1
|
Deng W, Huang S, Yu L, Gao B, Pan Y, Wang X, Li L. HIF-1α knockdown attenuates phenotypic transformation and oxidative stress induced by high salt in human aortic vascular smooth muscle cells. Sci Rep 2024; 14:28100. [PMID: 39543255 PMCID: PMC11564746 DOI: 10.1038/s41598-024-79892-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 11/13/2024] [Indexed: 11/17/2024] Open
Abstract
Increased dietary salt intake is a well-established risk factor for hypertension and related cardiovascular diseases, involving complex vascular remodeling processes. However, the specific role of hypoxia-inducible factor-1α (HIF-1α) in vascular pathophysiology under high-salt conditions remains poorly understood. This study investigates the role of HIF-1α in high-salt-induced vascular remodeling using human aortic vascular smooth muscle cells (HA-VSMCs) cultured in vitro. HA-VSMCs were divided into three groups: high-salt with HIF-1α knockdown (shHIF-1α + HS), negative control (shcontrol), and high-salt (HS). Cell viability, migration, gene expression, and protein levels were evaluated. High-salt conditions significantly increased mRNA expression of α-smooth muscle actin (α-SMA), smooth muscle protein 22 (SM22), angiotensin II type 1 receptor (AT1R), collagen I, and collagen III (p < 0.0001). HIF-1α knockdown partially attenuated these increases, particularly for α-SMA, SM22, and AT1R (p < 0.01). At the protein level, high-salt exposure markedly elevated expression of collagen III, HIF-1α, osteopontin (OPN), and angiotensin II (Ang II) (p < 0.0001). HIF-1α knockdown significantly reduced the high-salt-induced increases in collagen III and HIF-1α protein levels (p < 0.001) but had a limited effect on OPN and Ang II upregulation. Interestingly, SM22 protein expression was significantly decreased under high-salt conditions (p < 0.0001), an effect partially reversed by HIF-1α knockdown (p < 0.0001). These findings demonstrate that high-salt conditions induce complex changes in gene and protein expression in HA-VSMCs, with HIF-1α playing a crucial role in mediating many of these alterations. The study highlights the differential effects of HIF-1α on various markers of vascular remodeling and suggests that HIF-1α may be a potential therapeutic target for mitigating salt-induced vascular pathology. Further research is warranted to elucidate the mechanisms underlying the HIF-1α-dependent and -independent effects observed in this study.
Collapse
MESH Headings
- Humans
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Hypoxia-Inducible Factor 1, alpha Subunit/genetics
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/cytology
- Oxidative Stress/drug effects
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/drug effects
- Aorta/metabolism
- Aorta/cytology
- Gene Knockdown Techniques
- Sodium Chloride, Dietary/adverse effects
- Actins/metabolism
- Phenotype
- Cell Movement/drug effects
- Muscle Proteins/metabolism
- Muscle Proteins/genetics
- Microfilament Proteins/metabolism
- Microfilament Proteins/genetics
- Receptor, Angiotensin, Type 1/metabolism
- Receptor, Angiotensin, Type 1/genetics
- Vascular Remodeling/drug effects
- Cell Survival/drug effects
- Osteopontin/metabolism
- Osteopontin/genetics
- Cells, Cultured
Collapse
Affiliation(s)
- Wenbin Deng
- Department of Geriatrics, The First Affiliated Hospital of Dali University, Dali, 671000, Yunnan, China
| | - Shiqiong Huang
- Department of Geriatrics, The First Affiliated Hospital of Dali University, Dali, 671000, Yunnan, China
| | - Lisha Yu
- Department of Geriatrics, The First Affiliated Hospital of Dali University, Dali, 671000, Yunnan, China
| | - Bo Gao
- Department of Pathology, The First Affiliated Hospital of Dali University, Dali, 671000, Yunnan, China
| | - Yun Pan
- Department of Pathology, The First Affiliated Hospital of Dali University, Dali, 671000, Yunnan, China
| | - Xue Wang
- Department of Geriatrics, The First Affiliated Hospital of Dali University, Dali, 671000, Yunnan, China
| | - Lihua Li
- Department of Geriatrics, The First Affiliated Hospital of Dali University, Dali, 671000, Yunnan, China.
- Department of Gerontology, The First Affiliated Hospital of Dali University, Jiashibo Road 32, Dali, 671000, China.
| |
Collapse
|
2
|
Yu Y, Cai Y, Yang F, Yang Y, Cui Z, Shi D, Bai R. Vascular smooth muscle cell phenotypic switching in atherosclerosis. Heliyon 2024; 10:e37727. [PMID: 39309965 PMCID: PMC11416558 DOI: 10.1016/j.heliyon.2024.e37727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 08/25/2024] [Accepted: 09/09/2024] [Indexed: 09/25/2024] Open
Abstract
Atherosclerosis (AS) is a complex pathology process involving intricate interactions among various cells and biological processes. Vascular smooth muscle cells (VSMCs) are the predominant cell type in normal arteries, and under atherosclerotic stimuli, VSMCs respond to altered blood flow and microenvironment changes by downregulating contractile markers and switching their phenotype. This review overviews the diverse phenotypes of VSMCs, including the canonical contractile VSMCs, synthetic VSMCs, and phenotypes resembling macrophages, foam cells, myofibroblasts, osteoblasts/chondrocytes, and mesenchymal stem cells. We summarize their presumed protective and pro-atherosclerotic roles in AS development. Additionally, we underscore the molecular mechanisms and regulatory pathways governing VSMC phenotypic switching, encompassing transcriptional regulation, biochemical factors, plaque microenvironment, epigenetics, miRNAs, and the cytoskeleton, emphasizing their significance in AS development. Finally, we outline probable future research directions targeting VSMCs, offering insights into potential therapeutic strategies for AS management.
Collapse
Affiliation(s)
- Yanqiao Yu
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
- National Clinical Research Center for Chinese Medicine Cardiology, Beijing, 100091, China
- Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yajie Cai
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
- National Clinical Research Center for Chinese Medicine Cardiology, Beijing, 100091, China
| | - Furong Yang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
- National Clinical Research Center for Chinese Medicine Cardiology, Beijing, 100091, China
| | - Yankai Yang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
- National Clinical Research Center for Chinese Medicine Cardiology, Beijing, 100091, China
- Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Zhuorui Cui
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
- National Clinical Research Center for Chinese Medicine Cardiology, Beijing, 100091, China
- Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Dazhuo Shi
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
- National Clinical Research Center for Chinese Medicine Cardiology, Beijing, 100091, China
| | - Ruina Bai
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
- National Clinical Research Center for Chinese Medicine Cardiology, Beijing, 100091, China
| |
Collapse
|
3
|
Jia K, Luo X, Yi J, Zhang C. Hormonal influence: unraveling the impact of sex hormones on vascular smooth muscle cells. Biol Res 2024; 57:61. [PMID: 39227995 PMCID: PMC11373308 DOI: 10.1186/s40659-024-00542-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: 04/30/2024] [Accepted: 08/26/2024] [Indexed: 09/05/2024] Open
Abstract
Sex hormones play a pivotal role as endocrine hormones that exert profound effects on the biological characteristics and vascular function of vascular smooth muscle cells (VSMCs). By modulating intracellular signaling pathways, activating nuclear receptors, and regulating gene expression, sex hormones intricately influence the morphology, function, and physiological state of VSMCs, thereby impacting the biological properties of vascular contraction, relaxation, and growth. Increasing evidence suggests that abnormal phenotypic changes in VSMCs contribute to the initiation of vascular diseases, including atherosclerosis. Therefore, understanding the factors governing phenotypic alterations in VSMCs and elucidating the underlying mechanisms can provide crucial insights for refining interventions targeted at vascular diseases. Additionally, the varying levels of different types of sex hormones in the human body, influenced by sex and age, may also affect the phenotypic conversion of VSMCs. This review aims to explore the influence of sex hormones on the phenotypic switching of VSMCs and the development of associated vascular diseases in the human body.
Collapse
Affiliation(s)
- Keran Jia
- Department of Medical Cell Biology and Genetics, School of Basic Medical Sciences, Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Xin Luo
- Department of Medical Cell Biology and Genetics, School of Basic Medical Sciences, Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Southwest Medical University, Luzhou, Sichuan, 646000, China
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Jingyan Yi
- Department of Medical Cell Biology and Genetics, School of Basic Medical Sciences, Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Southwest Medical University, Luzhou, Sichuan, 646000, China.
| | - Chunxiang Zhang
- Department of Cardiology, The Affiliated Hospital, Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Southwest Medical University, Luzhou, Sichuan, 646000, China.
| |
Collapse
|
4
|
Wang B, Cui K, Zhu B, Dong Y, Wang D, Singh B, Wu H, Li K, Eisa-Beygi S, Sun Y, Wong S, Cowan DB, Chen Y, Du M, Chen H. Epsins oversee smooth muscle cell reprograming by influencing master regulators KLF4 and OCT4. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.08.574714. [PMID: 39131381 PMCID: PMC11312448 DOI: 10.1101/2024.01.08.574714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
Smooth muscle cells in major arteries play a crucial role in regulating coronary artery disease. Conversion of smooth muscle cells into other adverse cell types in the artery propels the pathogenesis of the disease. Curtailing artery plaque buildup by modulating smooth muscle cell reprograming presents us a new opportunity to thwart coronary artery disease. Here, our report how Epsins, a family of endocytic adaptor proteins oversee the smooth muscle cell reprograming by influencing master regulators OCT4 and KLF4. Using single-cell RNA sequencing, we characterized the phenotype of modulated smooth muscle cells in mouse atherosclerotic plaque and found that smooth muscle cells lacking epsins undergo profound reprogramming into not only beneficial myofibroblasts but also endothelial cells for injury repair of diseased endothelium. Our work lays concrete groundwork to explore an uncharted territory as we show that depleting Epsins bolsters smooth muscle cells reprograming to endothelial cells by augmenting OCT4 activity but restrain them from reprograming to harmful foam cells by destabilizing KLF4, a master regulator of adverse reprograming of smooth muscle cells. Moreover, the expression of Epsins in smooth muscle cells positively correlates with the severity of both human and mouse coronary artery disease. Integrating our scRNA-seq data with human Genome-Wide Association Studies (GWAS) identifies pivotal roles Epsins play in smooth muscle cells in the pathological process leading to coronary artery disease. Our findings reveal a previously unexplored direction for smooth muscle cell phenotypic modulation in the development and progression of coronary artery disease and unveil Epsins and their downstream new targets as promising novel therapeutic targets for mitigating metabolic disorders.
Collapse
Affiliation(s)
- Beibei Wang
- Vascular Biology Program, Boston Children’s Hospital and Department of Surgery, Harvard Medical School, Boston, MA, 02115, USA
| | - Kui Cui
- Vascular Biology Program, Boston Children’s Hospital and Department of Surgery, Harvard Medical School, Boston, MA, 02115, USA
| | - Bo Zhu
- Vascular Biology Program, Boston Children’s Hospital and Department of Surgery, Harvard Medical School, Boston, MA, 02115, USA
| | - Yunzhou Dong
- Vascular Biology Program, Boston Children’s Hospital and Department of Surgery, Harvard Medical School, Boston, MA, 02115, USA
| | - Donghai Wang
- Vascular Biology Program, Boston Children’s Hospital and Department of Surgery, Harvard Medical School, Boston, MA, 02115, USA
| | - Bandana Singh
- Vascular Biology Program, Boston Children’s Hospital and Department of Surgery, Harvard Medical School, Boston, MA, 02115, USA
| | - Hao Wu
- Vascular Biology Program, Boston Children’s Hospital and Department of Surgery, Harvard Medical School, Boston, MA, 02115, USA
| | - Kathryn Li
- Vascular Biology Program, Boston Children’s Hospital and Department of Surgery, Harvard Medical School, Boston, MA, 02115, USA
| | - Shahram Eisa-Beygi
- Vascular Biology Program, Boston Children’s Hospital and Department of Surgery, Harvard Medical School, Boston, MA, 02115, USA
| | - Yong Sun
- Department of Pathology, Birmingham, AL 35294, USA; University of Alabama at Birmingham, and the Birmingham Veterans Affairs Medical Center, Birmingham, AL 35294, USA
| | - Scott Wong
- Vascular Biology Program, Boston Children’s Hospital and Department of Surgery, Harvard Medical School, Boston, MA, 02115, USA
| | - Douglas B. Cowan
- Vascular Biology Program, Boston Children’s Hospital and Department of Surgery, Harvard Medical School, Boston, MA, 02115, USA
| | - Yabing Chen
- Department of Pathology, Birmingham, AL 35294, USA; University of Alabama at Birmingham, and the Birmingham Veterans Affairs Medical Center, Birmingham, AL 35294, USA
| | - Mulong Du
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, 655 Huntington Avenue, Boston, MA, 02115, USA
| | - Hong Chen
- Vascular Biology Program, Boston Children’s Hospital and Department of Surgery, Harvard Medical School, Boston, MA, 02115, USA
| |
Collapse
|
5
|
Sun R, Pan X, Ward E, Intrevado R, Morozan A, Lauzon AM, Martin JG. Serum Response Factor Expression in Excess Permits a Dual Contractile-Proliferative Phenotype of Airway Smooth Muscle. Am J Respir Cell Mol Biol 2024; 71:182-194. [PMID: 38775474 DOI: 10.1165/rcmb.2024-0081oc] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 04/18/2024] [Indexed: 08/02/2024] Open
Abstract
The transcription factors (TFs) MyoCD (myocardin) and Elk-1 (ETS Like-1 protein) competitively bind to SRF (serum response factor) and control myogenic- and mitogenic-related gene expression in smooth muscle, respectively. Their functions are therefore mutually inhibitory, which results in a contractile-versus-proliferative phenotype dichotomy. Airway smooth muscle cell (ASMC) phenotype alterations occur in various inflammatory airway diseases, promoting pathological remodeling and contributing to airflow obstruction. We characterized MyoCD and Elk-1 interactions and their roles in phenotype determination in human ASMCs. MyoCD overexpression in ASMCs increased smooth muscle gene expression, force generation, and partially restored the loss of smooth muscle protein associated with prolonged culturing while inhibiting Elk-1 transcriptional activities and proliferation induced by EGF (epidermal growth factor). However, MyoCD overexpression failed to suppress these responses induced by FBS, as FBS also upregulated SRF expression to a degree that allowed unopposed function of both TFs. Inhibition of the RhoA pathway reversed said SRF changes, allowing inhibition of Elk-1 by MyoCD overexpression and suppressing FBS-mediated contractile protein gene upregulation. Our study confirmed that MyoCD in increased abundance can competitively inhibit Elk-1 function. However, SRF upregulation permits a dual contractile-proliferative ASMC phenotype that is anticipated to exacerbate pathological alterations, whereas therapies targeting SRF may inhibit pathological ASMC proliferation and contractile protein gene expression.
Collapse
Affiliation(s)
- Rui Sun
- Meakins-Christie Laboratories, The Research Institute of McGill University Health Centre, Montréal, Québec, Canada
| | - Xingning Pan
- Meakins-Christie Laboratories, The Research Institute of McGill University Health Centre, Montréal, Québec, Canada
| | - Erin Ward
- Meakins-Christie Laboratories, The Research Institute of McGill University Health Centre, Montréal, Québec, Canada
| | - Rafael Intrevado
- Meakins-Christie Laboratories, The Research Institute of McGill University Health Centre, Montréal, Québec, Canada
| | - Arina Morozan
- Meakins-Christie Laboratories, The Research Institute of McGill University Health Centre, Montréal, Québec, Canada
| | - Anne-Marie Lauzon
- Meakins-Christie Laboratories, The Research Institute of McGill University Health Centre, Montréal, Québec, Canada
| | - James G Martin
- Meakins-Christie Laboratories, The Research Institute of McGill University Health Centre, Montréal, Québec, Canada
| |
Collapse
|
6
|
Sarkar A, Pawar SV, Chopra K, Jain M. Gamut of glycolytic enzymes in vascular smooth muscle cell proliferation: Implications for vascular proliferative diseases. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167021. [PMID: 38216067 DOI: 10.1016/j.bbadis.2024.167021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 01/05/2024] [Accepted: 01/05/2024] [Indexed: 01/14/2024]
Abstract
Vascular smooth muscle cells (VSMCs) are the predominant cell type in the media of the blood vessels and are responsible for maintaining vascular tone. Emerging evidence confirms that VSMCs possess high plasticity. During vascular injury, VSMCs switch from a "contractile" phenotype to an extremely proliferative "synthetic" phenotype. The balance between both strongly affects the progression of vascular remodeling in many cardiovascular pathologies such as restenosis, atherosclerosis and aortic aneurism. Proliferating cells demand high energy requirements and to meet this necessity, alteration in cellular bioenergetics seems to be essential. Glycolysis, fatty acid metabolism, and amino acid metabolism act as a fuel for VSMC proliferation. Metabolic reprogramming of VSMCs is dynamically variable that involves multiple mechanisms and encompasses the coordination of various signaling molecules, proteins, and enzymes. Here, we systemically reviewed the metabolic changes together with the possible treatments that are still under investigation underlying VSMC plasticity which provides a promising direction for the treatment of diseases associated with VSMC proliferation. A better understanding of the interaction between metabolism with associated signaling may uncover additional targets for better therapeutic strategies in vascular disorders.
