1
|
Shvetsova AA, Shateeva VS, Khlystova MA, Makukha YA, Tarasova OS, Gaynullina DK. NADPH oxidase derived ROS promote arterial contraction in early postnatal rats by activation of L-type voltage-gated Ca 2+ channels. Free Radic Res 2025; 59:49-60. [PMID: 39879138 DOI: 10.1080/10715762.2024.2448483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 12/04/2024] [Accepted: 12/26/2024] [Indexed: 01/31/2025]
Abstract
Reactive oxygen species (ROS) produced by NADPH oxidase promote contraction of peripheral arteries, which is especially pronounced in early postnatal period in comparison to adulthood, but the mechanisms of such vasomotor influence are poorly understood. We tested the hypothesis that Rho-kinase and protein kinase C (PKC) mediate procontractile influence of NADPH oxidase derived ROS in peripheral artery of early postnatal rats. In addition, we evaluated the involvement Src-kinase and L-type voltage-gated Ca2+ channels (LTCC) into procontractile influence of ROS, produced by NADPH oxidase, because of their known interplay with Rho-kinase and PKC pathways. Saphenous arteries from 11- to 15-day-old male rats were studied using quantitative PCR, isometric myography and lucigenin-enhanced chemiluminescence. Arterial tissue of early postnatal rats contained Nox2, Nox4, Duox1 and Duox2 mRNAs, among which Nox2 mRNA was the most abundant. Pan-NADPH oxidase inhibitor VAS2870 (10 µM) significantly reduced arterial contractile responses to methoxamine. The inhibitors of Rho-kinase (Y27632, 3 µM), PKC (GF109203X, 10 µM) and Src-kinase (PP2, 10 µM), as well as LTCC blockers (nimodipine, 0.1 µM, and verapamil, 0.1 μM) also reduced methoxamine-induced contraction. Importantly, the effect of VAS2870 persisted in the presence of Rho-kinase, PKC or Src-kinase inhibitors, but not in the presence of LTCC blocker. Notably, the blockade of LTCC did not affect either basal or NADPH-induced O2•- production. Our data show that LTCC, but not Rho-kinase, PKC or Src-kinase are involved into procontractile effect of ROS, produced by NADPH oxidase, in saphenous artery of young rats. Сalcium influx through LTCC does not activate ROS production by NADPH oxidase.
Collapse
Affiliation(s)
- Anastasia A Shvetsova
- Department of Human and Animal Physiology, Faculty of Biology, M.V. Lomonosov Moscow State University, Moscow, Russia
| | - Valentina S Shateeva
- Department of Human and Animal Physiology, Faculty of Biology, M.V. Lomonosov Moscow State University, Moscow, Russia
| | - Margarita A Khlystova
- Department of Human and Animal Physiology, Faculty of Biology, M.V. Lomonosov Moscow State University, Moscow, Russia
| | - Yulia A Makukha
- Department of Human and Animal Physiology, Faculty of Biology, M.V. Lomonosov Moscow State University, Moscow, Russia
| | - Olga S Tarasova
- Department of Human and Animal Physiology, Faculty of Biology, M.V. Lomonosov Moscow State University, Moscow, Russia
- Department of Physiology and Pathology, Faculty of Basic Medicine, M.V. Lomonosov Moscow State University, Moscow, Russia
| | - Dina K Gaynullina
- Department of Human and Animal Physiology, Faculty of Biology, M.V. Lomonosov Moscow State University, Moscow, Russia
| |
Collapse
|
2
|
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
|
3
|
Samara I, Moula AI, Moulas AN, Katsouras CS. The Effect of Retinoids in Vascular Smooth Muscle Cells: From Phenotyping Switching to Proliferation and Migration. Int J Mol Sci 2024; 25:10303. [PMID: 39408632 PMCID: PMC11477379 DOI: 10.3390/ijms251910303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 09/22/2024] [Accepted: 09/23/2024] [Indexed: 10/20/2024] Open
Abstract
Atherosclerosis, a term derived from the Greek "athero" (atheroma) and "sclerosis" (hardening), is a long-standing process that leads to the formation of atheromatous plaques in the arterial wall, contributing to the development of atherosclerotic cardiovascular disease. The proliferation and migration of vascular smooth muscle cells (VSMCs) and the switching of their phenotype play a crucial role in the whole process. Retinoic acid (RA), a natural derivative of vitamin A, has been used in the treatment of various inflammatory diseases and cell proliferation disorders. Numerous studies have demonstrated that RA has an important inhibitory effect on the proliferation, migration, and dedifferentiation of vascular smooth muscle cells, leading to a significant reduction in atherosclerotic lesions. In this review article, we explore the effects of RA on the pathogenesis of atherosclerosis, focusing on its regulatory action in VSMCs and its role in the phenotypic switching, proliferation, and migration of VSMCs. Despite the potential impact that RA may have on the process of atherosclerosis, further studies are required to examine its safety and efficacy in clinical practice.
Collapse
Affiliation(s)
- Ioanna Samara
- Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece;
| | - Amalia I. Moula
- Department of Surgery, “Achillopouleion” General Hospital, 38222 Volos, Greece;
| | | | - Christos S. Katsouras
- Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece;
| |
Collapse
|
4
|
Lorigo M, Cairrao E. UV-B filter octylmethoxycinnamate-induced vascular endothelial disruption on rat aorta: In silico and in vitro approach. CHEMOSPHERE 2022; 307:135807. [PMID: 35931261 DOI: 10.1016/j.chemosphere.2022.135807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 07/07/2022] [Accepted: 07/19/2022] [Indexed: 06/15/2023]
Abstract
Throughout human life, an extensive and varied range of emerging environmental contaminants, called endocrine disruptors (EDCs), cause adverse health effects, including in the cardiovascular (CV) system. Cardiovascular diseases (CVD) are worryingly one of the leading causes of all mortality and mobility worldwide. The UV-B filter octylmethoxycinnamate (also designated octinoxate, or ethylhexyl methoxycinnamate (CAS number: 5466-77-3)) is an EDC widely present in all personal care products. However, to date, there are no studies evaluating the OMC-induced effects on vasculature using animal models to improve human cardiovascular health. This work analysed the effects of OMC on rat aorta vasculature and explored the modes of action implicated in these effects. Our results indicated that OMC relaxes the rat aorta by endothelium-dependent mechanisms through the signaling pathways of cyclic nucleotides and by endothelium-independent mechanisms involving inhibition of L-Type voltage-operated Ca2+ channels (L-Type VOCC). Overall, OMC toxicity on rat aorta may produce hypotension via vasodilation due to excessive NO release and blockade of L-Type VOCC. Moreover, the OMC-induced endothelial dysfunction may also occur by promoting the endothelial release of endothelin-1. Therefore, our findings demonstrate that exposure to OMC alters the reactivity of the rat aorta and highlight that long-term OMC exposure may increase the risk of human CV diseases.
Collapse
Affiliation(s)
- Margarida Lorigo
- CICS-UBI, Health Sciences Research Centre, University of Beira Interior, 6200-506, Covilhã, Portugal; FCS - UBI, Faculty of Health Sciences, University of Beira Interior, 6200-506, Covilhã, Portugal; C4-UBI, Cloud Computing Competence Centre, University of Beira Interior, 6200-501, Covilhã, Portugal.
| | - Elisa Cairrao
- CICS-UBI, Health Sciences Research Centre, University of Beira Interior, 6200-506, Covilhã, Portugal; FCS - UBI, Faculty of Health Sciences, University of Beira Interior, 6200-506, Covilhã, Portugal; C4-UBI, Cloud Computing Competence Centre, University of Beira Interior, 6200-501, Covilhã, Portugal.
| |
Collapse
|
5
|
Deng Q, Chen J. Potential Therapeutic Effect of All-Trans Retinoic Acid on Atherosclerosis. Biomolecules 2022; 12:869. [PMID: 35883425 PMCID: PMC9312697 DOI: 10.3390/biom12070869] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 06/08/2022] [Accepted: 06/22/2022] [Indexed: 11/16/2022] Open
Abstract
Atherosclerosis is a major risk factor for myocardial infarction and ischemic stroke, which are the leading cause of death worldwide. All-trans retinoic acid (ATRA) is a natural derivative of essential vitamin A. Numerous studies have shown that ATRA plays an important role in cell proliferation, cell apoptosis, cell differentiation, and embryonic development. All-trans retinoic acid (ATRA) is a ligand of retinoic acid receptors that regulates various biological processes by activating retinoic acid signals. In this paper, the metabolic processes of ATRA were reviewed, with emphasis on the effects of ATRA on inflammatory cells involved in the process of atherosclerosis.
Collapse
Affiliation(s)
| | - Jixiang Chen
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China;
| |
Collapse
|
6
|
Intrauterine Nitric Oxide Deficiency Weakens Differentiation of Vascular Smooth Muscle in Newborn Rats. Int J Mol Sci 2021; 22:ijms22158003. [PMID: 34360769 PMCID: PMC8347173 DOI: 10.3390/ijms22158003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 07/22/2021] [Accepted: 07/24/2021] [Indexed: 12/22/2022] Open
Abstract
Nitric oxide (NO) deficiency during pregnancy is a key reason for preeclampsia development. Besides its important vasomotor role, NO is shown to regulate the cell transcriptome. However, the role of NO in transcriptional regulation of developing smooth muscle has never been studied before. We hypothesized that in early ontogeny, NO is important for the regulation of arterial smooth muscle-specific genes expression. Pregnant rats consumed NO-synthase inhibitor L-NAME (500 mg/L in drinking water) from gestational day 10 till delivery, which led to an increase in blood pressure, a key manifestation of preeclampsia. L-NAME reduced blood concentrations of NO metabolites in dams and their newborn pups, as well as relaxations of pup aortic rings to acetylcholine. Using qPCR, we demonstrated reduced abundances of the smooth muscle-specific myosin heavy chain isoform, α-actin, SM22α, and L-type Ca2+-channel mRNAs in the aorta of newborn pups from the L-NAME group compared to control pups. To conclude, the intrauterine NO deficiency weakens gene expression specific for a contractile phenotype of arterial smooth muscle in newborn offspring.
Collapse
|
7
|
Shawer H, Norman K, Cheng CW, Foster R, Beech DJ, Bailey MA. ORAI1 Ca 2+ Channel as a Therapeutic Target in Pathological Vascular Remodelling. Front Cell Dev Biol 2021; 9:653812. [PMID: 33937254 PMCID: PMC8083964 DOI: 10.3389/fcell.2021.653812] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 03/08/2021] [Indexed: 12/21/2022] Open
Abstract
In the adult, vascular smooth muscle cells (VSMC) are normally physiologically quiescent, arranged circumferentially in one or more layers within blood vessel walls. Remodelling of native VSMC to a proliferative state for vascular development, adaptation or repair is driven by platelet-derived growth factor (PDGF). A key effector downstream of PDGF receptors is store-operated calcium entry (SOCE) mediated through the plasma membrane calcium ion channel, ORAI1, which is activated by the endoplasmic reticulum (ER) calcium store sensor, stromal interaction molecule-1 (STIM1). This SOCE was shown to play fundamental roles in the pathological remodelling of VSMC. Exciting transgenic lineage-tracing studies have revealed that the contribution of the phenotypically-modulated VSMC in atherosclerotic plaque formation is more significant than previously appreciated, and growing evidence supports the relevance of ORAI1 signalling in this pathologic remodelling. ORAI1 has also emerged as an attractive potential therapeutic target as it is accessible to extracellular compound inhibition. This is further supported by the progression of several ORAI1 inhibitors into clinical trials. Here we discuss the current knowledge of ORAI1-mediated signalling in pathologic vascular remodelling, particularly in the settings of atherosclerotic cardiovascular diseases (CVDs) and neointimal hyperplasia, and the recent developments in our understanding of the mechanisms by which ORAI1 coordinates VSMC phenotypic remodelling, through the activation of key transcription factor, nuclear factor of activated T-cell (NFAT). In addition, we discuss advances in therapeutic strategies aimed at the ORAI1 target.
Collapse
Affiliation(s)
- Heba Shawer
- School of Medicine, The Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| | - Katherine Norman
- School of Medicine, The Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom.,School of Chemistry, University of Leeds, Leeds, United Kingdom
| | - Chew W Cheng
- School of Medicine, The Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| | - Richard Foster
- School of Medicine, The Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom.,School of Chemistry, University of Leeds, Leeds, United Kingdom
| | - David J Beech
- School of Medicine, The Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| | - Marc A Bailey
- School of Medicine, The Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| |
Collapse
|
8
|
A differentiated Ca 2+ signalling phenotype has minimal impact on myocardin expression in an automated differentiation assay using A7r5 cells. Cell Calcium 2021; 96:102369. [PMID: 33677175 DOI: 10.1016/j.ceca.2021.102369] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 02/06/2021] [Accepted: 02/08/2021] [Indexed: 01/12/2023]
Abstract
Vascular smooth muscle cells are unusual in that differentiated, contractile cells possess the capacity to "de-differentiate" into a synthetic phenotype that is characterized by being replicative, secretory, and migratory. One aspect of this phenotypic modulation is a shift from voltage-gated Ca2+ signalling in electrically coupled, differentiated cells to increased dependence on store-operated Ca2+ entry and sarcoplasmic reticulum Ca2+ release in synthetic cells. Conversely, an increased voltage-gated Ca2+ entry is seen when proliferating A7r5 smooth muscle cells quiesce. We asked whether this change in Ca2+ signalling was linked to changes in the expression of the phenotype-regulating transcriptional co-activator myocardin or α-smooth muscle actin, using correlative epifluorescence Ca2+ imaging and immunocytochemistry. Cells were cultured in growth media (DMEM, 10% serum, 25 mM glucose) or differentiation media (DMEM, 1% serum, 5 mM glucose). Coinciding with growth arrest, A7r5 cells became electrically coupled, and spontaneous Ca2+ signalling showed increasing dependence on L-type voltage-gated Ca2+ channels that were blocked with nifedipine (5 μM). These synchronized oscillations were modulated by ryanodine receptors, based on their sensitivity to dantrolene (5 μM). Actively growing cultures had spontaneous Ca2+ transients that were insensitive to nifedipine and dantrolene but were blocked by inhibition of the sarco-endoplasmic reticulum ATPase with cyclopiazonic acid (10 μM). In cells treated with differentiation media, myocardin and αSMA immunoreactivity increased prior to changes in the Ca2+ signalling phenotype, while chronic inhibition of voltage-gated Ca2+ entry modestly increased immunoreactivity of myocardin. Stepwise regression analyses suggested that changes in myocardin expression had a weak relationship with Ca2+ signalling synchronicity, but not frequency or amplitude. In conclusion, we report a 96-well assay and analytical pipeline to study the link between Ca2+ signalling and smooth muscle differentiation. This assay showed that changes in the expression of two molecular differentiation markers (myocardin and αSMA) tended to precede changes in the Ca2+ signalling phenotype.
