1
|
Shaw AA, Steketee JD, Bukiya AN, Dopico AM. Toluene is a cerebral artery constrictor acting via BK channels. Neuropharmacology 2025; 266:110272. [PMID: 39706291 PMCID: PMC11745904 DOI: 10.1016/j.neuropharm.2024.110272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 12/09/2024] [Accepted: 12/16/2024] [Indexed: 12/23/2024]
Abstract
Acute intoxication by toluene usually follows intentional inhalation to achieve a "high", which may lead to repeated use due to toluene's reinforcing properties. In both acute and chronic intoxication brain function is primarily affected. Neuronal and glial elements participate in toluene's reinforcing properties and chronic toxicity, yet the targets underlying acute toxicity remain unknown. Many signs of toluene's acute toxicity overlap with those of brain ischemia. Moreover, two studies in humans who abused toluene reveal brain hypoperfusion in middle cerebral artery (MCA) territories. Hypoperfusion, however, may result from either excessive vasoconstriction/increased vasodilation. Using rat and mouse models, we demonstrate that toluene at concentrations reached during recreational inhalation (8000 ppm) significantly decreases (-8%) MCA diameter in vivo in male and female animals. Using GC-MS, we determined toluene blood levels from inhalation (0.09-127 mM) and then show that <1 mM toluene constricts ex vivo-pressurized MCA independently of endothelium. Toluene action is blunted by deletion of KCNMA1, which codes for BK channels, key regulators of MCA diameter, and upon selective channel blockade by 1 μM paxilline. Lastly, when applied onto an isolated membrane patch several minutes after patch-excision from the SM cell, submM toluene reduces mildly yet statistically significantly (P < 0.05) both steady-state activity (-15%) and unitary current amplitude (-20%) of MCA myocyte BK channels. Thus, BK channels themselves and their immediate proteolipid microenvironment suffice for these drug actions. Collectively, data unveil a direct inhibition of MCA myocyte BK currents by intoxicating levels of toluene, which determines, or at least contributes to, MCA constriction by toluene levels reached during inhalation by humans who suffer acute brain intoxication.
Collapse
Affiliation(s)
- Andrew A Shaw
- Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN, 38103, USA
| | - Jeffery D Steketee
- Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN, 38103, USA
| | - Anna N Bukiya
- Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN, 38103, USA
| | - Alex M Dopico
- Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN, 38103, USA.
| |
Collapse
|
2
|
Al-Hosni R, Agostinelli E, Ilkan Z, Scofano L, Kaye R, Dinsdale RL, Acheson K, MacDonald A, Rivers D, Biosa A, Gunthorpe MJ, Platt F, Tammaro P. Pharmacological profiling of small molecule modulators of the TMEM16A channel and their implications for the control of artery and capillary function. Br J Pharmacol 2025. [PMID: 39829151 DOI: 10.1111/bph.17383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 09/26/2024] [Accepted: 10/01/2024] [Indexed: 01/22/2025] Open
Abstract
BACKGROUND AND PURPOSE TMEM16A chloride channels constitute a depolarising mechanism in arterial smooth muscle cells (SMCs) and contractile cerebral pericytes. TMEM16A pharmacology is incompletely defined. We elucidated the mode of action and selectivity of a recently identified positive allosteric modulator of TMEM16A (PAM_16A) and of a range of TMEM16A inhibitors. We also explore the consequences of selective modulation of TMEM16A activity on arterial and capillary function. EXPERIMENTAL APPROACH Patch-clamp electrophysiology, isometric tension recordings, live imaging of cerebral cortical capillaries and assessment of cell death were employed to explore the effect of selective pharmacological control of TMEM16A on vascular function. KEY RESULTS In low intracellular free Ca2+ concentrations ([Ca2+]i), nanomolar concentrations of PAM_16A activated heterologous TMEM16A channels, while being almost ineffective on the closely related TMEM16B channel. In either the absence of Ca2+ or in saturating [Ca2+]i, PAM_16A had no effect on TMEM16A currents at physiological potentials. PAM_16A selectively activated TMEM16A currents in SMCs and enhanced aortic contraction caused by phenylephrine or angiotensin-II and capillary (pericyte) constriction evoked by endothelin-1 or oxygen-glucose deprivation (OGD) to simulate cerebral ischaemia. Conversely, selective TMEM16A inhibition with Ani9 facilitated aortic, mesenteric and pericyte relaxation, and protected against OGD-mediated pericyte cell death. Unlike PAM_16A and Ani9, a range of other available modulators were found to interfere with endogenous cationic currents in SMCs. CONCLUSIONS AND IMPLICATIONS Arterial tone and capillary diameter can be controlled with TMEM16A modulators, highlighting TMEM16A as a target for disorders with a vascular component, including hypertension, stroke, Alzheimer's disease and vascular dementia.
Collapse
Affiliation(s)
| | | | - Zeki Ilkan
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - Lara Scofano
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - Rachel Kaye
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - Ria L Dinsdale
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - Kathryn Acheson
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - Andrew MacDonald
- Autifony Therapeutics Ltd, Stevenage Bioscience Catalyst, Stevenage, UK
| | - Dean Rivers
- Autifony Therapeutics Ltd, Stevenage Bioscience Catalyst, Stevenage, UK
| | - Alice Biosa
- Autifony Srl, Istituto di Ricerca Pediatrica Citta' della Speranza, Padua, Italy
| | | | - Frances Platt
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - Paolo Tammaro
- Department of Pharmacology, University of Oxford, Oxford, UK
| |
Collapse
|
3
|
Panerai RB, Davies A, Alshehri A, Beishon LC, Minhas JS. Subcomponent analysis of the directional sensitivity of dynamic cerebral autoregulation. Am J Physiol Heart Circ Physiol 2025; 328:H37-H46. [PMID: 39570199 DOI: 10.1152/ajpheart.00498.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 11/08/2024] [Accepted: 11/11/2024] [Indexed: 11/22/2024]
Abstract
The origin of the directional sensitivity (DS) of dynamic cerebral autoregulation (dCA) is not known. In 140 healthy participants (67 male, 27.5 ± 6.1 yr old), middle cerebral artery velocity (MCAv, transcranial Doppler), arterial blood pressure (ABP, Finometer), and end-tidal CO2 (EtCO2, capnography) were recorded at rest. Critical closing pressure (CrCP) and resistance-area product (RAP) were obtained for each cardiac cycle, as well as mean MCAv and ABP (MAP). The integrated positive and negative derivatives of MAP (MAP+D and MAP-D, respectively) were used as simultaneous inputs to an autoregressive moving average model to generate two distinct MCAv step responses. Similar models allowed the estimation of corresponding MAP-CrCP and MAP-RAP responses to step changes in MAP+D and MAP-D. The strength of DS (ΔDS) was expressed by the difference in mean values of the step responses for the time interval 12-18 s. ΔDS was significant for MCAv (8.5 ± 46.9% vs. 26.7 ± 42.0%, P < 0.001) and RAP (-93.9 ± 48.1 vs. -74.5 ± 43.0%, P < 0.001), respectively, for MAP+D and MAP-D inputs, but not for CrCP (2.2 ± 48.1% vs. 0.72 ± 42.9%, P = 0.76). Compared with males, female participants had higher MCAv (63.9 ± 15.6 cm/s vs. 55.4 ± 12.9 cm/s, P < 0.001) but lower EtCO2 (P < 0.001) and RAP (P = 0.015). Sex did not influence ΔDS for any of the three-step responses. The presence of directional sensitivity in the RAP, but not in the CrCP transfer function, suggests that the origin could be solely myogenic, without metabolic involvement.NEW & NOTEWORTHY The directional sensitivity of the cerebral blood velocity response to a sudden change in mean arterial blood pressure (MAP) is mediated by the resistance-area product, without involvement from the cerebral critical closing pressure. The reduced amplitude of MAP spontaneous fluctuations at rest suggests that it is less likely that directional sensitivity has origins in the sympathetic control of cerebral blood vessels, thus generating the need to consider other alternatives.
Collapse
Affiliation(s)
- Ronney B Panerai
- Cerebral Haemodynamics in Ageing and Stroke Medicine (CHiASM), Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom
- NIHR Leicester Biomedical Research Centre, BHF Cardiovascular Research Centre, Glenfield Hospital, Leicester, United Kingdom
| | - Aaron Davies
- Cerebral Haemodynamics in Ageing and Stroke Medicine (CHiASM), Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom
| | - Abdulaziz Alshehri
- Cerebral Haemodynamics in Ageing and Stroke Medicine (CHiASM), Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom
- Emergency Medical Services Department, College of Applied Medical Sciences, Najran University, Najran, Saudi Arabia
| | - Lucy C Beishon
- Cerebral Haemodynamics in Ageing and Stroke Medicine (CHiASM), Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom
- NIHR Leicester Biomedical Research Centre, BHF Cardiovascular Research Centre, Glenfield Hospital, Leicester, United Kingdom
| | - Jatinder S Minhas
- Cerebral Haemodynamics in Ageing and Stroke Medicine (CHiASM), Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom
- NIHR Leicester Biomedical Research Centre, BHF Cardiovascular Research Centre, Glenfield Hospital, Leicester, United Kingdom
| |
Collapse
|
4
|
Silva JF, Polk FD, Martin PE, Thai SH, Savu A, Gonzales M, Kath AM, Gee MT, Pires PW. Sex-specific mechanisms of cerebral microvascular BK Ca dysfunction in a mouse model of Alzheimer's disease. Alzheimers Dement 2024. [PMID: 39698895 DOI: 10.1002/alz.14438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 09/18/2024] [Accepted: 11/06/2024] [Indexed: 12/20/2024]
Abstract
INTRODUCTION Cerebrovascular dysfunction occurs in Alzheimer's disease (AD), impairing hemodynamic regulation. Large conductance Ca2+-activated K+ channels (BKCa) regulate cerebrovascular reactivity and are impaired in AD. BKCa activity depends on intracellular Ca2+ (Ca2+ sparks) and nitro-oxidative post-translational modifications. However, whether these mechanisms underlie BKCa impairment in AD remains unknown. METHODS Cerebral arteries from 5x-FAD and wild-type (WT) littermates were used for molecular biology, electrophysiology, ex vivo, and in vivo experiments. RESULTS Arterial BKCa activity is reduced in 5x-FAD via sex-dependent mechanisms: in males, there is lower BKα subunit expression and less Ca2+ sparks. In females, we observed reversible nitro-oxidative modification of BKCa. Further, BKCa is involved in hemodynamic regulation in WT mice, and its dysfunction is associated with vascular deficits in 5x-FAD. DISCUSSION Our data highlight the central role played by BKCa in cerebral hemodynamic regulation and that molecular mechanisms of its impairment diverge based on sex in 5x-FAD. HIGHLIGHTS Cerebral microvascular BKCa dysfunction occurs in both female and male 5x-FAD. Reduction in BKα subunit protein and Ca2+ sparks drive the dysfunction in males. Nitro-oxidative stress is present in females, but not males, 5x-FAD. Reversible nitro-oxidation of BKα underlies BKCa dysfunction in female 5x-FAD.
Collapse
Affiliation(s)
- Josiane F Silva
- Department of Physiology, University of Arizona College of Medicine, Tucson, Arizona, USA
| | - Felipe D Polk
- Department of Physiology, University of Arizona College of Medicine, Tucson, Arizona, USA
| | - Paige E Martin
- Department of Physiology, University of Arizona College of Medicine, Tucson, Arizona, USA
| | - Stephenie H Thai
- Department of Physiology, University of Arizona College of Medicine, Tucson, Arizona, USA
| | - Andrea Savu
- Department of Physiology, University of Arizona College of Medicine, Tucson, Arizona, USA
| | - Matthew Gonzales
- Department of Physiology, University of Arizona College of Medicine, Tucson, Arizona, USA
| | - Allison M Kath
- Department of Physiology, University of Arizona College of Medicine, Tucson, Arizona, USA
| | - Michael T Gee
- Department of Physiology, University of Arizona College of Medicine, Tucson, Arizona, USA
| | - Paulo W Pires
- Department of Physiology, University of Arizona College of Medicine, Tucson, Arizona, USA
- Sarver Heart Center, University of Arizona College of Medicine, Tucson, Arizona, USA
- Bio5 Institute, University of Arizona College of Medicine, Tucson, Arizona, USA
| |
Collapse
|
5
|
Zhang X, Tian H, Xie C, Yang Y, Li P, Cheng J. The role and mechanism of vascular wall cell ion channels in vascular fibrosis remodeling. Channels (Austin) 2024; 18:2418128. [PMID: 39425532 PMCID: PMC11492694 DOI: 10.1080/19336950.2024.2418128] [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: 06/23/2024] [Revised: 09/24/2024] [Accepted: 10/12/2024] [Indexed: 10/21/2024] Open
Abstract
Fibrosis is usually the final pathological state of many chronic inflammatory diseases and may lead to organ malfunction. Excessive deposition of extracellular matrix (ECM) molecules is a characteristic of most fibrotic tissues. The blood vessel wall contains three layers of membrane structure, including the intima, which is composed of endothelial cells; the media, which is composed of smooth muscle cells; and the adventitia, which is formed by the interaction of connective tissue and fibroblasts. The occurrence and progression of vascular remodeling are closely associated with cardiovascular diseases, and vascular remodeling can alter the original structure and function of the blood vessel. Dysregulation of the composition of the extracellular matrix in blood vessels leads to the continuous advancement of vascular stiffening and fibrosis. Vascular fibrosis reaction leads to excessive deposition of the extracellular matrix in the vascular adventitia, reduces vessel compliance, and ultimately alters key aspects of vascular biomechanics. The pathogenesis of fibrosis in the vasculature and strategies for its reversal have become interesting and important challenges. Ion channels are widely expressed in the cardiovascular system; they regulate blood pressure, maintain cardiovascular function homeostasis, and play important roles in ion transport, cell differentiation, proliferation. In blood vessels, different types of ion channels in fibroblasts, smooth muscle cells and endothelial cells may be relevant mediators of the development of fibrosis in organs or tissues. This review discusses the known roles of ion channels in vascular fibrosis remodeling and discusses potential therapeutic targets for regulating remodeling and repair after vascular injury.
