36
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Phenotypic switching of vascular smooth muscle cells in the ‘normal region’ of aorta from atherosclerosis patients is regulated by miR‐145

      research-article

      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.

          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 micro RNA‐145 ( miR‐145), its target gene Kruppel‐Like Factor 5 ( KLF5) and Myocardin ( MYOCD, a smooth muscle‐specific transcriptional coactivator) were analysed using RTqPCR, 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.

          Related collections

          Most cited references37

          • Record: found
          • Abstract: found
          • Article: not found

          Switching from repression to activation: microRNAs can up-regulate translation.

          AU-rich elements (AREs) and microRNA target sites are conserved sequences in messenger RNA (mRNA) 3' untranslated regions (3'UTRs) that control gene expression posttranscriptionally. Upon cell cycle arrest, the ARE in tumor necrosis factor-alpha (TNFalpha) mRNA is transformed into a translation activation signal, recruiting Argonaute (AGO) and fragile X mental retardation-related protein 1 (FXR1), factors associated with micro-ribonucleoproteins (microRNPs). We show that human microRNA miR369-3 directs association of these proteins with the AREs to activate translation. Furthermore, we document that two well-studied microRNAs-Let-7 and the synthetic microRNA miRcxcr4-likewise induce translation up-regulation of target mRNAs on cell cycle arrest, yet they repress translation in proliferating cells. Thus, activation is a common function of microRNPs on cell cycle arrest. We propose that translation regulation by microRNPs oscillates between repression and activation during the cell cycle.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            miR-145 and miR-143 Regulate Smooth Muscle Cell Fate Decisions

            SUMMARY microRNAs are regulators of myriad cellular events, but evidence for a single microRNA that can efficiently differentiate multipotent cells into a specific lineage or regulate direct reprogramming of cells into an alternate cell fate has been elusive. Here, we show that miR-145 and miR-143 are co-transcribed in multipotent cardiac progenitors before becoming localized to smooth muscle cells, including neural crest stem cell–derived vascular smooth muscle cells. miR-145 and miR-143 were direct transcriptional targets of serum response factor, myocardin and Nkx2.5, and were downregulated in injured or atherosclerotic vessels containing proliferating, less differentiated smooth muscle cells. miR-145 was necessary for myocardin-induced reprogramming of adult fibroblasts into smooth muscle cells and sufficient to induce differentiation of multipotent neural crest stem cells into vascular smooth muscle. Furthermore, miR-145 and miR-143 cooperatively targeted a network of transcription factors, including Klf4, myocardin, and Elk-1 to promote differentiation and repress proliferation of smooth muscle cells. These findings demonstrate that miR-145 can direct the smooth muscle fate and that miR-145 and miR-143 function to regulate the quiescent versus proliferative phenotype of smooth muscle cells.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Role of microRNAs in vascular diseases, inflammation, and angiogenesis.

              The integrity of the endothelial monolayer is fundamental for the homoeostasis of the vascular system. Functional endothelial cells are also required for the growth of new blood vessels during neovascularization. Although multiple growth factors have been shown to regulate angiogenesis and vascular development, little is known about the complex upstream regulation of gene expression and translation. MicroRNAs (miRNAs) are an emerging class of highly conserved, non-coding small RNAs that regulate gene expression on the post-transcriptional level by inhibiting the translation of protein from mRNA or by promoting the degradation of mRNA. More than 500 human miRNAs have been identified so far, and increasing evidence indicates that miRNAs have distinct expression profiles and play crucial roles in various physiological and pathological processes such as cardiogenesis, haematopoietic lineage differentiation, and oncogenesis. Meanwhile, a few specific miRNAs that regulate endothelial cell functions and angiogenesis have been described. Let7-f, miR-27b, and mir-130a were identified as pro-angiogenic miRNAs. In contrast, miR-221 and miR-222 inhibit endothelial cell migration, proliferation, and angiogenesis in vitro by targeting the stem cell factor receptor c-kit and indirectly regulating endothelial nitric oxide synthase expression. Moreover, some miRNAs are involved in tumour angiogenesis such as the miR-17-92 cluster and miR-378. Early studies also indicate the contribution of specific miRNAs (e.g. miR-155, miR-21, and miR-126) to vascular inflammation and diseases. Thus, the identification of miRNAs and their respective targets may offer new therapeutic strategies to treat vascular diseases such as atherosclerosis, to improve neovascularization after ischaemia, or to prevent tumour progression.
                Bookmark

                Author and article information

                Journal
                J Cell Mol Med
                J. Cell. Mol. Med
                10.1111/(ISSN)1582-4934
                JCMM
                Journal of Cellular and Molecular Medicine
                John Wiley and Sons Inc. (Hoboken )
                1582-1838
                1582-4934
                15 March 2016
                June 2016
                : 20
                : 6 ( doiID: 10.1111/jcmm.2016.20.issue-6 )
                : 1049-1061
                Affiliations
                [ 1 ] Department of Cardiovascular SurgerySecond Affiliated Hospital of Harbin Medical University HarbinChina
                [ 2 ] Key Laboratories of Myocardial Ischemia Mechanism and Treatment Harbin Medical UniversityMinistry of Education HarbinChina
                [ 3 ] Toronto General Research Institute University Health Network and Department of Surgery Division of Cardiac SurgeryUniversity of Toronto Toronto ONCanada
                Author notes
                [*] [* ] Correspondence to: Hai TIAN, M.D., Ph.D. or Ren‐Ke LI, M.D., Ph.D.

                E‐mail: doctor_tianhai@ 123456163.com (or) renkeli@ 123456uhnres.utoronto.ca

                Article
                JCMM12825
                10.1111/jcmm.12825
                4882986
                26992033
                d10e66b0-db57-4534-9640-de4a71f05f46
                © 2016 The Authors. Journal of Cellular and Molecular Medicine published by John Wiley & Sons Ltd and Foundation for Cellular and Molecular Medicine.

                This is an open access article under the terms of the Creative Commons Attribution License, which permits use, distribution and reproduction in any medium, provided the original work is properly cited.

                History
                : 06 October 2015
                : 04 February 2016
                Page count
                Pages: 13
                Funding
                Funded by: National Natural Science Foundation of China
                Award ID: 81270188
                Funded by: Natural Science Foundation of Heilongjiang Province China
                Award ID: JC2015020
                Funded by: Public Health Foundation of Heilongjiang Province China
                Award ID: 2013054
                Categories
                Original Article
                Original Articles
                Custom metadata
                2.0
                jcmm12825
                June 2016
                Converter:WILEY_ML3GV2_TO_NLMPMC version:4.9.8 mode:remove_FC converted:28.11.2016

                Molecular medicine
                mir‐145,vsmcs,atherosclerosis,proliferation,phenotypic switch
                Molecular medicine
                mir‐145, vsmcs, atherosclerosis, proliferation, phenotypic switch

                Comments

                Comment on this article