Collapse
|
9
|
Nguyen NT, Nguyen TT, Da Ly D, Xia JB, Qi XF, Lee IK, Cha SK, Park KS. Oxidative stress by Ca 2+ overload is critical for phosphate-induced vascular calcification. Am J Physiol Heart Circ Physiol 2020; 319:H1302-H1312. [PMID: 33095057 DOI: 10.1152/ajpheart.00305.2020] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Hyperphosphatemia is the primary risk factor for vascular calcification, which is closely associated with cardiovascular morbidity and mortality. Recent evidence showed that oxidative stress by high inorganic phosphate (Pi) mediates calcific changes in vascular smooth muscle cells (VSMCs). However, intracellular signaling responsible for Pi-induced oxidative stress remains unclear. Here, we investigated molecular mechanisms of Pi-induced oxidative stress related with intracellular Ca2+ ([Ca2+]i) disturbance, which is critical for calcification of VSMCs. VSMCs isolated from rat thoracic aorta or A7r5 cells were incubated with high Pi-containing medium. Extracellular signal-regulated kinase (ERK) and mammalian target of rapamycin were activated by high Pi that was required for vascular calcification. High Pi upregulated expressions of type III sodium-phosphate cotransporters PiT-1 and -2 and stimulated their trafficking to the plasma membrane. Interestingly, high Pi increased [Ca2+]i exclusively dependent on extracellular Na+ and Ca2+ as well as PiT-1/2 abundance. Furthermore, high-Pi induced plasma membrane depolarization mediated by PiT-1/2. Pretreatment with verapamil, as a voltage-gated Ca2+ channel (VGCC) blocker, inhibited Pi-induced [Ca2+]i elevation, oxidative stress, ERK activation, and osteogenic differentiation. These protective effects were reiterated by extracellular Ca2+-free condition, intracellular Ca2+ chelation, or suppression of oxidative stress. Mitochondrial superoxide scavenger also effectively abrogated ERK activation and osteogenic differentiation of VSMCs by high Pi. Taking all these together, we suggest that high Pi activates depolarization-triggered Ca2+ influx via VGCC, and subsequent [Ca2+]i increase elicits oxidative stress and osteogenic differentiation. PiT-1/2 mediates Pi-induced [Ca2+]i overload and oxidative stress but in turn, PiT-1/2 is upregulated by consequences of these alterations.NEW & NOTEWORTHY The novel findings of this study are type III sodium-phosphate cotransporters PiT-1 and -2-dependent depolarization by high Pi, leading to Ca2+ entry via voltage-gated Ca2+ channels in vascular smooth muscle cells. Cytosolic Ca2+ increase and subsequent oxidative stress are indispensable for osteogenic differentiation and calcification. In addition, plasmalemmal abundance of PiT-1/2 relies on Ca2+ overload and oxidative stress, establishing a positive feedback loop. Identification of mechanistic components of a vicious cycle could provide novel therapeutic strategies against vascular calcification in hyperphosphatemic patients.
Collapse
Affiliation(s)
- Nhung Thi Nguyen
- Department of Physiology, Yonsei University Wonju College of Medicine, Wonju, Korea.,Mitohormesis Research Center, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Tuyet Thi Nguyen
- Department of Physiology, Yonsei University Wonju College of Medicine, Wonju, Korea.,Internal Medicine Residency Program, College of Health Sciences, VinUniversity, Hanoi, Vietnam
| | - Dat Da Ly
- Department of Physiology, Yonsei University Wonju College of Medicine, Wonju, Korea.,Mitohormesis Research Center, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Jing-Bo Xia
- Key Laboratory of Regenerative Medicine, Ministry of Education, Department of Developmental and Regenerative Biology, Jinan University, Guangzhou, China
| | - Xu-Feng Qi
- Key Laboratory of Regenerative Medicine, Ministry of Education, Department of Developmental and Regenerative Biology, Jinan University, Guangzhou, China
| | - In-Kyu Lee
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Daegu, Korea
| | - Seung-Kuy Cha
- Department of Physiology, Yonsei University Wonju College of Medicine, Wonju, Korea.,Mitohormesis Research Center, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Kyu-Sang Park
- Department of Physiology, Yonsei University Wonju College of Medicine, Wonju, Korea.,Mitohormesis Research Center, Yonsei University Wonju College of Medicine, Wonju, Korea
| |
Collapse
|
10
|
Freichel M, Ottenheijm R. L-type blocker STIMulate Ca 2+ entry in synthetic VSMCs. Cell Calcium 2020; 91:102279. [PMID: 32892148 DOI: 10.1016/j.ceca.2020.102279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 08/25/2020] [Indexed: 11/25/2022]
Affiliation(s)
- Marc Freichel
- Pharmakologisches Institut, Ruprecht-Karls-Universität Heidelberg, 69120, Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Heidelberg, 69120, Germany.
| | - Roger Ottenheijm
- Pharmakologisches Institut, Ruprecht-Karls-Universität Heidelberg, 69120, Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Heidelberg, 69120, Germany
| |
Collapse
|
11
|
Johnson MT, Gudlur A, Zhang X, Xin P, Emrich SM, Yoast RE, Courjaret R, Nwokonko RM, Li W, Hempel N, Machaca K, Gill DL, Hogan PG, Trebak M. L-type Ca 2+ channel blockers promote vascular remodeling through activation of STIM proteins. Proc Natl Acad Sci U S A 2020; 117:17369-17380. [PMID: 32641503 PMCID: PMC7382247 DOI: 10.1073/pnas.2007598117] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Voltage-gated L-type Ca2+ channel (Cav1.2) blockers (LCCBs) are major drugs for treating hypertension, the preeminent risk factor for heart failure. Vascular smooth muscle cell (VSMC) remodeling is a pathological hallmark of chronic hypertension. VSMC remodeling is characterized by molecular rewiring of the cellular Ca2+ signaling machinery, including down-regulation of Cav1.2 channels and up-regulation of the endoplasmic reticulum (ER) stromal-interacting molecule (STIM) Ca2+ sensor proteins and the plasma membrane ORAI Ca2+ channels. STIM/ORAI proteins mediate store-operated Ca2+ entry (SOCE) and drive fibro-proliferative gene programs during cardiovascular remodeling. SOCE is activated by agonists that induce depletion of ER Ca2+, causing STIM to activate ORAI. Here, we show that the three major classes of LCCBs activate STIM/ORAI-mediated Ca2+ entry in VSMCs. LCCBs act on the STIM N terminus to cause STIM relocalization to junctions and subsequent ORAI activation in a Cav1.2-independent and store depletion-independent manner. LCCB-induced promotion of VSMC remodeling requires STIM1, which is up-regulated in VSMCs from hypertensive rats. Epidemiology showed that LCCBs are more associated with heart failure than other antihypertensive drugs in patients. Our findings unravel a mechanism of LCCBs action on Ca2+ signaling and demonstrate that LCCBs promote vascular remodeling through STIM-mediated activation of ORAI. Our data indicate caution against the use of LCCBs in elderly patients or patients with advanced hypertension and/or onset of cardiovascular remodeling, where levels of STIM and ORAI are elevated.
Collapse
Affiliation(s)
- Martin T Johnson
- Department of Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, Hershey, PA 17033
| | - Aparna Gudlur
- Division of Signaling and Gene Expression, La Jolla Institute for Immunology, La Jolla, CA 92037
| | - Xuexin Zhang
- Department of Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, Hershey, PA 17033
| | - Ping Xin
- Department of Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, Hershey, PA 17033
| | - Scott M Emrich
- Department of Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, Hershey, PA 17033
| | - Ryan E Yoast
- Department of Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, Hershey, PA 17033
| | - Raphael Courjaret
- Department of Physiology and Biophysics, Weill Cornell Medicine Qatar, Education City, Qatar Foundation, Doha, Qatar
| | - Robert M Nwokonko
- Department of Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, Hershey, PA 17033
| | - Wei Li
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA 17033
- Penn State Cancer Institute, The Pennsylvania State University College of Medicine, Hershey, PA 17033
- Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA 17033
| | - Nadine Hempel
- Penn State Cancer Institute, The Pennsylvania State University College of Medicine, Hershey, PA 17033
- Department of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, PA 17033
| | - Khaled Machaca
- Department of Physiology and Biophysics, Weill Cornell Medicine Qatar, Education City, Qatar Foundation, Doha, Qatar
| | - Donald L Gill
- Department of Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, Hershey, PA 17033
| | - Patrick G Hogan
- Division of Signaling and Gene Expression, La Jolla Institute for Immunology, La Jolla, CA 92037
| | - Mohamed Trebak
- Department of Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, Hershey, PA 17033;
- Penn State Cancer Institute, The Pennsylvania State University College of Medicine, Hershey, PA 17033
| |
Collapse
|
12
|
Activation of Inward Rectifier K + Channel 2.1 by PDGF-BB in Rat Vascular Smooth Muscle Cells through Protein Kinase A. BIOMED RESEARCH INTERNATIONAL 2020; 2020:4370832. [PMID: 32461988 PMCID: PMC7212311 DOI: 10.1155/2020/4370832] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Accepted: 03/19/2020] [Indexed: 11/17/2022]
Abstract
Platelet-derived growth factor-BB (PDGF-BB) can induce the proliferation, migration, and phenotypic modulation of vascular smooth muscle cells (VSMCs). We used patch clamp methods to study the effects of PDGF-BB on inward rectifier K+ channel 2.1 (Kir2.1) channels in rat thoracic aorta VSMCs (RASMCs). PDGF-BB (25 ng/mL) increased Kir2.x currents (−11.81 ± 2.47 pA/pF, P < 0.05 vs. CON, n = 10). Ba2+(50 μM) decreased Kir2.x currents (−2.13 ± 0.23 pA/pF, P < 0.05 vs. CON, n = 10), which were promoted by PDGF-BB (−6.98 ± 1.03 pA/pF). PDGF-BB specifically activates Kir2.1 but not Kir2.2 and Kir2.3 channels in HEK-293 cells. The PDGF-BB-induced stimulation of Kir2.1 currents was blocked by the PDGF-BB receptor β (PDGF-BBRβ) inhibitor AG1295 and was not affected by the PDGF-BBRα inhibitor AG1296. The PDGF-BB-induced stimulation of Kir2.1 currents was blocked by the protein kinase A inhibitor Rp-8-CPT-cAMPs; however, the antagonist of protein kinase B (GSK690693) had marginal effects on current activity. The PDGF-BB-induced stimulation of Kir2.1 currents was enhanced by forskolin, an adenylyl cyclase (AC) activator, and was blocked by the AC inhibitor SQ22536. We conclude that PDGF-BB increases Kir2.1 currents via PDGF-BBRβ through activation of cAMP-PKA signaling in RASMCs.
Collapse
|
13
|
Zhong C, Schleifenbaum J. Genetically Encoded Calcium Indicators: A New Tool in Renal Hypertension Research. Front Med (Lausanne) 2019; 6:128. [PMID: 31263699 PMCID: PMC6585435 DOI: 10.3389/fmed.2019.00128] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 05/23/2019] [Indexed: 11/13/2022] Open
Abstract
Hypertension is ranked as the third cause of disability-adjusted life-years. The percentage of the population suffering from hypertension will continue to increase over the next years. Renovascular disease is one of the most common causes of secondary hypertension. Vascular changes seen in hypertension are partially based on dysfunctional calcium signaling. This signaling can be studied using calcium indicators (loading dyes and genetically encoded calcium indicators; GECIs). Most progress in development has been seen in GECIs, which are used in an increasing number of publications concerning calcium signaling in vasculature and the kidney. The use of transgenic mouse models expressing GECIs will facilitate new possibilities to study dysfunctional calcium signaling in a cell type-specific manner, thus helping to identify more specific targets for treatment of (renal) hypertension.
Collapse
Affiliation(s)
- Cheng Zhong
- Institute of Vegetative Physiology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Johanna Schleifenbaum
- Institute of Vegetative Physiology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|
14
|
Fang Q, Tian M, Wang F, Zhang Z, Du T, Wang W, Yang Y, Li X, Chen G, Xiao L, Wei H, Wang Y, Chen C, Wang DW. Amlodipine induces vasodilation via Akt2/Sp1-activated miR-21 in smooth muscle cells. Br J Pharmacol 2019; 176:2306-2320. [PMID: 30927374 DOI: 10.1111/bph.14679] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 02/15/2019] [Accepted: 03/13/2019] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND AND PURPOSE The calcium antagonist amlodipine exerts important cardioprotective effects by modulating smooth muscle and endothelial functions. However, the mechanisms underlying these effects are incompletely understood. EXPERIMENTAL APPROACH Western blotting was used to compare the expression of key genes involved in vascular smooth muscle cell (VSMC) phenotype conversion. Recombinant adeno-associated virus system was used to regulate miRNA expression in rats via tail vein. Bioinformatics was used to predict the transcriptional regulation of miR-21 upstream followed by biochemical validation using quantitative real-time polymerase chain reaction, ChIP-qPCR and EMSA assays. KEY RESULTS Only the calcium antagonist amlodipine, and no other type of anti-hypertensive drug, induced miR-21 overexpression in plasma and aortic vessels in the animal model. Real-time PCR and luciferase assays showed that amlodipine induced miR-21 overexpression in vascular smooth muscle cells. Western blot and immunofluorescence assays demonstrated that amlodipine activated Akt2, rather than Akt1, followed by activation of transcription factor Sp1, which regulated VSMC phenotype conversion via binding to the miR-21 promoter. Furthermore, bioinformatic analyses and luciferase assays demonstrated that amlodipine activated miR-21 transcription at the -2034/-2027 Sp1-binding site, which was further demonstrated by ChIP-qPCR and EMSA assays. Consistently, small-interfering RNA-mediated knockdown of Akt2 and Sp1 significantly attenuated the effects of amlodipine on miR-21 expression in smooth muscle cells. CONCLUSION AND IMPLICATIONS These results indicate that amlodipine induces smooth muscle cell differentiation via miR-21, which is regulated by p-Akt2 and Sp1 nuclear translocation, thereby providing a novel target for cardiovascular diseases.