Collapse
Affiliation(s)
- Ankan Sarkar
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
| | - Sandip V Pawar
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
| | - Kanwaljit Chopra
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
| | - Manish Jain
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India.
| |
Collapse
|
7
|
Choo YY, Sakai T, Ikebe R, Jeffers A, Idell S, Tucker TA, Ikebe M. Role of ZIP kinase in development of myofibroblast differentiation from HPMCs. Am J Physiol Lung Cell Mol Physiol 2024; 326:L353-L366. [PMID: 38252666 PMCID: PMC11281797 DOI: 10.1152/ajplung.00251.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 12/14/2023] [Accepted: 01/15/2024] [Indexed: 01/24/2024] Open
Abstract
During the development of pleural fibrosis, pleural mesothelial cells (PMCs) undergo phenotypic switching from differentiated mesothelial cells to mesenchymal cells (MesoMT). Here, we investigated how external stimuli such as TGF-β induce HPMC-derived myofibroblast differentiation to facilitate the development of pleural fibrosis. TGF-β significantly increased di-phosphorylation but not mono-phosphorylation of myosin II regulatory light chain (RLC) in HPMCs. An increase in RLC di-phosphorylation was also found at the pleural layer of our carbon black bleomycin (CBB) pleural fibrosis mouse model, where it showed filamentous localization that coincided with alpha smooth muscle actin (αSMA) in the cells in the pleura. Among the protein kinases that can phosphorylate myosin II RLC, ZIPK (zipper-interacting kinase) protein expression was significantly augmented after TGF-β stimulation. Furthermore, ZIPK gene silencing attenuated RLC di-phosphorylation, suggesting that ZIPK is responsible for di-phosphorylation of myosin II in HPMCs. Although TGF-β significantly increased the expression of ZIP kinase protein, the change in ZIP kinase mRNA was marginal, suggesting a posttranscriptional mechanism for the regulation of ZIP kinase expression by TGF-β. ZIPK gene knockdown (KD) also significantly reduced TGF-β-induced upregulation of αSMA expression. This finding suggests that siZIPK attenuates myofibroblast differentiation of HPMCs. siZIPK diminished TGF-β-induced contractility of HPMCs consistent with siZIPK-induced decrease in the di-phosphorylation of myosin II RLC. The present results implicate ZIPK in the regulation of the contractility of HPMC-derived myofibroblasts, phenotype switching, and myofibroblast differentiation of HPMCs.NEW & NOTEWORTHY Here, we highlight that ZIP kinase is responsible for di-phosphorylation of myosin light chain, which facilitates stress fiber formation and actomyosin-based cell contraction during mesothelial to mesenchymal transition in human pleural mesothelial cells. This transition has a significant impact on tissue remodeling and subsequent stiffness of the pleura. This study provides insight into a new therapeutic strategy for the treatment of pleural fibrosis.
Collapse
Affiliation(s)
- Young-Yeon Choo
- Department of Cellular and Molecular Biology, The University of Texas at Tyler Health Science Center, Tyler, Texas, United States
| | - Tsuyoshi Sakai
- Department of Cellular and Molecular Biology, The University of Texas at Tyler Health Science Center, Tyler, Texas, United States
| | - Reiko Ikebe
- Department of Cellular and Molecular Biology, The University of Texas at Tyler Health Science Center, Tyler, Texas, United States
| | - Ann Jeffers
- Department of Cellular and Molecular Biology, The University of Texas at Tyler Health Science Center, Tyler, Texas, United States
| | - Steven Idell
- Department of Cellular and Molecular Biology, The University of Texas at Tyler Health Science Center, Tyler, Texas, United States
| | - Torry A Tucker
- Department of Cellular and Molecular Biology, The University of Texas at Tyler Health Science Center, Tyler, Texas, United States
| | - Mitsuo Ikebe
- Department of Cellular and Molecular Biology, The University of Texas at Tyler Health Science Center, Tyler, Texas, United States
| |
Collapse
|
8
|
Wong D, Auguste G, Cardenas CLL, Turner AW, Chen Y, Song Y, Ma L, Perry RN, Aherrahrou R, Kuppusamy M, Yang C, Mosquera JV, Dube CJ, Khan MD, Palmore M, Kalra JK, Kavousi M, Peyser PA, Matic L, Hedin U, Manichaikul A, Sonkusare SK, Civelek M, Kovacic JC, Björkegren JL, Malhotra R, Miller CL. FHL5 Controls Vascular Disease-Associated Gene Programs in Smooth Muscle Cells. Circ Res 2023; 132:1144-1161. [PMID: 37017084 PMCID: PMC10147587 DOI: 10.1161/circresaha.122.321692] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 03/21/2023] [Indexed: 04/06/2023]
Abstract
BACKGROUND Genome-wide association studies have identified hundreds of loci associated with common vascular diseases, such as coronary artery disease, myocardial infarction, and hypertension. However, the lack of mechanistic insights for many GWAS loci limits their translation into the clinic. Among these loci with unknown functions is UFL1-four-and-a-half LIM (LIN-11, Isl-1, MEC-3) domain 5 (FHL5; chr6q16.1), which reached genome-wide significance in a recent coronary artery disease/ myocardial infarction GWAS meta-analysis. UFL1-FHL5 is also associated with several vascular diseases, consistent with the widespread pleiotropy observed for GWAS loci. METHODS We apply a multimodal approach leveraging statistical fine-mapping, epigenomic profiling, and ex vivo analysis of human coronary artery tissues to implicate FHL5 as the top candidate causal gene. We unravel the molecular mechanisms of the cross-phenotype genetic associations through in vitro functional analyses and epigenomic profiling experiments in coronary artery smooth muscle cells. RESULTS We prioritized FHL5 as the top candidate causal gene at the UFL1-FHL5 locus through expression quantitative trait locus colocalization methods. FHL5 gene expression was enriched in the smooth muscle cells and pericyte population in human artery tissues with coexpression network analyses supporting a functional role in regulating smooth muscle cell contraction. Unexpectedly, under procalcifying conditions, FHL5 overexpression promoted vascular calcification and dysregulated processes related to extracellular matrix organization and calcium handling. Lastly, by mapping FHL5 binding sites and inferring FHL5 target gene function using artery tissue gene regulatory network analyses, we highlight regulatory interactions between FHL5 and downstream coronary artery disease/myocardial infarction loci, such as FOXL1 and FN1 that have roles in vascular remodeling. CONCLUSIONS Taken together, these studies provide mechanistic insights into the pleiotropic genetic associations of UFL1-FHL5. We show that FHL5 mediates vascular disease risk through transcriptional regulation of downstream vascular remodeling gene programs. These transacting mechanisms may explain a portion of the heritable risk for complex vascular diseases.
Collapse
Affiliation(s)
- Doris Wong
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, Virginia, USA
- Center for Public Health Genomics, University of Virginia, Charlottesville, Virginia, USA
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia, USA
| | - Gaëlle Auguste
- Center for Public Health Genomics, University of Virginia, Charlottesville, Virginia, USA
| | - Christian L. Lino Cardenas
- Cardiovascular Research Center, Cardiology Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Adam W. Turner
- Center for Public Health Genomics, University of Virginia, Charlottesville, Virginia, USA
| | - Yixuan Chen
- Center for Public Health Genomics, University of Virginia, Charlottesville, Virginia, USA
| | - Yipei Song
- Center for Public Health Genomics, University of Virginia, Charlottesville, Virginia, USA
| | - Lijiang Ma
- Department of Genetics and Genomic Sciences, Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, USA
| | - R. Noah Perry
- Center for Public Health Genomics, University of Virginia, Charlottesville, Virginia, USA
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia, USA
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA
| | - Redouane Aherrahrou
- Center for Public Health Genomics, University of Virginia, Charlottesville, Virginia, USA
| | - Maniselvan Kuppusamy
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia, USA
| | - Chaojie Yang
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, Virginia, USA
- Center for Public Health Genomics, University of Virginia, Charlottesville, Virginia, USA
| | - Jose Verdezoto Mosquera
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, Virginia, USA
- Center for Public Health Genomics, University of Virginia, Charlottesville, Virginia, USA
| | - Collin J. Dube
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia, USA
| | - Mohammad Daud Khan
- Center for Public Health Genomics, University of Virginia, Charlottesville, Virginia, USA
| | - Meredith Palmore
- Center for Public Health Genomics, University of Virginia, Charlottesville, Virginia, USA
| | - Jaspreet K. Kalra
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia, USA
| | - Maryam Kavousi
- Department of Epidemiology, Erasmus University Medical Center, The Netherlands
| | | | - Ljubica Matic
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Ulf Hedin
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Ani Manichaikul
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, Virginia, USA
- Center for Public Health Genomics, University of Virginia, Charlottesville, Virginia, USA
- Department of Public Health Sciences, University of Virginia, Charlottesville, Virginia, USA
| | - Swapnil K. Sonkusare
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia, USA
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia, USA
| | - Mete Civelek
- Center for Public Health Genomics, University of Virginia, Charlottesville, Virginia, USA
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia, USA
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA
| | - Jason C. Kovacic
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, USA
- Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia
- St. Vincent’s Clinical School, University of New South Wales, Sydney, Australia
| | - Johan L.M. Björkegren
- Department of Genetics and Genomic Sciences, Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, USA
- Integrated Cardio Metabolic Centre, Department of Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Rajeev Malhotra
- Cardiovascular Research Center, Cardiology Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Clint L. Miller
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, Virginia, USA
- Center for Public Health Genomics, University of Virginia, Charlottesville, Virginia, USA
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia, USA
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
9
|
Zhou X, Luo C, Fan J, Gao G, Wang T, Zhang H, Wei A. Myocardin Reverses Hypoxia-Inducible Factor-1α Mediated Phenotypic Modulation of Corpus Cavernosum Smooth Muscle Cells in Hypoxia Induced by Cobalt Chloride. World J Mens Health 2023; 41:363-372. [PMID: 35274501 PMCID: PMC10042661 DOI: 10.5534/wjmh.210226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 12/21/2021] [Accepted: 12/30/2021] [Indexed: 11/15/2022] Open
Abstract
PURPOSE We aimed to investigate the mechanism of phenotypic transformation of corporal cavernosum smooth muscle cells (CCSMCs) under hypoxic conditions in vitro. MATERIALS AND METHODS In this study, a hypoxia model was established using cobalt chloride (CoCl2). CCSMCs were treated with different concentrations of CoCl2 for varying time periods, and cell viability was assessed. Hypoxia-inducible factor-1α (HIF-1α), myocardin (Myocd) and phenotypic markers were detected in the CCSMCs. We also transfected the CCSMCs with si-HIF-1α and Ad-Myocd and evaluated the effects on phenotypic modulation of CCSMCs and the relationship between HIF-1α and Myocd was evaluated. RESULTS CoCl2 inhibited the viability of CCSMCs in a dose- and time-dependent manner, and treatment with 300 µM CoCl2 for 48 hours were the optimal conditions for establishing the hypoxia model. The results showed increased expression levels of HIF-1α and osteopontin and decreased Myocd, alpha-smooth muscle actin, and calponin levels in CCSMCs under hypoxia. HIF-1α knockdown reversed hypoxia-induced phenotypic transformation with elevated Myocd expression. Overexpression of Myocd also reversed the effect of hypoxia on the phenotypic switch, but did not affect HIF-1α expression. CONCLUSIONS Our findings showed that HIF-1α was involved in the effect of hypoxia induced by CoCl2 on CCSMC phenotypic modulation, and Myocd overexpression could inhibit this process. Thus, Myocd might be a potential therapeutic target for erectile dysfunction under hypoxia or HIF-1α activation.
Collapse
Affiliation(s)
- Xiongcai Zhou
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Department of Urology, Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Chao Luo
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Junhong Fan
- Department of Urology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Guangqian Gao
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Tao Wang
- Department of Andrology, The Third Affiliated Hospital of Guangdong Medical University, Foshan, Guangdong, China.
| | - Haibo Zhang
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
| | - Anyang Wei
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
10
|
Yu J, Wang W, Yang J, Zhang Y, Gong X, Luo H, Cao N, Xu Z, Tian M, Yang P, Mei Q, Chen Z, Li Z, Li C, Duan X, Lyu QR, Gao C, Zhang B, Wang Y, Wu G, Zeng C. LncRNA PSR Regulates Vascular Remodeling Through Encoding a Novel Protein Arteridin. Circ Res 2022; 131:768-787. [PMID: 36134578 PMCID: PMC9588624 DOI: 10.1161/circresaha.122.321080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 09/06/2022] [Indexed: 01/26/2023]
Abstract
RATIONALE Vascular smooth muscle cells (VSMCs) phenotype switch from contractile to proliferative phenotype is a pathological hallmark in various cardiovascular diseases. Recently, a subset of long noncoding RNAs was identified to produce functional polypeptides. However, the functional impact and regulatory mechanisms of long noncoding RNAs in VSMCs phenotype switching remain to be fully elucidated. OBJECTIVES To illustrate the biological function and mechanism of a VSMC-enriched long noncoding RNA and its encoded peptide in VSMC phenotype switching and vascular remodeling. RESULTS We identified a VSMC-enriched transcript encoded by a previously uncharacterized gene, which we called phenotype switching regulator (PSR), which was markedly upregulated during vascular remodeling. Although PSR was annotated as a long noncoding RNA, we demonstrated that the lncPSR (PSR transcript) also encoded a protein, which we named arteridin. In VSMCs, both arteridin and lncPSR were necessary and sufficient to induce phenotype switching. Mechanistically, arteridin and lncPSR regulate downstream genes by directly interacting with a transcription factor YBX1 (Y-box binding protein 1) and modulating its nuclear translocation and chromatin targeting. Intriguingly, the PSR transcription was also robustly induced by arteridin. More importantly, the loss of PSR gene or arteridin protein significantly attenuated the vascular remodeling induced by carotid arterial injury. In addition, VSMC-specific inhibition of lncPSR using adeno-associated virus attenuated Ang II (angiotensin II)-induced hypertensive vascular remodeling. CONCLUSIONS PSR is a VSMC-enriched gene, and its transcript IncPSR and encoded protein (arteridin) coordinately regulate transcriptional reprogramming through a shared interacting partner, YBX1. This is a previously uncharacterized regulatory circuit in VSMC phenotype switching during vascular remodeling, with lncPSR/arteridin as potential therapeutic targets for the treatment of VSMC phenotype switching-related vascular remodeling.
