10 mW/cm2 for 5 min) with a 10 min dark period between irradiations. Single-channel K+ currents were recorded at room temperature from cell-attached and excised inside-out membrane patches of freshly isolated mouse femoral arterial muscle cells using the patch-clamp technique. RESULTS R/NIR stimulated vasodilation requires functional activation of the large conductance potassium channels. There is a voltage dependent outward current in SMC with light stimulation, which is due to increases in the open state probability of channel opening. R/NIR modulation of channel opening is eliminated pharmacologically (paxilline) and genetically (BKca α subunit knockout). There is no direct action of light to modulate channel activity as excised patches did not increase the open state probability of channel opening. CONCLUSION R/NIR vasodilation requires indirect activation of the BKca channel.
Collapse
Affiliation(s)
- Debebe Gebremendhin
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, United States of America
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, United States of America
| | - Brian Lindemer
- Division of Cardiovascular Medicine, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States of America
- Clement J Zablocki VA Medical Center, Milwaukee, WI, United States of America
| | - Dorothee Weihrauch
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, United States of America
| | - David R. Harder
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, United States of America
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, United States of America
- Clement J Zablocki VA Medical Center, Milwaukee, WI, United States of America
| | - Nicole L. Lohr
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, United States of America
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, United States of America
- Division of Cardiovascular Medicine, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States of America
- Clement J Zablocki VA Medical Center, Milwaukee, WI, United States of America
- * E-mail:
| |
Collapse
|
42
|
Veteto AB, Peana D, Lambert MD, McDonald KS, Domeier TL. Transient receptor potential vanilloid-4 contributes to stretch-induced hypercontractility and time-dependent dysfunction in the aged heart. Cardiovasc Res 2021; 116:1887-1896. [PMID: 31693106 DOI: 10.1093/cvr/cvz287] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 09/29/2019] [Accepted: 11/04/2019] [Indexed: 01/06/2023] Open
Abstract
AIMS Cardiovascular disease remains the greatest cause of mortality in Americans over 65. The stretch-activated transient receptor potential vanilloid-4 (TRPV4) ion channel is expressed in cardiomyocytes of the aged heart. This investigation tests the hypothesis that TRPV4 alters Ca2+ handling and cardiac function in response to increased ventricular preload and cardiomyocyte stretch. METHODS AND RESULTS Left ventricular maximal pressure (PMax) was monitored in isolated working hearts of Aged (24-27 months) mice following preload elevation from 5 to 20mmHg, with and without TRPV4 antagonist HC067047 (HC, 1 µmol/L). In preload responsive hearts, PMax prior to and immediately following preload elevation (i.e. Frank-Starling response) was similar between Aged and Aged+HC. Within 1 min following preload elevation, Aged hearts demonstrated secondary PMax augmentation (Aged>Aged+HC) suggesting a role for stretch-activated TRPV4 in cardiac hypercontractility. However, after 20 min at 20 mmHg Aged exhibited depressed PMax (Aged<Aged+HC) suggestive of TRPV4-dependent contractile dysfunction with sustained stretch. To examine stretch-induced Ca2+ homeostasis at the single-cell level, isolated cardiomyocytes were stretched 10-15% of slack length while measuring intracellular Ca2+ with fura-2. Uniaxial longitudinal stretch increased intracellular Ca2+ levels and triggered Ca2+ overload and terminal cellular contracture in Aged, but not Aged+HC. Preload elevation in hearts of young/middle-age (3-12 months) mice produced an initial PMax increase (Frank-Starling response) without secondary PMax augmentation, and cardiomyocyte stretch did not affect intracellular Ca2+ levels. Hearts of transgenic mice with cardiac-specific TRPV4 expression exhibited PMax similar to 3- to 12-month control mice prior to and immediately following preload elevation but displayed secondary PMax augmentation. Cardiomyocytes of mice with transgenic TRPV4 expression were highly sensitive to mechanical stimulation and exhibited elevated Ca2+ levels, Ca2+ overload, and terminal contracture upon cellular attachment and stretch. CONCLUSION TRPV4 contributes to a stretch-induced increase in cardiomyocyte Ca2+ and cardiac hypercontractility, yet sustained stretch leads to cardiomyocyte Ca2+ overload and contractile dysfunction in the aged heart.
