951
|
Chacar S, Farès N, Bois P, Faivre JF. Basic Signaling in Cardiac Fibroblasts. J Cell Physiol 2016; 232:725-730. [DOI: 10.1002/jcp.25624] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 09/28/2016] [Indexed: 12/14/2022]
Affiliation(s)
- Stéphanie Chacar
- Laboratoire Signalisation et Transports Ioniques Membranaires (STIM); Université de Poitiers; CNRS; Poitiers France
- Laboratoire de recherche en Physiologie et Physiopathologie (LRPP); pôle technologie santé; Faculté de Médecine; Université Saint Joseph; Beyrouth Liban
| | - Nassim Farès
- Laboratoire de recherche en Physiologie et Physiopathologie (LRPP); pôle technologie santé; Faculté de Médecine; Université Saint Joseph; Beyrouth Liban
| | - Patrick Bois
- Laboratoire Signalisation et Transports Ioniques Membranaires (STIM); Université de Poitiers; CNRS; Poitiers France
| | - Jean-François Faivre
- Laboratoire Signalisation et Transports Ioniques Membranaires (STIM); Université de Poitiers; CNRS; Poitiers France
| |
Collapse
|
952
|
Affiliation(s)
- Rohan Dharmakumar
- From the Biomedical Imaging Research Institute and Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA; and Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles
| |
Collapse
|
953
|
Rubin N, Harrison MR, Krainock M, Kim R, Lien CL. Recent advancements in understanding endogenous heart regeneration-insights from adult zebrafish and neonatal mice. Semin Cell Dev Biol 2016; 58:34-40. [PMID: 27132022 PMCID: PMC5028242 DOI: 10.1016/j.semcdb.2016.04.011] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Revised: 02/13/2016] [Accepted: 04/17/2016] [Indexed: 02/06/2023]
Abstract
Enhancing the endogenous regenerative capacity of the mammalian heart is a promising strategy that can lead to potential treatment of injured cardiac tissues. Studies on heart regeneration in zebrafish and neonatal mice have shown that cardiomyocyte proliferation is essential for replenishing myocardium. We will review recent advancements that have demonstrated the importance of Neuregulin 1/ErbB2 and innervation in regulating cardiomyocyte proliferation using both adult zebrafish and neonatal mouse heart regeneration models. Emerging findings suggest that different populations of macrophages and inflammation might contribute to regenerative versus fibrotic responses. Finally, we will discuss variation in the severity of the cardiac injury and size of the wound, which may explain the range of outcomes observed in different injury models.
Collapse
Affiliation(s)
- Nicole Rubin
- Heart Institute and Program of Developmental Biology and Regenerative Medicine, The Saban Research Institute of Children's Hospital Los Angeles, United States; Division of Cardiothoracic Surgery, Department of Surgery, Keck School of Medicine, University of Southern California, United States
| | - Michael R Harrison
- Heart Institute and Program of Developmental Biology and Regenerative Medicine, The Saban Research Institute of Children's Hospital Los Angeles, United States; Division of Cardiothoracic Surgery, Department of Surgery, Keck School of Medicine, University of Southern California, United States
| | - Michael Krainock
- Heart Institute and Program of Developmental Biology and Regenerative Medicine, The Saban Research Institute of Children's Hospital Los Angeles, United States; Division of Cardiothoracic Surgery, Department of Surgery, Keck School of Medicine, University of Southern California, United States
| | - Richard Kim
- Heart Institute and Program of Developmental Biology and Regenerative Medicine, The Saban Research Institute of Children's Hospital Los Angeles, United States; Division of Cardiothoracic Surgery, Department of Surgery, Keck School of Medicine, University of Southern California, United States
| | - Ching-Ling Lien
- Heart Institute and Program of Developmental Biology and Regenerative Medicine, The Saban Research Institute of Children's Hospital Los Angeles, United States; Division of Cardiothoracic Surgery, Department of Surgery, Keck School of Medicine, University of Southern California, United States; Department of Biochemistry & Molecular Biology, Keck School of Medicine, University of Southern California, United States.
| |
Collapse
|
954
|
Jones DA, Khambata RS, Andiapen M, Rathod KS, Mathur A, Ahluwalia A. Intracoronary nitrite suppresses the inflammatory response following primary percutaneous coronary intervention. Heart 2016; 103:508-516. [DOI: 10.1136/heartjnl-2016-309748] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 08/27/2016] [Accepted: 08/31/2016] [Indexed: 02/06/2023] Open
|
955
|
Sun Z, Zhou D, Xie X, Wang S, Wang Z, Zhao W, Xu H, Zheng L. Cross-talk between macrophages and atrial myocytes in atrial fibrillation. Basic Res Cardiol 2016; 111:63. [PMID: 27660282 PMCID: PMC5033992 DOI: 10.1007/s00395-016-0584-z] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 09/13/2016] [Indexed: 12/26/2022]
Abstract
Increased macrophage accumulation occurs in the atria of patients with atrial fibrillation (AF). However, the phenotype and functions of the macrophages in AF remain unclear. We investigated the macrophage-atrial myocyte interaction in AF patients and found that the increased macrophages were mainly pro-inflammatory macrophages (iNOS+, Arg1−). Tachypacing of HL-1 atrial myocytes also led to pro-inflammatory macrophage polarization. In addition, lipopolysaccharide (LPS)-stimulated pro-inflammatory macrophages-induced atrial electrical remodeling, evidenced by increased AF incidence and decreased atrial effective refractory period and L-type calcium currents (ICa-L) in both canine and mouse AF models. Depletion of macrophages relieved LPS-induced atrial electrical remodeling, confirming the role of pro-inflammatory macrophages in the pathogenesis of AF. We also found that the effect of LPS-stimulated macrophages on atrial myocytes was mediated by secretion of interleukin 1 beta (IL-1β), which inhibited atrial myocyte quaking protein (QKI) expression. IL-1β knockout in macrophages restored the LPS-stimulated macrophage-induced inhibition of QKI and CACNA1C (α1C subunit of L-type calcium channel) in atrial myocytes. Meanwhile, QKI overexpression in atrial myocytes restored the LPS-stimulated macrophage-induced electrical remodeling through enhanced binding of QKI to CACNA1C mRNA, which upregulated the expression of CACNA1C as well as ICa-L. In contrast, QKI knockout inhibited CACNA1C expression. Finally, using transcription factor activation profiling plate array and chromatin immunoprecipitation, we revealed that special AT-rich sequence binding protein 1 activated QKI transcription. Taken together, our study uncovered the functional interaction between macrophages and atrial myocytes in AF. AF induced pro-inflammatory macrophage polarization while pro-inflammatory macrophages exacerbated atrial electrical remodeling by secreting IL-1β, further inhibiting QKI expression in atrial myocytes, which contributed to ICa-L downregulation. Our study demonstrates a novel molecular mechanism underlying the pathogenesis and progression of AF and suggests that QKI is a potential therapeutic target.
