201
|
Broughton KM, Sussman MA. Adult Cardiomyocyte Cell Cycle Detour: Off-ramp to Quiescent Destinations. Trends Endocrinol Metab 2019; 30:557-567. [PMID: 31262545 PMCID: PMC6703820 DOI: 10.1016/j.tem.2019.05.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 05/24/2019] [Accepted: 05/30/2019] [Indexed: 02/06/2023]
Abstract
Ability to promote completion of mitotic cycling of adult mammalian cardiomyocytes remains an intractable and vexing challenge, despite being one of the most sought after 'holy grails' of cardiovascular research. While some of the struggle is attributable to adult cardiomyocytes themselves that are notoriously post-mitotic, another contributory factor rests with difficulty in definitive tracking of adult cardiomyocyte cell cycle and lack of rigorous measures to track proliferation in situ. This review summarizes past, present, and future directions to promote adult mammalian cardiomyocyte cell cycle progression, proliferation, and renewal. Establishing relationship(s) between cardiomyocyte cell cycle progression and cellular biological properties is sorely needed to understand the mechanistic basis for cardiomyocyte cell cycle withdrawal to enhance cardiomyocyte cell cycle progression and mitosis.
Collapse
Affiliation(s)
- Kathleen M Broughton
- San Diego State University, Department of Biology and Integrated Regenerative Research Institute, San Diego, CA 92182, USA
| | - Mark A Sussman
- San Diego State University, Department of Biology and Integrated Regenerative Research Institute, San Diego, CA 92182, USA.
| |
Collapse
|
202
|
Shibamoto M, Higo T, Naito AT, Nakagawa A, Sumida T, Okada K, Sakai T, Kuramoto Y, Yamaguchi T, Ito M, Masumura Y, Higo S, Lee JK, Hikoso S, Komuro I, Sakata Y. Activation of DNA Damage Response and Cellular Senescence in Cardiac Fibroblasts Limit Cardiac Fibrosis After Myocardial Infarction. Int Heart J 2019; 60:944-957. [DOI: 10.1536/ihj.18-701] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Affiliation(s)
- Masato Shibamoto
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine
| | - Tomoaki Higo
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine
| | | | - Akito Nakagawa
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine
| | | | - Katsuki Okada
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine
| | - Taku Sakai
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine
| | - Yuki Kuramoto
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine
| | - Toshihiro Yamaguchi
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo
| | - Masamichi Ito
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo
| | - Yuki Masumura
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine
| | - Shuichirou Higo
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine
| | - Jong-Kook Lee
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine
- Department of Advanced Cardiovascular Regenerative Medicine, Osaka University Graduate School of Medicine
| | - Shungo Hikoso
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine
| | - Issei Komuro
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo
| | - Yasushi Sakata
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine
| |
Collapse
|
203
|
Scharf GM, Kilian K, Cordero J, Wang Y, Grund A, Hofmann M, Froese N, Wang X, Kispert A, Kist R, Conway SJ, Geffers R, Wollert KC, Dobreva G, Bauersachs J, Heineke J. Inactivation of Sox9 in fibroblasts reduces cardiac fibrosis and inflammation. JCI Insight 2019; 5:126721. [PMID: 31310588 DOI: 10.1172/jci.insight.126721] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Fibrotic scarring drives the progression of heart failure after myocardial infarction (MI). Therefore, the development of specific treatment regimens to counteract fibrosis is of high clinical relevance. The transcription factor SOX9 functions as an important regulator during embryogenesis, but recent data point towards an additional causal role in organ fibrosis. We show here that SOX9 is upregulated in the scar after MI in mice. Fibroblast specific deletion of Sox9 ameliorated MI-induced left ventricular dysfunction, dilatation and myocardial scarring in vivo. Unexpectedly, deletion of Sox9 also potently eliminated persisting leukocyte infiltration of the scar in the chronic phase after MI. RNA-sequencing from the infarct scar revealed that Sox9 deletion in fibroblasts resulted in strongly downregulated expression of genes related to extracellular matrix, proteolysis and inflammation. Importantly, Sox9 deletion in isolated cardiac fibroblasts in vitro similarly affected gene expression as in the cardiac scar and reduced fibroblast proliferation, migration and contraction capacity. Together, our data demonstrate that fibroblast SOX9 functions as a master regulator of cardiac fibrosis and inflammation and might constitute a novel therapeutic target during MI.
Collapse
Affiliation(s)
- Gesine M Scharf
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany.,Department of Cardiovascular Research and
| | - Katja Kilian
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Julio Cordero
- Department of Anatomy and Developmental Biology Center for Biomedicine and Medical Technology Mannheim, European Center for Angioscience, Medical Faculty Mannheim of Heidelberg University, Mannheim, Germany
| | - Yong Wang
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany.,Division of Molecular and Translational Cardiology, Department of Cardiology and Angiology, and
| | - Andrea Grund
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany.,Department of Cardiovascular Research and
| | - Melanie Hofmann
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Natali Froese
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Xue Wang
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Andreas Kispert
- Institute of Molecular Biology, Hannover Medical School, Hannover, Germany
| | - Ralf Kist
- Institute of Genetic Medicine, Faculty of Medical Sciences, and Centre for Oral Health Research, School of Dental Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Simon J Conway
- HB Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Robert Geffers
- Helmholtz Center for Infection Research, Braunschweig, Germany
| | - Kai C Wollert
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany.,Division of Molecular and Translational Cardiology, Department of Cardiology and Angiology, and
| | - Gergana Dobreva
- Department of Anatomy and Developmental Biology Center for Biomedicine and Medical Technology Mannheim, European Center for Angioscience, Medical Faculty Mannheim of Heidelberg University, Mannheim, Germany.,German Center for Cardiovascular Research partner site Mannheim/Heidelberg, Heidelberg, Germany
| | - Johann Bauersachs
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Joerg Heineke
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany.,Department of Cardiovascular Research and.,German Center for Cardiovascular Research partner site Mannheim/Heidelberg, Heidelberg, Germany
| |
Collapse
|
204
|
Ebrahimighaei R, McNeill MC, Smith SA, Wray JP, Ford KL, Newby AC, Bond M. Elevated cyclic-AMP represses expression of exchange protein activated by cAMP (EPAC1) by inhibiting YAP-TEAD activity and HDAC-mediated histone deacetylation. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2019; 1866:1634-1649. [PMID: 31255721 DOI: 10.1016/j.bbamcr.2019.06.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 06/19/2019] [Accepted: 06/26/2019] [Indexed: 02/05/2023]
Abstract
Ligand-induced activation of Exchange Protein Activated by cAMP-1 (EPAC1) is implicated in numerous physiological and pathological processes, including cardiac fibrosis where changes in EPAC1 expression have been detected. However, little is known about how EPAC1 expression is regulated. Therefore, we investigated regulation of EPAC1 expression by cAMP in cardiac fibroblasts. Elevation of cAMP using forskolin, cAMP-analogues or adenosine A2B-receptor activation significantly reduced EPAC1 mRNA and protein levels and inhibited formation of F-actin stress fibres. Inhibition of actin polymerisation with cytochalasin-D, latrunculin-B or the ROCK inhibitor, Y-27632, mimicked effects of cAMP on EPAC1 mRNA and protein levels. Elevated cAMP also inhibited activity of an EPAC1 promoter-reporter gene, which contained a consensus binding element for TEAD, which is a target for inhibition by cAMP. Inhibition of TEAD activity using siRNA-silencing of its co-factors YAP and TAZ, expression of dominant-negative TEAD or treatment with YAP-TEAD inhibitors, significantly inhibited EPAC1 expression. However, whereas expression of constitutively-active YAP completely reversed forskolin inhibition of EPAC1-promoter activity it did not rescue EPAC1 mRNA levels. Chromatin-immunoprecipitation detected a significant reduction in histone3-lysine27-acetylation at the EPAC1 proximal promoter in response to forskolin stimulation. HDAC1/3 inhibition partially reversed forskolin inhibition of EPAC1 expression, which was completely rescued by simultaneously expressing constitutively active YAP. Taken together, these data demonstrate that cAMP downregulates EPAC1 gene expression via disrupting the actin cytoskeleton, which inhibits YAP/TAZ-TEAD activity in concert with HDAC-mediated histone deacetylation at the EPAC1 proximal promoter. This represents a novel negative feedback mechanism controlling EPAC1 levels in response to cAMP elevation.
Collapse
Affiliation(s)
- Reza Ebrahimighaei
- School of Translational Health Sciences, Faculty of Health Sciences, University of Bristol, Research Floor Level 7, Bristol Royal Infirmary, Bristol BS2 8HW, UK
| | - Madeleine C McNeill
- School of Translational Health Sciences, Faculty of Health Sciences, University of Bristol, Research Floor Level 7, Bristol Royal Infirmary, Bristol BS2 8HW, UK
| | - Sarah A Smith
- School of Translational Health Sciences, Faculty of Health Sciences, University of Bristol, Research Floor Level 7, Bristol Royal Infirmary, Bristol BS2 8HW, UK
| | - Jason P Wray
- School of Translational Health Sciences, Faculty of Health Sciences, University of Bristol, Research Floor Level 7, Bristol Royal Infirmary, Bristol BS2 8HW, UK
| | - Kerrie L Ford
- School of Translational Health Sciences, Faculty of Health Sciences, University of Bristol, Research Floor Level 7, Bristol Royal Infirmary, Bristol BS2 8HW, UK
| | - Andrew C Newby
- School of Translational Health Sciences, Faculty of Health Sciences, University of Bristol, Research Floor Level 7, Bristol Royal Infirmary, Bristol BS2 8HW, UK
| | - Mark Bond
- School of Translational Health Sciences, Faculty of Health Sciences, University of Bristol, Research Floor Level 7, Bristol Royal Infirmary, Bristol BS2 8HW, UK.
| |
Collapse
|
205
|
Humeres C, Frangogiannis NG. Fibroblasts in the Infarcted, Remodeling, and Failing Heart. JACC Basic Transl Sci 2019; 4:449-467. [PMID: 31312768 PMCID: PMC6610002 DOI: 10.1016/j.jacbts.2019.02.006] [Citation(s) in RCA: 222] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 02/15/2019] [Accepted: 02/19/2019] [Indexed: 02/07/2023]
Abstract
Expansion and activation of fibroblasts following cardiac injury is important for repair but may also contribute to fibrosis, remodeling, and dysfunction. The authors discuss the dynamic alterations of fibroblasts in failing and remodeling myocardium. Emerging concepts suggest that fibroblasts are not unidimensional cells that act exclusively by secreting extracellular matrix proteins, thus promoting fibrosis and diastolic dysfunction. In addition to their involvement in extracellular matrix expansion, activated fibroblasts may also exert protective actions, preserving the cardiac extracellular matrix, transducing survival signals to cardiomyocytes, and regulating inflammation and angiogenesis. The functional diversity of cardiac fibroblasts may reflect their phenotypic heterogeneity.
Collapse
Key Words
- AT1, angiotensin type 1
- ECM, extracellular matrix
- FAK, focal adhesion kinase
- FGF, fibroblast growth factor
- IL, interleukin
- MAPK, mitogen-activated protein kinase
- MRTF, myocardin-related transcription factor
- PDGF, platelet-derived growth factor
- RNA, ribonucleic acid
- ROCK, Rho-associated coiled-coil containing kinase
- ROS, reactive oxygen species
- SMA, smooth muscle actin
- TGF, transforming growth factor
- TRP, transient receptor potential
- cytokines
- extracellular matrix
- fibroblast
- infarction
- lncRNA, long noncoding ribonucleic acid
- miRNA, micro–ribonucleic acid
- remodeling
Collapse
Affiliation(s)
- Claudio Humeres
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx, New York
| | - Nikolaos G Frangogiannis
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx, New York
| |
Collapse
|
206
|
Hara A, Kobayashi H, Asai N, Saito S, Higuchi T, Kato K, Okumura T, Bando YK, Takefuji M, Mizutani Y, Miyai Y, Saito S, Maruyama S, Maeda K, Ouchi N, Nagasaka A, Miyata T, Mii S, Kioka N, Worthley DL, Murohara T, Takahashi M, Enomoto A. Roles of the Mesenchymal Stromal/Stem Cell Marker Meflin in Cardiac Tissue Repair and the Development of Diastolic Dysfunction. Circ Res 2019; 125:414-430. [PMID: 31221024 DOI: 10.1161/circresaha.119.314806] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
RATIONALE Myofibroblasts have roles in tissue repair following damage associated with ischemia, aging, and inflammation and also promote fibrosis and tissue stiffening, causing organ dysfunction. One source of myofibroblasts is mesenchymal stromal/stem cells that exist as resident fibroblasts in multiple tissues. We previously identified meflin (mesenchymal stromal cell- and fibroblast-expressing Linx paralogue), a glycosylphosphatidylinositol-anchored membrane protein, as a specific marker of mesenchymal stromal/stem cells and a regulator of their undifferentiated state. The roles of meflin in the development of heart disease, however, have not been investigated. OBJECTIVE We examined the expression of meflin in the heart and its involvement in cardiac repair after ischemia, fibrosis, and the development of heart failure. METHODS AND RESULTS We found that meflin has an inhibitory role in myofibroblast differentiation of cultured mesenchymal stromal/stem cells. Meflin expression was downregulated by stimulation with TGF (transforming growth factor)-β, substrate stiffness, hypoxia, and aging. Histological analysis revealed that meflin-positive fibroblastic cells and their lineage cells proliferated in the hearts after acute myocardial infarction and pressure-overload heart failure mouse models. Analysis of meflin knockout mice revealed that meflin is essential for the increase in the number of cells that highly express type I collagen in the heart walls after myocardial infarction induction. When subjected to pressure overload by transverse aortic constriction, meflin knockout mice developed marked cardiac interstitial fibrosis with defective compensation mechanisms. Analysis with atomic force microscopy and hemodynamic catheterization revealed that meflin knockout mice developed stiff failing hearts with diastolic dysfunction. Mechanistically, we found that meflin interacts with bone morphogenetic protein 7, an antifibrotic cytokine that counteracts the action of TGF-β and augments its intracellular signaling. CONCLUSIONS These data suggested that meflin is involved in cardiac tissue repair after injury and has an inhibitory role in myofibroblast differentiation of cardiac fibroblastic cells and the development of cardiac fibrosis.
Collapse
Affiliation(s)
- Akitoshi Hara
- From the Department of Pathology (A.H., H.K., N.A., Y. Mizutani, Y. Miyai, S. Mii, M. Takahashi, A.E.), Nagoya University Graduate School of Medicine, Japan.,Department of Cardiology (A.H., K.K., T.O., Y.K.B., M. Takefuji, N.O., T. Murohara), Nagoya University Graduate School of Medicine, Japan
| | - Hiroki Kobayashi
- From the Department of Pathology (A.H., H.K., N.A., Y. Mizutani, Y. Miyai, S. Mii, M. Takahashi, A.E.), Nagoya University Graduate School of Medicine, Japan.,School of Medicine, University of Adelaide and South Australian Health and Medical Research Institute, Adelaide (H.K., D.L.W.)
| | - Naoya Asai
- From the Department of Pathology (A.H., H.K., N.A., Y. Mizutani, Y. Miyai, S. Mii, M. Takahashi, A.E.), Nagoya University Graduate School of Medicine, Japan.,Division of Molecular Pathology, Center for Neurological Disease and Cancer (N.A., M. Takahashi), Nagoya University Graduate School of Medicine, Japan
| | - Shigeyoshi Saito
- Department of Biomedical Imaging, National Cardiovascular and Cerebral Research Center, Osaka, Japan (Shigeyoshi Saito, T.H.)
| | - Takahiro Higuchi
- Department of Biomedical Imaging, National Cardiovascular and Cerebral Research Center, Osaka, Japan (Shigeyoshi Saito, T.H.)
| | - Katsuhiro Kato
- Department of Cardiology (A.H., K.K., T.O., Y.K.B., M. Takefuji, N.O., T. Murohara), Nagoya University Graduate School of Medicine, Japan
| | - Takahiro Okumura
- Department of Cardiology (A.H., K.K., T.O., Y.K.B., M. Takefuji, N.O., T. Murohara), Nagoya University Graduate School of Medicine, Japan
| | - Yasuko K Bando
- Department of Cardiology (A.H., K.K., T.O., Y.K.B., M. Takefuji, N.O., T. Murohara), Nagoya University Graduate School of Medicine, Japan
| | - Mikito Takefuji
- Department of Cardiology (A.H., K.K., T.O., Y.K.B., M. Takefuji, N.O., T. Murohara), Nagoya University Graduate School of Medicine, Japan
| | - Yasuyuki Mizutani
- From the Department of Pathology (A.H., H.K., N.A., Y. Mizutani, Y. Miyai, S. Mii, M. Takahashi, A.E.), Nagoya University Graduate School of Medicine, Japan.,Department of Gastroenterology and Hepatology (Y. Mizutani, K.M.), Nagoya University Graduate School of Medicine, Japan
| | - Yuki Miyai
- From the Department of Pathology (A.H., H.K., N.A., Y. Mizutani, Y. Miyai, S. Mii, M. Takahashi, A.E.), Nagoya University Graduate School of Medicine, Japan
| | - Shoji Saito
- Department of Nephrology (Shoji Saito, S. Maruyama), Nagoya University Graduate School of Medicine, Japan
| | - Shoichi Maruyama
- Department of Nephrology (Shoji Saito, S. Maruyama), Nagoya University Graduate School of Medicine, Japan
| | - Keiko Maeda
- Department of Gastroenterology and Hepatology (Y. Mizutani, K.M.), Nagoya University Graduate School of Medicine, Japan
| | - Noriyuki Ouchi
- Department of Cardiology (A.H., K.K., T.O., Y.K.B., M. Takefuji, N.O., T. Murohara), Nagoya University Graduate School of Medicine, Japan
| | - Arata Nagasaka
- Division of Anatomy, Department of Human Development and Fostering, Meikai University School of Dentistry, Saitama, Japan (A.N.)