Collapse
Affiliation(s)
- Qin Fang
- Division of Cardiology, Department of Internal Medicine, Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Min Tian
- Division of Cardiology, Department of Internal Medicine, Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Feng Wang
- Division of Cardiology, Department of Internal Medicine, Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Zhihao Zhang
- Division of Cardiology, Department of Internal Medicine, Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Tingyi Du
- Division of Cardiology, Department of Internal Medicine, Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Wang
- Division of Cardiology, Department of Internal Medicine, Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Yong Yang
- Division of Cardiology, Department of Internal Medicine, Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Xianqing Li
- Division of Cardiology, Department of Internal Medicine, Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Guangzhi Chen
- Division of Cardiology, Department of Internal Medicine, Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Lei Xiao
- Division of Cardiology, Department of Internal Medicine, Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Haoran Wei
- Division of Cardiology, Department of Internal Medicine, Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Wang
- Division of Cardiology, Department of Internal Medicine, Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Chen Chen
- Division of Cardiology, Department of Internal Medicine, Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Dao Wen Wang
- Division of Cardiology, Department of Internal Medicine, Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
15
|
Christie MJ, Romano T, Murphy RM, Posterino GS. The effect of intrauterine growth restriction on Ca 2+ -activated force and contractile protein expression in the mesenteric artery of adult (6-month-old) male and female Wistar-Kyoto rats. Physiol Rep 2018; 6:e13954. [PMID: 30592188 PMCID: PMC6308111 DOI: 10.14814/phy2.13954] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 11/22/2018] [Indexed: 12/29/2022] Open
Abstract
Intrauterine growth restriction (IUGR) is known to alter vascular smooth muscle reactivity, but it is currently unknown whether these changes are driven by downstream events that lead to force development, specifically, Ca2+ -regulated activation of the contractile apparatus or a shift in contractile protein content. This study investigated the effects of IUGR on Ca2+ -activated force production, contractile protein expression, and a potential phenotypic switch in the resistance mesenteric artery of both male and female Wistar-Kyoto (WKY) rats following two different growth restriction models. Pregnant female WKY rats were randomly assigned to either a control (C; N = 9) or food restriction diet (FR; 40% of control; N = 11) at gestational day-15 or underwent a bilateral uterine vessel ligation surgery restriction (SR; N = 10) or a sham surgery control model (SC; N = 12) on day-18 of gestation. At 6-months of age, vascular responsiveness of intact mesenteric arteries was studied, before chemically permeabilization using 50 μmol/L β-escin to investigate Ca2+ -activated force. Peak responsiveness to a K+ -induced depolarization was decreased (P ≤ 0.05) due to a reduction in maximum Ca2+ -activated force (P ≤ 0.05) in both male growth restricted experimental groups. Vascular responsiveness was unchanged between female experimental groups. Segments of mesenteric artery were analyzed using Western blotting revealed IUGR reduced the relative abundance of important receptor and contractile proteins in male growth restricted rats (P ≤ 0.05), suggesting a potential phenotypic switch, whilst no changes were observed in females. Results from this study suggest that IUGR alters the mesenteric artery reactivity due to a decrease in maximum Ca2+ -activated force, and likely contributed to by a reduction in contractile protein and receptor/channel content in 6-month-old male rats, while female WKY rats appear to be protected.
Collapse
Affiliation(s)
- Michael J. Christie
- Department of Physiology, Anatomy and MicrobiologyLa Trobe UniversityMelbourneVictoriaAustralia
| | - Tania Romano
- Department of Physiology, Anatomy and MicrobiologyLa Trobe UniversityMelbourneVictoriaAustralia
| | - Robyn M. Murphy
- Department of Biochemistry and GeneticsLa Trobe Institute for Molecular SciencesLa Trobe UniversityMelbourneVictoriaAustralia
| | - Giuseppe S. Posterino
- Department of Physiology, Anatomy and MicrobiologyLa Trobe UniversityMelbourneVictoriaAustralia
| |
Collapse
|
16
|
López-López JR, Cidad P, Pérez-García MT. Kv channels and vascular smooth muscle cell proliferation. Microcirculation 2018; 25. [PMID: 29110368 DOI: 10.1111/micc.12427] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 10/30/2017] [Indexed: 12/12/2022]
Abstract
Kv channels are present in virtually all VSMCs and strongly influence contractile responses. However, they are also instrumental in the proliferative, migratory, and secretory functions of synthetic, dedifferentiated VSMCs upon PM. In fact, Kv channels not only contribute to all these processes but also are active players in the phenotypic switch itself. This review is focused on the role(s) of Kv channels in VSMC proliferation, which is one of the best characterized functions of dedifferentiated VSMCs. VSMC proliferation is a complex process requiring specific Kv channels at specific time and locations. Their identification is further complicated by their large diversity and the differences in expression across vascular beds. Of interest, both conserved changes in some Kv channels and vascular bed-specific regulation of others seem to coexist and participate in VSMC proliferation through complementary mechanisms. Such a system will add flexibility to the process while providing the required robustness to preserve this fundamental cellular response.
Collapse
Affiliation(s)
- José R López-López
- Departamento de Bioquímica y Biología Molecular y Fisiología e Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid y Consejo Superior de Investigaciones Científicas (CSIC), Valladolid, Spain
| | - Pilar Cidad
- Departamento de Bioquímica y Biología Molecular y Fisiología e Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid y Consejo Superior de Investigaciones Científicas (CSIC), Valladolid, Spain
| | - M Teresa Pérez-García
- Departamento de Bioquímica y Biología Molecular y Fisiología e Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid y Consejo Superior de Investigaciones Científicas (CSIC), Valladolid, Spain
| |
Collapse
|
17
|
Fan G, Kaßmann M, Hashad AM, Welsh DG, Gollasch M. Differential targeting and signalling of voltage-gated T-type Ca v 3.2 and L-type Ca v 1.2 channels to ryanodine receptors in mesenteric arteries. J Physiol 2018; 596:4863-4877. [PMID: 30146760 PMCID: PMC6187032 DOI: 10.1113/jp276923] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 08/24/2018] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS In arterial smooth muscle, Ca2+ sparks are elementary Ca2+ -release events generated by ryanodine receptors (RyRs) to cause vasodilatation by opening maxi Ca2+ -sensitive K+ (BKCa ) channels. This study elucidated the contribution of T-type Cav 3.2 channels in caveolae and their functional interaction with L-type Cav 1.2 channels to trigger Ca2+ sparks in vascular smooth muscle cells (VSMCs). Our data demonstrate that L-type Cav 1.2 channels provide the predominant Ca2+ pathway for the generation of Ca2+ sparks in murine arterial VSMCs. T-type Cav 3.2 channels represent an additional source for generation of VSMC Ca2+ sparks. They are located in pit structures of caveolae to provide locally restricted, tight coupling between T-type Cav 3.2 channels and RyRs to ignite Ca2+ sparks. ABSTRACT Recent data suggest that T-type Cav 3.2 channels in arterial vascular smooth muscle cells (VSMCs) and pits structure of caveolae could contribute to elementary Ca2+ signalling (Ca2+ sparks) via ryanodine receptors (RyRs) to cause vasodilatation. While plausible, their precise involvement in igniting Ca2+ sparks remains largely unexplored. The goal of this study was to elucidate the contribution of caveolar Cav 3.2 channels and their functional interaction with Cav 1.2 channels to trigger Ca2+ sparks in VSMCs from mesenteric, tibial and cerebral arteries. We used tamoxifen-inducible smooth muscle-specific Cav 1.2-/- (SMAKO) mice and laser scanning confocal microscopy to assess Ca2+ spark generation in VSMCs. Ni2+ , Cd2+ and methyl-β-cyclodextrin were used to inhibit Cav 3.2 channels, Cav 1.2 channels and caveolae, respectively. Ni2+ (50 μmol L-1 ) and methyl-β-cyclodextrin (10 mmol L-1 ) decreased Ca2+ spark frequency by ∼20-30% in mesenteric VSMCs in a non-additive manner, but failed to inhibit Ca2+ sparks in tibial and cerebral artery VSMCs. Cd2+ (200 μmol L-1 ) suppressed Ca2+ sparks in mesenteric arteries by ∼70-80%. A similar suppression of Ca2+ sparks was seen in mesenteric artery VSMCs of SMAKO mice. The remaining Ca2+ sparks were fully abolished by Ni2+ or methyl-β-cyclodextrin. Our data demonstrate that Ca2+ influx through CaV 1.2 channels is the primary means of triggering Ca2+ sparks in murine arterial VSMCs. CaV 3.2 channels, localized to caveolae and tightly coupled to RyR, provide an additional Ca2+ source for Ca2+ spark generation in mesenteric, but not tibial and cerebral, arteries.
Collapse
Affiliation(s)
- Gang Fan
- Charité – Universitätsmedizin BerlinExperimental and Clinical Research Center (ECRC)Campus BuchBerlinGermany
| | - Mario Kaßmann
- Charité – Universitätsmedizin BerlinExperimental and Clinical Research Center (ECRC)Campus BuchBerlinGermany
- DZHK (German Centre for Cardiovascular Research)BerlinGermany
| | - Ahmed M. Hashad
- Department of Physiology and PharmacologyHotchkiss Brain and Libin Cardiovascular InstitutesUniversity of CalgaryAlbertaCanada
| | - Donald G. Welsh
- Department of Physiology and PharmacologyWestern UniversityLondonONCanada
| | - Maik Gollasch
- Charité – Universitätsmedizin BerlinExperimental and Clinical Research Center (ECRC)Campus BuchBerlinGermany
- DZHK (German Centre for Cardiovascular Research)BerlinGermany
- Charité – Universitätsmedizin BerlinMedical Clinic for Nephrology and Internal Intensive CareCampus VirchowBerlinGermany
| |
Collapse
|
18
|
Zhou F, Rao F, Deng YQ, Yang H, Kuang SJ, Wu FL, Wu SL, Xue YM, Wu XM, Deng CY. Atorvastatin ameliorates the contractile dysfunction of the aorta induced by organ culture. Naunyn Schmiedebergs Arch Pharmacol 2018; 392:19-28. [DOI: 10.1007/s00210-018-1559-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 08/15/2018] [Indexed: 11/29/2022]
|
19
|
Rainbow RD, Brennan S, Jackson R, Beech AJ, Bengreed A, Waldschmidt HV, Tesmer JJG, Challiss RAJ, Willets JM. Small-Molecule G Protein-Coupled Receptor Kinase Inhibitors Attenuate G Protein-Coupled Receptor Kinase 2-Mediated Desensitization of Vasoconstrictor-Induced Arterial Contractions. Mol Pharmacol 2018; 94:1079-1091. [PMID: 29980659 DOI: 10.1124/mol.118.112524] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 06/29/2018] [Indexed: 01/01/2023] Open
Abstract
Vasoconstrictor-driven G protein-coupled receptor (GPCR)/phospholipase C (PLC) signaling increases intracellular Ca2+ concentration to mediate arterial contraction. To counteract vasoconstrictor-induced contraction, GPCR/PLC signaling can be desensitized by G protein-coupled receptor kinases (GRKs), with GRK2 playing a predominant role in isolated arterial smooth muscle cells. In this study, we use an array of GRK2 inhibitors to assess their effects on the desensitization of UTP and angiotensin II (AngII)-mediated arterial contractions. The effects of GRK2 inhibitors on the desensitization of UTP- or AngII-stimulated mesenteric third-order arterial contractions, and PLC activity in isolated mesenteric smooth muscle cells (MSMC), were determined using wire myography and Ca2+ imaging, respectively. Applying a stimulation protocol to cause receptor desensitization resulted in reductions in UTP- and AngII-stimulated arterial contractions. Preincubation with the GRK2 inhibitor paroxetine almost completely prevented desensitization of UTP- and attenuated desensitization of AngII-stimulated arterial contractions. In contrast, fluoxetine was ineffective. Preincubation with alternative GRK2 inhibitors (Takeda compound 101 or CCG224063) also attenuated the desensitization of UTP-mediated arterial contractile responses. In isolated MSMC, paroxetine, Takeda compound 101, and CCG224063 also attenuated the desensitization of UTP- and AngII-stimulated increases in Ca2+, whereas fluoxetine did not. In human uterine smooth muscle cells, paroxetine reversed GRK2-mediated histamine H1 receptor desensitization, but not GRK6-mediated oxytocin receptor desensitization. Utilizing various small-molecule GRK2 inhibitors, we confirm that GRK2 plays a central role in regulating vasoconstrictor-mediated arterial tone, highlighting a potentially novel strategy for blood pressure regulation through targeting GRK2 function.