Collapse
Affiliation(s)
- Zewei Sun
- Department of Cardiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, No. 79 Qingchun Road, Hangzhou, 310003, China
| | - Dongchen Zhou
- Department of Cardiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, No. 79 Qingchun Road, Hangzhou, 310003, China
| | - Xudong Xie
- Department of Cardiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, No. 79 Qingchun Road, Hangzhou, 310003, China
| | - Shuai Wang
- Department of Cardiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, No. 79 Qingchun Road, Hangzhou, 310003, China
| | - Zhen Wang
- Department of Cardiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, No. 79 Qingchun Road, Hangzhou, 310003, China
| | - Wenting Zhao
- Department of Cardiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, No. 79 Qingchun Road, Hangzhou, 310003, China
| | - Hongfei Xu
- Department of Cardiothoracic Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, No.79 Qingchun Road, Hangzhou, 310003, China
| | - Liangrong Zheng
- Department of Cardiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, No. 79 Qingchun Road, Hangzhou, 310003, China.
| |
Collapse
|
956
|
Woodall MC, Woodall BP, Gao E, Yuan A, Koch WJ. Cardiac Fibroblast GRK2 Deletion Enhances Contractility and Remodeling Following Ischemia/Reperfusion Injury. Circ Res 2016; 119:1116-1127. [PMID: 27601479 DOI: 10.1161/circresaha.116.309538] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Accepted: 09/06/2016] [Indexed: 12/30/2022]
Abstract
RATIONALE G protein-coupled receptor kinase 2 (GRK2) is an important molecule upregulated after myocardial injury and during heart failure. Myocyte-specific GRK2 loss before and after myocardial ischemic injury improves cardiac function and remodeling. The cardiac fibroblast plays an important role in the repair and remodeling events after cardiac ischemia; the importance of GRK2 in these events has not been investigated. OBJECTIVE The aim of this study is to elucidate the in vivo implications of deleting GRK2 in the cardiac fibroblast after ischemia/reperfusion injury. METHODS AND RESULTS We demonstrate, using Tamoxifen inducible, fibroblast-specific GRK2 knockout mice, that GRK2 loss confers a protective advantage over control mice after myocardial ischemia/reperfusion injury. Fibroblast GRK2 knockout mice presented with decreased infarct size and preserved cardiac function 24 hours post ischemia/reperfusion as demonstrated by increased ejection fraction (59.1±1.8% versus 48.7±1.2% in controls; P<0.01). GRK2 fibroblast knockout mice also had decreased fibrosis and fibrotic gene expression. Importantly, these protective effects correlated with decreased infiltration of neutrophils to the ischemia site and decreased levels of tumor necrosis factor-α expression and secretion in GRK2 fibroblast knockout mice. CONCLUSIONS These novel data showing the benefits of inhibiting GRK2 in the cardiac fibroblast adds to previously published data showing the advantage of GRK2 ablation and reinforces the therapeutic potential of GRK2 inhibition in the heart after myocardial ischemia.
Collapse
Affiliation(s)
- Meryl C Woodall
- From the Department of Pharmacology, Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA (M.C.W., B.P.W., E.G., A.Y., W.J.K.); and Department of Cardiology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, China (A.Y.)
| | - Benjamin P Woodall
- From the Department of Pharmacology, Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA (M.C.W., B.P.W., E.G., A.Y., W.J.K.); and Department of Cardiology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, China (A.Y.)
| | - Erhe Gao
- From the Department of Pharmacology, Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA (M.C.W., B.P.W., E.G., A.Y., W.J.K.); and Department of Cardiology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, China (A.Y.)
| | - Ancai Yuan
- From the Department of Pharmacology, Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA (M.C.W., B.P.W., E.G., A.Y., W.J.K.); and Department of Cardiology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, China (A.Y.)
| | - Walter J Koch
- From the Department of Pharmacology, Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA (M.C.W., B.P.W., E.G., A.Y., W.J.K.); and Department of Cardiology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, China (A.Y.).
| |
Collapse
|
957
|
Huang Y, Wang D, Wang X, Zhang Y, Liu T, Chen Y, Tang Y, Wang T, Hu D, Huang C. Abrogation of CC chemokine receptor 9 ameliorates ventricular remodeling in mice after myocardial infarction. Sci Rep 2016; 6:32660. [PMID: 27585634 PMCID: PMC5009347 DOI: 10.1038/srep32660] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Accepted: 08/11/2016] [Indexed: 12/14/2022] Open
Abstract
CC chemokine receptor 9 (CCR9), which is a unique receptor for CC chemokine ligand (CCL25), is mainly expressed on lymphocytes, dendritic cells (DCs) and monocytes/macrophages. CCR9 mediates the chemotaxis of inflammatory cells and participates in the pathological progression of inflammatory diseases. However, the role of CCR9 in the pathological process of myocardial infarction (MI) remains unexplored; inflammation plays a key role in this process. Here, we used CCR9 knockout mice to determine the functional significance of CCR9 in regulating post-MI cardiac remodeling and its underlying mechanism. MI was induced by surgical ligation of the left anterior descending coronary artery in CCR9 knockout mice and their CCR9+/+ littermates. Our results showed that the CCR9 expression levels were up-regulated in the hearts of the MI mice. Abrogation of CCR9 improved the post-MI survival rate and left ventricular (LV) dysfunction and decreased the infarct size. In addition, the CCR9 knockout mice exhibited attenuated inflammation, apoptosis, structural and electrical remodeling compared with the CCR9+/+ MI mice. Mechanistically, CCR9 mainly regulated the pathological response by interfering with the NF-κB and MAPK signaling pathways. In conclusion, the data reveal that CCR9 serves as a novel modulator of pathological progression following MI through NF-κB and MAPK signaling.
Collapse
Affiliation(s)
- Yan Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China.,Cardiovascular Research Institute, Wuhan University, Wuhan 430060, PR China.,Hubei Key Laboratory of Cardiology, Wuhan 430060, PR China
| | - Dandan Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China.,Cardiovascular Research Institute, Wuhan University, Wuhan 430060, PR China.,Hubei Key Laboratory of Cardiology, Wuhan 430060, PR China
| | - Xin Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China
| | - Yijie Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China.,Cardiovascular Research Institute, Wuhan University, Wuhan 430060, PR China.,Hubei Key Laboratory of Cardiology, Wuhan 430060, PR China
| | - Tao Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China.,Cardiovascular Research Institute, Wuhan University, Wuhan 430060, PR China.,Hubei Key Laboratory of Cardiology, Wuhan 430060, PR China
| | - Yuting Chen
- Cardiovascular Research Institute, Wuhan University, Wuhan 430060, PR China.,Hubei Key Laboratory of Cardiology, Wuhan 430060, PR China
| | - Yanhong Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China.,Cardiovascular Research Institute, Wuhan University, Wuhan 430060, PR China.,Hubei Key Laboratory of Cardiology, Wuhan 430060, PR China
| | - Teng Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China.,Cardiovascular Research Institute, Wuhan University, Wuhan 430060, PR China.,Hubei Key Laboratory of Cardiology, Wuhan 430060, PR China
| | - Dan Hu
- Cardiovascular Research Institute, Wuhan University, Wuhan 430060, PR China.,Hubei Key Laboratory of Cardiology, Wuhan 430060, PR China.,Masonic Medical Research Laboratory, Utica, NY, USA
| | - Congxin Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China.,Cardiovascular Research Institute, Wuhan University, Wuhan 430060, PR China.,Hubei Key Laboratory of Cardiology, Wuhan 430060, PR China
| |
Collapse
|
958
|
Yuan MJ, Kong B, Wang T, Wang X, Huang H, Maghsoudi T. RETRACTED: Ghrelin protects infarcted myocardium by induction of autophagy and AMP-activated protein kinase pathway. Biochem Biophys Res Commun 2016; 476:365-370. [PMID: 27235554 DOI: 10.1016/j.bbrc.2016.05.128] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 05/25/2016] [Indexed: 11/16/2022]
Abstract
This article has been retracted: please see Elsevier Policy on Article Withdrawal (http://www.elsevier.com/locate/withdrawalpolicy). This article has been retracted at the request of the Editor-in-Chief. It contains LM images in figure 2E, which were originally published in Nat. Comm. 6/2015, p. 1-14 (http://doi:10.1038/ncomms8388). Permission to reproduce this figure was not requested nor granted by the authors of the original article for its re-use. The authors of this article would like to apologize to all affected parties.