| | - Takaki Miyata
- Department of Anatomy and Cell Biology (T. Miyata), Nagoya University Graduate School of Medicine, Japan
| | - Shinji Mii
- From the Department of Pathology (A.H., H.K., N.A., Y. Mizutani, Y. Miyai, S. Mii, M. Takahashi, A.E.), Nagoya University Graduate School of Medicine, Japan
| | - Noriyuki Kioka
- Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Japan (N.K.)
| | - Daniel L Worthley
- School of Medicine, University of Adelaide and South Australian Health and Medical Research Institute, Adelaide (H.K., D.L.W.)
| | - Toyoaki Murohara
- Department of Cardiology (A.H., K.K., T.O., Y.K.B., M. Takefuji, N.O., T. Murohara), Nagoya University Graduate School of Medicine, Japan
| | - Masahide Takahashi
- From the Department of Pathology (A.H., H.K., N.A., Y. Mizutani, Y. Miyai, S. Mii, M. Takahashi, A.E.), Nagoya University Graduate School of Medicine, Japan.,Division of Molecular Pathology, Center for Neurological Disease and Cancer (N.A., M. Takahashi), Nagoya University Graduate School of Medicine, Japan
| | - Atsushi Enomoto
- From the Department of Pathology (A.H., H.K., N.A., Y. Mizutani, Y. Miyai, S. Mii, M. Takahashi, A.E.), Nagoya University Graduate School of Medicine, Japan
| |
Collapse
|
207
|
Owen TJ, Harding SE. Multi-cellularity in cardiac tissue engineering, how close are we to native heart tissue? J Muscle Res Cell Motil 2019; 40:151-157. [PMID: 31222588 PMCID: PMC6726707 DOI: 10.1007/s10974-019-09528-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 06/15/2019] [Indexed: 12/25/2022]
Abstract
Tissue engineering is a complex field where the elements of biology and engineering are combined in an attempt to recapitulate the native environment of the body. Tissue engineering has shown one thing categorically; that the human body is extremely complex and it is truly a difficult task to generate this in the lab. There have been varied attempts at trying to generate a model for the heart with numerous cell types and different scaffolds or materials. The common underlying theme in these approaches is to combine together matrix material and different cell types to make something similar to heart tissue. Multi-cellularity is an essential aspect of the heart and therefore critical to any approach which would try to mimic such a complex tissue. The heart is made up of many cell types that combine to form complex structures like: deformable chambers, a tri-layered heart muscle, and vessels. Thus, in this review we will summarise how tissue engineering has progressed in modelling the heart and what gaps still exist in this dynamic field.
Collapse
Affiliation(s)
- Thomas J Owen
- National Heart and Lung Institute, Imperial College London Hammersmith Campus, Imperial Centre for Translational and Experimental Medicine, Du Cane Road, London, W12 0NN, UK.
| | - Sian E Harding
- National Heart and Lung Institute, Imperial College London Hammersmith Campus, Imperial Centre for Translational and Experimental Medicine, Du Cane Road, London, W12 0NN, UK
| |
Collapse
|
208
|
Broughton KM, Khieu T, Nguyen N, Rosa M, Mohsin S, Quijada P, Wang BJ, Echeagaray OH, Kubli DA, Kim T, Firouzi F, Monsanto MM, Gude NA, Adamson RM, Dembitsky WP, Davis ME, Sussman MA. Cardiac interstitial tetraploid cells can escape replicative senescence in rodents but not large mammals. Commun Biol 2019; 2:205. [PMID: 31231694 PMCID: PMC6565746 DOI: 10.1038/s42003-019-0453-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 05/02/2019] [Indexed: 12/26/2022] Open
Abstract
Cardiomyocyte ploidy has been described but remains obscure in cardiac interstitial cells. Ploidy of c-kit+ cardiac interstitial cells was assessed using confocal, karyotypic, and flow cytometric technique. Notable differences were found between rodent (rat, mouse) c-kit+ cardiac interstitial cells possessing mononuclear tetraploid (4n) content, compared to large mammals (human, swine) with mononuclear diploid (2n) content. In-situ analysis, confirmed with fresh isolates, revealed diploid content in human c-kit+ cardiac interstitial cells and a mixture of diploid and tetraploid content in mouse. Downregulation of the p53 signaling pathway provides evidence why rodent, but not human, c-kit+ cardiac interstitial cells escape replicative senescence. Single cell transcriptional profiling reveals distinctions between diploid versus tetraploid populations in mouse c-kit+ cardiac interstitial cells, alluding to functional divergences. Collectively, these data reveal notable species-specific biological differences in c-kit+ cardiac interstitial cells, which could account for challenges in extrapolation of myocardial from preclinical studies to clinical trials.
Collapse
Affiliation(s)
- Kathleen M. Broughton
- San Diego State University Heart Institute and the Integrated Regenerative Research Institute, 5500 Campanile Drive, San Diego, CA 92182 USA
| | - Tiffany Khieu
- San Diego State University Heart Institute and the Integrated Regenerative Research Institute, 5500 Campanile Drive, San Diego, CA 92182 USA
| | - Nicky Nguyen
- San Diego State University Heart Institute and the Integrated Regenerative Research Institute, 5500 Campanile Drive, San Diego, CA 92182 USA
| | - Michael Rosa
- San Diego State University Heart Institute and the Integrated Regenerative Research Institute, 5500 Campanile Drive, San Diego, CA 92182 USA
| | - Sadia Mohsin
- Cardiovascular Research Center, Temple University, 3500 N. Broad St., Philadelphia, 19140 PA USA
| | - Pearl Quijada
- San Diego State University Heart Institute and the Integrated Regenerative Research Institute, 5500 Campanile Drive, San Diego, CA 92182 USA
| | - Bingyan J. Wang
- San Diego State University Heart Institute and the Integrated Regenerative Research Institute, 5500 Campanile Drive, San Diego, CA 92182 USA
| | - Oscar H. Echeagaray
- San Diego State University Heart Institute and the Integrated Regenerative Research Institute, 5500 Campanile Drive, San Diego, CA 92182 USA
| | - Dieter A. Kubli
- San Diego State University Heart Institute and the Integrated Regenerative Research Institute, 5500 Campanile Drive, San Diego, CA 92182 USA
| | - Taeyong Kim
- San Diego State University Heart Institute and the Integrated Regenerative Research Institute, 5500 Campanile Drive, San Diego, CA 92182 USA
| | - Fareheh Firouzi
- San Diego State University Heart Institute and the Integrated Regenerative Research Institute, 5500 Campanile Drive, San Diego, CA 92182 USA
| | - Megan M. Monsanto
- San Diego State University Heart Institute and the Integrated Regenerative Research Institute, 5500 Campanile Drive, San Diego, CA 92182 USA
| | - Natalie A. Gude
- San Diego State University Heart Institute and the Integrated Regenerative Research Institute, 5500 Campanile Drive, San Diego, CA 92182 USA
| | - Robert M. Adamson
- Division of Cardiology, Sharp Memorial Hospital, 8010 Frost St., San Diego, 92123 CA USA
| | - Walter P. Dembitsky
- Division of Cardiology, Sharp Memorial Hospital, 8010 Frost St., San Diego, 92123 CA USA
| | - Michael E. Davis
- Biomedical Engineering and Medicine, Emory University, 1760 Haygood Dr., Atlanta, 30322 GA USA
| | - Mark A. Sussman
- San Diego State University Heart Institute and the Integrated Regenerative Research Institute, 5500 Campanile Drive, San Diego, CA 92182 USA
| |
Collapse
|
209
|
Ge W, Zhang W, Gao R, Li B, Zhu H, Wang J. IMM-H007 improves heart function via reducing cardiac fibrosis. Eur J Pharmacol 2019; 857:172442. [PMID: 31181209 DOI: 10.1016/j.ejphar.2019.172442] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 06/06/2019] [Accepted: 06/06/2019] [Indexed: 01/08/2023]
Abstract
Cardiac dysfunction is a pathological state characterized by damaged ability of the left ventricle (LV) to either eject or fill blood accompanied by cardiac hypertrophy and fibrosis. IMM-H007, an adenosine derivative, is an activator of AMP-Activated Protein Kinase (AMPK). AMPK can decrease the transforming growth factor-β1 (TGF-β1) expression during fibrosis. Therefore, we hypothesized that IMM-H007 contributed to cardiac dysfunction by mediating cardiac fibrosis. To test this hypothesis, we used angiotensin II (AngII)-induced cardiac remodeling model treated with IMM-H007 or vehicle. Echocardiography measurements showed that IMM-H007 significantly improved heart function indicated by increased LV ejection fraction (%LVEF) and LV fractional shortening (%LVFS). Histological staining and qRT-PCR analysis revealed that IMM-H007 markedly reduced AngII-induced cardiac fibroblast activation (α-smooth muscle actin and periostin) and matrix protein production (Collagen I and Collagen III). However, IMM-H007 did not affect AngII-induced cardiac hypertrophy. Immunoblotting analysis revealed that IMM-H007 activated AMPK, decreased the expression of TGF-β1, and inhibited the activation of Smad2 in heart tissues. In mouse primary cultured cardiac fibroblasts, pharmacological activation of AMPK by IMM-H007 significantly reduced AngII-induced TGF-β1 expression as well. Consistently, in human cardiac fibroblasts-adult ventricular (HCF-av), IMM-H007 activated AMPK and markedly suppressed AngII-induced TGF-β1 expression. These results together reveal that IMM-H007 improves heart function, and alleviates AngII-induced cardiac fibrosis by regulating AMPK-TGF-β1 signaling. These findings suggest IMM-H007 as a potential drug for treating cardiac dysfunction.
Collapse
Affiliation(s)
- Weipeng Ge
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Department of Pathophysiology, Peking Union Medical College, Beijing, China
| | - Wei Zhang
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Department of Pathophysiology, Peking Union Medical College, Beijing, China
| | - Ran Gao
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Department of Pathophysiology, Peking Union Medical College, Beijing, China
| | - Bolun Li
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Department of Pathophysiology, Peking Union Medical College, Beijing, China
| | - Haibo Zhu
- State Key Laboratory for Bioactive Substances and Functions of Natural Medicines, Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jing Wang
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Department of Pathophysiology, Peking Union Medical College, Beijing, China.
| |
Collapse
|
210
|
Andreadou I, Cabrera-Fuentes HA, Devaux Y, Frangogiannis NG, Frantz S, Guzik T, Liehn EA, Gomes CPC, Schulz R, Hausenloy DJ. Immune cells as targets for cardioprotection: new players and novel therapeutic opportunities. Cardiovasc Res 2019; 115:1117-1130. [PMID: 30825305 PMCID: PMC6529904 DOI: 10.1093/cvr/cvz050] [Citation(s) in RCA: 128] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 11/18/2018] [Accepted: 02/24/2019] [Indexed: 12/22/2022] Open
Abstract
New therapies are required to reduce myocardial infarct (MI) size and prevent the onset of heart failure in patients presenting with acute myocardial infarction (AMI), one of the leading causes of death and disability globally. In this regard, the immune cell response to AMI, which comprises an initial pro-inflammatory reaction followed by an anti-inflammatory phase, contributes to final MI size and post-AMI remodelling [changes in left ventricular (LV) size and function]. The transition between these two phases is critical in this regard, with a persistent and severe pro-inflammatory reaction leading to adverse LV remodelling and increased propensity for developing heart failure. In this review article, we provide an overview of the immune cells involved in orchestrating the complex and dynamic inflammatory response to AMI-these include neutrophils, monocytes/macrophages, and emerging players such as dendritic cells, lymphocytes, pericardial lymphoid cells, endothelial cells, and cardiac fibroblasts. We discuss potential reasons for past failures of anti-inflammatory cardioprotective therapies, and highlight new treatment targets for modulating the immune cell response to AMI, as a potential therapeutic strategy to improve clinical outcomes in AMI patients. This article is part of a Cardiovascular Research Spotlight Issue entitled 'Cardioprotection Beyond the Cardiomyocyte', and emerged as part of the discussions of the European Union (EU)-CARDIOPROTECTION Cooperation in Science and Technology (COST) Action, CA16225.
Collapse
Affiliation(s)
- Ioanna Andreadou
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Panepistimiopolis, Zografou, Athens, Greece
| | - Hector A Cabrera-Fuentes
- Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, 8 College Road, Singapore
- National Heart Research Institute Singapore, National Heart Centre Singapore, 5 Hospital Drive, Singapore
- Institute of Biochemistry, Medical School, Justus-Liebig University, Ludwigstrasse 23, Giessen, Germany
- Tecnologico de Monterrey, Centro de Biotecnologia-FEMSA, Av. Eugenio Garza Sada 2501 Sur, Nuevo Leon, Mexico
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, Kazan, Kremlyovskaya St, 18, Kazan, Respublika Tatarstan, Russia
| | - Yvan Devaux
- Cardiovascular Research Unit, Department of Population Health, Luxembourg Institute of Health, 1A-B rue Thomas Edison, Strassen, Luxembourg
| | - Nikolaos G Frangogiannis
- Wilf Family Cardiovascular Research Institute Department of Medicine (Cardiology) Albert Einstein College of Medicine, 1300 Morris Park Avenue, Forchheimer G46B Bronx NY USA
| | - Stefan Frantz
- Department of Internal Medicine I, University Hospital Würzburg, Oberdürrbacher Str. 6, Würzburg, Germany
| | - Tomasz Guzik
- Department of Internal and Agricultural Medicine, Jagiellonian University Medical College, Świętej Anny 12, Kraków, Poland
- Institute of Cardiovascular and Medical Sciences, University ofGlasgow, University Avenue, Glasgow, UK
| | - Elisa A Liehn
- Institute for Molecular Cardiovascular Research, Rheinisch Westfälische Technische Hochschule Aachen University,Templergraben 55, Aachen, Germany
- Human Genomics Laboratory, University of Medicine and Pharmacy Craiova, Strada Petru Rareș 2, Craiova, Romania
- Department of Cardiology, Pulmonology, Angiology and Intensive Care, University Hospital, Rheinisch Westfälische Technische Hochschule,Templergraben 55, Aachen, Germany
| | - Clarissa P C Gomes
- Cardiovascular Research Unit, Department of Population Health, Luxembourg Institute of Health, 1A-B rue Thomas Edison, Strassen, Luxembourg
| | - Rainer Schulz
- Physiologisches Institut Fachbereich Medizin der Justus-Liebig-Universität, Aulweg 129, Giessen, Germany
| | - Derek J Hausenloy
- Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, 8 College Road, Singapore
- National Heart Research Institute Singapore, National Heart Centre Singapore, 5 Hospital Drive, Singapore
- Tecnologico de Monterrey, Centro de Biotecnologia-FEMSA, Av. Eugenio Garza Sada 2501 Sur, Nuevo Leon, Mexico
- Yong Loo Lin School of Medicine, National University Singapore, 1E Kent Ridge Road, Singapore
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, London, UK
- The National Institute of Health Research University College London Hospitals Biomedical Research Centre, Research & Development, Maple House 1st floor, 149 Tottenham Court Road, London, UK
| |
Collapse
|
211
|
Affiliation(s)
- Mark A Sussman
- Department of Biology & Integrated Regenerative Research Institute, San Diego State University, San Diego, CA 92182, USA
| |
Collapse
|
212
|
Perbellini F, Watson SA, Scigliano M, Alayoubi S, Tkach S, Bardi I, Quaife N, Kane C, Dufton NP, Simon A, Sikkel MB, Faggian G, Randi AM, Gorelik J, Harding SE, Terracciano CM. Investigation of cardiac fibroblasts using myocardial slices. Cardiovasc Res 2019; 114:77-89. [PMID: 29016704 PMCID: PMC5852538 DOI: 10.1093/cvr/cvx152] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 08/17/2017] [Indexed: 12/15/2022] Open
Abstract
Aims Cardiac fibroblasts (CFs) are considered the principal regulators of cardiac fibrosis. Factors that influence CF activity are difficult to determine. When isolated and cultured in vitro, CFs undergo rapid phenotypic changes including increased expression of α-SMA. Here we describe a new model to study CFs and their response to pharmacological and mechanical stimuli using in vitro cultured mouse, dog and human myocardial slices. Methods and results Unloading of myocardial slices induced CF proliferation without α-SMA expression up to 7 days in culture. CFs migrating onto the culture plastic support or cultured on glass expressed αSMA within 3 days. The cells on the slice remained αSMA(−) despite transforming growth factor-β (20 ng/ml) or angiotensin II (200 µM) stimulation. When diastolic load was applied to myocardial slices using A-shaped stretchers, CF proliferation was significantly prevented at Days 3 and 7 (P < 0.001). Conclusions Myocardial slices allow the study of CFs in a multicellular environment and may be used to effectively study mechanisms of cardiac fibrosis and potential targets.