Collapse
Affiliation(s)
- Richard D Rainbow
- Department of Molecular and Cell Biology, University of Leicester, Leicester, United Kingdom (A.B., R.A.J.C., J.M.W.); Department of Cardiovascular Sciences, University of Leicester, Clinical Sciences Wing, Glenfield General Hospital, Leicester, United Kingdom (R.D.R., S.B., R.J., A.J.B.); Life Sciences Institute and Departments of Pharmacology, Biological Sciences, and Medicinal Chemistry, University of Michigan, Ann Arbor, Michigan (H.V.W., J.J.G.T.); and Department of Biological Sciences, Purdue University, West Lafayette, Indiana (J.J.G.T.)
| | - Sean Brennan
- Department of Molecular and Cell Biology, University of Leicester, Leicester, United Kingdom (A.B., R.A.J.C., J.M.W.); Department of Cardiovascular Sciences, University of Leicester, Clinical Sciences Wing, Glenfield General Hospital, Leicester, United Kingdom (R.D.R., S.B., R.J., A.J.B.); Life Sciences Institute and Departments of Pharmacology, Biological Sciences, and Medicinal Chemistry, University of Michigan, Ann Arbor, Michigan (H.V.W., J.J.G.T.); and Department of Biological Sciences, Purdue University, West Lafayette, Indiana (J.J.G.T.)
| | - Robert Jackson
- Department of Molecular and Cell Biology, University of Leicester, Leicester, United Kingdom (A.B., R.A.J.C., J.M.W.); Department of Cardiovascular Sciences, University of Leicester, Clinical Sciences Wing, Glenfield General Hospital, Leicester, United Kingdom (R.D.R., S.B., R.J., A.J.B.); Life Sciences Institute and Departments of Pharmacology, Biological Sciences, and Medicinal Chemistry, University of Michigan, Ann Arbor, Michigan (H.V.W., J.J.G.T.); and Department of Biological Sciences, Purdue University, West Lafayette, Indiana (J.J.G.T.)
| | - Alison J Beech
- Department of Molecular and Cell Biology, University of Leicester, Leicester, United Kingdom (A.B., R.A.J.C., J.M.W.); Department of Cardiovascular Sciences, University of Leicester, Clinical Sciences Wing, Glenfield General Hospital, Leicester, United Kingdom (R.D.R., S.B., R.J., A.J.B.); Life Sciences Institute and Departments of Pharmacology, Biological Sciences, and Medicinal Chemistry, University of Michigan, Ann Arbor, Michigan (H.V.W., J.J.G.T.); and Department of Biological Sciences, Purdue University, West Lafayette, Indiana (J.J.G.T.)
| | - Amal Bengreed
- Department of Molecular and Cell Biology, University of Leicester, Leicester, United Kingdom (A.B., R.A.J.C., J.M.W.); Department of Cardiovascular Sciences, University of Leicester, Clinical Sciences Wing, Glenfield General Hospital, Leicester, United Kingdom (R.D.R., S.B., R.J., A.J.B.); Life Sciences Institute and Departments of Pharmacology, Biological Sciences, and Medicinal Chemistry, University of Michigan, Ann Arbor, Michigan (H.V.W., J.J.G.T.); and Department of Biological Sciences, Purdue University, West Lafayette, Indiana (J.J.G.T.)
| | - Helen V Waldschmidt
- Department of Molecular and Cell Biology, University of Leicester, Leicester, United Kingdom (A.B., R.A.J.C., J.M.W.); Department of Cardiovascular Sciences, University of Leicester, Clinical Sciences Wing, Glenfield General Hospital, Leicester, United Kingdom (R.D.R., S.B., R.J., A.J.B.); Life Sciences Institute and Departments of Pharmacology, Biological Sciences, and Medicinal Chemistry, University of Michigan, Ann Arbor, Michigan (H.V.W., J.J.G.T.); and Department of Biological Sciences, Purdue University, West Lafayette, Indiana (J.J.G.T.)
| | - John J G Tesmer
- Department of Molecular and Cell Biology, University of Leicester, Leicester, United Kingdom (A.B., R.A.J.C., J.M.W.); Department of Cardiovascular Sciences, University of Leicester, Clinical Sciences Wing, Glenfield General Hospital, Leicester, United Kingdom (R.D.R., S.B., R.J., A.J.B.); Life Sciences Institute and Departments of Pharmacology, Biological Sciences, and Medicinal Chemistry, University of Michigan, Ann Arbor, Michigan (H.V.W., J.J.G.T.); and Department of Biological Sciences, Purdue University, West Lafayette, Indiana (J.J.G.T.)
| | - R A John Challiss
- Department of Molecular and Cell Biology, University of Leicester, Leicester, United Kingdom (A.B., R.A.J.C., J.M.W.); Department of Cardiovascular Sciences, University of Leicester, Clinical Sciences Wing, Glenfield General Hospital, Leicester, United Kingdom (R.D.R., S.B., R.J., A.J.B.); Life Sciences Institute and Departments of Pharmacology, Biological Sciences, and Medicinal Chemistry, University of Michigan, Ann Arbor, Michigan (H.V.W., J.J.G.T.); and Department of Biological Sciences, Purdue University, West Lafayette, Indiana (J.J.G.T.)
| | - Jonathon M Willets
- Department of Molecular and Cell Biology, University of Leicester, Leicester, United Kingdom (A.B., R.A.J.C., J.M.W.); Department of Cardiovascular Sciences, University of Leicester, Clinical Sciences Wing, Glenfield General Hospital, Leicester, United Kingdom (R.D.R., S.B., R.J., A.J.B.); Life Sciences Institute and Departments of Pharmacology, Biological Sciences, and Medicinal Chemistry, University of Michigan, Ann Arbor, Michigan (H.V.W., J.J.G.T.); and Department of Biological Sciences, Purdue University, West Lafayette, Indiana (J.J.G.T.)
| |
Collapse
|
20
|
Liu B, Shi R, Li X, Liu Y, Feng X, Chen X, Fan X, Zhang Y, Zhang W, Tang J, Zhou X, Li N, Lu X, Xu Z. Downregulation of L-Type Voltage-Gated Ca 2+, Voltage-Gated K +, and Large-Conductance Ca 2+-Activated K + Channels in Vascular Myocytes From Salt-Loading Offspring Rats Exposed to Prenatal Hypoxia. J Am Heart Assoc 2018; 7:JAHA.117.008148. [PMID: 29545262 PMCID: PMC5907567 DOI: 10.1161/jaha.117.008148] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Background Prenatal hypoxia is suggested to be associated with increased risks of hypertension in offspring. This study tested whether prenatal hypoxia resulted in salt‐sensitive offspring and its related mechanisms of vascular ion channel remodeling. Methods and Results Pregnant rats were housed in a normoxic (21% O2) or hypoxic (10.5% O2) chamber from gestation days 5 to 21. A subset of male offspring received a high‐salt diet (8% NaCl) from 4 to 12 weeks after birth. Blood pressure was significantly increased only in the salt‐loading offspring exposed to prenatal hypoxia, not in the offspring that received regular diets and in control offspring provided with high‐salt diets. In mesenteric artery myocytes from the salt‐loading offspring with prenatal hypoxia, depolarized resting membrane potential was associated with decreased density of L‐type voltage‐gated Ca2+ (Cav1.2) and voltage‐gated K+ channel currents and decreased calcium sensitive to the large‐conductance Ca2+‐activated K+ channels. Protein expression of the L‐type voltage‐gated Ca2+ α1C subunit, large‐conductance calcium‐activated K+ channel (β1, not α subunits), and voltage‐gated K+ channel (KV2.1, not KV1.5 subunits) was also decreased in the arteries of salt‐loading offspring with prenatal hypoxia. Conclusions The results demonstrated that chronic prenatal hypoxia may program salt‐sensitive hypertension in male offspring, providing new information of ion channel remodeling in hypertensive myocytes. This information paves the way for early prevention and treatments of salt‐induced hypertension related to developmental problems in fetal origins.
Collapse
Affiliation(s)
- Bailin Liu
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Ruixiu Shi
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Xiang Li
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Yanping Liu
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Xueqin Feng
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Xueyi Chen
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Xiaorong Fan
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Yingying Zhang
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Wenna Zhang
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Jiaqi Tang
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Xiuwen Zhou
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Na Li
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Xiyuan Lu
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Zhice Xu
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China .,Center for Perinatal Biology, Loma Linda University, Loma Linda, CA
| |
Collapse
|
21
|
Effects of Hyperglycemia on Vascular Smooth Muscle Ca 2+ Signaling. BIOMED RESEARCH INTERNATIONAL 2017; 2017:3691349. [PMID: 28713824 PMCID: PMC5497615 DOI: 10.1155/2017/3691349] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/04/2016] [Accepted: 05/24/2017] [Indexed: 12/25/2022]
Abstract
Diabetes is a complex disease that is characterized with hyperglycemia, dyslipidemia, and insulin resistance. These pathologies are associated with significant cardiovascular implications that affect both the macro- and microvasculature. It is therefore important to understand the effects of various pathologies associated with diabetes on the vasculature. Here we directly test the effects of hyperglycemia on vascular smooth muscle (VSM) Ca2+ signaling in an isolated in vitro system using the A7r5 rat aortic cell line as a model. We find that prolonged exposure of A7r5 cells to hyperglycemia (weeks) is associated with changes to Ca2+ signaling, including most prominently an inhibition of the passive ER Ca2+ leak and the sarcoplasmic reticulum Ca2+-ATPase (SERCA). To translate these findings to the in vivo condition, we used primary VSM cells from normal and diabetic subjects and find that only the inhibition of the ER Ca2+ leaks replicates in cells from diabetic donors. These results show that prolonged hyperglycemia in isolation alters the Ca2+ signaling machinery in VSM cells. However, these alterations are not readily translatable to the whole organism situation where alterations to the Ca2+ signaling machinery are different.
Collapse
|
22
|
Ets HK, Seow CY, Moreland RS. Sustained Contraction in Vascular Smooth Muscle by Activation of L-type Ca 2+ Channels Does Not Involve Ca 2+ Sensitization or Caldesmon. Front Pharmacol 2017; 7:516. [PMID: 28082901 PMCID: PMC5183594 DOI: 10.3389/fphar.2016.00516] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Accepted: 12/12/2016] [Indexed: 11/13/2022] Open
Abstract
Vascular smooth muscle (VSM) is unique in its ability to maintain an intrinsic level of contractile force, known as tone. Vascular tone is believed to arise from the constitutive activity of membrane-bound L-type Ca2+ channels (LTCC). This study used a pharmacological agonist of LTCC, Bay K8644, to elicit a sustained, sub-maximal contraction in VSM that mimics tone. Downstream signaling was investigated in order to determine what molecules are responsible for tone. Medial strips of swine carotid artery were stimulated with 100 nM Bay K8644 to induce a sustained level of force. Force and phosphorylation levels of myosin light chain (MLC), MAP kinase, MYPT1, CPI-17, and caldesmon were measured during Bay K8644 stimulation in the presence and absence of nifedipine, ML-7, U0126, bisindolylmaleimide (Bis), and H-1152. Nifedipine and ML-7 inhibited force and MLC phosphorylation in response to Bay K8644. Inhibition of Rho kinase (H-1152) but not PKC (Bis) inhibited Bay K8644 induced force. U0126 significantly increased Bay K8644-dependent force with no effect on MLC phosphorylation. Neither CPI-17 nor caldesmon phosphorylation were increased during the maintenance of sustained force. Our results suggest that force due to the influx of calcium through LTCCs is partially MLC phosphorylation-dependent but does not involve PKC or caldesmon. Interestingly, inhibition of MLC kinase (MLCK) and PKC significantly increased MAP kinase phosphorylation suggesting that MLCK and PKC may directly or indirectly inhibit MAP kinase activity during prolonged contractions induced by Bay K8544.
Collapse
Affiliation(s)
- Hillevi K Ets
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia PA, USA
| | - Chun Y Seow
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver BC, Canada
| | - Robert S Moreland
- Department of Pharmacology and Physiology, Drexel University College of Medicine, PhiladelphiaPA, USA; Department of Pathology and Laboratory Medicine, Drexel University College of Medicine, PhiladelphiaPA, USA
| |
Collapse
|
23
|
Lopez RJ, Byrne S, Vukcevic M, Sekulic-Jablanovic M, Xu L, Brink M, Alamelu J, Voermans N, Snoeck M, Clement E, Muntoni F, Zhou H, Radunovic A, Mohammed S, Wraige E, Zorzato F, Treves S, Jungbluth H. An RYR1 mutation associated with malignant hyperthermia is also associated with bleeding abnormalities. Sci Signal 2016; 9:ra68. [PMID: 27382027 DOI: 10.1126/scisignal.aad9813] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Malignant hyperthermia is a potentially fatal hypermetabolic disorder triggered by halogenated anesthetics and the myorelaxant succinylcholine in genetically predisposed individuals. About 50% of susceptible individuals carry dominant, gain-of-function mutations in RYR1 [which encodes ryanodine receptor type 1 (RyR1)], though they have normal muscle function and no overt clinical symptoms. RyR1 is predominantly found in skeletal muscle but also at lower amounts in immune and smooth muscle cells, suggesting that RYR1 mutations may have a wider range of effects than previously suspected. Mild bleeding abnormalities have been described in patients with malignant hyperthermia carrying gain-of-function RYR1 mutations. We sought to determine the frequency and molecular basis for this symptom. We found that some patients with specific RYR1 mutations had abnormally high bleeding scores, whereas their healthy relatives did not. Knock-in mice with the malignant hyperthermia susceptibility RYR1 mutation Y522S (MHS RYR1Y522S) had longer bleeding times than their wild-type littermates. Primary vascular smooth muscle cells from RYR1Y522S knock-in mice exhibited a higher frequency of subplasmalemmal Ca(2+) sparks, leading to a more negative resting membrane potential. The bleeding defect of RYR1Y522S mice and of one patient was reversed by treatment with the RYR1 antagonist dantrolene, and Ca(2+) sparks in primary vascular smooth muscle cells from the MHS RYR1Y522S mice were blocked by ryanodine or dantrolene. Thus, RYR1 mutations may lead to prolonged bleeding by altering vascular smooth muscle cell function. The reversibility of the bleeding phenotype emphasizes the potential therapeutic value of dantrolene in the treatment of such bleeding disorders.