Collapse
Affiliation(s)
- Ming-Jie Yuan
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute of Wuhan University, Hubei Key Laboratory of Cardiology, Jiefang Road 238, Wuchang, 430060, Wuhan, PR China.
| | - Bin Kong
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute of Wuhan University, Hubei Key Laboratory of Cardiology, Jiefang Road 238, Wuchang, 430060, Wuhan, PR China
| | - Tao Wang
- Cardiovascular Research Center, University of Virginia, MR5 Building, Room 1332, 415 Lane RD, Charlottesville, VA 22908, United States
| | - Xin Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute of Wuhan University, Hubei Key Laboratory of Cardiology, Jiefang Road 238, Wuchang, 430060, Wuhan, PR China
| | - He Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute of Wuhan University, Hubei Key Laboratory of Cardiology, Jiefang Road 238, Wuchang, 430060, Wuhan, PR China
| | - Taneen Maghsoudi
- Cardiovascular Research Center, University of Virginia, MR5 Building, Room 1332, 415 Lane RD, Charlottesville, VA 22908, United States
| |
Collapse
|
959
|
Széplaki G, Boros AM, Szilágyi S, Osztheimer I, Jenei Z, Kosztin A, Nagy KV, Karády J, Molnár L, Tahin T, Zima E, Gellér L, Prohászka Z, Merkely B. Complement C3a predicts outcome in cardiac resynchronization therapy of heart failure. Inflamm Res 2016; 65:933-940. [DOI: 10.1007/s00011-016-0976-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Revised: 06/23/2016] [Accepted: 07/19/2016] [Indexed: 12/19/2022] Open
|
960
|
Frunza O, Russo I, Shinde AV, Su Y, Frangogiannis NG. Authors' Reply. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:2234-2235. [PMID: 27461365 DOI: 10.1016/j.ajpath.2016.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Revised: 06/01/2016] [Accepted: 06/01/2016] [Indexed: 11/15/2022]
Abstract
This correspondence is a reply to Galectin-3, Cardiac Function, and Fibrosis by Wouter C. Meijers et al.
Collapse
Affiliation(s)
- Olga Frunza
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx, New York
| | - Ilaria Russo
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx, New York
| | - Arti V Shinde
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx, New York
| | - Ya Su
- 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
|
961
|
Rischpler C, Dirschinger RJ, Nekolla SG, Kossmann H, Nicolosi S, Hanus F, van Marwick S, Kunze KP, Meinicke A, Götze K, Kastrati A, Langwieser N, Ibrahim T, Nahrendorf M, Schwaiger M, Laugwitz KL. Prospective Evaluation of 18F-Fluorodeoxyglucose Uptake in Postischemic Myocardium by Simultaneous Positron Emission Tomography/Magnetic Resonance Imaging as a Prognostic Marker of Functional Outcome. Circ Cardiovasc Imaging 2016; 9:e004316. [PMID: 27056601 DOI: 10.1161/circimaging.115.004316] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 02/12/2016] [Indexed: 01/01/2023]
Abstract
BACKGROUND The immune system orchestrates the repair of infarcted myocardium. Imaging of the cellular inflammatory response by (18)F-fluorodeoxyglucose ((18)F-FDG) positron emission tomography/magnetic resonance imaging in the heart has been demonstrated in preclinical and clinical studies. However, the clinical relevance of post-MI (18)F-FDG uptake in the heart has not been elucidated. The objective of this study was to explore the value of (18)F-FDG positron emission tomography/magnetic resonance imaging in patients after acute myocardial infarction as a biosignal for left ventricular functional outcome. METHODS AND RESULTS We prospectively enrolled 49 patients with ST-segment-elevation myocardial infarction and performed (18)F-FDG positron emission tomography/magnetic resonance imaging 5 days after percutaneous coronary intervention and follow-up cardiac magnetic resonance imaging after 6 to 9 months. In a subset of patients, (99m)Tc-sestamibi single-photon emission computed tomography was performed with tracer injection before revascularization. Cellular innate immune response was analyzed at multiple time points. Segmental comparison of (18)F-FDG-uptake and late gadolinium enhancement showed substantial overlap (κ=0.66), whereas quantitative analysis demonstrated that (18)F-FDG extent exceeded late gadolinium enhancement extent (33.2±16.2% left ventricular myocardium versus 20.4±10.6% left ventricular myocardium, P<0.0001) and corresponded to the area at risk (r=0.87, P<0.0001). The peripheral blood count of CD14(high)/CD16(+) monocytes correlated with the infarction size and (18)F-FDG signal extent (r=0.53, P<0.002 and r=0.42, P<0.02, respectively). (18)F-FDG uptake in the infarcted myocardium was highest in areas with transmural scar, and the standardized uptake valuemean was associated with left ventricular functional outcome independent of infarct size (Δ ejection fraction: P<0.04, Δ end-diastolic volume: P<0.02, Δ end-systolic volume: P<0.005). CONCLUSIONS In this study, the intensity of (18)F-FDG uptake in the myocardium after acute myocardial infarction correlated inversely with functional outcome at 6 months. Thus, (18)F-FDG uptake in infarcted myocardium may represent a novel biosignal of myocardial injury.
Collapse
Affiliation(s)
- Christoph Rischpler
- From the Nuklearmedizinische Klinik und Poliklinik, Klinikum rechts der Isar (C.R., S.G.N., S.N., S.v.M., K.P.K., A.M., M.S.), Medizinische Klinik und Poliklinik I, Klinikum rechts der Isar (R.J.D., H.K., F.H., N.L., T.I., K.-L.L.), Medizinische Klinik und Poliklinik III, Klinikum rechts der Isar (K.G.), and Deutsches Herzzentrum (A.K.), Technische Universität München, Munich, Germany; DZKH (Deutsches Zentrum für Herz-Kreislauf-Forschung e.V.), partner site Munich Heart Alliance, Munich, Germany (C.R., S.G.N., A.K., M.S., K.-L.L.); and Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston (M.N.)
| | - Ralf J Dirschinger
- From the Nuklearmedizinische Klinik und Poliklinik, Klinikum rechts der Isar (C.R., S.G.N., S.N., S.v.M., K.P.K., A.M., M.S.), Medizinische Klinik und Poliklinik I, Klinikum rechts der Isar (R.J.D., H.K., F.H., N.L., T.I., K.-L.L.), Medizinische Klinik und Poliklinik III, Klinikum rechts der Isar (K.G.), and Deutsches Herzzentrum (A.K.), Technische Universität München, Munich, Germany; DZKH (Deutsches Zentrum für Herz-Kreislauf-Forschung e.V.), partner site Munich Heart Alliance, Munich, Germany (C.R., S.G.N., A.K., M.S., K.-L.L.); and Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston (M.N.)
| | - Stephan G Nekolla
- From the Nuklearmedizinische Klinik und Poliklinik, Klinikum rechts der Isar (C.R., S.G.N., S.N., S.v.M., K.P.K., A.M., M.S.), Medizinische Klinik und Poliklinik I, Klinikum rechts der Isar (R.J.D., H.K., F.H., N.L., T.I., K.-L.L.), Medizinische Klinik und Poliklinik III, Klinikum rechts der Isar (K.G.), and Deutsches Herzzentrum (A.K.), Technische Universität München, Munich, Germany; DZKH (Deutsches Zentrum für Herz-Kreislauf-Forschung e.V.), partner site Munich Heart Alliance, Munich, Germany (C.R., S.G.N., A.K., M.S., K.-L.L.); and Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston (M.N.)
| | - Hans Kossmann
- From the Nuklearmedizinische Klinik und Poliklinik, Klinikum rechts der Isar (C.R., S.G.N., S.N., S.v.M., K.P.K., A.M., M.S.), Medizinische Klinik und Poliklinik I, Klinikum rechts der Isar (R.J.D., H.K., F.H., N.L., T.I., K.-L.L.), Medizinische Klinik und Poliklinik III, Klinikum rechts der Isar (K.G.), and Deutsches Herzzentrum (A.K.), Technische Universität München, Munich, Germany; DZKH (Deutsches Zentrum für Herz-Kreislauf-Forschung e.V.), partner site Munich Heart Alliance, Munich, Germany (C.R., S.G.N., A.K., M.S., K.-L.L.); and Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston (M.N.)
| | - Stefania Nicolosi
- From the Nuklearmedizinische Klinik und Poliklinik, Klinikum rechts der Isar (C.R., S.G.N., S.N., S.v.M., K.P.K., A.M., M.S.), Medizinische Klinik und Poliklinik I, Klinikum rechts der Isar (R.J.D., H.K., F.H., N.L., T.I., K.-L.L.), Medizinische Klinik und Poliklinik III, Klinikum rechts der Isar (K.G.), and Deutsches Herzzentrum (A.K.), Technische Universität München, Munich, Germany; DZKH (Deutsches Zentrum für Herz-Kreislauf-Forschung e.V.), partner site Munich Heart Alliance, Munich, Germany (C.R., S.G.N., A.K., M.S., K.-L.L.); and Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston (M.N.)
