1
|
Sugita S, Kawai R, Ujihara Y, Nakamura M. Stress fiber strain is zero in normal aortic smooth muscle, elevated in hypertensive stretch, and minimal in wall thickening rats. Sci Rep 2024; 14:29731. [PMID: 39613822 PMCID: PMC11606938 DOI: 10.1038/s41598-024-81229-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 11/25/2024] [Indexed: 12/01/2024] Open
Abstract
Hypertension causes aortic wall thickening until the original wall stress is restored. We hypothesized that this regulation involves stress fiber (SF) tension transmission to the nucleus in smooth muscle cells (SMCs) and investigated the strain in the SF direction as a condition required for this transmission. Thoracic aortas from Wistar Kyoto (WKY) and spontaneously hypertensive rats (SHRs) were examined. SFs in aortic SMCs were fluorescently labeled and observed under a confocal microscope while stretched along the circumferential (θ) axis. Three conditions were studied: WKY physiological (WKYphys; blood pressure changes from diastolic to systolic for WKY), high-strain state (WKYhigh; diastolic to hypertensive level for WKY simulating initial hypertension), and SHR physiological (SHRphys; diastolic to systolic for SHR simulating after wall-thickening). SF strain and direction were measured. The SF inclination angle from the θ axis was 18° ± 3° in WKYphys, 13° ± 2° in WKYhigh, and 20° ± 1° in SHRphys. SF strain was 0.01 ± 0.02 in WKYphys, 0.20 ± 0.04 in WKYhigh, and 0.02 ± 0.02 SHRphys. SF strain was minimal in WKYphys, significantly increased in WKYhigh, and reduced to approximately zero in SHRphys. These findings support SFs function as mechanosensors in response to hypertension.
Collapse
Affiliation(s)
- Shukei Sugita
- Department of Electrical and Mechanical Engineering, Nagoya Institute of Technology, Gokiso-Cho, Showa-Ku, Nagoya, 466-8555, Japan.
- Center of Biomedical Physics and Information Technology, Nagoya Institute of Technology, Gokiso-Cho, Showa-Ku, Nagoya, Japan.
| | - Rintaro Kawai
- Department of Electrical and Mechanical Engineering, Nagoya Institute of Technology, Gokiso-Cho, Showa-Ku, Nagoya, 466-8555, Japan
| | - Yoshihiro Ujihara
- Department of Electrical and Mechanical Engineering, Nagoya Institute of Technology, Gokiso-Cho, Showa-Ku, Nagoya, 466-8555, Japan
| | - Masanori Nakamura
- Department of Electrical and Mechanical Engineering, Nagoya Institute of Technology, Gokiso-Cho, Showa-Ku, Nagoya, 466-8555, Japan
- Center of Biomedical Physics and Information Technology, Nagoya Institute of Technology, Gokiso-Cho, Showa-Ku, Nagoya, Japan
- Department of Nanopharmaceutical Sciences, Nagoya Institute of Technology, Gokiso-Cho, Showa-Ku, Nagoya, Japan
| |
Collapse
|
2
|
Rasmussen M, Jin JP. Mechanoregulation and function of calponin and transgelin. BIOPHYSICS REVIEWS 2024; 5:011302. [PMID: 38515654 PMCID: PMC10954348 DOI: 10.1063/5.0176784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Accepted: 02/26/2024] [Indexed: 03/23/2024]
Abstract
It is well known that chemical energy can be converted to mechanical force in biological systems by motor proteins such as myosin ATPase. It is also broadly observed that constant/static mechanical signals potently induce cellular responses. However, the mechanisms that cells sense and convert the mechanical force into biochemical signals are not well understood. Calponin and transgelin are a family of homologous proteins that participate in the regulation of actin-activated myosin motor activity. An isoform of calponin, calponin 2, has been shown to regulate cytoskeleton-based cell motility functions under mechanical signaling. The expression of the calponin 2 gene and the turnover of calponin 2 protein are both under mechanoregulation. The regulation and function of calponin 2 has physiological and pathological significance, as shown in platelet adhesion, inflammatory arthritis, arterial atherosclerosis, calcific aortic valve disease, post-surgical fibrotic peritoneal adhesion, chronic proteinuria, ovarian insufficiency, and tumor metastasis. The levels of calponin 2 vary in different cell types, reflecting adaptations to specific tissue environments and functional states. The present review focuses on the mechanoregulation of calponin and transgelin family proteins to explore how cells sense steady tension and convert the force signal to biochemical activities. Our objective is to present a current knowledge basis for further investigations to establish the function and mechanisms of calponin and transgelin in cellular mechanoregulation.
Collapse
Affiliation(s)
- Monica Rasmussen
- Medical Scientist Training Program, University of Miami Miller School of Medicine, Miami, Florida 33101, USA
| | - J.-P. Jin
- Department of Physiology and Biophysics, University of Illinois at Chicago College of Medicine, Chicago, Illinois 60612, USA
| |
Collapse
|
3
|
Daoud F, Arévalo Martinez M, Holmberg J, Alajbegovic A, Ali N, Rippe C, Swärd K, Albinsson S. YAP and TAZ in Vascular Smooth Muscle Confer Protection Against Hypertensive Vasculopathy. Arterioscler Thromb Vasc Biol 2022; 42:428-443. [PMID: 35196875 PMCID: PMC8939708 DOI: 10.1161/atvbaha.121.317365] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Hypertension remains a major risk factor for cardiovascular diseases, but the underlying mechanisms are not well understood. We hypothesize that appropriate mechanotransduction and contractile function in vascular smooth muscle cells are crucial to maintain vascular wall integrity. The Hippo pathway effectors YAP (yes-associated protein 1) and TAZ (WW domain containing transcription regulator 1) have been identified as mechanosensitive transcriptional coactivators. However, their role in vascular smooth muscle cell mechanotransduction has not been investigated in vivo. METHODS We performed physiological and molecular analyses utilizing an inducible smooth muscle-specific YAP/TAZ knockout mouse model. RESULTS Arteries lacking YAP/TAZ have reduced agonist-mediated contraction, decreased myogenic response, and attenuated stretch-induced transcriptional regulation of smooth muscle markers. Moreover, in established hypertension, YAP/TAZ knockout results in severe vascular lesions in small mesenteric arteries characterized by neointimal hyperplasia, elastin degradation, and adventitial thickening. CONCLUSIONS This study demonstrates a protective role of YAP/TAZ against hypertensive vasculopathy.
Collapse
Affiliation(s)
- Fatima Daoud
- Department of Experimental Medical Science (F.D., M.A.M., J.H., A.A., C.R., K.S., S.A.), Lund University, Sweden
| | - Marycarmen Arévalo Martinez
- Department of Experimental Medical Science (F.D., M.A.M., J.H., A.A., C.R., K.S., S.A.), Lund University, Sweden
| | - Johan Holmberg
- Department of Experimental Medical Science (F.D., M.A.M., J.H., A.A., C.R., K.S., S.A.), Lund University, Sweden
| | - Azra Alajbegovic
- Department of Experimental Medical Science (F.D., M.A.M., J.H., A.A., C.R., K.S., S.A.), Lund University, Sweden
| | - Neserin Ali
- Department of Clinical Sciences Lund, Orthopaedics, Clinical Epidemiology Unit (N.A.), Lund University, Sweden
| | - Catarina Rippe
- Department of Experimental Medical Science (F.D., M.A.M., J.H., A.A., C.R., K.S., S.A.), Lund University, Sweden
| | - Karl Swärd
- Department of Experimental Medical Science (F.D., M.A.M., J.H., A.A., C.R., K.S., S.A.), Lund University, Sweden
| | - Sebastian Albinsson
- Department of Experimental Medical Science (F.D., M.A.M., J.H., A.A., C.R., K.S., S.A.), Lund University, Sweden
| |
Collapse
|
4
|
Petzold J, Gentleman E. Intrinsic Mechanical Cues and Their Impact on Stem Cells and Embryogenesis. Front Cell Dev Biol 2021; 9:761871. [PMID: 34820380 PMCID: PMC8606660 DOI: 10.3389/fcell.2021.761871] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 10/14/2021] [Indexed: 12/25/2022] Open
Abstract
Although understanding how soluble cues direct cellular processes revolutionised the study of cell biology in the second half of the 20th century, over the last two decades, new insights into how mechanical cues similarly impact cell fate decisions has gained momentum. During development, extrinsic cues such as fluid flow, shear stress and compressive forces are essential for normal embryogenesis to proceed. Indeed, both adult and embryonic stem cells can respond to applied forces, but they can also detect intrinsic mechanical cues from their surrounding environment, such as the stiffness of the extracellular matrix, which impacts differentiation and morphogenesis. Cells can detect changes in their mechanical environment using cell surface receptors such as integrins and focal adhesions. Moreover, dynamic rearrangements of the cytoskeleton have been identified as a key means by which forces are transmitted from the extracellular matrix to the cell and vice versa. Although we have some understanding of the downstream mechanisms whereby mechanical cues are translated into changes in cell behaviour, many of the signalling pathways remain to be defined. This review discusses the importance of intrinsic mechanical cues on adult cell fate decisions, the emerging roles of cell surface mechano-sensors and the cytoskeleton in enabling cells to sense its microenvironment, and the role of intracellular signalling in translating mechanical cues into transcriptional outputs. In addition, the contribution of mechanical cues to fundamental processes during embryogenesis such as apical constriction and convergent extension is discussed. The continued development of tools to measure the biomechanical properties of soft tissues in vivo is likely to uncover currently underestimated contributions of these cues to adult stem cell fate decisions and embryogenesis, and may inform on regenerative strategies for tissue repair.
Collapse
Affiliation(s)
- Jonna Petzold
- Centre for Craniofacial and Regenerative Biology, King's College London, London, United Kingdom
| | - Eileen Gentleman
- Centre for Craniofacial and Regenerative Biology, King's College London, London, United Kingdom
| |
Collapse
|
5
|
Hyperglycemia-induced transcriptional regulation of ROCK1 and TGM2 expression is involved in small artery remodeling in obese diabetic Göttingen Minipigs. Clin Sci (Lond) 2020; 133:2499-2516. [PMID: 31830262 DOI: 10.1042/cs20191066] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 12/11/2019] [Accepted: 12/12/2019] [Indexed: 12/11/2022]
Abstract
Obesity and diabetes in humans are associated with hypertrophic remodeling and increased media:lumen ratio of small resistance arteries, which is an independent predictor of cardiovascular events. In order to minimize increases in media:lumen ratio, hypertrophic remodeling should be accompanied by outward remodeling. We aimed to investigate the mechanisms of structural remodeling in small pial arteries (PAs) and terminal mesenteric arteries (TMAs) from obese Göttingen Minipigs with or without diabetes. Göttingen Minipigs received either control diet (lean control (LC)), high fat/high fructose/high cholesterol diet (FFC), or FFC diet with streptozotocin (STZ)-induced diabetes (FFC/STZ) for 13 months. At the end of the study (20 months), we assessed body weight, fasting plasma biochemistry, passive vessel dimensions, mRNA expression (matrix metallopeptidases 2/9 (MMP2, MMP9), tissue inhibitor of metallopeptidase 1 (TIMP1), transglutaminase 2 (TGM2), Rho-kinase 1 (ROCK1), TGFβ-receptor 2 (TGFBR2), and IGF1-receptor (IGFR1) genes), and immunofluorescence in PAs and TMAs. We performed multiple linear correlation analyses using plasma values, structural data, and gene expression data. We detected outward hypertrophic remodeling in TMAs and hypertrophic remodeling in PAs from FFC/STZ animals. ROCK1 and TGM2 genes were up-regulated in PAs and TMAs from the FFC/STZ group. Passive lumen diameter (PLD) of TMAs was correlated with plasma values of glucose (GLU), fructosamine (FRA), total cholesterol (TC), and triglycerides (TGs). ROCK1 and TGM2 expressions in TMAs were correlated with PLD, plasma GLU, fructosamine, and TC. ROCK1 and TGM2 proteins were immunolocalized in the media of PAs and TMAs, and their fluorescence levels were increased in the FFC/STZ group. Hyperglycemia/hyperlipidemia is involved in regulation of ROCK1 and TGM2 expression leading to outward remodeling of small resistance arteries in obese diabetic Göttingen Minipigs.