Collapse
Affiliation(s)
- Junyi Yu
- Department of Cardiology, Daping Hospital, The Third Military Medical University (Army Medical University), Chongqing, P.R. China
- Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, P. R. China
| | - Wei Wang
- Department of Cardiology, Daping Hospital, The Third Military Medical University (Army Medical University), Chongqing, P.R. China
- Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, P. R. China
| | - Jining Yang
- Research Center for Nutrition and Food Safety, Chongqing Key Laboratory of Nutrition and Food Safety, Institute of Military Preventive Medicine, The Third Military Medical University, Chongqing, P.R. China
| | - Ye Zhang
- Department of Cardiology, Daping Hospital, The Third Military Medical University (Army Medical University), Chongqing, P.R. China
- Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, P. R. China
| | - Xue Gong
- Department of Cardiology, Daping Hospital, The Third Military Medical University (Army Medical University), Chongqing, P.R. China
- Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, P. R. China
| | - Hao Luo
- Department of Cardiology, Daping Hospital, The Third Military Medical University (Army Medical University), Chongqing, P.R. China
- Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, P. R. China
| | - Nian Cao
- Department of Cardiology, Daping Hospital, The Third Military Medical University (Army Medical University), Chongqing, P.R. China
- Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, P. R. China
| | - Zaicheng Xu
- Department of Cardiology, Daping Hospital, The Third Military Medical University (Army Medical University), Chongqing, P.R. China
- Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, P. R. China
| | - Miao Tian
- Department of Cardiology, Daping Hospital, The Third Military Medical University (Army Medical University), Chongqing, P.R. China
- Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, P. R. China
| | - Peili Yang
- Department of Cardiology, Daping Hospital, The Third Military Medical University (Army Medical University), Chongqing, P.R. China
- Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, P. R. China
| | - Qiao Mei
- Department of Cardiology, Daping Hospital, The Third Military Medical University (Army Medical University), Chongqing, P.R. China
- Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, P. R. China
| | - Zhi Chen
- Department of Cardiology, Daping Hospital, The Third Military Medical University (Army Medical University), Chongqing, P.R. China
- Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, P. R. China
| | - Zhuxin Li
- Department of Cardiology, Daping Hospital, The Third Military Medical University (Army Medical University), Chongqing, P.R. China
- Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, P. R. China
| | - Chuanwei Li
- Department of Cardiology, Daping Hospital, The Third Military Medical University (Army Medical University), Chongqing, P.R. China
- Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, P. R. China
| | - Xudong Duan
- Cardiovascular Research Center of Chongqing College, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Chongqing, P. R. China
| | - Qing Rex Lyu
- Cardiovascular Research Center of Chongqing College, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Chongqing, P. R. China
| | - Chen Gao
- Department of Pharmacology & Systems Physiology, University of Cincinnati College of Medicine, OH, USA
| | - Bing Zhang
- Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yibin Wang
- Signature Program in Cardiovascular and Metabolic Diseases, Duke-NUS School of Medicine, Singapore
| | - Gengze Wu
- Department of Cardiology, Daping Hospital, The Third Military Medical University (Army Medical University), Chongqing, P.R. China
- Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, P. R. China
| | - Chunyu Zeng
- Department of Cardiology, Daping Hospital, The Third Military Medical University (Army Medical University), Chongqing, P.R. China
- Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, P. R. China
- Cardiovascular Research Center of Chongqing College, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Chongqing, P. R. China
- State Key Laboratory of Trauma, Burns and Combined Injury, Daping Hospital, The Third Military Medical University, Chongqing, P.R. China
- Heart Center of Fujian Province, Union Hospital, Fujian Medical University, Fuzhou, P.R. China
- Department of Cardiology, Chongqing General Hospital, Chongqing, P. R. China
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, P.R. China
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing
| |
Collapse
|
11
|
Khachigian LM, Black BL, Ferdinandy P, De Caterina R, Madonna R, Geng YJ. Transcriptional regulation of vascular smooth muscle cell proliferation, differentiation and senescence: Novel targets for therapy. Vascul Pharmacol 2022; 146:107091. [PMID: 35896140 DOI: 10.1016/j.vph.2022.107091] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 07/21/2022] [Accepted: 07/21/2022] [Indexed: 10/16/2022]
Abstract
Vascular smooth muscle cells (SMC) possess a unique cytoplasticity, regulated by transcriptional, translational and phenotypic transformation in response to a diverse range of extrinsic and intrinsic pathogenic factors. The mature, differentiated SMC phenotype is physiologically typified transcriptionally by expression of genes encoding "contractile" proteins, such as SMα-actin (ACTA2), SM-MHC (myosin-11) and SM22α (transgelin). When exposed to various pathological conditions (e.g., pro-atherogenic risk factors, hypertension), SMC undergo phenotypic modulation, a bioprocess enabling SMC to de-differentiate in immature stages or trans-differentiate into other cell phenotypes. As recent studies suggest, the process of SMC phenotypic transformation involves five distinct states characterized by different patterns of cell growth, differentiation, migration, matrix protein expression and declined contractility. These changes are mediated via the action of several transcriptional regulators, including myocardin and serum response factor. Conversely, other factors, including Kruppel-like factor 4 and nuclear factor-κB, can inhibit SMC differentiation and growth arrest, while factors such as yin yang-1, can promote SMC differentiation whilst inhibiting proliferation. This article reviews recent advances in our understanding of regulatory mechanisms governing SMC phenotypic modulation. We propose the concept that transcription factors mediating this switching are important biomarkers and potential pharmacological targets for therapeutic intervention in cardiovascular disease.
Collapse
Affiliation(s)
- Levon M Khachigian
- Vascular Biology and Translational Research, Department of Pathology, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW 2052, Australia.
| | - Brian L Black
- Cardiovascular Research Institute, University of California, San Francisco, CA, United States of America
| | - Péter Ferdinandy
- Cardiovascular and Metabolic Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1089 Budapest, Hungary; Pharmahungary Group, 6722 Szeged, Hungary
| | - Raffaele De Caterina
- Cardiovascular Division, Pisa University Hospital & University of Pisa, Via Paradisa, 2, Pisa 56124, Italy
| | - Rosalinda Madonna
- Cardiovascular Division, Pisa University Hospital & University of Pisa, Via Paradisa, 2, Pisa 56124, Italy; Division of Cardiovascular Medicine, Department of Internal Medicine, The Center for Cardiovascular Biology and Atherosclerosis Research, McGovern School of Medicine, University of Texas Health Science Center at Houston, Houston, TX, United States of America
| | - Yong-Jian Geng
- Division of Cardiovascular Medicine, Department of Internal Medicine, The Center for Cardiovascular Biology and Atherosclerosis Research, McGovern School of Medicine, University of Texas Health Science Center at Houston, Houston, TX, United States of America
| |
Collapse
|
12
|
Winkler EA, Kim CN, Ross JM, Garcia JH, Gil E, Oh I, Chen LQ, Wu D, Catapano JS, Raygor K, Narsinh K, Kim H, Weinsheimer S, Cooke DL, Walcott BP, Lawton MT, Gupta N, Zlokovic BV, Chang EF, Abla AA, Lim DA, Nowakowski TJ. A single-cell atlas of the normal and malformed human brain vasculature. Science 2022; 375:eabi7377. [PMID: 35084939 PMCID: PMC8995178 DOI: 10.1126/science.abi7377] [Citation(s) in RCA: 157] [Impact Index Per Article: 78.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cerebrovascular diseases are a leading cause of death and neurologic disability. Further understanding of disease mechanisms and therapeutic strategies requires a deeper knowledge of cerebrovascular cells in humans. We profiled transcriptomes of 181,388 cells to define a cell atlas of the adult human cerebrovasculature, including endothelial cell molecular signatures with arteriovenous segmentation and expanded perivascular cell diversity. By leveraging this reference, we investigated cellular and molecular perturbations in brain arteriovenous malformations, which are a leading cause of stroke in young people, and identified pathologic endothelial transformations with abnormal vascular patterning and the ontology of vascularly derived inflammation. We illustrate the interplay between vascular and immune cells that contributes to brain hemorrhage and catalog opportunities for targeting angiogenic and inflammatory programs in vascular malformations.
Collapse
Affiliation(s)
- Ethan A Winkler
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
- Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA, USA
- Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
- Department of Neurosurgery, Barrow Neurological Institute, Phoenix, AZ, USA
| | - Chang N Kim
- Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA, USA
- Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
- Department of Anatomy, University of California, San Francisco, CA, USA
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, CA, USA
| | - Jayden M Ross
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
- Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA, USA
- Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
- Department of Anatomy, University of California, San Francisco, CA, USA
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, CA, USA
| | - Joseph H Garcia
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
| | - Eugene Gil
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
- Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA, USA
| | - Irene Oh
- Rebus Biosystems, Santa Clara, CA, USA
| | | | - David Wu
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
- Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA, USA
| | - Joshua S Catapano
- Department of Neurosurgery, Barrow Neurological Institute, Phoenix, AZ, USA
| | - Kunal Raygor
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
| | - Kazim Narsinh
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, USA
| | - Helen Kim
- Center for Cerebrovascular Research, Department of Anesthesia and Perioperative Care, University of California, San Francisco, CA, USA
| | - Shantel Weinsheimer
- Center for Cerebrovascular Research, Department of Anesthesia and Perioperative Care, University of California, San Francisco, CA, USA
| | - Daniel L Cooke
- Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, USA
| | - Brian P Walcott
- Department of Neurosurgery, NorthShore University HealthSystem, Evanston, IL, USA
| | - Michael T Lawton
- Department of Neurosurgery, Barrow Neurological Institute, Phoenix, AZ, USA
| | - Nalin Gupta
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
| | - Berislav V Zlokovic
- Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Edward F Chang
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
- Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Adib A Abla
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
- Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Daniel A Lim
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
- Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA, USA
- Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
- San Francisco Veterans Affairs Medical Center, San Francisco, CA, USA
| | - Tomasz J Nowakowski
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
- Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA, USA
- Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
- Department of Anatomy, University of California, San Francisco, CA, USA
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| |
Collapse
|
13
|
Neumann DP, Goodall GJ, Gregory PA. The Quaking RNA-binding proteins as regulators of cell differentiation. WILEY INTERDISCIPLINARY REVIEWS. RNA 2022; 13:e1724. [PMID: 35298877 PMCID: PMC9786888 DOI: 10.1002/wrna.1724] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 02/18/2022] [Accepted: 02/21/2022] [Indexed: 12/30/2022]
Abstract
The RNA-binding protein Quaking (QKI) has emerged as a potent regulator of cellular differentiation in developmental and pathological processes. The QKI gene is itself alternatively spliced to produce three major isoforms, QKI-5, QKI-6, and QKI-7, that possess very distinct functions. Here, we highlight roles of the different QKI isoforms in neuronal, vascular, muscle, and monocyte cell differentiation, and during epithelial-mesenchymal transition in cancer progression. QKI isoforms control cell differentiation through regulating alternative splicing, mRNA stability and translation, with activities in gene transcription now also becoming evident. These diverse functions of the QKI isoforms contribute to their broad influences on RNA metabolism and cellular differentiation. This article is categorized under: RNA Interactions with Proteins and Other Molecules > Protein-RNA Interactions: Functional Implications RNA Processing > Splicing Regulation/Alternative Splicing RNA in Disease and Development > RNA in Development.
Collapse
Affiliation(s)
- Daniel P. Neumann
- Centre for Cancer BiologyUniversity of South Australia and SA PathologyAdelaideSouth Australia
| | - Gregory J. Goodall
- Centre for Cancer BiologyUniversity of South Australia and SA PathologyAdelaideSouth Australia,Faculty of Health and Medical SciencesThe University of AdelaideAdelaideSouth Australia
| | - Philip A. Gregory
- Centre for Cancer BiologyUniversity of South Australia and SA PathologyAdelaideSouth Australia,Faculty of Health and Medical SciencesThe University of AdelaideAdelaideSouth Australia
| |
Collapse
|
14
|
Yap C, Mieremet A, de Vries CJ, Micha D, de Waard V. Six Shades of Vascular Smooth Muscle Cells Illuminated by KLF4 (Krüppel-Like Factor 4). Arterioscler Thromb Vasc Biol 2021; 41:2693-2707. [PMID: 34470477 PMCID: PMC8545254 DOI: 10.1161/atvbaha.121.316600] [Citation(s) in RCA: 126] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 08/20/2021] [Indexed: 12/23/2022]
Abstract
Multiple layers of vascular smooth muscle cells (vSMCs) are present in blood vessels forming the media of the vessel wall. vSMCs provide a vessel wall structure, enabling it to contract and relax, thus modulating blood flow. They also play a crucial role in the development of vascular diseases, such as atherosclerosis and aortic aneurysm formation. vSMCs display a remarkable high degree of plasticity. At present, the number of different vSMC phenotypes has only partially been characterized. By mapping vSMC phenotypes in detail and identifying triggers for phenotype switching, the relevance of the different phenotypes in vascular disease may be identified. Up until recently, vSMCs were classified as either contractile or dedifferentiated (ie, synthetic). However, single-cell RNA sequencing studies revealed such dedifferentiated arterial vSMCs to be highly diverse. Currently, no consensus exist about the number of vSMC phenotypes. Therefore, we reviewed the data from relevant single-cell RNA sequencing studies, and classified a total of 6 vSMC phenotypes. The central dedifferentiated vSMC type that we classified is the mesenchymal-like phenotype. Mesenchymal-like vSMCs subsequently seem to differentiate into fibroblast-like, macrophage-like, osteogenic-like, and adipocyte-like vSMCs, which contribute differentially to vascular disease. This phenotype switching between vSMCs requires the transcription factor KLF4 (Kruppel-like factor 4). Here, we performed an integrated analysis of the data about the recently identified vSMC phenotypes, their associated gene expression profiles, and previous vSMC knowledge to better understand the role of vSMC phenotype transitions in vascular pathology.
Collapse
Affiliation(s)
- Carmen Yap
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam UMC, Location Academic Medical Center, The Netherlands (C.Y., A.M., C.J.M.d.V., V.d.W.)
| | - Arnout Mieremet
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam UMC, Location Academic Medical Center, The Netherlands (C.Y., A.M., C.J.M.d.V., V.d.W.)
| | - Carlie J.M. de Vries
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam UMC, Location Academic Medical Center, The Netherlands (C.Y., A.M., C.J.M.d.V., V.d.W.)
| | - Dimitra Micha
- Department of Clinical Genetics, Amsterdam Cardiovascular Sciences, Vrije Universiteit Amsterdam, Amsterdam UMC, Location VU University Medical Center, Amsterdam, The Netherlands (D.M.)
| | - Vivian de Waard
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam UMC, Location Academic Medical Center, The Netherlands (C.Y., A.M., C.J.M.d.V., V.d.W.)
| |
Collapse
|
15
|
Inducible Prmt1 ablation in adult vascular smooth muscle leads to contractile dysfunction and aortic dissection. Exp Mol Med 2021; 53:1569-1579. [PMID: 34635781 PMCID: PMC8568946 DOI: 10.1038/s12276-021-00684-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 07/26/2021] [Accepted: 08/06/2021] [Indexed: 01/27/2023] Open
Abstract
Vascular smooth muscle cells (VSMCs) have remarkable plasticity in response to diverse environmental cues. Although these cells are versatile, chronic stress can trigger VSMC dysfunction, which ultimately leads to vascular diseases such as aortic aneurysm and atherosclerosis. Protein arginine methyltransferase 1 (Prmt1) is a major enzyme catalyzing asymmetric arginine dimethylation of proteins that are sources of asymmetric dimethylarginine (ADMA), an endogenous inhibitor of nitric oxide synthase. Although a potential role of Prmt1 in vascular pathogenesis has been proposed, its role in vascular function has yet to be clarified. Here, we investigated the role and underlying mechanism of Prmt1 in vascular smooth muscle contractility and function. The expression of PRMT1 and contractile-related genes was significantly decreased in the aortas of elderly humans and patients with aortic aneurysms. Mice with VSMC-specific Prmt1 ablation (smKO) exhibited partial lethality, low blood pressure and aortic dilation. The Prmt1-ablated aortas showed aortic dissection with elastic fiber degeneration and cell death. Ex vivo and in vitro analyses indicated that Prmt1 ablation significantly decreased the contractility of the aorta and traction forces of VSMCs. Prmt1 ablation downregulated the expression of contractile genes such as myocardin while upregulating the expression of synthetic genes, thus causing the contractile to synthetic phenotypic switch of VSMCs. In addition, mechanistic studies demonstrated that Prmt1 directly regulates myocardin gene activation by modulating epigenetic histone modifications in the myocardin promoter region. Thus, our study demonstrates that VSMC Prmt1 is essential for vascular homeostasis and that its ablation causes aortic dilation/dissection through impaired myocardin expression.
