151
|
Gualdrini F, Esnault C, Horswell S, Stewart A, Matthews N, Treisman R. SRF Co-factors Control the Balance between Cell Proliferation and Contractility. Mol Cell 2016; 64:1048-1061. [PMID: 27867007 PMCID: PMC5179500 DOI: 10.1016/j.molcel.2016.10.016] [Citation(s) in RCA: 125] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 09/26/2016] [Accepted: 10/11/2016] [Indexed: 12/20/2022]
Abstract
The ERK-regulated ternary complex factors (TCFs) act with the transcription factor serum response factor (SRF) to activate mitogen-induced transcription. However, the extent of their involvement in the immediate-early transcriptional response, and their wider functional significance, has remained unclear. We show that, in MEFs, TCF inactivation significantly inhibits over 60% of TPA-inducible gene transcription and impairs cell proliferation. Using integrated SRF ChIP-seq and Hi-C data, we identified over 700 TCF-dependent SRF direct target genes involved in signaling, transcription, and proliferation. These also include a significant number of cytoskeletal gene targets for the Rho-regulated myocardin-related transcription factor (MRTF) SRF cofactor family. The TCFs act as general antagonists of MRTF-dependent SRF target gene expression, competing directly with the MRTFs for access to SRF. As a result, TCF-deficient MEFs exhibit hypercontractile and pro-invasive behavior. Thus, competition between TCFs and MRTFs for SRF determines the balance between antagonistic proliferative and contractile programs of gene expression.
Collapse
Affiliation(s)
- Francesco Gualdrini
- Signalling and Transcription Group, Francis Crick Institute, 1 Midland Rd, London NW1 1AT, UK
| | - Cyril Esnault
- Signalling and Transcription Group, Francis Crick Institute, 1 Midland Rd, London NW1 1AT, UK
| | - Stuart Horswell
- Bioinformatics and Biostatistics STP, Francis Crick Institute, 1 Midland Rd, London NW1 1AT, UK
| | - Aengus Stewart
- Bioinformatics and Biostatistics STP, Francis Crick Institute, 1 Midland Rd, London NW1 1AT, UK
| | - Nik Matthews
- Advanced Sequencing STP, Francis Crick Institute, 1 Midland Rd, London NW1 1AT, UK
| | - Richard Treisman
- Signalling and Transcription Group, Francis Crick Institute, 1 Midland Rd, London NW1 1AT, UK.
| |
Collapse
|
152
|
Chettimada S, Joshi SR, Dhagia V, Aiezza A, Lincoln TM, Gupte R, Miano JM, Gupte SA. Vascular smooth muscle cell contractile protein expression is increased through protein kinase G-dependent and -independent pathways by glucose-6-phosphate dehydrogenase inhibition and deficiency. Am J Physiol Heart Circ Physiol 2016; 311:H904-H912. [PMID: 27521420 PMCID: PMC5114469 DOI: 10.1152/ajpheart.00335.2016] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Accepted: 07/06/2016] [Indexed: 11/22/2022]
Abstract
Homeostatic control of vascular smooth muscle cell (VSMC) differentiation is critical for contractile activity and regulation of blood flow. Recently, we reported that precontracted blood vessels are relaxed and the phenotype of VSMC is regulated from a synthetic to contractile state by glucose-6-phosphate dehydrogenase (G6PD) inhibition. In the current study, we investigated whether the increase in the expression of VSMC contractile proteins by inhibition and knockdown of G6PD is mediated through a protein kinase G (PKG)-dependent pathway and whether it regulates blood pressure. We found that the expression of VSMC-restricted contractile proteins, myocardin (MYOCD), and miR-1 and miR-143 are increased by G6PD inhibition or knockdown. Importantly, RNA-sequence analysis of aortic tissue from G6PD-deficient mice revealed uniform increases in VSMC-restricted genes, particularly those regulated by the MYOCD-serum response factor (SRF) switch. Conversely, expression of Krüppel-like factor 4 (KLF4) is decreased by G6PD inhibition. Interestingly, the G6PD inhibition-induced expression of miR-1 and contractile proteins was blocked by Rp-β-phenyl-1,N2-etheno-8-bromo-guanosine-3',5'-cyclic monophosphorothioate, a PKG inhibitor. On the other hand, MYOCD and miR-143 levels are increased by G6PD inhibition through a PKG-independent manner. Furthermore, blood pressure was lower in the G6PD-deficient compared with wild-type mice. Therefore, our results suggest that the expression of VSMC contractile proteins induced by G6PD inhibition occurs via PKG1α-dependent and -independent pathways.
Collapse
MESH Headings
- Animals
- Aorta/drug effects
- Aorta/metabolism
- Blotting, Western
- Cattle
- Chromatography, Liquid
- Contractile Proteins/drug effects
- Contractile Proteins/genetics
- Contractile Proteins/metabolism
- Cyclic GMP-Dependent Protein Kinase Type I/antagonists & inhibitors
- Cyclic GMP-Dependent Protein Kinase Type I/metabolism
- Cyclic GMP-Dependent Protein Kinases/antagonists & inhibitors
- Cyclic GMP-Dependent Protein Kinases/metabolism
- Gene Knockdown Techniques
- Glucosephosphate Dehydrogenase/antagonists & inhibitors
- Glucosephosphate Dehydrogenase/genetics
- Immunoprecipitation
- Kruppel-Like Factor 4
- Kruppel-Like Transcription Factors/drug effects
- Kruppel-Like Transcription Factors/genetics
- Kruppel-Like Transcription Factors/metabolism
- Mice
- MicroRNAs/drug effects
- MicroRNAs/genetics
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Nuclear Proteins/drug effects
- Nuclear Proteins/genetics
- Nuclear Proteins/metabolism
- Polymerase Chain Reaction
- Rats
- Serum Response Factor/drug effects
- Serum Response Factor/genetics
- Serum Response Factor/metabolism
- Tandem Mass Spectrometry
- Trans-Activators/drug effects
- Trans-Activators/genetics
- Trans-Activators/metabolism
Collapse
Affiliation(s)
- Sukrutha Chettimada
- Biochemistry & Molecular Biology, University of South Alabama, Mobile, Alabama; Pharmacology, New York Medical College, Valhalla, New York
| | - Sachindra Raj Joshi
- Biochemistry & Molecular Biology, University of South Alabama, Mobile, Alabama; Pharmacology, New York Medical College, Valhalla, New York
| | - Vidhi Dhagia
- Pharmacology, New York Medical College, Valhalla, New York
| | - Alessandro Aiezza
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, New York; and
| | | | - Rakhee Gupte
- Biochemistry & Molecular Biology, University of South Alabama, Mobile, Alabama; Pharmacology, New York Medical College, Valhalla, New York
| | - Joseph M Miano
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, New York; and
| | - Sachin A Gupte
- Biochemistry & Molecular Biology, University of South Alabama, Mobile, Alabama; Pharmacology, New York Medical College, Valhalla, New York
| |
Collapse
|
153
|
Krawczyk KK, Ekman M, Rippe C, Grossi M, Nilsson BO, Albinsson S, Uvelius B, Swärd K. Assessing the contribution of thrombospondin-4 induction and ATF6α activation to endoplasmic reticulum expansion and phenotypic modulation in bladder outlet obstruction. Sci Rep 2016; 6:32449. [PMID: 27581066 PMCID: PMC5007532 DOI: 10.1038/srep32449] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 08/08/2016] [Indexed: 12/15/2022] Open
Abstract
Phenotypic modulation of smooth muscle cells is a hallmark of disease. The associated expansion of endoplasmic reticulum (ER) volume remains unexplained. Thrombospondin-4 was recently found to promote ATF6α activation leading to ER expansion. Using bladder outlet obstruction as a paradigm for phenotypic modulation, we tested if thrombospondin-4 is induced in association with ATF6α activation and ER expansion. Thrombospondin-4 was induced and ATF6α was activated after outlet obstruction in rodents. Increased abundance of spliced of Xbp1, another ER-stress sensor, and induction of Atf4 and Creb3l2 was also seen. Downstream of ATF6α, Calr, Manf, Sdf2l1 and Pdi increased as did ER size, whereas contractile markers were reduced. Overexpression of ATF6α, but not of thrombospondin-4, increased Calr, Manf, Sdf2l1 and Pdi and caused ER expansion, but the contractile markers were inert. Knockout of thrombospondin-4 neither affected bladder growth nor expression of ATF6α target genes, and repression of contractile markers was the same, even if ATF6α activation was curtailed. Increases of Xbp1s, Atf4 and Creb3l2 were similar. Our findings demonstrate reciprocal regulation of the unfolded protein response, including ATF6α activation and ER expansion, and reduced contractile differentiation in bladder outlet obstruction occurring independently of thrombospondin-4, which however is a sensitive indicator of obstruction.
Collapse
Affiliation(s)
| | - Mari Ekman
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Catarina Rippe
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Mario Grossi
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Bengt-Olof Nilsson
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | | | - Bengt Uvelius
- Department of Urology, Clinical Sciences, Lund University, Lund, Sweden
| | - Karl Swärd
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| |
Collapse
|
154
|
Stress-dependent phosphorylation of myocardin-related transcription factor A (MRTF-A) by the p38(MAPK)/MK2 axis. Sci Rep 2016; 6:31219. [PMID: 27492266 PMCID: PMC4974569 DOI: 10.1038/srep31219] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Accepted: 07/14/2016] [Indexed: 12/23/2022] Open
Abstract
Myocardin-related transcription factor A (MRTF-A) is a known actin-regulated transcriptional coactivator of serum response factor (SRF). Stimulation of actin polymerization activates MRTF-A by releasing it from G-actin and thus allowing it to bind to and activate SRF. Here, we compared protein phosphorylation in MK2/3-deficient cells rescued or not by ectopic expression of MK2 in two independent phosphoproteomic approaches using anisomycin-treated MEF cells and LPS-stimulated mouse macrophages, respectively. Two MRTF-A sites, Ser351 (corresponding to Ser312 in human) and Ser371 (Ser333 in human), showed significantly stronger phosphorylation (12-fold and 6-fold increase) in the cells expressing MK2. MRTF-A is phosphorylated at these sites in a stress-, but not in a mitogen-induced manner, and p38MAPK/MK2 catalytic activities are indispensable for this phosphorylation. MK2-mediated phosphorylation of MRTF-A at Ser312 and Ser333 was further confirmed in an in vitro kinase assay and using the phospho-protein kinase-D (PKD)-consensus motif antibody (anti-LXRXXpS/pT), the p38MAPK inhibitor BIRB-796, MK2/3-deficient cells and MRTF-A phospho-site mutants. Unexpectedly, dimerization, subcellular localization and translocation, interaction with actin, SRF or SMAD3 and transactivating potential of MRTF-A seem to be unaffected by manipulating the p38MAPK/MK2-dependent phosphorylations. Hence, MRTF-A is stress-dependently phosphorylated by MK2 at Ser312 and Ser333 with so far undetected functional and physiological consequences.
Collapse
|
155
|
Differentiation of Human Adipose Derived Stem Cells into Smooth Muscle Cells Is Modulated by CaMKIIγ. Stem Cells Int 2016; 2016:1267480. [PMID: 27493668 PMCID: PMC4963582 DOI: 10.1155/2016/1267480] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 06/02/2016] [Accepted: 06/07/2016] [Indexed: 12/13/2022] Open
Abstract
The multifunctional Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) is known to participate in maintenance and switches of smooth muscle cell (SMC) phenotypes. However, which isoform of CaMKII is involved in differentiation of adult mesenchymal stem cells into contractile SMCs remains unclear. In the present study, we detected γ isoform of CaMKII in differentiation of human adipose derived stem cells (hASCs) into SMCs that resulted from treatment with TGF-β1 and BMP4 in combination for 7 days. The results showed that CaMKIIγ increased gradually during differentiation of hASCs as determined by real-time PCR and western blot analysis. The siRNA-mediated knockdown of CaMKIIγ decreased the protein levels and transcriptional levels of smooth muscle contractile markers (a-SMA, SM22a, calponin, and SM-MHC), while CaMKIIγ overexpression increases the transcriptional and protein levels of smooth muscle contractile markers. These results suggested that γ isoform of CaMKII plays a significant role in smooth muscle differentiation of hASCs.
