2
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      HDAC6 Regulates the MRTF-A/SRF Axis and Vascular Smooth Muscle Cell Plasticity

      research-article
      , MD, MS a , b , , , MD, PhD a , c , , , MD d , , , PhD c , d , , BS a , , MD c , ∗∗ , , PhD a ,
      JACC: Basic to Translational Science
      Elsevier
      dedifferentiation, HDAC6, MRTF-A, SRF, vascular smooth muscle cell, SMA, smooth muscle actin, DMEM, Dulbecco’s modified Eagle’s medium, DNA, deoxyribonucleic acid, EEL, external elastic lamina, FBS, fetal bovine serum, HDAC, histone deacetylase, IEL, internal elastic lamina, IgG, immunoglobulin G, IH, intimal hyperplasia, MMP, matrix metalloproteinase, MRTF-A, myocardin-related transcription factor A, PDGF-BB, platelet-derived growth factor-BB, siRNA, small interfering ribonucleic acid, SMC, vascular smooth muscle cell, SMHC, smooth muscle myosin heavy chain, SRF, serum response factor, TNF, tumor necrosis factor, TSA, trichostatin A

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Visual Abstract

          Highlights

          • Distinct from other histone deacetylases, HDAC6 primarily resides in the cytosol.

          • Unexpectedly, HDAC6-selective inhibition (or silencing) enhances the nuclear activity of SRF.

          • HDAC6 inhibition elevates acetylation and protein levels of myocardin-related transcription factor A, a cytoplasmic-nuclear shuttling co-activator of SRF. Myocardin-related transcription factor A/SRF are known to critically regulate vascular smooth muscle cell phenotypic stability.

          • HDAC6 inhibition prevents smooth muscle cell dedifferentiation in vitro and reduces neointima and restenosis in vivo.

          Summary

          Cellular plasticity is fundamental in biology and disease. Vascular smooth muscle cell (SMC) dedifferentiation (loss of contractile proteins) initiates and perpetrates vascular pathologies such as restenosis. Contractile gene expression is governed by the master transcription factor, serum response factor (SRF). Unlike other histone deacetylases, histone deacetylase 6 (HDAC6) primarily resides in the cytosol. Whether HDAC6 regulates SRF nuclear activity was previously unknown in any cell type. This study found that selective inhibition of HDAC6 with tubastatin A preserved the contractile protein (alpha-smooth muscle actin) that was otherwise diminished by platelet-derived growth factor-BB. Tubastatin A also enhanced SRF transcriptional (luciferase) activity, and this effect was confirmed by HDAC6 knockdown. Interestingly, HDAC6 inhibition increased acetylation and total protein of myocardin-related transcription factor A (MRTF-A), a transcription co-activator known to translocate from the cytosol to the nucleus, thereby activating SRF. Consistently, HDAC6 co-immunoprecipitated with MRTF-A. In vivo studies showed that tubastatin A treatment of injured rat carotid arteries mitigated neointimal lesion, which is known to be formed largely by dedifferentiated SMCs. This report is the first to show HDAC6 regulation of the MRTF-A/SRF axis and SMC plasticity, thus opening a new perspective for interventions of vascular pathologies.

          Related collections

          Most cited references19

          • Record: found
          • Abstract: found
          • Article: found
          Is Open Access

          HDAC and HDAC Inhibitor: From Cancer to Cardiovascular Diseases

          Histone deacetylases (HDACs) are epigenetic regulators that regulate the histone tail, chromatin conformation, protein-DNA interaction, and even transcription. HDACs are also post-transcriptional modifiers that regulate the protein acetylation implicated in several pathophysiologic states. HDAC inhibitors have been highlighted as a novel category of anti-cancer drugs. To date, four HDAC inhibitors, Vorinostat, Romidepsin, Panobinostat, and Belinostat, have been approved by the United States Food and Drug Administration. Principally, these HDAC inhibitors are used for hematologic cancers in clinic with less severe side effects. Clinical trials are continuously expanding to address other types of cancer and also nonmalignant diseases. HDAC inhibition also results in beneficial outcomes in various types of neurodegenerative diseases, inflammation disorders, and cardiovascular diseases. In this review, we will briefly discuss 1) the roles of HDACs in the acquisition of a cancer's phenotype and the general outcome of the HDAC inhibitors in cancer, 2) the functional relevance of HDACs in cardiovascular diseases and the possible therapeutic implications of HDAC inhibitors in cardiovascular disease.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Histone deacetylase 3 is critical in endothelial survival and atherosclerosis development in response to disturbed flow.