Collapse
|
16
|
Combination of PD98059 and TGF-β1 Efficiently Differentiates Human Urine-Derived Stem Cells into Smooth Muscle Cells. Int J Mol Sci 2021; 22:ijms221910532. [PMID: 34638875 PMCID: PMC8508912 DOI: 10.3390/ijms221910532] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 09/22/2021] [Accepted: 09/24/2021] [Indexed: 11/17/2022] Open
Abstract
Pluripotent adult stem cells have potential applications in cell therapy and tissue engineering. Urine-derived stem cells (UDSCs) differentiate into various cell types. Here, we attempted to differentiate human UDSCs (hUDSCs) into smooth muscle cells (SMCs) using transforming growth factor-beta 1 (TGF-β1) and/or PD98059, an extracellular signal-regulated kinase (ERK) inhibitor. Both quantitative polymerase chain reaction (qPCR) and Western blot analysis showed that the expression of messenger ribonucleic acid (mRNA) and proteins for alpha-smooth muscle actin (α-SMA), calponin (CNN1), and smooth muscle myosin heavy chain (SM-MHC), which are specific markers for SMCs, increased on day 9 after differentiation and again on day 14. The differentiated cells from human UDSCs (hUDSCs) with a combination of TGF-β1 and PD98059 showed the highest expression of SMC marker proteins. Immunocytochemical staining performed to assess the molecular expression revealed CNN and α-SMA colocalizing in the cytoplasm. The cells that differentiated from hUDSCs with a combination of TGF-β1 and PD98059 showed the strongest expression for CNN1, α-SMA, and SM-MHC. Functional testing of the differentiated cells revealed a stronger contractile capacity for the cells differentiated with a combination of PD98059 and TGF-β1 than those differentiated with a single factor. These results suggest the combination of PD98059 and TGF-β1 to be a more effective differentiation method and that differentiated SMCs could be used for restoring the functions of the sphincter muscle or bladder.
Collapse
|
17
|
Li Z, Li JN, Li Q, Liu C, Zhou LH, Zhang Q, Xu Y. miR-25-5p regulates endothelial progenitor cell differentiation in response to shear stress through targeting ABCA1. Cell Biol Int 2021; 45:1876-1886. [PMID: 33945659 DOI: 10.1002/cbin.11621] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 04/07/2021] [Accepted: 05/01/2021] [Indexed: 11/10/2022]
Abstract
The importance of flow shear stress (SS) on the differentiation of endothelial progenitor cells (EPCs) has been demonstrated in various studies. Cholesterol retention and microRNA regulation have been also proposed as relevant factors involved in this process, though evidence regarding their regulatory roles in the differentiation of EPCs is currently lacking. In the present study on high shear stress (HSS)-induced differentiation of EPCs, we investigated the importance of ATP-binding cassette transporter 1 (ABCA1), an important regulator in cholesterol efflux, and miR-25-5p, a potential regulator of endothelial reconstruction. We first revealed an inverse correlation between miR-25-5p and ABCA1 expression levels in EPCs under HSS treatment; their direct interaction was subsequently validated by a dual-luciferase reporter assay. Further studies using flow cytometry and quantitative polymerase chain reaction demonstrated that both miR-25-5p overexpression and ABCA1 inhibition led to elevated levels of specific markers of endothelial cells, with concomitant downregulation of smooth muscle cell markers. Finally, knockdown of ABCA1 in EPCs significantly promoted tube formation, which confirmed our conjecture. Our current results suggest that miR-25-5p might regulate the differentiation of EPCs partially through targeting ABCA1, and such a mechanism might account for HSS-induced differentiation of EPCs.
Collapse
Affiliation(s)
- Zhe Li
- Department of Cerebrovascular Diseases, Blue Cross Brain Hospital affiliated to Tongji University, Shanghai, China
| | - Jia-Nan Li
- Department of Neurosurgery, General Hospital of Northern Theater Command, Shenyang, Liaoning, China
| | - Qiang Li
- Department of Neurosurgery, Changhai Hospital of Shanghai affiliated to Naval Military Medical University, Shanghai, China
| | - Chun Liu
- Department of Cerebrovascular Diseases, Blue Cross Brain Hospital affiliated to Tongji University, Shanghai, China
| | - Lin-Hua Zhou
- Department of Cerebrovascular Diseases, Blue Cross Brain Hospital affiliated to Tongji University, Shanghai, China
| | - Qi Zhang
- Department of Cerebrovascular Diseases, Blue Cross Brain Hospital affiliated to Tongji University, Shanghai, China
| | - Yi Xu
- Department of Neurosurgery, Changhai Hospital of Shanghai affiliated to Naval Military Medical University, Shanghai, China
| |
Collapse
|
18
|
Tao Z, Zheng S, He X, Sun J, He C, Zhang L. Hsa_circ_0037897 may be a risk factor for essential hypertension via hsa-miR-145-5p. Clin Exp Hypertens 2021; 43:281-286. [PMID: 33307836 DOI: 10.1080/10641963.2020.1860081] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Objectives: This study aims to investigate the association between hsa_circ_0037897 and essential hypertension (EH) and to evaluate the diagnostic value of biomarker hsa_circ_0037897 in EH. Methods: This study included 92 EH patients and 92 sex- and age- (±3 years) matched subjects as control. qRT-PCR was performed to measure the expression level of circRNA and miRNA. Logistic regression analysis model was used to assess independent association between hsa_circ_0037897 and EH. Results: The expression level of hsa_circ_0037897 in EH patients was significantly higher (p < .001) compared to the control group, while hsa-miR-145-5p had significantly lower expression(p = .002) than the control group. The area under the ROC curve (AUC) of hsa_circ_0037897 was 0.656. Furthermore, the AUC increased to 0.714 when hsa_circ_0037897 was combined with hsa-miR-145-5p, BMI and smoking. Conclusion: The present results suggested that the high expression of hsa_circ_0037897 may be a risk factor for EH, and hsa_circ_0037897 has certain diagnostic value for EH.
Collapse
Affiliation(s)
- Zhenbo Tao
- Department of Preventative Medicine, Zhejiang Provincial Key Laboratory of Pathological and Physiological Technology, Medicine School of Ningbo University , Ningbo, China
| | - Shuying Zheng
- Department of Health Monitoring, Yinzhou District Center for Disease Control and Prevention , Ningbo, China
| | - Xin He
- Department of Preventative Medicine, Zhejiang Provincial Key Laboratory of Pathological and Physiological Technology, Medicine School of Ningbo University , Ningbo, China
| | - Jihan Sun
- Department of Clinical Medicine, Medicine School of Ningbo University , Ningbo, China
| | - Canxia He
- Department of Preventative Medicine, Zhejiang Provincial Key Laboratory of Pathological and Physiological Technology, Medicine School of Ningbo University , Ningbo, China
| | - Lina Zhang
- Department of Preventative Medicine, Zhejiang Provincial Key Laboratory of Pathological and Physiological Technology, Medicine School of Ningbo University , Ningbo, China
| |
Collapse
|
19
|
Purine-rich element binding protein B attenuates the coactivator function of myocardin by a novel molecular mechanism of smooth muscle gene repression. Mol Cell Biochem 2021; 476:2899-2916. [PMID: 33743134 DOI: 10.1007/s11010-021-04117-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 02/20/2021] [Indexed: 10/21/2022]
Abstract
Myocardin is a potent transcriptional coactivator protein, which functions as the master regulator of vascular smooth muscle cell differentiation. The cofactor activity of myocardin is mediated by its physical interaction with serum response factor, a ubiquitously expressed transactivator that binds to CArG boxes in genes encoding smooth muscle-restricted proteins. Purine-rich element binding protein B (Purβ) represses the transcription of the smooth muscle α-actin gene (Acta2) in fibroblasts and smooth muscle cells by interacting with single-stranded DNA sequences flanking two 5' CArG boxes in the Acta2 promoter. In this study, the ability of Purβ to modulate the cofactor activity of myocardin was investigated using a combination of cellular and biochemical approaches. Results of smooth muscle gene promoter-reporter assays indicated that Purβ specifically inhibits the coactivator function of myocardin in a manner requiring the presence of all three single-stranded DNA binding domains in the Purβ homodimer. DNA binding analyses demonstrated that Purβ interacts with CArG-containing DNA elements with a much lower affinity compared to other purine-rich target sequences present in the Acta2 promoter. Co-immunoprecipitation and DNA pull-down assays revealed that Purβ associates with myocardin and serum response factor when free or bound to duplex DNA containing one or more CArG boxes. Functional analysis of engineered Purβ point mutants identified several amino acid residues essential for suppression of myocardin activity. Collectively, these findings suggest an inhibitory mechanism involving direct protein-protein interaction between the homodimeric Purβ repressor and the myocardin-serum response factor-CArG complex.
Collapse
|
20
|
Sawant D, Lilly B. MicroRNA-145 targets in cancer and the cardiovascular system: evidence for common signaling pathways. VASCULAR BIOLOGY 2020; 2:R115-R128. [PMID: 33283158 PMCID: PMC7709916 DOI: 10.1530/vb-20-0012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 10/23/2020] [Indexed: 12/01/2022]
Abstract
miRNAs are small regulatory RNAs which govern gene expression post-transcriptionally by primarily binding to the 3'-UTR of mRNA target genes. miR-145 is a well-studied miRNA that has been implicated in controlling a range of biological processes. miR-145 is expressed in a variety of tissues and cell types and acts as a tumor-suppressor by regulating target gene signaling pathways involved in different aspects of tumor growth and progression. There is also strong evidence that highlights the important functions of miR-145 in the cardiovascular system. Here, we review the mechanisms of miR-145 in tumorigenesis and cancer progression and compare and contrast with the roles of miR-145 in cardiovascular development and disease. We discuss the important targets of miR-145 in cancer and their possible link to the cardiovascular system.
Collapse
Affiliation(s)
- Dwitiya Sawant
- Center for Cardiovascular Research and The Heart Center, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Brenda Lilly
- Center for Cardiovascular Research and The Heart Center, Nationwide Children's Hospital, Columbus, Ohio, USA.,Department of Pediatrics, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
21
|
Bersini S, Schulte R, Huang L, Tsai H, Hetzer MW. Direct reprogramming of human smooth muscle and vascular endothelial cells reveals defects associated with aging and Hutchinson-Gilford progeria syndrome. eLife 2020; 9:54383. [PMID: 32896271 PMCID: PMC7478891 DOI: 10.7554/elife.54383] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 08/18/2020] [Indexed: 12/14/2022] Open
Abstract
Vascular dysfunctions are a common feature of multiple age-related diseases. However, modeling healthy and pathological aging of the human vasculature represents an unresolved experimental challenge. Here, we generated induced vascular endothelial cells (iVECs) and smooth muscle cells (iSMCs) by direct reprogramming of healthy human fibroblasts from donors of different ages and Hutchinson-Gilford Progeria Syndrome (HGPS) patients. iVECs induced from old donors revealed upregulation of GSTM1 and PALD1, genes linked to oxidative stress, inflammation and endothelial junction stability, as vascular aging markers. A functional assay performed on PALD1 KD VECs demonstrated a recovery in vascular permeability. We found that iSMCs from HGPS donors overexpressed bone morphogenetic protein (BMP)−4, which plays a key role in both vascular calcification and endothelial barrier damage observed in HGPS. Strikingly, BMP4 concentrations are higher in serum from HGPS vs. age-matched mice. Furthermore, targeting BMP4 with blocking antibody recovered the functionality of the vascular barrier in vitro, hence representing a potential future therapeutic strategy to limit cardiovascular dysfunction in HGPS. These results show that iVECs and iSMCs retain disease-related signatures, allowing modeling of vascular aging and HGPS in vitro.
Collapse
Affiliation(s)
- Simone Bersini
- Molecular and Cell Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, United States.,Paul F. Glenn Center for Biology of Aging Research at The Salk Institute, La Jolla, United States
| | - Roberta Schulte
- Molecular and Cell Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, United States
| | - Ling Huang
- The Razavi Newman Integrative Genomics and Bioinformatics Core (IGC), The Salk Institute for Biological Studies, La Jolla, United States
| | - Hannah Tsai
- Molecular and Cell Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, United States
| | - Martin W Hetzer
- Molecular and Cell Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, United States
| |
Collapse
|
22
|
Nwadozi E, Rudnicki M, Haas TL. Metabolic Coordination of Pericyte Phenotypes: Therapeutic Implications. Front Cell Dev Biol 2020; 8:77. [PMID: 32117997 PMCID: PMC7033550 DOI: 10.3389/fcell.2020.00077] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 01/29/2020] [Indexed: 12/15/2022] Open
Abstract
Pericytes are mural vascular cells found predominantly on the abluminal wall of capillaries, where they contribute to the maintenance of capillary structural integrity and vascular permeability. Generally quiescent cells in the adult, pericyte activation and proliferation occur during both physiological and pathological vascular and tissue remodeling. A considerable body of research indicates that pericytes possess attributes of a multipotent adult stem cell, as they are capable of self-renewal as well as commitment and differentiation into multiple lineages. However, pericytes also display phenotypic heterogeneity and recent studies indicate that lineage potential differs between pericyte subpopulations. While numerous microenvironmental cues and cell signaling pathways are known to regulate pericyte functions, the roles that metabolic pathways play in pericyte quiescence, self-renewal or differentiation have been given limited consideration to date. This review will summarize existing data regarding pericyte metabolism and will discuss the coupling of signal pathways to shifts in metabolic pathway preferences that ultimately regulate pericyte quiescence, self-renewal and trans-differentiation. The association between dysregulated metabolic processes and development of pericyte pathologies will be highlighted. Despite ongoing debate regarding pericyte classification and their functional capacity for trans-differentiation in vivo, pericytes are increasingly exploited as a cell therapy tool to promote tissue healing and regeneration. Ultimately, the efficacy of therapeutic approaches hinges on the capacity to effectively control/optimize the fate of the implanted pericytes. Thus, we will identify knowledge gaps that need to be addressed to more effectively harness the opportunity for therapeutic manipulation of pericytes to control pathological outcomes in tissue remodeling.
Collapse
Affiliation(s)
| | | | - Tara L. Haas
- School of Kinesiology and Health Science, Angiogenesis Research Group and Muscle Health Research Centre, York University, Toronto, ON, Canada
| |
Collapse
|
23
|
Martinez AN, Pascale CL, Amenta PS, Israilevich R, Dumont AS. Cell Culture Model to Study Cerebral Aneurysm Biology. ACTA NEUROCHIRURGICA SUPPLEMENT 2020; 127:29-34. [DOI: 10.1007/978-3-030-04615-6_5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
|
24
|
Arévalo-Martínez M, Cidad P, García-Mateo N, Moreno-Estar S, Serna J, Fernández M, Swärd K, Simarro M, de la Fuente MA, López-López JR, Pérez-García MT. Myocardin-Dependent Kv1.5 Channel Expression Prevents Phenotypic Modulation of Human Vessels in Organ Culture. Arterioscler Thromb Vasc Biol 2019; 39:e273-e286. [DOI: 10.1161/atvbaha.119.313492] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Objective:
We have previously described that changes in the expression of Kv channels associate to phenotypic modulation (PM), so that Kv1.3/Kv1.5 ratio is a landmark of vascular smooth muscle cells phenotype. Moreover, we demonstrated that the Kv1.3 functional expression is relevant for PM in several types of vascular lesions. Here, we explore the efficacy of Kv1.3 inhibition for the prevention of remodeling in human vessels, and the mechanisms linking the switch in Kv1.3 /Kv1.5 ratio to PM.
Approach and Results:
Vascular remodeling was explored using organ culture and primary cultures of vascular smooth muscle cells obtained from human vessels. We studied the effects of Kv1.3 inhibition on serum-induced remodeling, as well as the impact of viral vector-mediated overexpression of Kv channels or myocardin knock-down. Kv1.3 blockade prevented remodeling by inhibiting proliferation, migration, and extracellular matrix secretion. PM activated Kv1.3 via downregulation of Kv1.5. Hence, both Kv1.3 blockers and Kv1.5 overexpression inhibited remodeling in a nonadditive fashion. Finally, myocardin knock-down induced vessel remodeling and Kv1.5 downregulation and myocardin overexpression increased Kv1.5, while Kv1.5 overexpression inhibited PM without changing myocardin expression.
