6
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      PLF‐1 (Proliferin‐1) Modulates Smooth Muscle Cell Proliferation and Development of Experimental Intimal Hyperplasia

      research-article
      , MD, PhD 1 , 2 , 3 , , MD, PhD 4 , , MD, PhD 5 , , MD, PhD 6 , , MD, PhD 5 , , PhD 3 , 10 , , MD, PhD 7 , , PhD 8 , , MD, PhD 2 , , MD, PhD 2 , , MD, PhD 2 , 3 , , MD, PhD 2 , , MD, PhD 2 , , MD, PhD 5 , , DSc 9 , , MD, PhD 5 , , MD, PhD 3 , 10 , , MD, PhD, FAHA 2 , 3 , 7 , 10 ,
      Journal of the American Heart Association: Cardiovascular and Cerebrovascular Disease
      John Wiley and Sons Inc.
      hyperplasia, proliferation, vascular remodeling, vascular smooth muscle, Animal Models of Human Disease, Basic Science Research

      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

          Background

          Although apoptosis and cell proliferation have been extensively investigated in atherosclerosis and restenosis postinjury, the communication between these 2 cellular events has not been evaluated. Here, we report an inextricable communicative link between apoptosis and smooth muscle cell proliferation in the promotion of vascular remodeling postinjury.

          Methods and Results

          Cathepsin K–mediated caspase‐8 maturation is a key initial step for oxidative stress–induced smooth muscle cell apoptosis. Apoptotic cells generate a potential growth‐stimulating signal to facilitate cellular mass changes in response to injury. One downstream mediator that cathepsin K regulates is PLF‐1 (proliferin‐1), which can potently stimulate growth of surviving neighboring smooth muscle cells through activation of PI3K/Akt/p38MAPK (phosphatidylinositol 3‐kinase/protein kinase B/p38 mitogen‐activated protein kinase)‐dependent and ‐independent mTOR (mammalian target of rapamycin) signaling cascades. We observed that cathepsin K deficiency substantially mitigated neointimal hyperplasia by reduction of Toll‐like receptor‐2/caspase‐8–mediated PLF‐1 expression. Interestingly, PLF‐1 blocking, with its neutralizing antibody, suppressed neointima formation and remodeling in response to injury in wild‐type mice. Contrarily, administration of recombinant mouse PLF‐1 accelerated injury‐induced vascular actions.

          Conclusions

          This is the first study detailing PLF‐1 as a communicator between apoptosis and proliferation during injury‐related vascular remodeling and neointimal hyperplasia. These data suggested that apoptosis‐driven expression of PLF‐1 is thus a novel target for treatment of apoptosis‐based hyperproliferative disorders.

          Related collections

          Most cited references36

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

          Expression of the elastolytic cathepsins S and K in human atheroma and regulation of their production in smooth muscle cells.

          Formation of the atherosclerotic intima must involve altered metabolism of the elastin-rich arterial extracellular matrix. Proteases potentially involved in these processes remain unclear. This study examined the expression of the potent elastases cathepsins S and K in human atheroma. Normal arteries contained little or no cathepsin K or S. In contrast, macrophages in atheroma contained abundant immunoreactive cathepsins K and S. Intimal smooth muscle cells (SMC), especially cells appearing to traverse the internal elastic laminae, also contained these enzymes. Extracts of atheromatous tissues had approximately twofold greater elastase-specific activity than extracts of uninvolved arteries, mostly due to cysteine proteases. Cultured human SMC displayed no immunoreactive cathepsins K and S and exhibited little or no elastolytic activity when incubated with insoluble elastin. SMC stimulated with the atheroma-associated cytokines IL-1beta or IFN-gamma secreted active cathepsin S and degraded substantial insoluble elastin (15-20 microg/10(6) cells/24 h). A selective inhibitor of cathepsin S blocked > 80% of this elastolytic activity. The presence of cathepsins K and S at sites of vascular matrix remodeling and the ability of SMC and macrophages to use these enzymes to degrade elastin supports a role for elastolytic cathepsins in vessel wall remodeling and identifies novel therapeutic targets in regulating plaque stability.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Lysosomal cysteine proteases: more than scavengers.

            Lysosomal cysteine proteases were believed to be mainly involved in intracellular protein degradation. Under special conditions they have been found outside lysosomes resulting in pathological conditions. With the discovery of a series of new cathepsins with restricted tissue distributions, it has become evident that these enzymes must be involved in a range of specific cellular tasks much broader than as simple housekeeping enzymes. It is therefore timely to review and discuss the various physiological roles of mammalian lysosomal papain-like cysteine proteases as well as their mechanisms of action and the regulation of their activity.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Cathepsin S controls angiogenesis and tumor growth via matrix-derived angiogenic factors.