Collapse
Affiliation(s)
- Adam B Veteto
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, 1 Hospital Drive, Columbia, MO 65212, USA
| | - Deborah Peana
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, 1 Hospital Drive, Columbia, MO 65212, USA
| | - Michelle D Lambert
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, 1 Hospital Drive, Columbia, MO 65212, USA
| | - Kerry S McDonald
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, 1 Hospital Drive, Columbia, MO 65212, USA
| | - Timothy L Domeier
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, 1 Hospital Drive, Columbia, MO 65212, USA
| |
Collapse
|
43
|
Kärki T, Tojkander S. TRPV Protein Family-From Mechanosensing to Cancer Invasion. Biomolecules 2021; 11:1019. [PMID: 34356643 PMCID: PMC8301805 DOI: 10.3390/biom11071019] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 06/30/2021] [Accepted: 07/09/2021] [Indexed: 02/08/2023] Open
Abstract
Biophysical cues from the cellular microenvironment are detected by mechanosensitive machineries that translate physical signals into biochemical signaling cascades. At the crossroads of extracellular space and cell interior are located several ion channel families, including TRP family proteins, that are triggered by mechanical stimuli and drive intracellular signaling pathways through spatio-temporally controlled Ca2+-influx. Mechanosensitive Ca2+-channels, therefore, act as critical components in the rapid transmission of physical signals into biologically compatible information to impact crucial processes during development, morphogenesis and regeneration. Given the mechanosensitive nature of many of the TRP family channels, they must also respond to the biophysical changes along the development of several pathophysiological conditions and have also been linked to cancer progression. In this review, we will focus on the TRPV, vanilloid family of TRP proteins, and their connection to cancer progression through their mechanosensitive nature.
Collapse
Affiliation(s)
- Tytti Kärki
- Department of Applied Physics, School of Science, Aalto University, 00076 Espoo, Finland;
| | - Sari Tojkander
- Department of Veterinary Biosciences, Section of Pathology, University of Helsinki, 00014 Helsinki, Finland
| |
Collapse
|
44
|
Zhou T, Wang Z, Guo M, Zhang K, Geng L, Mao A, Yang Y, Yu F. Puerarin induces mouse mesenteric vasodilation and ameliorates hypertension involving endothelial TRPV4 channels. Food Funct 2021; 11:10137-10148. [PMID: 33155599 DOI: 10.1039/d0fo02356f] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Puerarin (Pue) is an isoflavone derived from the root of Pueraria lobata, which has been widely used as food and a herb for treating cardiovascular and cerebrovascular diseases. Transient receptor potential vanilloid 4 (TRPV4), a Ca2+-permeable channel with multiple modes of activation, plays an important role in vascular endothelial function and vasodilation. However, no reports have shown the effects of Pue on TRPV4 channels and mouse small mesenteric arteries. In the present study, we performed a molecular docking assay by using Discovery Studio 3.5 software to predict the binding of Pue to TRPV4 protein. The activation of TRPV4 by Pue was determined by intracellular Ca2+ concentration ([Ca2+]i), live-cell fluorescent Ca2+ imaging and patch clamp assays. Molecular docking results indicated a high possibility of Pue-TPRV4 binding. [Ca2+]i and Ca2+ imaging assays showed that Pue activated TRPV4 channels and increased [Ca2+]i in TRPV4-overexpressing HEK293 (TRPV4-HEK293) cells and primary mouse mesenteric artery endothelial cells (MAECs). Patch clamp assay demonstrated that Pue stimulated the TRPV4-mediated cation currents. Additionally, Pue relaxed mouse mesenteric arteries involving the TRPV4-small-conductance Ca2+-activated K+ channel (SKCa)/intermediate-conductance Ca2+-activated K+ channel (IKCa) pathway, and reduced systolic blood pressure (SBP) in high-salt-induced hypertensive mice. Our study found for the first time that Pue acts as a TRPV4 agonist, induces endothelium-dependent vasodilation in mouse mesenteric arteries, and attenuates blood pressure in high-salt-induced hypertensive mice, highlighting the beneficial effect of Pue in treating endothelial dysfunction-related cardiovascular diseases.