Collapse
Affiliation(s)
- Xiaolin Zhang
- Key Lab of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Lab of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Public Center of Experimental Technology, Hemodynamics and Medical Engineering Combination Key Laboratory of Luzhou, Southwest Medical University, Luzhou, China
| | - Hai Tian
- Key Lab of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Lab of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Public Center of Experimental Technology, Hemodynamics and Medical Engineering Combination Key Laboratory of Luzhou, Southwest Medical University, Luzhou, China
| | - Cheng Xie
- Key Lab of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Lab of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Public Center of Experimental Technology, Hemodynamics and Medical Engineering Combination Key Laboratory of Luzhou, Southwest Medical University, Luzhou, China
| | - Yan Yang
- Key Lab of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Lab of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Public Center of Experimental Technology, Hemodynamics and Medical Engineering Combination Key Laboratory of Luzhou, Southwest Medical University, Luzhou, China
| | - Pengyun Li
- Key Lab of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Lab of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Public Center of Experimental Technology, Hemodynamics and Medical Engineering Combination Key Laboratory of Luzhou, Southwest Medical University, Luzhou, China
| | - Jun Cheng
- Key Lab of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Lab of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Public Center of Experimental Technology, Hemodynamics and Medical Engineering Combination Key Laboratory of Luzhou, Southwest Medical University, Luzhou, China
| |
Collapse
|
6
|
Kamran SA, Moghnieh H, Hossain KF, Bartlett A, Tavakkoli A, Drumm BT, Sanders KM, Baker SA. Automated denoising software for calcium imaging signals using deep learning. Heliyon 2024; 10:e39574. [PMID: 39524741 PMCID: PMC11546308 DOI: 10.1016/j.heliyon.2024.e39574] [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: 10/27/2023] [Revised: 10/16/2024] [Accepted: 10/17/2024] [Indexed: 11/16/2024] Open
Abstract
Dynamic Ca2+ signaling is crucial for cell survival and death, and Ca2+ imaging approaches are commonly used to study and measure cellular Ca2+ patterns within cells. However, the presence of image noise from instrumentation and experimentation protocols can impede the accurate extraction of Ca2+ signals. Removing noise from Ca2+ Spatio-Temporal Maps (STMaps) is essential for precisely analyzing Ca2+ datasets. Current methods for denoising STMaps can be time-consuming and subjective and rely mainly on image processing protocols. To address this, we developed CalDenoise, an automated software that employs robust image processing and deep learning models to remove noise and enhance Ca2+ signals in STMaps effectively. CalDenoise integrates four pipelines capable of efficiently removing salt-and-pepper, impulsive, and periodic noise and detecting and removing background noise. Comprising both an image-processing-based pipeline and three generative-adversarial-network-based (GAN) deep learning models, CalDenoise proficiently removes complex noise patterns. The software features adjustable parameters to enhance accuracy and is integrated into a user-friendly graphical interface for easy access and streamlined usage. CalDenoise can serve as a robust platform for denoising complex dynamic fluorescence signal images across diverse cell types, including Ca2+, voltage, ions, and pH signals.
Collapse
Affiliation(s)
- Sharif Amit Kamran
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV, 89557, USA
- Department of Computer Science and Engineering, University of Nevada, Reno, NV 89557, USA
| | - Hussein Moghnieh
- Department of Electrical and Computer Engineering], McGill University, Montréal, Québec, H3A 0E9, Canada
| | | | - Allison Bartlett
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV, 89557, USA
| | - Alireza Tavakkoli
- Department of Computer Science and Engineering, University of Nevada, Reno, NV 89557, USA
| | - Bernard T. Drumm
- Department of Life & Health Science, Dundalk Institute of Technology, Co. Louth, Ireland
| | - Kenton M. Sanders
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV, 89557, USA
| | - Salah A. Baker
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV, 89557, USA
| |
Collapse
|
7
|
Nelapudi N, Boskind M, Hu XQ, Mallari D, Chan M, Wilson D, Romero M, Albert-Minckler E, Zhang L, Blood AB, Wilson CG, Puglisi JL, Wilson SM. Long-term hypoxia modulates depolarization activation of BK Ca currents in fetal sheep middle cerebral arterial myocytes. Front Physiol 2024; 15:1479882. [PMID: 39563935 PMCID: PMC11573761 DOI: 10.3389/fphys.2024.1479882] [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: 08/12/2024] [Accepted: 10/17/2024] [Indexed: 11/21/2024] Open
Abstract
Introduction Previous evidence indicates that gestational hypoxia disrupts cerebrovascular development, increasing the risk of intracranial hemorrhage and stroke in the newborn. Due to the role of cytosolic Ca2+ in regulating vascular smooth muscle (VSM) tone and fetal cerebrovascular blood flow, understanding Ca2+ signals can offer insight into the pathophysiological disruptions taking place in hypoxia-related perinatal cerebrovascular disease. This study aimed to determine the extent to which gestational hypoxia disrupts local Ca2+ sparks and whole-cell Ca2+ signals and coupling with BKCa channel activity. Methods Confocal imaging of cytosolic Ca2+ and recording BKCa currents of fetal sheep middle cerebral arterial (MCA) myocytes was performed. MCAs were isolated from term fetal sheep (∼140 days of gestation) from ewes held at low- (700 m) and high-altitude (3,801 m) hypoxia (LTH) for 100+ days of gestation. Arteries were depolarized with 30 mM KCl (30K), in the presence or absence of 10 μM ryanodine (Ry), to block RyR mediated Ca2+ release. Results Membrane depolarization increased Ry-sensitive Ca2+ spark frequency in normoxic and LTH groups along with BKCa activity. LTH reduced Ca2+ spark and whole-cell Ca2+ activity and induced a large leftward shift in the voltage-dependence of BKCa current activation. The influence of LTH on the spatial and temporal aspects of Ca2+ sparks and whole-cell Ca2+ responses varied. Discussion Overall, LTH attenuates Ca2+ signaling while increasing the coupling of Ca2+ sparks to BKCa activity; a process that potentially helps maintain oxygen delivery to the developing brain.
Collapse
Affiliation(s)
- Nikitha Nelapudi
- Lawrence D Longo Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, CA, United States
| | - Madison Boskind
- Lawrence D Longo Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, CA, United States
| | - Xiang-Qun Hu
- Lawrence D Longo Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, CA, United States
| | - David Mallari
- Lawrence D Longo Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, CA, United States
| | - Michelle Chan
- Lawrence D Longo Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, CA, United States
| | - Devin Wilson
- Lawrence D Longo 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
| | - Eris Albert-Minckler
- Advanced Imaging and Microscopy Core, Loma Linda University School of Medicine, Loma Linda, CA, United States
| | - Lubo Zhang
- Lawrence D Longo Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, CA, United States
| | - Arlin B Blood
- Lawrence D Longo Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, CA, United States
| | - Christopher G Wilson
- Lawrence D Longo Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, CA, United States
| | - Jose Luis Puglisi
- Department of Biostatistics, California Northstate University School of Medicine, Elk Grove, CA, United States
| | - Sean M Wilson
- Lawrence D Longo 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
| |
Collapse
|
8
|
Yucesan E, Goncu B, Ozgul C, Kebapci A, Aslanger AD, Akyuz E, Yesil G. Functional characterization of KCNMA1 mutation associated with dyskinesia, seizure, developmental delay, and cerebellar atrophy. Int J Neurosci 2024; 134:1098-1103. [PMID: 37269313 DOI: 10.1080/00207454.2023.2221814] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 04/26/2023] [Accepted: 05/31/2023] [Indexed: 06/05/2023]
Abstract
KCNMA1 located on chromosome 10q22.3, encodes the pore-forming α subunit of the 'Big K+' (BK) large conductance calcium and voltage-activated K + channel. Numerous evidence suggests the functional BK channel alterations produced by different KCNMA1 alleles may associate with different symptoms, such as paroxysmal non kinesigenic dyskinesia with gain of function and ataxia with loss of function. Functional classifications revealed two major patterns, gain of function and loss of function effects on channel properties in different cell lines. In the literature, two mutations have been shown to confer gain of function properties to BK channels: D434G and N995S. In this study, we report the functional characterization of a variant which was previously reported the whole exome sequencing revealed bi-allelic nonsense variation of the cytoplasmic domain of calcium-activated potassium channel subunit alpha-1 protein. To detect functional consequences of the variation, we parallely conducted two independent approaches. One is immunostaining using and the other one is electrophysiological recording using patch-clamp on wild-type and R458X mutant cells to detect the differences between wild-type and the mutant cells. We detected the gain of function effect for the mutation (NM_001161352.1 (ENST00000286628.8):c.1372C > T;Arg458*) using two parallel approaches. According to the result we found, the reported mutation causes the loss of function in the cell. It should be noted that in future studies, it can be thought that the functions of genes associated with channelopathies may have a dual effect such as loss and gain.
Collapse
Affiliation(s)
- Emrah Yucesan
- Department of Neurogenetics, Institute of Neurological Sciences, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Beyza Goncu
- Department of Medical Services and Techniques, Vocational School of Health Services, Bezmialem Vakif University, Istanbul, Turkey
- Experimental Research Center, Bezmialem Vakif University, Istanbul, Turkey
| | - Cemil Ozgul
- Regenerative and Restorative Medicine Research Center (REMER), Research Institute for Health Sciences and Technologies (SABITA, Istanbul Medipol University, Istanbul, Turkey)
| | - Arda Kebapci
- Regenerative and Restorative Medicine Research Center (REMER), Research Institute for Health Sciences and Technologies (SABITA, Istanbul Medipol University, Istanbul, Turkey)
| | - Ayca Dilruba Aslanger
- Department of Medical Genetics, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Enes Akyuz
- Department of Biophysics, Faculty of International Medicine, University of Health Sciences, Istanbul, Turkey
| | - Gozde Yesil
- Department of Medical Genetics, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| |
Collapse
|
9
|
Drumm BT, Gupta N, Mircea A, Griffin CS. Cells and ionic conductances contributing to spontaneous activity in bladder and urethral smooth muscle. J Physiol 2024. [PMID: 39323077 DOI: 10.1113/jp284744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 09/02/2024] [Indexed: 09/27/2024] Open
Abstract
Smooth muscle organs of the lower urinary tract comprise the bladder detrusor and urethral wall, which have a reciprocal contractile relationship during urine storage and micturition. As the bladder fills with urine, detrusor smooth muscle cells (DSMCs) remain relaxed to accommodate increases in intravesical pressure while urethral smooth muscle cells (USMCs) sustain tone to occlude the urethral orifice, preventing leakage. While neither organ displays coordinated regular contractions as occurs in small intestine, lymphatics or renal pelvis, they do exhibit patterns of rhythmicity at cellular and tissue levels. In rabbit and guinea-pig urethra, electrical slow waves are recorded from USMCs. This activity is linked to cells expressing vimentin, c-kit and Ca2+-activated Cl- channels, like interstitial cells of Cajal in the gastrointestinal tract. In mouse, USMCs are rhythmically active (firing propagating Ca2+ waves linked to contraction), and this cellular rhythmicity is asynchronous across tissues and summates to form tone. Experiments in mice have failed to demonstrate a voltage-dependent mechanism for regulating this rhythmicity or contractions in vitro, suggesting that urethral tone results from an intrinsic ability of USMCs to 'pace' their own Ca2+ mobilization pathways required for contraction. DSMCs exhibit spontaneous transient contractions, increases in intracellular Ca2+ and action potentials. Consistent across numerous species, including humans, this activity relies on voltage-dependent Ca2+ influx in DSMCs. While interstitial cells are present in the bladder, they do not 'pace' the organ in an excitatory manner. Instead, specialized cells (PDGFRα+ interstitial cells) may 'negatively pace' DSMCs to prevent bladder overexcitability.
Collapse
Affiliation(s)
- Bernard T Drumm
- Smooth Muscle Research Centre, Department of Life & Health Science, Dundalk Institute of Technology, Dundalk, Ireland
| | - Neha Gupta
- Smooth Muscle Research Centre, Department of Life & Health Science, Dundalk Institute of Technology, Dundalk, Ireland
| | - Alexandru Mircea
- Smooth Muscle Research Centre, Department of Life & Health Science, Dundalk Institute of Technology, Dundalk, Ireland
| | - Caoimhin S Griffin
- Smooth Muscle Research Centre, Department of Life & Health Science, Dundalk Institute of Technology, Dundalk, Ireland
| |
Collapse
|
10
|
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
|
11
|
Mahapatra C, Thakkar R. In Silico Electrophysiological Investigation of Transient Receptor Potential Melastatin-4 Ion Channel Biophysics to Study Detrusor Overactivity. Int J Mol Sci 2024; 25:6875. [PMID: 38999984 PMCID: PMC11241520 DOI: 10.3390/ijms25136875] [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: 05/21/2024] [Revised: 06/18/2024] [Accepted: 06/20/2024] [Indexed: 07/14/2024] Open
Abstract
Enhanced electrical activity in detrusor smooth muscle (DSM) cells is a key factor in detrusor overactivity which causes overactive bladder pathological disorders. Transient receptor potential melastatin-4 (TRPM4) channels, which are calcium-activated cation channels, play a role in regulating DSM electrical activities. These channels likely contribute to depolarizing the DSM cell membrane, leading to bladder overactivity. Our research focuses on understanding TRPM4 channel function in the DSM cells of mice, using computational modeling. We aimed to create a detailed computational model of the TRPM4 channel based on existing electrophysiological data. We employed a modified Hodgkin-Huxley model with an incorporated TRP-like current to simulate action potential firing in response to current and synaptic stimulus inputs. Validation against experimental data showed close agreement with our simulations. Our model is the first to analyze the TRPM4 channel's role in DSM electrical activity, potentially revealing insights into bladder overactivity. In conclusion, TRPM4 channels are pivotal in regulating human DSM function, and TRPM4 channel inhibitors could be promising targets for treating overactive bladder.
Collapse
Affiliation(s)
- Chitaranjan Mahapatra
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA 94158, USA
- Paris Saclay Institute of Neuroscience, 91440 Saclay, France
| | - Ravindra Thakkar
- California Institute for Quantitative Biosciences, University of California Berkeley, Berkeley, CA 94720, USA
| |
Collapse
|
12
|
da Silva JF, Polk FD, Martin PE, Thai SH, Savu A, Gonzales M, Kath AM, Gee MT, Pires PW. Sex-specific mechanisms of cerebral microvascular BK Ca dysfunction in a mouse model of Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.06.06.543962. [PMID: 37333104 PMCID: PMC10274786 DOI: 10.1101/2023.06.06.543962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
BACKGROUND Cerebral microvascular dysfunction and nitro-oxidative stress are present in patients with Alzheimer's disease (AD) and may contribute to disease progression and severity. Large conductance Ca 2+ -activated K + channels (BK Ca ) play an essential role in vasodilatory responses and maintenance of myogenic tone in resistance arteries. BK Ca impairment can lead to microvascular dysfunction and hemodynamic deficits in the brain. We hypothesized that reduced BK Ca function in cerebral arteries mediates microvascular and neurovascular responses in the 5x-FAD model of AD. METHODS BK Ca activity in the cerebral microcirculation was assessed by patch clamp electrophysiology and pressure myography, in situ Ca 2+ sparks by spinning disk confocal microscopy, hemodynamics by laser speckle contrast imaging. Molecular and biochemical analyses were conducted by affinity-purification assays, qPCR, Western blots and immunofluorescence. RESULTS We observed that pial arteries from 5-6 months-old male and female 5x-FAD mice exhibited a hyper-contractile phenotype than wild-type (WT) littermates, which was linked to lower vascular BK Ca activity and reduced open probability. In males, BK Ca dysfunction is likely a consequence of an observed lower expression of the pore-forming subunit BKα and blunted frequency of Ca 2+ sparks, which are required for BK Ca activity. However, in females, impaired BK Ca function is, in part, a consequence of reversible nitro-oxidative changes in the BK α subunit, which reduces its open probability and regulation of vascular tone. We further show that BK Ca function is involved in neurovascular coupling in mice, and its dysfunction is linked to neurovascular dysfunction in the model. CONCLUSION These data highlight the central role played by BK Ca in cerebral microvascular and neurovascular regulation, as well as sex-dependent mechanisms underlying its dysfunction in a mouse model of AD.