Conclusions:
We demonstrate that Kv1.5 channel gene is a myocardin-regulated, vascular smooth muscle cells contractile marker. Kv1.5 downregulation upon PM leaves Kv1.3 as the dominant Kv1 channel expressed in dedifferentiated cells. We demonstrated that the inhibition of Kv1.3 channel function with selective blockers or by preventing Kv1.5 downregulation can represent an effective, novel strategy for the prevention of intimal hyperplasia and restenosis of the human vessels used for coronary angioplasty procedures.
Collapse
Affiliation(s)
- Marycarmen Arévalo-Martínez
- From the Departamento de Bioquímica y Biología Molecular y Fisiología, Universidad de Valladolid, Spain (M.A.-M., P.C., N.G.-M., S.M.-E., J.S., J.R.L.-L., M.T.P.-G.)
- Instituto de Biología y Genética Molecular (IBGM), CSIC, Universidad de Valladolid, Spain (M.A.-M., P.C., N.G.-M., S.M.-E., J.S., M.S., M.A.d.l.F.)
| | - Pilar Cidad
- From the Departamento de Bioquímica y Biología Molecular y Fisiología, Universidad de Valladolid, Spain (M.A.-M., P.C., N.G.-M., S.M.-E., J.S., J.R.L.-L., M.T.P.-G.)
- Instituto de Biología y Genética Molecular (IBGM), CSIC, Universidad de Valladolid, Spain (M.A.-M., P.C., N.G.-M., S.M.-E., J.S., M.S., M.A.d.l.F.)
| | - Nadia García-Mateo
- From the Departamento de Bioquímica y Biología Molecular y Fisiología, Universidad de Valladolid, Spain (M.A.-M., P.C., N.G.-M., S.M.-E., J.S., J.R.L.-L., M.T.P.-G.)
- Instituto de Biología y Genética Molecular (IBGM), CSIC, Universidad de Valladolid, Spain (M.A.-M., P.C., N.G.-M., S.M.-E., J.S., M.S., M.A.d.l.F.)
| | - Sara Moreno-Estar
- From the Departamento de Bioquímica y Biología Molecular y Fisiología, Universidad de Valladolid, Spain (M.A.-M., P.C., N.G.-M., S.M.-E., J.S., J.R.L.-L., M.T.P.-G.)
- Instituto de Biología y Genética Molecular (IBGM), CSIC, Universidad de Valladolid, Spain (M.A.-M., P.C., N.G.-M., S.M.-E., J.S., M.S., M.A.d.l.F.)
| | - Julia Serna
- From the Departamento de Bioquímica y Biología Molecular y Fisiología, Universidad de Valladolid, Spain (M.A.-M., P.C., N.G.-M., S.M.-E., J.S., J.R.L.-L., M.T.P.-G.)
- Instituto de Biología y Genética Molecular (IBGM), CSIC, Universidad de Valladolid, Spain (M.A.-M., P.C., N.G.-M., S.M.-E., J.S., M.S., M.A.d.l.F.)
| | - Mirella Fernández
- Cardiovascular Surgery Department, Hospital Clínico Universitario de Valladolid, Spain (M.F.)
| | - Karl Swärd
- Department of Experimental Medical Science, University of Lund, Sweden (K.S.)
| | - María Simarro
- Instituto de Biología y Genética Molecular (IBGM), CSIC, Universidad de Valladolid, Spain (M.A.-M., P.C., N.G.-M., S.M.-E., J.S., M.S., M.A.d.l.F.)
- Departamento de Enfermería, Universidad de Valladolid, Spain (M.S.)
| | - Miguel A. de la Fuente
- Instituto de Biología y Genética Molecular (IBGM), CSIC, Universidad de Valladolid, Spain (M.A.-M., P.C., N.G.-M., S.M.-E., J.S., M.S., M.A.d.l.F.)
- Departamento de Biología Celular, Universidad de Valladolid, Spain (M.A.d.l.F.)
| | - José R. López-López
- From the Departamento de Bioquímica y Biología Molecular y Fisiología, Universidad de Valladolid, Spain (M.A.-M., P.C., N.G.-M., S.M.-E., J.S., J.R.L.-L., M.T.P.-G.)
| | - M. Teresa Pérez-García
- From the Departamento de Bioquímica y Biología Molecular y Fisiología, Universidad de Valladolid, Spain (M.A.-M., P.C., N.G.-M., S.M.-E., J.S., J.R.L.-L., M.T.P.-G.)
| |
Collapse
|
25
|
Hirai H, Yang B, Garcia-Barrio MT, Rom O, Ma PX, Zhang J, Chen YE. Direct Reprogramming of Fibroblasts Into Smooth Muscle-Like Cells With Defined Transcription Factors-Brief Report. Arterioscler Thromb Vasc Biol 2019; 38:2191-2197. [PMID: 30026272 DOI: 10.1161/atvbaha.118.310870] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Objective- To identify the transcription factors that could contribute to direct reprogramming of fibroblasts toward smooth muscle cell fate. Approach and Results- We screened various combinations of transcription factors, including Myocd (myocardin), Mef2C (myocyte enhancer factor 2C), Mef2B (myocyte enhancer factor 2B), Mkl1 (MKL [megakaryoblastic leukemia]/Myocd-like 1), Gata4 (GATA-binding protein 4), Gata5 (GATA-binding protein 5), Gata6 (GATA-binding protein 6), Ets1 (E26 avian leukemia oncogene 1, 5' domain), and their corresponding carboxyterminal fusions to the transactivation domain of MyoD (myogenic differentiation 1)-indicated by *-for their effects on reprogramming mouse embryonic fibroblasts and human adult dermal fibroblasts to the smooth muscle cell fate as determined by the expression of specific markers. The combination of 3 transcription factors, Myocd (or Myocd*) with Mef2C (or Mef2C*) and Gata6, was the most efficient in enhancing the expression of smooth muscle marker genes and decreasing fibroblast gene expression. Additionally, the derived induced smooth muscle-like cells showed a contractile phenotype in response to carbachol. Conclusions- Combination of Myocd and Gata6 with Mef2C* (MG2*) could sufficiently and efficiently direct differentiation of mouse embryonic and human dermal fibroblasts into induced smooth muscle-like cells, thus opening new opportunities for disease modeling, tissue engineering, and personalized medicine.
Collapse
Affiliation(s)
- Hiroyuki Hirai
- From the Division of Cardiovascular Medicine, Department of Internal Medicine (H.H., M.T.G.-B., O.R., J.Z., Y.E.C.).,Department of Cardiac Surgery (H.H., B.Y., Y.E.C.)
| | - Bo Yang
- Department of Cardiac Surgery (H.H., B.Y., Y.E.C.)
| | - Minerva T Garcia-Barrio
- From the Division of Cardiovascular Medicine, Department of Internal Medicine (H.H., M.T.G.-B., O.R., J.Z., Y.E.C.)
| | - Oren Rom
- From the Division of Cardiovascular Medicine, Department of Internal Medicine (H.H., M.T.G.-B., O.R., J.Z., Y.E.C.)
| | - Peter X Ma
- Department of Materials Science and Engineering (P.X.M.), University of Michigan, Ann Arbor
| | - Jifeng Zhang
- From the Division of Cardiovascular Medicine, Department of Internal Medicine (H.H., M.T.G.-B., O.R., J.Z., Y.E.C.)
| | - Y Eugene Chen
- From the Division of Cardiovascular Medicine, Department of Internal Medicine (H.H., M.T.G.-B., O.R., J.Z., Y.E.C.).,Department of Cardiac Surgery (H.H., B.Y., Y.E.C.)
| |
Collapse
|
26
|
Zhang M, Wang Z. Downregulation of miR143/145 gene cluster expression promotes the aortic media degeneration process via the TGF-β1 signaling pathway. Am J Transl Res 2019; 11:370-378. [PMID: 30787994 PMCID: PMC6357338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 12/23/2018] [Indexed: 06/09/2023]
Abstract
Aortic dissection (AD) is a serious threat to human health; however, the cause of this condition has not yet been fully elucidated. In this study, we found significantly increased expression of phospho-Smad2/3 and phospho-ERK in AD tissues and downregulated expression of miR143 and miR145 in AD tissues. Knockdown of the miR143/145 gene cluster induced phenotypic switching of vascular smooth muscle cells (VSMCs) and activation of the TGF-β1 signaling pathway. When the TFG-β1 signaling pathway was blocked by pretreatment with an LY364947 inhibitor, expression of miR143 and miR145, and VSMC phenotypic markers were not affected by knockdown of the miR143/145 gene cluster. Immunohistochemical staining of aortic tissues donated by AD patients and organ donors showed decrease alpha-smooth muscle actin (α-SMA) expression in pathological tissue, while osteopontin (OPN) expression increased and the arrangement of smooth muscle cells in the tunica media was dysregulated. In conclusion, our study suggests that downregulated expression of the miR143/145 gene cluster promotes phenotypic switching of VSMCs via the TGF-β1 signaling pathway. This may play an important role in the pathogenesis of AD.
Collapse
Affiliation(s)
- Min Zhang
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University Wuhan 430000, Hubei Province, People's Republic of China
| | - Zhiwei Wang
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University Wuhan 430000, Hubei Province, People's Republic of China
| |
Collapse
|
27
|
Vacante F, Denby L, Sluimer JC, Baker AH. The function of miR-143, miR-145 and the MiR-143 host gene in cardiovascular development and disease. Vascul Pharmacol 2019; 112:24-30. [PMID: 30502421 PMCID: PMC6395947 DOI: 10.1016/j.vph.2018.11.006] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 11/22/2018] [Accepted: 11/22/2018] [Indexed: 02/09/2023]
Abstract
Noncoding RNAs (long noncoding RNAs and small RNAs) are emerging as critical modulators of phenotypic changes associated with physiological and pathological contexts in a variety of cardiovascular diseases (CVDs). Although it has been well established that hereditable genetic alterations and exposure to risk factors are crucial in the development of CVDs, other critical regulators of cell function impact on disease processes. Here we discuss noncoding RNAs have only recently been identified as key players involved in the progression of disease. In particular, we discuss micro RNA (miR)-143/145 since they represent one of the most characterised microRNA clusters regulating smooth muscle cell (SMC) differentiation and phenotypic switch in response to vascular injury and remodelling. MiR143HG is a well conserved long noncoding RNA (lncRNA), which is the host gene for miR-143/145 and recently implicated in cardiac specification during heart development. Although the lncRNA-miRNA interactions have not been completely characterised, their crosstalk is now beginning to emerge and likely requires further research focus. In this review we give an overview of the biology of the genomic axis that is miR-143/145 and MiR143HG, focusing on their important functional role(s) in the cardiovascular system.
Collapse
Affiliation(s)
- Francesca Vacante
- Centre for Cardiovascular Science, The Queen's Medical Research Institute, Edinburgh EH16 4TJ, UK
| | - Laura Denby
- Centre for Cardiovascular Science, The Queen's Medical Research Institute, Edinburgh EH16 4TJ, UK
| | - Judith C Sluimer
- Centre for Cardiovascular Science, The Queen's Medical Research Institute, Edinburgh EH16 4TJ, UK; Maastricht University Medical Centre, Dept. of Pathology, Maastricht 6229 HX, The Netherlands
| | - Andrew H Baker
- Centre for Cardiovascular Science, The Queen's Medical Research Institute, Edinburgh EH16 4TJ, UK.
| |
Collapse
|
28
|
Frismantiene A, Philippova M, Erne P, Resink TJ. Smooth muscle cell-driven vascular diseases and molecular mechanisms of VSMC plasticity. Cell Signal 2018; 52:48-64. [PMID: 30172025 DOI: 10.1016/j.cellsig.2018.08.019] [Citation(s) in RCA: 246] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 08/28/2018] [Accepted: 08/28/2018] [Indexed: 02/06/2023]
Abstract
Vascular smooth muscle cells (VSMCs) are the major cell type in blood vessels. Unlike many other mature cell types in the adult body, VSMC do not terminally differentiate but retain a remarkable plasticity. Fully differentiated medial VSMCs of mature vessels maintain quiescence and express a range of genes and proteins important for contraction/dilation, which allows them to control systemic and local pressure through the regulation of vascular tone. In response to vascular injury or alterations in local environmental cues, differentiated/contractile VSMCs are capable of switching to a dedifferentiated phenotype characterized by increased proliferation, migration and extracellular matrix synthesis in concert with decreased expression of contractile markers. Imbalanced VSMC plasticity results in maladaptive phenotype alterations that ultimately lead to progression of a variety of VSMC-driven vascular diseases. The nature, extent and consequences of dysregulated VSMC phenotype alterations are diverse, reflecting the numerous environmental cues (e.g. biochemical factors, extracellular matrix components, physical) that prompt VSMC phenotype switching. In spite of decades of efforts to understand cues and processes that normally control VSMC differentiation and their disruption in VSMC-driven disease states, the crucial molecular mechanisms and signalling pathways that shape the VSMC phenotype programme have still not yet been precisely elucidated. In this article we introduce the physiological functions of vascular smooth muscle/VSMCs, outline VSMC-driven cardiovascular diseases and the concept of VSMC phenotype switching, and review molecular mechanisms that play crucial roles in the regulation of VSMC phenotypic plasticity.
Collapse
Affiliation(s)
- Agne Frismantiene
- Department of Biomedicine, Laboratory for Signal Transduction, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Maria Philippova
- Department of Biomedicine, Laboratory for Signal Transduction, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Paul Erne
- Department of Biomedicine, Laboratory for Signal Transduction, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Therese J Resink
- Department of Biomedicine, Laboratory for Signal Transduction, University Hospital Basel and University of Basel, Basel, Switzerland.
| |
Collapse
|
29
|
Leimgruber C, Quintar AA, Peinetti N, Scalerandi MV, Nicola JP, Miano JM, Maldonado CA. Testosterone Rescues the De-Differentiation of Smooth Muscle Cells Through Serum Response Factor/Myocardin. J Cell Physiol 2017; 232:2806-2817. [PMID: 27861881 DOI: 10.1002/jcp.25679] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 11/07/2016] [Indexed: 12/23/2022]
Abstract
Prostatic smooth muscle cells (pSMCs) differentiation is a key factor for prostatic homeostasis, with androgens exerting multiple effects on these cells. Here, we demonstrated that the myodifferentiator complex Srf/Myocd is up-regulated by testosterone in a dose-dependent manner in primary cultures of rat pSMCs, which was associated to the increase in Acta2, Cnn1, and Lmod1 expressions. Blocking Srf or Myocd by siRNAs inhibited the myodifferentiator effect of testosterone. While LPS led to a dedifferentiated phenotype in pSMCs, characterized by down-regulation of Srf/Myocd and smooth muscle cell (SMC)-restricted genes, endotoxin treatment on Myocd-overexpressing cells did not result in phenotypic alterations. Testosterone at a physiological dose was able to restore the muscular phenotype by normalizing Srf/Myocd expression in inflammation-induced dedifferentiated pSMCs. Moreover, the androgen reestablished the proliferation rate and IL-6 secretion increased by LPS. These results provide novel evidence regarding the myodifferentiating role of testosterone on SMCs by modulating Srf/Myocd. Thus, androgens preserve prostatic SMC phenotype, which is essential to maintain the normal structure and function of the prostate. J. Cell. Physiol. 232: 2806-2817, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Carolina Leimgruber
- Centro de Microscopía Electrónica, Instituto de Investigaciones en Ciencias de la Salud (INICSA-CONICET), Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Amado A Quintar
- Centro de Microscopía Electrónica, Instituto de Investigaciones en Ciencias de la Salud (INICSA-CONICET), Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Nahuel Peinetti
- Centro de Microscopía Electrónica, Instituto de Investigaciones en Ciencias de la Salud (INICSA-CONICET), Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - María V Scalerandi
- Centro de Microscopía Electrónica, Instituto de Investigaciones en Ciencias de la Salud (INICSA-CONICET), Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Juan P Nicola
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Joseph M Miano
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, New York
| | - Cristina A Maldonado
- Centro de Microscopía Electrónica, Instituto de Investigaciones en Ciencias de la Salud (INICSA-CONICET), Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| |
Collapse
|
30
|
Veber M, Dolivo D, Rolle M, Dominko T. Pro-myogenic and low-oxygen culture increases expression of contractile smooth muscle markers in human fibroblasts. J Tissue Eng Regen Med 2017; 12:572-582. [PMID: 28513058 DOI: 10.1002/term.2473] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 04/25/2017] [Accepted: 05/09/2017] [Indexed: 01/23/2023]
Abstract
Smooth muscle cells (SMCs) are essential for tissue engineering strategies to fabricate organs such as blood vessels, the oesophagus and bladder, and to create disease models of these systems. In order for such therapies and models to be feasible, SMCs must be sourced effectively to enable production of large numbers of functional cells. In vitro, SMCs divide slowly and demonstrate short proliferative lifespans compared with other types of cells, including stem cells and fibroblasts, limiting the number of cells that can be derived from expansion in culture of a primary isolation. As such, it would be beneficial to better understand the factors underlying induction and maintenance of SMC phenotypes, in order to produce new sources of SMCs for tissue engineering and disease modelling. Here we report the ability of human dermal fibroblasts to display patterns of gene expression resembling contractile SMCs when cultured under conditions that are known to promote a contractile phenotype in SMCs, including culture on collagen IV, low-serum culture, TGF-β1 treatment and hypoxia. These factors drive expression of the myogenic transcription factor myocardin, as well as expression of several of its gene targets that are known contributors to contractile phenotype in SMCs, including smooth muscle alpha actin, calponin, and myosin heavy chain. Our results suggest that culture conditions associated with culture of SMCs may be sufficient to induce myogenic gene expression patterns and potential myogenic function in non-muscle cells.