Collapse
Affiliation(s)
- Tingting Zhou
- Wuxi School of Medicine, Jiangnan University, 1800 Lihu Road, Wuxi 214122, China.
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Mughal A, Sun C, O'Rourke ST. Apelin Does Not Impair Coronary Artery Relaxation Mediated by Nitric Oxide-Induced Activation of BK Ca Channels. Front Pharmacol 2021; 12:679005. [PMID: 34122102 PMCID: PMC8194342 DOI: 10.3389/fphar.2021.679005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 05/18/2021] [Indexed: 11/13/2022] Open
Abstract
Apelin-APJ receptor signaling regulates vascular tone in cerebral and peripheral arteries. We recently reported that apelin inhibits BKCa channel function in cerebral arteries, resulting in impaired endothelium-dependent relaxations. In contrast, apelin causes endothelium-dependent relaxation of coronary arteries. However, the effects of apelin on BKCa channel function in coronary arterial myocytes have not yet been explored. We hypothesized that apelin-APJ receptor signaling does not have an inhibitory effect on coronary arterial BKCa channels and hence does not alter nitric oxide (NO)-dependent relaxation of coronary arteries. Patch clamp recording was used to measure whole cell K+ currents in freshly isolated coronary smooth muscle cells. Apelin had no effect on the increases in current density in response to membrane depolarization or to NS1619 (a BKCa channel opener). Moreover, apelin did not inhibit NO/cGMP-dependent relaxations that required activation of BKCa channels in isolated coronary arteries. Apelin-APJ receptor signaling caused a marked increase in intracellular Ca2+ levels in coronary arterial smooth muscle cells, but failed to activate PI3-kinase to increase phosphorylation of Akt protein. Collectively, these data provide mechanistic evidence that apelin has no inhibitory effects on BKCa channel function in coronary arteries. The lack of inhibitory effect on BKCa channels makes it unlikely that activation of APJ receptors in coronary arteries would adversely affect coronary flow by creating a vasoconstrictive environment. It can be expected that apelin or other APJ receptor agonists in development will not interfere with the vasodilator effects of endogenous BKCa channel openers.
Collapse
Affiliation(s)
- Amreen Mughal
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND, United States
| | - Chengwen Sun
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND, United States
| | - Stephen T O'Rourke
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND, United States
| |
Collapse
|
46
|
Liu L, Guo M, Lv X, Wang Z, Yang J, Li Y, Yu F, Wen X, Feng L, Zhou T. Role of Transient Receptor Potential Vanilloid 4 in Vascular Function. Front Mol Biosci 2021; 8:677661. [PMID: 33981725 PMCID: PMC8107436 DOI: 10.3389/fmolb.2021.677661] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 04/06/2021] [Indexed: 12/19/2022] Open
Abstract
Transient receptor potential vanilloid 4 (TRPV4) channels are widely expressed in systemic tissues and can be activated by many stimuli. TRPV4, a Ca2+-permeable cation channel, plays an important role in the vasculature and is implicated in the regulation of cardiovascular homeostasis processes such as blood pressure, vascular remodeling, and pulmonary hypertension and edema. Within the vasculature, TRPV4 channels are expressed in smooth muscle cells, endothelial cells, and perivascular nerves. The activation of endothelial TRPV4 contributes to vasodilation involving nitric oxide, prostacyclin, and endothelial-derived hyperpolarizing factor pathways. TRPV4 activation also can directly cause vascular smooth muscle cell hyperpolarization and vasodilation. In addition, TRPV4 activation can evoke constriction in some specific vascular beds or under some pathological conditions. TRPV4 participates in the control of vascular permeability and vascular damage, particularly in the lung capillary endothelial barrier and lung injury. It also participates in vascular remodeling regulation mainly by controlling vasculogenesis and arteriogenesis. This review examines the role of TRPV4 in vascular function, particularly in vascular dilation and constriction, vascular permeability, vascular remodeling, and vascular damage, along with possible mechanisms, and discusses the possibility of targeting TRPV4 for therapy.