Collapse
|
6
|
Knock GA. NADPH oxidase in the vasculature: Expression, regulation and signalling pathways; role in normal cardiovascular physiology and its dysregulation in hypertension. Free Radic Biol Med 2019; 145:385-427. [PMID: 31585207 DOI: 10.1016/j.freeradbiomed.2019.09.029] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 08/29/2019] [Accepted: 09/23/2019] [Indexed: 02/06/2023]
Abstract
The last 20-25 years have seen an explosion of interest in the role of NADPH oxidase (NOX) in cardiovascular function and disease. In vascular smooth muscle and endothelium, NOX generates reactive oxygen species (ROS) that act as second messengers, contributing to the control of normal vascular function. NOX activity is altered in response to a variety of stimuli, including G-protein coupled receptor agonists, growth-factors, perfusion pressure, flow and hypoxia. NOX-derived ROS are involved in smooth muscle constriction, endothelium-dependent relaxation and smooth muscle growth, proliferation and migration, thus contributing to the fine-tuning of blood flow, arterial wall thickness and vascular resistance. Through reversible oxidative modification of target proteins, ROS regulate the activity of protein tyrosine phosphatases, kinases, G proteins, ion channels, cytoskeletal proteins and transcription factors. There is now considerable, but somewhat contradictory evidence that NOX contributes to the pathogenesis of hypertension through oxidative stress. Specific NOX isoforms have been implicated in endothelial dysfunction, hyper-contractility and vascular remodelling in various animal models of hypertension, pulmonary hypertension and pulmonary arterial hypertension, but also have potential protective effects, particularly NOX4. This review explores the multiplicity of NOX function in the healthy vasculature and the evidence for and against targeting NOX for antihypertensive therapy.
Collapse
Affiliation(s)
- Greg A Knock
- Dpt. of Inflammation Biology, School of Immunology & Microbial Sciences, Faculty of Life Sciences & Medicine, King's College London, UK.
| |
Collapse
|
7
|
Abstract
The human cerebral vasculature originates in the fourth week of gestation and continues to expand and diversify well into the first few years of postnatal life. A key feature of this growth is smooth muscle differentiation, whereby smooth muscle cells within cerebral arteries transform from migratory to proliferative to synthetic and finally to contractile phenotypes. These phenotypic transformations can be reversed by pathophysiological perturbations such as hypoxia, which causes loss of contractile capacity in immature cerebral arteries. In turn, loss of contractility affects all whole-brain cerebrovascular responses, including those involved in flow-metabolism coupling, vasodilatory responses to acute hypoxia and hypercapnia, cerebral autoregulation, and reactivity to activation of perivascular nerves. Future strategies to minimize cerebral injury following hypoxia-ischemic insults in the immature brain might benefit by targeting treatments to preserve and promote contractile differentiation in the fetal cerebrovasculature. This could potentially be achieved through inhibition of receptor tyrosine kinase-mediated growth factors, such as vascular endothelial growth factor and platelet-derived growth factor, which are mobilized by hypoxic and ischemic injury and which facilitate contractile dedifferentiation. Interruption of the effects of other vascular mitogens, such as endothelin and angiotensin-II, and even some miRNA species, also could be beneficial. Future experimental work that addresses these possibilities offers promise to improve current clinical management of neonates who have suffered and survived hypoxic, ischemic, asphyxic, or inflammatory cerebrovascular insults.
Collapse
Affiliation(s)
- William J Pearce
- From the Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, CA.
| |
Collapse
|
8
|
Ducsay CA, Goyal R, Pearce WJ, Wilson S, Hu XQ, Zhang L. Gestational Hypoxia and Developmental Plasticity. Physiol Rev 2018; 98:1241-1334. [PMID: 29717932 PMCID: PMC6088145 DOI: 10.1152/physrev.00043.2017] [Citation(s) in RCA: 123] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Hypoxia is one of the most common and severe challenges to the maintenance of homeostasis. Oxygen sensing is a property of all tissues, and the response to hypoxia is multidimensional involving complicated intracellular networks concerned with the transduction of hypoxia-induced responses. Of all the stresses to which the fetus and newborn infant are subjected, perhaps the most important and clinically relevant is that of hypoxia. Hypoxia during gestation impacts both the mother and fetal development through interactions with an individual's genetic traits acquired over multiple generations by natural selection and changes in gene expression patterns by altering the epigenetic code. Changes in the epigenome determine "genomic plasticity," i.e., the ability of genes to be differentially expressed according to environmental cues. The genomic plasticity defined by epigenomic mechanisms including DNA methylation, histone modifications, and noncoding RNAs during development is the mechanistic substrate for phenotypic programming that determines physiological response and risk for healthy or deleterious outcomes. This review explores the impact of gestational hypoxia on maternal health and fetal development, and epigenetic mechanisms of developmental plasticity with emphasis on the uteroplacental circulation, heart development, cerebral circulation, pulmonary development, and the hypothalamic-pituitary-adrenal axis and adipose tissue. The complex molecular and epigenetic interactions that may impact an individual's physiology and developmental programming of health and disease later in life are discussed.
Collapse
Affiliation(s)
- Charles A. Ducsay
- The Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California
| | - Ravi Goyal
- The Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California
| | - William J. Pearce
- The Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California
| | - Sean Wilson
- The Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California
| | - Xiang-Qun Hu
- The Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California
| | - Lubo Zhang
- The Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California
| |
Collapse
|
9
|
Mochalov SV, Tarasova NV, Kudryashova TV, Gaynullina DK, Kalenchuk VU, Borovik AS, Vorotnikov AV, Tarasova OS, Schubert R. Higher Ca 2+ -sensitivity of arterial contraction in 1-week-old rats is due to a greater Rho-kinase activity. Acta Physiol (Oxf) 2018; 223:e13044. [PMID: 29383848 DOI: 10.1111/apha.13044] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 01/23/2018] [Accepted: 01/24/2018] [Indexed: 12/22/2022]
Abstract
AIM During early post-natal development, arterial contraction depends less on Ca2+ -signalling pathways but more on changes in Ca2+ -sensitivity compared to adult animals. Whether this difference is related to Rho-kinase, one of the major players affecting Ca2+ -sensitivity, is unknown for intact vessels. Thus, we tested the hypothesis that Rho-kinase critically contributes to the higher Ca2+ -sensitivity of contraction in intact arteries of 1-week-old rats. METHODS We studied 1-week-old, 4- to 5-week-old and 10- to 12-week-old rats performing isometric myography, Ca2+ -fluorimetry and Western blotting using intact saphenous arteries and arterial pressure measurements under urethane anaesthesia. RESULTS In 10- to 12-week-old rats, methoxamine (MX) produced vasoconstriction associated with an increase in [Ca2+ ]i and Ca2+ -sensitivity. In contrast, in 1-week-old rats these contractions were accompanied only by an increase in Ca2+ -sensitivity. All MX-induced effects were reduced by the Rho-kinase inhibitor Y-27632; this reduction was complete only in 1-week-old rats. The Rho-kinase specific site Thr855 on MYPT1 was increasingly phosphorylated by MX in vessels of 1-week-old, but not 10- to 12-week-old rats; this effect was also inhibited completely by Y-27632. The Rho-kinase inhibitor fasudil in a dose not affecting the pressor response to MX in 4- to 5-week-old rats reduced it considerably in 1-week-old rats. CONCLUSION Our results suggest that the higher Ca2+ -sensitivity of arterial contraction in 1-week-old compared to 10- to 12-week-old rats is due to a greater Rho-kinase activity. Constitutively active Rho-kinase contributes to MX-induced contraction in 10- to 12-week-old rats. In 1-week-old rats, additional Rho-kinase activation is involved. This remodelling of the Rho-kinase pathway is associated with its increased contribution to adrenergic arterial pressure responses.
Collapse
Affiliation(s)
- S. V. Mochalov
- Medical Faculty Mannheim; Centre for Biomedicine and Medical Technology Mannheim (CBTM) and European Center of Angioscience (ECAS); Research Division Cardiovascular Physiology; Heidelberg University; Mannheim Germany
- Faculty of Biology, M.V. Lomonosov; Moscow State University; Moscow Russia
- ChemRar Research and Development Institute; Khimki Moscow Region Russia
| | - N. V. Tarasova
- Medical Faculty Mannheim; Centre for Biomedicine and Medical Technology Mannheim (CBTM) and European Center of Angioscience (ECAS); Research Division Cardiovascular Physiology; Heidelberg University; Mannheim Germany
- Faculty of Biology, M.V. Lomonosov; Moscow State University; Moscow Russia
- Molecular Medicine Institute; I.M. Sechenov First Moscow State Medical University; Moscow Russia
| | - T. V. Kudryashova
- Institute of Experimental Cardiology; Cardiology Research Center; Moscow Russia
| | - D. K. Gaynullina
- Medical Faculty Mannheim; Centre for Biomedicine and Medical Technology Mannheim (CBTM) and European Center of Angioscience (ECAS); Research Division Cardiovascular Physiology; Heidelberg University; Mannheim Germany
- Faculty of Biology, M.V. Lomonosov; Moscow State University; Moscow Russia
- Department of Physiology; Russian National Research Medical University; Moscow Russia
| | - V. U. Kalenchuk
- Faculty of Basic Medicine; M.V. Lomonosov Moscow State University; Moscow Russia
| | - A. S. Borovik
- State Research Center of the Russian Federation - Institute for Biomedical Problems; Russian Academy of Sciences; Moscow Russia
| | - A. V. Vorotnikov
- Institute of Experimental Cardiology; Cardiology Research Center; Moscow Russia
- Medical Center; M.V. Lomonosov Moscow State University; Moscow Russia
| | - O. S. Tarasova
- Faculty of Biology, M.V. Lomonosov; Moscow State University; Moscow Russia
- State Research Center of the Russian Federation - Institute for Biomedical Problems; Russian Academy of Sciences; Moscow Russia
| | - R. Schubert
- Medical Faculty Mannheim; Centre for Biomedicine and Medical Technology Mannheim (CBTM) and European Center of Angioscience (ECAS); Research Division Cardiovascular Physiology; Heidelberg University; Mannheim Germany
| |
Collapse
|
10
|
Hien TT, Garcia‐Vaz E, Stenkula KG, Sjögren J, Nilsson J, Gomez MF, Albinsson S. MicroRNA‐dependent regulation of KLF4 by glucose in vascular smooth muscle. J Cell Physiol 2018; 233:7195-7205. [DOI: 10.1002/jcp.26549] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Accepted: 02/12/2018] [Indexed: 12/20/2022]
Affiliation(s)
- Tran T. Hien
- Department of Experimental Medical ScienceLund UniversityLundSweden
| | - Eliana Garcia‐Vaz
- Department of Clinical Sciences in Malmö, Lund University Diabetes CentreLund UniversitySweden
| | | | - Johan Sjögren
- Department of Cardiothoracic SurgerySkåne University Hospital and Lund UniversityLundSweden
| | - Johan Nilsson
- Department of Cardiothoracic SurgerySkåne University Hospital and Lund UniversityLundSweden
| | - Maria F. Gomez
- Department of Clinical Sciences in Malmö, Lund University Diabetes CentreLund UniversitySweden
| | | |
Collapse
|
11
|
Huang CH, Ciou JS, Chen ST, Kok VC, Chung Y, Tsai JJP, Kurubanjerdjit N, Huang CYF, Ng KL. Identify potential drugs for cardiovascular diseases caused by stress-induced genes in vascular smooth muscle cells. PeerJ 2016; 4:e2478. [PMID: 27703845 PMCID: PMC5045879 DOI: 10.7717/peerj.2478] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 08/23/2016] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Abnormal proliferation of vascular smooth muscle cells (VSMC) is a major cause of cardiovascular diseases (CVDs). Many studies suggest that vascular injury triggers VSMC dedifferentiation, which results in VSMC changes from a contractile to a synthetic phenotype; however, the underlying molecular mechanisms are still unclear. METHODS In this study, we examined how VSMC responds under mechanical stress by using time-course microarray data. A three-phase study was proposed to investigate the stress-induced differentially expressed genes (DEGs) in VSMC. First, DEGs were identified by using the moderated t-statistics test. Second, more DEGs were inferred by using the Gaussian Graphical Model (GGM). Finally, the topological parameters-based method and cluster analysis approach were employed to predict the last batch of DEGs. To identify the potential drugs for vascular diseases involve VSMC proliferation, the drug-gene interaction database, Connectivity Map (cMap) was employed. Success of the predictions were determined using in-vitro data, i.e. MTT and clonogenic assay. RESULTS Based on the differential expression calculation, at least 23 DEGs were found, and the findings were qualified by previous studies on VSMC. The results of gene set enrichment analysis indicated that the most often found enriched biological processes are cell-cycle-related processes. Furthermore, more stress-induced genes, well supported by literature, were found by applying graph theory to the gene association network (GAN). Finally, we showed that by processing the cMap input queries with a cluster algorithm, we achieved a substantial increase in the number of potential drugs with experimental IC50 measurements. With this novel approach, we have not only successfully identified the DEGs, but also improved the DEGs prediction by performing the topological and cluster analysis. Moreover, the findings are remarkably validated and in line with the literature. Furthermore, the cMap and DrugBank resources were used to identify potential drugs and targeted genes for vascular diseases involve VSMC proliferation. Our findings are supported by in-vitro experimental IC50, binding activity data and clinical trials. CONCLUSION This study provides a systematic strategy to discover potential drugs and target genes, by which we hope to shed light on the treatments of VSMC proliferation associated diseases.