Collapse
Affiliation(s)
- Rubén J Lopez
- Departments of Biomedicine and Anesthesia, Basel University Hospital, Hebelstrasse 20, 4031 Basel, Switzerland
| | - Susan Byrne
- Department of Paediatric Neurology, Neuromuscular Service, Evelina Children's Hospital, St Thomas' Hospital, London SE1 7EH, UK
| | - Mirko Vukcevic
- Departments of Biomedicine and Anesthesia, Basel University Hospital, Hebelstrasse 20, 4031 Basel, Switzerland. Department of Biomedicine, Basel University Hospital, Hebelstrasse 20, 4031 Basel, Switzerland
| | - Marijana Sekulic-Jablanovic
- Departments of Biomedicine and Anesthesia, Basel University Hospital, Hebelstrasse 20, 4031 Basel, Switzerland
| | - Lifen Xu
- Department of Biomedicine, Basel University Hospital, Hebelstrasse 20, 4031 Basel, Switzerland
| | - Marijke Brink
- Department of Biomedicine, Basel University Hospital, Hebelstrasse 20, 4031 Basel, Switzerland
| | - Jay Alamelu
- Department of Haematology, Evelina Children's Hospital, St Thomas' Hospital, London SE1 7EH, UK
| | - Nicol Voermans
- Department of Neurology, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Marc Snoeck
- National MH Investigation Unit, Department of Anesthesiology, Canisius Wilhelmina Hospital, 6532 Nijmegen, Netherlands
| | - Emma Clement
- Department of Clinical Genetics, Guy's Hospital, London SE1 7EH, UK
| | - Francesco Muntoni
- Dubowitz Neuromuscular Centre, Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Haiyan Zhou
- Dubowitz Neuromuscular Centre, Institute of Child Health, University College London, London WC1N 1EH, UK
| | | | - Shehla Mohammed
- Department of Clinical Genetics, Guy's Hospital, London SE1 7EH, UK
| | - Elizabeth Wraige
- Department of Paediatric Neurology, Neuromuscular Service, Evelina Children's Hospital, St Thomas' Hospital, London SE1 7EH, UK
| | - Francesco Zorzato
- Departments of Biomedicine and Anesthesia, Basel University Hospital, Hebelstrasse 20, 4031 Basel, Switzerland. Department of Life Sciences, General Pathology Section, University of Ferrara, Via Borsari 46, 44100 Ferrara, Italy
| | - Susan Treves
- Departments of Biomedicine and Anesthesia, Basel University Hospital, Hebelstrasse 20, 4031 Basel, Switzerland. Department of Life Sciences, General Pathology Section, University of Ferrara, Via Borsari 46, 44100 Ferrara, Italy
| | - Heinz Jungbluth
- Department of Paediatric Neurology, Neuromuscular Service, Evelina Children's Hospital, St Thomas' Hospital, London SE1 7EH, UK. Randall Division of Cell and Molecular Biophysics, Muscle Signalling Section, King's College, London SE1 1UL, UK. Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London SE5 9RX, UK
| |
Collapse
|
24
|
MISÁRKOVÁ E, BEHULIAK M, BENCZE M, ZICHA J. Excitation-Contraction Coupling and Excitation-Transcription Coupling in Blood Vessels: Their Possible Interactions in Hypertensive Vascular Remodeling. Physiol Res 2016; 65:173-91. [DOI: 10.33549/physiolres.933317] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Vascular smooth muscle cells (VSMC) display considerable phenotype plasticity which can be studied in vivo on vascular remodeling which occurs during acute or chronic vascular injury. In differentiated cells, which represent contractile phenotype, there are characteristic rapid transient changes of intracellular Ca2+ concentration ([Ca2+]i), while the resting cytosolic [Ca2+]i concentration is low. It is mainly caused by two components of the Ca2+ signaling pathways: Ca2+ entry via L-type voltage-dependent Ca2+ channels and dynamic involvement of intracellular stores. Proliferative VSMC phenotype is characterized by long-lasting [Ca2+]i oscillations accompanied by sustained elevation of basal [Ca2+]i. During the switch from contractile to proliferative phenotype there is a general transition from voltage-dependent Ca2+ entry to voltage-independent Ca2+ entry into the cell. These changes are due to the altered gene expression which is dependent on specific transcription factors activated by various stimuli. It is an open question whether abnormal VSMC phenotype reported in rats with genetic hypertension (such as spontaneously hypertensive rats) might be partially caused by a shift from contractile to proliferative VSMC phenotype.
Collapse
Affiliation(s)
| | | | | | - J. ZICHA
- Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic
| |
Collapse
|
25
|
An overview of potential molecular mechanisms involved in VSMC phenotypic modulation. Histochem Cell Biol 2015; 145:119-30. [DOI: 10.1007/s00418-015-1386-3] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/03/2015] [Indexed: 12/21/2022]
|
26
|
Mesenchymal stromal cells for sphincter regeneration. Adv Drug Deliv Rev 2015; 82-83:123-36. [PMID: 25451135 DOI: 10.1016/j.addr.2014.10.026] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Revised: 09/29/2014] [Accepted: 10/15/2014] [Indexed: 02/06/2023]
Abstract
Stress urinary incontinence (SUI), defined as the involuntary loss of considerable amounts of urine during increased abdominal pressure (exertion, effort, sneezing, coughing, etc.), is a severe problem to the individuals affected and a significant medical, social and economic challenge. SUI is associated with pelvic floor debility, absence of detrusor contraction, or a loss of control over the sphincter muscle apparatus. The pathology includes an increasing loss of muscle cells, replacement of muscular tissue with fibrous tissue, and general aging associated processes of the sphincter complex. When current therapies fail to cure or improve SUI, application of regeneration-competent cells may be an alternative therapeutic option. Here we discuss different aspects of the biology of mesenchymal stromal cells, which are relevant to their clinical applications and for regenerating the sphincter complex. However, there are reports in favor of and against cell-based therapies. We therefore summarize the potential and the risks of cell-based therapies for the treatment of SUI.
Collapse
|
27
|
Wan XJ, Zhao HC, Zhang P, Huo B, Shen BR, Yan ZQ, Qi YX, Jiang ZL. Involvement of BK channel in differentiation of vascular smooth muscle cells induced by mechanical stretch. Int J Biochem Cell Biol 2015; 59:21-9. [DOI: 10.1016/j.biocel.2014.11.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Revised: 11/13/2014] [Accepted: 11/25/2014] [Indexed: 12/26/2022]
|
28
|
Ouyang QF, Han Y, Lin ZH, Xie H, Xu CS, Xie LD. Fluvastatin upregulates the α 1C subunit of CaV1.2 channel expression in vascular smooth muscle cells via RhoA and ERK/p38 MAPK pathways. DISEASE MARKERS 2014; 2014:237067. [PMID: 25614710 PMCID: PMC4295146 DOI: 10.1155/2014/237067] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Revised: 12/09/2014] [Accepted: 12/11/2014] [Indexed: 11/18/2022]
Abstract
Abnormal phenotypic switch of vascular smooth muscle cell (VSMC) is a hallmark of vascular disorders such as atherosclerosis and restenosis. And this process has been related to remodeling of L-type calcium channel (LTCC). We attempted to investigate whether fluvastatin has any effect on VSMC proliferation and LTCCα 1C subunit (LTCCα 1C) expression as well as the potential mechanisms involved. The VSMCs proliferation was assayed by osteopontin immunofluorescent staining and [(3)H]-thymidine incorporation. The cell cycle was detected by flow cytometric analysis. The activity of RhoA was determined with pull-down assay. MAPK activity and LTCCα 1C expression were assessed by western blotting. We demonstrated fluvastatin prevented the VSMCs dedifferentiating into a proliferative phenotype and induced cell cycle arrest in the G0/G1 phase in response to PDGF-BB stimulation. Fluvastatin dose-dependently reversed the downregulation of LTCCα 1C expression induced by PDGF-BB. Inhibition of ROCK, ERK, or p38 MAPK activation largely enhanced the upregulation effect of fluvastatin (P < 0.01). However, blockade of JNK pathway had no effect on LTCCα 1C expression. We concluded LTCCα 1C was a VSMC contractile phenotype marker gene. Fluvastatin upregulated LTCCα 1C expression, at least in part, by inhibiting ROCK, ERK1/2, and p38 MAPK activation. Fluvastatin may be a potential candidate for preventing or treating vascular diseases.
Collapse
MESH Headings
- Animals
- Calcium Channels, L-Type/genetics
- Calcium Channels, L-Type/metabolism
- Cell Cycle Checkpoints
- Cell Proliferation
- Fatty Acids, Monounsaturated/pharmacology
- Fluvastatin
- Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology
- Indoles/pharmacology
- MAP Kinase Signaling System
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Platelet-Derived Growth Factor/physiology
- Protein Transport/drug effects
- Rats, Inbred SHR
- Up-Regulation/drug effects
- rhoA GTP-Binding Protein/metabolism
Collapse
Affiliation(s)
- Qiu-Fang Ouyang
- Fujian Hypertension Research Institute, The First Affiliated Hospital of Fujian Medical University, Fujian 350005, China
- Ultrasound Department, The Second Affiliated People's Hospital, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350003, China
| | - Ying Han
- Fujian Hypertension Research Institute, The First Affiliated Hospital of Fujian Medical University, Fujian 350005, China
| | - Zhi-Hong Lin
- Fujian Hypertension Research Institute, The First Affiliated Hospital of Fujian Medical University, Fujian 350005, China
| | - Hong Xie
- Fujian Hypertension Research Institute, The First Affiliated Hospital of Fujian Medical University, Fujian 350005, China
| | - Chang-Sheng Xu
- Fujian Hypertension Research Institute, The First Affiliated Hospital of Fujian Medical University, Fujian 350005, China
| | - Liang-Di Xie
- Fujian Hypertension Research Institute, The First Affiliated Hospital of Fujian Medical University, Fujian 350005, China
| |
Collapse
|
29
|
Mugnai P, Durante M, Sgaragli G, Saponara S, Paliuri G, Bova S, Fusi F. L-type Ca(2+) channel current characteristics are preserved in rat tail artery myocytes after one-day storage. Acta Physiol (Oxf) 2014; 211:334-45. [PMID: 24666564 DOI: 10.1111/apha.12282] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2013] [Revised: 09/04/2013] [Accepted: 03/21/2014] [Indexed: 12/14/2022]
Abstract
AIM To develop a cheap and simple method of storing for 24-h vascular tissue and single myocytes while preserving therein the biophysical and pharmacological characteristics of L-type Ca(2+) channels and contractile activity. METHODS Rings or vascular smooth muscle cells obtained from the rat tail main artery were used either freshly (R0h and VSMC0h) or stored for 24 h (R24h and VSMC24h) at 4 °C, to record whole-cell L-type Ca(2+) currents (IC a(L) ) or measure contractile responses. RESULTS R0h/VSMC0h and R24h/VSMC24h comparably contracted when stimulated with phenylephrine, high KCl or ATP. In both VSMC0h and VSMC24h, IC a(L) was identified and characterized as a stable inward current for at least 35 min; IC a(L) was comparably inhibited by the Ca(2+) antagonists nifedipine, verapamil and diltiazem and increased by the Ca(2+) channel agonist (S)-(-)-Bay K 8644; current density and current-voltage relationships were similar; at more hyperpolarized holding potentials, IC a(L) intensity increased comparably; nifedipine shifted the steady-state inactivation curve towards more negative potentials, while verapamil blocked IC a(L) in a frequency-dependent manner and slowed down the rate of recovery from inactivation in a comparable way. CONCLUSION Findings show that smooth muscle contractile activity and the biophysical and pharmacological features of L-type Ca(2+) channels are similar in VSMC24h and VSMC0h. The fact that reproducible results were obtained in vascular myocytes up to 24 h after dissociation may facilitate vascular smooth muscle cell investigation by increasing throughput and reducing the number of animals required.
Collapse
Affiliation(s)
- P. Mugnai
- Dipartimento di Scienze della Vita; Università di Siena; Siena Italy
| | - M. Durante
- Dipartimento di Scienze della Vita; Università di Siena; Siena Italy
| | - G. Sgaragli
- Dipartimento di Scienze della Vita; Università di Siena; Siena Italy
| | - S. Saponara
- Dipartimento di Scienze della Vita; Università di Siena; Siena Italy
| | - G. Paliuri
- Dipartimento di Scienze del Farmaco; Università degli Studi di Padova; Padova Italy
| | - S. Bova
- Dipartimento di Scienze del Farmaco; Università degli Studi di Padova; Padova Italy
| | - F. Fusi
- Dipartimento di Scienze della Vita; Università di Siena; Siena Italy
| |
Collapse
|
30
|
Kudryavtseva O, Herum KM, Dam VS, Straarup MS, Kamaev D, Briggs Boedtkjer DM, Matchkov VV, Aalkjær C. Downregulation of L-type Ca2+ channel in rat mesenteric arteries leads to loss of smooth muscle contractile phenotype and inward hypertrophic remodeling. Am J Physiol Heart Circ Physiol 2014; 306:H1287-301. [DOI: 10.1152/ajpheart.00503.2013] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
L-type Ca2+ channels (LTCCs) are important for vascular smooth muscle cell (VSMC) contraction, as well as VSMC differentiation, as indicated by loss of LTCCs during VSMC dedifferentiation. However, it is not clear whether loss of LTCCs is a primary event underlying phenotypic modulation or whether loss of LTCCs has significance for vascular structure. We used small interference RNA (siRNA) transfection in vivo to investigate the role of LTCCs in VSMC phenotypic expression and structure of rat mesenteric arteries. siRNA reduced LTCC mRNA and protein expression in rat mesenteric arteries 3 days after siRNA transfection to 12.7 ± 0.7% and 47.3 ± 13%, respectively: this was associated with an increased resting intracellular Ca2+ concentration ([Ca2+]i). Despite the high [Ca2+]i, the contractility was reduced (tension development to norepinephrine was 3.5 ± 0.2 N/m and 0.8 ± 0.2 N/m for sham-transfected and downregulated arteries respectively; P < 0.05). Expression of contractile phenotype marker genes was reduced in arteries downregulated for LTCCs. Phenotypic changes were associated with a 45% increase in number of VSMCs and a consequent increase of media thickness and media area. Ten days after siRNA transfection arterial structure was again normalized. The contractile responses of LTCC-siRNA transfected arteries were elevated in comparison with matched controls 10 days after transfection. The study provides strong evidence for causal relationships between LTCC expression and VSMC contractile phenotype, as well as novel data addressing the complex relationship between VSMC contractility, phenotype, and vascular structure. These findings are relevant for understanding diseases, associated with phenotype changes of VSMC and vascular remodeling, such as atherosclerosis and hypertension.