Collapse
Affiliation(s)
- Filippo Perbellini
- Imperial Centre for Translational and Experimental Medicine, National Heart and Lung Institute, Imperial College London, Hammersmith Campus, Du Cane Road, London W12?0NN, UK
| | - Samuel A Watson
- Imperial Centre for Translational and Experimental Medicine, National Heart and Lung Institute, Imperial College London, Hammersmith Campus, Du Cane Road, London W12?0NN, UK
| | | | - Samha Alayoubi
- Imperial Centre for Translational and Experimental Medicine, National Heart and Lung Institute, Imperial College London, Hammersmith Campus, Du Cane Road, London W12?0NN, UK
| | - Sebastian Tkach
- Imperial Centre for Translational and Experimental Medicine, National Heart and Lung Institute, Imperial College London, Hammersmith Campus, Du Cane Road, London W12?0NN, UK
| | - Ifigeneia Bardi
- Imperial Centre for Translational and Experimental Medicine, National Heart and Lung Institute, Imperial College London, Hammersmith Campus, Du Cane Road, London W12?0NN, UK
| | - Nicholas Quaife
- Imperial Centre for Translational and Experimental Medicine, National Heart and Lung Institute, Imperial College London, Hammersmith Campus, Du Cane Road, London W12?0NN, UK
| | - Christopher Kane
- Imperial Centre for Translational and Experimental Medicine, National Heart and Lung Institute, Imperial College London, Hammersmith Campus, Du Cane Road, London W12?0NN, UK
| | - Neil P Dufton
- Imperial Centre for Translational and Experimental Medicine, National Heart and Lung Institute, Imperial College London, Hammersmith Campus, Du Cane Road, London W12?0NN, UK
| | - André Simon
- Department of Cardiothoracic Transplantation and Mechanical Circulatory Support, Royal Brompton and Harefield NHS Foundation Trust, Harefield, UK
| | - Markus B Sikkel
- Imperial Centre for Translational and Experimental Medicine, National Heart and Lung Institute, Imperial College London, Hammersmith Campus, Du Cane Road, London W12?0NN, UK
| | - Giuseppe Faggian
- Department of Cardiac Surgery, University of Verona, Verona, Italy
| | - Anna M Randi
- Imperial Centre for Translational and Experimental Medicine, National Heart and Lung Institute, Imperial College London, Hammersmith Campus, Du Cane Road, London W12?0NN, UK
| | - Julia Gorelik
- Imperial Centre for Translational and Experimental Medicine, National Heart and Lung Institute, Imperial College London, Hammersmith Campus, Du Cane Road, London W12?0NN, UK
| | - Sian E Harding
- Imperial Centre for Translational and Experimental Medicine, National Heart and Lung Institute, Imperial College London, Hammersmith Campus, Du Cane Road, London W12?0NN, UK
| | - Cesare M Terracciano
- Imperial Centre for Translational and Experimental Medicine, National Heart and Lung Institute, Imperial College London, Hammersmith Campus, Du Cane Road, London W12?0NN, UK
| |
Collapse
|
213
|
Huang S, Chen B, Su Y, Alex L, Humeres C, Shinde AV, Conway SJ, Frangogiannis NG. Distinct roles of myofibroblast-specific Smad2 and Smad3 signaling in repair and remodeling of the infarcted heart. J Mol Cell Cardiol 2019; 132:84-97. [PMID: 31085202 DOI: 10.1016/j.yjmcc.2019.05.006] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 04/26/2019] [Accepted: 05/09/2019] [Indexed: 12/21/2022]
Abstract
TGF-βs regulate fibroblast responses, by activating Smad2 or Smad3 signaling, or via Smad-independent pathways. We have previously demonstrated that myofibroblast-specific Smad3 is critically implicated in repair of the infarcted heart. However, the role of fibroblast Smad2 in myocardial infarction remains unknown. This study investigates the role of myofibroblast-specific Smad2 signaling in myocardial infarction, and explores the mechanisms responsible for the distinct effects of Smad2 and Smad3. In a mouse model of non-reperfused myocardial infarction, Smad2 activation in infarct myofibroblasts peaked 7 days after coronary occlusion. In vitro, TGF-β1, -β2 and -β3, but not angiotensin 2 and bone morphogenetic proteins-2, -4 and -7, activated fibroblast Smad2. Myofibroblast-specific Smad2 and Smad3 knockout mice (FS2KO, FS3KO) and corresponding control littermates underwent non-reperfused infarction. In contrast to the increase in rupture rates and adverse remodeling in FS3KO mice, FS2KO animals had mortality comparable to Smad2 fl/fl controls, and exhibited a modest but transient improvement in dysfunction after 7 days of coronary occlusion. At the 28 day timepoint, FS2KO and Smad2 fl/fl mice had comparable adverse remodeling. Although both FS3KO and FS2KO animals had increased myofibroblast density in the infarct, only FS3KO mice exhibited impaired scar organization, associated with perturbed alignment of infarct myofibroblasts. In vitro, Smad3 but not Smad2 knockdown downmodulated fibroblast α2 and α5 integrin expression. Moreover, Smad3 knockdown reduced expression of the GTPase RhoA, whereas Smad2 knockdown markedly increased fibroblast RhoA levels. Smad3-dependent integrin expression may be important for fibroblast activation, whereas RhoA may transduce planar cell polarity pathway signals, essential for fibroblast alignment. Myofibroblast-specific Smad3, but not Smad2 is required for formation of aligned myofibroblast arrays in the infarct. The distinct in vivo effects of myofibroblast Smad2 and Smad3 may involve Smad3-dependent integrin synthesis, and contrasting effects of Smad2 and Smad3 on RhoA expression.
Collapse
Affiliation(s)
- Shuaibo Huang
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx, NY, United States of America
| | - Bijun Chen
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx, NY, United States of America
| | - Ya Su
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx, NY, United States of America
| | - Linda Alex
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx, NY, United States of America
| | - Claudio Humeres
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx, NY, United States of America
| | - Arti V Shinde
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx, NY, United States of America
| | - Simon J Conway
- Department of Pediatrics, Indiana University, Indianapolis, IN, United States of America
| | - Nikolaos G Frangogiannis
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx, NY, United States of America.
| |
Collapse
|
214
|
Lin CF, Su CJ, Liu JH, Chen ST, Huang HL, Pan SL. Potential Effects of CXCL9 and CCL20 on Cardiac Fibrosis in Patients with Myocardial Infarction and Isoproterenol-Treated Rats. J Clin Med 2019; 8:jcm8050659. [PMID: 31083544 PMCID: PMC6572441 DOI: 10.3390/jcm8050659] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 05/02/2019] [Accepted: 05/09/2019] [Indexed: 02/07/2023] Open
Abstract
The chemokines CXCL9 and CCL20 have been reported to be associated with ventricular dysfunction. This study was aimed to investigate the effects of CXCL9/CCL20 on cardiac fibrosis following myocardial infarction (MI). Blood samples of patients with MI were obtained to determine the serum CXCL9, CCL20, tumor necrosis factor-α (TNF-α), and transforming growth factor-β (TGF-β). The expression of CXCL9 and CCL20 in hypoxia-incubated H9c2 cells and TNF-α/TGF-β-activated peripheral blood mononuclear cells (PBMCs) were examined. The experimental MI of rats was produced by the intraperitoneal injection of isoproterenol (ISO) (85 mg/kg/day) for two consecutive days. The growth and migration of CXCL9/CCL20-incubated cardiac fibroblasts in vitro were evaluated. TNF-α/TGF-β-activated PBMCs showed an enhanced expression of CXCL9 and CCL20, while hypoxic H9c2 cells did not. Patients with MI had significantly enhanced levels of serum TGF-β and CXCL9 compared to healthy subjects. ISO-treated rats had increased serum CXCL9 levels and marked cardiac fibrosis compared to control rats. The trend of increased serum CCL20 in patients with MI and ISO-treated rats was not significant. CXCL9-incubated cardiac fibroblasts showed enhanced proliferation and migration. The findings of this study suggest that an enhanced expression of CXCL9 following MI might play a role in post-MI cardiac fibrosis by activating cardiac fibroblasts.
Collapse
Affiliation(s)
- Chao-Feng Lin
- Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei 110, Taiwan.
- Department of Medicine, MacKay Medical College, New Taipei City 252, Taiwan.
- Division of Cardiology, Department of Internal Medicine, MacKay Memorial Hospital, Taipei 104, Taiwan.
| | - Chih-Jou Su
- Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei 110, Taiwan.
| | - Jia-Hong Liu
- Graduate Institute of Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan.
| | - Shui-Tien Chen
- Institute of Biological Chemistry, Academia Sinica, Taipei 115, Taiwan.
| | - Han-Li Huang
- TMU Biomedical Commercialization Center, Taipei Medical University, Taipei 110, Taiwan.
| | - Shiow-Lin Pan
- Graduate Institute of Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan.
- TMU Biomedical Commercialization Center, Taipei Medical University, Taipei 110, Taiwan.
- Ph.D. Program in Biotechnology Research and Development, College of Pharmacy, Taipei Medical University, Taipei 110, Taiwan.
| |
Collapse
|
215
|
|
216
|
Chou PC, Tsai YC, Chen SJ, Tsai LK, Chien CL. Intracerebral transplantation of erythropoietin-producing fibroblasts facilitates neurogenesis and functional recovery in an ischemic stroke model. Brain Behav 2019; 9:e01274. [PMID: 30920178 PMCID: PMC6520520 DOI: 10.1002/brb3.1274] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2018] [Revised: 02/28/2019] [Accepted: 03/06/2019] [Indexed: 12/18/2022] Open
Abstract
INTRODUCTION Erythropoietin (EPO) can enhance neurogenesis and fibroblasts can secrete growth factors; together, they may benefit ischemic stroke. We transplanted EPO-producing fibroblasts into the rodent infarcted brain to test their effect on neurogenesis and functional recovery. METHODS A total of 106 cells of EPO-producing NIH/3T3 fibroblasts (EPO/EGFP/3T3) or enhanced green fluorescence protein (EGFP)-expressing fibroblasts (EGFP/3T3) were stereotaxically injected into the infarcted striatum of adult rats that received transient middle cerebral artery occlusion (MCAO) surgery 1 day poststroke. On day 14 after MCAO, the animals were euthanized for the evaluation of neurogenesis via immunohistochemistry and of the expression of growth factors using enzyme-linked immunosorbent assay. The infarct volume was analyzed using magnetic resonance imaging and the neurological behavior was assessed using the neurological severity scoring performed within 14 days after MCAO. RESULTS The MCAO rats with EPO/EGFP/3T3 treatment showed high EPO expression in the infarcted brain for at least 1 week. The concentration of brain-derived neurotrophic factor was higher in both hemispheres of MCAO rats with either EGFP/3T3 or EPO/EGFP/3T3 treatment at 14 days poststroke compared with untreated MCAO rats. The number of Ki-67-, nestin-, or doublecortin-immunoreactive cells in bilateral subventricular zones was higher in EPO/EGFP/3T3-treated MCAO rats than it was in untreated MCAO control animals, indicating the enhancement of neurogenesis after EPO/EGFP/3T3 treatment. Notably, post-MCAO EPO/EGFP/3T3 treatment significantly reduced infarct size and improved functional recovery. CONCLUSION The intracerebral transplantation of EPO-producing fibroblasts benefited an ischemic stroke model probably via the enhancement of neurogenesis.
Collapse
Affiliation(s)
- Pin-Chun Chou
- Graduate Institute of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yi-Chieh Tsai
- Department of Neurology and Stroke Center, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Shiu-Jau Chen
- Department of Medicine, Mackay Medical College, New Taipei City, Taiwan
| | - Li-Kai Tsai
- Department of Neurology and Stroke Center, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Chung-Liang Chien
- Graduate Institute of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
217
|
Pattar SS, Fatehi Hassanabad A, Fedak PWM. Acellular Extracellular Matrix Bioscaffolds for Cardiac Repair and Regeneration. Front Cell Dev Biol 2019; 7:63. [PMID: 31080800 PMCID: PMC6497812 DOI: 10.3389/fcell.2019.00063] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 04/08/2019] [Indexed: 12/19/2022] Open
Abstract
Heart failure is a progressive deterioration of cardiac pump function over time and is often a manifestation of ischemic injury caused by myocardial infarction (MI). Post-MI, structural remodeling of the infarcted myocardium ensues. Dysregulation of extracellular matrix (ECM) homeostasis is a hallmark of structural cardiac remodeling and is largely driven by cardiac fibroblast activation. While initially adaptive, structural cardiac remodeling leads to irreversible heart failure due to the progressive loss of cardiac function. Loss of pump function is associated with myocardial fibrosis, wall thinning, and left ventricular (LV) dilatation. Surgical revascularization of the damaged myocardium via coronary artery bypass graft (CABG) surgery and/or percutaneous coronary intervention (PCI) can enhance myocardial perfusion and is beneficial. However, these interventions alone are unable to prevent progressive fibrotic remodeling and loss of heart function that leads to clinical end-stage heart failure. Acellular biologic ECM scaffolds can be surgically implanted onto injured myocardial regions during open-heart surgery as an adjunct therapy to surgical revascularization. This presents a novel therapeutic approach to alter maladaptive remodeling and promote functional recovery. Acellular ECM bioscaffolds have been shown to provide passive structural support to the damaged myocardium and also to act as a dynamic bioactive reservoir capable of promoting endogenous mechanisms of tissue repair, such as vasculogenesis. The composition and structure of xenogenic acellular ECM bioscaffolds are determined by the physiological requirements of the tissue from which they are derived. The capacity of different tissue-derived acellular bioscaffolds to attenuate cardiac remodeling and restore ECM homeostasis after injury may depend on such properties. Accordingly, the search and discovery of an optimal ECM bioscaffold for use in cardiac repair is warranted and may be facilitated by comparing bioscaffolds. This review will provide a summary of the acellular ECM bioscaffolds currently available for use in cardiac surgery with a focus on how they attenuate cardiac remodeling by providing the necessary environmental cues to promote endogenous mechanisms of tissue repair.
Collapse
Affiliation(s)
- Simranjit S Pattar
- Section of Cardiac Surgery, Department of Cardiac Science, Cumming School of Medicine, Libin Cardiovascular Institute of Alberta, University of Calgary, Calgary, AB, Canada
| | - Ali Fatehi Hassanabad
- Section of Cardiac Surgery, Department of Cardiac Science, Cumming School of Medicine, Libin Cardiovascular Institute of Alberta, University of Calgary, Calgary, AB, Canada
| | - Paul W M Fedak
- Section of Cardiac Surgery, Department of Cardiac Science, Cumming School of Medicine, Libin Cardiovascular Institute of Alberta, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
218
|
Shi Y, Yang Y, Guo Q, Gao Q, Ding Y, Wang H, Xu W, Yu B, Wang M, Zhao Y, Zhu W. Exosomes Derived from Human Umbilical Cord Mesenchymal Stem Cells Promote Fibroblast-to-Myofibroblast Differentiation in Inflammatory Environments and Benefit Cardioprotective Effects. Stem Cells Dev 2019; 28:799-811. [PMID: 30896296 DOI: 10.1089/scd.2018.0242] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Cardioprotective effects of exosomes derived from human umbilical cord mesenchymal stem cells (hucMSC-exosomes) postmyocardial infarction (post-MI) have been reported in our previous study. It is known that fibroblasts are pro-inflammatory phenotypes, while myofibroblasts are anti-inflammatory phenotypes. This study aimed to investigate whether hucMSC-exosomes promoted cardiac fibroblast-to-myofibroblast differentiation in inflammatory environments and protected cardiomyocytes. Rats were performed by permanent ligation of the left anterior descending coronary artery and underwent intramyocardial injection of hucMSC-exosomes or phosphate-buffered saline (PBS) in surgery. Fibroblasts were stimulated by lipopolysaccharide (LPS) to create inflammatory environments in vitro. Western blot and immunohistochemical and immunofluorescence staining for α-smooth muscle actin were used to demonstrate fibroblast-to-myofibroblast differentiation. Transwell migration assay and CCK-8 assay were used to evaluate migration and proliferation of fibroblasts. Reverse transcription-polymerase chain reaction, western blot, and immunohistochemical staining were used to detect expressions of inflammatory factors. To investigate cardioprotective effects, cardiomyocytes were treated with supernatant derived from fibroblasts pretreated with LPS or LPS plus hucMSC-exosomes in hypoxic environments. Cardiomyocyte apoptosis was determined using TUNEL assay and western blot. Results indicated that hucMSC-exosomes increased the density of myofibroblasts in infarct areas during inflammatory phases post-MI, promoted fibroblast-to-myofibroblast differentiation in inflammatory environments, and attenuated inflammatory responses in vitro and in vivo. Culture medium derived from fibroblasts pretreated with LPS plus hucMSC-exosomes reduced cardiomyocyte apoptosis. In vivo, apoptotic cells in acute myocardial infarction (AMI)+exosomes groups were also less than AMI+PBS groups. In conclusion, hucMSC-exosomes can promote fibroblast-to-myofibroblast differentiation in inflammatory environments, then protecting cardiomyocytes.