Collapse
|
156
|
Qiao Y, Tang C, Wang Q, Wang D, Yan G, Zhu B. Kir2.1 regulates rat smooth muscle cell proliferation, migration, and post-injury carotid neointimal formation. Biochem Biophys Res Commun 2016; 477:774-780. [PMID: 27387235 DOI: 10.1016/j.bbrc.2016.06.134] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2016] [Accepted: 06/26/2016] [Indexed: 12/15/2022]
Abstract
Phenotype switching of vascular smooth muscle cells (VSMC) from the contractile type to the synthetic type is a hallmark of vascular disorders such as atherosclerosis and restenosis after angioplasty. Inward rectifier K(+) channel 2.1 (Kir2.1) has been identified in VSMC. However, whether it plays a functional role in regulating cellular transformation remains obscure. In this study, we evaluated the role of Kir2.1 on VSMC proliferation, migration, phenotype switching, and post-injury carotid neointimal formation. Kir2.1 knockdown significantly suppressed platelet-derived growth factor BB-stimulated rat vascular smooth muscle cells (rat-VSMC) proliferation and migration. Deficiency in Kir2.1 contributed to the restoration of smooth muscle α-actin, smooth muscle 22α, and calponin and to a reduction in osteopontin expression in rat-VSMC. Moreover, the in vivo study showed that rat-VSMC switched to proliferative phenotypes and that knockdown of Kir2.1 significantly inhibited neointimal formation after rat carotid injury. Kir2.1 may be a potential therapeutic target in the treatment of cardiovascular diseases, such as atherosclerosis and restenosis following percutaneous coronary intervention.
Collapse
Affiliation(s)
- Yong Qiao
- Department of Cardiology, Zhongda Hospital of Southeast University Medical School, Nanjing, Jiangsu, China
| | - Chengchun Tang
- Department of Cardiology, Zhongda Hospital of Southeast University Medical School, Nanjing, Jiangsu, China.
| | - Qingjie Wang
- Department of Cardiology, Zhongda Hospital of Southeast University Medical School, Nanjing, Jiangsu, China
| | - Dong Wang
- Department of Cardiology, Zhongda Hospital of Southeast University Medical School, Nanjing, Jiangsu, China
| | - Gaoliang Yan
- Department of Cardiology, Zhongda Hospital of Southeast University Medical School, Nanjing, Jiangsu, China
| | - Boqian Zhu
- Department of Cardiology, Zhongda Hospital of Southeast University Medical School, Nanjing, Jiangsu, China
| |
Collapse
|
157
|
Matsumoto Y, Itami S, Kuroda M, Yoshizato K, Kawada N, Murakami Y. MiR-29a Assists in Preventing the Activation of Human Stellate Cells and Promotes Recovery From Liver Fibrosis in Mice. Mol Ther 2016; 24:1848-1859. [PMID: 27480597 DOI: 10.1038/mt.2016.127] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 06/02/2016] [Indexed: 12/22/2022] Open
Abstract
The microRNA-29 (miR-29) family is known to suppress the activation of hepatic stellate cells (HSCs) and reversibly control liver fibrosis; however, the mechanism of how miR-29a controls liver fibrosis remains largely unknown. This study was conducted to clarify the mechanism of anti-fibrotic effect of miR-29a and to explore if miR-29a is a promising candidate for nucleic acid medicine against liver fibrosis. Two liver fibrosis murine models (carbon tetrachloride or thioacetamide) were used. MiR-29a mixed with atelocollagen was systemically administered. Hepatic fibrosis was evaluated by histological analysis and the expression levels of fibrosis-related genes. We observed that miR-29a treatment dramatically accelerated the reversion of liver fibrosis in vivo. Additionally, miR-29a regulated the mRNA expression of collagen type I alpha 1 (COL1A1) and platelet-derived growth factor C (PDGFC). We also noted that miR-29a significantly suppressed COL1A1 mRNA expression and cell viability and significantly increased caspase-9 activity (P < 0.05) in LX-2 cells. Pretreatment of miR-29a inhibited activation of LX-2 cell by transforming growth factor beta treatment. MiR-29a exhibited anti-fibrotic effect without cell toxicity in vivo and directly suppressed the expression of PDGF-related genes as well as COL1A1 and induced apoptosis of LX-2 cells. MiR-29a is a promising nucleic acid inhibitor to target liver fibrosis.
Collapse
Affiliation(s)
- Yoshinari Matsumoto
- Department of Hepatology, Graduate School of Medicine, Osaka City University, Osaka, Japan.,Department of Medical Nutrition, Graduate School of Human Life Science, Osaka City University, Osaka, Japan.,Current address: Department of Nutrition Management, Osaka University Medical Hospital, Osaka, Japan
| | - Saori Itami
- Department of Hepatology, Graduate School of Medicine, Osaka City University, Osaka, Japan
| | - Masahiko Kuroda
- Department of Molecular Pathology, Tokyo Medical University, Tokyo, Japan
| | | | - Norifumi Kawada
- Department of Hepatology, Graduate School of Medicine, Osaka City University, Osaka, Japan
| | - Yoshiki Murakami
- Department of Hepatology, Graduate School of Medicine, Osaka City University, Osaka, Japan
| |
Collapse
|
158
|
Sahoo S, Meijles DN, Al Ghouleh I, Tandon M, Cifuentes-Pagano E, Sembrat J, Rojas M, Goncharova E, Pagano PJ. MEF2C-MYOCD and Leiomodin1 Suppression by miRNA-214 Promotes Smooth Muscle Cell Phenotype Switching in Pulmonary Arterial Hypertension. PLoS One 2016; 11:e0153780. [PMID: 27144530 PMCID: PMC4856285 DOI: 10.1371/journal.pone.0153780] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2015] [Accepted: 04/04/2016] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Vascular hyperproliferative disorders are characterized by excessive smooth muscle cell (SMC) proliferation leading to vessel remodeling and occlusion. In pulmonary arterial hypertension (PAH), SMC phenotype switching from a terminally differentiated contractile to synthetic state is gaining traction as our understanding of the disease progression improves. While maintenance of SMC contractile phenotype is reportedly orchestrated by a MEF2C-myocardin (MYOCD) interplay, little is known regarding molecular control at this nexus. Moreover, the burgeoning interest in microRNAs (miRs) provides the basis for exploring their modulation of MEF2C-MYOCD signaling, and in turn, a pro-proliferative, synthetic SMC phenotype. We hypothesized that suppression of SMC contractile phenotype in pulmonary hypertension is mediated by miR-214 via repression of the MEF2C-MYOCD-leiomodin1 (LMOD1) signaling axis. METHODS AND RESULTS In SMCs isolated from a PAH patient cohort and commercially obtained hPASMCs exposed to hypoxia, miR-214 expression was monitored by qRT-PCR. miR-214 was upregulated in PAH- vs. control subject hPASMCs as well as in commercially obtained hPASMCs exposed to hypoxia. These increases in miR-214 were paralleled by MEF2C, MYOCD and SMC contractile protein downregulation. Of these, LMOD1 and MEF2C were directly targeted by the miR. Mir-214 overexpression mimicked the PAH profile, downregulating MEF2C and LMOD1. AntagomiR-214 abrogated hypoxia-induced suppression of the contractile phenotype and its attendant proliferation. Anti-miR-214 also restored PAH-PASMCs to a contractile phenotype seen during vascular homeostasis. CONCLUSIONS Our findings illustrate a key role for miR-214 in modulation of MEF2C-MYOCD-LMOD1 signaling and suggest that an antagonist of miR-214 could mitigate SMC phenotype changes and proliferation in vascular hyperproliferative disorders including PAH.
Collapse
Affiliation(s)
- Sanghamitra Sahoo
- Department of Pharmacology and Chemical Biology University of Pittsburgh, Pittsburgh, Pennsylvania, 15261, United States of America
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, 15261, United States of America
| | - Daniel N. Meijles
- Department of Pharmacology and Chemical Biology University of Pittsburgh, Pittsburgh, Pennsylvania, 15261, United States of America
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, 15261, United States of America
| | - Imad Al Ghouleh
- Department of Pharmacology and Chemical Biology University of Pittsburgh, Pittsburgh, Pennsylvania, 15261, United States of America
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, 15261, United States of America
| | - Manuj Tandon
- Department of Pharmacology and Chemical Biology University of Pittsburgh, Pittsburgh, Pennsylvania, 15261, United States of America
| | - Eugenia Cifuentes-Pagano
- Department of Pharmacology and Chemical Biology University of Pittsburgh, Pittsburgh, Pennsylvania, 15261, United States of America
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, 15261, United States of America
| | - John Sembrat
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, 15261, United States of America
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, 15261, United States of America
| | - Mauricio Rojas
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, 15261, United States of America
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, 15261, United States of America
| | - Elena Goncharova
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, 15261, United States of America
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, 15261, United States of America
| | - Patrick J. Pagano
- Department of Pharmacology and Chemical Biology University of Pittsburgh, Pittsburgh, Pennsylvania, 15261, United States of America
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, 15261, United States of America
- * E-mail:
| |
Collapse
|
159
|
Nazari M, Ni NC, Lüdke A, Li SH, Guo J, Weisel RD, Li RK. Mast cells promote proliferation and migration and inhibit differentiation of mesenchymal stem cells through PDGF. J Mol Cell Cardiol 2016; 94:32-42. [PMID: 26996757 DOI: 10.1016/j.yjmcc.2016.03.007] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Revised: 01/12/2016] [Accepted: 03/15/2016] [Indexed: 01/31/2023]
Abstract
BACKGROUND Mast cells (MCs) dynamically participate in wound healing after myocardial infarction (MI) by releasing cytokines. Indeed, MC-deficient mice undergo rapid left ventricular dilation post-MI. Mesenchymal stem cells (MSCs) are recruited to the injured region following an MI and have potential for cardiac repair. In the current study, we evaluate the effect of MCs on MSC proliferation and myogenic differentiation. METHODS AND RESULTS MCs were cultured from mouse bone marrow and MC granulate (MCG) was extracted from MCs via freeze-thaw cycles followed by filtration. α-SMA (smooth muscle actin) expression was examined as an indicator of myogenic differentiation. MSC/MC co-culture resulted in decreased MSC differentiation indicated by α-SMA suppression in MSCs. MCG also suppressed α-SMA expression and increased MSC migration and proliferation in a dose-dependent manner. Removal of MCG rescued α-SMA expression and MSC differentiation. Platelet derived growth factor (PDGF) receptor blockade using AG1296 also rescued MSC differentiation even after MCG treatment. Real-time PCR and Western blot showed that MCG exerted its effects on MSCs via downregulation of miR-145 and miR-143, downregulation of myocardin, upregulation of Klf4, and increased Erk and Elk1 phosphorylation. CONCLUSIONS MCs promote MSC proliferation and migration by suppressing their myogenic differentiation. MCs accomplish this via activation of the PDGF pathway, downregulation of miR-145/143, and modulation of the myocardin-Klf4 axis. These data suggest a potential role for MSC/MC interaction in the infarcted heart where MCs may inhibit MSCs from differentiation and promote their proliferation whereby increased cardiac MSC accumulation promotes eventual cardiac regeneration after MCs cease activity.