Collapse
Affiliation(s)
| | - David Dolivo
- Department of Biology and Biotechnology, Worcester Polytechnic Institute, Worcester, Massachusetts, USA
| | - Marsha Rolle
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, Massachusetts, USA
| | - Tanja Dominko
- Department of Biology and Biotechnology, Worcester Polytechnic Institute, Worcester, Massachusetts, USA.,Center for Biomedical Sciences and Engineering, University of Nova Gorica, Vipava, Slovenia
| |
Collapse
|
31
|
Ignatieva E, Kostina D, Irtyuga O, Uspensky V, Golovkin A, Gavriliuk N, Moiseeva O, Kostareva A, Malashicheva A. Mechanisms of Smooth Muscle Cell Differentiation Are Distinctly Altered in Thoracic Aortic Aneurysms Associated with Bicuspid or Tricuspid Aortic Valves. Front Physiol 2017; 8:536. [PMID: 28790933 PMCID: PMC5524772 DOI: 10.3389/fphys.2017.00536] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Accepted: 07/10/2017] [Indexed: 12/30/2022] Open
Abstract
Cellular and molecular mechanisms of thoracic aortic aneurysm are not clear and therapeutic approaches are mostly absent. Thoracic aortic aneurysm is associated with defective differentiation of smooth muscle cells (SMC) of aortic wall. Bicuspid aortic valve (BAV) comparing to tricuspid aortic valve (TAV) significantly predisposes to a risk of thoracic aortic aneurysms. It has been suggested recently that BAV-associated aortopathies represent a separate pathology comparing to TAV-associated dilations. The only proven candidate gene that has been associated with BAV remains NOTCH1. In this study we tested the hypothesis that Notch-dependent and related TGF-β and BMP differentiation pathways are differently altered in aortic SMC of BAV- vs. TAV-associated aortic aneurysms. SMC were isolated from aortic tissues of the patients with BAV- or TAV-associated aortic aneurysms and from healthy donors used as controls. Gene expression was verified by qPCR and Western blotting. For TGF-β induced differentiation SMC were treated with the medium containing TGF-β1. To induce proosteogenic signaling we cultured SMC in the presence of specific osteogenic factors. Notch-dependent differentiation was induced via lentiviral transduction of SMC with activated Notch1 domain. MYOCD expression, a master gene of SMC differentiation, was down regulated in SMC of both BAV and TAV patients. Discriminant analysis of gene expression patterns included a set of contractile genes specific for SMC, Notch-related genes and proosteogenic genes and revealed that control cells form a separate cluster from both BAV and TAV group, while BAV- and TAV-derived SMC are partially distinct with some overlapping. In differentiation experiments TGF-β caused similar patterns of target gene expression for BAV- and TAV derived cells while the induction was higher in the diseased cells than in control ones. Osteogenic induction caused significant change in RUNX2 expression exclusively in BAV group. Notch activation induced significant ACTA2 expression also exclusively in BAV group. We show that Notch acts synergistically with proosteogenic factors to induce ACTA2 transcription and osteogenic differentiation. In conclusion we have found differences in responsiveness of SMC to Notch and to proosteogenic induction between BAV- and TAV-associated aortic aneurysms.
Collapse
Affiliation(s)
- Elena Ignatieva
- Laboratory of Molecular Cardiology, Almazov Federal Medical Research CentreSaint Petersburg, Russia
| | - Daria Kostina
- Laboratory of Molecular Cardiology, Almazov Federal Medical Research CentreSaint Petersburg, Russia.,Department of Medical Physics, Peter the Great Saint-Petersburg Polytechnic UniversitySaint Petersburg, Russia
| | - Olga Irtyuga
- Laboratory of Molecular Cardiology, Almazov Federal Medical Research CentreSaint Petersburg, Russia
| | - Vladimir Uspensky
- Laboratory of Molecular Cardiology, Almazov Federal Medical Research CentreSaint Petersburg, Russia
| | - Alexey Golovkin
- Laboratory of Molecular Cardiology, Almazov Federal Medical Research CentreSaint Petersburg, Russia
| | - Natalia Gavriliuk
- Laboratory of Molecular Cardiology, Almazov Federal Medical Research CentreSaint Petersburg, Russia
| | - Olga Moiseeva
- Laboratory of Molecular Cardiology, Almazov Federal Medical Research CentreSaint Petersburg, Russia
| | - Anna Kostareva
- Laboratory of Molecular Cardiology, Almazov Federal Medical Research CentreSaint Petersburg, Russia.,Laboratory of Bioinformatics and Genomics, Institute of Translational Medicine, ITMO UniversitySaint Petersburg, Russia
| | - Anna Malashicheva
- Laboratory of Molecular Cardiology, Almazov Federal Medical Research CentreSaint Petersburg, Russia.,Laboratory of Bioinformatics and Genomics, Institute of Translational Medicine, ITMO UniversitySaint Petersburg, Russia.,Faculty of Biology, Saint-Petersburg State UniversitySaint Petersburg, Russia
| |
Collapse
|
32
|
|
33
|
Liao XH, Xiang Y, Li H, Zheng DL, Xu Y, Xi Yu C, Li JP, Zhang XY, Xing WB, Cao DS, Bao LY, Zhang TC. VEGF-A Stimulates STAT3 Activity via Nitrosylation of Myocardin to Regulate the Expression of Vascular Smooth Muscle Cell Differentiation Markers. Sci Rep 2017; 7:2660. [PMID: 28572685 PMCID: PMC5453982 DOI: 10.1038/s41598-017-02907-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 04/20/2017] [Indexed: 01/21/2023] Open
Abstract
Vascular endothelial growth factor A (VEGF-A) is a pivotal player in angiogenesis. It is capable of influencing such cellular processes as tubulogenesis and vascular smooth muscle cell (VSMC) proliferation, yet very little is known about the actual signaling events that mediate VEGF-A induced VSMC phenotypic switch. In this report, we describe the identification of an intricate VEGF-A-induced signaling cascade that involves VEGFR2, STAT3, and Myocardin. We demonstrate that VEGF-A promotes VSMC proliferation via VEGFR2/STAT3-mediated upregulating the proliferation of markers like Cyclin D1 and PCNA. Specifically, VEGF-A leads to nitrosylation of Myocardin, weakens its effect on promoting the expression of contractile markers and is unable to inhibit the activation of STAT3. These observations reinforce the importance of nitric oxide and S-nitrosylation in angiogenesis and provide a mechanistic pathway for VEGF-A-induced VSMC phenotypic switch. In addition, Myocardin, GSNOR and GSNO can create a negative feedback loop to regulate the VSMC phenotypic switch. Thus, the discovery of this interactive network of signaling pathways provides novel and unexpected therapeutic targets for angiogenesis-dependent diseases.
Collapse
Affiliation(s)
- Xing Hua Liao
- Institute of Biology and Medicine, Wuhan University of Science and Technology, Wuhan, 430000, P. R. China. .,Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education and Tianjin, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, 300457, P. R. China.
| | - Yuan Xiang
- Institute of Biology and Medicine, Wuhan University of Science and Technology, Wuhan, 430000, P. R. China
| | - Hui Li
- Institute of Biology and Medicine, Wuhan University of Science and Technology, Wuhan, 430000, P. R. China
| | - De Liang Zheng
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education and Tianjin, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, 300457, P. R. China
| | - Yao Xu
- Institute of Biology and Medicine, Wuhan University of Science and Technology, Wuhan, 430000, P. R. China
| | - Cheng Xi Yu
- Institute of Biology and Medicine, Wuhan University of Science and Technology, Wuhan, 430000, P. R. China
| | - Jia Peng Li
- Institute of Biology and Medicine, Wuhan University of Science and Technology, Wuhan, 430000, P. R. China
| | - Xiao Yu Zhang
- Institute of Biology and Medicine, Wuhan University of Science and Technology, Wuhan, 430000, P. R. China
| | - Wei Bin Xing
- Institute of Biology and Medicine, Wuhan University of Science and Technology, Wuhan, 430000, P. R. China
| | - Dong Sun Cao
- Institute of Biology and Medicine, Wuhan University of Science and Technology, Wuhan, 430000, P. R. China
| | - Le Yuan Bao
- Institute of Biology and Medicine, Wuhan University of Science and Technology, Wuhan, 430000, P. R. China
| | - Tong Cun Zhang
- Institute of Biology and Medicine, Wuhan University of Science and Technology, Wuhan, 430000, P. R. China. .,Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education and Tianjin, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, 300457, P. R. China.
| |
Collapse
|
34
|
Li B, Wang Z, Hu Z, Zhang M, Ren Z, Zhou Z, Huang J, Hu X. P38 MAPK Signaling Pathway Mediates Angiotensin II-Induced miR143/145 Gene Cluster Downregulation during Aortic Dissection Formation. Ann Vasc Surg 2017; 40:262-273. [PMID: 28167124 DOI: 10.1016/j.avsg.2016.09.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 08/18/2016] [Accepted: 09/09/2016] [Indexed: 02/02/2023]
Abstract
BACKGROUND We endeavored to prove that angiotensin II (Ang II) regulates both the expression of micro-RNA143/145 (miR143/145) and differentiation of vascular smooth muscle cells (VSMCs) during the formation of aortic dissection (AD). We also studied the contribution of p38 mitogen-activated protein kinase (MAPK) signaling pathway toward this process. METHODS Ascending aortic tissues were harvested from the patients with AD and organ donors. Tissues were immunostained with labeled antibodies targeting p38 MAPK, phospho-p38 MAPK, alpha-smooth muscle actin (α-SMA), and osteopontin (OPN). Next, we treated mouse aortic VSMCs with different regimens of Ang II (duration and dosages) in vitro and determined expression levels of miR143/145 and VSMC phenotype marker proteins (α-SMA and OPN) by quantitative polymerase chain reaction and/or western blotting. SB203580 was used to inhibit the p38 MAPK signaling pathway. Finally, the VSMC phenotype was validated by immunofluorescence microscopy. RESULTS Expression of phospho-p38 MAPK was significantly greater in the AD tissue. Ang II induced the phenotypic switching of VSMCs along with the downregulation of an miR143/145 gene cluster. Expression of OPN and phospho-p38 was significantly increased in VSMCs treated with 0.1 μM Ang II for 12 hr. Furthermore, the expression of miR143 and miR145 was downregulated by Ang II treatment. When the p38 MAPK signaling pathway was blocked by pretreatment with an SB203580 inhibitor, the expression of miR143, miR145, and VSMC phenotypic markers was not affected by Ang II. Immunohistochemical staining of aortic tissues donated by AD patients and healthy donors showed that the expression of α-SMA decreased in pathological tissue, while the OPN increased and the arrangement of the smooth muscle cells of the media was dysregulated, which we verified in vitro. CONCLUSIONS Ang II could regulate the expression of miR143/145 gene cluster and the phenotypic switching of VSMCs via the p38 MAPK signaling pathway. This may play an important role in the pathogenesis of AD.
Collapse
MESH Headings
- Actins/metabolism
- Adult
- Aortic Dissection/enzymology
- Aortic Dissection/genetics
- Aortic Dissection/pathology
- Angiotensin II/pharmacology
- Animals
- Aortic Aneurysm/enzymology
- Aortic Aneurysm/genetics
- Aortic Aneurysm/pathology
- Case-Control Studies
- Cells, Cultured
- Dose-Response Relationship, Drug
- Female
- Humans
- Male
- Mice, Inbred C57BL
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Middle Aged
- Multigene Family
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/enzymology
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/enzymology
- Myocytes, Smooth Muscle/pathology
- Phosphorylation
- Signal Transduction/drug effects
- Time Factors
- p38 Mitogen-Activated Protein Kinases/metabolism
Collapse
Affiliation(s)
- Bowen Li
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, People's Republic of China
| | - Zhiwei Wang
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, People's Republic of China.
| | - Zhipeng Hu
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, People's Republic of China
| | - Min Zhang
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, People's Republic of China
| | - Zongli Ren
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, People's Republic of China
| | - Zhen Zhou
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, People's Republic of China
| | - Jizhen Huang
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, People's Republic of China
| | - Xiaoping Hu
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, People's Republic of China
| |
Collapse
|
35
|
Li H, Xiang Y, Fan LJ, Zhang XY, Li JP, Yu CX, Bao LY, Cao DS, Xing WB, Liao XH, Zhang TC. Myocardin inhibited the gap protein connexin 43 via promoted miR-206 to regulate vascular smooth muscle cell phenotypic switch. Gene 2017; 616:22-30. [PMID: 28342807 DOI: 10.1016/j.gene.2017.03.029] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Revised: 03/17/2017] [Accepted: 03/21/2017] [Indexed: 11/15/2022]
Abstract
Myocardin is regarded as a key mediator for the change of smooth muscle phenotype. The gap junction protein connexin 43 (Cx43) has been shown to be involved in vascular smooth muscle cells (VSMCs) proliferation and the development of atherosclerosis. However, the role of myocardin on gap junction of cell communication and the relation between myocardin and Cx43 in VSMC phenotypic switch has not been investigated. The goal of the present study is to investigate the molecular mechanism by which myocardin affects Cx43-regulated VSMC proliferation. Data presented in this study demonstrated that inhibition of the Cx43 activation process impaired VSMC proliferation. On the other hand, overexpression miR-206 inhibited VSMC proliferation. In additon, miR-206 silences the expression of Cx43 via targeting Cx43 3' Untranslated Regions. Importantly, myocardin can significantly promote the expression of miR-206. Cx43 regulates VSMCs' proliferation and metastasis through miR-206, which could be promoted by myocardin and used as a marker for diagnosis and a target for therapeutic intervention. Thus myocardin affected the gap junction by inhibited Cx43 and myocardin-miR-206-Cx43 formed a loop to regulate VSMC phenotypic switch.