| | - Franziska Hanus
- From the Nuklearmedizinische Klinik und Poliklinik, Klinikum rechts der Isar (C.R., S.G.N., S.N., S.v.M., K.P.K., A.M., M.S.), Medizinische Klinik und Poliklinik I, Klinikum rechts der Isar (R.J.D., H.K., F.H., N.L., T.I., K.-L.L.), Medizinische Klinik und Poliklinik III, Klinikum rechts der Isar (K.G.), and Deutsches Herzzentrum (A.K.), Technische Universität München, Munich, Germany; DZKH (Deutsches Zentrum für Herz-Kreislauf-Forschung e.V.), partner site Munich Heart Alliance, Munich, Germany (C.R., S.G.N., A.K., M.S., K.-L.L.); and Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston (M.N.)
| | - Sandra van Marwick
- From the Nuklearmedizinische Klinik und Poliklinik, Klinikum rechts der Isar (C.R., S.G.N., S.N., S.v.M., K.P.K., A.M., M.S.), Medizinische Klinik und Poliklinik I, Klinikum rechts der Isar (R.J.D., H.K., F.H., N.L., T.I., K.-L.L.), Medizinische Klinik und Poliklinik III, Klinikum rechts der Isar (K.G.), and Deutsches Herzzentrum (A.K.), Technische Universität München, Munich, Germany; DZKH (Deutsches Zentrum für Herz-Kreislauf-Forschung e.V.), partner site Munich Heart Alliance, Munich, Germany (C.R., S.G.N., A.K., M.S., K.-L.L.); and Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston (M.N.)
| | - Karl P Kunze
- From the Nuklearmedizinische Klinik und Poliklinik, Klinikum rechts der Isar (C.R., S.G.N., S.N., S.v.M., K.P.K., A.M., M.S.), Medizinische Klinik und Poliklinik I, Klinikum rechts der Isar (R.J.D., H.K., F.H., N.L., T.I., K.-L.L.), Medizinische Klinik und Poliklinik III, Klinikum rechts der Isar (K.G.), and Deutsches Herzzentrum (A.K.), Technische Universität München, Munich, Germany; DZKH (Deutsches Zentrum für Herz-Kreislauf-Forschung e.V.), partner site Munich Heart Alliance, Munich, Germany (C.R., S.G.N., A.K., M.S., K.-L.L.); and Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston (M.N.)
| | - Alexander Meinicke
- From the Nuklearmedizinische Klinik und Poliklinik, Klinikum rechts der Isar (C.R., S.G.N., S.N., S.v.M., K.P.K., A.M., M.S.), Medizinische Klinik und Poliklinik I, Klinikum rechts der Isar (R.J.D., H.K., F.H., N.L., T.I., K.-L.L.), Medizinische Klinik und Poliklinik III, Klinikum rechts der Isar (K.G.), and Deutsches Herzzentrum (A.K.), Technische Universität München, Munich, Germany; DZKH (Deutsches Zentrum für Herz-Kreislauf-Forschung e.V.), partner site Munich Heart Alliance, Munich, Germany (C.R., S.G.N., A.K., M.S., K.-L.L.); and Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston (M.N.)
| | - Katharina Götze
- From the Nuklearmedizinische Klinik und Poliklinik, Klinikum rechts der Isar (C.R., S.G.N., S.N., S.v.M., K.P.K., A.M., M.S.), Medizinische Klinik und Poliklinik I, Klinikum rechts der Isar (R.J.D., H.K., F.H., N.L., T.I., K.-L.L.), Medizinische Klinik und Poliklinik III, Klinikum rechts der Isar (K.G.), and Deutsches Herzzentrum (A.K.), Technische Universität München, Munich, Germany; DZKH (Deutsches Zentrum für Herz-Kreislauf-Forschung e.V.), partner site Munich Heart Alliance, Munich, Germany (C.R., S.G.N., A.K., M.S., K.-L.L.); and Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston (M.N.)
| | - Adnan Kastrati
- From the Nuklearmedizinische Klinik und Poliklinik, Klinikum rechts der Isar (C.R., S.G.N., S.N., S.v.M., K.P.K., A.M., M.S.), Medizinische Klinik und Poliklinik I, Klinikum rechts der Isar (R.J.D., H.K., F.H., N.L., T.I., K.-L.L.), Medizinische Klinik und Poliklinik III, Klinikum rechts der Isar (K.G.), and Deutsches Herzzentrum (A.K.), Technische Universität München, Munich, Germany; DZKH (Deutsches Zentrum für Herz-Kreislauf-Forschung e.V.), partner site Munich Heart Alliance, Munich, Germany (C.R., S.G.N., A.K., M.S., K.-L.L.); and Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston (M.N.)
| | - Nicolas Langwieser
- From the Nuklearmedizinische Klinik und Poliklinik, Klinikum rechts der Isar (C.R., S.G.N., S.N., S.v.M., K.P.K., A.M., M.S.), Medizinische Klinik und Poliklinik I, Klinikum rechts der Isar (R.J.D., H.K., F.H., N.L., T.I., K.-L.L.), Medizinische Klinik und Poliklinik III, Klinikum rechts der Isar (K.G.), and Deutsches Herzzentrum (A.K.), Technische Universität München, Munich, Germany; DZKH (Deutsches Zentrum für Herz-Kreislauf-Forschung e.V.), partner site Munich Heart Alliance, Munich, Germany (C.R., S.G.N., A.K., M.S., K.-L.L.); and Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston (M.N.)
| | - Tareq Ibrahim
- From the Nuklearmedizinische Klinik und Poliklinik, Klinikum rechts der Isar (C.R., S.G.N., S.N., S.v.M., K.P.K., A.M., M.S.), Medizinische Klinik und Poliklinik I, Klinikum rechts der Isar (R.J.D., H.K., F.H., N.L., T.I., K.-L.L.), Medizinische Klinik und Poliklinik III, Klinikum rechts der Isar (K.G.), and Deutsches Herzzentrum (A.K.), Technische Universität München, Munich, Germany; DZKH (Deutsches Zentrum für Herz-Kreislauf-Forschung e.V.), partner site Munich Heart Alliance, Munich, Germany (C.R., S.G.N., A.K., M.S., K.-L.L.); and Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston (M.N.)
| | - Matthias Nahrendorf
- From the Nuklearmedizinische Klinik und Poliklinik, Klinikum rechts der Isar (C.R., S.G.N., S.N., S.v.M., K.P.K., A.M., M.S.), Medizinische Klinik und Poliklinik I, Klinikum rechts der Isar (R.J.D., H.K., F.H., N.L., T.I., K.-L.L.), Medizinische Klinik und Poliklinik III, Klinikum rechts der Isar (K.G.), and Deutsches Herzzentrum (A.K.), Technische Universität München, Munich, Germany; DZKH (Deutsches Zentrum für Herz-Kreislauf-Forschung e.V.), partner site Munich Heart Alliance, Munich, Germany (C.R., S.G.N., A.K., M.S., K.-L.L.); and Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston (M.N.)
| | - Markus Schwaiger
- From the Nuklearmedizinische Klinik und Poliklinik, Klinikum rechts der Isar (C.R., S.G.N., S.N., S.v.M., K.P.K., A.M., M.S.), Medizinische Klinik und Poliklinik I, Klinikum rechts der Isar (R.J.D., H.K., F.H., N.L., T.I., K.-L.L.), Medizinische Klinik und Poliklinik III, Klinikum rechts der Isar (K.G.), and Deutsches Herzzentrum (A.K.), Technische Universität München, Munich, Germany; DZKH (Deutsches Zentrum für Herz-Kreislauf-Forschung e.V.), partner site Munich Heart Alliance, Munich, Germany (C.R., S.G.N., A.K., M.S., K.-L.L.); and Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston (M.N.)