Collapse
Affiliation(s)
- Mansoreh Nazari
- Toronto General Research Institute, University Health Network, Division of Cardiovascular Surgery, Toronto, Ontario, Canada
| | - Nathan C Ni
- Toronto General Research Institute, University Health Network, Division of Cardiovascular Surgery, Toronto, Ontario, Canada
| | - Ana Lüdke
- Toronto General Research Institute, University Health Network, Division of Cardiovascular Surgery, Toronto, Ontario, Canada
| | - Shu-Hong Li
- Toronto General Research Institute, University Health Network, Division of Cardiovascular Surgery, Toronto, Ontario, Canada
| | - Jian Guo
- Toronto General Research Institute, University Health Network, Division of Cardiovascular Surgery, Toronto, Ontario, Canada
| | - Richard D Weisel
- Toronto General Research Institute, University Health Network, Division of Cardiovascular Surgery, Toronto, Ontario, Canada; Department of Surgery, Division of Cardiac Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Ren-Ke Li
- Toronto General Research Institute, University Health Network, Division of Cardiovascular Surgery, Toronto, Ontario, Canada; Department of Surgery, Division of Cardiac Surgery, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
160
|
Ro S. Multi-phenotypic Role of Serum Response Factor in the Gastrointestinal System. J Neurogastroenterol Motil 2016; 22:193-200. [PMID: 26727951 PMCID: PMC4819857 DOI: 10.5056/jnm15183] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 12/26/2015] [Indexed: 12/12/2022] Open
Abstract
Serum response factor (SRF) is a master transcription factor of the actin cytoskeleton that binds to highly conserved CArG boxes located within the majority of smooth muscle cell (SMC)-restricted promoters/enhancers. Although most studies of SRF focus on skeletal muscle, cardiac muscle, and vascular SMCs, SRF research has recently expanded into the gastrointestinal (GI) system. Genome scale analyses of GI SMC transcriptome and CArG boxes (CArGome) have identified new SRF target genes. In addition to circular and longitudinal smooth muscle layers, SRF is also expressed in GI mucosa and cancers. In the GI tract, SRF is the central regulator of genes involved in apoptosis, dedifferentiation, proliferation, and migration of cells. Since SRF is the cell phenotypic modulator, it may play an essential role in the development of myopathy, hypertrophy, ulcers, gastric and colon cancers within the GI tract. Given the multifunctional role displayed by SRF in the digestive system, SRF has received more attention emerging as a potential therapeutic target. This review summarizes the findings in SRF research pertaining to the GI tract and provides valuable insight into future directions.
Collapse
Affiliation(s)
- Seungil Ro
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV,
USA
| |
Collapse
|
161
|
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: 2.7] [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
|
162
|
Hoon JL, Tan MH, Koh CG. The Regulation of Cellular Responses to Mechanical Cues by Rho GTPases. Cells 2016; 5:cells5020017. [PMID: 27058559 PMCID: PMC4931666 DOI: 10.3390/cells5020017] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 03/29/2016] [Accepted: 03/30/2016] [Indexed: 12/21/2022] Open
Abstract
The Rho GTPases regulate many cellular signaling cascades that modulate cell motility, migration, morphology and cell division. A large body of work has now delineated the biochemical cues and pathways, which stimulate the GTPases and their downstream effectors. However, cells also respond exquisitely to biophysical and mechanical cues such as stiffness and topography of the extracellular matrix that profoundly influence cell migration, proliferation and differentiation. As these cellular responses are mediated by the actin cytoskeleton, an involvement of Rho GTPases in the transduction of such cues is not unexpected. In this review, we discuss an emerging role of Rho GTPase proteins in the regulation of the responses elicited by biophysical and mechanical stimuli.
Collapse
Affiliation(s)
- Jing Ling Hoon
- School of Biological Sciences, Nanyang Technological University, Singapore 637551, Singapore.
| | - Mei Hua Tan
- School of Biological Sciences, Nanyang Technological University, Singapore 637551, Singapore.
| | - Cheng-Gee Koh
- School of Biological Sciences, Nanyang Technological University, Singapore 637551, Singapore.
- Mechanobiology Institute, Singapore 117411, Singapore.
| |
Collapse
|
163
|
Tizioto PC, Coutinho LL, Mourão GB, Gasparin G, Malagó-Jr W, Bressani FA, Tullio RR, Nassu RT, Taylor JF, Regitano LCA. Variation inmyogenic differentiation 1mRNA abundance is associated with beef tenderness in Nelore cattle. Anim Genet 2016; 47:491-4. [DOI: 10.1111/age.12434] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/10/2016] [Indexed: 01/12/2023]
Affiliation(s)
| | - L. L. Coutinho
- Department of Animal Science; University of São Paulo/ESALQ; Piracicaba SP Brazil
| | - G. B. Mourão
- Department of Animal Science; University of São Paulo/ESALQ; Piracicaba SP Brazil
| | - G. Gasparin
- Department of Animal Science; University of São Paulo/ESALQ; Piracicaba SP Brazil
| | - W. Malagó-Jr
- Embrapa Southeast Livestock; São Carlos SP Brazil
| | | | - R. R. Tullio
- Embrapa Southeast Livestock; São Carlos SP Brazil
| | - R. T. Nassu
- Embrapa Southeast Livestock; São Carlos SP Brazil
| | - J. F. Taylor
- Division of Animal Sciences; University of Missouri; Columbia MO USA
| | | |
Collapse
|
164
|
Gurdziel K, Vogt KR, Walton KD, Schneider GK, Gumucio DL. Transcriptome of the inner circular smooth muscle of the developing mouse intestine: Evidence for regulation of visceral smooth muscle genes by the hedgehog target gene, cJun. Dev Dyn 2016; 245:614-26. [PMID: 26930384 DOI: 10.1002/dvdy.24399] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Revised: 01/29/2016] [Accepted: 02/16/2016] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Digestion is facilitated by coordinated contractions of the intestinal muscularis externa, a bilayered smooth muscle structure that is composed of inner circular muscles (ICM) and outer longitudinal muscles (OLM). We performed transcriptome analysis of intestinal mesenchyme tissue at E14.5, when the ICM, but not the OLM, is present, to investigate the transcriptional program of the ICM. RESULTS We identified 3967 genes enriched in E14.5 intestinal mesenchyme. The gene expression profiles were clustered and annotated to known muscle genes, identifying a muscle-enriched subcluster. Using publically available in situ data, 127 genes were verified as expressed in ICM. Examination of the promoter and regulatory regions for these co-expressed genes revealed enrichment for cJUN transcription factor binding sites, and cJUN protein was enriched in ICM. cJUN ChIP-seq, performed at E14.5, revealed that cJUN regulatory regions contain characteristics of muscle enhancers. Finally, we show that cJun is a target of Hedgehog (Hh), a signaling pathway known to be important in smooth muscle development, and identify a cJun genomic enhancer that is responsive to Hh. CONCLUSIONS This work provides the first transcriptional catalog for the developing ICM and suggests that cJun regulates gene expression in the ICM downstream of Hh signaling. Developmental Dynamics 245:614-626, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Katherine Gurdziel
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI, 48109.,Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, 48109
| | - Kyle R Vogt
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI, 48109
| | - Katherine D Walton
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI, 48109
| | - Gary K Schneider
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI, 48109
| | - Deborah L Gumucio
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI, 48109
| |
Collapse
|
165
|
Zhang YN, Xie BD, Sun L, Chen W, Jiang SL, Liu W, Bian F, Tian H, Li RK. Phenotypic switching of vascular smooth muscle cells in the 'normal region' of aorta from atherosclerosis patients is regulated by miR-145. J Cell Mol Med 2016; 20:1049-61. [PMID: 26992033 PMCID: PMC4882986 DOI: 10.1111/jcmm.12825] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 02/04/2016] [Indexed: 12/22/2022] Open
Abstract
Switching of vascular smooth muscle cells (VSMCs) from a contractile phenotype to an adverse proliferative phenotype is a hallmark of atherosclerosis or vascular restenosis. However, the genetic modulators responsible for this switch have not been fully elucidated in humans nor have they been correlated with clinical abnormalities. This study investigated genetic mechanisms involved in phenotypic switching of VSMCs at non-defect areas of the aorta in patients with atherosclerosis. Aortic wall samples were obtained from patients with (N = 53) and without (N = 27) atherosclerosis undergoing cardiovascular surgery. Vascular smooth muscle cell cultures were generated, and expression of microRNA-145 (miR-145), its target gene Kruppel-Like Factor 5 (KLF5) and Myocardin (MYOCD, a smooth muscle-specific transcriptional coactivator) were analysed using RT-qPCR, along with expression of relevant proteins. Vascular smooth muscle cells were transduced with miR-145 inhibitor and mimic to determine the effect of miR-145 expression on VSMC proliferation. miR-145 expression decreased while KLF5 expression increased in atherosclerotic aortas. Atherosclerotic samples and VSMCs had decreased expression of contractile markers calponin and alpha smooth muscle actin (α-SMA) and MYOCD. miR-145 inhibitor-transduced VSMCs from non-atherosclerotic patients showed decreased expression of calponin and α-SMA and increased proliferation compared with non-transduced controls, and these levels were close to those of atherosclerotic patients. miR-145 mimic-transduced VSMCs from atherosclerotic patients showed increased expression of calponin and α-SMA and decreased proliferation compared with non-transduced controls, and these levels were close to those found in non-atherosclerotic patients. These data demonstrate that miR-145 modulates the phenotypic switch of VSMCs from a contractile to a proliferative state via KLF5 and MYOCD in atherosclerosis.
Collapse
Affiliation(s)
- Yu-Nan Zhang
- Department of Cardiovascular Surgery, Second Affiliated Hospital of Harbin Medical University, Harbin, China.,Key Laboratories of Myocardial Ischemia Mechanism and Treatment, Harbin Medical University, Ministry of Education, Harbin, China
| | - Bao-Dong Xie
- Department of Cardiovascular Surgery, Second Affiliated Hospital of Harbin Medical University, Harbin, China.,Key Laboratories of Myocardial Ischemia Mechanism and Treatment, Harbin Medical University, Ministry of Education, Harbin, China
| | - Lu Sun
- Department of Cardiovascular Surgery, Second Affiliated Hospital of Harbin Medical University, Harbin, China.,Key Laboratories of Myocardial Ischemia Mechanism and Treatment, Harbin Medical University, Ministry of Education, Harbin, China
| | - Wei Chen
- Department of Cardiovascular Surgery, Second Affiliated Hospital of Harbin Medical University, Harbin, China.,Key Laboratories of Myocardial Ischemia Mechanism and Treatment, Harbin Medical University, Ministry of Education, Harbin, China
| | - Shu-Lin Jiang
- Department of Cardiovascular Surgery, Second Affiliated Hospital of Harbin Medical University, Harbin, China.,Key Laboratories of Myocardial Ischemia Mechanism and Treatment, Harbin Medical University, Ministry of Education, Harbin, China
| | - Wei Liu
- Key Laboratories of Myocardial Ischemia Mechanism and Treatment, Harbin Medical University, Ministry of Education, Harbin, China
| | - Fei Bian
- Key Laboratories of Myocardial Ischemia Mechanism and Treatment, Harbin Medical University, Ministry of Education, Harbin, China
| | - Hai Tian
- Department of Cardiovascular Surgery, Second Affiliated Hospital of Harbin Medical University, Harbin, China.,Key Laboratories of Myocardial Ischemia Mechanism and Treatment, Harbin Medical University, Ministry of Education, Harbin, China
| | - Ren-Ke Li
- Toronto General Research Institute, University Health Network and Department of Surgery, Division of Cardiac Surgery, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
166
|
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: 57] [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
|
167
|
Eschrich J, Meyer R, Kuk H, Wagner AH, Noppeney T, Debus S, Hecker M, Korff T. Varicose Remodeling of Veins Is Suppressed by 3-Hydroxy-3-Methylglutaryl Coenzyme A Reductase Inhibitors. J Am Heart Assoc 2016; 5:e002405. [PMID: 26908399 PMCID: PMC4802467 DOI: 10.1161/jaha.115.002405] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 01/07/2016] [Indexed: 11/16/2022]
Abstract
BACKGROUND Despite the high prevalence of chronic venous insufficiency and varicose veins in the Western world, suitable pharmaceutical therapies for these venous diseases have not been explored to date. In this context, we recently reported that a chronic increase in venous wall stress or biomechanical stretch is sufficient to cause development of varicose veins through the activation of the transcription factor activator protein 1. METHODS AND RESULTS We investigated whether deleterious venous remodeling is suppressed by the pleiotropic effects of statins. In vitro, activator protein 1 activity was inhibited by two 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors, rosuvastatin and atorvastatin, in stretch-stimulated human venous smooth muscle cells. Correspondingly, both statins inhibited venous smooth muscle cell proliferation as well as mRNA expression of the activator protein 1 target gene monocyte chemotactic protein 1 (MCP1). In isolated mouse veins exposed to an increased level of intraluminal pressure, statin treatment diminished proliferation of venous smooth muscle cells and protein abundance of MCP1 while suppressing the development of varicose veins in a corresponding animal model by almost 80%. Further analyses of human varicose vein samples from patients chronically treated with statins indicated a decrease in venous smooth muscle cell proliferation and MCP1 abundance compared with samples from untreated patients. CONCLUSIONS Our findings imply that both atorvastatin and rosuvastatin effectively inhibit the development of varicose veins, at least partially, by interfering with wall stress-mediated activator protein 1 activity in venous smooth muscle cells. For the first time, this study reveals a potential pharmacological treatment option that may be suitable to prevent growth of varicose veins and to limit formation of recurrence after varicose vein surgery.