Collapse
|
13
|
Davis MJ, Zawieja SD. Pacemaking in the lymphatic system. J Physiol 2024. [PMID: 38520402 DOI: 10.1113/jp284752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 02/08/2024] [Indexed: 03/25/2024] Open
Abstract
Lymphatic collecting vessels exhibit spontaneous phasic contractions that are critical for lymph propulsion and tissue fluid homeostasis. This rhythmic activity is driven by action potentials conducted across the lymphatic muscle cell (LMC) layer to produce entrained contractions. The contraction frequency of a lymphatic collecting vessel displays exquisite mechanosensitivity, with a dynamic range from <1 to >20 contractions per minute. A myogenic pacemaker mechanism intrinsic to the LMCs was initially postulated to account for pressure-dependent chronotropy. Further interrogation into the cellular constituents of the lymphatic vessel wall identified non-muscle cell populations that shared some characteristics with interstitial cells of Cajal, which have pacemaker functions in the gastrointestinal and lower urinary tracts, thus raising the possibility of a non-muscle cell pacemaker. However, recent genetic knockout studies in mice support LMCs and a myogenic origin of the pacemaker activity. LMCs exhibit stochastic, but pressure-sensitive, sarcoplasmic reticulum calcium release (puffs and waves) from IP3R1 receptors, which couple to the calcium-activated chloride channel Anoctamin 1, causing depolarisation. The resulting electrical activity integrates across the highly coupled lymphatic muscle electrical syncytia through connexin 45 to modulate diastolic depolarisation. However, multiple other cation channels may also contribute to the ionic pacemaking cycle. Upon reaching threshold, a voltage-gated calcium channel-dependent action potential fires, resulting in a nearly synchronous calcium global calcium flash within the LMC layer to drive an entrained contraction. This review summarizes the key ion channels potentially responsible for the pressure-dependent chronotropy of lymphatic collecting vessels and various mechanisms of IP3R1 regulation that could contribute to frequency tuning.
Collapse
Affiliation(s)
- Michael J Davis
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, MO, USA
| | - Scott D Zawieja
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, MO, USA
| |
Collapse
|
14
|
Hernandez-Hernandez G, O'Dwyer SC, Yang PC, Matsumoto C, Tieu M, Fong Z, Lewis TJ, Santana LF, Clancy CE. A computational model predicts sex-specific responses to calcium channel blockers in mammalian mesenteric vascular smooth muscle. eLife 2024; 12:RP90604. [PMID: 38335126 PMCID: PMC10942543 DOI: 10.7554/elife.90604] [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] [Indexed: 02/12/2024] Open
Abstract
The function of the smooth muscle cells lining the walls of mammalian systemic arteries and arterioles is to regulate the diameter of the vessels to control blood flow and blood pressure. Here, we describe an in silico model, which we call the 'Hernandez-Hernandez model', of electrical and Ca2+ signaling in arterial myocytes based on new experimental data indicating sex-specific differences in male and female arterial myocytes from murine resistance arteries. The model suggests the fundamental ionic mechanisms underlying membrane potential and intracellular Ca2+ signaling during the development of myogenic tone in arterial blood vessels. Although experimental data suggest that KV1.5 channel currents have similar amplitudes, kinetics, and voltage dependencies in male and female myocytes, simulations suggest that the KV1.5 current is the dominant current regulating membrane potential in male myocytes. In female cells, which have larger KV2.1 channel expression and longer time constants for activation than male myocytes, predictions from simulated female myocytes suggest that KV2.1 plays a primary role in the control of membrane potential. Over the physiological range of membrane potentials, the gating of a small number of voltage-gated K+ channels and L-type Ca2+ channels are predicted to drive sex-specific differences in intracellular Ca2+ and excitability. We also show that in an idealized computational model of a vessel, female arterial smooth muscle exhibits heightened sensitivity to commonly used Ca2+ channel blockers compared to male. In summary, we present a new model framework to investigate the potential sex-specific impact of antihypertensive drugs.
Collapse
Affiliation(s)
| | - Samantha C O'Dwyer
- Department of Physiology & Membrane Biology, University of California, DavisDavisUnited States
| | - Pei-Chi Yang
- Department of Physiology & Membrane Biology, University of California, DavisDavisUnited States
| | - Collin Matsumoto
- Department of Physiology & Membrane Biology, University of California, DavisDavisUnited States
| | - Mindy Tieu
- Department of Physiology & Membrane Biology, University of California, DavisDavisUnited States
| | - Zhihui Fong
- Department of Physiology & Membrane Biology, University of California, DavisDavisUnited States
| | - Timothy J Lewis
- Department of Mathematics, University of California, DavisDavisUnited States
| | - L Fernando Santana
- Department of Physiology & Membrane Biology, University of California, DavisDavisUnited States
| | - Colleen E Clancy
- Department of Physiology & Membrane Biology, University of California, DavisDavisUnited States
- Center for Precision Medicine and Data Sciences, University of California, DavisDavisUnited States
| |
Collapse
|
15
|
Longden TA, Lederer WJ. Electro-metabolic signaling. J Gen Physiol 2024; 156:e202313451. [PMID: 38197953 PMCID: PMC10783436 DOI: 10.1085/jgp.202313451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 10/27/2023] [Accepted: 12/14/2023] [Indexed: 01/11/2024] Open
Abstract
Precise matching of energy substrate delivery to local metabolic needs is essential for the health and function of all tissues. Here, we outline a mechanistic framework for understanding this critical process, which we refer to as electro-metabolic signaling (EMS). All tissues exhibit changes in metabolism over varying spatiotemporal scales and have widely varying energetic needs and reserves. We propose that across tissues, common signatures of elevated metabolism or increases in energy substrate usage that exceed key local thresholds rapidly engage mechanisms that generate hyperpolarizing electrical signals in capillaries that then relax contractile elements throughout the vasculature to quickly adjust blood flow to meet changing needs. The attendant increase in energy substrate delivery serves to meet local metabolic requirements and thus avoids a mismatch in supply and demand and prevents metabolic stress. We discuss in detail key examples of EMS that our laboratories have discovered in the brain and the heart, and we outline potential further EMS mechanisms operating in tissues such as skeletal muscle, pancreas, and kidney. We suggest that the energy imbalance evoked by EMS uncoupling may be central to cellular dysfunction from which the hallmarks of aging and metabolic diseases emerge and may lead to generalized organ failure states-such as diverse flavors of heart failure and dementia. Understanding and manipulating EMS may be key to preventing or reversing these dysfunctions.
Collapse
Affiliation(s)
- Thomas A. Longden
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
- Laboratory of Neurovascular Interactions, Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - W. Jonathan Lederer
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
- Laboratory of Molecular Cardiology, Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
16
|
Panerai RB, Davies A, Clough RH, Beishon LC, Robinson TG, Minhas JS. The effect of hypercapnia on the directional sensitivity of dynamic cerebral autoregulation and the influence of age and sex. J Cereb Blood Flow Metab 2024; 44:272-283. [PMID: 37747437 PMCID: PMC10993882 DOI: 10.1177/0271678x231203475] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 08/24/2023] [Accepted: 09/05/2023] [Indexed: 09/26/2023]
Abstract
The cerebral circulation responds differently to increases in mean arterial pressure (MAP), compared to reductions in MAP. We tested the hypothesis that this directional sensitivity is reduced by hypercapnia. Retrospective analysis of 104 healthy subjects (46 male (44%), age range 19-74 years), with five minute recordings of middle cerebral blood velocity (MCAv, transcranial Doppler), non-invasive MAP (Finometer) and end-tidal CO2 (capnography) at rest, during both poikilocapnia and hypercapnia (5% CO2 breathing in air) produced MCAv step responses allowing estimation of the classical Autoregulation Index (ARIORIG), and corresponding values for both positive (ARI+D) and negative (ARI-D) changes in MAP. Hypercapnia led to marked reductions in ARIORIG, ARI+D and ARI-D (p < 0.0001, all cases). Females had a lower value of ARIORIG compared to males (p = 0.030) at poikilocapnia (4.44 ± 1.74 vs 4.74 ± 1.48) and hypercapnia (2.44 ± 1.93 vs 3.33 ± 1.61). The strength of directional sensitivity (ARI+D-ARI-D) was not influenced by hypercapnia (p = 0.46), sex (p = 0.76) or age (p = 0.61). During poikilocapnia, ARI+D decreased with age in females (p = 0.027), but not in males. Directional sensitivity was not affected by hypercapnia, suggesting that its origins are more likely to be inherent to the mechanics of vascular smooth muscle than to myogenic pathways.
Collapse
Affiliation(s)
- Ronney B Panerai
- Department of Cardiovascular Sciences, University of Leicester, Robert Kilpatrick Clinical Sciences Building, Leicester, UK
- NIHR Leicester Biomedical Research Centre, BHF Cardiovascular Research Centre, Glenfield Hospital, Leicester, UK
| | - Aaron Davies
- Department of Cardiovascular Sciences, University of Leicester, Robert Kilpatrick Clinical Sciences Building, Leicester, UK
| | - Rebecca H Clough
- Department of Cardiovascular Sciences, University of Leicester, Robert Kilpatrick Clinical Sciences Building, Leicester, UK
| | - Lucy C Beishon
- Department of Cardiovascular Sciences, University of Leicester, Robert Kilpatrick Clinical Sciences Building, Leicester, UK
| | - Thompson G Robinson
- Department of Cardiovascular Sciences, University of Leicester, Robert Kilpatrick Clinical Sciences Building, Leicester, UK
- NIHR Leicester Biomedical Research Centre, BHF Cardiovascular Research Centre, Glenfield Hospital, Leicester, UK
| | - Jatinder S Minhas
- Department of Cardiovascular Sciences, University of Leicester, Robert Kilpatrick Clinical Sciences Building, Leicester, UK
- NIHR Leicester Biomedical Research Centre, BHF Cardiovascular Research Centre, Glenfield Hospital, Leicester, UK
| |
Collapse
|
17
|
Hernandez-Hernandez G, O’Dwyer SC, Matsumoto C, Tieu M, Fong Z, Yang PC, Lewis TJ, Fernando Santana L, Clancy CE. A computational model predicts sex-specific responses to calcium channel blockers in mammalian mesenteric vascular smooth muscle. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.06.24.546394. [PMID: 37425682 PMCID: PMC10327109 DOI: 10.1101/2023.06.24.546394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
The function of the smooth muscle cells lining the walls of mammalian systemic arteries and arterioles is to regulate the diameter of the vessels to control blood flow and blood pressure. Here, we describe an in-silico model, which we call the "Hernandez-Hernandez model", of electrical and C a 2+ signaling in arterial myocytes based on new experimental data indicating sex-specific differences in male and female arterial myocytes from murine resistance arteries. The model suggests the fundamental ionic mechanisms underlying membrane potential and intracellular C a 2+ signaling during the development of myogenic tone in arterial blood vessels. Although experimental data suggest that KV1.5 channel currents have similar amplitudes, kinetics, and voltage dependencies in male and female myocytes, simulations suggest that the KV1.5 current is the dominant current regulating membrane potential in male myocytes. In female cells, which have larger KV2.1 channel expression and longer time constants for activation than male myocytes, predictions from simulated female myocytes suggest that KV2.1 plays a primary role in the control of membrane potential. Over the physiological range of membrane potentials, the gating of a small number of voltage-gated K+ channels and L-type C a 2+ channels are predicted to drive sex-specific differences in intracellular C a 2+ and excitability. We also show that in an idealized computational model of a vessel, female arterial smooth muscle exhibits heightened sensitivity to commonly used C a 2+ channel blockers compared to male. In summary, we present a new model framework to investigate the potential sex-specific impact of anti-hypertensive drugs.
Collapse
Affiliation(s)
- Gonzalo Hernandez-Hernandez
- Department of Physiology & Membrane Biology, Center for Precision Medicine and Data Science, University of California School of Medicine, Davis, California, 95616
- Department of Mathematics, University of California, Davis, California, 95616
| | - Samantha C. O’Dwyer
- Department of Physiology & Membrane Biology, Center for Precision Medicine and Data Science, University of California School of Medicine, Davis, California, 95616
- Department of Mathematics, University of California, Davis, California, 95616
| | - Collin Matsumoto
- Department of Physiology & Membrane Biology, Center for Precision Medicine and Data Science, University of California School of Medicine, Davis, California, 95616
- Department of Mathematics, University of California, Davis, California, 95616
| | - Mindy Tieu
- Department of Physiology & Membrane Biology, Center for Precision Medicine and Data Science, University of California School of Medicine, Davis, California, 95616
- Department of Mathematics, University of California, Davis, California, 95616
| | - Zhihui Fong
- Department of Physiology & Membrane Biology, Center for Precision Medicine and Data Science, University of California School of Medicine, Davis, California, 95616
- Department of Mathematics, University of California, Davis, California, 95616
| | - Pei-Chi Yang
- Department of Physiology & Membrane Biology, Center for Precision Medicine and Data Science, University of California School of Medicine, Davis, California, 95616
- Department of Mathematics, University of California, Davis, California, 95616
| | - Timothy J. Lewis
- Department of Mathematics, University of California, Davis, California, 95616
| | | | - Colleen E. Clancy
- Department of Physiology & Membrane Biology, Center for Precision Medicine and Data Science, University of California School of Medicine, Davis, California, 95616
| |
Collapse
|
18
|
Sanders KM, Drumm BT, Cobine CA, Baker SA. Ca 2+ dynamics in interstitial cells: foundational mechanisms for the motor patterns in the gastrointestinal tract. Physiol Rev 2024; 104:329-398. [PMID: 37561138 PMCID: PMC11281822 DOI: 10.1152/physrev.00036.2022] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 06/29/2023] [Accepted: 08/06/2023] [Indexed: 08/11/2023] Open
Abstract
The gastrointestinal (GI) tract displays multiple motor patterns that move nutrients and wastes through the body. Smooth muscle cells (SMCs) provide the forces necessary for GI motility, but interstitial cells, electrically coupled to SMCs, tune SMC excitability, transduce inputs from enteric motor neurons, and generate pacemaker activity that underlies major motor patterns, such as peristalsis and segmentation. The interstitial cells regulating SMCs are interstitial cells of Cajal (ICC) and PDGF receptor (PDGFR)α+ cells. Together these cells form the SIP syncytium. ICC and PDGFRα+ cells express signature Ca2+-dependent conductances: ICC express Ca2+-activated Cl- channels, encoded by Ano1, that generate inward current, and PDGFRα+ cells express Ca2+-activated K+ channels, encoded by Kcnn3, that generate outward current. The open probabilities of interstitial cell conductances are controlled by Ca2+ release from the endoplasmic reticulum. The resulting Ca2+ transients occur spontaneously in a stochastic manner. Ca2+ transients in ICC induce spontaneous transient inward currents and spontaneous transient depolarizations (STDs). Neurotransmission increases or decreases Ca2+ transients, and the resulting depolarizing or hyperpolarizing responses conduct to other cells in the SIP syncytium. In pacemaker ICC, STDs activate voltage-dependent Ca2+ influx, which initiates a cluster of Ca2+ transients and sustains activation of ANO1 channels and depolarization during slow waves. Regulation of GI motility has traditionally been described as neurogenic and myogenic. Recent advances in understanding Ca2+ handling mechanisms in interstitial cells and how these mechanisms influence motor patterns of the GI tract suggest that the term "myogenic" should be replaced by the term "SIPgenic," as this review discusses.