Collapse
Affiliation(s)
- Hui Li
- Institute of Biology and Medicine, Wuhan University of Science and Technology, 430000, PR China
| | - Yuan Xiang
- Institute of Biology and Medicine, Wuhan University of Science and Technology, 430000, PR China
| | - Li-Juan Fan
- Institute of Biology and Medicine, Wuhan University of Science and Technology, 430000, PR China
| | - Xiao-Yu Zhang
- Institute of Biology and Medicine, Wuhan University of Science and Technology, 430000, PR China
| | - Jia-Peng Li
- Institute of Biology and Medicine, Wuhan University of Science and Technology, 430000, PR China
| | - Cheng-Xi Yu
- Institute of Biology and Medicine, Wuhan University of Science and Technology, 430000, PR China
| | - Le-Yuan Bao
- Institute of Biology and Medicine, Wuhan University of Science and Technology, 430000, PR China
| | - Dong-Sun Cao
- Institute of Biology and Medicine, Wuhan University of Science and Technology, 430000, PR China
| | - Wei-Bing Xing
- Institute of Biology and Medicine, Wuhan University of Science and Technology, 430000, PR China
| | - Xing-Hua Liao
- Institute of Biology and Medicine, Wuhan University of Science and Technology, 430000, PR China; Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education and Tianjin, College of Biotechnology, Tianjin University of Science and Technology, 300457, PR China.
| | - Tong-Cun Zhang
- Institute of Biology and Medicine, Wuhan University of Science and Technology, 430000, PR China; Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education and Tianjin, College of Biotechnology, Tianjin University of Science and Technology, 300457, PR China.
| |
Collapse
|
36
|
Jehanno C, Flouriot G, Nicol-Benoît F, Le Page Y, Le Goff P, Michel D. Envisioning metastasis as a transdifferentiation phenomenon clarifies discordant results on cancer. Breast Dis 2017; 36:47-59. [PMID: 27177343 DOI: 10.3233/bd-150210] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Cancer is generally conceived as a dedifferentiation process in which quiescent post-mitotic differentiated cells acquire stem-like properties and the capacity to proliferate. This view holds for the initial stages of carcinogenesis but is more questionable for advanced stages when the cells can transdifferentiate into the contractile phenotype associated to migration and metastasis. Singularly from this perspective, the hallmark of the most aggressive cancers would correspond to a genuine differentiation status, even if it is different from the original one. This seeming paradox could help reconciling discrepancies in the literature about the pro- or anti-tumoral functions of candidate molecules involved in cancer and whose actual effects depend on the tumoral grade. These ambiguities which are likely to concern a myriad of molecules and pathways, are illustrated here with the selected examples of chromatin epigenetics and myocardin-related transcription factors, using the human MCF10A and MCF7 breast cancer cells. Self-renewing stem like cells are characterized by a loose chromatin with low levels of the H3K9 trimetylation, but high levels of this mark can also appear in cancer cells acquiring a contractile-type differentiation state associated to metastasis. Similarly, the myocardin-related transcription factor MRTF-A is involved in metastasis and epithelial-mesenchymal transition, whereas this factor is naturally enriched in the quiescent cells which are precisely the most resistant to cancer: cardiomyocytes. These seeming paradoxes reflect the bistable epigenetic landscape of cancer in which dedifferentiated self-renewing and differentiated migrating states are incompatible at the single cell level, though coexisting at the population level.
Collapse
|
37
|
Xia XD, Zhou Z, Yu XH, Zheng XL, Tang CK. Myocardin: A novel player in atherosclerosis. Atherosclerosis 2017; 257:266-278. [DOI: 10.1016/j.atherosclerosis.2016.12.002] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2016] [Revised: 11/29/2016] [Accepted: 12/01/2016] [Indexed: 12/21/2022]
|
38
|
Uremia modulates the phenotype of aortic smooth muscle cells. Atherosclerosis 2017; 257:64-70. [DOI: 10.1016/j.atherosclerosis.2016.12.022] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 12/02/2016] [Accepted: 12/21/2016] [Indexed: 12/12/2022]
|
39
|
Tuano NK, Okabe J, Ziemann M, Cooper ME, El-Osta A. Set7 mediated interactions regulate transcriptional networks in embryonic stem cells. Nucleic Acids Res 2016; 44:9206-9217. [PMID: 27439711 PMCID: PMC5100561 DOI: 10.1093/nar/gkw621] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Accepted: 07/01/2016] [Indexed: 12/17/2022] Open
Abstract
Histone methylation by lysine methyltransferase enzymes regulate the expression of genes implicated in lineage specificity and cellular differentiation. While it is known that Set7 catalyzes mono-methylation of histone and non-histone proteins, the functional importance of this enzyme in stem cell differentiation remains poorly understood. We show Set7 expression is increased during mouse embryonic stem cell (mESC) differentiation and is regulated by the pluripotency factors, Oct4 and Sox2. Transcriptional network analyses reveal smooth muscle (SM) associated genes are subject to Set7-mediated regulation. Furthermore, pharmacological inhibition of Set7 activity confirms this regulation. We observe Set7-mediated modification of serum response factor (SRF) and mono-methylation of histone H4 lysine 4 (H3K4me1) regulate gene expression. We conclude the broad substrate specificity of Set7 serves to control key transcriptional networks in embryonic stem cells.
Collapse
Affiliation(s)
- Natasha K Tuano
- Epigenetics in Human Health and Disease Laboratory, Baker IDI Heart and Diabetes Institute, The Alfred Medical Research and Education Precinct, Melbourne, Victoria 3004, Australia
| | - Jun Okabe
- Epigenetics in Human Health and Disease Laboratory, Baker IDI Heart and Diabetes Institute, The Alfred Medical Research and Education Precinct, Melbourne, Victoria 3004, Australia .,Faculty of Medicine, Nursing and Health Sciences, Monash University, Victoria, Australia
| | - Mark Ziemann
- Epigenetics in Human Health and Disease Laboratory, Baker IDI Heart and Diabetes Institute, The Alfred Medical Research and Education Precinct, Melbourne, Victoria 3004, Australia.,Epigenomic Profiling Facility, Baker IDI Heart and Diabetes Institute, The Alfred Medical Research and Education Precinct, Melbourne, Victoria 3004, Australia
| | - Mark E Cooper
- Junvenile Diabetes Research Foundation (JDRF) Danielle Alberti Centre for Diabetic Complications, Diabetes Division, Baker IDI Heart and Diabetes Institute, The Alfred Medical Research and Education Precinct, Melbourne, Victoria 3004, Australia
| | - Assam El-Osta
- Epigenetics in Human Health and Disease Laboratory, Baker IDI Heart and Diabetes Institute, The Alfred Medical Research and Education Precinct, Melbourne, Victoria 3004, Australia .,Faculty of Medicine, Nursing and Health Sciences, Monash University, Victoria, Australia.,Epigenomic Profiling Facility, Baker IDI Heart and Diabetes Institute, The Alfred Medical Research and Education Precinct, Melbourne, Victoria 3004, Australia.,Department of Pathology, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
40
|
Rodriguez A, Tripurani SK, Burton JC, Clementi C, Larina I, Pangas SA. SMAD Signaling Is Required for Structural Integrity of the Female Reproductive Tract and Uterine Function During Early Pregnancy in Mice. Biol Reprod 2016; 95:44. [PMID: 27335065 PMCID: PMC5029477 DOI: 10.1095/biolreprod.116.139477] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Accepted: 06/10/2016] [Indexed: 12/20/2022] Open
Abstract
Pregnancy is a complex physiological process tightly controlled by the interplay among hormones, morphogens, transcription factors, and signaling pathways. Although recent studies using genetically engineered mouse models have revealed that ligands and receptors of transforming growth factor beta (TGFbeta) and bone morphogenetic protein (BMP) signaling pathways are essential for multiple reproductive events during pregnancy, the functional role of SMAD transcription factors, which serve as the canonical signaling platform for the TGFbeta/BMP pathways, in the oviduct and uterus is undefined. Here, we used a mouse model containing triple conditional deletion of the BMP receptor signaling Smads (Smad1 and Smad5) and Smad4, the central mediator of both TGFbeta and BMP signaling, to investigate the role of the SMADs in reproductive tract structure and function in cells from the Amhr2 lineage. Unlike the respective single- or double-knockouts, female Smad1(flox/flox) Smad5(flox/flox) Smad4(flox/flox) Amhr2(cre/+)conditional knockout (i.e., Smad1/5/4-Amhr2-cre KO) mice are sterile. We discovered that Smad1/5/4-Amhr2-cre KO females have malformed oviducts that subsequently develop oviductal diverticuli. These oviducts showed dysregulation of multiple genes essential for oviduct and smooth muscle development. In addition, uteri from Smad1/5/4-Amhr2-cre KO females exhibit multiple defects in stroma, epithelium, and smooth muscle layers and fail to assemble a closed uterine lumen upon embryo implantation, with defective uterine decidualization that led to pregnancy loss at early to mid-gestation. Taken together, our study uncovers a new role for the SMAD transcription factors in maintaining the structural and functional integrity of oviduct and uterus, required for establishment and maintenance of pregnancy.
Collapse
Affiliation(s)
- Amanda Rodriguez
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas Graduate Program in Molecular and Cell Biology, Baylor College of Medicine, Houston, Texas
| | - Swamy K Tripurani
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas
| | - Jason C Burton
- Graduate Program in Integrative Molecular and Biomedical Sciences, Baylor College of Medicine, Houston, Texas
| | - Caterina Clementi
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas Graduate Program in Developmental Biology, Baylor College of Medicine, Houston, Texas
| | - Irina Larina
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas
| | - Stephanie A Pangas
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas Center for Reproductive Medicine, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
41
|
Frismantiene A, Dasen B, Pfaff D, Erne P, Resink TJ, Philippova M. T-cadherin promotes vascular smooth muscle cell dedifferentiation via a GSK3β-inactivation dependent mechanism. Cell Signal 2016; 28:516-530. [DOI: 10.1016/j.cellsig.2016.02.014] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 02/12/2016] [Accepted: 02/18/2016] [Indexed: 11/24/2022]
|
42
|
Coll-Bonfill N, de la Cruz-Thea B, Pisano MV, Musri MM. Noncoding RNAs in smooth muscle cell homeostasis: implications in phenotypic switch and vascular disorders. Pflugers Arch 2016; 468:1071-87. [PMID: 27109570 DOI: 10.1007/s00424-016-1821-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 04/04/2016] [Indexed: 12/16/2022]
Abstract
Vascular smooth muscle cells (SMC) are a highly specialized cell type that exhibit extraordinary plasticity in adult animals in response to a number of environmental cues. Upon vascular injury, SMC undergo phenotypic switch from a contractile-differentiated to a proliferative/migratory-dedifferentiated phenotype. This process plays a major role in vascular lesion formation and during the development of vascular remodeling. Vascular remodeling comprises the accumulation of dedifferentiated SMC in the intima of arteries and is central to a number of vascular diseases such as arteriosclerosis, chronic obstructive pulmonary disease or pulmonary hypertension. Therefore, it is critical to understand the molecular mechanisms that govern SMC phenotype. In the last decade, a number of new classes of noncoding RNAs have been described. These molecules have emerged as key factors controlling tissue homeostasis during physiological and pathological conditions. In this review, we will discuss the role of noncoding RNAs, including microRNAs and long noncoding RNAs, in the regulation of SMC plasticity.
Collapse
Affiliation(s)
- N Coll-Bonfill
- Department of Pulmonary Medicine Hospital Clínic-Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - B de la Cruz-Thea
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET, Universidad Nacional de Córdoba, Friuli 2434, 5016, Córdoba, Argentina
| | - M V Pisano
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET, Universidad Nacional de Córdoba, Friuli 2434, 5016, Córdoba, Argentina
| | - M M Musri
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET, Universidad Nacional de Córdoba, Friuli 2434, 5016, Córdoba, Argentina.
| |
Collapse
|
43
|
Nuclear PTEN functions as an essential regulator of SRF-dependent transcription to control smooth muscle differentiation. Nat Commun 2016; 7:10830. [PMID: 26940659 PMCID: PMC5411712 DOI: 10.1038/ncomms10830] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Accepted: 01/25/2016] [Indexed: 12/13/2022] Open
Abstract
Vascular disease progression is associated with marked changes in vascular smooth muscle cell (SMC) phenotype and function. SMC contractile gene expression and, thus differentiation, is under direct transcriptional control by the transcription factor, serum response factor (SRF); however, the mechanisms dynamically regulating SMC phenotype are not fully defined. Here we report that the lipid and protein phosphatase, PTEN, has a novel role in the nucleus by functioning as an indispensible regulator with SRF to maintain the differentiated SM phenotype. PTEN interacts with the N-terminal domain of SRF and PTEN–SRF interaction promotes SRF binding to essential promoter elements in SM-specific genes. Factors inducing phenotypic switching promote loss of nuclear PTEN through nucleo-cytoplasmic translocation resulting in reduced myogenically active SRF, but enhanced SRF activity on target genes involved in proliferation. Overall decreased expression of PTEN was observed in intimal SMCs of human atherosclerotic lesions underlying the potential clinical importance of these findings. The transcription factor, serum response factor, SRF regulates critical smooth muscle (SM) contractile gene expression but what else controls SM differentiation is unclear. Here, Horita et al. demonstrate that nuclear PTEN acts with SRF at the transcriptional level to maintain the differentiated SM phenotype.
Collapse
|
44
|
Ji H, Atchison L, Chen Z, Chakraborty S, Jung Y, Truskey GA, Christoforou N, Leong KW. Transdifferentiation of human endothelial progenitors into smooth muscle cells. Biomaterials 2016; 85:180-194. [PMID: 26874281 DOI: 10.1016/j.biomaterials.2016.01.066] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Revised: 01/23/2016] [Accepted: 01/28/2016] [Indexed: 12/17/2022]
Abstract
Access to smooth muscle cells (SMC) would create opportunities for tissue engineering, drug testing, and disease modeling. Herein we report the direct conversion of human endothelial progenitor cells (EPC) to induced smooth muscle cells (iSMC) by induced expression of MYOCD. The EPC undergo a cytoskeletal rearrangement resembling that of mesenchymal cells within 3 days post initiation of MYOCD expression. By day 7, the reprogrammed cells show upregulation of smooth muscle markers ACTA2, MYH11, and TAGLN by qRT-PCR and ACTA2 and MYH11 expression by immunofluorescence. By two weeks, they resemble umbilical artery SMC in microarray gene expression analysis. The iSMC, in contrast to EPC control, show calcium transients in response to phenylephrine stimulation and a contractility an order of magnitude higher than that of EPC as determined by traction force microscopy. Tissue-engineered blood vessels constructed using iSMC show functionality with respect to flow- and drug-mediated vasodilation and vasoconstriction.
Collapse
Affiliation(s)
- HaYeun Ji
- Department of Biomedical Engineering, Columbia University, Mail Code 8904, 1210 Amsterdam Avenue, New York, NY, 10027, USA
| | - Leigh Atchison
- Department of Biomedical Engineering, Duke University, 136 Hudson Hall, Box 90281, Durham, NC, 27708, USA
| | - Zaozao Chen
- Department of Biomedical Engineering, Columbia University, Mail Code 8904, 1210 Amsterdam Avenue, New York, NY, 10027, USA
| | - Syandan Chakraborty
- Department of Biomedical Engineering, Columbia University, Mail Code 8904, 1210 Amsterdam Avenue, New York, NY, 10027, USA
| | - Youngmee Jung
- Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, Korea
| | - George A Truskey
- Department of Biomedical Engineering, Duke University, 136 Hudson Hall, Box 90281, Durham, NC, 27708, USA
| | - Nicolas Christoforou
- Department of Biomedical Engineering, Duke University, 136 Hudson Hall, Box 90281, Durham, NC, 27708, USA.,Department of Biomedical Engineering, Khalifa University, P. O. Box 127788, Abu Dhabi, UAE
| | - Kam W Leong
- Department of Biomedical Engineering, Columbia University, Mail Code 8904, 1210 Amsterdam Avenue, New York, NY, 10027, USA
| |
Collapse
|
45
|
Basu S, Proweller A. Autoregulatory Control of Smooth Muscle Myosin Light Chain Kinase Promoter by Notch Signaling. J Biol Chem 2015; 291:2988-99. [PMID: 26703474 DOI: 10.1074/jbc.m115.679803] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2015] [Indexed: 11/06/2022] Open
Abstract
Smooth muscle myosin light chain kinase (SM-MLCK) is the key enzyme responsible for phosphorylation of regulatory myosin light chain (MLC20), resulting in actin-myosin cross-bridging and force generation in vascular smooth muscle required for physiological vasoreactivity and blood pressure control. In this study, we investigated the combinatorial role of myocardin/serum response factor (SRF) and Notch signaling in the transcriptional regulation of MLCK gene expression. Promoter reporter analyses in rat A10 smooth muscle cells revealed a bimodal pattern of MLCK promoter activity and gene expression upon stimulation with constitutively active Notch1 in presence of myocardin or by Jagged1 ligand stimulation. An initial Notch1-induced increase in MLCK transcription was followed by loss in promoter sensitivity, which could be restored with further Notch1 dose escalation. Real-time PCR analyses revealed that endogenous levels of Hairy Related Transcription (HRT) factor 2 (HRT2) peaked concurrently with inhibitory concentrations of Notch1. Forced expression of HRT2 demonstrated simultaneous repression of both myocardin- and Notch1-induced MLCK promoter activity. HRT2-mediated repression was further confirmed by HRT2 truncations and siHRT2 treatments that rescued MLCK promoter activity and gene expression. Chromatin immunoprecipitation studies revealed both Jagged1 ligand- and Notch1-enhanced myocardin/SRF complex formation at the promoter CArG element. In contrast, heightened levels of HRT2 concomitantly disrupted myocardin/SRF and Notch transcription complex formation at respective CArG and CSL binding elements. Taken together, SM-MLCK promoter activity appears highly sensitive to the relative levels of Notch1 signaling, HRT2, and myocardin. These findings identify a novel Notch-dependent HRT2 autoregulatory circuit coordinating transcriptional regulation of SM-MLCK.