Collapse
Affiliation(s)
- Yu Shi
- 1 School of Medicine, Jiangsu University, Zhenjiang, China
| | - Yuqi Yang
- 2 Changzhou Maternity and Child Health Care Hospital Affiliated to Nanjing Medical University, Changzhou, China
| | - Qinyu Guo
- 1 School of Medicine, Jiangsu University, Zhenjiang, China
| | - Qiuzhi Gao
- 1 School of Medicine, Jiangsu University, Zhenjiang, China
| | - Ying Ding
- 1 School of Medicine, Jiangsu University, Zhenjiang, China
| | - Hua Wang
- 3 The Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Wenrong Xu
- 1 School of Medicine, Jiangsu University, Zhenjiang, China
| | - Bin Yu
- 2 Changzhou Maternity and Child Health Care Hospital Affiliated to Nanjing Medical University, Changzhou, China
| | - Mei Wang
- 1 School of Medicine, Jiangsu University, Zhenjiang, China
| | - Yuanyuan Zhao
- 1 School of Medicine, Jiangsu University, Zhenjiang, China
| | - Wei Zhu
- 1 School of Medicine, Jiangsu University, Zhenjiang, China
| |
Collapse
|
219
|
Inhibition of Mitofusin-2 Promotes Cardiac Fibroblast Activation via the PERK/ATF4 Pathway and Reactive Oxygen Species. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:3649808. [PMID: 31178957 PMCID: PMC6501253 DOI: 10.1155/2019/3649808] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 12/06/2018] [Indexed: 02/05/2023]
Abstract
Mitofusin-2 (Mfn2) is a key outer mitochondrial membrane protein, which maintains normal mitochondrial dynamics and function. However, its role in cardiac fibroblast activation remains poorly understood. In the present study, a rat model of transverse aortic constriction (TAC) was established to observe the cardiac fibroblast activation in vivo. TGF-β1 treatment for 24 hours was used to induce cardiac fibroblast activation in vitro. As a result, the expression of Mfn2 decreased in the hypertrophic heart tissues and cardiac fibroblasts treated with TGF-β1. siMfn2 and adenovirus were applied to mediate Mfn2 gene silencing and overexpression in cardiac fibroblasts to elucidate the relationship between Mfn2 and cardiac fibroblast activation, as well as the possible underlying mechanisms. Knockdown of Mfn2 further promoted TGF-β1-induced cardiac fibroblast activation, while forced expression of Mfn2 attenuated this pathological reaction. The PERK/ATF4 pathway, one of the branches of endoplasmic reticulum (ER) stress, was identified to be involved in this process. Knockdown and overexpression of Mfn2 lead to aggravation or alleviation of the PERK/ATF4 pathway. Blocking this pathway by silencing ATF4 with siATF4 attenuated the pathological process. During the activation of cardiac fibroblasts, knockdown of Mfn2 also increased the production of reactive oxygen species (ROS), while ROS scavenger N-acetyl-l-cysteine (NAC) could attenuate the effect caused by knockdown of Mfn2. Our data suggested that inhibition of Mfn2 could promote cardiac fibroblast activation by activating the PERK/ATF4 signaling pathway and increasing the generation of ROS.
Collapse
|
220
|
Psarras S, Beis D, Nikouli S, Tsikitis M, Capetanaki Y. Three in a Box: Understanding Cardiomyocyte, Fibroblast, and Innate Immune Cell Interactions to Orchestrate Cardiac Repair Processes. Front Cardiovasc Med 2019; 6:32. [PMID: 31001541 PMCID: PMC6454035 DOI: 10.3389/fcvm.2019.00032] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 03/11/2019] [Indexed: 12/11/2022] Open
Abstract
Following an insult by both intrinsic and extrinsic pathways, complex cellular, and molecular interactions determine a successful recovery or inadequate repair of damaged tissue. The efficiency of this process is particularly important in the heart, an organ characterized by very limited regenerative and repair capacity in higher adult vertebrates. Cardiac insult is characteristically associated with fibrosis and heart failure, as a result of cardiomyocyte death, myocardial degeneration, and adverse remodeling. Recent evidence implies that resident non-cardiomyocytes, fibroblasts but also macrophages -pillars of the innate immunity- form part of the inflammatory response and decisively affect the repair process following a cardiac insult. Multiple studies in model organisms (mouse, zebrafish) of various developmental stages (adult and neonatal) combined with genetically engineered cell plasticity and differentiation intervention protocols -mainly targeting cardiac fibroblasts or progenitor cells-reveal particular roles of resident and recruited innate immune cells and their secretome in the coordination of cardiac repair. The interplay of innate immune cells with cardiac fibroblasts and cardiomyocytes is emerging as a crucial platform to help our understanding and, importantly, to allow the development of effective interventions sufficient to minimize cardiac damage and dysfunction after injury.
Collapse
Affiliation(s)
- Stelios Psarras
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Dimitris Beis
- Center of Clinical, Experimental Surgery & Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Sofia Nikouli
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Mary Tsikitis
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Yassemi Capetanaki
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| |
Collapse
|
221
|
Lai SL, Marín-Juez R, Stainier DYR. Immune responses in cardiac repair and regeneration: a comparative point of view. Cell Mol Life Sci 2019; 76:1365-1380. [PMID: 30578442 PMCID: PMC6420886 DOI: 10.1007/s00018-018-2995-5] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 11/26/2018] [Accepted: 12/10/2018] [Indexed: 12/13/2022]
Abstract
Immediately after cardiac injury, the immune system plays major roles in repair and regeneration as it becomes involved in a number of processes including damage-associated signaling, inflammation, revascularization, cardiomyocyte dedifferentiation and replenishment, and fibrotic scar formation/resolution. Recent studies have revealed that different immune responses occur in the various experimental models capable or incapable of cardiac regeneration, and that harnessing these immune responses might improve cardiac repair. In light of this concept, this review analyzes current knowledge about the immune responses to cardiac injury from a comparative perspective. Insights gained from such comparative analyses may provide ways to modulate the immune response as a potential therapeutic strategy for cardiac disease.
Collapse
Affiliation(s)
- Shih-Lei Lai
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan.
| | - Rubén Marín-Juez
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Didier Y R Stainier
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.
| |
Collapse
|
222
|
Farbehi N, Patrick R, Dorison A, Xaymardan M, Janbandhu V, Wystub-Lis K, Ho JW, Nordon RE, Harvey RP. Single-cell expression profiling reveals dynamic flux of cardiac stromal, vascular and immune cells in health and injury. eLife 2019; 8:43882. [PMID: 30912746 PMCID: PMC6459677 DOI: 10.7554/elife.43882] [Citation(s) in RCA: 358] [Impact Index Per Article: 59.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 03/25/2019] [Indexed: 12/11/2022] Open
Abstract
Besides cardiomyocytes (CM), the heart contains numerous interstitial cell types which play key roles in heart repair, regeneration and disease, including fibroblast, vascular and immune cells. However, a comprehensive understanding of this interactive cell community is lacking. We performed single-cell RNA-sequencing of the total non-CM fraction and enriched (Pdgfra-GFP+) fibroblast lineage cells from murine hearts at days 3 and 7 post-sham or myocardial infarction (MI) surgery. Clustering of >30,000 single cells identified >30 populations representing nine cell lineages, including a previously undescribed fibroblast lineage trajectory present in both sham and MI hearts leading to a uniquely activated cell state defined in part by a strong anti-WNT transcriptome signature. We also uncovered novel myofibroblast subtypes expressing either pro-fibrotic or anti-fibrotic signatures. Our data highlight non-linear dynamics in myeloid and fibroblast lineages after cardiac injury, and provide an entry point for deeper analysis of cardiac homeostasis, inflammation, fibrosis, repair and regeneration. In our bodies, heart attacks lead to cell death and inflammation. This is then followed by a healing phase where the organ repairs itself. There are many types of heart cells, from muscle and pacemaker cells that help to create the beating motion, to so-called fibroblasts that act as a supporting network. Yet, it is still unclear how individual cells participate in the heart's response to injury. All cells possess the same genetic information, but they turn on or off different genes depending on the specific tasks that they need to perform. Spotting which genes are activated in individual cells can therefore provide clues about their exact roles in the body. Until recently, technological limitations meant that this information was difficult to access, because it was only possible to capture the global response of a group of cells in a sample. A new method called single-cell RNA sequencing is now allowing researchers to study the activities of many genes in thousands of individual cells at the same time. Here, Farbehi, Patrick et al. performed single-cell RNA sequencing on over 30,000 individual cells from healthy and injured mouse hearts. Computational approaches were then used to cluster cells into groups according to the activities of their genes. The experiments identified over 30 distinct sub-types of cell, including several that were previously unknown. For example, a group of fibroblasts that express a gene called Wif1 was discovered. Previous genetic studies have shown that Wif1 is essential for the heart's response to injury. Further experiments by Farbehi, Patrick et al. indicated that this new sub-type of cells may control the timing of the different aspects of heart repair after damage. Tens of millions of people around the world suffer from heart attacks and other heart diseases. Knowing how different types of heart cells participate in repair mechanisms may help to find new targets for drugs and other treatments.
Collapse
Affiliation(s)
- Nona Farbehi
- Victor Chang Cardiac Research Institute, Darlinghurst, Australia.,Stem Cells Australia, Melbourne Brain Centre, University of Melbourne, Victoria, Australia.,Garvan Weizmann Centre for Cellular Genomics, Garvan Institute of Medical Research, Sydney, Australia.,Graduate School of Biomedical Engineering, UNSW Sydney, Kensington, Australia
| | - Ralph Patrick
- Victor Chang Cardiac Research Institute, Darlinghurst, Australia.,Stem Cells Australia, Melbourne Brain Centre, University of Melbourne, Victoria, Australia.,St. Vincent's Clinical School, UNSW Sydney, Kensington, Australia
| | - Aude Dorison
- Victor Chang Cardiac Research Institute, Darlinghurst, Australia.,Stem Cells Australia, Melbourne Brain Centre, University of Melbourne, Victoria, Australia
| | - Munira Xaymardan
- Victor Chang Cardiac Research Institute, Darlinghurst, Australia.,Stem Cells Australia, Melbourne Brain Centre, University of Melbourne, Victoria, Australia.,School of Dentistry, Faculty of Medicine and Health, University of Sydney, Westmead Hospital, Westmead, Australia
| | - Vaibhao Janbandhu
- Victor Chang Cardiac Research Institute, Darlinghurst, Australia.,Stem Cells Australia, Melbourne Brain Centre, University of Melbourne, Victoria, Australia.,St. Vincent's Clinical School, UNSW Sydney, Kensington, Australia
| | | | - Joshua Wk Ho
- Victor Chang Cardiac Research Institute, Darlinghurst, Australia.,St. Vincent's Clinical School, UNSW Sydney, Kensington, Australia
| | - Robert E Nordon
- Stem Cells Australia, Melbourne Brain Centre, University of Melbourne, Victoria, Australia.,Graduate School of Biomedical Engineering, UNSW Sydney, Kensington, Australia
| | - Richard P Harvey
- Victor Chang Cardiac Research Institute, Darlinghurst, Australia.,Stem Cells Australia, Melbourne Brain Centre, University of Melbourne, Victoria, Australia.,School of Biotechnology and Biomolecular Science, UNSW Sydney, Kensington, Australia
| |
Collapse
|
223
|
Hinz B, McCulloch CA, Coelho NM. Mechanical regulation of myofibroblast phenoconversion and collagen contraction. Exp Cell Res 2019; 379:119-128. [PMID: 30910400 DOI: 10.1016/j.yexcr.2019.03.027] [Citation(s) in RCA: 121] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 02/21/2019] [Accepted: 03/19/2019] [Indexed: 12/17/2022]
Abstract
Activated fibroblasts promote physiological wound repair following tissue injury. However, dysregulation of fibroblast activation contributes to the development of fibrosis by enhanced production and contraction of collagen-rich extracellular matrix. At the peak of their activities, fibroblasts undergo phenotypic conversion into highly contractile myofibroblasts by developing muscle-like features, including formation of contractile actin-myosin bundles. The phenotype and function of fibroblasts and myofibroblasts are mechanically regulated by matrix stiffness using a feedback control system that is integrated with the progress of tissue remodelling. The actomyosin contraction machinery and cell-matrix adhesion receptors are critical elements that are needed for mechanosensing by fibroblasts and the translation of mechanical signals into biological responses. Here, we focus on mechanical and chemical regulation of collagen contraction by fibroblasts and the involvement of these factors in their phenotypic conversion to myofibroblasts.
Collapse
Affiliation(s)
- Boris Hinz
- Laboratory of Tissue Repair and Regeneration, Canada; Faculty of Dentistry, University of Toronto, Toronto, ON, M5G 1G6, Canada
| | | | - Nuno M Coelho
- Faculty of Dentistry, University of Toronto, Toronto, ON, M5G 1G6, Canada.
| |
Collapse
|
224
|
Inflammation and fibrosis in murine models of heart failure. Basic Res Cardiol 2019; 114:19. [PMID: 30887214 DOI: 10.1007/s00395-019-0722-5] [Citation(s) in RCA: 243] [Impact Index Per Article: 40.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 02/12/2019] [Indexed: 02/07/2023]
Abstract
Heart failure is a consequence of various cardiovascular diseases and associated with poor prognosis. Despite progress in the treatment of heart failure in the past decades, prevalence and hospitalisation rates are still increasing. Heart failure is typically associated with cardiac remodelling. Here, inflammation and fibrosis are thought to play crucial roles. During cardiac inflammation, immune cells invade the cardiac tissue and modulate tissue-damaging responses. Cardiac fibrosis, however, is characterised by an increased amount and a disrupted composition of extracellular matrix proteins. As evidence exists that cardiac inflammation and fibrosis are potentially reversible in experimental and clinical set ups, they are interesting targets for innovative heart failure treatments. In this context, animal models are important as they mimic clinical conditions of heart failure patients. The advantages of mice in this respect are short generation times and genetic modifications. As numerous murine models of heart failure exist, the selection of a proper disease model for a distinct research question is demanding. To facilitate this selection, this review aims to provide an overview about the current understanding of the pathogenesis of cardiac inflammation and fibrosis in six frequently used murine models of heart failure. Hence, it compares the models of myocardial infarction with or without reperfusion, transverse aortic constriction, chronic subjection to angiotensin II or deoxycorticosterone acetate, and coxsackievirus B3-induced viral myocarditis in this context. It furthermore provides information about the clinical relevance and the limitations of each model, and, if applicable, about the recent advancements in their methodological proceedings.
Collapse
|
225
|
Li C, Li J, Xue K, Zhang J, Wang C, Zhang Q, Chen X, Gao C, Yu X, Sun L. MicroRNA-143-3p promotes human cardiac fibrosis via targeting sprouty3 after myocardial infarction. J Mol Cell Cardiol 2019; 129:281-292. [PMID: 30878395 DOI: 10.1016/j.yjmcc.2019.03.005] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 02/23/2019] [Accepted: 03/08/2019] [Indexed: 12/22/2022]
Abstract
Myocardial infarction (MI) is one of the most catastrophic diseases threatening human health in the world. Because cardiomyocytes have a minuscule regenerative potential, the natural repair of infarct healing after MI shows fibrotic scar. MicroRNA-143-3p (miR-143-3p) plays a critical regulatory role in various pathophysiological processes in the heart. Sprouty3 (SPRY3) is predicted to be a potential fibrosis-associated target gene of miR-143-3p. The aim was to explore the role and mechanism of miR-143-3p in the infarct healing after MI in vivo and in vitro. Myocardial samples were obtained during autopsy from 12 human patients with or without MI. An increase in miR-143-3p mRNA levels was detected in the infarct zone of human MI samples. Moreover, silencing expression of miR-143-3p by antagomir-143-3p alleviated fibrotic scar in MI model of mice. To assess the mechanism by which miR-143-3p may function in fibrosis, human cardiac fibroblasts (HCFs) were transfected with miR-143-3p mimics and inhibitors. MiR-143-3p overexpression promoted HCFs proliferation, migration, transformation, and extracellular matrix (ECM) excessive accumulation. Additionally, miR-143-3p inhibitors reversed the fibrosis effect of HCFs treated with transforming growth β1 (TGFβ1) in vitro. Importantly, a luciferase reporter assay demonstrated that miR-143-3p could directly bind to the 3'-untranslational region (3'-UTR) of SPRY3 mRNA. Lastly, HCFs transfected with SPRY3 siRNA (si-SPRY3) enhanced the activation of the P38, ERK, and JNK pathways in the process of fibrosis. MiR-143-3p promoted fibrosis along with SPRY3 degradation and the activation of its downstream P38, ERK, and JNK pathways. Our results may contribute to improve the quality of life in MI patients by interfering with the role of miR-143-3p in MI area.