Collapse
Affiliation(s)
- Liangliang Liu
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Mengting Guo
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Xiaowang Lv
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Zhiwei Wang
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Jigang Yang
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Yanting Li
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Fan Yu
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Xin Wen
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Lei Feng
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Tingting Zhou
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| |
Collapse
|
47
|
Villegas D, Giard O, Brochu-Gaudreau K, Rousseau É. Activation of TRPV4 channels leads to a consistent tocolytic effect on human myometrial tissues. Eur J Obstet Gynecol Reprod Biol X 2021; 10:100124. [PMID: 33733088 PMCID: PMC7941160 DOI: 10.1016/j.eurox.2021.100124] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 02/26/2021] [Indexed: 11/21/2022] Open
Abstract
Specific pharmacological activation of alternative Ca2+ conductance. Activation of TRPV channels, abolishes the rhythmic contractile activity. Tocolysis was consistently induced on human myometrial strips.
Background Human myometrium is a therapeutic target for labor induction and preterm labor. Objective This study aimed to assess the physiological role of alternative calcium conductance on contractions triggered by uterotonic drugs in human myometrium. Membrane conductances, supported by TRPV channels, may provide alternative pathways to control either free intracellular and/or submembrane Ca2+-concentration, which in turn will modulate membrane polarization and contractile responses. Study design Uterine biopsies were obtained from consenting women undergoing elective caesarean delivery at term without labor (N = 22). Isometric tension measurements were performed on uterine smooth muscle strips (n = 132). Amplitude, frequency, and area under the curve (AUC) of phasic contractions, as well as resting tone, were measured under various experimental conditions. Immuno histo- and cyto-chemistry, as well as Western blot analyses, have been performed with specific antibodies against TRPV1, TRPV3, and TRPV4 proteins. TRPV4 agonists; GSK1016790A, 4αPDD, and 5,6-EET were used to assess the role of TRPV4 channels on rhythmic activity triggered by 30–300 nM oxytocin. 5 μM of ruthenium red was used as an efficient blocker of ionic current through TRPV4 channels. Nanomolar concentrations of iberiotoxin (IbTX) were also used to confirm the downstream involvement of BKCa channels in controlling uterine reactivity and contractility. Results The expression of TRPV3 and TRPV4 isoforms has now been demonstrated in human myometrial tissue and cell culture. Nanomolar concentrations of the TRPV4 agonists, (either GSK1016790A or 4αPDD) abolished the rhythmic contractions, resulting in a rapid and consistent tocolytic effect. While 5 μM of ruthenium reversed this tocolytic effect. The addition of IbTX (a BKCa channel blocker) reversed the effects of GSK1016790A. Carvacrol, a TRPV3 agonist, had similar tocolytic effects on rhythmic contractions albeit at higher concentrations. This inhibitory effect was also reversed by ruthenium red. Conclusion Collectively, these data suggest that activation of TRPV4 leads to a Ca2+ entry and subsequent BKCa channel activation (increase in open state probability), which in turn hyperpolarizes the myometrial cell membrane, inactivating L-type Ca2+ channels and efficiently abrogates contractile activity. Consequently, alternative Ca2+ conductance supported by TRPV4 plays a physiological role in the modulation of myometrial reactivity.