Collapse
Affiliation(s)
- Sanchita Basu
- From the Department of Medicine, Case Cardiovascular Research Institute and University Hospitals Harrington Heart and Vascular Institute, Case Western Reserve University, Cleveland, Ohio 44106
| | - Aaron Proweller
- From the Department of Medicine, Case Cardiovascular Research Institute and University Hospitals Harrington Heart and Vascular Institute, Case Western Reserve University, Cleveland, Ohio 44106
| |
Collapse
|
46
|
Saddouk FZ, Sun LY, Liu YF, Jiang M, Singer DV, Backs J, Van Riper D, Ginnan R, Schwarz JJ, Singer HA. Ca2+/calmodulin-dependent protein kinase II-γ (CaMKIIγ) negatively regulates vascular smooth muscle cell proliferation and vascular remodeling. FASEB J 2015; 30:1051-64. [PMID: 26567004 DOI: 10.1096/fj.15-279158] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 10/28/2015] [Indexed: 01/15/2023]
Abstract
Vascular smooth muscle (VSM) expresses calcium/calmodulin-dependent protein kinase II (CaMKII)-δ and -γ isoforms. CaMKIIδ promotes VSM proliferation and vascular remodeling. We tested CaMKIIγ function in vascular remodeling after injury. CaMKIIγ protein decreased 90% 14 d after balloon injury in rat carotid artery. Intraluminal transduction of adenovirus encoding CaMKIIγC rescued expression to 35% of uninjured controls, inhibited neointima formation (>70%), inhibited VSM proliferation (>60%), and increased expression of the cell-cycle inhibitor p21 (>2-fold). Comparable doses of CaMKIIδ2 adenovirus had no effect. Similar dynamics in CaMKIIγ mRNA and protein expression were observed in ligated mouse carotid arteries, correlating closely with expression of VSM differentiation markers. Targeted deletion of CaMKIIγ in smooth muscle resulted in a 20-fold increase in neointimal area, with a 3-fold increase in the cell proliferation index, no change in apoptosis, and a 60% decrease in p21 expression. In cultured VSM, CaMKIIγ overexpression induced p53 mRNA (1.7 fold) and protein (1.8-fold) expression; induced the p53 target gene p21 (3-fold); decreased VSM cell proliferation (>50%); and had no effect on expression of apoptosis markers. We conclude that regulated CaMKII isoform composition is an important determinant of the injury-induced vasculoproliferative response and that CaMKIIγ and -δ isoforms have nonequivalent, opposing functions.
Collapse
Affiliation(s)
- Fatima Z Saddouk
- *Center for Cardiovascular Sciences, Albany Medical College, Albany, New York, USA; and Department of Cardiology, Angiology and Pneumology, University of Heidelberg, Heidelberg, Germany
| | - Li-Yan Sun
- *Center for Cardiovascular Sciences, Albany Medical College, Albany, New York, USA; and Department of Cardiology, Angiology and Pneumology, University of Heidelberg, Heidelberg, Germany
| | - Yong Feng Liu
- *Center for Cardiovascular Sciences, Albany Medical College, Albany, New York, USA; and Department of Cardiology, Angiology and Pneumology, University of Heidelberg, Heidelberg, Germany
| | - Miao Jiang
- *Center for Cardiovascular Sciences, Albany Medical College, Albany, New York, USA; and Department of Cardiology, Angiology and Pneumology, University of Heidelberg, Heidelberg, Germany
| | - Diane V Singer
- *Center for Cardiovascular Sciences, Albany Medical College, Albany, New York, USA; and Department of Cardiology, Angiology and Pneumology, University of Heidelberg, Heidelberg, Germany
| | - Johannes Backs
- *Center for Cardiovascular Sciences, Albany Medical College, Albany, New York, USA; and Department of Cardiology, Angiology and Pneumology, University of Heidelberg, Heidelberg, Germany
| | - Dee Van Riper
- *Center for Cardiovascular Sciences, Albany Medical College, Albany, New York, USA; and Department of Cardiology, Angiology and Pneumology, University of Heidelberg, Heidelberg, Germany
| | - Roman Ginnan
- *Center for Cardiovascular Sciences, Albany Medical College, Albany, New York, USA; and Department of Cardiology, Angiology and Pneumology, University of Heidelberg, Heidelberg, Germany
| | - John J Schwarz
- *Center for Cardiovascular Sciences, Albany Medical College, Albany, New York, USA; and Department of Cardiology, Angiology and Pneumology, University of Heidelberg, Heidelberg, Germany
| | - Harold A Singer
- *Center for Cardiovascular Sciences, Albany Medical College, Albany, New York, USA; and Department of Cardiology, Angiology and Pneumology, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
47
|
Rinaldi B, Finicelli M, Donniacuo M, Di Bernardo G, Gritti G, Gaudio SD, Forte A, Peluso G, Cipollaro M, Rossi F, Galderisi U. G-CSF contributes at the healing of tunica media of arteriotomy-injured rat carotids by promoting differentiation of vascular smooth muscle cells. J Cell Physiol 2015; 231:215-23. [DOI: 10.1002/jcp.25074] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Accepted: 06/08/2015] [Indexed: 12/24/2022]
Affiliation(s)
- Barbara Rinaldi
- Department of Experimental Medicine, Section of Pharmacology “L. Donatelli”, Centre of Excellence for Cardiovascular Diseases; Second University of Naples; Naples Italy
- Department of Experimental Medicine, Section of Pharmacology L. Donatelli, Regional Centre for Pharmacovigilance and Pharmacoepidemiology; Second University of Naples; Naples Italy
| | - Mauro Finicelli
- Institute of Biomedicine and Bioresources (IBBR); CNR; Naples Italy
| | - Maria Donniacuo
- Department of Experimental Medicine, Section of Pharmacology L. Donatelli, Regional Centre for Pharmacovigilance and Pharmacoepidemiology; Second University of Naples; Naples Italy
| | - Giovanni Di Bernardo
- Department of Experimental Medicine, Biotechnology, and Molecular Biology Section; Second University of Naples; Naples Italy
| | - Giulia Gritti
- Department of Experimental Medicine, Section of Pharmacology L. Donatelli, Regional Centre for Pharmacovigilance and Pharmacoepidemiology; Second University of Naples; Naples Italy
| | - Stefania Del Gaudio
- Department of Experimental Medicine, Biotechnology, and Molecular Biology Section; Second University of Naples; Naples Italy
| | - Amalia Forte
- Department of Experimental Medicine, Biotechnology, and Molecular Biology Section; Second University of Naples; Naples Italy
| | | | - Marilena Cipollaro
- Department of Experimental Medicine, Biotechnology, and Molecular Biology Section; Second University of Naples; Naples Italy
| | - Francesco Rossi
- Department of Experimental Medicine, Section of Pharmacology L. Donatelli, Regional Centre for Pharmacovigilance and Pharmacoepidemiology; Second University of Naples; Naples Italy
- Institute of Biomedicine and Bioresources (IBBR); CNR; Naples Italy
| | - Umberto Galderisi
- Department of Experimental Medicine, Biotechnology, and Molecular Biology Section; Second University of Naples; Naples Italy
| |
Collapse
|
48
|
Liao XH, Wang N, Zhao DW, Zheng DL, Zheng L, Xing WJ, Ma WJ, Bao LY, Dong J, Zhang TC. STAT3 Protein Regulates Vascular Smooth Muscle Cell Phenotypic Switch by Interaction with Myocardin. J Biol Chem 2015; 290:19641-52. [PMID: 26100622 DOI: 10.1074/jbc.m114.630111] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Indexed: 11/06/2022] Open
Abstract
The JAK-STAT3 signaling pathway is one of the critical pathways regulating cell proliferation, differentiation, and apoptosis. Myocardin is regarded as a key mediator for the change of smooth muscle phenotypes. However, the relationship between STAT3 and myocardin in the vascular smooth muscle cell (VSMC) phenotypic switch has not been investigated. The goal of this study was to investigate the molecular mechanism by which STAT3 affects the myocardin-regulated VSMC phenotypic switch. Data presented in this study demonstrated that STAT3 was rapidly up-regulated after stimulation with VEGF. Inhibition of the STAT3 activation process impaired VSMC proliferation and enhanced the expression of VSMC contractile genes by increasing serum-response factor binding to the CArG-containing regions of VSMC-specific contractile genes. In contrast, the interaction between serum-response factor and its co-activator myocardin was reduced by overexpression of STAT3. In addition, treated VEGF inhibited the transcription activity of myocardin, and overexpression of STAT3 inhibited myocardin-induced up-regulation of VSMC contractile phenotype-specific genes. Although myocardin and STAT3 are negatively correlated, interestingly, both of them can enhance the expression of VEGF, suggesting a feedback loop to regulate the VSMC phenotypic switch. Taken together, these results indicate that the JAK-STAT3 signaling pathway plays a key role in controlling the phenotypic switch of VSMCs through the interactions between STAT3 and myocardin by various coordinated gene regulation pathways and feedback loops.
Collapse
Affiliation(s)
- Xing-Hua Liao
- From the Institute of Biology and Medicine, Wuhan University of Science and Technology, Wuhan 430000 and the Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Nan Wang
- the Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Dong-Wei Zhao
- the Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - De-Liang Zheng
- the Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Li Zheng
- the Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Wen-Jing Xing
- the Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Wen-Jian Ma
- the Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Le-Yuan Bao
- From the Institute of Biology and Medicine, Wuhan University of Science and Technology, Wuhan 430000 and
| | - Jian Dong
- From the Institute of Biology and Medicine, Wuhan University of Science and Technology, Wuhan 430000 and
| | - Tong-Cun Zhang
- From the Institute of Biology and Medicine, Wuhan University of Science and Technology, Wuhan 430000 and the Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| |
Collapse
|
49
|
Ghosh S, Kollar B, Nahar T, Suresh Babu S, Wojtowicz A, Sticht C, Gretz N, Wagner AH, Korff T, Hecker M. Loss of the mechanotransducer zyxin promotes a synthetic phenotype of vascular smooth muscle cells. J Am Heart Assoc 2015; 4:e001712. [PMID: 26071033 PMCID: PMC4599528 DOI: 10.1161/jaha.114.001712] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Background Exposure of vascular smooth muscle cells (VSMCs) to excessive cyclic stretch such as in hypertension causes a shift in their phenotype. The focal adhesion protein zyxin can transduce such biomechanical stimuli to the nucleus of both endothelial cells and VSMCs, albeit with different thresholds and kinetics. However, there is no distinct vascular phenotype in young zyxin-deficient mice, possibly due to functional redundancy among other gene products belonging to the zyxin family. Analyzing zyxin function in VSMCs at the cellular level might thus offer a better mechanistic insight. We aimed to characterize zyxin-dependent changes in gene expression in VSMCs exposed to biomechanical stretch and define the functional role of zyxin in controlling the resultant VSMC phenotype. Methods and Results DNA microarray analysis was used to identify genes and pathways that were zyxin regulated in static and stretched human umbilical artery–derived and mouse aortic VSMCs. Zyxin-null VSMCs showed a remarkable shift to a growth-promoting, less apoptotic, promigratory and poorly contractile phenotype with ≈90% of the stretch-responsive genes being zyxin dependent. Interestingly, zyxin-null cells already seemed primed for such a synthetic phenotype, with mechanical stretch further accentuating it. This could be accounted for by higher RhoA activity and myocardin-related transcription factor-A mainly localized to the nucleus of zyxin-null VSMCs, and a condensed and localized accumulation of F-actin upon stretch. Conclusions At the cellular level, zyxin is a key regulator of stretch-induced gene expression. Loss of zyxin drives VSMCs toward a synthetic phenotype, a process further consolidated by exaggerated stretch.
Collapse
Affiliation(s)
- Subhajit Ghosh
- Institute of Physiology and Pathophysiology, University of Heidelberg, Germany (S.G., B.K., T.N., A.H.W., T.K.)
| | - Branislav Kollar
- Institute of Physiology and Pathophysiology, University of Heidelberg, Germany (S.G., B.K., T.N., A.H.W., T.K.)
| | - Taslima Nahar
- Institute of Physiology and Pathophysiology, University of Heidelberg, Germany (S.G., B.K., T.N., A.H.W., T.K.)
| | - Sahana Suresh Babu
- Department of Cardiovascular Regeneration, Houston Methodist Research Institute, Houston, TX (S.S.B.)
| | - Agnieszka Wojtowicz
- Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland (A.W.)
| | - Carsten Sticht
- ZMF, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany (C.S., N.G.)
| | - Norbert Gretz
- ZMF, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany (C.S., N.G.)
| | - Andreas H Wagner
- Institute of Physiology and Pathophysiology, University of Heidelberg, Germany (S.G., B.K., T.N., A.H.W., T.K.)
| | - Thomas Korff
- Institute of Physiology and Pathophysiology, University of Heidelberg, Germany (S.G., B.K., T.N., A.H.W., T.K.)
| | - Markus Hecker
- Institute of Physiology and Pathophysiology, University of Heidelberg and Deutsches Zentrum Für Herz-Kreislauf-Forschung E.V. (DZHK), Partner site Heidelberg/Mannheim, Germany (M.H.)
| |
Collapse
|
50
|
Miano JM, Long X. The short and long of noncoding sequences in the control of vascular cell phenotypes. Cell Mol Life Sci 2015; 72:3457-88. [PMID: 26022065 DOI: 10.1007/s00018-015-1936-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Revised: 05/21/2015] [Accepted: 05/22/2015] [Indexed: 12/13/2022]
Abstract
The two principal cell types of importance for normal vessel wall physiology are smooth muscle cells and endothelial cells. Much progress has been made over the past 20 years in the discovery and function of transcription factors that coordinate proper differentiation of these cells and the maintenance of vascular homeostasis. More recently, the converging fields of bioinformatics, genomics, and next generation sequencing have accelerated discoveries in a number of classes of noncoding sequences, including transcription factor binding sites (TFBS), microRNA genes, and long noncoding RNA genes, each of which mediates vascular cell differentiation through a variety of mechanisms. Alterations in the nucleotide sequence of key TFBS or deviations in transcription of noncoding RNA genes likely have adverse effects on normal vascular cell phenotype and function. Here, the subject of noncoding sequences that influence smooth muscle cell or endothelial cell phenotype will be summarized as will future directions to further advance our understanding of the increasingly complex molecular circuitry governing normal vascular cell differentiation and how such information might be harnessed to combat vascular diseases.
Collapse
Affiliation(s)
- Joseph M Miano
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, 601 Elmwood Avenue, Rochester, NY, 14642, USA,
| | | |
Collapse
|