| | - Karl-Ludwig Laugwitz
- From the Nuklearmedizinische Klinik und Poliklinik, Klinikum rechts der Isar (C.R., S.G.N., S.N., S.v.M., K.P.K., A.M., M.S.), Medizinische Klinik und Poliklinik I, Klinikum rechts der Isar (R.J.D., H.K., F.H., N.L., T.I., K.-L.L.), Medizinische Klinik und Poliklinik III, Klinikum rechts der Isar (K.G.), and Deutsches Herzzentrum (A.K.), Technische Universität München, Munich, Germany; DZKH (Deutsches Zentrum für Herz-Kreislauf-Forschung e.V.), partner site Munich Heart Alliance, Munich, Germany (C.R., S.G.N., A.K., M.S., K.-L.L.); and Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston (M.N.).
| |
Collapse
|
962
|
Wang J, Ma Y, Sachs F, Li J, Suchyna TM. GsMTx4-D is a cardioprotectant against myocardial infarction during ischemia and reperfusion. J Mol Cell Cardiol 2016; 98:83-94. [PMID: 27423272 DOI: 10.1016/j.yjmcc.2016.07.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Revised: 06/25/2016] [Accepted: 07/12/2016] [Indexed: 01/01/2023]
Abstract
GsMTx4 is a selective inhibitor of cationic mechanosensitive ion channels (MSCs) and has helped establish the role of MSCs in cardiac physiology. Inhomogeneous local mechanical stresses due to hypercontracture and swelling during ischemic reperfusion injury (IRI) likely induce elevated MSC activity that can contribute to cation imbalance. The aim of this study was to determine if the D enantiomer of GsMTx4 can act as a cardioprotectant in a mouse IRI model. Ischemia and reperfusion involved ligating a coronary artery followed by release of the ligature. GsMTx4-D was tested by either acute intravenous injection during the ischemic event or by two day pretreatment by intraperitoneal injection, both methods achieving similar results. Based on pharmacokinetic studies, GsMTx4-D dosage was set to achieve expected plasma concentrations between 50 and 5000nM and heart tissue concentrations between 1 and 200nM by intravenous injection. Relative to vehicle injected animals, GsMTx4-D reduced infarct area by ~40% for acute and pretreated animals for both 20 and 45min ischemic challenges. Many indicators of cardiac output were indistinguishable from sham-treated control hearts after GsMTx4-D treatment showing improvement at both 4 and 48h post ischemia, and premature ventricular beats immediately following reperfusion were also significantly reduced. To determine if GsMTx4-D cardioprotection could act directly at the level of cardiomyocytes, we tested its effects in vitro on indicators of IRI damage like cation influx and activation of inflammatory kinases in isolated myocytes cultured under hypoxic conditions. Hypoxia challenged cardiomyocytes treated with 10μM GsMTx4-D showed improved contractility and near normal contraction-related Ca(2+) influx. GsMTx4-D inhibited indicators of ischemic damage such as the apoptotic signaling system JNK/c-Jun, but also inhibited the energy response signaling system Akt kinase. We conclude that GsMTx4-D is a potent cardioprotectant in vivo that may act directly on cardiomyocytes and potentially be useful in multidrug strategies to treat IRI.
Collapse
Affiliation(s)
- Jinli Wang
- Department of Pharmacology and Toxicology, School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14214, United States
| | - Yina Ma
- Department of Pharmacology and Toxicology, School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14214, United States
| | - Frederick Sachs
- Department of Physiology and Biophysics, School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14214, United States
| | - Ji Li
- Department of Pharmacology and Toxicology, School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14214, United States
| | - Thomas M Suchyna
- Department of Physiology and Biophysics, School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14214, United States.
| |
Collapse
|
963
|
Mummidi S, Das NA, Carpenter AJ, Kandikattu H, Krenz M, Siebenlist U, Valente AJ, Chandrasekar B. Metformin inhibits aldosterone-induced cardiac fibroblast activation, migration and proliferation in vitro, and reverses aldosterone+salt-induced cardiac fibrosis in vivo. J Mol Cell Cardiol 2016; 98:95-102. [PMID: 27423273 DOI: 10.1016/j.yjmcc.2016.07.006] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Revised: 07/08/2016] [Accepted: 07/13/2016] [Indexed: 11/29/2022]
Abstract
The overall goals of this study were to investigate whether metformin exerts anti-fibrotic effects in aldosterone (Aldo)+salt-treated wild type mouse hearts, and determine the underlying molecular mechanisms in isolated adult cardiac fibroblasts (CF). In vitro, Aldo induced CF activation, migration, and proliferation, and these effects were inhibited by metformin. Further, Aldo induced PPM1A (Protein Phosphatase Magnesium Dependent 1A) activation and inhibited AMPK phosphorylation. At a pharmacologically relevant concentration, metformin restored AMPK activation, and inhibited Aldo-induced Nox4/H2O2-dependent TRAF3IP2 induction, pro-inflammatory cytokine expression, and CF migration and proliferation. Further, metformin potentiated the inhibitory effects of spironolactone, a mineralocorticoid receptor antagonist, on Aldo-induced collagen expression, and CF migration and proliferation. These results were recapitulated in vivo, where metformin reversed Aldo+salt-induced oxidative stress, suppression of AMPK activation, TRAF3IP2 induction, pro-inflammatory cytokine expression, and cardiac fibrosis, without significantly modulating systolic blood pressure. These in vitro and in vivo data indicate that metformin has the potential to reduce adverse cardiac remodeling in hypertensive heart disease.
Collapse
Affiliation(s)
- Srinivas Mummidi
- Department of Medicine, University of Texas Health Science Center, San Antonio, TX 78229, USA
| | - Nitin A Das
- Department of Cardiothoracic Surgery, University of Texas Health Science Center, San Antonio, TX 78229, USA
| | - Andrea J Carpenter
- Department of Cardiothoracic Surgery, University of Texas Health Science Center, San Antonio, TX 78229, USA
| | | | - Maike Krenz
- Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, MO 65211, USA
| | | | - Anthony J Valente
- Department of Medicine, University of Texas Health Science Center, San Antonio, TX 78229, USA
| | - Bysani Chandrasekar
- Medicine/Cardiology, University of Missouri School of Medicine, Columbia, MO 65211, USA; Research Service, Harry S. Truman Memorial Veterans Hospital, Columbia, MO 65201, USA.
| |
Collapse
|
964
|
Kubota A, Hasegawa H, Tadokoro H, Hirose M, Kobara Y, Yamada-Inagawa T, Takemura G, Kobayashi Y, Takano H. Deletion of CD28 Co-stimulatory Signals Exacerbates Left Ventricular Remodeling and Increases Cardiac Rupture After Myocardial Infarction. Circ J 2016; 80:1971-9. [PMID: 27396441 DOI: 10.1253/circj.cj-16-0327] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND Inflammatory responses, especially by CD4(+)T cells activated by dendritic cells, are known to be important in the pathophysiology of cardiac repair after myocardial infarction (MI). Although co-stimulatory signals through B7 (CD80/86) and CD28 are necessary for CD4(+)T cell activation and survival, the roles of these signals in cardiac repair after MI are still unclear. METHODS AND RESULTS C57BL/6 (Control) mice and CD28 knockout (CD28KO) mice were subjected to left coronary artery permanent ligation. The ratio of death by cardiac rupture within 5 days after MI was significantly higher in CD28KO mice compared with Control mice. Although there were no significant differences in the infarct size between the 2 groups, left ventricular end-diastolic and end-systolic diameters were significantly increased, and fractional shortening was significantly decreased in CD28KO mice compared with Control mice. Electron microscopic observation revealed that the extent of extracellular collagen fiber was significantly decreased in CD28KO mice compared with Control mice. The number of α-smooth muscle actin-positive myofibroblasts was significantly decreased, and matrix metalloproteinase-9 activity and the mRNA expression of interleukin-1β were significantly increased in CD28KO mice compared with Control mice. CONCLUSIONS Deletion of CD28 co-stimulatory signals exacerbates left ventricular remodeling and increases cardiac rupture after MI through prolongation of the inflammatory period and reduction of collagen fiber in the infarct scars. (Circ J 2016; 80: 1971-1979).