Collapse
Affiliation(s)
- Kenton M Sanders
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada-Reno, Reno, Nevada, United States
| | - Bernard T Drumm
- Smooth Muscle Research Centre, Dundalk Institute of Technology, Dundalk, Ireland
| | - Caroline A Cobine
- Smooth Muscle Research Centre, Dundalk Institute of Technology, Dundalk, Ireland
| | - Salah A Baker
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada-Reno, Reno, Nevada, United States
| |
Collapse
|
19
|
Ameen AAM. Uterodilation effect of unripe fruit extract of Crataegus azarolus var. aronia L. on rat uterine smooth muscles. Prostaglandins Other Lipid Mediat 2023; 169:106783. [PMID: 37726053 DOI: 10.1016/j.prostaglandins.2023.106783] [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: 07/25/2023] [Revised: 09/14/2023] [Accepted: 09/16/2023] [Indexed: 09/21/2023]
Abstract
Crataegus azarolus var. aronia L. (C. aronia) is one of the most important medicinal plants used widely in folk medicine for the prevention of several diseases due to its content of several bioactive compounds like phenolic acid, aromatic amines, proanthocyanidins and flavonoids. This study investigated the uterodilation effect of methanol extract (ME) of C. aronia unripe fruit on the uterine smooth muscle in rats. The mechanism of action underlying the plant's extract was also screened. The unripe fruits were cleaned and extracted in methanol. The extract (1.9-4 mg/ml) was tested on rat uterine relaxation in calcium-free Kreb's solution and potassium chloride-induced uterine contraction. The plant extract was also studied in the presence of antagonists in separate experiments to determine the role of various ion channels and hyperpolarizing agents. The plant extract showed an uterodilation effect on the uterus, in which the ME produced a considerable relaxant effect. The results confirmed that the induced dilation was mediated mainly by the nitric oxide pathway and the activation of potassium channels with a limited role of the prostaglandin pathway and calcium channel activation. This in-vitro study provides the first scientific evidence of the claimed effect of C. aronia on uterine relaxation.
Collapse
|
20
|
Yogendran V, Mele L, Prysyazhna O, Budhram-Mahadeo VS. Vascular dysfunction caused by loss of Brn-3b/POU4F2 transcription factor in aortic vascular smooth muscle cells is linked to deregulation of calcium signalling pathways. Cell Death Dis 2023; 14:770. [PMID: 38007517 PMCID: PMC10676411 DOI: 10.1038/s41419-023-06306-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 09/14/2023] [Accepted: 11/07/2023] [Indexed: 11/27/2023]
Abstract
Phenotypic and functional changes in vascular smooth muscle cells (VSMCs) contribute significantly to cardiovascular diseases (CVD) but factors driving early adverse vascular changes are poorly understood. We report on novel and important roles for the Brn-3b/POU4F2 (Brn-3b) transcription factor (TF) in controlling VSMC integrity and function. Brn-3b protein is expressed in mouse aorta with localisation to VSMCs. Male Brn-3b knock-out (KO) aortas displayed extensive remodelling with increased extracellular matrix (ECM) deposition, elastin fibre disruption and small but consistent narrowing/coarctation in the descending aortas. RNA sequencing analysis showed that these effects were linked to deregulation of genes required for calcium (Ca2+) signalling, vascular contractility, sarco-endoplasmic reticulum (S/ER) stress responses and immune function in Brn-3b KO aortas and validation studies confirmed changes in Ca2+ signalling genes linked to increased intracellular Ca2+ and S/ER Ca2+ depletion [e.g. increased, Cacna1d Ca2+ channels; ryanodine receptor 2, (RyR2) and phospholamban (PLN) but reduced ATP2a1, encoding SERCA1 pump] and chaperone proteins, Hspb1, HspA8, DnaJa1 linked to increased S/ER stress, which also contributes to contractile dysfunction. Accordingly, vascular rings from Brn-3b KO aortas displayed attenuated contractility in response to KCl or phenylephrine (PE) while Brn-3b KO-derived VSMC displayed abnormal Ca2+ signalling following ATP stimulation. This data suggests that Brn-3b target genes are necessary to maintain vascular integrity /contractile function and deregulation upon loss of Brn-3b will contribute to contractile dysfunction linked to CVD.
Collapse
Affiliation(s)
- Vaishaali Yogendran
- Molecular Biology Development and Disease, UCL Institute of Cardiovascular Science, London, UK
| | - Laura Mele
- Molecular Biology Development and Disease, UCL Institute of Cardiovascular Science, London, UK
| | - Oleksandra Prysyazhna
- Clinical Pharmacology Centre, William Harvey Research Institute, Queen Mary University of London, London, UK
| | | |
Collapse
|
21
|
Akin EJ, Aoun J, Jimenez C, Mayne K, Baeck J, Young MD, Sullivan B, Sanders KM, Ward SM, Bulley S, Jaggar JH, Earley S, Greenwood IA, Leblanc N. ANO1, CaV1.2, and IP3R form a localized unit of EC-coupling in mouse pulmonary arterial smooth muscle. J Gen Physiol 2023; 155:e202213217. [PMID: 37702787 PMCID: PMC10499037 DOI: 10.1085/jgp.202213217] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 08/01/2023] [Accepted: 08/29/2023] [Indexed: 09/14/2023] Open
Abstract
Pulmonary arterial (PA) smooth muscle cells (PASMC) generate vascular tone in response to agonists coupled to Gq-protein receptor signaling. Such agonists stimulate oscillating calcium waves, the frequency of which drives the strength of contraction. These Ca2+ events are modulated by a variety of ion channels including voltage-gated calcium channels (CaV1.2), the Tmem16a or Anoctamin-1 (ANO1)-encoded calcium-activated chloride (CaCC) channel, and Ca2+ release from the sarcoplasmic reticulum through inositol-trisphosphate receptors (IP3R). Although these calcium events have been characterized, it is unclear how these calcium oscillations underly a sustained contraction in these muscle cells. We used smooth muscle-specific ablation of ANO1 and pharmacological tools to establish the role of ANO1, CaV1.2, and IP3R in the contractile and intracellular Ca2+ signaling properties of mouse PA smooth muscle expressing the Ca2+ biosensor GCaMP3 or GCaMP6. Pharmacological block or genetic ablation of ANO1 or inhibition of CaV1.2 or IP3R, or Ca2+ store depletion equally inhibited 5-HT-induced tone and intracellular Ca2+ waves. Coimmunoprecipitation experiments showed that an anti-ANO1 antibody was able to pull down both CaV1.2 and IP3R. Confocal and superresolution nanomicroscopy showed that ANO1 coassembles with both CaV1.2 and IP3R at or near the plasma membrane of PASMC from wild-type mice. We conclude that the stable 5-HT-induced PA contraction results from the integration of stochastic and localized Ca2+ events supported by a microenvironment comprising ANO1, CaV1.2, and IP3R. In this model, ANO1 and CaV1.2 would indirectly support cyclical Ca2+ release events from IP3R and propagation of intracellular Ca2+ waves.
Collapse
Affiliation(s)
- Elizabeth J. Akin
- Department of Pharmacology and Center of Biomedical Research Excellence (COBRE) for Molecular and Cellular Signal Transduction in the Cardiovascular System, Reno, NV, USA
| | - Joydeep Aoun
- Department of Pharmacology and Center of Biomedical Research Excellence (COBRE) for Molecular and Cellular Signal Transduction in the Cardiovascular System, Reno, NV, USA
| | - Connor Jimenez
- Department of Pharmacology and Center of Biomedical Research Excellence (COBRE) for Molecular and Cellular Signal Transduction in the Cardiovascular System, Reno, NV, USA
| | - Katie Mayne
- Department of Pharmacology and Center of Biomedical Research Excellence (COBRE) for Molecular and Cellular Signal Transduction in the Cardiovascular System, Reno, NV, USA
| | - Julius Baeck
- Department of Pharmacology and Center of Biomedical Research Excellence (COBRE) for Molecular and Cellular Signal Transduction in the Cardiovascular System, Reno, NV, USA
| | - Michael D. Young
- Department of Pharmacology and Center of Biomedical Research Excellence (COBRE) for Molecular and Cellular Signal Transduction in the Cardiovascular System, Reno, NV, USA
| | - Brennan Sullivan
- Department of Pharmacology and Center of Biomedical Research Excellence (COBRE) for Molecular and Cellular Signal Transduction in the Cardiovascular System, Reno, NV, USA
| | - Kenton M. Sanders
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV, USA
| | - Sean M. Ward
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV, USA
| | - Simon Bulley
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Jonathan H. Jaggar
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Scott Earley
- Department of Pharmacology and Center of Biomedical Research Excellence (COBRE) for Molecular and Cellular Signal Transduction in the Cardiovascular System, Reno, NV, USA
| | - Iain A. Greenwood
- Department of Vascular Pharmacology, Molecular and Clinical Science Research Institute, St. George’s University of London, London, UK
| | - Normand Leblanc
- Department of Pharmacology and Center of Biomedical Research Excellence (COBRE) for Molecular and Cellular Signal Transduction in the Cardiovascular System, Reno, NV, USA
| |
Collapse
|
22
|
Barenco-Marins TS, Seara FAC, Ponte CG, Nascimento JHM. Pulmonary Circulation Under Pressure: Pathophysiological and Therapeutic Implications of BK Channel. Cardiovasc Drugs Ther 2023:10.1007/s10557-023-07503-7. [PMID: 37624526 DOI: 10.1007/s10557-023-07503-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/10/2023] [Indexed: 08/26/2023]
Abstract
The large-conductance Ca2+-activated K+ (BK) channel is widely expressed in the pulmonary blood vessels and plays a significant role in regulating pulmonary vascular tonus. It opens under membrane depolarization, increased intracellular Ca+2 concentration, and chronic hypoxia, resulting in massive K+ efflux, membrane hyperpolarization, decreased L-type Ca+2 channel opening, and smooth muscle relaxation. Several reports have demonstrated an association between BK channel dysfunction and pulmonary hypertension (PH) development. Decreased BK channel subunit expression and impaired regulation by paracrine hormones result in decreased BK channel opening, increased pulmonary vascular resistance, and pulmonary arterial pressure being the cornerstone of PH. The resulting right ventricular pressure overload ultimately leads to ventricular remodeling and failure. Therefore, it is unsurprising that the BK channel has arisen as a potential target for treating PH. Recently, a series of selective, synthetic BK channel agonists have proven effective in attenuating the pathophysiological progression of PH without adverse effects in animal models.
Collapse
Affiliation(s)
- Thais S Barenco-Marins
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
- Programa de Pós-Graduação Em Cardiologia, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Fernando A C Seara
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
- Instituto de Ciências Biológicas E da Saúde, Universidade Federal Rural Do Rio de Janeiro, Seropédica, RJ, Brazil.
- Programa de Pós-Graduação Multicêntrico Em Ciências Fisiológicas, Sociedade Brasileira de Fisiologia, São Paulo, Brazil.
| | - Cristiano G Ponte
- Instituto Federal de Educação, Ciências e Tecnologia do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Jose H M Nascimento
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
- Programa de Pós-Graduação Em Cardiologia, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| |
Collapse
|
23
|
Niloy SI, Shen Y, Guo L, O'Rourke ST, Sun C. Loss of IP3R-BK Ca Coupling Is Involved in Vascular Remodeling in Spontaneously Hypertensive Rats. Int J Mol Sci 2023; 24:10903. [PMID: 37446080 DOI: 10.3390/ijms241310903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 06/23/2023] [Accepted: 06/27/2023] [Indexed: 07/15/2023] Open
Abstract
Mechanisms by which BKCa (large-conductance calcium-sensitive potassium) channels are involved in vascular remodeling in hypertension are not fully understood. Vascular smooth muscle cell (VSMC) proliferation and vascular morphology were compared between hypertensive and normotensive rats. BKCa channel activity, protein expression, and interaction with IP3R (inositol 1,4,5-trisphosphate receptor) were examined using patch clamp, Western blot analysis, and coimmunoprecipitation. On inside-out patches of VSMCs, the Ca2+-sensitivity and voltage-dependence of BKCa channels were similar between hypertensive and normotensive rats. In whole-cell patch clamp configuration, treatment of cells with the IP3R agonist, Adenophostin A (AdA), significantly increased BKCa channel currents in VSMCs of both strains of rats, suggesting IP3R-BKCa coupling; however, the AdA-induced increases in BKCa currents were attenuated in VSMCs of hypertensive rats, indicating possible IP3R-BKCa decoupling, causing BKCa dysfunction. Co-immunoprecipitation and Western blot analysis demonstrated that BKCa and IP3R proteins were associated together in VSMCs; however, the association of BKCa and IP3R proteins was dramatically reduced in VSMCs of hypertensive rats. Genetic disruption of IP3R-BKCa coupling using junctophilin-2 shRNA dramatically augmented Ang II-induced proliferation in VSMCs of normotensive rats. Subcutaneous infusion of NS1619, a BKCa opener, to reverse BKCa dysfunction caused by IP3R-BKCa decoupling significantly attenuated vascular hypertrophy in hypertensive rats. In summary, the data from this study demonstrate that loss of IP3R-BKCa coupling in VSMCs induces BKCa channel dysfunction, enhances VSMC proliferation, and thus, may contribute to vascular hypertrophy in hypertension.