Collapse
Affiliation(s)
- Cong Li
- Department of Pathology and Forensic Medicine, College of Basic Medical Sciences, Dalian Medical University, Dalian 116044, Liaoning Province, China
| | - Jing Li
- Department of Pathology and Forensic Medicine, College of Basic Medical Sciences, Dalian Medical University, Dalian 116044, Liaoning Province, China
| | - Ke Xue
- Department of Pathology and Forensic Medicine, College of Basic Medical Sciences, Dalian Medical University, Dalian 116044, Liaoning Province, China
| | - Jun Zhang
- Department of Pathology and Forensic Medicine, College of Basic Medical Sciences, Dalian Medical University, Dalian 116044, Liaoning Province, China
| | - Cong Wang
- Department of Pathology and Forensic Medicine, College of Basic Medical Sciences, Dalian Medical University, Dalian 116044, Liaoning Province, China
| | - Qingqing Zhang
- Department of Pathology and Forensic Medicine, College of Basic Medical Sciences, Dalian Medical University, Dalian 116044, Liaoning Province, China
| | - Xianlu Chen
- Department of Pathology and Forensic Medicine, College of Basic Medical Sciences, Dalian Medical University, Dalian 116044, Liaoning Province, China
| | - Chuanzhou Gao
- Department of electron microscope, College of Basic Medical Sciences, Dalian Medical University, Dalian 116044, Liaoning Province, China
| | - Xiao Yu
- Department of Pathology and Forensic Medicine, College of Basic Medical Sciences, Dalian Medical University, Dalian 116044, Liaoning Province, China.
| | - Lei Sun
- Department of Pathology and Forensic Medicine, College of Basic Medical Sciences, Dalian Medical University, Dalian 116044, Liaoning Province, China.
| |
Collapse
|
226
|
Cai L, Qi B, Wu X, Peng S, Zhou G, Wei Y, Xu J, Chen S, Liu S. Circular RNA Ttc3 regulates cardiac function after myocardial infarction by sponging miR-15b. J Mol Cell Cardiol 2019; 130:10-22. [PMID: 30876857 DOI: 10.1016/j.yjmcc.2019.03.007] [Citation(s) in RCA: 102] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 02/12/2019] [Accepted: 03/10/2019] [Indexed: 12/24/2022]
Abstract
The apoptotic death of cardiomyocytes critically contributes to cardiac remodeling after myocardial infarction (MI). Circular RNAs (circRNAs) are important regulators for a variety of biological functions. Circ-Ttc3 represents one of the top highest expressed circRNAs in the heart; however, its role in MI remains unknown. Herein, we found that circ-Ttc3 was markedly upregulated in the ischemic myocardium and the cardiomyocytes subjected to hypoxic insult. Forced expression of circ-Ttc3 in cardiomyocytes counteracted hypoxia-induced ATP depletion and apoptotic death, in sharp contrast to circ-Ttc3 knockdown. Accordingly, experiments with AAV9-cTnt-mediated knockdown of cardiac circ-Ttc3 in a rat model of MI recapitulated the in vitro findings, and showed the deterioration of cardiac dysfunction after MI. Furthermore, we identified that circ-Ttc3 sponged an endogenous miR-15b-5p to sequester and inhibit its activity, leading to the increased Arl2 expression. Conversely, knockdown of Arl2 partially abolished the beneficial effects of circ-Ttc3 overexpression on ATP production and apoptosis of cardiomyocytes. Thus, our findings revealed the cardioprotective role of circ-Ttc3 in MI. The miR-15b-Arl2 regulatory cascade underlies the protection against MI-induced cardiomyocyte apoptosis by circ-Ttc3.
Collapse
Affiliation(s)
- Lidong Cai
- Department of Cardiology, Shanghai General Hospital, School of Medicine, Shanghai Jiaotong University, Hongkou District, Shanghai, China
| | - Baozhen Qi
- Department of Cardiology, Shanghai Institute of Cardiovascular Disease, Zhongshan Hospital, Fudan University, Shanghai 200080, China
| | - Xiaoyu Wu
- Department of Cardiology, Shanghai General Hospital, School of Medicine, Shanghai Jiaotong University, Hongkou District, Shanghai, China
| | - Shi Peng
- Department of Cardiology, Shanghai General Hospital, School of Medicine, Shanghai Jiaotong University, Hongkou District, Shanghai, China
| | - Genqing Zhou
- Department of Cardiology, Shanghai General Hospital, School of Medicine, Shanghai Jiaotong University, Hongkou District, Shanghai, China
| | - Yong Wei
- Department of Cardiology, Shanghai General Hospital, School of Medicine, Shanghai Jiaotong University, Hongkou District, Shanghai, China
| | - Juan Xu
- Department of Cardiology, Shanghai General Hospital, School of Medicine, Shanghai Jiaotong University, Hongkou District, Shanghai, China
| | - Songwen Chen
- Department of Cardiology, Shanghai General Hospital, School of Medicine, Shanghai Jiaotong University, Hongkou District, Shanghai, China.
| | - Shaowen Liu
- Department of Cardiology, Shanghai General Hospital, School of Medicine, Shanghai Jiaotong University, Hongkou District, Shanghai, China.
| |
Collapse
|
227
|
The whole transcriptome and proteome changes in the early stage of myocardial infarction. Cell Death Discov 2019; 5:73. [PMID: 30854232 PMCID: PMC6399251 DOI: 10.1038/s41420-019-0152-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 02/10/2019] [Indexed: 12/31/2022] Open
Abstract
As the most severe manifestation of coronary artery disease, myocardial infarction (MI) is a complex and multifactorial pathophysiologic process. However, the pathogenesis that underlies MI remains unclear. Here, we generated a MI mouse model by ligation of the proximal left anterior descending coronary artery. The transcriptome and proteome, at different time points after MI, were detected and analysed. Immune-related pathways, cell cycle-related pathways, and extracellular matrix remodelling-related pathways were significantly increased after MI. Not only innate immune cells but also adaptive immune cells participated in the early stage of MI. Proteins that functioned in blood agglutination, fibrinolysis, secretion, and immunity were significantly changed after MI. Nppa, Serpina3n, and Anxa1, three secreted proteins that can easily be detected in blood, were significantly changed after MI. Our discoveries not only reveal the molecular and cellular changes in MI but also identify potential candidate biomarkers of MI for clinical diagnosis or treatment.
Collapse
|
228
|
Liu Z, Xu Q, Yang Q, Cao J, Wu C, Peng H, Zhang X, Chen J, Cheng G, Wu Y, Shi R, Zhang G. Vascular peroxidase 1 is a novel regulator of cardiac fibrosis after myocardial infarction. Redox Biol 2019; 22:101151. [PMID: 30844643 PMCID: PMC6402381 DOI: 10.1016/j.redox.2019.101151] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Revised: 02/14/2019] [Accepted: 02/21/2019] [Indexed: 12/11/2022] Open
Abstract
Cardiac fibrosis is the most important mechanism contributing to cardiac remodeling after myocardial infarction (MI). VPO1 is a heme enzyme that uses hydrogen peroxide (H2O2) to produce hypochlorous acid (HOCl). Our previous study has demonstrated that VPO1 regulates myocardial ischemic reperfusion and renal fibrosis. We investigated the role of VPO1 in cardiac fibrosis after MI. The results showed that VPO1 expression was robustly upregulated in the failing human heart with ischemic cardiomyopathy and in a murine model of MI accompanied by severe cardiac fibrosis. Most importantly, knockdown of VPO1 by tail vein injection of VPO1 siRNA significantly reduced cardiac fibrosis and improved cardiac function and survival rate. In VPO1 knockdown mouse model and cardiac fibroblasts cultured with TGF-β1, VPO1 contributes to cardiac fibroblasts differentiation, migration, collagen I synthesis and proliferation. Mechanistically, the fibrotic effects following MI of VPO1 manifested partially through HOCl formation to activate Smad2/3 and ERK1/2. Thus, we conclude that VPO1 is a crucial regulator of cardiac fibrosis after MI by mediating HOCl/Smad2/3 and ERK1/2 signaling pathways, implying a promising therapeutic target in ischemic cardiomyopathy.
Collapse
Affiliation(s)
- Zhaoya Liu
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Qian Xu
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Qixin Yang
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jing Cao
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Cong Wu
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Huihui Peng
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xinyi Zhang
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jia Chen
- Department of Humanistic Nursing, Xiangya Nursing School, Central South University, Changsha, Hunan, China
| | - Guangjie Cheng
- Division of Pulmonary, Allergy & Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, USA
| | - Yueheng Wu
- Department of Cardiovascular Medicine, Guangdong General Hospital, Guangzhou, Guangdong China
| | - Ruizheng Shi
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Guogang Zhang
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
229
|
Vainio LE, Szabó Z, Lin R, Ulvila J, Yrjölä R, Alakoski T, Piuhola J, Koch WJ, Ruskoaho H, Fouse SD, Seeley TW, Gao E, Signore P, Lipson KE, Magga J, Kerkelä R. Connective Tissue Growth Factor Inhibition Enhances Cardiac Repair and Limits Fibrosis After Myocardial Infarction. ACTA ACUST UNITED AC 2019; 4:83-94. [PMID: 30847422 PMCID: PMC6390503 DOI: 10.1016/j.jacbts.2018.10.007] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 10/22/2018] [Accepted: 10/23/2018] [Indexed: 12/11/2022]
Abstract
Myocardial infarction (MI)-induced cardiac fibrosis attenuates cardiac contractile function, and predisposes to arrhythmias and sudden cardiac death. Expression of connective tissue growth factor (CTGF) is elevated in affected organs in virtually every fibrotic disorder and in the diseased human myocardium. Mice were subjected to treatment with a CTGF monoclonal antibody (mAb) during infarct repair, post-MI left ventricular (LV) remodeling, or acute ischemia-reperfusion injury. CTGF mAb therapy during infarct repair improved survival and reduced LV dysfunction, and reduced post-MI LV hypertrophy and fibrosis. Mechanistically, CTGF mAb therapy induced expression of cardiac developmental and/or repair genes and attenuated expression of inflammatory and/or fibrotic genes.
Collapse
Key Words
- CTGF, connective tissue growth factor
- ECM, extracellular matrix
- ERK, extracellular signal-regulated kinase
- FB, fibroblast
- HF, heart failure
- I/R, ischemia−reperfusion
- Ig, immunoglobulin
- JNK, c-Jun N-terminal kinase
- LV, left ventricular
- MI, myocardial infarction
- TGF, transforming growth factor
- connective tissue growth factor monoclonal antibody
- fibrosis
- heart failure
- ischemia−reperfusion injury
- left ventricle
- mAb, monoclonal antibody
- myocardial infarction
Collapse
Affiliation(s)
- Laura E Vainio
- Research Unit of Biomedicine, Department of Pharmacology and Toxicology, University of Oulu, Oulu, Finland.,Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Zoltán Szabó
- Research Unit of Biomedicine, Department of Pharmacology and Toxicology, University of Oulu, Oulu, Finland.,Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Ruizhu Lin
- Research Unit of Biomedicine, Department of Pharmacology and Toxicology, University of Oulu, Oulu, Finland
| | - Johanna Ulvila
- Research Unit of Biomedicine, Department of Pharmacology and Toxicology, University of Oulu, Oulu, Finland
| | - Raisa Yrjölä
- Research Unit of Biomedicine, Department of Pharmacology and Toxicology, University of Oulu, Oulu, Finland
| | - Tarja Alakoski
- Research Unit of Biomedicine, Department of Pharmacology and Toxicology, University of Oulu, Oulu, Finland.,Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Jarkko Piuhola
- Division of Cardiology, Department of Internal Medicine, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Walter J Koch
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania
| | - Heikki Ruskoaho
- Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | | | | | - Erhe Gao
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania
| | | | | | - Johanna Magga
- Research Unit of Biomedicine, Department of Pharmacology and Toxicology, University of Oulu, Oulu, Finland.,Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Risto Kerkelä
- Research Unit of Biomedicine, Department of Pharmacology and Toxicology, University of Oulu, Oulu, Finland.,Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
| |
Collapse
|
230
|
de Boer RA, De Keulenaer G, Bauersachs J, Brutsaert D, Cleland JG, Diez J, Du XJ, Ford P, Heinzel FR, Lipson KE, McDonagh T, Lopez-Andres N, Lunde IG, Lyon AR, Pollesello P, Prasad SK, Tocchetti CG, Mayr M, Sluijter JPG, Thum T, Tschöpe C, Zannad F, Zimmermann WH, Ruschitzka F, Filippatos G, Lindsey ML, Maack C, Heymans S. Towards better definition, quantification and treatment of fibrosis in heart failure. A scientific roadmap by the Committee of Translational Research of the Heart Failure Association (HFA) of the European Society of Cardiology. Eur J Heart Fail 2019; 21:272-285. [PMID: 30714667 PMCID: PMC6607480 DOI: 10.1002/ejhf.1406] [Citation(s) in RCA: 182] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 11/28/2018] [Accepted: 12/03/2018] [Indexed: 12/19/2022] Open
Abstract
Fibrosis is a pivotal player in heart failure development and progression. Measurements of (markers of) fibrosis in tissue and blood may help to diagnose and risk stratify patients with heart failure, and its treatment may be effective in preventing heart failure and its progression. A lack of pathophysiological insights and uniform definitions has hampered the research in fibrosis and heart failure. The Translational Research Committee of the Heart Failure Association discussed several aspects of fibrosis in their workshop. Early insidious perturbations such as subclinical hypertension or inflammation may trigger first fibrotic events, while more dramatic triggers such as myocardial infarction and myocarditis give rise to full blown scar formation and ongoing fibrosis in diseased hearts. Aging itself is also associated with a cardiac phenotype that includes fibrosis. Fibrosis is an extremely heterogeneous phenomenon, as several stages of the fibrotic process exist, each with different fibrosis subtypes and a different composition of various cells and proteins — resulting in a very complex pathophysiology. As a result, detection of fibrosis, e.g. using current cardiac imaging modalities or plasma biomarkers, will detect only specific subforms of fibrosis, but cannot capture all aspects of the complex fibrotic process. Furthermore, several anti‐fibrotic therapies are under investigation, but such therapies generally target aspecific aspects of the fibrotic process and suffer from a lack of precision. This review discusses the mechanisms and the caveats and proposes a roadmap for future research.
Collapse
Affiliation(s)
- Rudolf A de Boer
- University Medical Center Groningen, University of Groningen, Department of Cardiology, Groningen, The Netherlands
| | | | - Johann Bauersachs
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Dirk Brutsaert
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| | - John G Cleland
- Robertson Centre for Biostatistics & Clinical Trials, University of Glasgow, Glasgow, UK
| | - Javier Diez
- Program of Cardiovascular Diseases, Center for Applied Medical Research, Departments of Nephrology, and Cardiology and Cardiac Surgery, University Clinic, University of Navarra, Pamplona, Spain
| | - Xiao-Jun Du
- Baker Heart and Diabetes Institute, Melbourne, Australia
| | | | - Frank R Heinzel
- Department of Cardiology, Campus Virchow-Klinikum, Charite Universitaetsmedizin Berlin, Berlin, Germany
| | | | | | - Natalia Lopez-Andres
- Cardiovascular Translational Research, Navarrabiomed, Complejo Hospitalario de Navarra, Universidad Publica de Navarra, Idisna, Spain
| | - Ida G Lunde
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Alexander R Lyon
- Royal Brompton Hospital, and Imperial College London, London, UK
| | | | | | - Carlo G Tocchetti
- Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Manuel Mayr
- The James Black Centre, King's College, University of London, London, UK
| | - Joost P G Sluijter
- University Medical Centre Utrecht, Experimental Cardiology Laboratory, UMC Utrecht Regenerative Medicine Center, University Utrecht, Utrecht, The Netherlands
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany.,REBIRTH Excellence Cluster, Hannover Medical School, Hannover, Germany.,DZHK (German Center for Cardiovascular Research) partner site Berlin, Berlin, Germany
| | - Carsten Tschöpe
- Department of Cardiology, Campus Virchow-Klinikum, Charite Universitaetsmedizin Berlin, Berlin, Germany
| | - Faiez Zannad
- Centre d'Investigation Clinique, CHU de Nancy, Nancy, France
| | - Wolfram-Hubertus Zimmermann
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Göttingen, Germany.,DZHK (German Center for Cardiovascular Research) partner site Göttingen, Göttingen, Germany
| | - Frank Ruschitzka
- Department of Cardiology, University Heart Center, University Hospital Zurich, Zurich, Switzerland
| | - Gerasimos Filippatos
- Heart Failure Unit, Department of Cardiology, School of Medicine, Athens University Hospital Attikon, National and Kapodistrian University of Athens, Athens, Greece
| | - Merry L Lindsey
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center and Research Service, G.V. (Sonny) Montgomery Veterans Affairs Medical Center, Jackson, MS, USA
| | - Christoph Maack
- Comprehensive Heart Failure Centre, University and University Hospital Würzburg, Würzburg, Germany
| | - Stephane Heymans
- Department of Cardiology, CARIM School for Cardiovascular Diseases Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands.,Department of Cardiovascular Sciences, Centre for Molecular and Vascular Biology, KU Leuven, Leuven, Belgium.,The Netherlands Heart Institute, Nl-HI, Utrecht, The Netherlands
| |
Collapse
|
231
|
Cai W, Zhong S, Zheng F, Zhang Y, Gao F, Xu H, Cai X, Lan J, Huang D, Shi G. Angiotensin II confers resistance to apoptosis in cardiac myofibroblasts through the AT1/ERK1/2/RSK1 pathway. IUBMB Life 2019; 71:261-276. [PMID: 30452117 DOI: 10.1002/iub.1967] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 10/05/2018] [Accepted: 10/10/2018] [Indexed: 02/05/2023]
Abstract
Myofibroblast apoptosis is essential for normal resolution of wound repair, including cardiac infarction repair. Impaired cardiac myofibroblast (CMF) apoptosis is associated with excessive extracellular matrix (ECM) deposition, which could be responsible for pathological cardiac fibrosis. Conventionally, angiotensin II (Ang II), a soluble peptide, is implicated in fibrogenesis because it induces cardiac fibroblast (CFb) proliferation, differentiation, and collagen synthesis. However, the role of Ang II in regulation of CMF survival and apoptosis has not been fully clarified. In this report, we cultured neonatal rat CFbs, which transform into CMFs after passage 3 (6-8 days), and investigated the effects of Ang II on CMFs challenged by TNF-α combined with cycloheximide and the underlying mechanisms. Here, we show that Ang II rapidly activates MAPKs but not AKT in CMFs and confers apoptosis resistance, as evidenced by the inhibition of caspase-3 cleavage, early apoptotic cells and late apoptotic cells. This inhibitory effect of Ang II was reversed by blockade of AT1 or inactivation of ERK1/2 or RSK1 but not AT2, indicating that activation of the prosurvival AT1/ERK1/2/RSK1 signaling pathway mediates apoptosis resistance. TGF-β, a latent fibrotic factor, was found to have no relation to Ang II-induced apoptosis resistance in our study. Furthermore, Ang II-mediated apoptosis resistance, which was conferred by activation of the AT1/ERK1/2/RSK1 signaling pathway, was also confirmed in human adult ventricular cardiac myofibroblasts. Collectively, our findings suggest a novel profibrotic mechanism of Ang II in which it promotes myofibroblast resistance to apoptosis in addition to classical mechanisms, providing a potential novel therapeutic approach by targeting prosurvival signaling pathways. © 2018 IUBMB Life, 71(1):261-276, 2019.