Collapse
Affiliation(s)
- Akihiko Kubota
- Department of Cardiovascular Medicine, Chiba University Graduate School of Medicine
| | | | | | | | | | | | | | | | | |
Collapse
|
965
|
Grisanti LA, Gumpert AM, Traynham CJ, Gorsky JE, Repas AA, Gao E, Carter RL, Yu D, Calvert JW, García AP, Ibáñez B, Rabinowitz JE, Koch WJ, Tilley DG. Leukocyte-Expressed β2-Adrenergic Receptors Are Essential for Survival After Acute Myocardial Injury. Circulation 2016; 134:153-67. [PMID: 27364164 DOI: 10.1161/circulationaha.116.022304] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Accepted: 05/17/2016] [Indexed: 11/16/2022]
Abstract
BACKGROUND Immune cell-mediated inflammation is an essential process for mounting a repair response after myocardial infarction (MI). The sympathetic nervous system is known to regulate immune system function through β-adrenergic receptors (βARs); however, their role in regulating immune cell responses to acute cardiac injury is unknown. METHODS Wild-type (WT) mice were irradiated followed by isoform-specific βAR knockout (βARKO) or WT bone-marrow transplantation (BMT) and after full reconstitution underwent MI surgery. Survival was monitored over time, and alterations in immune cell infiltration after MI were examined through immunohistochemistry. Alterations in splenic function were identified through the investigation of altered adhesion receptor expression. RESULTS β2ARKO BMT mice displayed 100% mortality resulting from cardiac rupture within 12 days after MI compared with ≈20% mortality in WT BMT mice. β2ARKO BMT mice displayed severely reduced post-MI cardiac infiltration of leukocytes with reciprocally enhanced splenic retention of the same immune cell populations. Splenic retention of the leukocytes was associated with an increase in vascular cell adhesion molecule-1 expression, which itself was regulated via β-arrestin-dependent β2AR signaling. Furthermore, vascular cell adhesion molecule-1 expression in both mouse and human macrophages was sensitive to β2AR activity, and spleens from human tissue donors treated with β-blocker showed enhanced vascular cell adhesion molecule-1 expression. The impairments in splenic retention and cardiac infiltration of leukocytes after MI were restored to WT levels via lentiviral-mediated re-expression of β2AR in β2ARKO bone marrow before transplantation, which also resulted in post-MI survival rates comparable to those in WT BMT mice. CONCLUSIONS Immune cell-expressed β2AR plays an essential role in regulating the early inflammatory repair response to acute myocardial injury by facilitating cardiac leukocyte infiltration.
Collapse
Affiliation(s)
- Laurel A Grisanti
- From Center for Translational Medicine (L.A.G., A.M.G., C.J.T., J.E.G., A.A.R., E.G., R.L.C., J.E.R., W.J.K., D.G.T.), Department of Pharmacology (E.G., J.E.R., W.J.K., D.G.T.), and Department of Clinical Sciences (D.Y.), Temple University School of Medicine, Philadelphia, PA; Department of Surgery, Division of Cardiothoracic Surgery, Emory University School of Medicine and Carlyle Fraser Heart Center, Atlanta, GA (J.W.C.); and Spanish National Center for Cardiovascular Research, Madrid, Spain (A.P.G., B.I.)
| | - Anna M Gumpert
- From Center for Translational Medicine (L.A.G., A.M.G., C.J.T., J.E.G., A.A.R., E.G., R.L.C., J.E.R., W.J.K., D.G.T.), Department of Pharmacology (E.G., J.E.R., W.J.K., D.G.T.), and Department of Clinical Sciences (D.Y.), Temple University School of Medicine, Philadelphia, PA; Department of Surgery, Division of Cardiothoracic Surgery, Emory University School of Medicine and Carlyle Fraser Heart Center, Atlanta, GA (J.W.C.); and Spanish National Center for Cardiovascular Research, Madrid, Spain (A.P.G., B.I.)
| | - Christopher J Traynham
- From Center for Translational Medicine (L.A.G., A.M.G., C.J.T., J.E.G., A.A.R., E.G., R.L.C., J.E.R., W.J.K., D.G.T.), Department of Pharmacology (E.G., J.E.R., W.J.K., D.G.T.), and Department of Clinical Sciences (D.Y.), Temple University School of Medicine, Philadelphia, PA; Department of Surgery, Division of Cardiothoracic Surgery, Emory University School of Medicine and Carlyle Fraser Heart Center, Atlanta, GA (J.W.C.); and Spanish National Center for Cardiovascular Research, Madrid, Spain (A.P.G., B.I.)
| | - Joshua E Gorsky
- From Center for Translational Medicine (L.A.G., A.M.G., C.J.T., J.E.G., A.A.R., E.G., R.L.C., J.E.R., W.J.K., D.G.T.), Department of Pharmacology (E.G., J.E.R., W.J.K., D.G.T.), and Department of Clinical Sciences (D.Y.), Temple University School of Medicine, Philadelphia, PA; Department of Surgery, Division of Cardiothoracic Surgery, Emory University School of Medicine and Carlyle Fraser Heart Center, Atlanta, GA (J.W.C.); and Spanish National Center for Cardiovascular Research, Madrid, Spain (A.P.G., B.I.)
| | - Ashley A Repas
- From Center for Translational Medicine (L.A.G., A.M.G., C.J.T., J.E.G., A.A.R., E.G., R.L.C., J.E.R., W.J.K., D.G.T.), Department of Pharmacology (E.G., J.E.R., W.J.K., D.G.T.), and Department of Clinical Sciences (D.Y.), Temple University School of Medicine, Philadelphia, PA; Department of Surgery, Division of Cardiothoracic Surgery, Emory University School of Medicine and Carlyle Fraser Heart Center, Atlanta, GA (J.W.C.); and Spanish National Center for Cardiovascular Research, Madrid, Spain (A.P.G., B.I.)
| | - Erhe Gao
- From Center for Translational Medicine (L.A.G., A.M.G., C.J.T., J.E.G., A.A.R., E.G., R.L.C., J.E.R., W.J.K., D.G.T.), Department of Pharmacology (E.G., J.E.R., W.J.K., D.G.T.), and Department of Clinical Sciences (D.Y.), Temple University School of Medicine, Philadelphia, PA; Department of Surgery, Division of Cardiothoracic Surgery, Emory University School of Medicine and Carlyle Fraser Heart Center, Atlanta, GA (J.W.C.); and Spanish National Center for Cardiovascular Research, Madrid, Spain (A.P.G., B.I.)
| | - Rhonda L Carter
- From Center for Translational Medicine (L.A.G., A.M.G., C.J.T., J.E.G., A.A.R., E.G., R.L.C., J.E.R., W.J.K., D.G.T.), Department of Pharmacology (E.G., J.E.R., W.J.K., D.G.T.), and Department of Clinical Sciences (D.Y.), Temple University School of Medicine, Philadelphia, PA; Department of Surgery, Division of Cardiothoracic Surgery, Emory University School of Medicine and Carlyle Fraser Heart Center, Atlanta, GA (J.W.C.); and Spanish National Center for Cardiovascular Research, Madrid, Spain (A.P.G., B.I.)
| | - Daohai Yu
- From Center for Translational Medicine (L.A.G., A.M.G., C.J.T., J.E.G., A.A.R., E.G., R.L.C., J.E.R., W.J.K., D.G.T.), Department of Pharmacology (E.G., J.E.R., W.J.K., D.G.T.), and Department of Clinical Sciences (D.Y.), Temple University School of Medicine, Philadelphia, PA; Department of Surgery, Division of Cardiothoracic Surgery, Emory University School of Medicine and Carlyle Fraser Heart Center, Atlanta, GA (J.W.C.); and Spanish National Center for Cardiovascular Research, Madrid, Spain (A.P.G., B.I.)