              The cysteine protease cathepsin S is highly expressed in malignant tissues. By using a mouse model of multistage murine pancreatic islet cell carcinogenesis in which cysteine cathepsin activity has been functionally implicated, we demonstrated that selective cathepsin S deficiency impaired angiogenesis and tumor cell proliferation, thereby impairing angiogenic islet formation and the growth of solid tumors, whereas the absence of its endogenous inhibitor cystatin C resulted in opposite phenotypes. Although mitogenic vascular endothelial growth factor, transforming growth factor-beta1, and the anti-angiogenic endostatin levels in either serum or carcinoma tissue extracts did not change in cathepsin S- or cystatin C-null mice, tumor tissue basic fibroblast growth factor and serum type 1 insulin growth factor levels were higher in cystatin C-null mice, and serum type 1 insulin growth factor levels were also increased in cathepsin S-null mice. Furthermore, cathepsin S affected the production of type IV collagen-derived anti-angiogenic peptides and the generation of bioactive pro-angiogenic gamma2 fragments from laminin-5, revealing a functional role for cathepsin S in angiogenesis and neoplastic progression.
                Bookmark

                Author and article information

                Contributors
                chengxw0908@163.com , xianwu@med.nagoya-u.ac.jp
                Journal
                J Am Heart Assoc
                J Am Heart Assoc
                10.1002/(ISSN)2047-9980
                JAH3
                ahaoa
                Journal of the American Heart Association: Cardiovascular and Cerebrovascular Disease
                John Wiley and Sons Inc. (Hoboken )
                2047-9980
                16 December 2019
                17 December 2019
                : 8
                : 24 ( doiID: 10.1002/jah3.v8.24 )
                : e005886
                Affiliations
                [ 1 ] Department of Public Health Guilin Medical College Guilin Guangxi China
                [ 2 ] Department of Cardiology/Hypertension and Heart Center Yanbian University Hospital Yanji Jilin China
                [ 3 ] Department of Community & Geriatrics Nagoya University Graduate School of Medicine Nagoya Japan
                [ 4 ] Department of Neurology Occupational and Environmental Health Kitakyushu Hukuoka Japan
                [ 5 ] Department of Cardiology Nagoya University Graduate School of Medicine Nagoya Japan
                [ 6 ] Department of Cardiology Shanghai General Hospital Shanghai Jiao Tong University School of Medicine Shanghai China
                [ 7 ] Division of Cardiology Department of Internal Medicine Kyung Hee University Seoul South Korea
                [ 8 ] Department of Physiology and Pathophysiology Yanbian University School of Medicine Yanji Jinlin China
                [ 9 ] Department of Cardiovascular Medicine Brigham and Women's Hospital and Harvard Medical School Boston MA
                [ 10 ] Institute of Innovation for Future Society Nagoya University Nagoya Japan
                Author notes
                [*] [* ] Correspondence to: Xian Wu Cheng, MD, PhD, FAHA, Department of Cardiology/Hypertension and Heart Center, Yanbian University Hospital, Juzijie 1327, Yanji, Jilin 133000, China or Institute of Innovation for Future Society, Nagoya University Graduate School of Medicine, 65 Tsuruma‐cho, Showa‐ku, Nagoya 466‐8550, Japan. E‐mails: chengxw0908@ 123456163.com or xianwu@ 123456med.nagoya-u.ac.jp
                [†]

                Dr Hu and Dr Huang contributed equally to this study.

                Article
                JAH33259
                10.1161/JAHA.117.005886
                6951060
                31838975
                26cd953c-7ccd-4af2-aa75-6108c9ad8ccf
                © 2019 The Authors. Published on behalf of the American Heart Association, Inc., by Wiley.

                This is an open access article under the terms of the http://creativecommons.org/licenses/by-nc/4.0/ License, which permits use, distribution and reproduction in any medium, provided the original work is properly cited and is not used for commercial purposes.

                History
                : 15 February 2017
                : 22 March 2018
                Page count
                Figures: 12, Tables: 0, Pages: 17, Words: 9802
                Funding
                Funded by: Scientific Research Fund of the Chinese Ministry of Education
                Award ID: 81260068
                Award ID: 81560240
                Award ID: 81660240
                Award ID: 81770485
                Award ID: 81760091
                Funded by: Ministry of Education, Culture, Sports, Science and Technology of Japan
                Award ID: 15H04801
                Award ID: 15H04802
                Funded by: Japan Society for Promotion of Science
                Award ID: 26‐04418
                Categories
                Original Research
                Original Research
                Vascular Medicine
                Custom metadata
                2.0
                jah33259
                17 December 2019
                Converter:WILEY_ML3GV2_TO_NLMPMC version:5.7.2 mode:remove_FC converted:16.12.2019

                Cardiovascular Medicine
                hyperplasia,proliferation,vascular remodeling,vascular smooth muscle,animal models of human disease,basic science research

                Comments

                Comment on this article