Collapse
Affiliation(s)
- Sayeman Islam Niloy
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND 58105, USA
| | - Yue Shen
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND 58105, USA
| | - Lirong Guo
- School of Nursing, Jilin University, Changchun 130021, China
| | - Stephen T O'Rourke
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND 58105, USA
| | - Chengwen Sun
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND 58105, USA
| |
Collapse
|
24
|
Panerai RB, Barnes SC, Batterham AP, Robinson TG, Haunton VJ. Directional sensitivity of dynamic cerebral autoregulation during spontaneous fluctuations in arterial blood pressure at rest. J Cereb Blood Flow Metab 2023; 43:552-564. [PMID: 36420777 PMCID: PMC10063834 DOI: 10.1177/0271678x221142527] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Directional sensitivity, the more efficient response of cerebral autoregulation to increases, compared to decreases, in mean arterial pressure (MAP), has been demonstrated with repeated squat-stand maneuvers (SSM). In 43 healthy subjects (26 male, 23.1 ± 4.2 years old), five min. recordings of cerebral blood velocity (bilateral Doppler ultrasound), MAP (Finometer), end-tidal CO2 (capnograph), and heart rate (ECG) were obtained during sitting (SIT), standing (STA) and SSM. A new analytical procedure, based on autoregressive-moving average models, allowed distinct estimates of the autoregulation index (ARI) by separating the MAP signal into its positive (MAP+D) and negative (MAP-D) derivatives. ARI+D was higher than ARI-D (p < 0.0001), SIT: 5.61 ± 1.58 vs 4.31 ± 2.16; STA: 5.70 ± 1.24 vs 4.63 ± 1.92; SSM: 4.70 ± 1.11 vs 3.31 ± 1.53, but the difference ARI+D-ARI-D was not influenced by the condition. A bootstrap procedure determined the critical number of subjects needed to identify a significant difference between ARI+D and ARI-D, corresponding to 24, 37 and 38 subjects, respectively, for SSM, STA and SIT. Further investigations are needed on the influences of sex, aging and other phenotypical characteristics on the phenomenon of directional sensitivity of dynamic autoregulation.
Collapse
Affiliation(s)
- Ronney B Panerai
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK.,NIHR Leicester Biomedical Research Centre, BHF Cardiovascular Research Centre, Glenfield Hospital, Leicester, UK
| | - Sam C Barnes
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
| | - Angus P Batterham
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
| | - Thompson G Robinson
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK.,NIHR Leicester Biomedical Research Centre, BHF Cardiovascular Research Centre, Glenfield Hospital, Leicester, UK
| | - Victoria J Haunton
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK.,NIHR Leicester Biomedical Research Centre, BHF Cardiovascular Research Centre, Glenfield Hospital, Leicester, UK
| |
Collapse
|
25
|
Yang HM, Lee JE, Kim JY, You J, Kim J, Lee HS, Yoo HM, Kong MG, Han JK, Cho HJ, Park KW, Kang HJ, Koo BK, Park YB, Kim HS. Identification of cell-biologic mechanisms of coronary artery spasm and its ex vivo diagnosis using peripheral blood-derived iPSCs. Biomater Res 2023; 27:16. [PMID: 36803875 PMCID: PMC9938986 DOI: 10.1186/s40824-023-00345-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 01/25/2023] [Indexed: 02/20/2023] Open
Abstract
BACKGROUND Although vasospastic angina (VSA) is known to be caused by coronary artery spasm, no study has fully elucidated the exact underlying mechanism. Moreover, in order to confirm VSA, patients should undergo invasive coronary angiography with spasm provocation test. Herein, we investigated the pathophysiology of VSA using peripheral blood-derived induced pluripotent stem cells (iPSCs) and developed an ex vivo diagnostic method for VSA. METHODS AND RESULTS With 10 mL of peripheral blood from patients with VSA, we generated iPSCs and differentiated these iPSCs into target cells. As compared with vascular smooth muscle cells (VSMCs) differentiated from iPSCs of normal subjects with negative provocation test, VSA patient-specific iPSCs-derived VSMCs showed very strong contraction in response to stimulants. Moreover, VSA patient-specific VSMCs exhibited a significant increase in stimulation-induced intracellular calcium efflux (Changes in the relative fluorescence unit [ΔF/F]; Control group vs. VSA group, 2.89 ± 0.34 vs. 10.32 ± 0.51, p < 0.01), and exclusively induced a secondary or tertiary peak of calcium efflux, suggesting that those findings could be diagnostic cut-off values for VSA. The observed hyperreactivity of VSA patient-specific VSMCs were caused by the upregulation of sarco/endoplasmic reticulum Ca2+-ATPase 2a (SERCA2a) due to its enhanced small ubiquitin-related modifier (SUMO)ylation. This increased activity of SERCA2a was reversed by treatment with ginkgolic acid, an inhibitor of SUMOylated E1 molecules (pi/µg protein; VSA group vs. VSA + ginkgolic acid, 52.36 ± 0.71 vs. 31.93 ± 1.13, p < 0.01). CONCLUSIONS Our findings showed that abnormal calcium handling in sarco/endoplasmic reticulum could be induced by the enhanced SERCA2a activity in patients with VSA, leading to spasm. Such novel mechanisms of coronary artery spasm could be useful for drug development and diagnosis of VSA.
Collapse
Affiliation(s)
- Han-Mo Yang
- grid.412484.f0000 0001 0302 820XDepartment of Internal Medicine, Seoul National University Hospital, 101 Daekak-Ro, Chongno-Gu, Seoul, 03080 Korea ,National Research Laboratory for Stem Cell Niche, Seoul, Korea ,grid.412484.f0000 0001 0302 820XInnovative Research Institute for Cell Therapy, Seoul National University Hospital, Seoul, Korea
| | - Joo-Eun Lee
- National Research Laboratory for Stem Cell Niche, Seoul, Korea ,grid.412484.f0000 0001 0302 820XInnovative Research Institute for Cell Therapy, Seoul National University Hospital, Seoul, Korea
| | - Ju-Young Kim
- National Research Laboratory for Stem Cell Niche, Seoul, Korea ,grid.412484.f0000 0001 0302 820XInnovative Research Institute for Cell Therapy, Seoul National University Hospital, Seoul, Korea
| | - Jihye You
- National Research Laboratory for Stem Cell Niche, Seoul, Korea ,grid.412484.f0000 0001 0302 820XInnovative Research Institute for Cell Therapy, Seoul National University Hospital, Seoul, Korea
| | - Joonoh Kim
- National Research Laboratory for Stem Cell Niche, Seoul, Korea ,grid.412484.f0000 0001 0302 820XInnovative Research Institute for Cell Therapy, Seoul National University Hospital, Seoul, Korea
| | - Hak Seung Lee
- grid.412484.f0000 0001 0302 820XDepartment of Internal Medicine, Seoul National University Hospital, 101 Daekak-Ro, Chongno-Gu, Seoul, 03080 Korea ,National Research Laboratory for Stem Cell Niche, Seoul, Korea ,grid.412484.f0000 0001 0302 820XInnovative Research Institute for Cell Therapy, Seoul National University Hospital, Seoul, Korea
| | - Hee Min Yoo
- grid.410883.60000 0001 2301 0664Biometrology Group, Korea Research Institute of Standards and Science (KRISS), Daejeon, Korea
| | - Min Gyu Kong
- grid.412484.f0000 0001 0302 820XDepartment of Internal Medicine, Seoul National University Hospital, 101 Daekak-Ro, Chongno-Gu, Seoul, 03080 Korea ,grid.412678.e0000 0004 0634 1623Department of Internal Medicine, Soon Chun Hyang University Hospital, Bucheon, Korea
| | - Jung-Kyu Han
- grid.412484.f0000 0001 0302 820XDepartment of Internal Medicine, Seoul National University Hospital, 101 Daekak-Ro, Chongno-Gu, Seoul, 03080 Korea ,National Research Laboratory for Stem Cell Niche, Seoul, Korea ,grid.412484.f0000 0001 0302 820XInnovative Research Institute for Cell Therapy, Seoul National University Hospital, Seoul, Korea
| | - Hyun-Jai Cho
- grid.412484.f0000 0001 0302 820XDepartment of Internal Medicine, Seoul National University Hospital, 101 Daekak-Ro, Chongno-Gu, Seoul, 03080 Korea ,National Research Laboratory for Stem Cell Niche, Seoul, Korea ,grid.412484.f0000 0001 0302 820XInnovative Research Institute for Cell Therapy, Seoul National University Hospital, Seoul, Korea
| | - Kyung Woo Park
- grid.412484.f0000 0001 0302 820XDepartment of Internal Medicine, Seoul National University Hospital, 101 Daekak-Ro, Chongno-Gu, Seoul, 03080 Korea ,National Research Laboratory for Stem Cell Niche, Seoul, Korea ,grid.412484.f0000 0001 0302 820XInnovative Research Institute for Cell Therapy, Seoul National University Hospital, Seoul, Korea
| | - Hyun-Jae Kang
- grid.412484.f0000 0001 0302 820XDepartment of Internal Medicine, Seoul National University Hospital, 101 Daekak-Ro, Chongno-Gu, Seoul, 03080 Korea ,National Research Laboratory for Stem Cell Niche, Seoul, Korea ,grid.412484.f0000 0001 0302 820XInnovative Research Institute for Cell Therapy, Seoul National University Hospital, Seoul, Korea
| | - Bon-Kwon Koo
- grid.412484.f0000 0001 0302 820XDepartment of Internal Medicine, Seoul National University Hospital, 101 Daekak-Ro, Chongno-Gu, Seoul, 03080 Korea ,National Research Laboratory for Stem Cell Niche, Seoul, Korea ,grid.412484.f0000 0001 0302 820XInnovative Research Institute for Cell Therapy, Seoul National University Hospital, Seoul, Korea
| | - Young-Bae Park
- grid.412484.f0000 0001 0302 820XDepartment of Internal Medicine, Seoul National University Hospital, 101 Daekak-Ro, Chongno-Gu, Seoul, 03080 Korea ,National Research Laboratory for Stem Cell Niche, Seoul, Korea ,grid.412484.f0000 0001 0302 820XInnovative Research Institute for Cell Therapy, Seoul National University Hospital, Seoul, Korea
| | - Hyo-Soo Kim
- Department of Internal Medicine, Seoul National University Hospital, 101 Daekak-Ro, Chongno-Gu, Seoul, 03080, Korea. .,National Research Laboratory for Stem Cell Niche, Seoul, Korea. .,Innovative Research Institute for Cell Therapy, Seoul National University Hospital, Seoul, Korea. .,Molecular Medicine and Biopharmaceutical Sciences, Seoul National University, Seoul, 03080, Korea.
| |
Collapse
|
26
|
Abstract
Pericytes, attached to the surface of capillaries, play an important role in regulating local blood flow. Using optogenetic tools and genetically encoded reporters in conjunction with confocal and multiphoton imaging techniques, the 3D structure, anatomical organization, and physiology of pericytes have recently been the subject of detailed examination. This work has revealed novel functions of pericytes and morphological features such as tunneling nanotubes in brain and tunneling microtubes in heart. Here, we discuss the state of our current understanding of the roles of pericytes in blood flow control in brain and heart, where functions may differ due to the distinct spatiotemporal metabolic requirements of these tissues. We also outline the novel concept of electro-metabolic signaling, a universal mechanistic framework that links tissue metabolic state with blood flow regulation by pericytes and vascular smooth muscle cells, with capillary KATP and Kir2.1 channels as primary sensors. Finally, we present major unresolved questions and outline how they can be addressed.
Collapse
Affiliation(s)
- Thomas A Longden
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, USA; ,
- Laboratory of Neurovascular Interactions, Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Guiling Zhao
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, USA; ,
- Laboratory of Molecular Cardiology, Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Ashwini Hariharan
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, USA; ,
- Laboratory of Neurovascular Interactions, Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - W Jonathan Lederer
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, USA; ,
- Laboratory of Molecular Cardiology, Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
27
|
Oliveira N, Marcelino H, Azevedo R, Verde I. Effects of bisphenol A on human umbilical arteries. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:27670-27681. [PMID: 36385337 DOI: 10.1007/s11356-022-24069-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 11/03/2022] [Indexed: 06/16/2023]
Abstract
Bisphenol A (BPA) is an endocrine-disrupting chemical widely used in the plastics industry, including food container, toys, and medical equipment. We analyzed the effect of BPA in human umbilical artery contractility and expression of some proteins modulating this function, such as ionic channels and proteins involved in the cGMP pathway. Using standard organ bath technique, rings of human umbilical arteries without endothelium were contracted by 5-HT (1 μM) and histamine (10 μM) and the effect of different concentrations of BPA (1 nM-100 μM) was analyzed. The results showed that BPA is a vasodilator of these arteries in a concentration-dependent way. Besides, qPCR studies on human umbilical smooth muscle cells (HUSMC) allowed to analyze the effects of BPA on gene expression. Thus, 12-h exposition to BPA induced reduction of expression of L-type calcium channels (LTCC), alpha subunit of BKCa channels, and Kvβ1 and Kvβ3 from Kv channels. BPA also decreased the expression of soluble guanylate cyclase (sGC) and natriuretic peptide receptor type A (NPRA), meanwhile increasing that of PKG, proteins involved in vasodilation of human umbilical arteries (HUA) by cGMP. Further studies will be necessary to increase knowledge about the implications of these changes induced by BPA exposure.
Collapse
Affiliation(s)
- Nádia Oliveira
- Faculty of Health Sciences & Health Sciences Research Centre (CICS-UBI; Centro de Investigação em Ciências da Saúde), University of Beira Interior, Av. Infante D. Henrique S/N, 6200-506, Covilhã, Portugal
| | - Helena Marcelino
- Faculty of Health Sciences & Health Sciences Research Centre (CICS-UBI; Centro de Investigação em Ciências da Saúde), University of Beira Interior, Av. Infante D. Henrique S/N, 6200-506, Covilhã, Portugal
| | - Regina Azevedo
- Faculty of Health Sciences & Health Sciences Research Centre (CICS-UBI; Centro de Investigação em Ciências da Saúde), University of Beira Interior, Av. Infante D. Henrique S/N, 6200-506, Covilhã, Portugal
| | - Ignacio Verde
- Faculty of Health Sciences & Health Sciences Research Centre (CICS-UBI; Centro de Investigação em Ciências da Saúde), University of Beira Interior, Av. Infante D. Henrique S/N, 6200-506, Covilhã, Portugal.
| |
Collapse
|
28
|
Myosin light chain phosphorylation exhibits a gradient across the wall of cerebellar arteries under sustained ex vivo vascular tone. Sci Rep 2023; 13:909. [PMID: 36650375 PMCID: PMC9845333 DOI: 10.1038/s41598-023-28092-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 01/12/2023] [Indexed: 01/18/2023] Open
Abstract
Small blood vessel diseases are often associated with impaired regulation of vascular tone. The current understanding of resistance arteries often focuses on how a level of vascular tone is achieved in the acute phase, while less emphasis is placed on mechanisms that maintain vascular tone. In this study, cannulated rat superior cerebellar arteries (SCA) developed spontaneous myogenic tone and showed a marked and sustained constriction in the presence of diluted serum (10%), a stimulus relevant to cerebrovascular disease. Both phosphorylated myosin light chain (MLC-p) and smooth muscle alpha actin (SM-α-actin) aligned with phalloidin-stained actin filaments in the vessel wall, while exhibiting a 'high to low' gradient across the layers of vascular smooth muscle cells (VSMC), peaking in the outer layer. The MLC-p distribution profile shifted towards the adventitia in serum treated vessels, while removal of the serum reversed it. Furthermore, a positive correlation between the MLC-p signal and vessel wall tension was also evident. The gradients of phosphorylated MLC and SM-α-actin are consistent with a spatial regulation of the myosin-actin apparatus in the vessel wall during the maintenance of vascular tone. Further, the changing profiles of MLC-p and SM-α-actin are consistent with SCA vasoconstriction being accompanied by VSMC cytoskeletal reorganization.