| | - John W Calvert
- From Center for Translational Medicine (L.A.G., A.M.G., C.J.T., J.E.G., A.A.R., E.G., R.L.C., J.E.R., W.J.K., D.G.T.), Department of Pharmacology (E.G., J.E.R., W.J.K., D.G.T.), and Department of Clinical Sciences (D.Y.), Temple University School of Medicine, Philadelphia, PA; Department of Surgery, Division of Cardiothoracic Surgery, Emory University School of Medicine and Carlyle Fraser Heart Center, Atlanta, GA (J.W.C.); and Spanish National Center for Cardiovascular Research, Madrid, Spain (A.P.G., B.I.)
| | - Andrés Pun García
- From Center for Translational Medicine (L.A.G., A.M.G., C.J.T., J.E.G., A.A.R., E.G., R.L.C., J.E.R., W.J.K., D.G.T.), Department of Pharmacology (E.G., J.E.R., W.J.K., D.G.T.), and Department of Clinical Sciences (D.Y.), Temple University School of Medicine, Philadelphia, PA; Department of Surgery, Division of Cardiothoracic Surgery, Emory University School of Medicine and Carlyle Fraser Heart Center, Atlanta, GA (J.W.C.); and Spanish National Center for Cardiovascular Research, Madrid, Spain (A.P.G., B.I.)
| | - Borja Ibáñez
- From Center for Translational Medicine (L.A.G., A.M.G., C.J.T., J.E.G., A.A.R., E.G., R.L.C., J.E.R., W.J.K., D.G.T.), Department of Pharmacology (E.G., J.E.R., W.J.K., D.G.T.), and Department of Clinical Sciences (D.Y.), Temple University School of Medicine, Philadelphia, PA; Department of Surgery, Division of Cardiothoracic Surgery, Emory University School of Medicine and Carlyle Fraser Heart Center, Atlanta, GA (J.W.C.); and Spanish National Center for Cardiovascular Research, Madrid, Spain (A.P.G., B.I.)
| | - Joseph E Rabinowitz
- From Center for Translational Medicine (L.A.G., A.M.G., C.J.T., J.E.G., A.A.R., E.G., R.L.C., J.E.R., W.J.K., D.G.T.), Department of Pharmacology (E.G., J.E.R., W.J.K., D.G.T.), and Department of Clinical Sciences (D.Y.), Temple University School of Medicine, Philadelphia, PA; Department of Surgery, Division of Cardiothoracic Surgery, Emory University School of Medicine and Carlyle Fraser Heart Center, Atlanta, GA (J.W.C.); and Spanish National Center for Cardiovascular Research, Madrid, Spain (A.P.G., B.I.)
| | - Walter J Koch
- From Center for Translational Medicine (L.A.G., A.M.G., C.J.T., J.E.G., A.A.R., E.G., R.L.C., J.E.R., W.J.K., D.G.T.), Department of Pharmacology (E.G., J.E.R., W.J.K., D.G.T.), and Department of Clinical Sciences (D.Y.), Temple University School of Medicine, Philadelphia, PA; Department of Surgery, Division of Cardiothoracic Surgery, Emory University School of Medicine and Carlyle Fraser Heart Center, Atlanta, GA (J.W.C.); and Spanish National Center for Cardiovascular Research, Madrid, Spain (A.P.G., B.I.)
| | - Douglas G Tilley
- From Center for Translational Medicine (L.A.G., A.M.G., C.J.T., J.E.G., A.A.R., E.G., R.L.C., J.E.R., W.J.K., D.G.T.), Department of Pharmacology (E.G., J.E.R., W.J.K., D.G.T.), and Department of Clinical Sciences (D.Y.), Temple University School of Medicine, Philadelphia, PA; Department of Surgery, Division of Cardiothoracic Surgery, Emory University School of Medicine and Carlyle Fraser Heart Center, Atlanta, GA (J.W.C.); and Spanish National Center for Cardiovascular Research, Madrid, Spain (A.P.G., B.I.).
| |
Collapse
|
966
|
Talman V, Ruskoaho H. Cardiac fibrosis in myocardial infarction-from repair and remodeling to regeneration. Cell Tissue Res 2016; 365:563-81. [PMID: 27324127 PMCID: PMC5010608 DOI: 10.1007/s00441-016-2431-9] [Citation(s) in RCA: 578] [Impact Index Per Article: 72.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 05/07/2016] [Indexed: 12/11/2022]
Abstract
Ischemic cell death during a myocardial infarction leads to a multiphase reparative response in which the damaged tissue is replaced with a fibrotic scar produced by fibroblasts and myofibroblasts. This also induces geometrical, biomechanical, and biochemical changes in the uninjured ventricular wall eliciting a reactive remodeling process that includes interstitial and perivascular fibrosis. Although the initial reparative fibrosis is crucial for preventing rupture of the ventricular wall, an exaggerated fibrotic response and reactive fibrosis outside the injured area are detrimental as they lead to progressive impairment of cardiac function and eventually to heart failure. In this review, we summarize current knowledge of the mechanisms of both reparative and reactive cardiac fibrosis in response to myocardial infarction, discuss the potential of inducing cardiac regeneration through direct reprogramming of fibroblasts and myofibroblasts into cardiomyocytes, and review the currently available and potential future therapeutic strategies to inhibit cardiac fibrosis. Graphical abstract Reparative response following a myocardial infarction. Hypoxia-induced cardiomyocyte death leads to the activation of myofibroblasts and a reparative fibrotic response in the injured area. Right top In adult mammals, the fibrotic scar formed at the infarcted area is permanent and promotes reactive fibrosis in the uninjured myocardium. Right bottom In teleost fish and newts and in embryonic and neonatal mammals, the initial formation of a fibrotic scar is followed by regeneration of the cardiac muscle tissue. Induction of post-infarction cardiac regeneration in adult mammals is currently the target of intensive research and drug discovery attempts.
Collapse
Affiliation(s)
- Virpi Talman
- Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, FI-00014, Helsinki, Finland.
| | - Heikki Ruskoaho
- Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, FI-00014, Helsinki, Finland
| |
Collapse
|
967
|
Targeting the Innate Immune Response to Improve Cardiac Graft Recovery after Heart Transplantation: Implications for the Donation after Cardiac Death. Int J Mol Sci 2016; 17:ijms17060958. [PMID: 27322252 PMCID: PMC4926491 DOI: 10.3390/ijms17060958] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Revised: 05/30/2016] [Accepted: 06/01/2016] [Indexed: 12/26/2022] Open
Abstract
Heart transplantation (HTx) is the ultimate treatment for end-stage heart failure. The number of patients on waiting lists for heart transplants, however, is much higher than the number of available organs. The shortage of donor hearts is a serious concern since the population affected by heart failure is constantly increasing. Furthermore, the long-term success of HTx poses some challenges despite the improvement in the management of the short-term complications and in the methods to limit graft rejection. Myocardial injury occurs during transplantation. Injury initiated in the donor as result of brain or cardiac death is exacerbated by organ procurement and storage, and is ultimately amplified by reperfusion injury at the time of transplantation. The innate immune system is a mechanism of first-line defense against pathogens and cell injury. Innate immunity is activated during myocardial injury and produces deleterious effects on the heart structure and function. Here, we briefly discuss the role of the innate immunity in the initiation of myocardial injury, with particular focus on the Toll-like receptors and inflammasome, and how to potentially expand the donor population by targeting the innate immune response.
