11
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      K Ca3.1 Channels Promote Cardiac Fibrosis Through Mediating Inflammation and Differentiation of Monocytes Into Myofibroblasts in Angiotensin II–Treated Rats

      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

          Background

          Cardiac fibrosis is a core pathological process associated with heart failure. The recruitment and differentiation of primitive fibroblast precursor cells of bone marrow origin play a critical role in pathological interstitial cardiac fibrosis. The K C a3.1 channels are expressed in both ventricular fibroblasts and circulating mononuclear cells in rats and are upregulated by angiotensin II. We hypothesized that K C a3.1 channels mediate the inflammatory microenvironment in the heart, promoting the infiltrated bone marrow–derived circulating mononuclear cells to differentiate into myofibroblasts, leading to myocardial fibrosis.

          Methods and Results

          We established a cardiac fibrosis model in rats by infusing angiotensin II to evaluate the impact of the specific K C a3.1 channel blocker TRAM‐34 on cardiac fibrosis. At the same time, mouse CD4 + T cells and rat circulating mononuclear cells were separated to investigate the underlying mechanism of the TRAM‐34 anti–cardiac fibrosis effect. TRAM‐34 significantly attenuated cardiac fibrosis and the inflammatory reaction and reduced the number of fibroblast precursor cells and myofibroblasts. Inhibition of K C a3.1 channels suppressed angiotensin II–stimulated expression and secretion of interleukin‐4 and interleukin‐13 in CD4 + T cells and interleukin‐4– or interleukin‐13–induced differentiation of monocytes into fibrocytes.

          Conclusions

          K C a3.1 channels facilitate myocardial inflammation and the differentiation of bone marrow‐derived monocytes into myofibroblasts in cardiac fibrosis caused by angiotensin II infusion.

          Related collections

          Most cited references37

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

          ACE inhibition, ACE2 and angiotensin-(1-7) axis in kidney and cardiac inflammation and fibrosis.

          The Renin Angiotensin System (RAS) is a pivotal physiological regulator of heart and kidney homeostasis, but also plays an important role in the pathophysiology of heart and kidney diseases. Recently, new components of the RAS have been discovered, including angiotensin converting enzyme 2 (ACE2), Angiotensin(Ang)-(1-7), Mas receptor, Ang-(1-9) and Alamandine. These new components of RAS are formed by the hydrolysis of Ang I and Ang II and, in general, counteract the effects of Ang II. In experimental models of heart and renal diseases, Ang-(1-7), Ang-(1-9) and Alamandine produced vasodilation, inhibition of cell growth, anti-thrombotic, anti-inflammatory and anti-fibrotic effects. Recent pharmacological strategies have been proposed to potentiate the effects or to enhance the formation of Ang-(1-7) and Ang-(1-9), including ACE2 activators, Ang-(1-7) in hydroxypropyl β-cyclodextrin, cyclized form of Ang-(1-7) and nonpeptide synthetic Mas receptor agonists. Here, we review the role and effects of ACE2, ACE2 activators, Ang-(1-7) and synthetic Mas receptor agonists in the control of inflammation and fibrosis in cardiovascular and renal diseases and as counter-regulators of the ACE-Ang II-AT1 axis. We briefly comment on the therapeutic potential of the novel members of RAS, Ang-(1-9) and alamandine, and the interactions between classical RAS inhibitors and new players in heart and kidney diseases.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Pivotal Advance: Th-1 cytokines inhibit, and Th-2 cytokines promote fibrocyte differentiation.

            CD14+ peripheral blood monocytes can differentiate into fibroblast-like cells called fibrocytes, which are associated with and are at least partially responsible for wound healing and fibrosis in multiple organ systems. Signals regulating fibrocyte differentiation are poorly understood. In this study, we find that when added to human PBMCs cultured in serum-free medium, the profibrotic cytokines IL-4 and IL-13 promote fibrocyte differentiation without inducing fibrocyte or fibrocyte precursor proliferation. We also find that the potent, antifibrotic cytokines IFN-gamma and IL-12 inhibit fibrocyte differentiation. In our culture system, IL-1beta, IL-3, IL-6, IL-7, IL-16, GM-CSF, M-CSF, fetal liver tyrosine kinase 3, insulin growth factor 1, vascular endothelial growth factor, and TNF-alpha had no significant effect on fibrocyte differentiation. IL-4, IL-13, and IFN-gamma act directly on monocytes to regulate fibrocyte differentiation, and IL-12 acts indirectly, possibly through CD16-positive NK cells. We previously identified the plasma protein serum amyloid P (SAP) as a potent inhibitor of fibrocyte differentiation. When added together, the fibrocyte-inhibitory activity of SAP dominates the profibrocyte activities of IL-4 and IL-13. The profibrocyte activities of IL-4 and IL-13 and the fibrocyte-inhibitory activities of IFN-gamma and IL-12 counteract each other in a concentration-dependent manner. These results indicate that the complex mix of cytokines and plasma proteins present in inflammatory lesions, wounds, and fibrosis will influence fibrocyte differentiation.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Bone marrow-derived fibroblast precursors mediate ischemic cardiomyopathy in mice.