Collapse
MESH Headings
- Angiotensin II/pharmacology
- Angiotensin II Type 2 Receptor Blockers/pharmacology
- Animals
- Apoptosis/drug effects
- Apoptosis/genetics
- Butadienes/pharmacology
- Cell Survival/drug effects
- Cycloheximide/pharmacology
- Flavonoids/pharmacology
- Gene Expression Regulation
- Humans
- Imidazoles/pharmacology
- Losartan/pharmacology
- Mitogen-Activated Protein Kinase 1/antagonists & inhibitors
- Mitogen-Activated Protein Kinase 1/genetics
- Mitogen-Activated Protein Kinase 1/metabolism
- Mitogen-Activated Protein Kinase 3/antagonists & inhibitors
- Mitogen-Activated Protein Kinase 3/genetics
- Mitogen-Activated Protein Kinase 3/metabolism
- Myocardium/cytology
- Myocardium/metabolism
- Myofibroblasts/cytology
- Myofibroblasts/drug effects
- Myofibroblasts/metabolism
- Nitriles/pharmacology
- Primary Cell Culture
- Protein Kinase Inhibitors/pharmacology
- Pyridines/pharmacology
- RNA, Small Interfering/genetics
- RNA, Small Interfering/metabolism
- Rats
- Rats, Sprague-Dawley
- Receptor, Angiotensin, Type 1/genetics
- Receptor, Angiotensin, Type 1/metabolism
- Ribosomal Protein S6 Kinases, 90-kDa/antagonists & inhibitors
- Ribosomal Protein S6 Kinases, 90-kDa/genetics
- Ribosomal Protein S6 Kinases, 90-kDa/metabolism
- Signal Transduction
- Tumor Necrosis Factor-alpha/pharmacology
Collapse
Affiliation(s)
- Wenfeng Cai
- Department of Pharmacology, Shantou University Medical College, Shantou, Guangdong, China
- Pharmaceutical Laboratory, The First Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong, China
| | - Shuping Zhong
- Department of Biochemistry and Molecular Biology, University of Southern California, Los Angeles, California, USA
| | - Fuchun Zheng
- Pharmaceutical Laboratory, The First Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong, China
| | - Yanmei Zhang
- Department of Pharmacology, Shantou University Medical College, Shantou, Guangdong, China
- Pharmaceutical Laboratory, The First Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong, China
| | - Fenfei Gao
- Department of Pharmacology, Shantou University Medical College, Shantou, Guangdong, China
- Pharmaceutical Laboratory, The First Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong, China
| | - Hongquan Xu
- Department of Plastic Surgery, The First Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong, China
| | - Xiangna Cai
- Department of Plastic Surgery, The First Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong, China
| | - Junhong Lan
- Department of Plastic Surgery, The First Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong, China
| | - Danmei Huang
- Department of Pharmacology, Shantou University Medical College, Shantou, Guangdong, China
- Pharmaceutical Laboratory, The First Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong, China
| | - Ganggang Shi
- Department of Pharmacology, Shantou University Medical College, Shantou, Guangdong, China
- Pharmaceutical Laboratory, The First Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong, China
- Department of Cardiovascular Diseases, The First Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong, China
| |
Collapse
|
232
|
Li D, Wang H, He JY, Wang CL, Feng WJ, Shen C, Zhu JF, Wang DL, Chen XD. Inflammatory and fibrosis infiltration in synovium associated with the progression in developmental dysplasia of the hip. Mol Med Rep 2019; 19:2808-2816. [PMID: 30720141 DOI: 10.3892/mmr.2019.9910] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 06/13/2018] [Indexed: 11/05/2022] Open
Abstract
Developmental dysplasia of the hip (DDH) is a common musculoskeletal disorder characterized by progressive joint soreness and limited mobility. The aim of the present study was to investigate the pathological changes and inflammatory infiltration in the hypertrophic synovium of the hip joint associated with the progression of DDH. Synovial biopsies in the hip joint are obtained from patients with moderate DDH and severe DDH during surgery. These biopsies are processed for histological and immunohistochemical (IHC) analysis and investigation of the pathological processes in a synovium, including types of inflammatory cell infiltration, synovial angiogenesis and fibrosis, neuron endings and neuropeptide invasion. Correlation analysis was performed between the mean optical density (MOD) of each antibody, and Harris hip score (HHS) and visual analogue score (VAS) using the Spearman correlation test. Chronic inflammation in the synovium was observed via the positive IHC staining of inflammatory cells, such as T cells, B cells, macrophages and leukocytes. Excessive staining of vimentin and α smooth muscle actin in the synovium of severe DDH represented significant fibrosis and angiogenesis. These targets were also significantly correlated with HHS in severe DDH. The MOD levels of CD68 (indicators of macrophage) indicated apparent correlations with HHS and VAS in patients with severe DDH. The labels of nerve fibers and pain transmission indicators were as follows: Neurofilament‑200 and substance P. Calcitonin gene‑related peptide was upregulated in the synovium of severe DDH in contrast to that in the synovium of moderate DDH. The MOD levels of NF‑200, SP and CGRP were correlated with VAS in severe DDH. The pathology of DDH includes chronic inflammatory cell infiltration corresponding with nerve fibers and fibroblastic proliferation, which might contribute to arthritis progression and joint soreness in DDH.
Collapse
Affiliation(s)
- De Li
- Department of Orthopedic Surgery, Xin‑Hua Hospital, Shanghai Jiao‑Tong University School of Medicine, Shanghai 200092, P.R. China
| | - Hui Wang
- Department of Orthopedic Surgery, Xin‑Hua Hospital, Shanghai Jiao‑Tong University School of Medicine, Shanghai 200092, P.R. China
| | - Ji-Ye He
- Department of Orthopedic Surgery, Xin‑Hua Hospital, Shanghai Jiao‑Tong University School of Medicine, Shanghai 200092, P.R. China
| | - Cheng-Long Wang
- Department of Orthopedic Surgery, Xin‑Hua Hospital, Shanghai Jiao‑Tong University School of Medicine, Shanghai 200092, P.R. China
| | - Wei-Jia Feng
- Department of Orthopedic Surgery, Xin‑Hua Hospital, Shanghai Jiao‑Tong University School of Medicine, Shanghai 200092, P.R. China
| | - Chao Shen
- Department of Orthopedic Surgery, Xin‑Hua Hospital, Shanghai Jiao‑Tong University School of Medicine, Shanghai 200092, P.R. China
| | - Jun-Feng Zhu
- Department of Orthopedic Surgery, Xin‑Hua Hospital, Shanghai Jiao‑Tong University School of Medicine, Shanghai 200092, P.R. China
| | - Dong-Liang Wang
- Department of Orthopedic Surgery, Xin‑Hua Hospital, Shanghai Jiao‑Tong University School of Medicine, Shanghai 200092, P.R. China
| | - Xiao-Dong Chen
- Department of Orthopedic Surgery, Xin‑Hua Hospital, Shanghai Jiao‑Tong University School of Medicine, Shanghai 200092, P.R. China
| |
Collapse
|
233
|
Specialized Pro-resolving Mediators Directs Cardiac Healing and Repair with Activation of Inflammation and Resolution Program in Heart Failure. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1161:45-64. [PMID: 31562621 DOI: 10.1007/978-3-030-21735-8_6] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
After myocardial infarction, splenic leukocytes direct biosynthesis of specialized pro-resolving mediators (SPMs) that are essential for the resolution of inflammation and tissue repair. In a laboratory environment, after coronary ligation of healthy risk free rodents (young adult mice) leukocytes biosynthesize SPMs with induced activity of lipoxygenases and cyclooxygenases, which facilitate cardiac repair. Activated monocytes/macrophages drive the biosynthesis of SPMs following experimental myocardial infarction in mice during the acute heart failure. In the presented review, we provided the recent updates on SPMs (resolvins, lipoxins and maresins) in cardiac repair that may serve as novel therapeutics for future heart failure therapy/management. We incorporated the underlying causes of non-resolving inflammation following cardiac injury if superimposed with obesity, hypertension, diabetes, disrupted circadian rhythm, co-medication (painkillers or oncological therapeutics), and/or aging that may delay or impair the biosynthesis of SPMs, intensifying pathological remodeling in heart failure.
Collapse
|
234
|
Pan Y, Zhou Z, Zhang H, Zhou Y, Li Y, Li C, Chen X, Yang S, Liao Y, Qiu Z. The ATRQβ-001 vaccine improves cardiac function and prevents postinfarction cardiac remodeling in mice. Hypertens Res 2018; 42:329-340. [PMID: 30587854 DOI: 10.1038/s41440-018-0185-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 09/09/2018] [Accepted: 09/09/2018] [Indexed: 11/09/2022]
Abstract
We invented the ATRQβ-001 hypertension vaccine, which targeted angiotensin II type 1 receptor (AT1R) and showed a desirable blocking effect for AT1R. The purpose of this study was to investigate whether the ATRQβ-001 vaccine could improve cardiac function and prevent cardiac remodeling after acute myocardial infarction (AMI). C57BL/6 male mice were randomly assigned into four groups: sham + VLP, MI + VLP, MI + ATRQβ-001, and MI + valsartan. Mice were administered Qβ virus-like particle (Qβ-VLP, 100 μg/time), ATRQβ-001 vaccine (100 μg/time), and valsartan (6 mg/kg/day) before AMI, which was induced by permanently ligating the left anterior descending coronary artery. The effect of the ATRQβ-001 vaccine on cardiac function and cardiac remodeling was observed by following up for 1 week, 4 weeks, and 12 weeks post MI. The ATRQβ-001 vaccine significantly reduced sudden cardiac death and increased survival rates (compared with MI + VLP, 80% versus 55% and mean estimate (days) 68.4 ± 7.0 versus 47.8 ± 8.9, respectively; p = 0.046) post MI. Echocardiography showed that the ATRQβ-001 vaccine remarkably improved cardiac function (left ventricular ejection fraction, 24.8 ± 7.0% versus 13.2 ± 3.8%, p = 0.005) post MI. Histological analysis revealed that the ATRQβ-001 vaccine obviously mitigated myocardial inflammation, apoptosis, and fibrosis after AMI. Further, the ATRQβ-001 vaccine significantly inhibited the TGF-β1/Smad2/3 signaling pathway. Assessment of the renin-angiotensin system (RAS) demonstrated that the ATRQβ-001 vaccine did not cause obvious feedback of circulating RAS, but prominently attenuated the expression of AT1R, compared with the other groups at 4 and 12 weeks after AMI. In conclusion, the ATRQβ-001 vaccine decreased mortality and improved cardiac function and remodeling after AMI.
Collapse
Affiliation(s)
- Yajie Pan
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Institute of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Key Lab of Molecular Biological Targeted Therapies of the Ministry of Education, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Zihua Zhou
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Institute of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Key Lab of Molecular Biological Targeted Therapies of the Ministry of Education, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Hongrong Zhang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Institute of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Key Lab of Molecular Biological Targeted Therapies of the Ministry of Education, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yanzhao Zhou
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Institute of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Key Lab of Molecular Biological Targeted Therapies of the Ministry of Education, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yingying Li
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Institute of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Key Lab of Molecular Biological Targeted Therapies of the Ministry of Education, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Chang Li
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Institute of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Key Lab of Molecular Biological Targeted Therapies of the Ministry of Education, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xiao Chen
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Institute of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Key Lab of Molecular Biological Targeted Therapies of the Ministry of Education, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Shijun Yang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Institute of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Key Lab of Molecular Biological Targeted Therapies of the Ministry of Education, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yuhua Liao
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Institute of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Key Lab of Molecular Biological Targeted Therapies of the Ministry of Education, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Zhihua Qiu
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China. .,Institute of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China. .,Key Lab of Molecular Biological Targeted Therapies of the Ministry of Education, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
235
|
Li YL, Hao WJ, Chen BY, Chen J, Li GQ. Cardiac Fibroblast-Specific Activating Transcription Factor 3 Promotes Myocardial Repair after Myocardial Infarction. Chin Med J (Engl) 2018; 131:2302-2309. [PMID: 30246716 PMCID: PMC6166466 DOI: 10.4103/0366-6999.241794] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Background: Myocardial ischemia injury is one of the leading causes of death and disability worldwide. Cardiac fibroblasts (CFs) have central roles in modulating cardiac function under pathophysiological conditions. Activating transcription factor 3 (ATF3) plays a self-protective role in counteracting CF dysfunction. However, the precise function of CF-specific ATF3 during myocardial infarction (MI) injury/repair remains incompletely understood. The aim of this study was to determine whether CF-specific ATF3 affected cardiac repair after MI. Methods: Fifteen male C57BL/6 wild-type mice were performed with MI operation to observe the expression of ATF3 at 0, 0.5, 1.0, 3.0, and 7.0 days postoperation. Model for MI was constructed in ATF3TGfl/flCol1a2-Cre+ (CF-specific ATF3 overexpression group, n = 5) and ATF3TGfl/flCol1a2-Cre− male mice (without CF-specific ATF3 overexpression group, n = 5). In addition, five mice of ATF3TGfl/flCol1a2-Cre+ and ATF3TGfl/flCol1a2-Cre− were subjected to sham MI operation. Heart function was detected by ultrasound and left ventricular remodeling was observed by Masson staining (myocardial fibrosis area was detected by blue collagen deposition area) at the 28th day after MI surgery in ATF3TGfl/flCol1a2-Cre+ and ATF3TGfl/flCol1a2-Cre− mice received sham or MI operation. Quantitative real-time polymerase chain reaction (qRT-PCR) was used to detect cell proliferation/cell cycle-related gene expression in cardiac tissue. BrdU staining was used to detect fibroblast proliferation. Results: After establishment of an MI model, we found that ATF3 proteins were increased in the heart of mice after MI surgery and dominantly expressed in CFs. Genetic overexpression of ATF3 in CFs (ATF3TGfl/flCol1a2-Cre+ group) resulted in an improvement in the heart function as indicated by increased cardiac ejection fraction (41.0% vs. 30.5%, t = 8.610, P = 0.001) and increased fractional shortening (26.8% vs. 18.1%, t = 7.173, P = 0.002), which was accompanied by a decrease in cardiac scar area (23.1% vs. 11.0%, t = 8.610, P = 0.001). qRT-PCR analysis of CFs isolated from ATF3TGfl/flCol1a2-Cre+ and ATF3TGfl/flCol1a2-Cre− ischemic hearts revealed a distinct transcriptional profile in ATF3-overexpressing CFs, displaying pro-proliferation properties. BrdU-positive cells significantly increased in ATF3-overexpressing CFs than control CFs under angiotensin II stimuli (11.5% vs. 6.8%, t = 31.599, P = 0.001) or serum stimuli (31.6% vs. 20.1%, t = 31.599, P = 0.001). The 5(6)-carboxyfluorescein N-hydroxysuccinimidyl ester assay showed that the cell numbers of the P2 and P3 generations were higher in the ATF3-overexpressing CFs at 24 h (P2: 91.6% vs. 71.8%, t = 8.465, P = 0.015) and 48 h (P3: 81.6% vs. 51.1%, t = 9.029, P = 0.012) after serum stimulation. Notably, ATF3 overexpression-induced CF proliferation was clearly increased in the heart after MI injury. Conclusions: We identify that CF-specific ATF3 might contribute to be MI repair through upregulating the expression of cell cycle/proliferation-related genes and enhancing cell proliferation.
Collapse
Affiliation(s)
- Yu-Lin Li
- Key Laboratory of Remodeling-Related Cardiovascular Disease (Ministry of Education), Beijing Anzhen Hospital Affiliated to Captital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing 100029, China
| | - Wen-Jing Hao
- Key Laboratory of Remodeling-Related Cardiovascular Disease (Ministry of Education), Beijing Anzhen Hospital Affiliated to Captital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing 100029, China
| | - Bo-Ya Chen
- Key Laboratory of Remodeling-Related Cardiovascular Disease (Ministry of Education), Beijing Anzhen Hospital Affiliated to Captital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing 100029, China
| | - Jing Chen
- Key Laboratory of Remodeling-Related Cardiovascular Disease (Ministry of Education), Beijing Anzhen Hospital Affiliated to Captital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing 100029, China
| | - Guo-Qi Li
- Key Laboratory of Remodeling-Related Cardiovascular Disease (Ministry of Education), Beijing Anzhen Hospital Affiliated to Captital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing 100029, China
| |
Collapse
|
236
|
Kaiser NJ, Kant RJ, Minor AJ, Coulombe KLK. Optimizing Blended Collagen-Fibrin Hydrogels for Cardiac Tissue Engineering with Human iPSC-derived Cardiomyocytes. ACS Biomater Sci Eng 2018; 5:887-899. [PMID: 30775432 PMCID: PMC6372981 DOI: 10.1021/acsbiomaterials.8b01112] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 12/10/2018] [Indexed: 01/08/2023]
Abstract
![]()
Natural
polymer hydrogels are used ubiquitously as scaffold materials
for cardiac tissue engineering as well as for soft tissue engineering
more broadly because of FDA approval, minimal immunogenicity, and
well-defined physiological clearance pathways. However, the relationships
between natural polymer hydrogels and resident cell populations in
directing the development of engineered tissues are poorly defined.