Collapse
Affiliation(s)
- Chien-Hung Huang
- Department of Computer Science and Information Engineering, National Formosa University, Yun-Lin, Taiwan
| | - Jin-Shuei Ciou
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung, Taiwan
| | - Shun-Tsung Chen
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung, Taiwan
| | - Victor C. Kok
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung, Taiwan
- Division of Medical Oncology, Kuang Tien General Hospital Cancer Center, Taichung, Taiwan
| | - Yi Chung
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung, Taiwan
| | - Jeffrey J. P. Tsai
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung, Taiwan
| | | | - Chi-Ying F. Huang
- Institute of Biopharmaceutical Sciences, National Yang-Ming University, Taipei, Taiwan
| | - Ka-Lok Ng
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
| |
Collapse
|
12
|
Hien TT, Turczyńska KM, Dahan D, Ekman M, Grossi M, Sjögren J, Nilsson J, Braun T, Boettger T, Garcia-Vaz E, Stenkula K, Swärd K, Gomez MF, Albinsson S. Elevated Glucose Levels Promote Contractile and Cytoskeletal Gene Expression in Vascular Smooth Muscle via Rho/Protein Kinase C and Actin Polymerization. J Biol Chem 2016; 291:3552-68. [PMID: 26683376 PMCID: PMC4751395 DOI: 10.1074/jbc.m115.654384] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Revised: 12/17/2015] [Indexed: 12/22/2022] Open
Abstract
Both type 1 and type 2 diabetes are associated with increased risk of cardiovascular disease. This is in part attributed to the effects of hyperglycemia on vascular endothelial and smooth muscle cells, but the underlying mechanisms are not fully understood. In diabetic animal models, hyperglycemia results in hypercontractility of vascular smooth muscle possibly due to increased activation of Rho-kinase. The aim of the present study was to investigate the regulation of contractile smooth muscle markers by glucose and to determine the signaling pathways that are activated by hyperglycemia in smooth muscle cells. Microarray, quantitative PCR, and Western blot analyses revealed that both mRNA and protein expression of contractile smooth muscle markers were increased in isolated smooth muscle cells cultured under high compared with low glucose conditions. This effect was also observed in hyperglycemic Akita mice and in diabetic patients. Elevated glucose activated the protein kinase C and Rho/Rho-kinase signaling pathways and stimulated actin polymerization. Glucose-induced expression of contractile smooth muscle markers in cultured cells could be partially or completely repressed by inhibitors of advanced glycation end products, L-type calcium channels, protein kinase C, Rho-kinase, actin polymerization, and myocardin-related transcription factors. Furthermore, genetic ablation of the miR-143/145 cluster prevented the effects of glucose on smooth muscle marker expression. In conclusion, these data demonstrate a possible link between hyperglycemia and vascular disease states associated with smooth muscle contractility.
Collapse
MESH Headings
- Actin Cytoskeleton/metabolism
- Actin Cytoskeleton/pathology
- Aged
- Animals
- Atherosclerosis/enzymology
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- Cells, Cultured
- Contractile Proteins/agonists
- Contractile Proteins/genetics
- Contractile Proteins/metabolism
- Cytoskeletal Proteins/agonists
- Cytoskeletal Proteins/genetics
- Cytoskeletal Proteins/metabolism
- Diabetes Mellitus, Type 1/complications
- Diabetes Mellitus, Type 2/complications
- Diabetic Angiopathies/enzymology
- Diabetic Angiopathies/metabolism
- Diabetic Angiopathies/pathology
- Gene Expression Regulation
- Humans
- Male
- Mice, Knockout
- Mice, Mutant Strains
- MicroRNAs/metabolism
- Muscle, Smooth, Vascular/enzymology
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Protein Kinase C/chemistry
- Protein Kinase C/metabolism
- Signal Transduction
- rho GTP-Binding Proteins/agonists
- rho GTP-Binding Proteins/metabolism
- rho-Associated Kinases/chemistry
- rho-Associated Kinases/metabolism
Collapse
Affiliation(s)
- Tran Thi Hien
- From the Departments of Experimental Medical Sciences and
| | | | - Diana Dahan
- From the Departments of Experimental Medical Sciences and
| | - Mari Ekman
- From the Departments of Experimental Medical Sciences and
| | - Mario Grossi
- From the Departments of Experimental Medical Sciences and
| | - Johan Sjögren
- Clinical Sciences, Lund University, BMC D12, SE-221 84 Lund, Sweden and
| | - Johan Nilsson
- Clinical Sciences, Lund University, BMC D12, SE-221 84 Lund, Sweden and
| | - Thomas Braun
- the Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany, and
| | - Thomas Boettger
- the Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany, and
| | - Eliana Garcia-Vaz
- the Department of Clinical Sciences in Malmö, Lund University, 205 02 Malmö, Sweden
| | - Karin Stenkula
- From the Departments of Experimental Medical Sciences and
| | - Karl Swärd
- From the Departments of Experimental Medical Sciences and
| | - Maria F Gomez
- the Department of Clinical Sciences in Malmö, Lund University, 205 02 Malmö, Sweden
| | | |
Collapse
|
13
|
Postolow F, Fediuk J, Nolette N, Hinton M, Dakshinamurti S. Thromboxane promotes smooth muscle phenotype commitment but not remodeling of hypoxic neonatal pulmonary artery. FIBROGENESIS & TISSUE REPAIR 2015; 8:20. [PMID: 26583045 PMCID: PMC4650498 DOI: 10.1186/s13069-015-0037-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Accepted: 10/20/2015] [Indexed: 12/19/2022]
Abstract
Background Persistent pulmonary hypertension of the newborn (PPHN) is characterized by vasoconstriction and pulmonary vascular remodeling. Remodeling is believed to be a response to physical or chemical stimuli including pro-mitotic inflammatory mediators such as thromboxane. Our objective was to examine the effects of hypoxia and thromboxane signaling ex vivo and in vitro on phenotype commitment, cell cycle entry, and proliferation of PPHN and control neonatal pulmonary artery (PA) myocytes in tissue culture. Methods To examine concurrent effects of hypoxia and thromboxane on myocyte growth, serum-fed first-passage newborn porcine PA myocytes were randomized into normoxic (21 % O2) or hypoxic (10 % O2) culture for 3 days, with daily addition of thromboxane mimetic U46619 (10−9 to 10−5 M) or diluent. Cell survival was detected by MTT assay. To determine the effect of chronic thromboxane exposure (versus whole serum) on activation of arterial remodeling, PPHN was induced in newborn piglets by a 3-day hypoxic exposure (FiO2 0.10); controls were 3 day-old normoxic and day 0 piglets. Third-generation PA were segmented and cultured for 3 days in physiologic buffer, Ham’s F-12 media (in the presence or absence of 10 % fetal calf serum), or media with 10−6 M U46619. DNA synthesis was measured by 3H-thymidine uptake, protein synthesis by 3H-leucine uptake, and proliferation by immunostaining for Ki67. Cell cycle entry was studied by laser scanning cytometry of nuclei in arterial tunica media after propidium iodide staining. Phenotype commitment was determined by immunostaining tunica media for myosin heavy chain and desmin, quantified by laser scanning cytometry. Results Contractile and synthetic myocyte subpopulations had differing responses to thromboxane challenge. U46619 decreased proliferation of synthetic and contractile myocytes. PPHN arteries exhibited decreased protein synthesis under all culture conditions. Serum-supplemented PA treated with U46619 had decreased G1/G0 phase myocytes and an increase in S and G2/M. When serum-deprived, PPHN PA incubated with U46619 showed arrested cell cycle entry (increased G0/G1, decreased S and G2/M) and increased abundance of contractile phenotype markers. Conclusions We conclude that thromboxane does not initiate phenotypic dedifferentiation and proliferative activation in PPHN PA. Exposure to thromboxane triggers cell cycle exit and myocyte commitment to contractile phenotype.
Collapse
Affiliation(s)
- Fabiana Postolow
- Department of Pediatrics, University of Manitoba, 715 McDermot Avenue, Winnipeg, MB R3E 3P4 Canada
| | - Jena Fediuk
- Department of Physiology, University of Manitoba, 715 McDermot Avenue, Winnipeg, MB R3E 3P4 Canada ; Biology of Breathing Group, Manitoba Institute of Child Health, 715 McDermot Avenue, Winnipeg, MB R3E 3P4 Canada
| | - Nora Nolette
- Biology of Breathing Group, Manitoba Institute of Child Health, 715 McDermot Avenue, Winnipeg, MB R3E 3P4 Canada
| | - Martha Hinton
- Biology of Breathing Group, Manitoba Institute of Child Health, 715 McDermot Avenue, Winnipeg, MB R3E 3P4 Canada
| | - Shyamala Dakshinamurti
- Department of Pediatrics, University of Manitoba, 715 McDermot Avenue, Winnipeg, MB R3E 3P4 Canada ; Department of Physiology, University of Manitoba, 715 McDermot Avenue, Winnipeg, MB R3E 3P4 Canada ; Biology of Breathing Group, Manitoba Institute of Child Health, 715 McDermot Avenue, Winnipeg, MB R3E 3P4 Canada ; Section of Neonatology, WS012 Women's Hospital, 735 Notre Dame Ave, Winnipeg, MB R3E 0L8 Canada
| |
Collapse
|
14
|
Blirando K, Blaise R, Gorodnaya N, Rouxel C, Meilhac O, Vincent P, Limon I. The stellate vascular smooth muscle cell phenotype is induced by IL-1β via the secretion of PGE2 and subsequent cAMP-dependent protein kinase A activation. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1853:3235-47. [PMID: 26403276 DOI: 10.1016/j.bbamcr.2015.09.019] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Revised: 09/11/2015] [Accepted: 09/14/2015] [Indexed: 12/12/2022]
Abstract
Atherosclerosis development is associated with morphological changes to intimal cells, leading to a stellate cell phenotype. In this study, we aimed to determine whether and how key pro-atherogenic cytokines present in atherosclerotic plaques (IL-1β, TNFα and IFNγ) could induce this phenotype, as these molecules are known to trigger the transdifferentiation of vascular smooth muscle cells (VSMCs). We found that, IL-1β was the only major inflammatory mediator tested capable of inducing a stellate morphology in VSMCs. This finding was confirmed by staining for F-actin and vinculin at focal adhesions, as these two markers were disrupted only by IL-1β. We then investigated the possible association of this IL-1β-dependent change in morphology with an increase in intracellular cAMP concentration ([cAMP]), using the FRET-based biosensor for cAMP (T)Epac(VV). Experiments in the presence of IL-1β or medium conditioned by IL-1β-treated VSMCs and pharmacological tools demonstrated that the long-term increase in intracellular cAMP concentration was induced by the secretion of an autocrine/paracrine mediator, prostaglandin E₂(PGE₂), acting through the EP4 receptor. Finally, by knocking down the expression of the regulatory subunit PKAR1α, thereby reproducing the effects of IL-1β and PGE₂ on VSMCs, we demonstrated the contribution of PKA activity to the observed behavior of VSMCs.
Collapse
Affiliation(s)
- Karl Blirando
- Sorbonne Universités, UPMC Univ Paris 06CNRS UMR 8256 B2A, IBPS, F-75005, Paris, France
| | - Régis Blaise
- Sorbonne Universités, UPMC Univ Paris 06CNRS UMR 8256 B2A, IBPS, F-75005, Paris, France
| | - Natalia Gorodnaya
- Sorbonne Universités, UPMC Univ Paris 06CNRS UMR 8256 B2A, IBPS, F-75005, Paris, France
| | - Clotilde Rouxel
- Sorbonne Universités, UPMC Univ Paris 06CNRS UMR 8256 B2A, IBPS, F-75005, Paris, France
| | - Olivier Meilhac
- Inserm U1188 Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI) CYROI, 2, rue Maxime Rivière, 97490 Sainte Clotilde, La Réunion, France
| | - Pierre Vincent
- Sorbonne Universités, UPMC Univ Paris 06CNRS UMR 8256 B2A, IBPS, F-75005, Paris, France
| | - Isabelle Limon
- Sorbonne Universités, UPMC Univ Paris 06CNRS UMR 8256 B2A, IBPS, F-75005, Paris, France.
| |
Collapse
|
15
|
Turczyńska KM, Swärd K, Hien TT, Wohlfahrt J, Mattisson IY, Ekman M, Nilsson J, Sjögren J, Murugesan V, Hultgårdh-Nilsson A, Cidad P, Hellstrand P, Pérez-García MT, Albinsson S. Regulation of Smooth Muscle Dystrophin and Synaptopodin 2 Expression by Actin Polymerization and Vascular Injury. Arterioscler Thromb Vasc Biol 2015; 35:1489-97. [DOI: 10.1161/atvbaha.114.305065] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Accepted: 03/27/2015] [Indexed: 01/25/2023]
Abstract
Objective—
Actin dynamics in vascular smooth muscle is known to regulate contractile differentiation and may play a role in the pathogenesis of vascular disease. However, the list of genes regulated by actin polymerization in smooth muscle remains incomprehensive. Thus, the objective of this study was to identify actin-regulated genes in smooth muscle and to demonstrate the role of these genes in the regulation of vascular smooth muscle phenotype.