Collapse
MESH Headings
- Animals
- Atorvastatin/pharmacology
- Case-Control Studies
- Cell Proliferation/drug effects
- Cells, Cultured
- Chemokine CCL2/metabolism
- Disease Models, Animal
- Dose-Response Relationship, Drug
- Female
- Humans
- Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology
- Male
- Mice
- Middle Aged
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Rosuvastatin Calcium/pharmacology
- Signal Transduction/drug effects
- Time Factors
- Transcription Factor AP-1/genetics
- Transcription Factor AP-1/metabolism
- Varicose Veins/metabolism
- Varicose Veins/pathology
- Varicose Veins/prevention & control
- Vascular Remodeling/drug effects
- Veins/drug effects
- Veins/metabolism
- Veins/pathology
Collapse
Affiliation(s)
- Johannes Eschrich
- Division of Cardiovascular Physiology, Institute of Physiology and Pathophysiology, Heidelberg University, Heidelberg, Germany
| | - Ralph Meyer
- Division of Cardiovascular Physiology, Institute of Physiology and Pathophysiology, Heidelberg University, Heidelberg, Germany
| | - Hanna Kuk
- Division of Cardiovascular Physiology, Institute of Physiology and Pathophysiology, Heidelberg University, Heidelberg, Germany
| | - Andreas H Wagner
- Division of Cardiovascular Physiology, Institute of Physiology and Pathophysiology, Heidelberg University, Heidelberg, Germany
| | | | - Sebastian Debus
- Department of Vascular Medicine, German Aortic Center, Hamburg, Germany
| | - Markus Hecker
- Division of Cardiovascular Physiology, Institute of Physiology and Pathophysiology, Heidelberg University, Heidelberg, Germany
| | - Thomas Korff
- Division of Cardiovascular Physiology, Institute of Physiology and Pathophysiology, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
168
|
Saxton J, Ferjentsik Z, Ducker C, Johnson AD, Shaw PE. Stepwise evolution of Elk-1 in early deuterostomes. FEBS J 2016; 283:1025-38. [PMID: 26613204 DOI: 10.1111/febs.13607] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Revised: 11/16/2015] [Accepted: 11/23/2015] [Indexed: 11/29/2022]
Abstract
Metazoans have multiple ETS paralogues with overlapping or indiscriminate biological functions. Elk-1, one of three mammalian ternary complex factors (TCFs), is a well-conserved, ETS domain-containing transcriptional regulator of mitogen-responsive genes that operates in concert with serum response factor (SRF). Nonetheless, its genetic role remains unresolved because the elk-1 gene could be deleted from the mouse genome seemingly without adverse effect. Here we have explored the evolution of Elk-1 to gain insight into its conserved biological role. We identified antecedent Elk-1 proteins in extant early metazoans and used amino acid sequence alignments to chart the appearance of domains characteristic of human Elk-1. We then performed biochemical studies to determine whether putative domains apparent in the Elk-1 protein of a primitive hemichordate were functionally orthologous to those of human Elk-1. Our findings imply the existence of primordial Elk-1 proteins in primitive deuterostomes that could operate as mitogen-responsive ETS transcription factors but not as TCFs. The role of TCF was acquired later, but presumably prior to the whole genome duplications in the basal vertebrate lineage. Thus its evolutionary origins link Elk-1 to the appearance of mesoderm.
Collapse
Affiliation(s)
- Janice Saxton
- School of Life Sciences, University of Nottingham, UK
| | | | | | | | - Peter E Shaw
- School of Life Sciences, University of Nottingham, UK
| |
Collapse
|
169
|
Scirocco A, Matarrese P, Carabotti M, Ascione B, Malorni W, Severi C. Cellular and Molecular Mechanisms of Phenotypic Switch in Gastrointestinal Smooth Muscle. J Cell Physiol 2016; 231:295-302. [PMID: 26206426 DOI: 10.1002/jcp.25105] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 07/21/2015] [Indexed: 10/16/2023]
Abstract
As a general rule, smooth muscle cells (SMC) are able to switch from a contractile phenotype to a less mature synthetic phenotype. This switch is accompanied by a loss of differentiation with decreased expression of contractile markers, increased proliferation as well as the synthesis and the release of several signaling molecules such as pro-inflammatory cytokines, chemotaxis-associated molecules, and growth factors. This SMC phenotypic plasticity has extensively been investigated in vascular diseases, but interest is also emerging in the field of gastroenterology. It has in fact been postulated that altered microenvironmental conditions, including the composition of microbiota, could trigger the remodeling of the enteric SMC, with phenotype changes and consequent alterations of contraction and impairment of gut motility. Several molecular actors participate in this phenotype remodeling. These include extracellular molecules such as cytokines and extracellular matrix proteins, as well as intracellular proteins, for example, transcription factors. Epigenetic control mechanisms and miRNA have also been suggested to participate. In this review key roles and actors of smooth muscle phenotypic switch, mainly in GI tissue, are described and discussed in the light of literature data available so far. J. Cell. Physiol. 231: 295-302, 2016. © 2015 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Annunziata Scirocco
- Department of Internal Medicine and Medical Specialties, University Sapienza Rome, Rome, Italy
| | - Paola Matarrese
- Department of Drug Research and Evaluation, Istituto Superiore di Sanit, à, Rome, Italy
- Center of Metabolomics, Rome, Italy
| | - Marilia Carabotti
- Department of Internal Medicine and Medical Specialties, University Sapienza Rome, Rome, Italy
| | - Barbara Ascione
- Department of Drug Research and Evaluation, Istituto Superiore di Sanit, à, Rome, Italy
| | - Walter Malorni
- Department of Drug Research and Evaluation, Istituto Superiore di Sanit, à, Rome, Italy
- San Raffaele Pisana Institute, Rome, Italy
| | - Carola Severi
- Department of Internal Medicine and Medical Specialties, University Sapienza Rome, Rome, Italy
| |
Collapse
|
170
|
Meyer zu Reckendorf C, Anastasiadou S, Bachhuber F, Franz-Wachtel M, Macek B, Knöll B. Proteomic analysis of SRF associated transcription complexes identified TFII-I as modulator of SRF function in neurons. Eur J Cell Biol 2016; 95:42-56. [DOI: 10.1016/j.ejcb.2015.11.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Revised: 10/30/2015] [Accepted: 11/05/2015] [Indexed: 11/25/2022] Open
|
171
|
An overview of potential molecular mechanisms involved in VSMC phenotypic modulation. Histochem Cell Biol 2015; 145:119-30. [DOI: 10.1007/s00418-015-1386-3] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/03/2015] [Indexed: 12/21/2022]
|
172
|
Rourke JL, Dranse HJ, Sinal CJ. CMKLR1 and GPR1 mediate chemerin signaling through the RhoA/ROCK pathway. Mol Cell Endocrinol 2015; 417:36-51. [PMID: 26363224 DOI: 10.1016/j.mce.2015.09.002] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Revised: 07/31/2015] [Accepted: 09/01/2015] [Indexed: 12/14/2022]
Abstract
Chemerin is an adipose-derived hormone that regulates immunity and energy homesotasis. To date, all known chemerin functions have been attributed to activation of the G protein-coupled receptor chemokine-like receptor-1 (CMKLR1). Chemerin is also the only known ligand for a second receptor, G protein-coupled receptor-1 (GPR1), whose signaling and function remains unknown. This study investigated the in vitro signal transduction mechanisms of CMKLR1 and GPR1 using a panel of luciferase-reporters and pathway-specific inhibitors. Herein we report the novel finding that chemerin signals through a RhoA and rho-associated protein kinase (ROCK)-dependent pathway for activation of the transcriptional regulator serum-response factor (SRF). Despite similarities in RhoA/ROCK, Gαi/o, and MAPK signaling, we also demonstrate species-specific and receptor-dependent variations in GPR1 and CMKLR1 signaling and expression of the SRF target genes EGR1, FOS and VCL. Moreover, we demonstrate that signaling through p38, Gαi/o, RhoA, and ROCK is required for chemerin-mediated chemotaxis of L1.2 lymphocytes and AGS gastric adenocarcinoma cells. These results provide, to our knowledge, the first empirical evidence that GPR1 is a functional chemerin receptor and identify RhoA/SRF as a novel chemerin-signaling axis via both CMKLR1 and GPR1.
Collapse
Affiliation(s)
- Jillian L Rourke
- Department of Pharmacology, Dalhousie University, Halifax, NS, Canada
| | - Helen J Dranse
- Department of Pharmacology, Dalhousie University, Halifax, NS, Canada
| | | |
Collapse
|
173
|
Grossi M, Phanstiel O, Rippe C, Swärd K, Alajbegovic A, Albinsson S, Forte A, Persson L, Hellstrand P, Nilsson BO. Inhibition of Polyamine Uptake Potentiates the Anti-Proliferative Effect of Polyamine Synthesis Inhibition and Preserves the Contractile Phenotype of Vascular Smooth Muscle Cells. J Cell Physiol 2015; 231:1334-42. [PMID: 26529275 DOI: 10.1002/jcp.25236] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 11/02/2015] [Indexed: 12/29/2022]
Abstract
Increased vascular smooth muscle cell (VSMC) proliferation is a factor in atherosclerosis and injury-induced arterial (re) stenosis. Inhibition of polyamine synthesis by α-difluoro-methylornithine (DFMO), an irreversible inhibitor of ornithine decarboxylase, attenuates VSMC proliferation with high sensitivity and specificity. However, cells can escape polyamine synthesis blockade by importing polyamines from the environment. To address this issue, polyamine transport inhibitors (PTIs) have been developed. We investigated the effects of the novel trimer44NMe (PTI-1) alone and in combination with DFMO on VSMC polyamine uptake, proliferation and phenotype regulation. PTI-1 efficiently inhibited polyamine uptake in primary mouse aortic and human coronary VSMCs in the absence as well as in the presence of DFMO. Interestingly, culture with DFMO for 2 days substantially (>95%) reduced putrescine (Put) and spermidine (Spd) contents without any effect on proliferation. Culture with PTI-1 alone had no effect on either polyamine levels or proliferation rate, but the combination of both treatments reduced Put and Spd levels below the detection limit and inhibited proliferation. Treatment with DFMO for a longer time period (4 days) reduced Put and Spd below their detection limits and reduced proliferation, showing that only a small pool of polyamines is needed to sustain VSMC proliferation. Inhibited proliferation by polyamine depletion was associated with maintained expression of contractile smooth marker genes. In cultured intact mouse aorta, PTI-1 potentiated the DFMO-induced inhibition of cell proliferation. The combination of endogenous polyamine synthesis inhibition with uptake blockade is thus a viable approach for targeting unwanted vascular cell proliferation in vivo, including vascular restenosis.