            Histone deacetylase 3 (HDAC3) is known to play a crucial role in the differentiation of endothelial progenitors. The role of HDAC3 in mature endothelial cells, however, is not well understood. Here, we investigated the function of HDAC3 in preserving endothelial integrity in areas of disturbed blood flow, ie, bifurcation areas prone to atherosclerosis development. En face staining of aortas from apolipoprotein E-knockout mice revealed increased expression of HDAC3, specifically in these branching areas in vivo, whereas rapid upregulation of HDAC3 protein was observed in endothelial cells exposed to disturbed flow in vitro. Interestingly, phosphorylation of HDAC3 at serine/threonine was observed in these cells, suggesting that disturbed flow leads to posttranscriptional modification and stabilization of the HDAC3 protein. Coimmunoprecipitation experiments showed that HDAC3 and Akt form a complex. Using a series of constructs harboring deletions, we found residues 136 to 206 of HDAC3 to be crucial in this interaction. Enforced expression of HDAC3 resulted in increased phosphorylation of Akt and upregulation of its kinase activity. In line with these findings, knockdown of HDAC3 with lentiviral vectors (shHDAC3) led to a dramatic decrease in cell survival accompanied by apoptosis in endothelial cells. In aortic isografts of apolipoprotein E-knockout mice treated with shHDAC3, a robust atherosclerotic lesion was formed. Surprisingly, 3 of the 8 mice that received shHDAC3-infected grafts died within 2 days after the operation. Miller staining of the isografts revealed disruption of the basement membrane and rupture of the vessel. Our findings demonstrated that HDAC3 serves as an essential prosurvival molecule with a critical role in maintaining the endothelial integrity via Akt activation and that severe atherosclerosis and vessel rupture in isografted vessels of apolipoprotein E-knockout mice occur when HDAC3 is knocked down.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Epigenetic regulation of vascular smooth muscle cell proliferation and neointima formation by histone deacetylase inhibition.

              Proliferation of smooth muscle cells (SMC) in response to vascular injury is central to neointimal vascular remodeling. There is accumulating evidence that histone acetylation constitutes a major epigenetic modification for the transcriptional control of proliferative gene expression; however, the physiological role of histone acetylation for proliferative vascular disease remains elusive. In the present study, we investigated the role of histone deacetylase (HDAC) inhibition in SMC proliferation and neointimal remodeling. We demonstrate that mitogens induce transcription of HDAC 1, 2, and 3 in SMC. Short interfering RNA-mediated knockdown of either HDAC 1, 2, or 3 and pharmacological inhibition of HDAC prevented mitogen-induced SMC proliferation. The mechanisms underlying this reduction of SMC proliferation by HDAC inhibition involve a growth arrest in the G(1) phase of the cell cycle that is due to an inhibition of retinoblastoma protein phosphorylation. HDAC inhibition resulted in a transcriptional and posttranscriptional regulation of the cyclin-dependent kinase inhibitors p21(Cip1) and p27(Kip). Furthermore, HDAC inhibition repressed mitogen-induced cyclin D1 mRNA expression and cyclin D1 promoter activity. As a result of this differential cell cycle-regulatory gene expression by HDAC inhibition, the retinoblastoma protein retains a transcriptional repression of its downstream target genes required for S phase entry. Finally, we provide evidence that these observations are applicable in vivo by demonstrating that HDAC inhibition decreased neointima formation and expression of cyclin D1 in a murine model of vascular injury. These findings identify HDAC as a critical component of a transcriptional cascade regulating SMC proliferation and suggest that HDAC might play a pivotal role in the development of proliferative vascular diseases, including atherosclerosis and in-stent restenosis.
                Bookmark

                Author and article information

                Contributors
                Journal
                JACC Basic Transl Sci
                JACC Basic Transl Sci
                JACC: Basic to Translational Science
                Elsevier
                2452-302X
                31 December 2018
                December 2018
                31 December 2018
                : 3
                : 6
                : 782-795
                Affiliations
                [a ]Department of Surgery and Department of Physiology and Cell Biology, College of Medicine, and the Davis Heart and Lung Research Institute, Wexner Medical Center, The Ohio State University, Columbus, Ohio
                [b ]Cellular and Molecular Pathology Graduate Program, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin
                [c ]Department of Surgery, College of Medicine, and the Davis Heart and Lung Research Institute, Wexner Medical Center, The Ohio State University, Columbus, Ohio
                [d ]Department of Surgery, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin
                Author notes
                [] Address for correspondence: Dr. Lian-Wang Guo, Department of Surgery and Department of Physiology and Cell Biology, The Ohio State University, Columbus, Ohio 43210. lianwang.guo@ 123456osumc.edu
                [∗∗ ]Dr. K. Craig Kent, Department of Surgery, College of Medicine, Davis Heart and Lung Research Institute, Wexner Medical Center, The Ohio State University, Columbus, Ohio 43210. kc.kent@ 123456osumc.edu
                [∗]

                Drs. Zhang, Urabe, and Little contributed equally to this work and are joint first authors.

                Article
                S2452-302X(18)30232-8
                10.1016/j.jacbts.2018.08.010
                6314972
                30623138
                90157538-0967-4e54-8e6d-eb4ba60dd8fb
                © 2018 The Authors

                This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/).

                History
                : 5 July 2018
                : 31 July 2018
                : 23 August 2018
                Categories
                PRECLINICAL RESEARCH

                dedifferentiation,hdac6,mrtf-a,srf,vascular smooth muscle cell,sma, smooth muscle actin,dmem, dulbecco’s modified eagle’s medium,dna, deoxyribonucleic acid,eel, external elastic lamina,fbs, fetal bovine serum,hdac, histone deacetylase,iel, internal elastic lamina,igg, immunoglobulin g,ih, intimal hyperplasia,mmp, matrix metalloproteinase,mrtf-a, myocardin-related transcription factor a,pdgf-bb, platelet-derived growth factor-bb,sirna, small interfering ribonucleic acid,smc, vascular smooth muscle cell,smhc, smooth muscle myosin heavy chain,srf, serum response factor,tnf, tumor necrosis factor,tsa, trichostatin a

                Comments

                Comment on this article