Approach and Results—
Mouse aortic smooth muscle cells were treated with an actin-stabilizing agent, jasplakinolide, and analyzed by microarrays. Several transcripts were upregulated including both known and previously unknown actin-regulated genes. Dystrophin and synaptopodin 2 were selected for further analysis in models of phenotypic modulation and vascular disease. These genes were highly expressed in differentiated versus synthetic smooth muscle and their expression was promoted by the transcription factors myocardin and myocardin-related transcription factor A. Furthermore, the expression of both synaptopodin 2 and dystrophin was significantly reduced in balloon-injured human arteries. Finally, using a dystrophin mutant
mdx
mouse and synaptopodin 2 knockdown, we demonstrate that these genes are involved in the regulation of smooth muscle differentiation and function.
Conclusions—
This study demonstrates novel genes that are promoted by actin polymerization, that regulate smooth muscle function, and that are deregulated in models of vascular disease. Thus, targeting actin polymerization or the genes controlled in this manner can lead to novel therapeutic options against vascular pathologies that involve phenotypic modulation of smooth muscle cells.
Collapse
Affiliation(s)
- Karolina M. Turczyńska
- From the Department of Experimental Medical Science (K.M.T., K.S., T.T.H., J.W., I.Y.M., M.E., V.M., A.H.-N., P.H., S.A.) and Department of Clinical Science (J.N., J.S.), Lund University, Lund, Sweden; and Departamento de Bioquímica y Biología Molecular y Fisiología and Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid and Consejo Superior de Investigaciones Científicas (CSIC), Valladolid, Spain (P.C., M.T.P.-G.)
| | - Karl Swärd
- From the Department of Experimental Medical Science (K.M.T., K.S., T.T.H., J.W., I.Y.M., M.E., V.M., A.H.-N., P.H., S.A.) and Department of Clinical Science (J.N., J.S.), Lund University, Lund, Sweden; and Departamento de Bioquímica y Biología Molecular y Fisiología and Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid and Consejo Superior de Investigaciones Científicas (CSIC), Valladolid, Spain (P.C., M.T.P.-G.)
| | - Tran Thi Hien
- From the Department of Experimental Medical Science (K.M.T., K.S., T.T.H., J.W., I.Y.M., M.E., V.M., A.H.-N., P.H., S.A.) and Department of Clinical Science (J.N., J.S.), Lund University, Lund, Sweden; and Departamento de Bioquímica y Biología Molecular y Fisiología and Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid and Consejo Superior de Investigaciones Científicas (CSIC), Valladolid, Spain (P.C., M.T.P.-G.)
| | - Johan Wohlfahrt
- From the Department of Experimental Medical Science (K.M.T., K.S., T.T.H., J.W., I.Y.M., M.E., V.M., A.H.-N., P.H., S.A.) and Department of Clinical Science (J.N., J.S.), Lund University, Lund, Sweden; and Departamento de Bioquímica y Biología Molecular y Fisiología and Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid and Consejo Superior de Investigaciones Científicas (CSIC), Valladolid, Spain (P.C., M.T.P.-G.)
| | - Ingrid Yao Mattisson
- From the Department of Experimental Medical Science (K.M.T., K.S., T.T.H., J.W., I.Y.M., M.E., V.M., A.H.-N., P.H., S.A.) and Department of Clinical Science (J.N., J.S.), Lund University, Lund, Sweden; and Departamento de Bioquímica y Biología Molecular y Fisiología and Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid and Consejo Superior de Investigaciones Científicas (CSIC), Valladolid, Spain (P.C., M.T.P.-G.)
| | - Mari Ekman
- From the Department of Experimental Medical Science (K.M.T., K.S., T.T.H., J.W., I.Y.M., M.E., V.M., A.H.-N., P.H., S.A.) and Department of Clinical Science (J.N., J.S.), Lund University, Lund, Sweden; and Departamento de Bioquímica y Biología Molecular y Fisiología and Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid and Consejo Superior de Investigaciones Científicas (CSIC), Valladolid, Spain (P.C., M.T.P.-G.)
| | - Johan Nilsson
- From the Department of Experimental Medical Science (K.M.T., K.S., T.T.H., J.W., I.Y.M., M.E., V.M., A.H.-N., P.H., S.A.) and Department of Clinical Science (J.N., J.S.), Lund University, Lund, Sweden; and Departamento de Bioquímica y Biología Molecular y Fisiología and Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid and Consejo Superior de Investigaciones Científicas (CSIC), Valladolid, Spain (P.C., M.T.P.-G.)
| | - Johan Sjögren
- From the Department of Experimental Medical Science (K.M.T., K.S., T.T.H., J.W., I.Y.M., M.E., V.M., A.H.-N., P.H., S.A.) and Department of Clinical Science (J.N., J.S.), Lund University, Lund, Sweden; and Departamento de Bioquímica y Biología Molecular y Fisiología and Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid and Consejo Superior de Investigaciones Científicas (CSIC), Valladolid, Spain (P.C., M.T.P.-G.)
| | - Vignesh Murugesan
- From the Department of Experimental Medical Science (K.M.T., K.S., T.T.H., J.W., I.Y.M., M.E., V.M., A.H.-N., P.H., S.A.) and Department of Clinical Science (J.N., J.S.), Lund University, Lund, Sweden; and Departamento de Bioquímica y Biología Molecular y Fisiología and Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid and Consejo Superior de Investigaciones Científicas (CSIC), Valladolid, Spain (P.C., M.T.P.-G.)
| | - Anna Hultgårdh-Nilsson
- From the Department of Experimental Medical Science (K.M.T., K.S., T.T.H., J.W., I.Y.M., M.E., V.M., A.H.-N., P.H., S.A.) and Department of Clinical Science (J.N., J.S.), Lund University, Lund, Sweden; and Departamento de Bioquímica y Biología Molecular y Fisiología and Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid and Consejo Superior de Investigaciones Científicas (CSIC), Valladolid, Spain (P.C., M.T.P.-G.)
| | - Pilar Cidad
- From the Department of Experimental Medical Science (K.M.T., K.S., T.T.H., J.W., I.Y.M., M.E., V.M., A.H.-N., P.H., S.A.) and Department of Clinical Science (J.N., J.S.), Lund University, Lund, Sweden; and Departamento de Bioquímica y Biología Molecular y Fisiología and Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid and Consejo Superior de Investigaciones Científicas (CSIC), Valladolid, Spain (P.C., M.T.P.-G.)
| | - Per Hellstrand
- From the Department of Experimental Medical Science (K.M.T., K.S., T.T.H., J.W., I.Y.M., M.E., V.M., A.H.-N., P.H., S.A.) and Department of Clinical Science (J.N., J.S.), Lund University, Lund, Sweden; and Departamento de Bioquímica y Biología Molecular y Fisiología and Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid and Consejo Superior de Investigaciones Científicas (CSIC), Valladolid, Spain (P.C., M.T.P.-G.)
| | - M. Teresa Pérez-García
- From the Department of Experimental Medical Science (K.M.T., K.S., T.T.H., J.W., I.Y.M., M.E., V.M., A.H.-N., P.H., S.A.) and Department of Clinical Science (J.N., J.S.), Lund University, Lund, Sweden; and Departamento de Bioquímica y Biología Molecular y Fisiología and Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid and Consejo Superior de Investigaciones Científicas (CSIC), Valladolid, Spain (P.C., M.T.P.-G.)
| | - Sebastian Albinsson
- From the Department of Experimental Medical Science (K.M.T., K.S., T.T.H., J.W., I.Y.M., M.E., V.M., A.H.-N., P.H., S.A.) and Department of Clinical Science (J.N., J.S.), Lund University, Lund, Sweden; and Departamento de Bioquímica y Biología Molecular y Fisiología and Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid and Consejo Superior de Investigaciones Científicas (CSIC), Valladolid, Spain (P.C., M.T.P.-G.)
| |
Collapse
|
16
|
Albinsson S, Bhattachariya A, Hellstrand P. Stretch-dependent smooth muscle differentiation in the portal vein-role of actin polymerization, calcium signaling, and microRNAs. Microcirculation 2015; 21:230-8. [PMID: 24238368 DOI: 10.1111/micc.12106] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Accepted: 11/09/2013] [Indexed: 11/28/2022]
Abstract
The mechanical forces acting on SMC in the vascular wall are known to regulate processes such as vascular remodeling and contractile differentiation. However, investigations to elucidate the underlying mechanisms of mechanotransduction in smooth muscle have been hampered by technical limitations associated with mechanical studies on pressurized small arteries, due primarily to the small amount of available tissue. The murine portal vein is a relatively large vessel showing myogenic tone that in many respects recapitulates the properties of small resistance vessels. Studies on stretched portal veins to elucidate mechanisms of mechanotransduction in the vascular wall have shown that stretch-sensitive regulation of contractile differentiation is mediated via Rho-activation and actin polymerization, while stretch-induced growth is regulated by the MAPK pathway. In this review, we have summarized findings on mechanotransduction in the portal vein with focus on stretch-induced contractile differentiation and the role of calcium, actin polymerization and miRNAs in this response.
Collapse
|
17
|
Smooth muscle strips for intestinal tissue engineering. PLoS One 2014; 9:e114850. [PMID: 25486279 PMCID: PMC4259486 DOI: 10.1371/journal.pone.0114850] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Accepted: 11/14/2014] [Indexed: 01/04/2023] Open
Abstract
Functionally contracting smooth muscle is an essential part of the engineered intestine that has not been replicated in vitro. The purpose of this study is to produce contracting smooth muscle in culture by maintaining the native smooth muscle organization. We employed intact smooth muscle strips and compared them to dissociated smooth muscle cells in culture for 14 days. Cells isolated by enzymatic digestion quickly lost maturity markers for smooth muscle cells and contained few enteric neural and glial cells. Cultured smooth muscle strips exhibited periodic contraction and maintained neural and glial markers. Smooth muscle strips cultured for 14 days also exhibited regular fluctuation of intracellular calcium, whereas cultured smooth muscle cells did not. After implantation in omentum for 14 days on polycaprolactone scaffolds, smooth muscle strip constructs expressed high levels of smooth muscle maturity markers as well as enteric neural and glial cells. Intact smooth muscle strips may be a useful component for engineered intestinal smooth muscle.
Collapse
|
18
|
Fan Z, Li C, Qin C, Xie L, Wang X, Gao Z, Qiangbacuozhen, Wang T, Yu L, Liu H. Role of the PI3K/AKT pathway in modulating cytoskeleton rearrangements and phenotype switching in rat pulmonary arterial vascular smooth muscle cells. DNA Cell Biol 2013; 33:12-9. [PMID: 24283363 DOI: 10.1089/dna.2013.2022] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Pulmonary arterial smooth muscle cell (PASMC) phenotype switching, which is characterized by changes in smooth muscle (SM)-specific gene expression, contributes to vascular remodeling in pulmonary hypertension. In addition, it has been shown that the transcription of SM-specific genes is modulated by cytoskeleton rearrangement. However, the intracellular mechanisms and signaling pathways that regulate these relationships are largely unknown. In the present study, we aimed to investigate the roles that phosphoinositide 3-kinase (PI3K) and protein kinase B (PKB), also known as AKT, play in modulating the cytoskeleton and phenotype of rat PASMCs. To observe the downstream effects of inhibiting or enhancing PI3K/AKT pathway activity, we used various approaches to manipulate protein function and gene expression. Treatment of PASMCs with platelet-derived growth factor (PDGF)-BB or PIK3CA-adenovirus induced cytoskeleton rearrangements and downregulated SM22α and α-SM actin gene expression. Inhibition of PI3K led to blocking of AKT phosphorylation and attenuated the PDGF-BB-induced downregulation of F-actin and SM-specific genes, the downstream effector of PI3K. The decrease in SM22α and α-SM actin mRNA levels induced by PDGF-BB was markedly and reproducibly blocked by LY294002. PI3K/AKT pathway plays a vital role in the modulation of PASMCs cytoskeleton rearrangement and phenotype switching.