Collapse
Affiliation(s)
- Mario Grossi
- Department of Experimental Medical Science, Lund University, Sweden
| | - Otto Phanstiel
- Department of Medical Education, University of Central Florida, Orlando, Florida
| | - Catarina Rippe
- Department of Experimental Medical Science, Lund University, Sweden
| | - Karl Swärd
- Department of Experimental Medical Science, Lund University, Sweden
| | - Azra Alajbegovic
- Department of Experimental Medical Science, Lund University, Sweden
| | | | - Amalia Forte
- Department of Experimental Medicine, Second University of Naples, Italy
| | - Lo Persson
- Department of Experimental Medical Science, Lund University, Sweden
| | - Per Hellstrand
- Department of Experimental Medical Science, Lund University, Sweden
| | | |
Collapse
|
174
|
Shi N, Chen SY. Smooth Muscle Cell Differentiation: Model Systems, Regulatory Mechanisms, and Vascular Diseases. J Cell Physiol 2015; 231:777-87. [DOI: 10.1002/jcp.25208] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 09/29/2015] [Indexed: 02/06/2023]
Affiliation(s)
- Ning Shi
- Department of Physiology and Pharmacology; University of Georgia; Athens Georgia
| | - Shi-You Chen
- Department of Physiology and Pharmacology; University of Georgia; Athens Georgia
| |
Collapse
|
175
|
Park C, Lee MY, Park PJ, Ha SE, Berent RM, Fuchs R, Miano JM, Becker LS, Sanders KM, Ro S. Serum Response Factor Is Essential for Prenatal Gastrointestinal Smooth Muscle Development and Maintenance of Differentiated Phenotype. J Neurogastroenterol Motil 2015; 21:589-602. [PMID: 26424044 PMCID: PMC4622142 DOI: 10.5056/jnm15063] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Revised: 06/20/2015] [Accepted: 07/12/2015] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND/AIMS Smooth muscle cells (SMCs) characteristically express serum response factor (SRF), which regulates their development. The role of SRF in SMC plasticity in the pathophysiological conditions of gastrointestinal (GI) tract is less characterized. METHODS We generated SMC-specific Srf knockout mice and characterized the prenatally lethal phenotype using ultrasound biomicroscopy and histological analysis. We used small bowel partial obstruction surgeries and primary cell culture using cell-specific enhanced green fluorescent protein (EGFP) mouse lines to study phenotypic and molecular changes of SMCs by immunofluorescence, Western blotting, and quantitative polymerase chain reaction. Finally we examined SRF change in human rectal prolapse tissue by immunofluorescence. RESULTS Congenital SMC-specific Srf knockout mice died before birth and displayed severe GI and cardiac defects. Partial obstruction resulted in an overall increase in SRF protein expression. However, individual SMCs appeared to gradually lose SRF in the hypertrophic muscle. Cells expressing low levels of SRF also expressed low levels of platelet-derived growth factor receptor alpha (PDGFRα(low)) and Ki67. SMCs grown in culture recaptured the phenotypic switch from differentiated SMCs to proliferative PDGFRα(low) cells. The immediate and dramatic reduction of Srf and Myh11 mRNA expression confirmed the phenotypic change. Human rectal prolapse tissue also demonstrated significant loss of SRF expression. CONCLUSIONS SRF expression in SMCs is essential for prenatal development of the GI tract and heart. Following partial obstruction, SMCs down-regulate SRF to transition into proliferative PDGFRα(low) cells that may represent a phenotype responsible for their plasticity. These findings demonstrate that SRF also plays a critical role in the remodeling process following GI injury.
Collapse
Affiliation(s)
- Chanjae Park
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada,
USA
| | - Moon Young Lee
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada,
USA
- Department of Physiology, Wonkwang Digestive Disease Research Institute and Institute of Wonkwang Medical Science, School of Medicine, Wonkwang University, Iksan, Jeollabuk-do,
Korea
| | - Paul J Park
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada,
USA
| | - Se Eun Ha
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada,
USA
| | - Robyn M Berent
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada,
USA
| | - Robert Fuchs
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada,
USA
| | - Joseph M Miano
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, New York,
USA
| | - Laren S Becker
- Gastroenterology and Hepatology, Stanford University School of Medicine, Stanford, California,
USA
| | - Kenton M Sanders
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada,
USA
| | - Seungil Ro
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada,
USA
| |
Collapse
|
176
|
Myocardin-related transcription factors are required for cardiac development and function. Dev Biol 2015; 406:109-16. [PMID: 26386146 DOI: 10.1016/j.ydbio.2015.09.006] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Revised: 09/09/2015] [Accepted: 09/10/2015] [Indexed: 01/24/2023]
Abstract
Myocardin-Related Transcription Factors A and B (MRTF-A and MRTF-B) are highly homologous proteins that function as powerful coactivators of serum response factor (SRF), a ubiquitously expressed transcription factor essential for cardiac development. The SRF/MRTF complex binds to CArG boxes found in the control regions of genes that regulate cytoskeletal dynamics and muscle contraction, among other processes. While SRF is required for heart development and function, the role of MRTFs in the developing or adult heart has not been explored. Through cardiac-specific deletion of MRTF alleles in mice, we show that either MRTF-A or MRTF-B is dispensable for cardiac development and function, whereas deletion of both MRTF-A and MRTF-B causes a spectrum of structural and functional cardiac abnormalities. Defects observed in MRTF-A/B null mice ranged from reduced cardiac contractility and adult onset heart failure to neonatal lethality accompanied by sarcomere disarray. RNA-seq analysis on neonatal hearts identified the most altered pathways in MRTF double knockout hearts as being involved in cytoskeletal organization. Together, these findings demonstrate redundant but essential roles of the MRTFs in maintenance of cardiac structure and function and as indispensible links in cardiac cytoskeletal gene regulatory networks.
Collapse
|
177
|
Li W, Wang N, Li M, Gong H, Liao X, Yang X, Zhang T. Protein kinase Cα inhibits myocardin-induced cardiomyocyte hypertrophy through the promotion of myocardin phosphorylation. Acta Biochim Biophys Sin (Shanghai) 2015. [PMID: 26206583 DOI: 10.1093/abbs/gmv067] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Myocardin plays a key role in the development of cardiac hypertrophy. However, the upstream signals that control the stability and transactivity of myocardin remain to be fully understood. The expression of protein kinase Cα (PKCα) also induces cardiac hypertrophy. An essential downstream molecule of PKCα, extracellular signal-regulated kinase 1/2, was reported to negatively regulate the activities of myocardin. But, the effect of cooperation between PKCα and myocardin and the potential molecular mechanism by which PKCα regulates myocardin-mediated cardiac hypertrophy are unclear. In this study, a luciferase assay was performed using H9C2 cells transfected with expression plasmids for PKCα and myocardin. Surprisingly, the results showed that PKCα inhibited the transcriptional activity of myocardin. PKCα inhibited myocardin-induced cardiomyocyte hypertrophy, demonstrated by the decrease in cell surface area and fetal gene expression, in cardiomyocyte cells overexpressing PKCα and myocardin. The potential mechanism underlying the inhibition effect of PKCα on the function of myocardin is further explored. PKCα directly promoted the basal phosphorylation of endogenous myocardin at serine and threonine residues. In myocardin-overexpressing cardiomyocyte cells, PKCα induced the excessive phosphorylation of myocardin, resulting in the degradation of myocardin and a transcriptional suppression of hypertrophic genes. These results demonstrated that PKCα inhibits myocardin-induced cardiomyocyte hypertrophy through the promotion of myocardin phosphorylation.
Collapse
Affiliation(s)
- Weizong Li
- Key Laboratory of Industrial Microbiology, Ministry of Education and Tianjin City, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Nan Wang
- Key Laboratory of Industrial Microbiology, Ministry of Education and Tianjin City, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Man Li
- Key Laboratory of Industrial Microbiology, Ministry of Education and Tianjin City, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Huiqin Gong
- Key Laboratory of Industrial Microbiology, Ministry of Education and Tianjin City, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Xinghua Liao
- Key Laboratory of Industrial Microbiology, Ministry of Education and Tianjin City, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China Department of Biochemistry, Medical College, Wuhan University of Science and Technology, Wuhan 430081, China
| | - Xiaolong Yang
- Key Laboratory of Industrial Microbiology, Ministry of Education and Tianjin City, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Tongcun Zhang
- Key Laboratory of Industrial Microbiology, Ministry of Education and Tianjin City, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China Department of Biochemistry, Medical College, Wuhan University of Science and Technology, Wuhan 430081, China
| |
Collapse
|
178
|
Yan G, Wang Q, Hu S, Wang D, Qiao Y, Ma G, Tang C, Gu Y. Digoxin inhibits PDGF-BB-induced VSMC proliferation and migration through an increase in ILK signaling and attenuates neointima formation following carotid injury. Int J Mol Med 2015; 36:1001-11. [PMID: 26311435 PMCID: PMC4564091 DOI: 10.3892/ijmm.2015.2320] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2014] [Accepted: 07/31/2015] [Indexed: 11/06/2022] Open
Abstract
The increased proliferation and migration of vascular smooth muscle cells (VSMCs) are key events in the development of artery restenosis following percutaneous coronary intervention. Digoxin has long been used in the treatment of heart failure and has been shown to inhibit the proliferation of cancer cells through multiple pathways. However, the potential role of digoxin in the regulation of VSMC proliferation and migration and its effectiveness in the treatment of cardiovascular diseases, such as restenosis, remains unexplored. In the present study, we demonstrate that digoxin-induced growth inhibition is associated with the downregulation of CDK activation and the restoration of p27Kip1 levels in platelet-derived growth factor (PDGF)-stimulated VSMCs. In addition, we found that digoxin restored the PDGF‑BB-induced inhibition of integrin linked kinase (ILK) expression and prevented the PDGF‑BB-induced activation of glycogen synthase kinase (GSK)-3β. Furthermore, digoxin inhibited adhesion molecule and extracellular matrix relative protein expression. Finally, we found that digoxin significantly inhibited neointima formation, accompanied by a decrease in cell proliferation following vascular injury in rats. These effects of digoxin were shown to be mediated, at least in part, through an increase in ILK/Akt signaling and a decrease in GSK-3β signaling in PDGF‑BB-stimulated VSMCs. In conclusion, our data demonstrate that digoxin exerts an inhibitory effect on the PDGF‑BB-induced proliferation, migration and phenotypic modulation of VSMCs, and prevents neointima formation in rats. These observations indicate the potential therapeutic application of digoxin in the treatment of cardiovascular diseases, such as restenosis.
Collapse
Affiliation(s)
- Gaoliang Yan
- Department of Cardiology, Zhongda Hospital of Southeast University Medical School, Nanjing, Jiangsu 210009, P.R. China
| | - Qingjie Wang
- Department of Cardiology, Zhongda Hospital of Southeast University Medical School, Nanjing, Jiangsu 210009, P.R. China
| | - Shengda Hu
- Department of Cardiology, Zhongda Hospital of Southeast University Medical School, Nanjing, Jiangsu 210009, P.R. China
| | - Dong Wang
- Department of Cardiology, Zhongda Hospital of Southeast University Medical School, Nanjing, Jiangsu 210009, P.R. China
| | - Yong Qiao
- Department of Cardiology, Zhongda Hospital of Southeast University Medical School, Nanjing, Jiangsu 210009, P.R. China
| | - Genshan Ma
- Department of Cardiology, Zhongda Hospital of Southeast University Medical School, Nanjing, Jiangsu 210009, P.R. China
| | - Chengchun Tang
- Department of Cardiology, Zhongda Hospital of Southeast University Medical School, Nanjing, Jiangsu 210009, P.R. China
| | - Yuchun Gu
- Institute of Molecular Medicine (IMM), Peking University, Beijing 100190, P.R. China
| |
Collapse
|
179
|
Abstract
Caveolae are membrane organelles that play roles in glucose and lipid metabolism and in vascular function. Formation of caveolae requires caveolins and cavins. The make-up of caveolae and their density is considered to reflect cell-specific transcriptional control mechanisms for caveolins and cavins, but knowledge regarding regulation of caveolae genes is incomplete. Myocardin (MYOCD) and its relative MRTF-A (MKL1) are transcriptional coactivators that control genes which promote smooth muscle differentiation. MRTF-A communicates changes in actin polymerization to nuclear gene transcription. Here we tested if myocardin family proteins control biogenesis of caveolae via activation of caveolin and cavin transcription. Using human coronary artery smooth muscle cells we found that jasplakinolide and latrunculin B (LatB), substances that promote and inhibit actin polymerization, increased and decreased protein levels of caveolins and cavins, respectively. The effect of LatB was associated with reduced mRNA levels for these genes and this was replicated by the MRTF inhibitor CCG-1423 which was non-additive with LatB. Overexpression of myocardin and MRTF-A caused 5-10-fold induction of caveolins whereas cavin-1 and cavin-2 were induced 2-3-fold. PACSIN2 also increased, establishing positive regulation of caveolae genes from three families. Full regulation of CAV1 was retained in its proximal promoter. Knock down of the serum response factor (SRF), which mediates many of the effects of myocardin, decreased cavin-1 but increased caveolin-1 and -2 mRNAs. Viral transduction of myocardin increased the density of caveolae 5-fold in vitro. A decrease of CAV1 was observed concomitant with a decrease of the smooth muscle marker calponin in aortic aneurysms from mice (C57Bl/6) infused with angiotensin II. Human expression data disclosed correlations of MYOCD with CAV1 in a majority of human tissues and in the heart, correlation with MKL2 (MRTF-B) was observed. The myocardin family of transcriptional coactivators therefore drives formation of caveolae and this effect is largely independent of SRF.