Collapse
Affiliation(s)
- Daniela Villegas
- Department of Obstetrics and Gynecology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, and Centre de Recherche du CHUS, Sherbrooke, QC, Canada
| | - Olivier Giard
- Department of Obstetrics and Gynecology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, and Centre de Recherche du CHUS, Sherbrooke, QC, Canada
| | - Karine Brochu-Gaudreau
- Department of Obstetrics and Gynecology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, and Centre de Recherche du CHUS, Sherbrooke, QC, Canada
| | - Éric Rousseau
- Department of Obstetrics and Gynecology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, and Centre de Recherche du CHUS, Sherbrooke, QC, Canada
| |
Collapse
|
48
|
Alghanem AF, Abello J, Maurer JM, Kumar A, Ta CM, Gunasekar SK, Fatima U, Kang C, Xie L, Adeola O, Riker M, Elliot-Hudson M, Minerath RA, Grueter CE, Mullins RF, Stratman AN, Sah R. The SWELL1-LRRC8 complex regulates endothelial AKT-eNOS signaling and vascular function. eLife 2021; 10:61313. [PMID: 33629656 PMCID: PMC7997661 DOI: 10.7554/elife.61313] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 02/22/2021] [Indexed: 12/15/2022] Open
Abstract
The endothelium responds to numerous chemical and mechanical factors in regulating vascular tone, blood pressure, and blood flow. The endothelial volume-regulated anion channel (VRAC) has been proposed to be mechanosensitive and thereby sense fluid flow and hydrostatic pressure to regulate vascular function. Here, we show that the leucine-rich repeat-containing protein 8a, LRRC8A (SWELL1), is required for VRAC in human umbilical vein endothelial cells (HUVECs). Endothelial LRRC8A regulates AKT-endothelial nitric oxide synthase (eNOS) signaling under basal, stretch, and shear-flow stimulation, forms a GRB2-Cav1-eNOS signaling complex, and is required for endothelial cell alignment to laminar shear flow. Endothelium-restricted Lrrc8a KO mice develop hypertension in response to chronic angiotensin-II infusion and exhibit impaired retinal blood flow with both diffuse and focal blood vessel narrowing in the setting of type 2 diabetes (T2D). These data demonstrate that LRRC8A regulates AKT-eNOS in endothelium and is required for maintaining vascular function, particularly in the setting of T2D.
Collapse
Affiliation(s)
- Ahmad F Alghanem
- Department of Internal Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, United States.,Eastern Region, King Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, Al Hasa, Saudi Arabia
| | - Javier Abello
- Department of Cell Biology and Physiology, Washington University in St. Louis, School of Medicine, St. Louis, United States
| | - Joshua M Maurer
- Department of Internal Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, United States
| | - Ashutosh Kumar
- Department of Internal Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, United States
| | - Chau My Ta
- Department of Internal Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, United States
| | - Susheel K Gunasekar
- Department of Internal Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, United States
| | - Urooj Fatima
- Department of Internal Medicine, Cardiovascular Division, University of Iowa, Iowa City, United States
| | - Chen Kang
- Department of Internal Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, United States
| | - Litao Xie
- Department of Internal Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, United States
| | - Oluwaseun Adeola
- Department of Internal Medicine, Cardiovascular Division, University of Iowa, Iowa City, United States
| | - Megan Riker
- Department of Ophthalmology, University of Iowa, Carver College of Medicine, Iowa City, United States
| | - Macaulay Elliot-Hudson
- Department of Internal Medicine, Cardiovascular Division, University of Iowa, Iowa City, United States
| | - Rachel A Minerath
- Department of Internal Medicine, Cardiovascular Division, University of Iowa, Iowa City, United States
| | - Chad E Grueter
- Department of Internal Medicine, Cardiovascular Division, University of Iowa, Iowa City, United States
| | - Robert F Mullins
- Department of Ophthalmology, University of Iowa, Carver College of Medicine, Iowa City, United States
| | - Amber N Stratman