Collapse
Affiliation(s)
- Zhiyu Fan
- 1 Pulmonary Vascular Remodeling Research Unit, West China Institute of Women's and Children's Health, West China Second University Hospital, Sichuan University , Chengdu, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Chiu CZ, Wang BW, Shyu KG. Effects of cyclic stretch on the molecular regulation of myocardin in rat aortic vascular smooth muscle cells. J Biomed Sci 2013; 20:50. [PMID: 23855625 PMCID: PMC3734126 DOI: 10.1186/1423-0127-20-50] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2013] [Accepted: 07/10/2013] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The expression of myocardin, a cardiac-restricted gene, increases during environmental stress. How mechanical stretch affects the regulation of myocardin in vascular smooth muscle cells (VSMCs) is not fully understood. We identify the mechanisms and pathways through which mechanical stretch induces myocardin expression in VSMCs. RESULTS Rat VSMCs grown on a flexible membrane base were stretched to 20% of maximum elongation, at 60 cycles per min. An in vivo model of aorta-caval shunt in adult rats was also used to investigate myocardin expression. Cyclic stretch significantly increased myocardin and angiotensin II (AngII) expression after 18 and 6 h of stretch. Addition of extracellular signal-regulated kinases (ERK) pathway inhibitor (PD98059), ERK small interfering RNA (siRNA), and AngII receptor blocker (ARB; losartan) before stretch inhibited the expression of myocardin protein. Gel shift assay showed that myocardin-DNA binding activity increased after stretch. PD98059, ERK siRNA and ARB abolished the binding activity induced by stretch. Stretch increased while myocardin-mutant plasmid, PD98059, and ARB abolished the promoter activity. Protein synthesis by measuring [3H]proline incorporation into the cells increased after cyclic stretch, which represented hypertrophic change of VSMCs. An in vivo model of aorta-caval shunt also demonstrated increased myocardin protein expression in the aorta. Confocal microscopy showed increased VSMC size 24 h after cyclic stretch and VSMC hypertrophy after creation of aorta-caval shunt for 3 days. CONCLUSIONS Cyclic stretch enhanced myocardin expression mediated by AngII through the ERK pathway in cultured rat VSMCs. These findings suggest that myocardin plays a role in stretch-induced VSMC hypertrophy.
Collapse
|
20
|
Turczyńska KM, Hellstrand P, Swärd K, Albinsson S. Regulation of vascular smooth muscle mechanotransduction by microRNAs and L-type calcium channels. Commun Integr Biol 2013; 6:e22278. [PMID: 23802033 PMCID: PMC3689564 DOI: 10.4161/cib.22278] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2012] [Revised: 09/17/2012] [Accepted: 09/18/2012] [Indexed: 11/19/2022] Open
Abstract
The phenotype of smooth muscle cells is regulated by multiple environmental factors including mechanical forces. Mechanical stretch of mouse portal veins ex vivo has been shown to promote contractile differentiation by activation of the Rho-pathway, an effect that is dependent on the influx of calcium via L-type calcium channels. MicroRNAs have recently been demonstrated to play a significant role in the control of smooth muscle phenotype and in a recent report we investigated their role in vascular mechanosensing. By smooth muscle specific deletion of Dicer, we found that microRNAs are essential for smooth muscle differentiation in response to stretch by regulating CamKIIδ and L-type calcium channel expression. Furthermore, we suggest that loss of L-type calcium channels in Dicer KO is due to reduced expression of the smooth muscle-enriched microRNA, miR-145, which targets CamKIIδ. These results unveil a novel mechanism for miR-145 dependent regulation of smooth muscle phenotype.
Collapse
|
21
|
Turczyńska KM, Bhattachariya A, Säll J, Göransson O, Swärd K, Hellstrand P, Albinsson S. Stretch-sensitive down-regulation of the miR-144/451 cluster in vascular smooth muscle and its role in AMP-activated protein kinase signaling. PLoS One 2013; 8:e65135. [PMID: 23705032 PMCID: PMC3660603 DOI: 10.1371/journal.pone.0065135] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2013] [Accepted: 04/22/2013] [Indexed: 11/25/2022] Open
Abstract
Vascular smooth muscle cells are constantly exposed to mechanical force by the blood pressure, which is thought to regulate smooth muscle growth, differentiation and contractile function. We have previously shown that the expression of microRNAs (miRNAs), small non-coding RNAs, is essential for regulation of smooth muscle phenotype including stretch-dependent contractile differentiation. In this study, we have investigated the effect of mechanical stretch on miRNA expression and the role of stretch-sensitive miRNAs for intracellular signaling in smooth muscle. MiRNA array analysis, comparing miRNA levels in stretched versus non-stretched portal veins, revealed a dramatic decrease in the miR-144/451 cluster level. Because this miRNA cluster is predicted to target AMPK pathway components, we next examined activation of this pathway. Diminished miR-144/451 expression was inversely correlated with increased phosphorylation of AMPKα at Thr172 in stretched portal vein. Similar to the effect of stretch, contractile differentiation could be induced in non-stretched portal veins by the AMPK activator, AICAR. Transfection with miR-144/451 mimics reduced the protein expression level of mediators in the AMPK pathway including MO25α, AMPK and ACC. This effect also decreased AICAR-induced activation of the AMPK signaling pathway. In conclusion, our results suggest that stretch-induced activation of AMPK in vascular smooth muscle is in part regulated by reduced levels of miR-144/451 and that this effect may play a role in promoting contractile differentiation of smooth muscle cells.
Collapse
|
22
|
Richardson WJ, Wilson E, Moore JE. Altered phenotypic gene expression of 10T1/2 mesenchymal cells in nonuniformly stretched PEGDA hydrogels. Am J Physiol Cell Physiol 2013; 305:C100-10. [PMID: 23657569 DOI: 10.1152/ajpcell.00340.2012] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Disease-related phenotype modulation of many cell types has been shown to be closely related to mechanical loading conditions; for example, vascular smooth muscle cell (SMC) phenotype shift from a mature, contractile state to a proliferative, synthetic state contributes to the formation of neointimal tissue during atherosclerosis and restenosis development and is related to SMC mechanical loading in vivo. The majority of past in vitro cell-stretching experiments have employed simplistic (uniform, uniaxial or biaxial) stretching environments to elucidate mechanobiological pathways involved in phenotypic shifts. However, the in vivo mechanics of the vascular wall consists of highly nonuniform stretch. Here we subjected 10T1/2 murine mesenchymal cells (an SMC precursor) to two- and three-dimensional nonuniform stretch environments. After 24 h of stretch, cells on an elastomeric membrane demonstrated varied proliferation [assessed by 5-bromo-2'-deoxyuridine (BrdU) incorporation] depending on location upon the membrane, with maximal proliferation occurring in a region of high, uniaxial stretch. Cells subjected to a nonuniform stretching regimen within three-dimensional polyethylene glycol diacrylate (PEGDA) hydrogel constructs demonstrated marked changes in mRNA expression of several phenotype-related proteins, indicating a sort of "hybrid" phenotype with contractile and synthetic markers being both upregulated and downregulated. Furthermore, expression levels of mRNAs were significantly different between various locations within the stretched gel. With the proliferation results, these data exhibit the capability of nonuniform stretching devices to induce heterogeneous cell responses, potentially indicative of spatial distributions of disease-related behaviors in vivo.
Collapse
Affiliation(s)
- W J Richardson
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA
| | | | | |
Collapse
|
23
|
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
|
24
|
Yang XR, Lin AHY, Hughes JM, Flavahan NA, Cao YN, Liedtke W, Sham JSK. Upregulation of osmo-mechanosensitive TRPV4 channel facilitates chronic hypoxia-induced myogenic tone and pulmonary hypertension. Am J Physiol Lung Cell Mol Physiol 2011; 302:L555-68. [PMID: 22207590 DOI: 10.1152/ajplung.00005.2011] [Citation(s) in RCA: 91] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Chronic hypoxia causes pulmonary hypertension with vascular remodeling, increase in vascular tone, and altered reactivity to agonists. These changes involve alterations in multiple Ca(2+) pathways in pulmonary arterial smooth muscle cells (PASMCs). We have previously shown that vanilloid (TRPV)- and melastatin-related transient receptor potential (TRPM) channels are expressed in pulmonary arteries (PAs). Here we found that TRPV4 was the only member of the TRPV and TRPM subfamilies upregulated in PAs of chronic hypoxic rats. The increase in TRPV4 expression occurred within 1 day of hypoxia exposure, indicative of an early hypoxic response. TRPV4 in PASMCs were found to be mechanosensitive. Osmo-mechanical stress imposed by hypotonic solution activated Ca(2+) transients; they were inhibited by TRPV4 specific short interfering RNA, the TRPV blocker ruthenium red, and the cytochrome P450 epoxygenase inhibitor N-(methylsulfonyl)-2-(2-propynyloxy)-benzenehexanamide. Consistent with TRPV4 upregulation, the Ca(2+) response induced by the TRPV4 agonist 4α-phorbol 12,13-didecanoate and hypotonicity was potentiated in hypoxic PASMCs. Moreover, a significant myogenic tone, sensitive to ruthenium red, was observed in pressurized endothelium denuded small PAs of hypoxic but not normoxic rats. The elevated basal intracellular Ca(2+) concentration in hypoxic PASMCs was also reduced by ruthenium red. In extension of these results, the development of pulmonary hypertension, right heart hypertrophy, and vascular remodeling was significantly delayed and suppressed in hypoxic trpv4(-/-) mice. These results suggest the novel concept that TRPV4 serves as a signal pathway crucial for the development of hypoxia-induced pulmonary hypertension. Its upregulation may provide a pathogenic feed-forward mechanism that promotes pulmonary hypertension via facilitated Ca(2+) influx, subsequently enhanced myogenic tone and vascular remodeling.
Collapse
Affiliation(s)
- Xiao-Ru Yang
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | | | | | | | | | | | | |
Collapse
|
25
|
Albinsson S, Skoura A, Yu J, DiLorenzo A, Fernández-Hernando C, Offermanns S, Miano JM, Sessa WC. Smooth muscle miRNAs are critical for post-natal regulation of blood pressure and vascular function. PLoS One 2011; 6:e18869. [PMID: 21526127 PMCID: PMC3081311 DOI: 10.1371/journal.pone.0018869] [Citation(s) in RCA: 102] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2010] [Accepted: 03/22/2011] [Indexed: 01/19/2023] Open
Abstract
Phenotypic modulation of smooth muscle cells (SMCs) plays a key role in vascular disease, including atherosclerosis. Several transcription factors have been suggested to regulate phenotypic modulation of SMCs but the decisive mechanisms remain unknown. Recent reports suggest that specific microRNAs (miRNAs) are involved in SMC differentiation and vascular disease but the global role of miRNAs in postnatal vascular SMC has not been elucidated. Thus, the objective of this study was to identify the role of Dicer-dependent miRNAs for blood pressure regulation and vascular SMC contractile function and differentiation in vivo. Tamoxifen-inducible and SMC specific deletion of Dicer was achieved by Cre-Lox recombination. Deletion of Dicer resulted in a global loss of miRNAs in aortic SMC. Furthermore, Dicer-deficient mice exhibited a dramatic reduction in blood pressure due to significant loss of vascular contractile function and SMC contractile differentiation as well as vascular remodeling. Several of these results are consistent with our previous observations in SM-Dicer deficient embryos. Therefore, miRNAs are essential for maintaining blood pressure and contractile function in resistance vessels. Although the phenotype of miR-143/145 deficient mice resembles the loss of Dicer, the phenotypes of SM-Dicer KO mice were far more severe suggesting that additional miRNAs are involved in maintaining postnatal SMC differentiation.
Collapse
Affiliation(s)
- Sebastian Albinsson
- Department of Pharmacology, Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Athanasia Skoura
- Department of Pharmacology, Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Jun Yu
- Department of Pharmacology, Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Annarita DiLorenzo
- Department of Pharmacology, Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Carlos Fernández-Hernando
- Department of Pharmacology, Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Stefan Offermanns
- Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Joseph M. Miano
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
| | - William C. Sessa
- Department of Pharmacology, Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, Connecticut, United States of America
| |
Collapse
|
26
|
Vazão H, Neves RPD, Grãos M, Ferreira L. Towards the maturation and characterization of smooth muscle cells derived from human embryonic stem cells. PLoS One 2011; 6:e17771. [PMID: 21423769 PMCID: PMC3053392 DOI: 10.1371/journal.pone.0017771] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2010] [Accepted: 02/14/2011] [Indexed: 01/04/2023] Open
Abstract
In this study we demonstrate that CD34(+) cells derived from human embryonic stem cells (hESCs) have higher smooth muscle cell (SMC) potential than CD34(-) cells. We report that from all inductive signals tested, retinoic acid (RA) and platelet derived growth factor (PDGF(BB)) are the most effective agents in guiding the differentiation of CD34(+) cells into smooth muscle progenitor cells (SMPCs) characterized by the expression of SMC genes and proteins, secretion of SMC-related cytokines, contraction in response to depolarization agents and vasoactive peptides and expression of SMC-related genes in a 3D environment. These cells are also characterized by a low organization of the contractile proteins and the contractility response is mediated by Ca(2+), which involves the activation of Rho A/Rho kinase- and Ca(2+)/calmodulin (CaM)/myosin light chain kinase (MLCK)-dependent pathways. We further show that SMPCs obtained from the differentiation of CD34(+) cells with RA, but not with PDGF(BB,) can be maturated in medium supplemented with endothelin-1 showing at the end individualized contractile filaments. Overall the hESC-derived SMCs presented in this work might be an unlimited source of SMCs for tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Helena Vazão
- CNC - Center of Neurosciences and Cell Biology, University of Coimbra, Coimbra, Portugal
- Biocant - Center of Innovation in Biotechnology, Cantanhede, Portugal
| | - Ricardo Pires das Neves
- CNC - Center of Neurosciences and Cell Biology, University of Coimbra, Coimbra, Portugal
- Biocant - Center of Innovation in Biotechnology, Cantanhede, Portugal
| | - Mário Grãos
- Biocant - Center of Innovation in Biotechnology, Cantanhede, Portugal
| | - Lino Ferreira
- CNC - Center of Neurosciences and Cell Biology, University of Coimbra, Coimbra, Portugal
- Biocant - Center of Innovation in Biotechnology, Cantanhede, Portugal
| |
Collapse
|
27
|
Mohamed JS, Lopez MA, Boriek AM. Mechanical stretch up-regulates microRNA-26a and induces human airway smooth muscle hypertrophy by suppressing glycogen synthase kinase-3β. J Biol Chem 2010; 285:29336-47. [PMID: 20525681 PMCID: PMC2937966 DOI: 10.1074/jbc.m110.101147] [Citation(s) in RCA: 170] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2010] [Revised: 05/28/2010] [Indexed: 12/19/2022] Open
Abstract
Airway smooth muscle hypertrophy is one of the hallmarks of airway remodeling in severe asthma. Several human diseases have been now associated with dysregulated microRNA (miRNA) expression. miRNAs are a class of small non-coding RNAs, which negatively regulate gene expression at the post-transcriptional level. Here, we identify miR-26a as a hypertrophic miRNA of human airway smooth muscle cells (HASMCs). We show that stretch selectively induces the transcription of miR-26a located in the locus 3p21.3 of human chromosome 3. The transcription factor CCAAT enhancer-binding protein α (C/EBPα) directly activates miR-26a expression through the transcriptional machinery upon stretch. Furthermore, stretch or enforced expression of miR-26a induces HASMC hypertrophy, and miR-26 knockdown reverses this effect, suggesting that miR-26a is a hypertrophic gene. We identify glycogen synthase kinase-3β (GSK-3β), an anti-hypertrophic protein, as a target gene of miR-26a. Luciferase reporter assays demonstrate that miR-26a directly interact with the 3'-untranslated repeat of the GSK-3β mRNA. Stretch or enforced expression of miR-26a attenuates the endogenous GSK-3β protein levels followed by the induction of HASMC hypertrophy. miR-26 knockdown reverses this effect, suggesting that miR-26a-induced hypertrophy occurs via its target gene GSK-3β. Overall, as a first time, our study unveils that miR-26a is a mechanosensitive gene, and it plays an important role in the regulation of HASMC hypertrophy.