Collapse
|
180
|
Mutations in ARID2 are associated with intellectual disabilities. Neurogenetics 2015; 16:307-14. [DOI: 10.1007/s10048-015-0454-0] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 07/21/2015] [Indexed: 12/30/2022]
|
181
|
Ha JM, Yun SJ, Kim YW, Jin SY, Lee HS, Song SH, Shin HK, Bae SS. Platelet-derived growth factor regulates vascular smooth muscle phenotype via mammalian target of rapamycin complex 1. Biochem Biophys Res Commun 2015; 464:57-62. [DOI: 10.1016/j.bbrc.2015.05.097] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Accepted: 05/28/2015] [Indexed: 10/23/2022]
|
182
|
Shyu KG, Cheng WP, Wang BW. Angiotensin II Downregulates MicroRNA-145 to Regulate Kruppel-like Factor 4 and Myocardin Expression in Human Coronary Arterial Smooth Muscle Cells under High Glucose Conditions. Mol Med 2015; 21:616-25. [PMID: 26181633 DOI: 10.2119/molmed.2015.00041] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Accepted: 07/13/2015] [Indexed: 01/26/2023] Open
Abstract
MicroRNA (miR)-145 is the most abundant miR in vascular smooth muscle cells (VSMCs). However, the effect of hyperglycemia on the regulation of miR-145 is unknown. We hypothesized that the hyperglycemic condition activates a proinflammatory response that mediates the expression of miR-145 in VSMCs. We investigated whether miR-145 serves as a critical regulator to regulate the downstream proliferation factors (including Kruppel-like factor 4 [Klf4] and myocardin) in VSMCs under hyperglycemic conditions. Human coronary artery smooth muscle cells (HCASMCs) were cultured under high glucose conditions. Sustained high glucose at 25 mmol/L significantly decreased the expression of miR-145 in HCASMCs. High glucose significantly increased angiotensin II (Ang II) secretion from HCASMCs and Ang II suppressed miR-145 expression in HCASMCs. Ang II repression of miR145 expression resulted in increased Klf4 and decreased myocardin expression under conditions of high glucose. Overexpression of miR-145 significantly decreased Klf4 and increased myocardin expression and inhibited HCASMC proliferation and migration induced by a high glucose state. Balloon injury of the carotid artery in diabetic rats was performed to investigate miR-145, Klf and myocardin expression. The expression of miR-145 was maximally increased at 7 d after carotid injury and gradually declined thereafter. Overexpression of miR-145 and treatment with valsartan reversed Klf4 and myocardin protein expression induced by balloon injury and improved vascular injury. In conclusion, our study reveals that Ang II downregulates miR-145 to regulate Klf4 and myocardin expression in HCASMCs under high glucose conditions. Ang II plays a critical role in the regulation of miR-145 under hyperglycemic conditions.
Collapse
Affiliation(s)
- Kou-Gi Shyu
- Division of Cardiology, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan.,Graduate Institute of Clinical Medicine, Taipei Medical University, Taipei, Taiwan
| | - Wen-Ping Cheng
- Department of Medical Education and Research, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
| | - Bao-Wei Wang
- Department of Medical Education and Research, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
| |
Collapse
|
183
|
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: 69] [Impact Index Per Article: 6.9] [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
|
184
|
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: 25] [Impact Index Per Article: 2.5] [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
|
185
|
Gutierrez-Pajares JL, Iturrieta J, Dulam V, Wang Y, Pavlides S, Malacari G, Lisanti MP, Frank PG. Caveolin-3 Promotes a Vascular Smooth Muscle Contractile Phenotype. Front Cardiovasc Med 2015; 2:27. [PMID: 26664898 PMCID: PMC4671348 DOI: 10.3389/fcvm.2015.00027] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 05/24/2015] [Indexed: 01/12/2023] Open
Abstract
Epidemiological studies have demonstrated the importance of cardiovascular diseases in Western countries. Among the cell types associated with a dysfunctional vasculature, smooth muscle (SM) cells are believed to play an essential role in the development of these illnesses. Vascular SM cells are key regulators of the vascular tone and also have an important function in the development of atherosclerosis and restenosis. While in the normal vasculature, contractile SM cells are predominant, in atherosclerotic vascular lesions, synthetic cells migrate toward the neointima, proliferate, and synthetize extracellular matrix proteins. In the present study, we have examined the role of caveolin-3 in the regulation of SM cell phenotype. Caveolin-3 is expressed in vivo in normal arterial SM cells, but its expression appears to be lost in cultured SM cells. Our data show that caveolin-3 expression in the A7r5 SM cell line is associated with increased expression of contractility markers such as SM α-actin, SM myosin heavy chain but decreased expression of the synthetic phenotype markers such as p-Elk and Klf4. Moreover, we also show that caveolin-3 expression can reduce proliferation upon treatment with LDL or PDGF. Finally, we show that caveolin-3-expressing SM cells are less sensitive to apoptosis than control cells upon treatment with oxidized LDL. Taken together, our data suggest that caveolin-3 can regulate the phenotypic switch between contractile and synthetic SM cells. A better understanding of the factors regulating caveolin-3 expression and function in this cell type will permit the development of a better comprehension of the factors regulating SM function in atherosclerosis and restenosis.
Collapse
Affiliation(s)
- Jorge L Gutierrez-Pajares
- Faculté de Médecine, INSERM UMR1069 "Nutrition, Croissance et Cancer", Université François Rabelais de Tours , Tours , France ; Department of Stem Cell Biology and Regenerative Medicine, Kimmel Cancer Center, Thomas Jefferson University , Philadelphia, PA , USA ; Department of Cancer Biology, Kimmel Cancer Center, Thomas Jefferson University , Philadelphia, PA , USA
| | - Jeannette Iturrieta
- Department of Stem Cell Biology and Regenerative Medicine, Kimmel Cancer Center, Thomas Jefferson University , Philadelphia, PA , USA ; Department of Cancer Biology, Kimmel Cancer Center, Thomas Jefferson University , Philadelphia, PA , USA
| | - Vipin Dulam
- Department of Stem Cell Biology and Regenerative Medicine, Kimmel Cancer Center, Thomas Jefferson University , Philadelphia, PA , USA ; Department of Cancer Biology, Kimmel Cancer Center, Thomas Jefferson University , Philadelphia, PA , USA
| | - Yu Wang
- Department of Stem Cell Biology and Regenerative Medicine, Kimmel Cancer Center, Thomas Jefferson University , Philadelphia, PA , USA ; Department of Cancer Biology, Kimmel Cancer Center, Thomas Jefferson University , Philadelphia, PA , USA
| | - Stephanos Pavlides
- The Manchester Centre for Cellular Metabolism (MCCM), Institute of Cancer Sciences, University of Manchester , Manchester , UK ; The Breakthrough Breast Cancer Research Unit, Institute of Cancer Sciences, University of Manchester , Manchester , UK
| | - Gabriella Malacari
- Department of Stem Cell Biology and Regenerative Medicine, Kimmel Cancer Center, Thomas Jefferson University , Philadelphia, PA , USA ; Department of Cancer Biology, Kimmel Cancer Center, Thomas Jefferson University , Philadelphia, PA , USA
| | - Michael P Lisanti
- The Manchester Centre for Cellular Metabolism (MCCM), Institute of Cancer Sciences, University of Manchester , Manchester , UK ; The Breakthrough Breast Cancer Research Unit, Institute of Cancer Sciences, University of Manchester , Manchester , UK
| | - Philippe G Frank
- Faculté de Médecine, INSERM UMR1069 "Nutrition, Croissance et Cancer", Université François Rabelais de Tours , Tours , France ; Department of Stem Cell Biology and Regenerative Medicine, Kimmel Cancer Center, Thomas Jefferson University , Philadelphia, PA , USA ; Department of Cancer Biology, Kimmel Cancer Center, Thomas Jefferson University , Philadelphia, PA , USA ; Department of Biochemistry and Molecular Biology, Thomas Jefferson University , Philadelphia, PA , USA
| |
Collapse
|
186
|
|
187
|
Wu Y, Shen Y, Kang K, Zhang Y, Ao F, Wan Y, Song J. Effects of estrogen on growth and smooth muscle differentiation of vascular wall-resident CD34(+) stem/progenitor cells. Atherosclerosis 2015; 240:453-61. [PMID: 25898000 DOI: 10.1016/j.atherosclerosis.2015.04.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Revised: 04/03/2015] [Accepted: 04/04/2015] [Indexed: 01/12/2023]
Abstract
OBJECTIVES To investigate the effects of estrogen on growth and smooth muscle cell (SMC)-differentiation of vascular wall-resident CD34(+) stem/progenitor cells (VRS/Pcs). METHODS AND RESULTS The existence of CD34(+) VRS/Pcs was confirmed by immunohistochemistry in the adventitia of arteries of young (2-month-old) and old (24-month-old) female SD rats with less CD34(+) adventitial cells detected in the old. The VRS/Pcs isolated from young animals were grown in Stem cell growth medium or induced to differentiate into SMC with PDGF-BB in the presence or absence of 17β-estrodiol (E2). Flow cytometry, RT-qPCR and Western blot showed that E2 promoted Brdu incorporation of the CD34(+) VRS/Pcs growing in Stem cell growth medium; but when the cells were incubated in PDGF-BB, the hormone enhanced their expression of SMC marker SM22. ChIP and IP assays showed that E2 significantly promoted the binding of pELK1-SRF complex to the promoter of c-fos gene in CD34(+) VRS/Pcs growing in the Stem cell growth medium; but when the cells were stimulated with PDGF-BB, an E2-enhanced binding of myocardin-SRF to the promoter of SM22 gene was found with enhanced expression of SRC3 and its binding to myocardin. The effects of E2 above could be blocked by the estrogen receptor antagonist ICI 182,780 or inhibited by SRF-siRNA. CONCLUSION Estrogen has dual effects on CD34(+) VRS/Pcs. For the undifferentiated VRS/Pcs, it accelerates their proliferation by enhancing binding of pELK1-SRF complex to c-fos gene; while for the differentiating VRS/Pcs, it promotes their differentiation to SMC through a mechanism of SRC3-mediated interaction of myocardin-SRF complex with SM22 gene.
Collapse
Affiliation(s)
- Yan Wu
- Department of Anatomy and Embryology, Wuhan University School of Basic Medical Sciences, 135 Donghu Road, Wuhan 430071, Hubei, PR China
| | - Yan Shen
- Department of Physiology, Wuhan University School of Basic Medical Sciences, 135 Donghu Road, Wuhan 430071, Hubei, PR China
| | - Kai Kang
- Department of Anatomy and Embryology, Wuhan University School of Basic Medical Sciences, 135 Donghu Road, Wuhan 430071, Hubei, PR China
| | - Yanhong Zhang
- Department of Anatomy and Embryology, Wuhan University School of Basic Medical Sciences, 135 Donghu Road, Wuhan 430071, Hubei, PR China
| | - Feng Ao
- Department of Anatomy and Embryology, Wuhan University School of Basic Medical Sciences, 135 Donghu Road, Wuhan 430071, Hubei, PR China
| | - Yu Wan
- Department of Physiology, Wuhan University School of Basic Medical Sciences, 135 Donghu Road, Wuhan 430071, Hubei, PR China.
| | - Jian Song
- Department of Anatomy and Embryology, Wuhan University School of Basic Medical Sciences, 135 Donghu Road, Wuhan 430071, Hubei, PR China.
| |
Collapse
|
188
|
Briot A, Jaroszewicz A, Warren CM, Lu J, Touma M, Rudat C, Hofmann JJ, Airik R, Weinmaster G, Lyons K, Wang Y, Kispert A, Pellegrini M, Iruela-Arispe ML. Repression of Sox9 by Jag1 is continuously required to suppress the default chondrogenic fate of vascular smooth muscle cells. Dev Cell 2015; 31:707-21. [PMID: 25535917 DOI: 10.1016/j.devcel.2014.11.023] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Revised: 09/11/2014] [Accepted: 11/13/2014] [Indexed: 01/15/2023]
Abstract
Acquisition and maintenance of vascular smooth muscle fate are essential for the morphogenesis and function of the circulatory system. Loss of contractile properties or changes in the identity of vascular smooth muscle cells (vSMCs) can result in structural alterations associated with aneurysms and vascular wall calcification. Here we report that maturation of sclerotome-derived vSMCs depends on a transcriptional switch between mouse embryonic days 13 and 14.5. At this time, Notch/Jag1-mediated repression of sclerotome transcription factors Pax1, Scx, and Sox9 is necessary to fully enable vSMC maturation. Specifically, Notch signaling in vSMCs antagonizes sclerotome and cartilage transcription factors and promotes upregulation of contractile genes. In the absence of the Notch ligand Jag1, vSMCs acquire a chondrocytic transcriptional repertoire that can lead to ossification. Importantly, our findings suggest that sustained Notch signaling is essential throughout vSMC life to maintain contractile function, prevent vSMC reprogramming, and promote vascular wall integrity.