              We previously described a mouse model of fibrotic ischemia/reperfusion cardiomyopathy (I/RC) arising from daily, brief coronary occlusion. One characteristic of I/RC was the prolonged elevation of monocyte chemoattractant protein 1 (MCP-1), which was obligate to its phenotype and may contribute to the uptake of bloodborne cells. Here we describe in I/RC hearts a population of small spindle-shaped fibroblasts that were highly proliferative and expressed collagen I and alpha-smooth muscle actin (myofibroblast markers), CD34 (a precursor marker), and CD45 (a hematopoietic marker). These cells represented 3% of all nonmyocyte live cells. To confirm the cells' bone marrow origin, chimeric mice were created by the rescue of irradiated C57BL/6 mice with marrow from ROSA26, a congenic line expressing lacZ. I/RC resulted in a large population of spindle-shaped fibroblasts containing lacZ. We postulated that the fibroblast precursors represented a developmental path for a subset of monocytes, whose phenotype we have shown to be influenced by serum amyloid P (SAP). Thus, we administered SAP in vivo, which markedly reduced the number of proliferative spindle-shaped fibroblasts and completely prevented I/RC-induced fibrosis and global ventricular dysfunction. By contrast, SAP did not suppress the inflammation or chemokine expression seen in I/RC. SAP, a member of the pentraxin family, binds to Fcgamma receptors and modifies the pathophysiological function of monocytes. Our data suggest that SAP interferes with assumption of a fibroblast phenotype in a subset of monocytes and that SAP may be an important regulator in the linkage between inflammation and nonadaptive fibrosis in the heart.
                Bookmark

                Author and article information

                Contributors
                dengxl@mail.xjtu.edu.cn
                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
                19 December 2018
                08 January 2019
                : 8
                : 1 ( doiID: 10.1002/jah3.2019.8.issue-1 )
                : e010418
                Affiliations
                [ 1 ] Department of Physiology and Pathophysiology School of Basic Medical Sciences Xi'an Jiaotong University Health Science Center Xi'an Shaanxi China
                [ 2 ] Department of Pathology School of Basic Medical Sciences Xi'an Jiaotong University Health Science Center Xi'an Shaanxi China
                [ 3 ] Basic Experiment Teaching Center School of Basic Medical Sciences Xi'an Jiaotong University Health Science Center Xi'an Shaanxi China
                [ 4 ] Cardiovascular Research Centre School of Basic Medical Sciences Xi'an Jiaotong University Health Science Center Xi'an Shaanxi China
                [ 5 ] Department of Pathology Xi'an Guangren Hospital Affiliated to Xi'an Jiaotong University Health Science Center Xi'an Shaanxi China
                [ 6 ] Baker Heart and Diabetes Institute Melbourne Victoria Australia
                Author notes
                [*] [* ] Correspondence to: Xiu‐Ling Deng, MD, PhD, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an, 710061 Shaanxi, China. E‐mail: dengxl@ 123456mail.xjtu.edu.cn
                Article
                JAH33731
                10.1161/JAHA.118.010418
                6405723
                30563389
                68b10b89-0ff5-4d79-98b6-f69772419582
                © 2018 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-nd/4.0/ License, which permits use and distribution in any medium, provided the original work is properly cited, the use is non‐commercial and no modifications or adaptations are made.

                History
                : 24 July 2018
                : 14 November 2018
                Page count
                Figures: 9, Tables: 0, Pages: 17, Words: 8715
                Funding
                Funded by: National Natural Science Foundation of China
                Award ID: 81570214
                Award ID: 81370191
                Award ID: 81600656
                Categories
                Original Research
                Original Research
                Heart Failure
                Custom metadata
                2.0
                jah33731
                08 January 2019
                Converter:WILEY_ML3GV2_TO_NLMPMC version:version=5.5.4 mode:remove_FC converted:08.01.2019

                Cardiovascular Medicine
                angiotensin ii,cardiac remodeling,differentiation,inflammation,k‐channel,fibrosis,ion channels/membrane transport,pathophysiology,mechanisms

                Comments

                Comment on this article