Collapse
Affiliation(s)
- Junaith S. Mohamed
- From Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Baylor College of Medicine, Houston, Texas 77030
| | - Michael A. Lopez
- From Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Baylor College of Medicine, Houston, Texas 77030
| | - Aladin M. Boriek
- From Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Baylor College of Medicine, Houston, Texas 77030
| |
Collapse
|
28
|
Xiao Y, Huang Z, Yin H, Zhang H, Wang S. Desmuslin gene knockdown causes altered expression of phenotype markers and differentiation of saphenous vein smooth muscle cells. J Vasc Surg 2010; 52:684-90. [PMID: 20573469 DOI: 10.1016/j.jvs.2010.03.069] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2010] [Revised: 03/23/2010] [Accepted: 03/28/2010] [Indexed: 01/12/2023]
Abstract
OBJECTIVE Phenotypic alterations of vascular smooth muscle cells (VSMCs) appear critical to the development of primary varicose veins. Previous study indicated desmuslin, an intermediate filament protein, was differentially expressed in smooth muscle cells (SMCs) isolated from varicose veins; thus, it was naturally hypothesized that altered desmuslin expression might in turn affect the functioning of VSMCs, leading to the phenotypic alterations and varicose vein development. METHODS In this study, expression of desmuslin in normal human saphenous vein SMCs was knocked down using small interfering RNA (siRNA), and control cells were treated with a scrambled siRNA sequence. The levels of several phenotypic markers including smooth muscle (SM) alpha-actin and smooth muscle myosin heavy chain (SM-MHC) were assessed. Collagen formation, matrix metalloproteinase expression (MMP-2), and cytoskeletal and morphological changes were also examined. RESULTS SMCs treated with desmuslin siRNA exhibited significantly increased levels of collagen synthesis and MMP-2 expression and decreased expression levels of SM alpha-actin, SM-MHC, and smoothelin and exhibited disassembly of actin stress fibers when compared with the control cells. Changes in cell morphology and actin fiber networks in VSMCs treated with desmuslin siRNA were consistent with a lower degree of differentiation. CONCLUSIONS These results indicated desmuslin expression is required for the maintenance of VSMC phenotype. Decreased desmuslin expression may affect differentiation of VSMCs and ultimately contribute to the development of varicose veins.
Collapse
Affiliation(s)
- Ying Xiao
- Vascular Surgery Institute, Department of Vascular Surgery, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China
| | | | | | | | | |
Collapse
|
29
|
Angiotensin II and the ERK pathway mediate the induction of myocardin by hypoxia in cultured rat neonatal cardiomyocytes. Clin Sci (Lond) 2010; 119:273-82. [PMID: 20446923 PMCID: PMC2890999 DOI: 10.1042/cs20100084] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Hypoxic injury to cardiomyocytes is a stress that causes cardiac pathology through cardiac-restricted gene expression. SRF (serum-response factor) and myocardin are important for cardiomyocyte growth and differentiation in response to myocardial injuries. Previous studies have indicated that AngII (angiotensin II) stimulates both myocardin expression and cardiomyocyte hypertrophy. In the present study, we evaluated the expression of myocardin and AngII after hypoxia in regulating gene transcription in neonatal cardiomyocytes. Cultured rat neonatal cardiomyocytes were subjected to hypoxia, and the expression of myocardin and AngII were evaluated. Different signal transduction pathway inhibitors were used to identify the pathway(s) responsible for myocardin expression. An EMSA (electrophoretic mobility-shift assay) was used to identify myocardin/SRF binding, and a luciferase assay was used to identify transcriptional activity of myocardin/SRF in neonatal cardiomyocytes. Both myocardin and AngII expression increased after hypoxia, with AngII appearing at an earlier time point than myocardin. Myocardin expression was stimulated by AngII and ERK (extracellular-signal-regulated kinase) phosphorylation, but was suppressed by an ARB (AngII type 1 receptor blocker), an ERK pathway inhibitor and myocardin siRNA (small interfering RNA). AngII increased both myocardin expression and transcription in neonatal cardiomyocytes. Binding of myocardin/SRF was identified using an EMSA, and a luciferase assay indicated the transcription of myocardin/SRF in neonatal cardiomyocytes. Increased BNP (B-type natriuretic peptide), MHC (myosin heavy chain) and [3H]proline incorporation into cardiomyocytes was identified after hypoxia with the presence of myocardin in hypertrophic cardiomyocytes. In conclusion, hypoxia in cardiomyocytes increased myocardin expression, which is mediated by the induction of AngII and the ERK pathway, to cause cardiomyocyte hypertrophy. Myocardial hypertrophy was identified as an increase in transcriptional activities, elevated hypertrophic and cardiomyocyte phenotype markers, and morphological hypertrophic changes in cardiomyocytes.
Collapse
|
30
|
Foster CD, Varghese LS, Gonzales LW, Margulies SS, Guttentag SH. The Rho pathway mediates transition to an alveolar type I cell phenotype during static stretch of alveolar type II cells. Pediatr Res 2010; 67:585-90. [PMID: 20220547 PMCID: PMC3063400 DOI: 10.1203/pdr.0b013e3181dbc708] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Stretch is an essential mechanism for lung growth and development. Animal models in which fetal lungs have been chronically over or underdistended demonstrate a disrupted mix of type II and type I cells, with static overdistention typically promoting a type I cell phenotype. The Rho GTPase family, key regulators of cytoskeletal signaling, are known to mediate cellular differentiation in response to stretch in other organs. Using a well-described model of alveolar epithelial cell differentiation and a validated stretch device, we investigated the effects of supraphysiologic stretch on human fetal lung alveolar epithelial cell phenotype. Static stretch applied to epithelial cells suppressed type II cell markers (SP-B and Pepsinogen C, PGC), and induced type I cell markers (Caveolin-1, Claudin 7 and Plasminogen Activator Inhibitor-1, PAI-1) as predicted. Static stretch was also associated with Rho A activation. Furthermore, the Rho kinase inhibitor Y27632 decreased Rho A activation and blunted the stretch-induced changes in alveolar epithelial cell marker expression. Together these data provide further evidence that mechanical stimulation of the cytoskeleton and Rho activation are key upstream events in mechanotransduction-associated alveolar epithelial cell differentiation.
Collapse
Affiliation(s)
- Cherie D Foster
- Department of Pediatrics, The Children's Hospital of Philadelphia, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | | | | | | | | |
Collapse
|
31
|
Wang X, Sun Z. Thyroid hormone induces artery smooth muscle cell proliferation: discovery of a new TRalpha1-Nox1 pathway. J Cell Mol Med 2010; 14:368-80. [PMID: 20414976 PMCID: PMC2888973 DOI: 10.1111/j.1582-4934.2008.00489.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2008] [Accepted: 08/22/2008] [Indexed: 11/30/2022] Open
Abstract
Thyroid hormone (T3) can stimulate protein synthesis and cell growth. NOX1 is a mitogenic oxidase. The aim of this study was to test a novel hypothesis that T3 induces artery smooth muscle cell proliferation by up-regulating NOX1. Immunofluoresence confocal microscopy was used to visualize the sub-cellular localization of NOX1 and TRalpha1 in rat aorta smooth muscle (RASM) cells. Optical sectioning showed that TRalpha1 and NOX1 co-localized around the nucleus. T3 promoted RASM cell proliferation as determined by the fact that T3 significantly increased the number of cytokinesis cells, proliferating cellular nuclear antigen (PCNA) and smooth muscle alpha-actin (SM alpha-actin). T3 increased NOX1 expression at both the transcription (mRNA) and translation (protein) levels as evaluated by RT-PCR and Western blot, respectively. T3 also significantly increased the intracellular ROS production based on the oxidation of 2',7'-dichlorodihydrofluoresein (H2DCF) to a fluorescent 2',7'-dichlorofluoresein (DCF). RNAi silence of TRalpha1 or NOX1 abolished T3-induced intracellular ROS generation and PCNA and SM alpha-actin expression, indicating that TRalpha1 and NOX1 mediated T3-induced RASM cell proliferation. Notably, RNAi silence of TRalpha1 blocked the T3-induced increase in NOX1 expression, whereas silence of NOX1 did not affect TRalpha1 expression, disclosing a new pathway, i.e. T3-TRalpha1-NOX1-cell proliferation. TRalpha1 and NOX1 co-localized around the nucleus. T3 induced RASM cell proliferation by up-regulating NOX1 in a TRalpha1-dependent manner.
Collapse
Affiliation(s)
- Xiuqing Wang
- Department of Physiology, College of Medicine, University of Oklahoma Health Sciences Center (OUHSC)Oklahoma City, OK, USA
| | - Zhongjie Sun
- Department of Physiology, College of Medicine, University of Oklahoma Health Sciences Center (OUHSC)Oklahoma City, OK, USA
| |
Collapse
|
32
|
Zheng JP, Ju D, Shen J, Yang M, Li L. Disruption of actin cytoskeleton mediates loss of tensile stress induced early phenotypic modulation of vascular smooth muscle cells in organ culture. Exp Mol Pathol 2009; 88:52-7. [PMID: 19874818 DOI: 10.1016/j.yexmp.2009.10.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2009] [Accepted: 10/19/2009] [Indexed: 12/29/2022]
Abstract
Aorta organ culture has been widely used as an ex vivo model for studying vessel pathophysiology. Recent studies show that the vascular smooth muscle cells (VSMCs) in organ culture undergo drastic dedifferentiation within the first few hours (termed early phenotypic modulation). Loss of tensile stress to which aorta is subject in vivo is the cause of this early phenotypic modulation. However, no underlying molecular mechanism has been discovered thus far. The purpose of the present study is to identify intracellular signals involved in the early phenotypic modulation of VSMC in organ culture. We find that the drastic VSMC dedifferentiation is accompanied by accelerated actin cytoskeleton dynamics and downregulation of SRF and myocardin. Among the variety of signal pathways examined, increasing actin polymerization by jasplakinolide is the only one hindering VSMC dedifferentiation in organ culture. Moreover, jasplakinolide reverses actin dynamics during organ culture. Latrunculin B (disrupting actin cytoskeleton) and jasplakinolide respectively suppressed and enhanced the expression of VSMC markers, SRF, myocardin, and CArG-box-mediated SMC promoters in PAC1, a VSMC line. These results identify actin cytoskeleton degradation as a major intracellular signal for loss of tensile stress-induced early phenotypic modulation of VSMC in organ culture. This study suggests that disrupting actin cytoskeleton integrity may contribute to the pathogenesis of vascular diseases.