Collapse
Affiliation(s)
- Anaïs Briot
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Artur Jaroszewicz
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Carmen M Warren
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Jing Lu
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Marlin Touma
- Division of Anesthesiology, Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Carsten Rudat
- Institut fur Molekularbiologie, Medizinische Hochschule Hannover, 30625 Hannover, Germany
| | - Jennifer J Hofmann
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Rannar Airik
- Institut fur Molekularbiologie, Medizinische Hochschule Hannover, 30625 Hannover, Germany
| | - Gerry Weinmaster
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Karen Lyons
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Yibin Wang
- Division of Anesthesiology, Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Andreas Kispert
- Institut fur Molekularbiologie, Medizinische Hochschule Hannover, 30625 Hannover, Germany
| | - Matteo Pellegrini
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - M Luisa Iruela-Arispe
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
189
|
Trembley MA, Velasquez LS, de Mesy Bentley KL, Small EM. Myocardin-related transcription factors control the motility of epicardium-derived cells and the maturation of coronary vessels. Development 2015; 142:21-30. [PMID: 25516967 DOI: 10.1242/dev.116418] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
An important pool of cardiovascular progenitor cells arises from the epicardium, a single layer of mesothelium lining the heart. Epicardium-derived progenitor cell (EPDC) formation requires epithelial-to-mesenchymal transition (EMT) and the subsequent migration of these cells into the sub-epicardial space. Although some of the physiological signals that promote EMT are understood, the functional mediators of EPDC motility and differentiation are not known. Here, we identify a novel regulatory mechanism of EPDC mobilization. Myocardin-related transcription factor (MRTF)-A and MRTF-B (MKL1 and MKL2, respectively) are enriched in the perinuclear space of epicardial cells during development. Transforming growth factor (TGF)-β signaling and disassembly of cell contacts leads to nuclear accumulation of MRTFs and the activation of the motile gene expression program. Conditional ablation of Mrtfa and Mrtfb specifically in the epicardium disrupts cell migration and leads to sub-epicardial hemorrhage, partially stemming from the depletion of coronary pericytes. Using lineage-tracing analyses, we demonstrate that sub-epicardial pericytes arise from EPDCs in a process that requires the MRTF-dependent motile gene expression program. These findings provide novel mechanisms linking EPDC motility and differentiation, shed light on the transcriptional control of coronary microvascular maturation and suggest novel therapeutic strategies to manipulate epicardium-derived progenitor cells for cardiac repair.
Collapse
Affiliation(s)
- Michael A Trembley
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY 14624, USA Department of Pharmacology and Physiology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14624, USA
| | - Lissette S Velasquez
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY 14624, USA
| | - Karen L de Mesy Bentley
- Department of Pathology and Laboratory Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14624, USA
| | - Eric M Small
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY 14624, USA Department of Pharmacology and Physiology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14624, USA
| |
Collapse
|
190
|
MicroRNAs and cardiovascular diseases. BIOMED RESEARCH INTERNATIONAL 2015; 2015:682857. [PMID: 25710020 PMCID: PMC4331324 DOI: 10.1155/2015/682857] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Accepted: 10/25/2014] [Indexed: 12/19/2022]
Abstract
Coronary artery diseases (CAD) and heart failure have high mortality rate in the world, although much progress has been made in this field in last two decades. There is still a clinical need for a novel diagnostic approach and a therapeutic strategy to decrease the incidence of CAD. MicroRNAs (miRNAs) are highly conserved noncoding small RNA molecules that regulate a large fraction of the genome by binding to complementary messenger RNA sequences, resulting in posttranscriptional gene silencing. Recent studies have shown that specific miRNAs are involved in whole stage of atherosclerosis, from endothelium dysfunction to plaque rupture. These findings suggest that miRNAs are potential biomarkers in early diagnosis and therapeutic targets in CAD. In the present review, we highlight the role of miRNAs in every stage of atherosclerosis, and discuss the prospects of miRNAs in the near future.
Collapse
|
191
|
Chettimada S, Gupte R, Rawat D, Gebb SA, McMurtry IF, Gupte SA. Hypoxia-induced glucose-6-phosphate dehydrogenase overexpression and -activation in pulmonary artery smooth muscle cells: implication in pulmonary hypertension. Am J Physiol Lung Cell Mol Physiol 2015; 308:L287-300. [PMID: 25480333 PMCID: PMC4338932 DOI: 10.1152/ajplung.00229.2014] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Accepted: 12/01/2014] [Indexed: 11/22/2022] Open
Abstract
Severe pulmonary hypertension is a debilitating disease with an alarmingly low 5-yr life expectancy. Hypoxia, one of the causes of pulmonary hypertension, elicits constriction and remodeling of the pulmonary arteries. We now know that pulmonary arterial remodeling is a consequence of hyperplasia and hypertrophy of pulmonary artery smooth muscle (PASM), endothelial, myofibroblast, and stem cells. However, our knowledge about the mechanisms that cause these cells to proliferate and hypertrophy in response to hypoxic stimuli is still incomplete, and, hence, the treatment for severe pulmonary arterial hypertension is inadequate. Here we demonstrate that the activity and expression of glucose-6-phosphate dehydrogenase (G6PD), the rate-limiting enzyme of the pentose phosphate pathway, are increased in hypoxic PASM cells and in lungs of chronic hypoxic rats. G6PD overexpression and -activation is stimulated by H2O2. Increased G6PD activity contributes to PASM cell proliferation by increasing Sp1 and hypoxia-inducible factor 1α (HIF-1α), which directs the cells to synthesize less contractile (myocardin and SM22α) and more proliferative (cyclin A and phospho-histone H3) proteins. G6PD inhibition with dehydroepiandrosterone increased myocardin expression in remodeled pulmonary arteries of moderate and severe pulmonary hypertensive rats. These observations suggest that altered glucose metabolism and G6PD overactivation play a key role in switching the PASM cells from the contractile to synthetic phenotype by increasing Sp1 and HIF-1α, which suppresses myocardin, a key cofactor that maintains smooth muscle cell in contractile state, and increasing hypoxia-induced PASM cell growth, and hence contribute to pulmonary arterial remodeling and pathogenesis of pulmonary hypertension.
Collapse
Affiliation(s)
- Sukrutha Chettimada
- Department of Biochemistry and Molecular Biology, College of Medicine, University of South Alabama, Mobile, Alabama
| | - Rakhee Gupte
- Department of Biochemistry and Molecular Biology, College of Medicine, University of South Alabama, Mobile, Alabama
| | - Dhwajbahadur Rawat
- Department of Biochemistry and Molecular Biology, College of Medicine, University of South Alabama, Mobile, Alabama
| | - Sarah A Gebb
- Department of Cell Biology and Neurosciences, College of Medicine, University of South Alabama, Mobile, Alabama
| | - Ivan F McMurtry
- Department of Pharmacology, College of Medicine, University of South Alabama, Mobile, Alabama; Department of Medicine, College of Medicine, University of South Alabama, Mobile, Alabama; and Center for Lung Biology, College of Medicine, University of South Alabama, Mobile, Alabama
| | - Sachin A Gupte
- Department of Biochemistry and Molecular Biology, College of Medicine, University of South Alabama, Mobile, Alabama; Center for Lung Biology, College of Medicine, University of South Alabama, Mobile, Alabama
| |
Collapse
|
192
|
Knipe RS, Tager AM, Liao JK. The Rho kinases: critical mediators of multiple profibrotic processes and rational targets for new therapies for pulmonary fibrosis. Pharmacol Rev 2015; 67:103-17. [PMID: 25395505 PMCID: PMC4279074 DOI: 10.1124/pr.114.009381] [Citation(s) in RCA: 163] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is characterized by progressive lung scarring, short median survival, and limited therapeutic options, creating great need for new pharmacologic therapies. IPF is thought to result from repetitive environmental injury to the lung epithelium, in the context of aberrant host wound healing responses. Tissue responses to injury fundamentally involve reorganization of the actin cytoskeleton of participating cells, including epithelial cells, fibroblasts, endothelial cells, and macrophages. Actin filament assembly and actomyosin contraction are directed by the Rho-associated coiled-coil forming protein kinase (ROCK) family of serine/threonine kinases (ROCK1 and ROCK2). As would therefore be expected, lung ROCK activation has been demonstrated in humans with IPF and in animal models of this disease. ROCK inhibitors can prevent fibrosis in these models, and more importantly, induce the regression of already established fibrosis. Here we review ROCK structure and function, upstream activators and downstream targets of ROCKs in pulmonary fibrosis, contributions of ROCKs to profibrotic cellular responses to lung injury, ROCK inhibitors and their efficacy in animal models of pulmonary fibrosis, and potential toxicities of ROCK inhibitors in humans, as well as involvement of ROCKs in fibrosis in other organs. As we discuss, ROCK activation is required for multiple profibrotic responses, in the lung and multiple other organs, suggesting ROCK participation in fundamental pathways that contribute to the pathogenesis of a broad array of fibrotic diseases. Multiple lines of evidence therefore indicate that ROCK inhibition has great potential to be a powerful therapeutic tool in the treatment of fibrosis, both in the lung and beyond.
Collapse
Affiliation(s)
- Rachel S Knipe
- Pulmonary and Critical Care Unit and Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts (R.S.K., A.M.T.); and Section of Cardiology, Department of Medicine, University of Chicago, Chicago, Illinois (J.K.L.)
| | - Andrew M Tager
- Pulmonary and Critical Care Unit and Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts (R.S.K., A.M.T.); and Section of Cardiology, Department of Medicine, University of Chicago, Chicago, Illinois (J.K.L.)
| | - James K Liao
- Pulmonary and Critical Care Unit and Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts (R.S.K., A.M.T.); and Section of Cardiology, Department of Medicine, University of Chicago, Chicago, Illinois (J.K.L.)
| |
Collapse
|
193
|
Hödebeck M, Scherer C, Wagner AH, Hecker M, Korff T. TonEBP/NFAT5 regulates ACTBL2 expression in biomechanically activated vascular smooth muscle cells. Front Physiol 2014; 5:467. [PMID: 25520667 PMCID: PMC4253659 DOI: 10.3389/fphys.2014.00467] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Accepted: 11/14/2014] [Indexed: 12/25/2022] Open
Abstract
Cytoskeletal reorganization and migration are critical responses which enable vascular smooth muscle cells (VSMCs) cells to evade, compensate, or adapt to alterations in biomechanical stress. An increase in wall stress or biomechanical stretch as it is elicited by arterial hypertension promotes their reorganization in the vessel wall which may lead to arterial stiffening and contractile dysfunction. This adaptive remodeling process is dependent on and driven by subtle phenotype changes including those controlling the cytoskeletal architecture and motility of VSMCs. Recently, it has been reported that the transcription factor nuclear factor of activated T-cells 5 (TonEBP/NFAT5) controls critical aspects of the VSMC phenotype and is activated by biomechanical stretch. We therefore hypothesized that NFAT5 controls the expression of gene products orchestrating cytoskeletal reorganization in stretch-stimulated VSMCs. Automated immunofluorescence and Western blot analyses revealed that biomechanical stretch enhances the expression and nuclear translocation of NFAT5 in VSMCs. Subsequent in silico analyses suggested that this transcription factor binds to the promotor region of ACTBL2 encoding kappa-actin which was shown to be abundantly expressed in VSMCs upon exposure to biomechanical stretch. Furthermore, ACTBL2 expression was inhibited in these cells upon siRNA-mediated knockdown of NFAT5. Kappa-actin appeared to be aligned with stress fibers under static culture conditions, dispersed in lamellipodia and supported VSMC migration as its knockdown diminishes lateral migration of these cells. In summary, our findings delineated biomechanical stretch as a determinant of NFAT5 expression and nuclear translocation controlling the expression of the cytoskeletal protein ACTBL2. This response may orchestrate the migratory activity of VSMCs and thus promote maladaptive rearrangement of the arterial vessel wall during hypertension.