Collapse
Affiliation(s)
- Olga Kudryavtseva
- Department of Biomedicine, Membranes, Aarhus University, Aarhus C, Denmark; and
| | - Kate Møller Herum
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Vibeke Secher Dam
- Department of Biomedicine, Membranes, Aarhus University, Aarhus C, Denmark; and
| | | | - Dmitry Kamaev
- Department of Biomedicine, Membranes, Aarhus University, Aarhus C, Denmark; and
| | | | | | - Christian Aalkjær
- Department of Biomedicine, Membranes, Aarhus University, Aarhus C, Denmark; and
| |
Collapse
|
31
|
Duckles H, Boycott HE, Al-Owais MM, Elies J, Johnson E, Dallas ML, Porter KE, Giuntini F, Boyle JP, Scragg JL, Peers C. Heme oxygenase-1 regulates cell proliferation via carbon monoxide-mediated inhibition of T-type Ca2+ channels. Pflugers Arch 2014; 467:415-27. [PMID: 24744106 PMCID: PMC4293494 DOI: 10.1007/s00424-014-1503-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Revised: 03/14/2014] [Accepted: 03/14/2014] [Indexed: 11/24/2022]
Abstract
Induction of the antioxidant enzyme heme oxygenase-1 (HO-1) affords cellular protection and suppresses proliferation of vascular smooth muscle cells (VSMCs) associated with a variety of pathological cardiovascular conditions including myocardial infarction and vascular injury. However, the underlying mechanisms are not fully understood. Over-expression of Cav3.2 T-type Ca2+ channels in HEK293 cells raised basal [Ca2+]i and increased proliferation as compared with non-transfected cells. Proliferation and [Ca2+]i levels were reduced to levels seen in non-transfected cells either by induction of HO-1 or exposure of cells to the HO-1 product, carbon monoxide (CO) (applied as the CO releasing molecule, CORM-3). In the aortic VSMC line A7r5, proliferation was also inhibited by induction of HO-1 or by exposure of cells to CO, and patch-clamp recordings indicated that CO inhibited T-type (as well as L-type) Ca2+ currents in these cells. Finally, in human saphenous vein smooth muscle cells, proliferation was reduced by T-type channel inhibition or by HO-1 induction or CO exposure. The effects of T-type channel blockade and HO-1 induction were non-additive. Collectively, these data indicate that HO-1 regulates proliferation via CO-mediated inhibition of T-type Ca2+ channels. This signalling pathway provides a novel means by which proliferation of VSMCs (and other cells) may be regulated therapeutically.
Collapse
Affiliation(s)
- Hayley Duckles
- Division of Cardiovascular and Diabetes Research, LIGHT, Faculty of Medicine and Health, University of Leeds, Clarendon Way, Leeds, LS2 9JT, UK
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Kinikoglu B, Kong Y, Liao EC. Characterization of cultured multipotent zebrafish neural crest cells. Exp Biol Med (Maywood) 2013; 239:159-68. [PMID: 24326414 DOI: 10.1177/1535370213513997] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The neural crest is a unique cell population associated with vertebrate evolution. Neural crest cells (NCCs) are characterized by their multipotent and migratory potentials. While zebrafish is a powerful genetic model organism, the isolation and culture of zebrafish NCCs would provide a useful adjunct to fully interrogate the genetic networks that regulate NCC development. Here we report for the first time the isolation, in vitro culture, and characterization of NCCs from zebrafish embryos. NCCs were isolated from transgenic sox10:egfp embryos using fluorescence activated cell sorting and cultured in complex culture medium without feeder layers. NCC multilineage differentiation was determined by immunocytochemistry and real-time qPCR, cell migration was assessed by wound healing assay, and the proliferation index was calculated by immunostaining against the mitosis marker phospho-histone H3. Cultured NCCs expressed major neural crest lineage markers such as sox10, sox9a, hnk1, p75, dlx2a, and pax3, and the pluripotency markers c-myc and klf4. We showed that the cultured NCCs can be differentiated into multiple neural crest lineages, contributing to neurons, glial cells, smooth muscle cells, melanocytes, and chondrocytes. We applied the NCC in vitro model to study the effect of retinoic acid on NCC development. We showed that retinoic acid had a profound effect on NCC morphology and differentiation, significantly inhibited proliferation and enhanced cell migration. The availability of high numbers of NCCs and reproducible functional assays offers new opportunities for mechanistic studies of neural crest development, in genetic and chemical biology applications.
Collapse
Affiliation(s)
- Beste Kinikoglu
- Center for Regenerative Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | | | | |
Collapse
|
33
|
Shi XZ, Sarna SK. Cell culture retains contractile phenotype but epigenetically modulates cell-signaling proteins of excitation-contraction coupling in colon smooth muscle cells. Am J Physiol Gastrointest Liver Physiol 2013; 304:G337-45. [PMID: 23238936 PMCID: PMC3566616 DOI: 10.1152/ajpgi.00369.2012] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2012] [Accepted: 12/11/2012] [Indexed: 01/31/2023]
Abstract
Smooth muscle cell cultures are used frequently to investigate the cellular mechanisms of contraction. We tested the hypothesis that cell culture alters the expression of select cell-signaling proteins of excitation-contraction coupling in colon smooth muscle cells without altering the contractile phenotype. We used muscularis externa (ME) tissues, freshly dispersed cells (FC), primary cell cultures (PC), and resuspensions of cell cultures (RC). Colon smooth muscle cells retained their phenotype in all states. We investigated expression of 10 cell-signaling proteins of excitation-contraction coupling in all four types of tissue. Expression of all these proteins did not differ between ME and FC (P > 0.05). However, expression of the α(1C)-subunit of Ca(v)1.2b, myosin light chain kinase, myosin phosphatase target subunit 1, and 17-kDa C kinase-potentiated protein phosphatase-1 inhibitor (CPI-17) decreased in PC and RC vs. ME and FC (all P < 0.05). Expression of Gα(i3), serine/threonine protein phosphatase-1 β-catalytic subunit, and Rho kinase 1 increased in PC and RC vs. ME and FC (all P < 0.05). Cell culture and resuspension downregulated expression of α-actin and calponin, but not myosin heavy chain. The net effect of these molecular changes was suppression of cell reactivity to ACh in RC vs. FC. Overexpression of CPI-17 in PC partially reversed the suppression of contractility in resuspended cells. Methylation-specific PCR showed increased methylation of the Cpi-17 gene promoter in PC vs. ME (P < 0.05). We concluded that smooth muscle cells retain their contractile phenotype in culture. However, reactivity to ACh declines because of altered expression of specific cell-signaling proteins involved in excitation-contraction coupling. DNA methylation of the Cpi-17 promoter may contribute to its gene suppression.
Collapse
Affiliation(s)
- Xuan-Zheng Shi
- Enteric Neuromuscular Disorders and Visceral Pain Center, Division of Gastroenterology, Department of Internal Medicine, University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA
| | | |
Collapse
|
34
|
Nguemo F, Fleischmann BK, Gupta MK, Šarić T, Malan D, Liang H, Pfannkuche K, Bloch W, Schunkert H, Hescheler J, Reppel M. The L-type Ca2+ channels blocker nifedipine represses mesodermal fate determination in murine embryonic stem cells. PLoS One 2013; 8:e53407. [PMID: 23320083 PMCID: PMC3539992 DOI: 10.1371/journal.pone.0053407] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2012] [Accepted: 11/28/2012] [Indexed: 01/20/2023] Open
Abstract
Dihydropyridines (DHP), which nifedipine is a member of, preferentially block Ca(2+) channels of different cell types. Moreover, influx of Ca(2+) through L-type Ca(2+) channels (LTCCs) activates Ca(2+) signaling pathways, which in turn contribute to numerous cellular processes. Although LTCCs are expressed in undifferentiated cells, very little is known about its contributions to the transcriptional regulation of mesodermal and cardiac genes. This study aimed to examine the contribution of LTCCs and the effect of nifedipine on the commitment of pluripotent stem cells toward the cardiac lineage in vitro. The murine embryonic stem (ES, cell line D3) and induced pluripotent stem (iPS, cell clone 09) cells were differentiated into enhanced green fluorescence protein (EGFP) expressing spontaneously beating cardiomyocytes (CMs). Early treatment of differentiating cells with 10 µM nifedipine led to a significant inhibition of the cardiac mesoderm formation and cardiac lineage commitment as revealed by gene regulation analysis. This was accompanied by the inhibition of spontaneously occurring Ca(2+) transient and reduction of LTCCs current density (I(CaL)) of differentiated CMs. In addition, nifedipine treatment instigated a pronounced delay of the spontaneous beating embryoid body (EB) and led to a poor surface localization of L-type Ca(2+) channel α(1C) (Ca(V)1.2) subunits. Contrary late incubation of pluripotent stem cells with nifedipine was without any impact on the differentiation process and did not affect the derived CMs function. Our data indicate that nifedipine blocks the determined path of pluripotent stem cells to cardiomyogenesis by inhibition of mesodermal commitment at early stages of differentiation, thus the proper upkeep Ca(2+) concentration and pathways are essentially required for cardiac gene expression, differentiation and function.
Collapse
Affiliation(s)
- Filomain Nguemo
- Institute of Neurophysiology, University of Cologne, Cologne, Germany
| | - Bernd K. Fleischmann
- Institute of Physiology I, Life and Brain Center, University of Bonn, Bonn, Germany
| | - Manoj K. Gupta
- Institute of Neurophysiology, University of Cologne, Cologne, Germany
| | - Tomo Šarić
- Institute of Neurophysiology, University of Cologne, Cologne, Germany
| | - Daniela Malan
- Institute of Physiology I, Life and Brain Center, University of Bonn, Bonn, Germany
| | - Huamin Liang
- Department of Physiology, Huazhong University of Science and Technology, Tongji Medical College, Wuhan, China
| | - Kurt Pfannkuche
- Institute of Neurophysiology, University of Cologne, Cologne, Germany
| | - Wilhelm Bloch
- Department of Molecular and Cellular Sport Medicine, German Sport University, Cologne, Germany
| | | | - Jürgen Hescheler
- Institute of Neurophysiology, University of Cologne, Cologne, Germany
| | - Michael Reppel
- Institute of Neurophysiology, University of Cologne, Cologne, Germany
- Department of Cardiology, Medical University of Lübeck, Lübeck, Germany
| |
Collapse
|
35
|
Wang T, Kendig DM, Chang S, Trappanese DM, Chacko S, Moreland RS. Bladder smooth muscle organ culture preparation maintains the contractile phenotype. Am J Physiol Renal Physiol 2012; 303:F1382-97. [PMID: 22896042 PMCID: PMC3518193 DOI: 10.1152/ajprenal.00261.2011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2011] [Accepted: 08/13/2012] [Indexed: 01/26/2023] Open
Abstract
Smooth muscle cells, when subjected to culture, modulate from a contractile to a secretory phenotype. This has hampered the use of cell culture for molecular techniques to study the regulation of smooth muscle biology. The goal of this study was to develop a new organ culture model of bladder smooth muscle (BSM) that would maintain the contractile phenotype and aid in the study of BSM biology. Our results showed that strips of BSM subjected to up to 9 days of organ culture maintained their contractile phenotype, including the ability to achieve near-control levels of force with a temporal profile similar to that of noncultured tissues. The technical aspects of our organ culture preparation that were responsible, in part, for the maintenance of the contractile phenotype were a slight longitudinal stretch during culture and subjection of the strips to daily contraction-relaxation. The tissues contained viable cells throughout the cross section of the strips. There was an increase in extracellular collagenous matrix, resulting in a leftward shift in the passive length-tension relationship. There were no significant changes in the content of smooth muscle-specific α-actin, calponin, h-caldesmon, total myosin heavy chain, protein kinase G, Rho kinase-I, or the ratio of SM1 to SM2 myosin isoforms. Moreover the organ cultured tissues maintained functional voltage-gated calcium channels and large-conductance calcium-activated potassium channels. Therefore, we propose that this novel BSM organ culture model maintains the contractile phenotype and will be a valuable tool for the use in cellular/molecular biology studies of bladder myocytes.