Collapse
Affiliation(s)
- Jian-Pu Zheng
- Department of Internal Medicine, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | | | | | | | | |
Collapse
|
33
|
Kim SG, Jang SJ, Soh J, Lee K, Park JK, Chang WK, Park EW, Chun SY. Expression of ectodermal neural cortex 1 and its association with actin during the ovulatory process in the rat. Endocrinology 2009; 150:3800-6. [PMID: 19372205 DOI: 10.1210/en.2008-1587] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Ectodermal neural cortex (ENC) 1, a member of the kelch family of genes, is an actin-binding protein and plays a pivotal role in neuronal and adipocyte differentiation. The present study was designed to examine the gonadotropin regulation and action of ENC1 during the ovulatory process in immature rats. The levels of ENC1 mRNA and protein were stimulated by LH/human chorionic gonadotropin (hCG) within 3 h both in vivo and in vitro. In situ hybridization analysis revealed that ENC1 mRNA was localized not only in theca/interstitial cells but also in granulosa cells of preovulatory follicles but not of growing follicles in pregnant mare's serum gonadotropin/hCG-treated ovaries. LH-induced ENC1 expression was suppressed by a high dose of protein kinase C inhibitor RO 31-8220 (10 microM) but not by low doses of RO 31-8220 (0.1-1.0 microM), suggesting the involvement of atypical protein kinase C. ENC1 was detected in both nucleus and cytoplasm that was increased by LH/hCG treatment. Both biochemical and morphological analysis revealed that LH/hCG treatment increased actin polymerization within 3 h in granulosa cells. Interestingly, ENC1 physically associated with actin and treatment with cytochalasin D, an actin-depolymerizing agent, abolished this association. Confocal microscopy further demonstrated the colocalization of ENC1 with filamentous actin (F-actin). The present study demonstrates that LH/hCG stimulates ENC1 expression and increases F-actin formation in granulosa cells. The present study further shows the physical association of ENC1 and F-actin, implicating the role of ENC1 in cytoskeletal reorganization during the differentiation of granulosa cells.
Collapse
Affiliation(s)
- Sun-Gyun Kim
- Hormone Research Center and School of Biological Sciences and Technology, Chonnam National University, Kwangju 500-712, Republic of Korea
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Pérot G, Derré J, Coindre JM, Tirode F, Lucchesi C, Mariani O, Gibault L, Guillou L, Terrier P, Aurias A. Strong smooth muscle differentiation is dependent on myocardin gene amplification in most human retroperitoneal leiomyosarcomas. Cancer Res 2009; 69:2269-78. [PMID: 19276386 DOI: 10.1158/0008-5472.can-08-1443] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Myocardin (MYOCD), a serum response factor (SRF) transcriptional cofactor, is essential for cardiac and smooth muscle development and differentiation. We show here by array-based comparative genomic hybridization, fluorescence in situ hybridization, and expression analysis approaches that MYOCD gene is highly amplified and overexpressed in human retroperitoneal leiomyosarcomas (LMS), a very aggressive well-differentiated tumor. MYOCD inactivation by shRNA in a human LMS cell line with MYOCD locus amplification leads to a dramatic decrease of smooth muscle differentiation and strongly reduces cell migration. Moreover, forced MYOCD expression in three undifferentiated sarcoma cell lines and in one liposarcoma cell line confers a strong smooth muscle differentiation phenotype and increased migration abilities. Collectively, these results show that human retroperitoneal LMS differentiation is dependent on MYOCD amplification/overexpression, suggesting that in these well-differentiated LMS, differentiation could be a consequence of an acquired genomic alteration. In this hypothesis, these tumors would not necessarily derive from cells initially committed to smooth muscle differentiation. These data also provide new insights on the cellular origin of these sarcomas and on the complex connections between oncogenesis and differentiation in mesenchymal tumors.
Collapse
Affiliation(s)
- Gaëlle Pérot
- Genetics and Biology of Cancers, Institut Curie, Paris, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Shyu KG. Cellular and molecular effects of mechanical stretch on vascular cells and cardiac myocytes. Clin Sci (Lond) 2009; 116:377-389. [DOI: 10.1042/cs20080163] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Abstract
Cells in the cardiovascular system are permanently subjected to mechanical forces due to the pulsatile nature of blood flow and shear stress, created by the beating heart. These haemodynamic forces play an important role in the regulation of vascular development, remodelling, wound healing and atherosclerotic lesion formation. Mechanical stretch can modulate several different cellular functions in VSMCs (vascular smooth muscle cells). These functions include, but are not limited to, cell alignment and differentiation, migration, survival or apoptosis, vascular remodelling, and autocrine and paracrine functions. Laminar shear stress exerts anti-apoptotic, anti-atherosclerotic and antithrombotic effects on ECs (endothelial cells). Mechanical stretch of cardiac myocytes can modulate growth, apoptosis, electric remodelling, alterations in gene expression, and autocrine and paracrine effects. The aim of the present review is primarily to summarize the cellular and molecular effects of mechanical stretch on vascular cells and cardiac myocytes, emphasizing the molecular mechanisms underlying the regulation. Knowledge of the impact of mechanical stretch on the cardiovascular system is vital to the understanding of the pathogenesis of cardiovascular diseases, and is also crucial to provide new insights into the prevention and therapy of cardiovascular diseases.
Collapse
Affiliation(s)
- Kou-Gi Shyu
- Division of Cardiology, Shin Kong Wu Ho-Su Memorial Hospital, 95 Wen-Chang Rd, Taipei 111, Taiwan, and Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| |
Collapse
|
36
|
Luykenaar KD, El-Rahman RA, Walsh MP, Welsh DG. Rho-kinase-mediated suppression of KDR current in cerebral arteries requires an intact actin cytoskeleton. Am J Physiol Heart Circ Physiol 2009; 296:H917-26. [PMID: 19218502 DOI: 10.1152/ajpheart.01206.2008] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
This study examined the role of the actin cytoskeleton in Rho-kinase-mediated suppression of the delayed-rectifier K(+) (K(DR)) current in cerebral arteries. Myocytes from rat cerebral arteries were enzymatically isolated, and whole cell K(DR) currents were monitored using conventional patch-clamp electrophysiology. At +40 mV, the K(DR) current averaged 19.8 +/- 1.6 pA/pF (mean +/- SE) and was potently inhibited by UTP (3 x 10(-5) M). This suppression was observed to depend on Rho signaling and was abolished by the Rho-kinase inhibitors H-1152 (3 x 10(-7) M) and Y-27632 (3 x 10(-5) M). Rho-kinase was also found to concomitantly facilitate actin polymerization in response to UTP. We therefore examined whether actin dynamics played a role in the ability of Rho-kinase to suppress K(DR) current and found that actin disruption using either cytochalasin D (1 x 10(-5) M) or latrunculin A (1 x 10(-8) M) prevented current modulation. Consistent with our electrophysiological observations, both Rho-kinase inhibition and actin disruption significantly attenuated UTP-induced depolarization and constriction of cerebral arteries. We propose that UTP initiates Rho-kinase-mediated remodeling of the actin cytoskeleton and consequently suppresses the K(DR) current, thereby facilitating the depolarization and constriction of cerebral arteries.
Collapse
|
37
|
Abstract
Mechanotransduction research has focused historically on how externally applied forces can affect cell signalling and function. A growing body of evidence suggests that contractile forces that are generated internally by the actomyosin cytoskeleton are also important in regulating cell behaviour, and suggest a broader role for mechanotransduction in biology. Although the molecular basis for these cellular forces in mechanotransduction is being pursued in cell culture, researchers are also beginning to appreciate their contribution to in vivo developmental processes. Here, we examine the role for mechanical forces and contractility in regulating cell and tissue structure and function during development.
Collapse
Affiliation(s)
- Michele A. Wozniak
- Department of Bioengineering, University of Pennsylvania, Philadelphia PA 19104
| | - Christopher S. Chen
- Department of Bioengineering, University of Pennsylvania, Philadelphia PA 19104
| |
Collapse
|
38
|
Gunst SJ, Zhang W. Actin cytoskeletal dynamics in smooth muscle: a new paradigm for the regulation of smooth muscle contraction. Am J Physiol Cell Physiol 2008; 295:C576-87. [PMID: 18596210 PMCID: PMC2544441 DOI: 10.1152/ajpcell.00253.2008] [Citation(s) in RCA: 276] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2008] [Accepted: 06/30/2008] [Indexed: 11/22/2022]
Abstract
A growing body of data supports a view of the actin cytoskeleton of smooth muscle cells as a dynamic structure that plays an integral role in regulating the development of mechanical tension and the material properties of smooth muscle tissues. The increase in the proportion of filamentous actin that occurs in response to the stimulation of smooth muscle cells and the essential role of stimulus-induced actin polymerization and cytoskeletal dynamics in the generation of mechanical tension has been convincingly documented in many smooth muscle tissues and cells using a wide variety of experimental approaches. Most of the evidence suggests that the functional role of actin polymerization during contraction is distinct and separately regulated from the actomyosin cross-bridge cycling process. The molecular basis for the regulation of actin polymerization and its physiological roles may vary in diverse types of smooth muscle cells and tissues. However, current evidence supports a model for smooth muscle contraction in which contractile stimulation initiates the assembly of cytoskeletal/extracellular matrix adhesion complex proteins at the membrane, and proteins within this complex orchestrate the polymerization and organization of a submembranous network of actin filaments. This cytoskeletal network may serve to strengthen the membrane for the transmission of force generated by the contractile apparatus to the extracellular matrix, and to enable the adaptation of smooth muscle cells to mechanical stresses. Better understanding of the physiological function of these dynamic cytoskeletal processes in smooth muscle may provide important insights into the physiological regulation of smooth muscle tissues.
Collapse
Affiliation(s)
- Susan J Gunst
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA.
| | | |
Collapse
|
39
|
Milewicz DM, Guo DC, Tran-Fadulu V, Lafont AL, Papke CL, Inamoto S, Kwartler CS, Pannu H. Genetic Basis of Thoracic Aortic Aneurysms and Dissections: Focus on Smooth Muscle Cell Contractile Dysfunction. Annu Rev Genomics Hum Genet 2008; 9:283-302. [DOI: 10.1146/annurev.genom.8.080706.092303] [Citation(s) in RCA: 315] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Dianna M. Milewicz
- Department of Internal Medicine, University of Texas, Houston, Texas 77030;
| | - Dong-Chuan Guo
- Department of Internal Medicine, University of Texas, Houston, Texas 77030;
| | - Van Tran-Fadulu
- Department of Internal Medicine, University of Texas, Houston, Texas 77030;
| | - Andrea L. Lafont
- Department of Internal Medicine, University of Texas, Houston, Texas 77030;
| | - Christina L. Papke
- Department of Internal Medicine, University of Texas, Houston, Texas 77030;
| | - Sakiko Inamoto
- Department of Internal Medicine, University of Texas, Houston, Texas 77030;
| | - Carrie S. Kwartler
- Department of Internal Medicine, University of Texas, Houston, Texas 77030;
| | - Hariyadarshi Pannu
- Department of Internal Medicine, University of Texas, Houston, Texas 77030;
| |
Collapse
|
40
|
O'Cearbhaill ED, Punchard MA, Murphy M, Barry FP, McHugh PE, Barron V. Response of mesenchymal stem cells to the biomechanical environment of the endothelium on a flexible tubular silicone substrate. Biomaterials 2008; 29:1610-9. [PMID: 18194813 DOI: 10.1016/j.biomaterials.2007.11.042] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2007] [Accepted: 11/28/2007] [Indexed: 12/21/2022]
Abstract
Understanding the response of mesenchymal stem cells (MSCs) to forces in the vasculature is very important in the field of cardiovascular intervention for a number of reasons. These include the development of MSC seeded tissue engineered vascular grafts, targeted or systemic delivery of MSCs in the dynamic environment of the coronary artery and understanding the potential pathological calcifying role of mechanically conditioned multipotent cells already present in the vessel wall. In vivo, cells present in the coronary artery are exposed to the primary biomechanical forces of shear stress, radial stress and hoop stress. To date, many studies have examined the effect of these stresses in isolation, thereby not presenting the complete picture. Therefore, the main aim of this study is to examine the combined role of these stresses on MSC behaviour. To this end, a bioreactor was configured to expose MSCs seeded on flexible silicone substrates to physiological forces - namely, a pulsatile pressure between 40 and 120mmHg (5.33-1.6x10(4)Pa), radial distention of 5% and a shear stress of 10dyn/cm(2) (1Pa) at frequency of 1Hz for up to 24h. Thereafter, the 'pseudovessel' was assessed for changes in morphology, orientation and expression of endothelial and smooth muscle cell (SMC) specific markers. Hematoxylin and eosin (H&E) staining revealed that MSCs exhibit a similar mechanosensitive response to that of endothelial cells (ECs); they reorientate parallel with direction of flow and have adapted their morphology to be similar to that of ECs. However, gene expression results show the cells exhibit greater levels of SMC-associated markers alpha-smooth muscle actin and calponin (p<0.05).