Collapse
|
29
|
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: 2] [Impact Index Per Article: 1.0] [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
|
30
|
Cao M, Ong MTY, Yung PSH, Tuan RS, Jiang Y. Role of synovial lymphatic function in osteoarthritis. Osteoarthritis Cartilage 2022; 30:1186-1197. [PMID: 35487439 DOI: 10.1016/j.joca.2022.04.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 04/01/2022] [Accepted: 04/20/2022] [Indexed: 02/02/2023]
Abstract
BACKGROUND Osteoarthritis (OA) affects the entire joint, initially with a low degree of inflammation. Synovitis is correlated with the severity of OA clinical symptoms and cartilage degradation. The synovial lymphatic system (SLS) plays a prominent role in clearing macromolecules within the joint, including the pro-inflammatory cytokines in arthritic status. Scattered evidence shows that impaired SLS drainage function leads to the accumulation of inflammatory factors in the joint and aggravates the progression of OA, and the role of SLS function in OA is less studied. DESIGN This review summarizes the current understanding of synovial lymphatic function in OA progression and potential regulatory pathways and aims to provide a framework of knowledge for the development of OA treatments targeting lymphatic structure and functions. RESULTS SLS locates in the subintima layer of the synovium and consists of lymphatic capillaries and lymphatic collecting vessels. Vascular endothelial growth factor C (VEGF-C) is the most critical regulating factor of lymphatic endothelial cells (LECs) and SLS. Nitric oxide production-induced impairment of lymphatic muscle cells (LMCs) and contractile function may attribute to drainage dysfunction. Preclinical evidence suggests that promoting lymphatic drainage may help restore intra-articular homeostasis to attenuate the progression of OA. CONCLUSION SLS is actively involved in the homeostatic maintenance of the joint. Understanding the drainage function of the SLS at different stages of OA development is essential for further design of therapies targeting the function of these vessels.
Collapse
Affiliation(s)
- M Cao
- Department of Orthopaedics & Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - M T Y Ong
- Department of Orthopaedics & Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - P S H Yung
- Department of Orthopaedics & Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Institute for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - R S Tuan
- Institute for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Y Jiang
- Department of Orthopaedics & Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Institute for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China.
| |
Collapse
|
31
|
A molecular complex of Ca v1.2/CaMKK2/CaMK1a in caveolae is responsible for vascular remodeling via excitation-transcription coupling. Proc Natl Acad Sci U S A 2022; 119:e2117435119. [PMID: 35412911 PMCID: PMC9169798 DOI: 10.1073/pnas.2117435119] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Excitation–transcription (E-T) coupling can initiate and modulate essential physiological or pathological responses in cells, such as neurons and cardiac myocytes. Although vascular myocytes also exhibit E-T coupling in response to membrane depolarization, the underlying molecular mechanisms are unknown. Our study reveals that E-T coupling in vascular myocytes converts intracellular Ca2+ signals into selective gene transcription related to chemotaxis, leukocyte adhesion, and inflammation. Our discovery identifies a mechanism for vascular remodeling as an adaptation to increased circumferential stretch. Elevation of intracellular Ca2+ concentration ([Ca2+]i) activates Ca2+/calmodulin-dependent kinases (CaMK) and promotes gene transcription. This signaling pathway is referred to as excitation–transcription (E-T) coupling. Although vascular myocytes can exhibit E-T coupling, the molecular mechanisms and physiological/pathological roles are unknown. Multiscale analysis spanning from single molecules to whole organisms has revealed essential steps in mouse vascular myocyte E-T coupling. Upon a depolarizing stimulus, Ca2+ influx through Cav1.2 voltage-dependent Ca2+ channels activates CaMKK2 and CaMK1a, resulting in intranuclear CREB phosphorylation. Within caveolae, the formation of a molecular complex of Cav1.2/CaMKK2/CaMK1a is promoted in vascular myocytes. Live imaging using a genetically encoded Ca2+ indicator revealed direct activation of CaMKK2 by Ca2+ influx through Cav1.2 localized to caveolae. CaMK1a is phosphorylated by CaMKK2 at caveolae and translocated to the nucleus upon membrane depolarization. In addition, sustained depolarization of a mesenteric artery preparation induced genes related to chemotaxis, leukocyte adhesion, and inflammation, and these changes were reversed by inhibitors of Cav1.2, CaMKK2, and CaMK, or disruption of caveolae. In the context of pathophysiology, when the mesenteric artery was loaded by high pressure in vivo, we observed CREB phosphorylation in myocytes, macrophage accumulation at adventitia, and an increase in thickness and cross-sectional area of the tunica media. These changes were reduced in caveolin1-knockout mice or in mice treated with the CaMKK2 inhibitor STO609. In summary, E-T coupling depends on Cav1.2/CaMKK2/CaMK1a localized to caveolae, and this complex converts [Ca2+]i changes into gene transcription. This ultimately leads to macrophage accumulation and media remodeling for adaptation to increased circumferential stretch.
Collapse
|
32
|
Salazar-Enciso R, Guerrero-Hernández A, Gómez AM, Benitah JP, Rueda A. Aldosterone-Induced Sarco/Endoplasmic Reticulum Ca2+ Pump Upregulation Counterbalances Cav1.2-Mediated Ca2+ Influx in Mesenteric Arteries. Front Physiol 2022; 13:834220. [PMID: 35360237 PMCID: PMC8963271 DOI: 10.3389/fphys.2022.834220] [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: 12/13/2021] [Accepted: 02/08/2022] [Indexed: 11/26/2022] Open
Abstract
In mesenteric arteries (MAs), aldosterone (ALDO) binds to the endogenous mineralocorticoid receptor (MR) and increases the expression of the voltage-gated L-type Cav1.2 channel, an essential ion channel for vascular contraction, sarcoplasmic reticulum (SR) Ca2+ store refilling, and Ca2+ spark generation. In mesenteric artery smooth muscle cells (MASMCs), Ca2+ influx through Cav1.2 is the indirect mechanism for triggering Ca2+ sparks. This process is facilitated by plasma membrane-sarcoplasmic reticulum (PM-SR) nanojunctions that drive Ca2+ from the extracellular space into the SR via Sarco/Endoplasmic Reticulum Ca2+ (SERCA) pump. Ca2+ sparks produced by clusters of Ryanodine receptors (RyRs) at PM-SR nanodomains, decrease contractility by activating large-conductance Ca2+-activated K+ channels (BKCa channels), which generate spontaneous transient outward currents (STOCs). Altogether, Cav1.2, SERCA pump, RyRs, and BKCa channels work as a functional unit at the PM-SR nanodomain, regulating intracellular Ca2+ and vascular function. However, the effect of the ALDO/MR signaling pathway on this functional unit has not been completely explored. Our results show that short-term exposure to ALDO (10 nM, 24 h) increased the expression of Cav1.2 in rat MAs. The depolarization-induced Ca2+ entry increased SR Ca2+ load, and the frequencies of both Ca2+ sparks and STOCs, while [Ca2+]cyt and vasoconstriction remained unaltered in Aldo-treated MAs. ALDO treatment significantly increased the mRNA and protein expression levels of the SERCA pump, which counterbalanced the augmented Cav1.2-mediated Ca2+ influx at the PM-SR nanodomain, increasing SR Ca2+ content, Ca2+ spark and STOC frequencies, and opposing to hyperpolarization-induced vasoconstriction while enhancing Acetylcholine-mediated vasorelaxation. This work provides novel evidence for short-term ALDO-induced upregulation of the functional unit comprising Cav1.2, SERCA2 pump, RyRs, and BKCa channels; in which the SERCA pump buffers ALDO-induced upregulation of Ca2+ entry at the superficial SR-PM nanodomain of MASMCs, preventing ALDO-triggered depolarization-induced vasoconstriction and enhancing vasodilation. Pathological conditions that lead to SERCA pump downregulation, for instance, chronic exposure to ALDO, might favor the development of ALDO/MR-mediated augmented vasoconstriction of mesenteric arteries.
Collapse
Affiliation(s)
- Rogelio Salazar-Enciso
- Departamento de Bioquímica, Centro de Investigación y de Estudios Avanzados del IPN, Mexico City, Mexico
- Signaling and Cardiovascular Pathophysiology - UMR-S 1180, Inserm, Université Paris-Saclay, Châtenay-Malabry, France
| | - Agustín Guerrero-Hernández
- Departamento de Bioquímica, Centro de Investigación y de Estudios Avanzados del IPN, Mexico City, Mexico
| | - Ana M. Gómez
- Signaling and Cardiovascular Pathophysiology - UMR-S 1180, Inserm, Université Paris-Saclay, Châtenay-Malabry, France
| | - Jean-Pierre Benitah
- Signaling and Cardiovascular Pathophysiology - UMR-S 1180, Inserm, Université Paris-Saclay, Châtenay-Malabry, France
| | - Angélica Rueda
- Departamento de Bioquímica, Centro de Investigación y de Estudios Avanzados del IPN, Mexico City, Mexico
- *Correspondence: Angélica Rueda,
| |
Collapse
|
33
|
MacKay CE, Floen M, Leo MD, Hasan R, Garrud TAC, Fernández-Peña C, Singh P, Malik KU, Jaggar JH. A plasma membrane-localized polycystin-1/polycystin-2 complex in endothelial cells elicits vasodilation. eLife 2022; 11:e74765. [PMID: 35229718 PMCID: PMC8933003 DOI: 10.7554/elife.74765] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 02/25/2022] [Indexed: 11/25/2022] Open
Abstract
Polycystin-1 (PC-1, PKD1), a receptor-like protein expressed by the Pkd1 gene, is present in a wide variety of cell types, but its cellular location, signaling mechanisms, and physiological functions are poorly understood. Here, by studying tamoxifen-inducible, endothelial cell (EC)-specific Pkd1 knockout (Pkd1 ecKO) mice, we show that flow activates PC-1-mediated, Ca2+-dependent cation currents in ECs. EC-specific PC-1 knockout attenuates flow-mediated arterial hyperpolarization and vasodilation. PC-1-dependent vasodilation occurs over the entire functional shear stress range and via the activation of endothelial nitric oxide synthase (eNOS) and intermediate (IK)- and small (SK)-conductance Ca2+-activated K+ channels. EC-specific PC-1 knockout increases systemic blood pressure without altering kidney anatomy. PC-1 coimmunoprecipitates with polycystin-2 (PC-2, PKD2), a TRP polycystin channel, and clusters of both proteins locate in nanoscale proximity in the EC plasma membrane. Knockout of either PC-1 or PC-2 (Pkd2 ecKO mice) abolishes surface clusters of both PC-1 and PC-2 in ECs. Single knockout of PC-1 or PC-2 or double knockout of PC-1 and PC-2 (Pkd1/Pkd2 ecKO mice) similarly attenuates flow-mediated vasodilation. Flow stimulates nonselective cation currents in ECs that are similarly inhibited by either PC-1 or PC-2 knockout or by interference peptides corresponding to the C-terminus coiled-coil domains present in PC-1 or PC-2. In summary, we show that PC-1 regulates arterial contractility through the formation of an interdependent signaling complex with PC-2 in ECs. Flow stimulates PC-1/PC-2 clusters in the EC plasma membrane, leading to eNOS, IK channel, and SK channel activation, vasodilation, and a reduction in blood pressure.
Collapse
Affiliation(s)
- Charles E MacKay
- Department of Physiology, University of Tennessee Health Science CenterMemphisUnited States
| | - Miranda Floen
- Department of Physiology, University of Tennessee Health Science CenterMemphisUnited States
| | - M Dennis Leo
- Department of Physiology, University of Tennessee Health Science CenterMemphisUnited States
| | - Raquibul Hasan
- Department of Physiology, University of Tennessee Health Science CenterMemphisUnited States
| | - Tessa AC Garrud
- Department of Physiology, University of Tennessee Health Science CenterMemphisUnited States
| | - Carlos Fernández-Peña
- Department of Physiology, University of Tennessee Health Science CenterMemphisUnited States
| | - Purnima Singh
- Department of Physiology, University of Tennessee Health Science CenterMemphisUnited States
| | - Kafait U Malik
- Department of Physiology, University of Tennessee Health Science CenterMemphisUnited States
| | - Jonathan H Jaggar
- Department of Physiology, University of Tennessee Health Science CenterMemphisUnited States
| |
Collapse
|
34
|
Gil D, Diercks BP, Guse AH, Dupont G. Three-Dimensional Model of Sub-Plasmalemmal Ca2+ Microdomains Evoked by T Cell Receptor/CD3 Complex Stimulation. Front Mol Biosci 2022; 9:811145. [PMID: 35281279 PMCID: PMC8906516 DOI: 10.3389/fmolb.2022.811145] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 01/24/2022] [Indexed: 12/31/2022] Open
Abstract
Ca2+ signalling plays an essential role in T cell activation, which is a key step to start an adaptive immune response. During the transition from a quiescent to a fully activated state, Ca2+ microdomains of reduced spatial and temporal extents develop in the junctions between the plasma membrane and the endoplasmic reticulum (ER). These microdomains rely on Ca2+ entry from the extracellular medium, via the ORAI1/STIM1/STIM2 system that mediates store operated Ca2+ entry Store operated calcium entry. The mechanism leading to local store depletion and subsequent Ca2+ entry depends on the activation state of the cells. The initial, smaller microdomains are triggered by D-myo-inositol 1,4,5-trisphosphate (IP3) signalling in response to T cell adhesion. T cell receptor (TCR)/CD3 stimulation then initiates nicotinic acid adenine dinucleotide phosphate signalling, which activates ryanodine receptors (RYR). We have recently developed a mathematical model to elucidate the spatiotemporal Ca2+ dynamics of the microdomains triggered by IP3 signalling in response to T cell adhesion (Gil et al., 2021). This reaction-diffusion model describes the evolution of the cytosolic and endoplasmic reticulum Ca2+ concentrations in a three-dimensional ER-PM junction and was solved using COMSOL Multiphysics. Modelling predicted that adhesion-dependent microdomains result from the concerted activity of IP3 receptors and pre-formed ORAI1-STIM2 complexes. In the present study, we extend this model to include the role of RYRs rapidly after TCR/CD3 stimulation. The involvement of STIM1, which has a lower KD for Ca2+ than STIM2, is also considered. Detailed 3D spatio-temporal simulations show that these Ca2+ microdomains rely on the concerted opening of ∼7 RYRs that are simultaneously active in response to the increase in NAADP induced by T cell stimulation. Opening of these RYRs provoke a local depletion of ER Ca2+ that triggers Ca2+ flux through the ORAI1 channels. Simulations predict that RYRs are most probably located around the junction and that the increase in junctional Ca2+ concentration results from the combination between diffusion of Ca2+ released through the RYRs and Ca2+ entry through ORAI1 in the junction. The computational model moreover provides a tool allowing to investigate how Ca2+ microdomains occur, extend and interact in various states of T cell activation.