This interaction is of particular concern for tissues prepared
with iPSC-derived cell populations, in which population purity and
batch-to-batch variability become additional critical factors to consider.
Herein, the design space for a blended fibrin and collagen scaffold
is characterized for applications in creating engineered myocardium
with human iPSC-derived cardiomyocytes. Stiffness values of the acellular
hydrogel formulations approach those of native myocardium in compression,
but deviate significantly in tension when compared to rat myocardium
in both transverse and longitudinal fiber orientations. A response
surface methodology approach to understanding the relationship between
collagen concentration, fibrin concentration, seeding density, and
cardiac purity found a statistically significant predictive model
across three repeated studies that confirms that all of these factors
contribute to tissue compaction. In these constructs, increased fibrin
concentration and seeding density were each associated with increased
compaction, while increased collagen concentration was associated
with decreased compaction. Both the lowest (24.4% cTnT+) and highest (60.2% cTnT+) cardiomyocyte purities evaluated
were associated with decreased compaction, whereas the greatest compaction
was predicted to occur in constructs prepared with a 40–50%
cTnT+ population. Constructs prepared with purified cardiomyocytes
(≥75.5% cTnT+) compacted and formed syncytia well,
although increased fibrin concentration in these groups was associated
with decreased compaction, a reversal of the trend observed in unpurified
cardiomyocytes. This study demonstrates an analytical approach to
understanding cell–scaffold interactions in engineered tissues
and provides a foundation for the development of more sophisticated
and customized scaffold platforms for human cardiac tissue engineering.
Collapse
Affiliation(s)
- Nicholas J Kaiser
- Center for Biomedical Engineering, Brown University, Providence, Rhode Island 02912, United States
| | - Rajeev J Kant
- Center for Biomedical Engineering, Brown University, Providence, Rhode Island 02912, United States
| | - Alicia J Minor
- Center for Biomedical Engineering, Brown University, Providence, Rhode Island 02912, United States
| | - Kareen L K Coulombe
- Center for Biomedical Engineering, Brown University, Providence, Rhode Island 02912, United States
| |
Collapse
|
237
|
Dong C, Ma A, Shang L. Nanoparticles for postinfarct ventricular remodeling. Nanomedicine (Lond) 2018; 13:3037-3050. [PMID: 30354963 DOI: 10.2217/nnm-2018-0264] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
In recent years, tremendous progress has been made in the treatment of acute myocardial infarction, but pathological ventricular remodeling often causes survivors to suffer from fatal heart failure. Currently, there is no effective therapy to attenuate ventricular remodeling. Recently, nanoparticle-based drug delivery systems are widely applied in biomedicine especially in cancer and liver fibrosis, owing to its excellent physical, chemical and biological properties. Therefore, the use of nanoparticles as delivery vehicles of small molecules, polypeptides, etc. to improve postinfarct ventricular remodeling is expected. In this review, we summarize the updated researches in this fast-growing area and suggest further works needed.
Collapse
Affiliation(s)
- Caijuan Dong
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Aiqun Ma
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Lijun Shang
- School of Chemistry & Biosciences, University of Bradford, Bradford, BD7 1DP, UK
| |
Collapse
|
238
|
Felisbino MB, McKinsey TA. Epigenetics in Cardiac Fibrosis: Emphasis on Inflammation and Fibroblast Activation. JACC Basic Transl Sci 2018; 3:704-715. [PMID: 30456341 PMCID: PMC6234501 DOI: 10.1016/j.jacbts.2018.05.003] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 05/24/2018] [Accepted: 05/25/2018] [Indexed: 12/18/2022]
Abstract
Chemical modifications to nucleosomal DNA and histone tails greatly influence transcription of adjacent and distant genes, a mode of gene regulation referred to as epigenetic control. Here, the authors summarize recent findings that have illustrated crucial roles for epigenetic regulatory enzymes and reader proteins in the control of cardiac fibrosis. Particular emphasis is placed on epigenetic regulation of stress-induced inflammation and fibroblast activation in the heart. The potential of developing innovative small molecule "epigenetic therapies" to combat cardiac fibrosis is highlighted.
Collapse
Key Words
- Ang II, angiotensin II
- BET, bromodomain and extraterminal protein
- DNMT, DNA methyltransferase
- ECM, extracellular matrix
- HAT, histone acetyltransferase
- HDAC, histone deacetylase
- IL, interleukin
- KDM, lysine demethylase
- KMT, lysine methyltransferase
- LPS, lipopolysaccharide
- MI, myocardial infarction
- NF-κB, nuclear factor-κB
- SASP, senescent-associated secretory phenotype
- SE, super-enhancer
- SMA, smooth muscle actin
- TET, ten-eleven translocation
- TNF, tumor necrosis factor
- TSA, trichostatin A
- Treg, regulatory T cell
- VPA, valproic acid
- epigenetics
- fibroblast
- fibrosis
- inflammation
Collapse
Affiliation(s)
- Marina B Felisbino
- Department of Medicine, Division of Cardiology and Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Timothy A McKinsey
- Department of Medicine, Division of Cardiology and Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| |
Collapse
|
239
|
Tao H, Song ZY, Ding XS, Yang JJ, Shi KH, Li J. LncRNAs and miRs as epigenetic signatures in diabetic cardiac fibrosis: new advances and perspectives. Endocrine 2018; 62:281-291. [PMID: 30054866 DOI: 10.1007/s12020-018-1688-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 07/16/2018] [Indexed: 12/13/2022]
Abstract
PURPOSE Diabetic cardiomyopathy (DCM) is a serious cardiac complication of diabetes, which further lead to heartfailure. It is known that diabetes-induced cardiac fibrosis is a key pathogenic factor contributing topathological changes in DCM. However, pathogenetic mechanisms underlying diabetes cardiac fibrosis arestill elusive. Recent studies have indicated that noncoding RNAs (ncRNAs) play a key role in diabetescardiac fibrosis. The increasing complexity of epigenetic regulator poses great challenges to ourconventional conceptions regarding how ncRNAs regulate diabetes cardiac fibrosis. METHODS We searched PubMed, Web of Science, and Scopus for manuscripts published prior to April 2018 using keywords "Diabetic cardiomyopathy" AND " diabetes cardiac fibrosis " OR " noncoding RNAs " OR " longnoncoding RNAs " OR " microRNAs " OR "epigenetic". Manuscripts were collated, studied and carriedforward for discussion where appropriate. RESULTS Based on the view that during diabetic cardiac fibrosis, ncRNAs are able to regulate diabetic cardiac fibrosisby targeting genes involved in epigenetic pathways. Many studies have focused on ncRNAs, an epigeneticregulator deregulating protein-coding genes in diabetic cardiac fibrosis, to identify potential therapeutictargets. Recent advances and new perspectives have found that long noncoding RNAs and microRNAs,exert their own effects on the progression of diabetic cardiac fibrosis. CONCLUSION We firstly examine the growing role of ncRNAs characteristics and ncRNAs-regulated genes involved indiabetic cardiac fibrosis. Then, we provide several possible therapeutic strategies and highlight the potentialof molecular mechanisms in which targeting epigenetic regulators are considered as an effective means of treating diabetic cardiac fibrosis.
Collapse
Affiliation(s)
- Hui Tao
- School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, 210009, Nanjing, China
- Department of Cardiothoracic Surgery, The Second Hospital of Anhui Medical University, 230601, Hefei, China
| | - Zheng-Yu Song
- School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, 210009, Nanjing, China
| | - Xuan-Sheng Ding
- School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, 210009, Nanjing, China.
| | - Jing-Jing Yang
- Department of Pharmacology, The Second Hospital of Anhui Medical University, 230601, Hefei, China
| | - Kai-Hu Shi
- Department of Cardiothoracic Surgery, The Second Hospital of Anhui Medical University, 230601, Hefei, China.
- Department of Cardiothoracic Surgery, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Jiangsu Province, 210028, Nanjing, China.
| | - Jun Li
- School of Pharmacy, Anhui Medical University, 230032, Hefei, China
| |
Collapse
|
240
|
Pan W, Zhu Y, Meng X, Zhang C, Yang Y, Bei Y. Immunomodulation by Exosomes in Myocardial Infarction. J Cardiovasc Transl Res 2018; 12:28-36. [PMID: 30374796 DOI: 10.1007/s12265-018-9836-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 09/14/2018] [Indexed: 02/06/2023]
Abstract
Exosomes are important carriers of biological information that facilitate intercellular communication and participate in the pathophysiology of different cardiovascular diseases. Myocardial infarction is among the leading causes of death worldwide. Upon myocardial infarction, massive cardiomyocyte death triggers a strong inflammatory response which is a vital process of cardiac injury, repair, and remodeling. Increasing evidence has unveiled that exosomes are involved in the inflammatory response and immune regulation after myocardial infarction. In this review, we will summarize the biological function of exosomes in the pathophysiology of myocardial infarction, especially focusing on their roles in the modulation of inflammation and immune response after myocardial infarction which further influences myocardial repair and remodeling. We will also discuss the immunomodulation by exosomes derived from stem and progenitor cells in the treatment of myocardial infarction. A deep understanding of immunomodulation by exosomes may represent a promising therapeutic option for the treatment of myocardial infarction.
Collapse
Affiliation(s)
- Wen Pan
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science, Shanghai University, 333 Nan Chen Road, Shanghai, 200444, China
| | - Yujiao Zhu
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science, Shanghai University, 333 Nan Chen Road, Shanghai, 200444, China.,Shanghai Key Laboratory of Bio-Energy Crops, School of Life Sciences, Shanghai University, Shanghai, 200444, China
| | - Xiangmin Meng
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science, Shanghai University, 333 Nan Chen Road, Shanghai, 200444, China
| | - Chenlin Zhang
- Department of Anesthesiology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200082, China
| | - Yan Yang
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, China.
| | - Yihua Bei
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science, Shanghai University, 333 Nan Chen Road, Shanghai, 200444, China.
| |
Collapse
|
241
|
Lubbers ER, Price MV, Mohler PJ. Arrhythmogenic Substrates for Atrial Fibrillation in Obesity. Front Physiol 2018; 9:1482. [PMID: 30405438 PMCID: PMC6204377 DOI: 10.3389/fphys.2018.01482] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2018] [Accepted: 10/01/2018] [Indexed: 12/19/2022] Open
Abstract
Global obesity rates have nearly tripled since 1975. This obesity rate increase is mirrored by increases in atrial fibrillation (AF) that now impacts nearly 10% of Americans over the age of 65. Numerous epidemiologic studies have linked incidence of AF and obesity and other obesity-related diseases, including hypertension and diabetes. Due to the wealth of epidemiologic data linking AF with obesity-related disease, mechanisms of AF pathogenesis in the context of obesity are an area of ongoing investigation. However, progress has been somewhat slowed by the complex phenotype of obesity; separating the effects of obesity from those of related sequelae is problematic. While the initiation of pathogenic pathways leading to AF varies with disease (including increased glycosylation in diabetes, increased renin angiotensin aldosterone system activation in hypertension, atrial ischemia in coronary artery disease, and sleep apnea) the pathogenesis of AF is united by shared mediators of altered conduction in the atria. We suggest focusing on these downstream mediators of AF in obesity is likely to yield more broadly applicable data. In the context of obesity, AF is driven by the interrelated processes of inflammation, atrial remodeling, and oxidative stress. Obesity is characterized by a constant low-grade inflammation that leads to increased expression of pro-inflammatory cytokines. These cytokines contribute to changes in cardiomyocyte excitability. Atrial structural remodeling, including fibrosis, enlargement, and fatty infiltration is a prominent feature of AF and contributes to the altered conduction. Finally, obesity impacts oxidative stress. Within the cardiomyocyte, oxidative stress is increased through both increased production of reactive oxygen species and by downregulation of scavenging enzymes. This increased oxidative stress modulates of cardiomyocyte excitability, increasing susceptibility to AF. Although the initiating insults vary, inflammation, atrial remodeling, and oxidative stress are conserved mechanisms in the pathophysiology of AF in the obese patients. In this review, we highlight mechanisms that have been shown to be relevant in the pathogenesis of AF across obesity-related disease.
Collapse
Affiliation(s)
- Ellen R. Lubbers
- The Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- Medical Scientist Training Program, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Morgan V. Price
- The Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Peter J. Mohler
- The Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- Department of Physiology & Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| |
Collapse
|
242
|
Peña C, Vargas R, Hernandez-Fonseca JP, Mosquera J. Cardiac myofibroblast induces decreased expression of major histocompatibility complex class II (Ia) on rat monocyte/macrophages. Tissue Cell 2018; 54:72-79. [PMID: 30309513 DOI: 10.1016/j.tice.2018.08.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 08/08/2018] [Accepted: 08/21/2018] [Indexed: 10/28/2022]
Abstract
The up-regulation of HLA antigens is important during heart inflammatory events and myofibroblasts may modulate the expression of this molecule in tissues. To test this possibility, the effect of cardiac myofibroblast:macrophage contact and the production of myofibroblast inhibitor factor(s) on the macrophage HLA (Ia) expression were studied. Listeria monocytogenes-elicited Ia + peritoneal macrophages (high Ia expression) were co-cultured with cardiac myofibroblasts for 3 and 7 days (myofibroblast contact). Proteosa peptone-elicited macrophages (low Ia expression) were cultured for 3 days with interferon gamma (INF-γ) and myofibroblast conditioned medium (FCM). Ia expression was analyzed by immunofluorescence and by radioimmune assay. Myofibroblast contact induced decreased expression of Ia molecule on macrophages (p < 0.001). This was confirmed by the radioimmune analysis in macrophage: myofibroblast co-cultures (p < 0.001). Double staining for Ia and CD14 showed that only CD14 positive cells (macrophages) expressed Ia molecule. FCM was capable of diminishing Ia expression induced by INF-γ on macrophages (p < 0.001). Decreased Ia macrophage expression induced by myofibroblasts could be important in the heart inflammation's resolution, probably involving Ia redistribution on cell: cell contact and myofibroblast inhibitor factor production.
Collapse
Affiliation(s)
- Caterina Peña
- Cátedra de Genética, Escuela de Bioanálisis, Facultad de Medicina, Universidad del Zulia, Maracaibo, Venezuela.
| | - Renata Vargas
- Instituto de Investigaciones Clínicas "Dr. Américo Negrette", Facultad de Medicina, Universidad del Zulia, Maracaibo, Venezuela.
| | - Juan Pablo Hernandez-Fonseca
- Instituto de Investigaciones Clínicas "Dr. Américo Negrette", Facultad de Medicina, Universidad del Zulia, Maracaibo, Venezuela.
| | - Jesús Mosquera
- Instituto de Investigaciones Clínicas "Dr. Américo Negrette", Facultad de Medicina, Universidad del Zulia, Maracaibo, Venezuela.
| |
Collapse
|
243
|
Dubnika A, Manoukian MA, Mohammadi MR, Parekh MB, Gurjarpadhye AA, Inayathullah M, Dubniks V, Lakey JR, Rajadas J. Cytokines as therapeutic agents and targets in heart disease. Cytokine Growth Factor Rev 2018; 43:54-68. [DOI: 10.1016/j.cytogfr.2018.08.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 08/01/2018] [Accepted: 08/13/2018] [Indexed: 02/02/2023]
|
244
|
Chrysin attenuates interstitial fibrosis and improves cardiac function in a rat model of acute myocardial infarction. J Mol Histol 2018; 49:555-565. [DOI: 10.1007/s10735-018-9793-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 08/23/2018] [Indexed: 12/19/2022]
|
245
|
Dong X, Yang H, Li C, Liu Q, Bai Q, Zhang Z. Triiodothyronine alleviates alcoholic liver disease injury through the negative regulation of the NLRP3 signaling pathway. Exp Ther Med 2018; 16:1866-1872. [PMID: 30186412 PMCID: PMC6122124 DOI: 10.3892/etm.2018.6409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 05/02/2018] [Indexed: 11/06/2022] Open
Abstract
The aim of the present study was to investigate the effect and mechanism of triiodothyronine (T3) on alcoholic liver disease (ALD)-induced injuries in mice. A total of 40 male C57/BL6 mice were randomly divided into the Control, ALD, ALD+T3 and ALD+T3+AMP-activated protein kinase inhibitor (CC) groups. Mice were administered alcohol (4 g/kg/day) intragastrically for 4 weeks except for Control group. Mice in the ALD+T3 group were given T3 (0.1 mg/kg/day) while mice in ALD+T3+CC group were given T3 (0.1 mg/kg/day) and CC (10 mg/kg/day) for 1 week. Control and ALD groups were treated with saline. Liver tissue and blood samples were obtained for testing. Alanine aminotransferase (ALT), aspartate aminotransferase (AST) and total bilirubin (TBIL) levels were determined and morphological changes in the liver tissues were observed under the optical microscope. Inflammatory factors, including IL-1β and transforming growth factor (TGF)-β/1, α-smooth muscle actin (SMA) and protein levels of nucleotide-binding oligomerization domain, leucine rich repeat containing family, pyrin domain containing 3 (NLRP3), caspase-1 and pro-IL-1β were measured. Serum ALT, AST and TBIL levels in the ALD+T3 group were significantly reduced compared with the ALD group, while they were significantly increased in the ALD+T3+CC group (P<0.05). The number of hepatic lobules in the ALD+T3 group was significantly reduced compared with the ALD group, whereas the number in the ALD+T3+CC group was significantly increased (P<0.05). IL-1β and TGF-β1 levels in the ALD+T3 group were significantly decreased compared with the ALD group; however, levels in the ALD+T3+CC group were significantly increased compared with the ALD+T3 group (P<0.05). In addition, it was revealed that the expression of α-SMA mRNA and protein in the ALD+T3 group was significantly decreased compared with the ALD group, whereas it was significantly increased in the ALD+T3+CC group compared with the ALD+T3 group. Expression of NLRP3, caspase-1, IL-1β and TGF-β1 in the ALD+T3 group was significantly decreased compared with the ALD group, while expression was significantly increased in the ALD+T3+CC group. Conversely, compared with the ALD group, expression of pro-IL-1β was significantly increased in the ALD+T3 group and decreased in the ALD+T3+CC group. In conclusion, T3 may reduce the inflammatory response and severity of liver cirrhosis in mice with ALD by negatively regulating the NLRP3 signaling pathway.