Collapse
Affiliation(s)
- Eoin D O'Cearbhaill
- National Centre for Biomedical Engineering Science, National University of Ireland, Galway, Galway, Ireland
| | | | | | | | | | | |
Collapse
|
41
|
Albinsson S, Shakirova Y, Rippe A, Baumgarten M, Rosengren BI, Rippe C, Hallmann R, Hellstrand P, Rippe B, Swärd K. Arterial remodeling and plasma volume expansion in caveolin-1-deficient mice. Am J Physiol Regul Integr Comp Physiol 2007; 293:R1222-31. [PMID: 17626133 DOI: 10.1152/ajpregu.00092.2007] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Caveolin-1 (Cav-1) is essential for the morphology of membrane caveolae and exerts a negative influence on a number of signaling systems, including nitric oxide (NO) production and activity of the MAP kinase cascade. In the vascular system, ablation of caveolin-1 may thus be expected to cause arterial dilatation and increased vessel wall mass (remodeling). This was tested in Cav-1 knockout (KO) mice by a detailed morphometric and functional analysis of mesenteric resistance arteries, shown to lack caveolae. Quantitative morphometry revealed increased media thickness and media-to-lumen ratio in KO. Pressure-induced myogenic tone and flow-induced dilatation were decreased in KO arteries, but both were increased toward wild-type (WT) levels following NO synthase (NOS) inhibition. Isometric force recordings following NOS inhibition showed rightward shifts of passive and active length-force relationships in KO, and the force response to alpha(1)-adrenergic stimulation was increased. In contrast, media thickness and force response of the aorta were unaltered in KO vs. WT, whereas lumen diameter was increased. Mean arterial blood pressure during isoflurane anesthesia was not different in KO vs. WT, but greater fluctuation in blood pressure over time was noted. Following NOS inhibition, fluctuations disappeared and pressure increased twice as much in KO (38 +/- 6%) compared with WT (17 +/- 3%). Tracer-dilution experiments showed increased plasma volume in KO. We conclude that NO affects blood pressure more in Cav-1 KO than in WT mice and that restructuring of resistance vessels and an increased responsiveness to adrenergic stimulation compensate for a decreased tone in Cav-1 KO mice.
Collapse
Affiliation(s)
- Sebastian Albinsson
- Department of Experimental Medical Science, Lund University, SE-221 84 Lund, Sweden
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Rice KM, Desai DH, Preston DL, Wehner PS, Blough ER. Uniaxial stretch-induced regulation of mitogen-activated protein kinase, Akt and p70 S6 kinase in the ageing Fischer 344 x Brown Norway rat aorta. Exp Physiol 2007; 92:963-70. [PMID: 17526558 DOI: 10.1113/expphysiol.2007.037275] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The effects of ageing on the cardiovascular system contribute to substantial alterations in cellular morphology and function. The variables regulating these changes are unknown; however, one set of signalling molecules that may be of particular importance in mediating numerous cellular responses, including control of cell growth, differentiation and adaptation, are the proteins associated with the mitogen-activated protein kinase (MAPK) signalling systems. The MAPKs, in conjunction with the p70 S6k signalling cascade, have emerged as critical components for regulating numerous mechanotransduction-related cellular responses. Here we investigate the ability of uniaxial stretch to activate the MAPK and p70 S6k pathways in adult (6-month-old), aged (30-month-old) and very aged (36-month-old) Fischer 344/NNiaHSd x Brown Norway/BiNia (FBN) rats. Western blotting of the MAPK family proteins extracellular signal-regulated kinase (Erk) 1/2, p38- and c-Jun NH(2)-terminal kinase (Jnk)-MAPKs showed differential expression and activation between these proteins with age. An acute 15 min interval of 20% uniaxial stretch using an ex vivo aortic preparation demonstrated similar regulation of Erk1/2, p38- and Jnk-MAPK. However, ageing altered uniaxial induced p70 S6k pathway signalling. These observations confirm previous data demonstrating that MAPK proteins are mechanically regulated and also suggest that p70 S6k signalling expression and activation are controlled differently with ageing. Taken together, these data may help to explain, in part, the age-related changes in vascular morphology, function and response to injury.
Collapse
Affiliation(s)
- Kevin M Rice
- Department of Biological Sciences, Marshall University, Huntington, WV 25755-1090, USA
| | | | | | | | | |
Collapse
|
43
|
Halka AT, Turner NJ, Carter A, Ghosh J, Murphy MO, Kirton JP, Kielty CM, Walker MG. The effects of stretch on vascular smooth muscle cell phenotype in vitro. Cardiovasc Pathol 2007; 17:98-102. [PMID: 18329554 DOI: 10.1016/j.carpath.2007.03.001] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2007] [Revised: 03/06/2007] [Accepted: 03/13/2007] [Indexed: 12/16/2022] Open
Abstract
Vascular smooth muscle cells (VSMC) situated in the tunica media of veins and arteries are central to maintaining conduit integrity in the face of mechanical forces inherent within the cardiovascular system. The predominant mechanical force influencing VSMC structural organisation and signalling is cyclic stretch. VSMC phenotype is manipulated by externally applied stretch which regulates the activity of their contractile apparatus. Stretch modulates cell shape, cytoplasmic organisation, and intracellular processes leading to migration, proliferation, or contraction. Drug therapy directed at the components of the signalling pathways influenced by stretch may ultimately prevent cardiovascular pathology such as myointimal hyperplasia.
Collapse
Affiliation(s)
- Anastassi T Halka
- Department of Vascular Surgery, Manchester Royal Infirmary, Manchester, UK
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Jiang Z, Yu P, Tao M, Fernandez C, Ifantides C, Moloye O, Schultz GS, Ozaki CK, Berceli SA. TGF-beta- and CTGF-mediated fibroblast recruitment influences early outward vein graft remodeling. Am J Physiol Heart Circ Physiol 2007; 293:H482-8. [PMID: 17369455 DOI: 10.1152/ajpheart.01372.2006] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Luminal shearing forces have been shown to impact both geometric remodeling and the development of intimal hyperplasia. Less well studied is the influence of intramural wall stresses on vessel growth and adaptation. Using a vein graft-fistula configuration to isolate the impact of circumferential wall stress, we identify the reorganization of adventitial myofibroblasts as the dominant histological event that limits early outward remodeling of vein grafts in response to elevated wall stress. We hypothesize that increased production of transforming growth factor-beta (TGF-beta) and connective tissue growth factor (CTGF) induces recruitment of myofibroblasts, promotes adventitial reorganization, and limits early outward remodeling in response to increased intramural wall stress. Vein grafts with a distal arteriovenous fistula in the neck of rabbits were constructed, resulting in a fourfold differential in circumferential wall stress. Using this model, we demonstrate 1) elevated wall stress augments the production of TGF-beta and CTGF, 2) increased TGF-beta expression and CTGF expression are correlated with the enhanced differentiation from fibroblasts to myofibroblasts, as evidenced by the significant increase in the alpha-actin-positive cells in adventitia, and 3) the levels of TGF-beta, CTGF, and alpha-actin are inversely correlated with the magnitude of outward remodeling of the graft wall. Increased wall stress after vein graft implantation appears to induce a TGF-beta- and CTGF-mediated recruitment of adventitial fibroblasts and a conversion to a myofibroblast phenotype. Although important in the maintenance of wall stability in the face of an increased mechanical load, this adventitial adaptation limits early outward remodeling of the vein conduit and may prove deleterious in maintaining long-term vein graft patency.
Collapse
Affiliation(s)
- Zhihua Jiang
- University of Florida College of Medicine, Gainesville, FL 32610-0286, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Corteling RL, Brett SE, Yin H, Zheng XL, Walsh MP, Welsh DG. The functional consequence of RhoA knockdown by RNA interference in rat cerebral arteries. Am J Physiol Heart Circ Physiol 2007; 293:H440-7. [PMID: 17369454 DOI: 10.1152/ajpheart.01374.2006] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Uridine triphosphate (UTP) constricts cerebral arteries by activating transduction pathways that increase cytosolic [Ca(2+)] and myofilament Ca(2+) sensitivity. The signaling proteins that comprise these pathways remain uncertain with recent studies implicating a role for several G proteins. To start clarifying which G proteins enable UTP-induced vasoconstriction, a small interfering RNA (siRNA) approach was developed to knock down specified targets in rat cerebral arteries. siRNA directed against G(q) and RhoA was introduced into isolated cerebral arteries using reverse permeabilization. Following a defined period of organ culture, arteries were assayed for contractile function, mRNA levels, and protein expression. Targeted siRNA reduced RhoA or G(q) mRNA expression by 60-70%, which correlated with a reduction in RhoA but not G(q) protein expression. UTP-induced constriction was abolished in RhoA-depleted arteries, but this was not due to a reduction in myosin light chain phosphorylation. UTP-induced actin polymerization was attenuated in RhoA-depleted arteries, which would explain the loss of agonist-induced constriction. In summary, this study illustrates that siRNA approaches can be effectively used on intact arteries to induce targeted knockdown given that the protein turnover rate is sufficiently high. It also demonstrates that the principal role of RhoA in agonist-induced constriction is to facilitate the formation of F-actin, the physical structure to which phosphorylated myosin binds to elicit arterial constriction.
Collapse
Affiliation(s)
- Randolph L Corteling
- Department of Physiology and Biophysics, University of Calgary, 3330 Hospital Drive NW, Calgary, Alberta, Canada
| | | | | | | | | | | |
Collapse
|
46
|
Zargham R, Wamhoff BR, Thibault G. RNA interference targeting alpha8 integrin attenuates smooth muscle cell growth. FEBS Lett 2007; 581:939-43. [PMID: 17300786 DOI: 10.1016/j.febslet.2007.01.069] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2007] [Accepted: 01/26/2007] [Indexed: 01/10/2023]
Abstract
alpha8 integrin gene silencing has been shown to result in the stress fibre disassembly. Stress fibres are required for cell adhesion to promote passage through cell cycle. Thus, we hypothesized that alpha8 integrin gene silencing might affect vascular smooth muscle cell (VSMC) growth. Short interference RNA (siRNA) targeting alpha8 integrin in rat VSMCs resulted in reduced DNA synthesis. Moreover, siRNA-alpha8 integrin prevented thrombin-induced proliferation. RhoA plays a critical role in regulating VSMC growth. alpha8 integrin co-immunoprecipitated with RhoA and siRNA-alpha8 reduced membrane associated RhoA. Our data suggest that alpha8 integrin expression is critical for VSMC growth, which has potential implications in postangioplasty neointimal hyperplasia.
Collapse
Affiliation(s)
- Ramin Zargham
- Institut de Recherches Cliniques de Montréal, Université de Montréal and Experimental Medicine Department, McGill University, 110 Avenue Des Pins Ouest, Montréal, Que., Canada.
| | | | | |
Collapse
|
47
|
Li FH, Xia W, Li AW, Zhao CF, Sun RP. Inhibition of rho kinase attenuates high flow induced pulmonary hypertension in rats. Chin Med J (Engl) 2007; 120:22-29. [PMID: 17254483 DOI: 10.1097/00029330-200701010-00005] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND The RhoA/Rho kinase pathway may participate in the pathogenesis of hypoxia and monocrotaline induced pulmonary hypertension. This study tested whether RhoA/Rho kinase pathway is involved in the pathogenesis of high flow induced pulmonary hypertension in rats. METHODS Male Wistar rats (4 weeks) were randomly divided into 4 shunt groups, 4 treated groups and 4 control groups. Shunt and treated groups underwent left common carotid artery/external jugular vein shunt operation. Control groups underwent sham operation. Treated groups received fasudil treatment and the others received same dose of saline. At weeks 1, 2, 4 and 8 of the study, right ventricular systolic pressure was measured and blood gases were analysed to calculate Qp/Qs. The weight ratio of right ventricle to left ventricle plus septum and the mean percentage of medial wall thickness in moderate sized pulmonary arteries were obtained. RhoA activity in pulmonary arteries was detected using Rho activity assay reagent. Rho kinase activity was quantified by the extent of MYPT1 phosphorylation with Western blot. Proliferating cells were evaluated using proliferating cell nuclear antigen immunohistological staining. RESULTS Carotid artery/jugular vein shunt resulted in high pulmonary blood flow, both an acute and a chronic elevation of right ventricular systolic pressure, significant medial wall thickening characterized by smooth muscle cells proliferation, right ventricular hypertrophy and increased activation of RhoA and Rho kinase. Fasudil treatment lowered pulmonary artery systolic pressure, suppressed pulmonary artery smooth muscle cells proliferation, attenuated pulmonary artery medial wall thickening and inhibited right ventricular hypertrophy together with significant suppression of Rho kinase activity but not Rho activity. CONCLUSIONS Activated RhoA/Rho kinase pathway is associated with both the acute pulmonary vasoconstriction and the chronic pulmonary artery remodelling of high flow induced pulmonary hypertension. Fasudil treatment could improve pulmonary hypertension by inhibiting Rho kinase activity.
Collapse
Affiliation(s)
- Fu-Hai Li
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Shandong University Qilu Hospital, Jinan 250012, China
| | | | | | | | | |
Collapse
|