Collapse
Affiliation(s)
- Tanchun Wang
- Dept. of Pharmacology and Physiology, Drexel Univ. College of Medicine, 245 N 15th St., MS 488, Philadelphia, PA 19102, USA
| | | | | | | | | | | |
Collapse
|
36
|
MEF2 is regulated by CaMKIIδ2 and a HDAC4-HDAC5 heterodimer in vascular smooth muscle cells. Biochem J 2012; 444:105-14. [PMID: 22360269 DOI: 10.1042/bj20120152] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
VSMCs (vascular smooth muscle cells) dedifferentiate from the contractile to the synthetic phenotype in response to acute vascular diseases such as restenosis and chronic vascular diseases such as atherosclerosis, and contribute to growth of the neointima. We demonstrated previously that balloon catheter injury of rat carotid arteries resulted in increased expression of CaMKII (Ca(2+)/calmodulin-dependent protein kinase) IIδ(2) in the medial wall and the expanding neointima [House and Singer (2008) Arterioscler. Thromb. Vasc. Biol. 28, 441-447]. These findings led us to hypothesize that increased expression of CaMKIIδ(2) is a positive mediator of synthetic VSMCs. HDAC (histone deacetylase) 4 and HDAC5 function as transcriptional co-repressors and are regulated in a CaMKII-dependent manner. In the present paper, we report that endogenous HDAC4 and HDAC5 in VSMCs are activated in a Ca(2+)- and CaMKIIδ(2)-dependent manner. We show further that AngII (angiotensin II)- and PDGF (platelet-derived growth factor)-dependent phosphorylation of HDAC4 and HDAC5 is reduced when CaMKIIδ(2) expression is suppressed or CaMKIIδ(2) activity is attenuated. The transcriptional activator MEF2 (myocyte-enhancer factor 2) is an important determinant of VSMC phenotype and is regulated in an HDAC-dependent manner. In the present paper, we report that stimulation of VSMCs with ionomycin or AngII potentiates MEF2's ability to bind DNA and increases the expression of established MEF2 target genes Nur77 (nuclear receptor 77) (NR4A1) and MCP1 (monocyte chemotactic protein 1) (CCL2). Suppression of CaMKIIδ(2) attenuates increased MEF2 DNA-binding activity and up-regulation of Nur77 and MCP1. Finally, we show that HDAC5 is regulated by HDAC4 in VSMCs. Suppression of HDAC4 expression and activity prevents AngII- and PDGF-dependent phosphorylation of HDAC5. Taken together, these results illustrate a mechanism by which CaMKIIδ(2) mediates MEF2-dependent gene transcription in VSMCs through regulation of HDAC4 and HDAC5.
Collapse
|
37
|
SERCA2a gene transfer prevents intimal proliferation in an organ culture of human internal mammary artery. Gene Ther 2012; 20:396-406. [PMID: 22763406 PMCID: PMC3465616 DOI: 10.1038/gt.2012.50] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Coronary restenosis, a major complication of percutaneous balloon angioplasty, results from neointimal proliferation of vascular smooth muscle cells (VSMCs). The sarco/endoplasmic reticulum calcium ATPase isoform 2a (SERCA2a), specific to contractile VSMCs, has been reported previously to be involved in the control of the Ca2+-signaling pathways governing proliferation and migration. Moreover, SERCA2a gene transfer was reported to inhibit in vitro VSMC proliferation and to prevent neointimal thickening in a rat carotid injury model. The aim of this study was to evaluate the potential therapeutic interest of SERCA2a gene transfer for prevention of in-stent restenosis using a human ex vivo model of left internal mammary artery (hIMA) intimal thickening. Left hIMAs, obtained at the time of aorto-coronary bypass surgeries, were subjected to balloon dilatation followed by infection for 30 min with adenoviruses encoding either human SERCA2 and GFP or control gene (beta-galactosidase) and GFP. Proliferation of subendothelial VSMCs and neointimal thickening were observed in balloon-injured hIMA maintained 14 days in organ culture under constant pressure and perfusion. SERCA2a gene transfer prevented vascular remodeling and significantly (p<0.01, n=5) reduced neointimal thickening in injured arteries (intima/media ratio was 0.07 ± 0.01 vs 0.40 ± 0.03 in βGal-infected arteries). These findings could have potential implications for treatment of pathological in stent-restenosis.
Collapse
|
38
|
Turczyńska KM, Sadegh MK, Hellstrand P, Swärd K, Albinsson S. MicroRNAs are essential for stretch-induced vascular smooth muscle contractile differentiation via microRNA (miR)-145-dependent expression of L-type calcium channels. J Biol Chem 2012; 287:19199-206. [PMID: 22474293 PMCID: PMC3365952 DOI: 10.1074/jbc.m112.341073] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2012] [Revised: 03/26/2012] [Indexed: 11/06/2022] Open
Abstract
Stretch of the vascular wall is an important stimulus to maintain smooth muscle contractile differentiation that is known to depend on L-type calcium influx, Rho-activation, and actin polymerization. The role of microRNAs in this response was investigated using tamoxifen-inducible and smooth muscle-specific Dicer KO mice. In the absence of Dicer, which is required for microRNA maturation, smooth muscle microRNAs were completely ablated. Stretch-induced contractile differentiation and Rho-dependent cofilin-2 phosphorylation were dramatically reduced in Dicer KO vessels. On the other hand, acute stretch-sensitive growth signaling, which is independent of influx through L-type calcium channels, was not affected by Dicer KO. Contractile differentiation induced by the actin polymerizing agent jasplakinolide was not altered by deletion of Dicer, suggesting an effect upstream of actin polymerization. Basal and stretch-induced L-type calcium channel expressions were both decreased in Dicer KO portal veins, and inhibition of L-type channels in control vessels mimicked the effects of Dicer deletion. Furthermore, inhibition of miR-145, a highly expressed microRNA in smooth muscle, resulted in a similar reduction of L-type calcium channel expression. This was abolished by the Ca(2+)/calmodulin-dependent protein kinase II inhibitor KN93, suggesting that Ca(2+)/calmodulin-dependent protein kinase IIδ, a target of miR-145 and up-regulated in Dicer KO, plays a role in the regulation of L-type channel expression. These results show that microRNAs play a crucial role in stretch-induced contractile differentiation in the vascular wall in part via miR-145-dependent regulation of L-type calcium channels.
Collapse
MESH Headings
- Animals
- Calcium Channels, L-Type/biosynthesis
- Calcium Channels, L-Type/genetics
- Calcium Signaling/drug effects
- Calcium Signaling/physiology
- Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics
- Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism
- Cells, Cultured
- DEAD-box RNA Helicases/genetics
- DEAD-box RNA Helicases/metabolism
- Male
- Mice
- Mice, Knockout
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Muscle Proteins/genetics
- Muscle Proteins/metabolism
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/cytology
- Myocytes, Smooth Muscle/metabolism
- Protein Kinase Inhibitors/pharmacology
- Ribonuclease III/genetics
- Ribonuclease III/metabolism
- Up-Regulation/drug effects
- Up-Regulation/physiology
Collapse
Affiliation(s)
- Karolina M. Turczyńska
- From the Department of Experimental Medical Science, Lund University, 221 84 Lund, Sweden
| | | | - Per Hellstrand
- From the Department of Experimental Medical Science, Lund University, 221 84 Lund, Sweden
| | - Karl Swärd
- From the Department of Experimental Medical Science, Lund University, 221 84 Lund, Sweden
| | - Sebastian Albinsson
- From the Department of Experimental Medical Science, Lund University, 221 84 Lund, Sweden
| |
Collapse
|
39
|
Povlsen GK, Waldsee R, Ahnstedt H, Kristiansen KA, Johansen FF, Edvinsson L. In vivo experimental stroke and in vitro organ culture induce similar changes in vasoconstrictor receptors and intracellular calcium handling in rat cerebral arteries. Exp Brain Res 2012; 219:507-20. [PMID: 22585122 DOI: 10.1007/s00221-012-3108-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2012] [Accepted: 04/25/2012] [Indexed: 12/11/2022]
Abstract
Cerebral arteries subjected to different types of experimental stroke upregulate their expression of certain G-protein-coupled vasoconstrictor receptors, a phenomenon that worsens the ischemic brain damage. Upregulation of contractile endothelin B (ET(B)) and 5-hydroxytryptamine 1B (5-HT(1B)) receptors has been demonstrated after subarachnoid hemorrhage and global ischemic stroke, but the situation is less clear after focal ischemic stroke. Changes in smooth muscle calcium handling have been implicated in different vascular diseases but have not hitherto been investigated in cerebral arteries after stroke. Here, we evaluate changes of ET(B) and 5-HT(1B) receptors, intracellular calcium levels, and calcium channel expression in rat middle cerebral artery (MCA) after focal cerebral ischemia and in vitro organ culture, a proposed model of vasoconstrictor receptor changes after stroke. Rats were subjected to 2 h MCA occlusion followed by reperfusion for 1 or 24 h. Alternatively, MCAs from naïve rats were cultured for 1 or 24 h. ET(B) and 5-HT(1B) receptor-mediated contractions were evaluated by wire myography. Receptor and channel expressions were measured by real-time PCR and immunohistochemistry. Intracellular calcium was measured by FURA-2. Expression and contractile functions of ET(B) and 5-HT(1B) receptors were strongly upregulated and slightly downregulated, respectively, 24 h after experimental stroke or organ culture. ET(B) receptor-mediated contraction was mediated by calcium from intracellular and extracellular sources, whereas 5-HT(1B) receptor-mediated contraction was solely dependent on extracellular calcium. Organ culture and stroke increased basal intracellular calcium levels in MCA smooth muscle cells and decreased the expression of inositol triphosphate receptor and transient receptor potential canonical calcium channels, but not voltage-operated calcium channels.
Collapse
MESH Headings
- Animals
- Calcium/metabolism
- Cerebral Arteries/drug effects
- Cerebral Arteries/metabolism
- Disease Models, Animal
- Dose-Response Relationship, Drug
- Intracellular Fluid/drug effects
- Intracellular Fluid/metabolism
- Male
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Organ Culture Techniques
- Rats
- Rats, Wistar
- Receptor, Endothelin B/biosynthesis
- Receptor, Serotonin, 5-HT1B/biosynthesis
- Stroke/metabolism
- Vasoconstriction/drug effects
- Vasoconstriction/physiology
- Viper Venoms/pharmacology
Collapse
Affiliation(s)
- Gro Klitgaard Povlsen
- Department of Clinical Experimental Research, Glostrup Research Institute, Ndr. Ringvej 69, 2600, Glostrup, Denmark.
| | | | | | | | | | | |
Collapse
|
40
|
Sadegh MK, Ekman M, Rippe C, Uvelius B, Swärd K, Albinsson S. Deletion of Dicer in smooth muscle affects voiding pattern and reduces detrusor contractility and neuroeffector transmission. PLoS One 2012; 7:e35882. [PMID: 22558254 PMCID: PMC3338793 DOI: 10.1371/journal.pone.0035882] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2012] [Accepted: 03/23/2012] [Indexed: 11/19/2022] Open
Abstract
MicroRNAs have emerged as important regulators of smooth muscle phenotype and may play important roles in pathogenesis of various smooth muscle related disease states. The aim of this study was to investigate the role of miRNAs for urinary bladder function. We used an inducible and smooth muscle specific Dicer knockout (KO) mouse which resulted in significantly reduced levels of miRNAs, including miR-145, miR-143, miR-22, miR125b-5p and miR-27a, from detrusor preparations without mucosa. Deletion of Dicer resulted in a disturbed micturition pattern in vivo and reduced depolarization-induced pressure development in the isolated detrusor. Furthermore, electrical field stimulation revealed a decreased cholinergic but maintained purinergic component of neurogenic activation in Dicer KO bladder strips. The ultrastructure of detrusor smooth muscle cells was well maintained, and the density of nerve terminals was similar. Western blotting demonstrated reduced contents of calponin and desmin. Smooth muscle α-actin, SM22α and myocardin were unchanged. Activation of strips with exogenous agonists showed that depolarization-induced contraction was preferentially reduced; ATP- and calyculin A-induced contractions were unchanged. Quantitative real time PCR and western blotting demonstrated reduced expression of Cav1.2 (Cacna1c). It is concluded that smooth muscle miRNAs play an important role for detrusor contractility and voiding pattern of unrestrained mice. This is mediated in part via effects on expression of smooth muscle differentiation markers and L-type Ca(2+) channels in the detrusor.
Collapse
Affiliation(s)
| | - Mari Ekman
- Department of Experimental Medical Science, Biomedical Centre, Lund University, Lund, Sweden
| | - Catarina Rippe
- Department of Experimental Medical Science, Biomedical Centre, Lund University, Lund, Sweden
| | - Bengt Uvelius
- Department of Urology, Clinical Sciences, Lund University, Lund, Sweden
| | - Karl Swärd
- Department of Experimental Medical Science, Biomedical Centre, Lund University, Lund, Sweden
| | - Sebastian Albinsson
- Department of Experimental Medical Science, Biomedical Centre, Lund University, Lund, Sweden
| |
Collapse
|
41
|
Sun Z, Li Z, Meininger GA. Mechanotransduction through fibronectin-integrin focal adhesion in microvascular smooth muscle cells: is calcium essential? Am J Physiol Heart Circ Physiol 2012; 302:H1965-73. [PMID: 22427509 DOI: 10.1152/ajpheart.00598.2011] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
It is believed that increased transmural pressure exerts force on vascular smooth muscle cells (VSMCs) and triggers Ca(2+) signaling as an initiating event responsible for the arteriolar myogenic response. However, the mechanisms linking the pressure increase to Ca(2+) signaling are unclear. We have shown previously using atomic force microscopy (AFM) that mechanical force induces a VSMC contractile response when applied to single fibronectin (FN; Sun Z, Martinez-Lemus LA, Hill MA, Meininger GA. Am J Physiol Cell Physiol 295; C268-C278, 2008) focal adhesion sites. This current study seeks to determine whether application of force to single focal adhesions can cause a change in VSMC Ca(2+). Experiments were performed in low passage (p3∼10) as well as in freshly isolated skeletal muscle arteriole VSMCs. AFM-attached microbeads (5 μm) were coated with FN or collagen type I (CN-I) or type IV (CN-IV) and placed on a VSMC for 20 min, resulting in formation of a focal adhesion between the cell and the microbead. In low passage VSMCs, mechanically pulling on the FN-coated beads (800∼3000 pN) did not induce a Ca(2+) increase but did cause a contractile response. In freshly isolated VSMCs, application of an FN or CN-I-coated bead onto the cell surface induced global Ca(2+) increases. However, these Ca(2+) increases were not correlated with the application of AFM pulling force to the bead or with the VSMC contractile responses to FN-coupled pulling. Chelating cytosolic Ca(2+) using BAPTA loading had no negative effect on the focal adhesion-related contractile response in both freshly isolated and low passage VSMCs, while the Rho-kinase inhibitor Y27632 abolished the micromyogenic response in both cases. These observations suggest that, in freshly isolated and cultured VSMCs, application of mechanical force to a focal adhesion does not invoke an acute global Ca(2+) increase. On the other hand, our data support a role for Rho-linked signaling mechanism involved in mechanotransduction leading to focal contraction that is independent of the need for a global increase in VSMC Ca(2+).