- Department of Cell Biology and Physiology, Washington University in St. Louis, School of Medicine, St. Louis, United States
| | - Rajan Sah
- Department of Internal Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, United States.,Center for Cardiovascular Research, Washington University, St Louis, United States
| |
Collapse
|
49
|
Kim K, Hong KS. Transient receptor potential channel-dependent myogenic responsiveness in small-sized resistance arteries. J Exerc Rehabil 2021; 17:4-10. [PMID: 33728282 PMCID: PMC7939990 DOI: 10.12965/jer.2040836.418] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 12/19/2020] [Indexed: 11/22/2022] Open
Abstract
It is well documented that the inherent ability of small arteries and arterioles to regulate intraluminal diameter in response to alterations in intravascular pressure determines peripheral vascular resistance and blood flow (termed myogenic response or pressure-induced vasoconstriction/dilation). This autoregulatory property of resistance arteries is primarily originated from mechanosensitive vascular smooth muscle cells (VSMCs). There are diverse biological apparatuses in the plasma membrane of VSMCs that sense mechanical stimuli and generate intracellular signals for the contractility of VSMCs. Although the roles of transient receptor potential (TRP) channels in pressure-induced vasoconstriction are not fully understood to date, TRP channels that are directly activated by mechanical stimuli (e.g., stretch of VSMCs) or indirectly evoked by intracellular molecules (e.g., inositol trisphosphate) provide the major sources of Ca2+ (e.g., Ca2+ influx or release from the sarcoplasmic reticulum) and in turn, evoke vascular reactivity. This review sought to summarize mounting evidence over several decades that the activation of TRP canonical, TRP melastatin, TRP vanilloid, and TRP polycystin channels contributes to myogenic vasoconstriction.
Collapse
Affiliation(s)
- Kijeong Kim
- School of Exercise & Sport Science, College of Natural Sciences, University of Ulsan, Ulsan, Korea
| | - Kwang-Seok Hong
- Department of Physical Education, College of Education, Chung-Ang University, Seoul, Korea
| |
Collapse
|
50
|
Abstract
Members of the transient receptor potential (TRP) channels that are expressed in the kidney have gained prominence in recent years following discoveries of their role in maintaining the integrity of the filtration barrier, regulating tubular reabsorption of Ca2+ and Mg2+, and sensing osmotic stimuli. Furthermore, evidence has linked mutations in TRP channels to kidney disease pathophysiological mechanisms, including focal segmental glomerulosclerosis, disturbances in Mg2+ homeostasis, and polycystic kidney disease. Several subtypes of TRP channels are expressed in the renal vasculature, from preglomerular arteries and arterioles to the descending vasa recta. Although investigations on the physiological and pathological significance of renal vascular TRP channels are sparse, studies on isolated vessels and cells have suggested their involvement in renal vasoregulation. Renal blood flow (RBF) is an essential determinant of kidney function, including glomerular filtration, water and solute reabsorption, and waste product excretion. Functional alterations in ion channels that are expressed in the endothelium and smooth muscle of renal vessels can modulate renal vascular resistance, arterial pressure, and RBF. Hence, renal vascular TRP channels are potential therapeutic targets for the treatment of kidney disease. This review summarizes the current knowledge of TRP channel expression in renal vasculature and their role in controlling kidney function in health and disease. TRP channels are widely distributed in mammalian kidneys in glomerular, tubular, and vascular cells. TRPC and TRPV channels are functionally expressed in afferent arterioles. TRPC4 may regulate Ca2+ signaling in the descending vasa recta. Smooth muscle, endothelial, and pericyte TRP channels may participate in signal transduction mechanisms. TRP channels underlie renal autoregulation and regional kidney perfusion in health and disease.
Collapse
Affiliation(s)
- Praghalathan Kanthakumar
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Adebowale Adebiyi
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| |
Collapse
|