Collapse
Affiliation(s)
- Diana Gil
- The Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Björn-Philipp Diercks
- The Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Andreas H. Guse
- The Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Geneviève Dupont
- Unit of Theoretical Chronobiology, Faculté des Sciences CP231, Université Libre de Bruxelles (ULB), Brussels, Belgium
- *Correspondence: Geneviève Dupont,
| |
Collapse
|
35
|
Imaizumi Y. Reciprocal Relationship between Ca 2+ Signaling and Ca 2+-Gated Ion Channels as a Potential Target for Drug Discovery. Biol Pharm Bull 2022; 45:1-18. [PMID: 34980771 DOI: 10.1248/bpb.b21-00896] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Cellular Ca2+ signaling functions as one of the most common second messengers of various signal transduction pathways in cells and mediates a number of physiological roles in a cell-type dependent manner. Ca2+ signaling also regulates more general and fundamental cellular activities, including cell proliferation and apoptosis. Among ion channels, Ca2+-permeable channels in the plasma membrane as well as endo- and sarcoplasmic reticulum membranes play important roles in Ca2+ signaling by directly contributing to the influx of Ca2+ from extracellular spaces or its release from storage sites, respectively. Furthermore, Ca2+-gated ion channels in the plasma membrane often crosstalk reciprocally with Ca2+ signals and are central to the regulation of cellular functions. This review focuses on the physiological and pharmacological impact of i) Ca2+-gated ion channels as an apparatus for the conversion of cellular Ca2+ signals to intercellularly propagative electrical signals and ii) the opposite feedback regulation of Ca2+ signaling by Ca2+-gated ion channel activities in excitable and non-excitable cells.
Collapse
Affiliation(s)
- Yuji Imaizumi
- Department of Molecular and Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University
| |
Collapse
|
36
|
Johnson S, Pleshinger DJ, Jalkh J, Ijaz Z, Annamdevula N, Britain AL, Francis CM, Deshpande D, Leavesley SJ, Rich TC. Measurement of agonist-induced Ca 2+ signals in human airway smooth muscle cells using excitation scan-based hyperspectral imaging and image analysis approaches. PROCEEDINGS OF SPIE--THE INTERNATIONAL SOCIETY FOR OPTICAL ENGINEERING 2022; 11964:119640J. [PMID: 35755606 PMCID: PMC9215168 DOI: 10.1117/12.2608276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
Ca2+ and cAMP are ubiquitous second messengers known to differentially regulate a variety of cellular functions over a wide range of timescales. Studies from a variety of groups support the hypothesis that these signals can be localized to discrete locations within cells, and that this subcellular localization is a critical component of signaling specificity. However, to date, it has been difficult to track second messenger signals at multiple locations. To overcome this limitation, we utilized excitation scan-based hyperspectral imaging approaches to track second messenger signals as well as labeled cellular structures and/or proteins in the same cell. We have previously reported that hyperspectral imaging techniques improve the signal-to-noise ratios of both fluorescence measurements, and are thus well suited for the measurement of localized Ca2+ signals. We investigated the spatial spread and intensities of agonist-induced Ca2+ signals in primary human airway smooth muscle cells (HASMCs) using the Ca2+ indicator Cal520. We measured responses triggered by three agonists, carbachol, histamine, and chloroquine. We utilized custom software coded in MATLAB and Python to assess agonist induced changes in Ca2+ levels. Software algorithms removed the background and applied correction coefficients to spectral data prior to linear unmixing, spatial and temporal filtering, adaptive thresholding, and automated region of interest (ROI) detection. All three agonists triggered transient Ca2+ responses that were spatially and temporally complex. We are currently analyzing differences in both ROI area and intensity distributions triggered by these agonists. This work was supported by NIH awards P01HL066299, K25HL136869, and R01HL137030 and NSF award MRI1725937.
Collapse
Affiliation(s)
| | - D J Pleshinger
- Pharmacology, University of South Alabama, Mobile, AL 36688
| | - Josephine Jalkh
- Biomedical Sciences, University of South Alabama, Mobile, AL 36688
| | - Zara Ijaz
- Pharmacology, University of South Alabama, Mobile, AL 36688
| | | | | | - C Michael Francis
- Physiology and Cell Biology, University of South Alabama, Mobile, AL 36688
| | - Deepak Deshpande
- Center for Translational Medicine, Thomas Jefferson University, Philadelphia, PA 19107
| | - Silas J Leavesley
- Pharmacology, University of South Alabama, Mobile, AL 36688
- Chemical and Biomolecular Engineering, University of South Alabama, Mobile, AL 36688
| | - Thomas C Rich
- Pharmacology, University of South Alabama, Mobile, AL 36688
| |
Collapse
|
37
|
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: 8.3] [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
|
38
|
Augmented K Ca2.3 Channel Feedback Regulation of Oxytocin Stimulated Uterine Strips from Nonpregnant Mice. Int J Mol Sci 2021; 22:ijms222413585. [PMID: 34948381 PMCID: PMC8709448 DOI: 10.3390/ijms222413585] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 12/07/2021] [Accepted: 12/11/2021] [Indexed: 11/16/2022] Open
Abstract
Uterine contractions prior to 37 weeks gestation can result in preterm labor with significant risk to the infant. Current tocolytic therapies aimed at suppressing premature uterine contractions are largely ineffective and cause serious side effects. Calcium (Ca2+) dependent contractions of uterine smooth muscle are physiologically limited by the opening of membrane potassium (K+) channels. Exploiting such inherent negative feedback mechanisms may offer new strategies to delay labor and reduce risk. Positive modulation of small conductance Ca2+-activated K+ (KCa2.3) channels with cyclohexyl-[2-(3,5-dimethyl-pyrazol-1-yl)-6-methyl-pyrimidin-4-yl]-amine (CyPPA), effectively decreases uterine contractions. This study investigates whether the receptor agonist oxytocin might solicit KCa2.3 channel feedback that facilitates CyPPA suppression of uterine contractions. Using isometric force myography, we found that spontaneous phasic contractions of myometrial tissue from nonpregnant mice were suppressed by CyPPA and, in the presence of CyPPA, oxytocin failed to augment contractions. In tissues exposed to oxytocin, depletion of internal Ca2+ stores with cyclopiazonic acid (CPA) impaired CyPPA relaxation, whereas blockade of nonselective cation channels (NSCC) using gadolinium (Gd3+) had no significant effect. Immunofluorescence revealed close proximity of KCa2.3 channels and ER inositol trisphosphate receptors (IP3Rs) within myometrial smooth muscle cells. The findings suggest internal Ca2+ stores play a role in KCa2.3-dependent feedback control of uterine contraction and offer new insights for tocolytic therapies.
Collapse
|
39
|
González-Cota AL, Santana-Calvo C, Servín-Vences R, Orta G, Balderas E. Regulatory mechanisms of mitochondrial BK Ca channels. Channels (Austin) 2021; 15:424-437. [PMID: 33955332 PMCID: PMC8117780 DOI: 10.1080/19336950.2021.1919463] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 04/05/2021] [Accepted: 04/05/2021] [Indexed: 02/06/2023] Open
Abstract
The mitochondrial BKCa channel (mitoBKCa) is a splice variant of plasma membrane BKCa (Maxi-K, BKCa, Slo1, KCa1.1). While a high-resolution structure of mitoBKCa is not available yet, functional and structural studies of the plasma membrane BKCa have provided important clues on the gating of the channel by voltage and Ca2+, as well as the interaction with auxiliary subunits. To date, we know that the control of expression of mitoBKCa, targeting and voltage-sensitivity strongly depends on its association with its regulatory β1-subunit, which overall participate in the control of mitochondrial Ca2+-overload in cardiac myocytes. Moreover, novel regulatory mechanisms of mitoBKCa such as β-subunits and amyloid-β have recently been proposed. However, major basic questions including how the regulatory BKCa-β1-subunit reaches mitochondria and the mechanism through which amyloid-β impairs mitoBKCa channel function remain to be addressed.
Collapse
Affiliation(s)
- Ana L. González-Cota
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, UNAM. Av. Universidad 2001, Cuernavaca, Morelos, México
| | - Carmen Santana-Calvo
- Instituto Gulbenkian de Ciência. Rua da Quinta Grande 6, Oeiras, Portugal
- Instituto de Tecnologia Química e Biológica, Universida de Nova de Lisboa. Av. da República, Oeiras, Portugal
| | - Rocío Servín-Vences
- Department of Neuroscience, The Scripps Research Institute. 10550 North Torrey Pines Road, La Jolla, CA, USA
| | - Gerardo Orta
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, UNAM. Av. Universidad 2001, Cuernavaca, Morelos, México
| | - Enrique Balderas
- Nora Eccles Harrison Cardiovascular Research & Training Institute, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
40
|
Alleyne J, Dopico AM. Alcohol Use Disorders and Their Harmful Effects on the Contractility of Skeletal, Cardiac and Smooth Muscles. ADVANCES IN DRUG AND ALCOHOL RESEARCH 2021; 1:10011. [PMID: 35169771 PMCID: PMC8843239 DOI: 10.3389/adar.2021.10011] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 09/21/2021] [Indexed: 06/14/2023]
Abstract
Alcohol misuse has deleterious effects on personal health, family, societal units, and global economies. Moreover, alcohol misuse usually leads to several diseases and conditions, including alcoholism, which is a chronic condition and a form of addiction. Alcohol misuse, whether as acute intoxication or alcoholism, adversely affects skeletal, cardiac and/or smooth muscle contraction. Ethanol (ethyl alcohol) is the main effector of alcohol-induced dysregulation of muscle contractility, regardless of alcoholic beverage type or the ethanol metabolite (with acetaldehyde being a notable exception). Ethanol, however, is a simple and "promiscuous" ligand that affects many targets to mediate a single biological effect. In this review, we firstly summarize the processes of excitation-contraction coupling and calcium homeostasis which are critical for the regulation of contractility in all muscle types. Secondly, we present the effects of acute and chronic alcohol exposure on the contractility of skeletal, cardiac, and vascular/ nonvascular smooth muscles. Distinctions are made between in vivo and in vitro experiments, intoxicating vs. sub-intoxicating ethanol levels, and human subjects vs. animal models. The differential effects of alcohol on biological sexes are also examined. Lastly, we show that alcohol-mediated disruption of muscle contractility, involves a wide variety of molecular players, including contractile proteins, their regulatory factors, membrane ion channels and pumps, and several signaling molecules. Clear identification of these molecular players constitutes a first step for a rationale design of pharmacotherapeutics to prevent, ameliorate and/or reverse the negative effects of alcohol on muscle contractility.
Collapse
Affiliation(s)
| | - Alex M. Dopico
- Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN, United States
| |
Collapse
|
41
|
Rahman F, Muthaiah N, Kumaramanickavel G. Current concepts and molecular mechanisms in pharmacogenetics of essential hypertension. Indian J Pharmacol 2021; 53:301-309. [PMID: 34414909 PMCID: PMC8411967 DOI: 10.4103/ijp.ijp_593_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Hypertension is a leading age-related disease in our society and if left untreated, leads to fatal cardiovascular complications. The prevalence of hypertension has increased and becomes a significant global health economic burden, particularly in lower-income societies. Many loci associated with blood pressure and hypertension have been reported by genome-wide association studies that provided potential targets for pharmacotherapy. Pharmacogenetic research had shown interindividual variations in drug efficacy, safety, and tolerability. This could be due to genetic polymorphisms in the pharmacokinetics (genes involved in a transporter, plasma protein binding, and metabolism) or pharmacodynamic pathway (receptors, ion channels, enzymes). Pharmacogenetics promises great hope toward targeted therapy, but challenges remain in implementing pharmacogenetic aided antihypertensive therapy in clinical practice. Using various databases, we analyzed the underlying mechanisms between the candidate gene polymorphisms and antihypertensive drug interactions and the challenges of implementing precision medicine. We review the emergence of pharmacogenetics and its relevance to clinical pharmacological practice.
Collapse
Affiliation(s)
- Farhana Rahman
- Department of Pharmacology, Sree Balaji Medical College and Hospital, Bharat University, Chennai, Tamil Nadu, India
| | - Nagasundaram Muthaiah
- Department of Pharmacology, Sree Balaji Medical College and Hospital, Bharat University, Chennai, Tamil Nadu, India
| | - Govindasamy Kumaramanickavel
- Genomic Research Centre, Sree Balaji Medical College and Hospital, Bharat University, Chennai, Tamil Nadu, India
| |
Collapse
|
42
|
Chronic exercise mediates epigenetic suppression of L-type Ca2+ channel and BKCa channel in mesenteric arteries of hypertensive rats. J Hypertens 2021; 38:1763-1776. [PMID: 32384389 DOI: 10.1097/hjh.0000000000002457] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Regular exercise is a lifestyle intervention for controlling hypertension and has an improving effect on vascular function. Voltage-gated L-type Ca (LTCC) and large-conductance Ca-activated K (BKCa) channels are two principal mediators of vascular smooth muscle cell contractility and arterial tone. The present study tested the hypothesis that DNA methylation dynamics plays a key role in exercise-induced reprogramming and downregulation of LTCC and BKCa channel in mesenteric arteries from spontaneously hypertensive rats (SHRs). METHODS SHRs and Wistar-Kyoto (WKY) rats were subjected to exercise training or kept sedentary, and vascular molecular and functional properties were evaluated. RESULTS Exercise inhibited hypertension-induced upregulation of LTCC and BKCa channel function in mesenteric arteries by repressing LTCC α1c and BKCa β1 subunit expression. In accordance, exercise triggered hypermethylation of α1c and β1 gene in SHR, with concomitant decreasing TET1, increasing DNMT1 and DNMT3b expression in mesenteric arteries, as well as altering peripheral α-KG and S-adenosylmethionine/ S-adenosylhomocysteine ratio. Acting synergistically, these exercise-induced functional and molecular amelioration could allow for attenuating hypertension-induced elevation in arterial blood pressure. CONCLUSION Our results indicate that exercise suppresses LTCC and BKCa channel function via hypermethylation of α1c and β1 subunits, which contributes to the restoration of mesenteric arterial function and vasodilation during hypertension.
Collapse
|
43
|
Ottolini M, Sonkusare SK. The Calcium Signaling Mechanisms in Arterial Smooth Muscle and Endothelial Cells. Compr Physiol 2021; 11:1831-1869. [PMID: 33792900 PMCID: PMC10388069 DOI: 10.1002/cphy.c200030] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The contractile state of resistance arteries and arterioles is a crucial determinant of blood pressure and blood flow. Physiological regulation of arterial contractility requires constant communication between endothelial and smooth muscle cells. Various Ca2+ signals and Ca2+ -sensitive targets ensure dynamic control of intercellular communications in the vascular wall. The functional effect of a Ca2+ signal on arterial contractility depends on the type of Ca2+ -sensitive target engaged by that signal. Recent studies using advanced imaging methods have identified the spatiotemporal signatures of individual Ca2+ signals that control arterial and arteriolar contractility. Broadly speaking, intracellular Ca2+ is increased by ion channels and transporters on the plasma membrane and endoplasmic reticular membrane. Physiological roles for many vascular Ca2+ signals have already been confirmed, while further investigation is needed for other Ca2+ signals. This article focuses on endothelial and smooth muscle Ca2+ signaling mechanisms in resistance arteries and arterioles. We discuss the Ca2+ entry pathways at the plasma membrane, Ca2+ release signals from the intracellular stores, the functional and physiological relevance of Ca2+ signals, and their regulatory mechanisms. Finally, we describe the contribution of abnormal endothelial and smooth muscle Ca2+ signals to the pathogenesis of vascular disorders. © 2021 American Physiological Society. Compr Physiol 11:1831-1869, 2021.