Collapse
Affiliation(s)
- Xiang Dong
- Department of Gastroenterology, Jining First People's Hospital, Jining, Shandong 272000, P.R. China
| | - Hongmei Yang
- Laboratory Medicine, Yantaishan Hospital, Yantai, Shandong 264000, P.R. China
| | - Cong Li
- Department of Otolaryngology, People's Hospital of Zhangqiu District, Jinan, Shandong 250200, P.R. China
| | - Qi Liu
- Department of Endocrinology, People's Hospital of Zhangqiu District, Jinan, Shandong 250200, P.R. China
| | - Qinglin Bai
- Department of Operation Room, People's Hospital of Zhangqiu District, Jinan, Shandong 250200, P.R. China
| | - Zhaoran Zhang
- Department of Gastroenterology, Jining First People's Hospital, Jining, Shandong 272000, P.R. China
| |
Collapse
|
246
|
Spinale FG, Frangogiannis NG, Hinz B, Holmes JW, Kassiri Z, Lindsey ML. Crossing Into the Next Frontier of Cardiac Extracellular Matrix Research. Circ Res 2018; 119:1040-1045. [PMID: 27789578 DOI: 10.1161/circresaha.116.309916] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Affiliation(s)
- Francis G Spinale
- From the University of South Carolina College of Engineering and Computing, Columbia (F.G.S.); Cardiovascular Translational Research Center (F.G.S.) and Department of Cell Biology and Anatomy (F.G.S.), University of South Carolina School of Medicine, Columbia; WJB Dorn Veteran Affairs Medical Center, Columbia, SC (F.G.S.); Department of Medicine (Cardiology), The Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY (N.G.F.); Laboratory of Tissue Repair and Regeneration, Matrix Dynamics Group, Faculty of Dentistry, University of Toronto, ON, Canada (B.H.); Departments of Biomedical Engineering (J.W.H.) and Medicine (J.W.H.), Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville; Department of Physiology, Cardiovascular Research Centre, Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada (Z.K.); Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson (M.L.L.); and Research Service, G.V. (Sonny) Montgomery Veterans Affairs Medical Center, Jackson, MS (M.L.L.)
| | - Nikolaos G Frangogiannis
- From the University of South Carolina College of Engineering and Computing, Columbia (F.G.S.); Cardiovascular Translational Research Center (F.G.S.) and Department of Cell Biology and Anatomy (F.G.S.), University of South Carolina School of Medicine, Columbia; WJB Dorn Veteran Affairs Medical Center, Columbia, SC (F.G.S.); Department of Medicine (Cardiology), The Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY (N.G.F.); Laboratory of Tissue Repair and Regeneration, Matrix Dynamics Group, Faculty of Dentistry, University of Toronto, ON, Canada (B.H.); Departments of Biomedical Engineering (J.W.H.) and Medicine (J.W.H.), Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville; Department of Physiology, Cardiovascular Research Centre, Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada (Z.K.); Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson (M.L.L.); and Research Service, G.V. (Sonny) Montgomery Veterans Affairs Medical Center, Jackson, MS (M.L.L.)
| | - Boris Hinz
- From the University of South Carolina College of Engineering and Computing, Columbia (F.G.S.); Cardiovascular Translational Research Center (F.G.S.) and Department of Cell Biology and Anatomy (F.G.S.), University of South Carolina School of Medicine, Columbia; WJB Dorn Veteran Affairs Medical Center, Columbia, SC (F.G.S.); Department of Medicine (Cardiology), The Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY (N.G.F.); Laboratory of Tissue Repair and Regeneration, Matrix Dynamics Group, Faculty of Dentistry, University of Toronto, ON, Canada (B.H.); Departments of Biomedical Engineering (J.W.H.) and Medicine (J.W.H.), Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville; Department of Physiology, Cardiovascular Research Centre, Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada (Z.K.); Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson (M.L.L.); and Research Service, G.V. (Sonny) Montgomery Veterans Affairs Medical Center, Jackson, MS (M.L.L.)
| | - Jeffrey W Holmes
- From the University of South Carolina College of Engineering and Computing, Columbia (F.G.S.); Cardiovascular Translational Research Center (F.G.S.) and Department of Cell Biology and Anatomy (F.G.S.), University of South Carolina School of Medicine, Columbia; WJB Dorn Veteran Affairs Medical Center, Columbia, SC (F.G.S.); Department of Medicine (Cardiology), The Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY (N.G.F.); Laboratory of Tissue Repair and Regeneration, Matrix Dynamics Group, Faculty of Dentistry, University of Toronto, ON, Canada (B.H.); Departments of Biomedical Engineering (J.W.H.) and Medicine (J.W.H.), Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville; Department of Physiology, Cardiovascular Research Centre, Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada (Z.K.); Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson (M.L.L.); and Research Service, G.V. (Sonny) Montgomery Veterans Affairs Medical Center, Jackson, MS (M.L.L.)
| | - Zamaneh Kassiri
- From the University of South Carolina College of Engineering and Computing, Columbia (F.G.S.); Cardiovascular Translational Research Center (F.G.S.) and Department of Cell Biology and Anatomy (F.G.S.), University of South Carolina School of Medicine, Columbia; WJB Dorn Veteran Affairs Medical Center, Columbia, SC (F.G.S.); Department of Medicine (Cardiology), The Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY (N.G.F.); Laboratory of Tissue Repair and Regeneration, Matrix Dynamics Group, Faculty of Dentistry, University of Toronto, ON, Canada (B.H.); Departments of Biomedical Engineering (J.W.H.) and Medicine (J.W.H.), Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville; Department of Physiology, Cardiovascular Research Centre, Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada (Z.K.); Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson (M.L.L.); and Research Service, G.V. (Sonny) Montgomery Veterans Affairs Medical Center, Jackson, MS (M.L.L.)
| | - Merry L Lindsey
- From the University of South Carolina College of Engineering and Computing, Columbia (F.G.S.); Cardiovascular Translational Research Center (F.G.S.) and Department of Cell Biology and Anatomy (F.G.S.), University of South Carolina School of Medicine, Columbia; WJB Dorn Veteran Affairs Medical Center, Columbia, SC (F.G.S.); Department of Medicine (Cardiology), The Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY (N.G.F.); Laboratory of Tissue Repair and Regeneration, Matrix Dynamics Group, Faculty of Dentistry, University of Toronto, ON, Canada (B.H.); Departments of Biomedical Engineering (J.W.H.) and Medicine (J.W.H.), Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville; Department of Physiology, Cardiovascular Research Centre, Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada (Z.K.); Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson (M.L.L.); and Research Service, G.V. (Sonny) Montgomery Veterans Affairs Medical Center, Jackson, MS (M.L.L.).
| |
Collapse
|
247
|
Affiliation(s)
- Nikolaos G Frangogiannis
- From the Department of Medicine (Cardiology), The Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY.
| |
Collapse
|
248
|
Lefort C, Benoist L, Chadet S, Piollet M, Heraud A, Babuty D, Baron C, Ivanes F, Angoulvant D. Stimulation of P2Y11 receptor modulates cardiac fibroblasts secretome toward immunomodulatory and protective roles after Hypoxia/Reoxygenation injury. J Mol Cell Cardiol 2018; 121:212-222. [PMID: 30031814 DOI: 10.1016/j.yjmcc.2018.07.245] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 07/10/2018] [Accepted: 07/19/2018] [Indexed: 12/21/2022]
Abstract
Cardiac fibroblasts are important regulators of myocardial structure and function. Their implications in pathological processes such as Ischemia/Reperfusion are well characterized. Cardiac fibroblasts respond to stress by excessive proliferation and secretion of pro-inflammatory cytokines and other factors, e.g. ATP, leading to purinergic receptors activation. P2Y11 receptor (P2Y11R) is an ATP-sensitive GPCR playing an immunomodulatory role in human dendritic cells (DC). We hypothesized that P2Y11R stimulation modulated the pro-inflammatory responses of human cardiac fibroblasts (HCF) to Hypoxia/Reoxygenation (H/R) mainly by acting on their secretome. P2Y11R stimulation in HCF at the onset of reoxygenation significantly limited H/R-induced proliferation (-19%) and pro-inflammatory cytokines and ATP secretion (-44% and -83% respectively). Exposure of DC to HCF secretome increased their expression of CD83, CD25 and CD86, suggesting a switch from immature to mature phenotype. Under LPS stimulation, DC had a pro-inflammatory profile (high IL-12/IL-10 ratio) that was decreased by HCF secretome (-3,8-fold), indicating induction of a tolerogenic profile. Moreover, P2Y11R inhibition in HCF led to high IL-12 secretion in DC, suggesting that the immunomodulatory effect of HCF secretome is P2Y11R-dependant. HCF secretome reduced H/R-induced cardiomyocytes death (-23%) through RISK pathway activation. P2Y11R inhibition in HCF induced a complete loss of HCF secretome protective effect, highlighting the cardioprotective role of P2Y11R. Our data demonstrated paracrine interactions between HCF, cardiomyocytes and DC following H/R, suggesting a key role of HCF in the cellular responses to reperfusion. These results also demonstrated a beneficial role of P2Y11R in HCF during H/R and strongly support the hypothesis that P2Y11R is a modulator of I/R injury.
Collapse
Affiliation(s)
- Claudie Lefort
- EA 4245 "Transplantation, Immunologie et Inflammation", Loire Valley Cardiovascular Collaboration & Université de Tours, 10 Boulevard Tonnellé, 37032 Tours, France
| | - Lauriane Benoist
- EA 4245 "Transplantation, Immunologie et Inflammation", Loire Valley Cardiovascular Collaboration & Université de Tours, 10 Boulevard Tonnellé, 37032 Tours, France
| | - Stéphanie Chadet
- EA 4245 "Transplantation, Immunologie et Inflammation", Loire Valley Cardiovascular Collaboration & Université de Tours, 10 Boulevard Tonnellé, 37032 Tours, France
| | - Marie Piollet
- EA 4245 "Transplantation, Immunologie et Inflammation", Loire Valley Cardiovascular Collaboration & Université de Tours, 10 Boulevard Tonnellé, 37032 Tours, France
| | - Audrey Heraud
- EA 4245 "Transplantation, Immunologie et Inflammation", Loire Valley Cardiovascular Collaboration & Université de Tours, 10 Boulevard Tonnellé, 37032 Tours, France
| | - Dominique Babuty
- EA 4245 "Transplantation, Immunologie et Inflammation", Loire Valley Cardiovascular Collaboration & Université de Tours, 10 Boulevard Tonnellé, 37032 Tours, France; Service de Cardiologie, Centre Hospitalier Régional Universitaire de Tours, 37044 Tours, France
| | - Christophe Baron
- EA 4245 "Transplantation, Immunologie et Inflammation", Loire Valley Cardiovascular Collaboration & Université de Tours, 10 Boulevard Tonnellé, 37032 Tours, France; Service de Néphrologie et d'Immunologie Clinique, Centre Hospitalier Régional Universitaire de Tours, 37044 Tours, France
| | - Fabrice Ivanes
- EA 4245 "Transplantation, Immunologie et Inflammation", Loire Valley Cardiovascular Collaboration & Université de Tours, 10 Boulevard Tonnellé, 37032 Tours, France; Service de Cardiologie, Centre Hospitalier Régional Universitaire de Tours, 37044 Tours, France.
| | - Denis Angoulvant
- EA 4245 "Transplantation, Immunologie et Inflammation", Loire Valley Cardiovascular Collaboration & Université de Tours, 10 Boulevard Tonnellé, 37032 Tours, France; Service de Cardiologie, Centre Hospitalier Régional Universitaire de Tours, 37044 Tours, France
| |
Collapse
|
249
|
MicroRNA-21 prevents excessive inflammation and cardiac dysfunction after myocardial infarction through targeting KBTBD7. Cell Death Dis 2018; 9:769. [PMID: 29991775 PMCID: PMC6039462 DOI: 10.1038/s41419-018-0805-5] [Citation(s) in RCA: 115] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 05/28/2018] [Accepted: 05/29/2018] [Indexed: 11/18/2022]
Abstract
The excessive inflammation triggered by damage-associated molecular patterns (DAMPs) after myocardial infarction (MI) is responsible for the development of cardiac dysfunction and adverse remodeling, while the mechanisms by which inflammation is fine tuned remain to be fully elucidated. MicroRNA-21 (miR-21) has been shown to function in cardiovascular diseases, while its role in inflammatory responses and cardiac function post MI in mice remains unknown. Here, we found that miR-21 expression was markedly increased in border and infarct areas of cardiac tissues during the early inflammatory phase of MI model established by ligating the left-anterior descending coronary artery. MiR-21 knockout mice had decreased survival rates, worse cardiac dysfunction, and increased infarct and scar areas after MI compared with WT mice. MiR-21 knockout mice showed significantly higher levels of inflammatory cytokines including IL-1β, IL-6, and TNF-α in cardiac tissues, as well as infiltration of CD11b+ monocytes/macrophages with higher expression level of inflammatory cytokines. MI induced the great release of high mobility group protein B1 (HMGB1) and heat shock protein 60 (HSP60) in cardiac tissue. MiR-21 deficiency significantly promoted the inflammatory cytokine production triggered by DAMPs in macrophages, whereas, miR-21 overexpression markedly inhibited the inflammatory cytokine production. Mechanistically, miR-21 deficiency enhanced p38 and NF-κB signaling activation in cardiac tissue post MI and macrophages treated with DAMPs. MiR-21 was found to directly target kelch repeat and BTB (POZ) domain containing 7 (KBTBD7), which promoted DAMP-triggered inflammatory responses in macrophages. Furthermore, KBTBD7 interacted with MKK3/6 and promoted their activation, which in turn enhanced the activation of downstream p38 and NF-κB signaling induced by DAMPs. Therefore, our findings demonstrate that miR-21 attenuates inflammation, cardiac dysfunction, and maladaptive remodeling post MI through targeting KBTBD7 and inhibiting p38 and NF-κB signaling activation, suggesting that miR-21 may function as a novel potential therapeutic target for MI.
Collapse
|
250
|
Trial J, Cieslik KA. Changes in cardiac resident fibroblast physiology and phenotype in aging. Am J Physiol Heart Circ Physiol 2018; 315:H745-H755. [PMID: 29906228 DOI: 10.1152/ajpheart.00237.2018] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
The cardiac fibroblast plays a central role in tissue homeostasis and in repair after injury. With aging, dysregulated cardiac fibroblasts have a reduced capacity to activate a canonical transforming growth factor-β-Smad pathway and differentiate poorly into contractile myofibroblasts. That results in the formation of an insufficient scar after myocardial infarction. In contrast, in the uninjured aged heart, fibroblasts are activated and acquire a profibrotic phenotype that leads to interstitial fibrosis, ventricular stiffness, and diastolic dysfunction, all conditions that may lead to heart failure. There is an apparent paradox in aging, wherein reparative fibrosis is impaired but interstitial, adverse fibrosis is augmented. This could be explained by analyzing the effectiveness of signaling pathways in resident fibroblasts from young versus aged hearts. Whereas defective signaling by transforming growth factor-β leads to insufficient scar formation by myofibroblasts, enhanced activation of the ERK1/2 pathway may be responsible for interstitial fibrosis mediated by activated fibroblasts. Listen to this article's corresponding podcast at https://ajpheart.podbean.com/e/fibroblast-phenotypic-changes-in-the-aging-heart/ .
Collapse
Affiliation(s)
- JoAnn Trial
- Division of Cardiovascular Sciences, Department of Medicine, Baylor College of Medicine , Houston, Texas
| | - Katarzyna A Cieslik
- Division of Cardiovascular Sciences, Department of Medicine, Baylor College of Medicine , Houston, Texas
| |
Collapse
|