Collapse
Affiliation(s)
- Maren Hödebeck
- Division of Cardiovascular Physiology, Institute of Physiology and Pathophysiology, University of Heidelberg Heidelberg, Germany
| | - Clemens Scherer
- Division of Cardiovascular Physiology, Institute of Physiology and Pathophysiology, University of Heidelberg Heidelberg, Germany
| | - Andreas H Wagner
- Division of Cardiovascular Physiology, Institute of Physiology and Pathophysiology, University of Heidelberg Heidelberg, Germany
| | - Markus Hecker
- Division of Cardiovascular Physiology, Institute of Physiology and Pathophysiology, University of Heidelberg Heidelberg, Germany
| | - Thomas Korff
- Division of Cardiovascular Physiology, Institute of Physiology and Pathophysiology, University of Heidelberg Heidelberg, Germany
| |
Collapse
|
194
|
Vasudevan HN, Soriano P. SRF regulates craniofacial development through selective recruitment of MRTF cofactors by PDGF signaling. Dev Cell 2014; 31:332-344. [PMID: 25453829 DOI: 10.1016/j.devcel.2014.10.005] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Revised: 09/02/2014] [Accepted: 10/03/2014] [Indexed: 02/07/2023]
Abstract
Receptor tyrosine kinase signaling is critical for mammalian craniofacial development, but the key downstream transcriptional effectors remain unknown. We demonstrate that serum response factor (SRF) is induced by both platelet-derived growth factor (PDGF) and fibroblast growth factor (FGF) signaling in mouse embryonic palatal mesenchyme cells and that Srf neural crest conditional mutants exhibit facial clefting accompanied by proliferation and migration defects. Srf and Pdgfra mutants interact genetically in craniofacial development, but Srf and Fgfr1 mutants do not. This signal specificity is recapitulated at the level of cofactor activation: while both PDGF and FGF target gene promoters show enriched genome-wide overlap with SRF ChIP-seq peaks, PDGF selectively activates a network of MRTF-dependent cytoskeletal genes. Collectively, our results identify a role for SRF in proliferation and migration during craniofacial development and delineate a mechanism of receptor tyrosine kinase specificity mediated through differential cofactor usage, leading to a PDGF-responsive SRF-driven transcriptional program in the midface.
Collapse
Affiliation(s)
- Harish N Vasudevan
- Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Philippe Soriano
- Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
195
|
Affiliation(s)
- Kathleen A Martin
- From the Departments of Internal Medicine (K.A.M., K.K.H.) and Pharmacology (K.A.M.), Yale Cardiovascular Research Center and Vascular Biology and Therapeutics Program, Yale University, New Haven, CT
| | - Karen K Hirschi
- From the Departments of Internal Medicine (K.A.M., K.K.H.) and Pharmacology (K.A.M.), Yale Cardiovascular Research Center and Vascular Biology and Therapeutics Program, Yale University, New Haven, CT.
| |
Collapse
|
196
|
Espinoza-Lewis RA, Wang DZ. Generation of a Cre knock-in into the Myocardin locus to mark early cardiac and smooth muscle cell lineages. Genesis 2014; 52:879-87. [PMID: 25174608 DOI: 10.1002/dvg.22819] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Revised: 08/25/2014] [Accepted: 08/26/2014] [Indexed: 11/10/2022]
Abstract
The molecular events that control cell fate determination in cardiac and smooth muscle lineages remain elusive. Myocardin is an important transcription cofactor that regulates cell proliferation, differentiation, and development of the cardiovascular system. Here, we describe the construction and analysis of a dual Cre and enhanced green fluorescent protein (EGFP) knock-in mouse line in the Myocardin locus (Myocd(KI)). We report that the Myocd(KI) allele expresses the Cre enzyme and the EGFP in a manner that recapitulates endogenous Myocardin expression patterns. We show that Myocardin expression marks the earliest cardiac and smooth muscle lineages. Furthermore, this genetic model allows for the identification of a cardiac cell population, which maintains both Myocardin and Isl-1 expression, in E7.75-E8.0 embryos, highlighting the contribution and merging of the first and second heart fields during cardiogenesis. Therefore, the Myocd(KI) allele is a unique tool for studying cardiovascular development and lineage-specific gene manipulation.
Collapse
Affiliation(s)
- Ramón A Espinoza-Lewis
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | | |
Collapse
|
197
|
Shan F, Li J, Huang QY. HIF-1 alpha-induced up-regulation of miR-9 contributes to phenotypic modulation in pulmonary artery smooth muscle cells during hypoxia. J Cell Physiol 2014; 229:1511-20. [PMID: 24615545 DOI: 10.1002/jcp.24593] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2013] [Accepted: 02/20/2014] [Indexed: 12/12/2022]
Abstract
Pulmonary artery smooth muscle cells (PASMCs) are associated with the development of hypoxic pulmonary hypertension (HPH). Recent studies have implicated a critical role for microRNAs (miRNAs) in HPH; however, their expression and regulation in hypoxia-mediated phenotypic modulation of PASMCs remains largely unclear. Here, we report that miR-9 was induced in hypoxia and involved in a hypoxia-induced phenotypic switch in rat primary PASMCs. Knockdown of miR-9 followed by hypoxia exposure attenuated PASMCs proliferation and enhanced the expression of contractile genes in vascular smooth muscle cells (VSMCs), while overexpression of miR-9 in normoxia promoted a proliferative phenotype in PASMCs. The primary transcripts of miR-9-1 and miR-9-3, but not miR-9-2, increased dramatically after hypoxia, whereas silencing of the hypoxia-associated transcription factor HIF-1α following hypoxia exposure abolished the enhancement of both primary transcripts in PASMCs. Using in silico analysis, we found three putative HIF-1α binding motifs on miR-9-1 and one motif on miR-9-3 located within the 5-kb region upstream of the transcriptional start sites. Chromatin immunoprecipitation assay revealed that hypoxia enhanced the direct interaction between HIF-1α and the regulatory elements of miR-9-1 and miR-9-3. Reporter assays showed that the regulatory regions of miR-9-1 and miR-9-3 behaved as enhancers in a HIF-1α-dependent manner during hypoxia. Taken together, our data uncover a regulatory mechanism involving HIF-1α-mediated up-regulation of miR-9, which plays a role in the hypoxia-induced phenotypic switch of PASMCs.
Collapse
Affiliation(s)
- Fabo Shan
- Department of Pathophysiology and High Altitude Physiology, College of High Altitude Military Medicine, Chongqing, China; Key Laboratory of High Altitude Medicine, Ministry of Education, Chongqing, China; Key Laboratory of High Altitude Medicine, PLA, Third Military Medical University, Chongqing, China
| | | | | |
Collapse
|
198
|
Zhang X, Azhar G, Rogers SC, Foster SR, Luo S, Wei JY. Overexpression of p49/STRAP alters cellular cytoskeletal structure and gross anatomy in mice. BMC Cell Biol 2014; 15:32. [PMID: 25183317 PMCID: PMC4160719 DOI: 10.1186/1471-2121-15-32] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Accepted: 08/04/2014] [Indexed: 11/12/2022] Open
Abstract
Background The protein p49/STRAP (SRFBP1) is a transcription cofactor of serum response factor (SRF) which regulates cytoskeletal and muscle-specific genes. Results Two conserved domains were found in the p49/STRAP protein. The SRF-binding domain was at its N-terminus and was highly conserved among mammalian species, xenopus and zebrafish. A BUD22 domain was found at its C-terminus in three sequence databases. The BUD22 domain was conserved among mammalian p49/STRAP proteins, and yeast cellular morphogenesis proteins, which is involved in ribosome biogenesis that affects growth rate and cell size. The endogenous p49/SRAP protein was localized mainly in the nucleus but also widely distributed in the cytoplasm, and was in close proximity to the actin. Transfected GFP-p49/STRAP protein co-localized with nucleolin within the nucleolus. Overexpression of p49/STRAP reduced actin content in cultured cells and resulted in smaller cell size versus control cells. Increased expression of p49/STRAP in transgenic mice resulted in newborns with malformations, which included asymmetric abdominal and thoracic cavities, and substantial changes in cardiac morphology. p49/STRAP altered the expression of certain muscle-specific genes, including that of the SRF gene, which is a key regulator of cardiac genes at the developmental, structural and maintenance level and has two SRE binding sites. Conclusions Since p49/STRAP is a co-factor of SRF, our data suggest that p49/STRAP likely regulates cell size and morphology through SRF target genes. The function of its BUD22 domain warrants further investigation. The observed increase in p49/STRAP expression during cellular aging may contribute to observed morphological changes in senescence.
Collapse
Affiliation(s)
| | | | | | | | | | - Jeanne Y Wei
- Reynolds Institute on Aging & Department of Geriatrics, University of Arkansas for Medical Sciences, 4301 West Markham St, #748, Little Rock, AR 72205, USA.
| |
Collapse
|
199
|
Lechuga S, Baranwal S, Li C, Naydenov NG, Kuemmerle JF, Dugina V, Chaponnier C, Ivanov AI. Loss of γ-cytoplasmic actin triggers myofibroblast transition of human epithelial cells. Mol Biol Cell 2014; 25:3133-46. [PMID: 25143399 PMCID: PMC4196865 DOI: 10.1091/mbc.e14-03-0815] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Transdifferentiation of epithelial cells into mesenchymal cells and myofibroblasts plays an important role in tumor progression and tissue fibrosis. Such epithelial plasticity is accompanied by dramatic reorganizations of the actin cytoskeleton, although mechanisms underlying cytoskeletal effects on epithelial transdifferentiation remain poorly understood. In the present study, we observed that selective siRNA-mediated knockdown of γ-cytoplasmic actin (γ-CYA), but not β-cytoplasmic actin, induced epithelial-to-myofibroblast transition (EMyT) of different epithelial cells. The EMyT manifested by increased expression of α-smooth muscle actin and other contractile proteins, along with inhibition of genes responsible for cell proliferation. Induction of EMyT in γ-CYA-depleted cells depended on activation of serum response factor and its cofactors, myocardial-related transcriptional factors A and B. Loss of γ-CYA stimulated formin-mediated actin polymerization and activation of Rho GTPase, which appear to be essential for EMyT induction. Our findings demonstrate a previously unanticipated, unique role of γ-CYA in regulating epithelial phenotype and suppression of EMyT that may be essential for cell differentiation and tissue fibrosis.
Collapse
Affiliation(s)
- Susana Lechuga
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA 23298
| | - Somesh Baranwal
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA 23298
| | - Chao Li
- Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA 23298
| | - Nayden G Naydenov
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA 23298
| | - John F Kuemmerle
- Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA 23298
| | - Vera Dugina
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119991, Russia
| | - Christine Chaponnier
- Department of Pathology and Immunology, University Medical Center, University of Geneva, Geneva 4, Switzerland
| | - Andrei I Ivanov
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA 23298 Virginia Institute of Molecular Medicine, Virginia Commonwealth University, Richmond, VA 23298 VCU Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 23298
| |
Collapse
|
200
|
Hwang KC. The Role of MicroRNAs in Vascular Diseases; Smooth Muscle Cell Differentiation and De-Differentiation. Korean Circ J 2014; 44:218-9. [PMID: 25089132 PMCID: PMC4117841 DOI: 10.4070/kcj.2014.44.4.218] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Affiliation(s)
- Ki-Chul Hwang
- Severance Biomedical Science Institute, Cardiovascular Research Institute, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|