Collapse
Affiliation(s)
- Matteo Ottolini
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia, USA
| | - Swapnil K Sonkusare
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia, USA.,Department of Molecular Physiology & Biological Physics, University of Virginia, Charlottesville, Virginia, USA.,Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
44
|
Hu XQ, Dasgupta C, Song R, Romero M, Wilson SM, Zhang L. MicroRNA-210 Mediates Hypoxia-Induced Repression of Spontaneous Transient Outward Currents in Sheep Uterine Arteries During Gestation. Hypertension 2021; 77:1412-1427. [PMID: 33641365 DOI: 10.1161/hypertensionaha.120.16831] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Xiang-Qun Hu
- From the Lawrence D. Longo, MD, Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, CA
| | - Chiranjib Dasgupta
- From the Lawrence D. Longo, MD, Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, CA
| | - Rui Song
- From the Lawrence D. Longo, MD, Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, CA
| | - Monica Romero
- From the Lawrence D. Longo, MD, Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, CA
| | - Sean M Wilson
- From the Lawrence D. Longo, MD, Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, CA
| | - Lubo Zhang
- From the Lawrence D. Longo, MD, Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, CA
| |
Collapse
|
45
|
Drumm BT, Thornbury KD, Hollywood MA, Sergeant GP. Role of Ano1 Ca 2+-activated Cl - channels in generating urethral tone. Am J Physiol Renal Physiol 2021; 320:F525-F536. [PMID: 33554780 DOI: 10.1152/ajprenal.00520.2020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Urinary continence is maintained in the lower urinary tract by the contracture of urethral sphincters, including smooth muscle of the internal urethral sphincter. These contractions occlude the urethral lumen, preventing urine leakage from the bladder to the exterior. Over the past 20 years, research on the ionic conductances that contribute to urethral smooth muscle contractility has greatly accelerated. A debate has emerged over the role of interstitial cell of Cajal (ICC)-like cells in the urethra and their expression of Ca2+-activated Cl- channels encoded by anoctamin-1 [Ano1; transmembrane member 16 A (Tmem16a) gene]. It has been proposed that Ano1 channels expressed in urethral ICC serve as a source of depolarization for smooth muscle cells, increasing their excitability and contributing to tone. Although a clear role for Ano1 channels expressed in ICC is evident in other smooth muscle organs, such as the gastrointestinal tract, the role of these channels in the urethra is unclear, owing to differences in the species (rabbit, rat, guinea pig, sheep, and mouse) examined and experimental approaches by different groups. The importance of clarifying this situation is evident as effective targeting of Ano1 channels may lead to new treatments for urinary incontinence. In this review, we summarize the key findings from different species on the role of ICC and Ano1 channels in urethral contractility. Finally, we outline proposals for clarifying this controversial and important topic by addressing how cell-specific optogenetic and inducible cell-specific genetic deletion strategies coupled with advances in Ano1 channel pharmacology may clarify this area in future studies.NEW & NOTEWORTHY Studies from the rabbit have shown that anoctamin-1 (Ano1) channels expressed in urethral interstitial cells of Cajal (ICC) serve as a source of depolarization for smooth muscle cells, increasing excitability and tone. However, the role of urethral Ano1 channels is unclear, owing to differences in the species examined and experimental approaches. We summarize findings from different species on the role of urethral ICC and Ano1 channels in urethral contractility and outline proposals for clarifying this topic using cell-specific optogenetic approaches.
Collapse
Affiliation(s)
- Bernard T Drumm
- Smooth Muscle Research Centre, Dundalk Institute of Technology, Dundalk, Ireland
| | - Keith D Thornbury
- Smooth Muscle Research Centre, Dundalk Institute of Technology, Dundalk, Ireland
| | - Mark A Hollywood
- Smooth Muscle Research Centre, Dundalk Institute of Technology, Dundalk, Ireland
| | - Gerard P Sergeant
- Smooth Muscle Research Centre, Dundalk Institute of Technology, Dundalk, Ireland
| |
Collapse
|
46
|
Nieves-Cintrón M, Flores-Tamez VA, Le T, Baudel MMA, Navedo MF. Cellular and molecular effects of hyperglycemia on ion channels in vascular smooth muscle. Cell Mol Life Sci 2021; 78:31-61. [PMID: 32594191 PMCID: PMC7765743 DOI: 10.1007/s00018-020-03582-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 06/10/2020] [Accepted: 06/22/2020] [Indexed: 12/13/2022]
Abstract
Diabetes affects millions of people worldwide. This devastating disease dramatically increases the risk of developing cardiovascular disorders. A hallmark metabolic abnormality in diabetes is hyperglycemia, which contributes to the pathogenesis of cardiovascular complications. These cardiovascular complications are, at least in part, related to hyperglycemia-induced molecular and cellular changes in the cells making up blood vessels. Whereas the mechanisms mediating endothelial dysfunction during hyperglycemia have been extensively examined, much less is known about how hyperglycemia impacts vascular smooth muscle function. Vascular smooth muscle function is exquisitely regulated by many ion channels, including several members of the potassium (K+) channel superfamily and voltage-gated L-type Ca2+ channels. Modulation of vascular smooth muscle ion channels function by hyperglycemia is emerging as a key contributor to vascular dysfunction in diabetes. In this review, we summarize the current understanding of how diabetic hyperglycemia modulates the activity of these ion channels in vascular smooth muscle. We examine underlying mechanisms, general properties, and physiological relevance in the context of myogenic tone and vascular reactivity.
Collapse
Affiliation(s)
- Madeline Nieves-Cintrón
- Department of Pharmacology, University of California Davis, One Shields Avenue, Davis, CA, 95616, USA
| | - Víctor A Flores-Tamez
- Department of Pharmacology, University of California Davis, One Shields Avenue, Davis, CA, 95616, USA
| | - Thanhmai Le
- Department of Pharmacology, University of California Davis, One Shields Avenue, Davis, CA, 95616, USA
| | | | - Manuel F Navedo
- Department of Pharmacology, University of California Davis, One Shields Avenue, Davis, CA, 95616, USA.
| |
Collapse
|
47
|
Bukiya AN, Leo MD, Jaggar JH, Dopico AM. Cholesterol activates BK channels by increasing KCNMB1 protein levels in the plasmalemma. J Biol Chem 2021; 296:100381. [PMID: 33556372 PMCID: PMC7950327 DOI: 10.1016/j.jbc.2021.100381] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 01/11/2021] [Accepted: 02/02/2021] [Indexed: 01/03/2023] Open
Abstract
Calcium-/voltage-gated, large-conductance potassium channels (BKs) control critical physiological processes, including smooth muscle contraction. Numerous observations concur that elevated membrane cholesterol (CLR) inhibits the activity of homomeric BKs consisting of channel-forming alpha subunits. In mammalian smooth muscle, however, native BKs include accessory KCNMB1 (β1) subunits, which enable BK activation at physiological intracellular calcium. Here, we studied the effect of CLR enrichment on BK currents from rat cerebral artery myocytes. Using inside-out patches from middle cerebral artery (MCA) myocytes at [Ca2+]free=30 μM, we detected BK activation in response to in vivo and in vitro CLR enrichment of myocytes. While a significant increase in myocyte CLR was achieved within 5 min of CLR in vitro loading, this brief CLR enrichment of membrane patches decreased BK currents, indicating that BK activation by CLR requires a protracted cellular process. Indeed, blocking intracellular protein trafficking with brefeldin A (BFA) not only prevented BK activation but led to channel inhibition upon CLR enrichment. Surface protein biotinylation followed by Western blotting showed that BFA blocked the increase in plasmalemmal KCNMB1 levels achieved via CLR enrichment. Moreover, CLR enrichment of arteries with naturally high KCNMB1 levels, such as basilar and coronary arteries, failed to activate BK currents. Finally, CLR enrichment failed to activate BK channels in MCA myocytes from KCNMB1-/- mouse while activation was detected in their wild-type (C57BL/6) counterparts. In conclusion, the switch in CLR regulation of BK from inhibition to activation is determined by a trafficking-dependent increase in membrane levels of KCNMB1 subunits.
Collapse
Affiliation(s)
- Anna N Bukiya
- Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee, USA.
| | - M Dennis Leo
- Department of Physiology, College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Jonathan H Jaggar
- Department of Physiology, College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Alex M Dopico
- Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee, USA.
| |
Collapse
|
48
|
Igniting Ca 2+ sparks with TRPML1. Proc Natl Acad Sci U S A 2020; 117:32836-32838. [PMID: 33262276 DOI: 10.1073/pnas.2022896117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
49
|
The intracellular Ca 2+ release channel TRPML1 regulates lower urinary tract smooth muscle contractility. Proc Natl Acad Sci U S A 2020; 117:30775-30786. [PMID: 33199609 PMCID: PMC7720193 DOI: 10.1073/pnas.2016959117] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
TRPML1 (transient receptor potential mucolipin 1) is a Ca2+-permeable, nonselective cation channel that is localized to late endosomes and lysosomes. Here, we investigated the function of TRPML1 channels in regulating lower urinary tract (LUT) smooth muscle cell (SMC) contractility. We found that TRPML1 forms a stable signaling complex with ryanodine receptors (RyRs) in the sarcoplasmic reticulum (SR). We further showed that TRPML1 channels are important for initiating an essential Ca2+-signaling negative feedback mechanism between RyRs on SR membranes and K+ channels on the plasma membrane. Knockout of TRPML1 channels in mice impaired this pathway, resulting in LUT smooth muscle hypercontractility and symptoms of overactive bladder. Our findings demonstrate a critical role for TRPML1 in LUT function. TRPML1 (transient receptor potential mucolipin 1) is a Ca2+-permeable, nonselective cation channel that is predominantly localized to the membranes of late endosomes and lysosomes (LELs). Intracellular release of Ca2+ through TRPML1 is thought to be pivotal for maintenance of intravesicular acidic pH as well as the maturation, fusion, and trafficking of LELs. Interestingly, genetic ablation of TRPML1 in mice (Mcoln1−/−) induces a hyperdistended/hypertrophic bladder phenotype. Here, we investigated this phenomenon further by exploring an unconventional role for TRPML1 channels in the regulation of Ca2+-signaling activity and contractility in bladder and urethral smooth muscle cells (SMCs). Four-dimensional (4D) lattice light-sheet live-cell imaging showed that the majority of LELs in freshly isolated bladder SMCs were essentially immobile. Superresolution microscopy revealed distinct nanoscale colocalization of LEL-expressing TRPML1 channels with ryanodine type 2 receptors (RyR2) in bladder SMCs. Spontaneous intracellular release of Ca2+ from the sarcoplasmic reticulum (SR) through RyR2 generates localized elevations of Ca2+ (“Ca2+ sparks”) that activate plasmalemmal large-conductance Ca2+-activated K+ (BK) channels, a critical negative feedback mechanism that regulates smooth muscle contractility. This mechanism was impaired in Mcoln1−/− mice, which showed diminished spontaneous Ca2+ sparks and BK channel activity in bladder and urethra SMCs. Additionally, ex vivo contractility experiments showed that loss of Ca2+ spark–BK channel signaling in Mcoln1−/− mice rendered both bladder and urethra smooth muscle hypercontractile. Voiding activity analyses revealed bladder overactivity in Mcoln1−/− mice. We conclude that TRPML1 is critically important for Ca2+ spark signaling, and thus regulation of contractility and function, in lower urinary tract SMCs.
Collapse
|
50
|
Hu XQ, Song R, Romero M, Dasgupta C, Min J, Hatcher D, Xiao D, Blood A, Wilson SM, Zhang L. Gestational Hypoxia Inhibits Pregnancy-Induced Upregulation of Ca 2+ Sparks and Spontaneous Transient Outward Currents in Uterine Arteries Via Heightened Endoplasmic Reticulum/Oxidative Stress. Hypertension 2020; 76:930-942. [PMID: 32683903 PMCID: PMC7429261 DOI: 10.1161/hypertensionaha.120.15235] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Hypoxia during pregnancy profoundly affects uterine vascular adaptation and increases the risk of pregnancy complications, including preeclampsia and fetal intrauterine growth restriction. We recently demonstrated that increases in Ca2+ sparks and spontaneous transient outward currents (STOCs) played an essential role in pregnancy-induced uterine vascular adaptation. In the present study, we hypothesize that gestational hypoxia suppresses Ca2+ sparks/STOCs coupling leading to increased uterine vascular tone via enhanced endoplasmic reticulum (ER)/oxidative stress. Uterine arteries were obtained from nonpregnant and near-term pregnant sheep residing in low altitude or acclimatizing to high-altitude (3801 m) hypoxia for ≈110 days. High-altitude hypoxia suppressed pregnancy-induced upregulation of RyR1 and RyR2 (ryanodine receptor 1 and 2) protein abundance, Ca2+ sparks, and STOCs in uterine arteries. Inhibition of Ca2+ sparks/STOCs with the RyR inhibitor ryanodine significantly increased pressure-dependent myogenic tone in uterine arteries from low-altitude normoxic pregnant animals but not those from high-altitude hypoxic pregnant animals. Gestational hypoxia significantly increased ER/oxidative stress in uterine arteries. Of importance, the hypoxia-mediated suppression of Ca2+ sparks/STOCs and increase in myogenic tone in uterine arteries of pregnant animals were reversed by inhibiting ER/oxidative stress. Of great interest, the impaired sex hormonal regulation of STOCs in high-altitude animals was annulled by scavenging reactive oxygen species but not by inhibiting ER stress. Together, the findings reveal the differential mechanisms of ER and oxidative stresses in suppressing Ca2+ sparks/STOCs and increasing myogenic tone of uterine arteries in hypoxia during gestation, providing new insights into the understanding of pregnancy complications associated with hypoxia.
Collapse
Affiliation(s)
- Xiang-Qun Hu
- From the Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, CA
| | - Rui Song
- From the Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, CA
| | - Monica Romero
- From the Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, CA
| | - Chiranjib Dasgupta
- From the Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, CA
| | - Joseph Min
- From the Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, CA
| | - Daisy Hatcher
- From the Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, CA
| | - Daliao Xiao
- From the Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, CA
| | - Arlin Blood
- From the Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, CA
| | - Sean M Wilson
- From the Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, CA
| | - Lubo Zhang
- From the Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, CA
| |
Collapse
|