Collapse
Affiliation(s)
- Zhe Sun
- Dalton Cardiovascular Research Center and Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO 65211, USA.
| | | | | |
Collapse
|
42
|
Deniset JF, Hedley TE, Dibrov E, Pierce GN. Chlamydophila pneumoniae infection induces alterations in vascular contractile responses. THE AMERICAN JOURNAL OF PATHOLOGY 2012; 180:1264-1272. [PMID: 22214836 DOI: 10.1016/j.ajpath.2011.11.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2010] [Revised: 10/11/2011] [Accepted: 11/11/2011] [Indexed: 11/29/2022]
Abstract
Chlamydophila pneumoniae infection has been associated in previous studies with coronary artery disease. The live bacterium has been detected within atherosclerotic plaques and can induce the structural remodeling of the vessel wall. However, the direct effects of infection on the contractile characteristics of the arteries remain unknown. Left anterior descending coronary arteries isolated from porcine hearts were dissected and placed in culture medium for 72 hours before infection with C. pneumoniae. Contractile responses to high molar KCl and u46619 levels and relaxation responses to bradykinin and sodium nitroprusside were assessed at days 5 and 10 postinfection. C. pneumoniae induced decreases in both KCl- and u46619-induced contractile responses at both time points. The altered contractile responses coincided with a down-regulation of L-type Ca(2+) channels at both time points and inositol 1,4,5-triphosphate receptor (IP3R) levels at day 10 postinfection. Infection also induced attenuation of the endothelial-dependent relaxation response to bradykinin at day 10 postinfection. A decrease in endothelial nitric oxide synthase expression levels was noted at day 10 postinfection. Furthermore, an increase in superoxide production combined with an increase in p22phox expression levels was also observed at this time point. These findings indicate that C. pneumoniae infection can directly alter the vascular contractile responses in porcine coronary arteries, providing additional evidence for the role of C. pneumoniae infection in cardiovascular disease.
Collapse
Affiliation(s)
- Justin F Deniset
- Institute of Cardiovascular Sciences, St. Boniface Hospital Research Centre, Department of Physiology, Faculties of Medicine and Pharmacy, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Thomas E Hedley
- Institute of Cardiovascular Sciences, St. Boniface Hospital Research Centre, Department of Physiology, Faculties of Medicine and Pharmacy, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Elena Dibrov
- Institute of Cardiovascular Sciences, St. Boniface Hospital Research Centre, Department of Physiology, Faculties of Medicine and Pharmacy, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Grant N Pierce
- Institute of Cardiovascular Sciences, St. Boniface Hospital Research Centre, Department of Physiology, Faculties of Medicine and Pharmacy, University of Manitoba, Winnipeg, Manitoba, Canada.
| |
Collapse
|
43
|
Matchkov VV, Kudryavtseva O, Aalkjaer C. Intracellular Ca2+ Signalling and Phenotype of Vascular Smooth Muscle Cells. Basic Clin Pharmacol Toxicol 2011; 110:42-8. [DOI: 10.1111/j.1742-7843.2011.00818.x] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
44
|
Cecchettini A, Rocchiccioli S, Boccardi C, Citti L. Vascular smooth-muscle-cell activation: proteomics point of view. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2011; 288:43-99. [PMID: 21482410 DOI: 10.1016/b978-0-12-386041-5.00002-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Vascular smooth-muscle cells (VSMCs) are the main component of the artery medial layer. Thanks to their great plasticity, when stimulated by external inputs, VSMCs react by changing morphology and functions and activating new signaling pathways while switching others off. In this way, they are able to increase the cell proliferation, migration, and synthetic capacity significantly in response to vascular injury assuming a more dedifferentiated state. In different states of differentiation, VSMCs are characterized by various repertories of activated pathways and differentially expressed proteins. In this context, great interest is addressed to proteomics technology, in particular to differential proteomics. In recent years, many authors have investigated proteomics in order to identify the molecular factors putatively involved in VSMC phenotypic modulation, focusing on metabolic networks linking the differentially expressed proteins. Some of the identified proteins may be markers of pathology and become useful tools of diagnosis. These proteins could also represent appropriately validated targets and be useful either for prevention, if related to early events of atherosclerosis, or for treatment, if specific of the acute, mid, and late phases of the pathology. RNA-dependent gene silencing, obtained against the putative targets with high selective and specific molecular tools, might be able to reverse a pathological drift and be suitable candidates for innovative therapeutic approaches.
Collapse
|
45
|
SERCA2a controls the mode of agonist-induced intracellular Ca2+ signal, transcription factor NFAT and proliferation in human vascular smooth muscle cells. J Mol Cell Cardiol 2010; 50:621-33. [PMID: 21195084 DOI: 10.1016/j.yjmcc.2010.12.016] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2010] [Revised: 11/23/2010] [Accepted: 12/21/2010] [Indexed: 01/05/2023]
Abstract
In blood vessels, tone is maintained by agonist-induced cytosolic Ca(2+) oscillations of quiescent/contractile vascular smooth muscle cells (VSMCs). However, in synthetic/proliferative VSMCs, Gq/phosphoinositide receptor-coupled agonists trigger a steady-state increase in cytosolic Ca(2+) followed by a Store Operated Calcium Entry (SOCE) which translates into activation of the proliferation-associated transcription factor NFAT. Here, we report that in human coronary artery smooth muscle cells (hCASMCs), the sarco/endoplasmic reticulum calcium ATPase type 2a (SERCA2a) expressed in the contractile form of the hCASMCs, controls the nature of the agonist-induced Ca(2+) transient and the resulting down-stream signaling pathway. Indeed, restoring SERCA2a expression by gene transfer in synthetic hCASMCs 1) increased Ca(2+) storage capacity; 2) modified agonist-induced IP(3)R Ca(2+) release from steady-state to oscillatory mode (the frequency of agonist-induced IP(3)R Ca(2+) signal was 11.66 ± 1.40/100 s in SERCA2a-expressing cells (n=39) vs 1.37 ± 0.20/100 s in control cells (n=45), p<0.01); 3) suppressed SOCE by preventing interactions between SR calcium sensor STIM1 and pore forming unit ORAI1; 4) inhibited calcium regulated transcription factor NFAT and its down-stream physiological function such as proliferation and migration. This study provides evidence for the first time that oscillatory and steady-state patterns of Ca(2+) transients have different effects on calcium-dependent physiological functions in smooth muscle cells.
Collapse
|
46
|
|
47
|
Cheong A, Li J, Sukumar P, Kumar B, Zeng F, Riches K, Munsch C, Wood IC, Porter KE, Beech DJ. Potent suppression of vascular smooth muscle cell migration and human neointimal hyperplasia by KV1.3 channel blockers. Cardiovasc Res 2010; 89:282-9. [PMID: 20884640 PMCID: PMC3020133 DOI: 10.1093/cvr/cvq305] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
AIM The aim of the study was to determine the potential for K(V)1 potassium channel blockers as inhibitors of human neoinitimal hyperplasia. METHODS AND RESULTS Blood vessels were obtained from patients or mice and studied in culture. Reverse transcriptase-polymerase chain reaction and immunocytochemistry were used to detect gene expression. Whole-cell patch-clamp, intracellular calcium measurement, cell migration assays, and organ culture were used to assess channel function. K(V)1.3 was unique among the K(V)1 channels in showing preserved and up-regulated expression when the vascular smooth muscle cells switched to the proliferating phenotype. There was strong expression in neointimal formations. Voltage-dependent potassium current in proliferating cells was sensitive to three different blockers of K(V)1.3 channels. Calcium entry was also inhibited. All three blockers reduced vascular smooth muscle cell migration and the effects were non-additive. One of the blockers (margatoxin) was highly potent, suppressing cell migration with an IC(50) of 85 pM. Two of the blockers were tested in organ-cultured human vein samples and both inhibited neointimal hyperplasia. CONCLUSION K(V)1.3 potassium channels are functional in proliferating mouse and human vascular smooth muscle cells and have positive effects on cell migration. Blockers of the channels may be useful as inhibitors of neointimal hyperplasia and other unwanted vascular remodelling events.
Collapse
Affiliation(s)
- Alex Cheong
- Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds LS2 9JT, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Kaimoto T, Yasuda O, Ohishi M, Mogi M, Takemura Y, Suhara T, Ogihara T, Fukuo K, Rakugi H. Nifedipine inhibits vascular smooth muscle cell dedifferentiation via downregulation of Akt signaling. Hypertension 2010; 56:247-52. [PMID: 20530298 DOI: 10.1161/hypertensionaha.110.149781] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Calcium is an essential signaling molecule that controls vascular smooth muscle cell (VSMC) contraction, proliferation, and differentiation. Here, we show that the calcium antagonist nifedipine inhibits VSMC dedifferentiation in vitro and in vivo. Differentiated VSMCs cultured on laminin-coated dishes were transferred to laminin-free dishes to induce dedifferentiation. Induction of dedifferentiation resulted in the upregulation of nonmuscle myosin heavy chain expression, a marker of dedifferentiation, and the downregulation of smooth muscle myosin heavy chain expression, a marker of differentiation. Nifedipine significantly inhibited both the induction of these phenotypic changes and upregulation of Akt signaling in these cells. Administration of nifedipine at a low concentration that did not affect blood pressure could inhibit the increase in nonmuscle myosin heavy chain expression and decrease in smooth muscle myosin heavy chain expression in a rat balloon-injury model. Furthermore, nifedipine suppressed neointimal hyperplasia and upregulation of Akt signaling. However, phospho-Akt expression was not suppressed in the regenerating arterial endothelium of the nifedipine-treated rats. The inhibitory effect of the downregulation of Akt signaling by dominant-negative Akt on the induction of VSMC dedifferentiation in the intima was identical to that of nifedipine. In contrast, upregulation of Akt signaling by transfection of the cells with a constitutively active Akt reversed the nifedipine-induced inhibition of VSMC dedifferentiation. In conclusion, nifedipine inhibits VSMC dedifferentiation by suppressing Akt signaling, thereby preventing neointimal thickening.
Collapse
Affiliation(s)
- Taeko Kaimoto
- Department of Cardiovascular Clinical and Translational Research, Kumamoto University Hospital, Kumamoto 860-8556, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Cidad P, Moreno-Domínguez A, Novensá L, Roqué M, Barquín L, Heras M, Pérez-García MT, López-López JR. Characterization of ion channels involved in the proliferative response of femoral artery smooth muscle cells. Arterioscler Thromb Vasc Biol 2010; 30:1203-11. [PMID: 20299686 DOI: 10.1161/atvbaha.110.205187] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Vascular smooth muscle cells (VSMCs) contribute significantly to occlusive vascular diseases by virtue of their ability to switch to a noncontractile, migratory, and proliferating phenotype. Although the participation of ion channels in this phenotypic modulation (PM) has been described previously, changes in their expression are poorly defined because of their large molecular diversity. We obtained a global portrait of ion channel expression in contractile versus proliferating mouse femoral artery VSMCs, and explored the functional contribution to the PM of the most relevant changes that we observed. METHODS AND RESULTS High-throughput real-time polymerase chain reaction of 87 ion channel genes was performed in 2 experimental paradigms: an in vivo model of endoluminal lesion and an in vitro model of cultured VSMCs obtained from explants. mRNA expression changes showed a good correlation between the 2 proliferative models, with only 2 genes, Kv1.3 and Kvbeta2, increasing their expression on proliferation. The functional characterization demonstrates that Kv1.3 currents increased in proliferating VSMC and that their selective blockade inhibits migration and proliferation. CONCLUSIONS These findings establish the involvement of Kv1.3 channels in the PM of VSMCs, providing a new therapeutical target for the treatment of intimal hyperplasia.
Collapse
MESH Headings
- Animals
- Cell Movement
- Cell Proliferation/drug effects
- Cells, Cultured
- Cluster Analysis
- Disease Models, Animal
- Femoral Artery/metabolism
- Femoral Artery/pathology
- Gene Expression Profiling
- Genotype
- Hyperplasia
- Kv1.3 Potassium Channel/antagonists & inhibitors
- Kv1.3 Potassium Channel/genetics
- Kv1.3 Potassium Channel/metabolism
- Membrane Potentials
- Mice
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/injuries
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Phenotype
- Potassium Channel Blockers/pharmacology
- RNA, Messenger/metabolism
- Shaker Superfamily of Potassium Channels/genetics
- Shaker Superfamily of Potassium Channels/metabolism
- Up-Regulation
- Vasoconstriction
Collapse
Affiliation(s)
- Pilar Cidad
- Departamento de Bioquímica y Biología Molecular y Fisiología e Instituto de Biología y Genética Molecular, Universidad de Valladolid y Consejo superior de Investigaciones Cientificas, Valladolid, Spain
| | | | | | | | | | | | | | | |
Collapse
|
50
|
A Specific Subset of Mouse Bone Marrow Cells Has Smooth Muscle Cell Differentiation Capacity–Brief Report. Arterioscler Thromb Vasc Biol 2010; 30:533-5. [DOI: 10.1161/atvbaha.109.200097] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Objective—
To determine whether CX
3
CR1
+
bone marrow cells have the capacity for smooth muscle cell (SMC) differentiation.
Methods and Results—
CX
3
CR1
+
and CX
3
CR1
−
cells were isolated from marrow of CX
3
CR1 transgenic mice and cultured in SMC differentiation media. Phenotypic and functional analyses showed only CX
3
CR1
+
bone marrow cells exhibit colony cell outgrowth with SMC-specific protein expression, calcium signaling, and contraction responses similar to mature SMC.
Conclusion—
CX
3
CR1 marks a bone marrow cells population that enriches for progenitors with capacity to differentiate in vitro into SMC-like cells.
Collapse
|