Collapse
|
968
|
Sanders LN, Schoenhard JA, Saleh MA, Mukherjee A, Ryzhov S, McMaster WG, Nolan K, Gumina RJ, Thompson TB, Magnuson MA, Harrison DG, Hatzopoulos AK. BMP Antagonist Gremlin 2 Limits Inflammation After Myocardial Infarction. Circ Res 2016; 119:434-49. [PMID: 27283840 DOI: 10.1161/circresaha.116.308700] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 06/09/2016] [Indexed: 11/16/2022]
Abstract
RATIONALE We have recently shown that the bone morphogenetic protein (BMP) antagonist Gremlin 2 (Grem2) is required for early cardiac development and cardiomyocyte differentiation. Our initial studies discovered that Grem2 is strongly induced in the adult heart after experimental myocardial infarction (MI). However, the function of Grem2 and BMP-signaling inhibitors after cardiac injury is currently unknown. OBJECTIVE To investigate the role of Grem2 during cardiac repair and assess its potential to improve ventricular function after injury. METHODS AND RESULTS Our data show that Grem2 is transiently induced after MI in peri-infarct area cardiomyocytes during the inflammatory phase of cardiac tissue repair. By engineering loss- (Grem2(-/-)) and gain- (TG(Grem2)) of-Grem2-function mice, we discovered that Grem2 controls the magnitude of the inflammatory response and limits infiltration of inflammatory cells in peri-infarct ventricular tissue, improving cardiac function. Excessive inflammation in Grem2(-/-) mice after MI was because of overactivation of canonical BMP signaling, as proven by the rescue of the inflammatory phenotype through administration of the canonical BMP inhibitor, DMH1. Furthermore, intraperitoneal administration of Grem2 protein in wild-type mice was sufficient to reduce inflammation after MI. Cellular analyses showed that BMP2 acts with TNFα to induce expression of proinflammatory proteins in endothelial cells and promote adhesion of leukocytes, whereas Grem2 specifically inhibits the BMP2 effect. CONCLUSIONS Our results indicate that Grem2 provides a molecular barrier that controls the magnitude and extent of inflammatory cell infiltration by suppressing canonical BMP signaling, thereby providing a novel mechanism for limiting the adverse effects of excessive inflammation after MI.
Collapse
Affiliation(s)
- Lehanna N Sanders
- From the Division of Cardiovascular Medicine, Department of Medicine (L.N.S., J.A.S., A.M., R.J.G., A.K.H.), Department of Cell and Developmental Biology (L.N.S., A.K.H.), Division of Clinical Pharmacology, Department of Medicine (M.A.S., W.G.M., D.G.H.), and Division of General Surgery, Department of Surgery (W.G.M.), Vanderbilt University Medical Center, Nashville, TN; Maine Medical Center Research Institute, Scarborough (S.R.); Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati, OH (K.N., T.B.T.); CentraCare Health, St. Cloud, MN (J.A.S.); Cincinnati Children's Hospital Medical Center, OH (A.M.); Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt (M.A.S.); and Center for Stem Cell Biology, Vanderbilt University School of Medicine, Nashville, TN (M.A.M.)
| | - John A Schoenhard
- From the Division of Cardiovascular Medicine, Department of Medicine (L.N.S., J.A.S., A.M., R.J.G., A.K.H.), Department of Cell and Developmental Biology (L.N.S., A.K.H.), Division of Clinical Pharmacology, Department of Medicine (M.A.S., W.G.M., D.G.H.), and Division of General Surgery, Department of Surgery (W.G.M.), Vanderbilt University Medical Center, Nashville, TN; Maine Medical Center Research Institute, Scarborough (S.R.); Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati, OH (K.N., T.B.T.); CentraCare Health, St. Cloud, MN (J.A.S.); Cincinnati Children's Hospital Medical Center, OH (A.M.); Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt (M.A.S.); and Center for Stem Cell Biology, Vanderbilt University School of Medicine, Nashville, TN (M.A.M.)
| | - Mohamed A Saleh
- From the Division of Cardiovascular Medicine, Department of Medicine (L.N.S., J.A.S., A.M., R.J.G., A.K.H.), Department of Cell and Developmental Biology (L.N.S., A.K.H.), Division of Clinical Pharmacology, Department of Medicine (M.A.S., W.G.M., D.G.H.), and Division of General Surgery, Department of Surgery (W.G.M.), Vanderbilt University Medical Center, Nashville, TN; Maine Medical Center Research Institute, Scarborough (S.R.); Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati, OH (K.N., T.B.T.); CentraCare Health, St. Cloud, MN (J.A.S.); Cincinnati Children's Hospital Medical Center, OH (A.M.); Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt (M.A.S.); and Center for Stem Cell Biology, Vanderbilt University School of Medicine, Nashville, TN (M.A.M.)
| | - Amrita Mukherjee
- From the Division of Cardiovascular Medicine, Department of Medicine (L.N.S., J.A.S., A.M., R.J.G., A.K.H.), Department of Cell and Developmental Biology (L.N.S., A.K.H.), Division of Clinical Pharmacology, Department of Medicine (M.A.S., W.G.M., D.G.H.), and Division of General Surgery, Department of Surgery (W.G.M.), Vanderbilt University Medical Center, Nashville, TN; Maine Medical Center Research Institute, Scarborough (S.R.); Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati, OH (K.N., T.B.T.); CentraCare Health, St. Cloud, MN (J.A.S.); Cincinnati Children's Hospital Medical Center, OH (A.M.); Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt (M.A.S.); and Center for Stem Cell Biology, Vanderbilt University School of Medicine, Nashville, TN (M.A.M.)
| | - Sergey Ryzhov
- From the Division of Cardiovascular Medicine, Department of Medicine (L.N.S., J.A.S., A.M., R.J.G., A.K.H.), Department of Cell and Developmental Biology (L.N.S., A.K.H.), Division of Clinical Pharmacology, Department of Medicine (M.A.S., W.G.M., D.G.H.), and Division of General Surgery, Department of Surgery (W.G.M.), Vanderbilt University Medical Center, Nashville, TN; Maine Medical Center Research Institute, Scarborough (S.R.); Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati, OH (K.N., T.B.T.); CentraCare Health, St. Cloud, MN (J.A.S.); Cincinnati Children's Hospital Medical Center, OH (A.M.); Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt (M.A.S.); and Center for Stem Cell Biology, Vanderbilt University School of Medicine, Nashville, TN (M.A.M.)
| | - William G McMaster
- From the Division of Cardiovascular Medicine, Department of Medicine (L.N.S., J.A.S., A.M., R.J.G., A.K.H.), Department of Cell and Developmental Biology (L.N.S., A.K.H.), Division of Clinical Pharmacology, Department of Medicine (M.A.S., W.G.M., D.G.H.), and Division of General Surgery, Department of Surgery (W.G.M.), Vanderbilt University Medical Center, Nashville, TN; Maine Medical Center Research Institute, Scarborough (S.R.); Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati, OH (K.N., T.B.T.); CentraCare Health, St. Cloud, MN (J.A.S.); Cincinnati Children's Hospital Medical Center, OH (A.M.); Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt (M.A.S.); and Center for Stem Cell Biology, Vanderbilt University School of Medicine, Nashville, TN (M.A.M.)
| | - Kristof Nolan
- From the Division of Cardiovascular Medicine, Department of Medicine (L.N.S., J.A.S., A.M., R.J.G., A.K.H.), Department of Cell and Developmental Biology (L.N.S., A.K.H.), Division of Clinical Pharmacology, Department of Medicine (M.A.S., W.G.M., D.G.H.), and Division of General Surgery, Department of Surgery (W.G.M.), Vanderbilt University Medical Center, Nashville, TN; Maine Medical Center Research Institute, Scarborough (S.R.); Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati, OH (K.N., T.B.T.); CentraCare Health, St. Cloud, MN (J.A.S.); Cincinnati Children's Hospital Medical Center, OH (A.M.); Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt (M.A.S.); and Center for Stem Cell Biology, Vanderbilt University School of Medicine, Nashville, TN (M.A.M.)
| | - Richard J Gumina
- From the Division of Cardiovascular Medicine, Department of Medicine (L.N.S., J.A.S., A.M., R.J.G., A.K.H.), Department of Cell and Developmental Biology (L.N.S., A.K.H.), Division of Clinical Pharmacology, Department of Medicine (M.A.S., W.G.M., D.G.H.), and Division of General Surgery, Department of Surgery (W.G.M.), Vanderbilt University Medical Center, Nashville, TN; Maine Medical Center Research Institute, Scarborough (S.R.); Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati, OH (K.N., T.B.T.); CentraCare Health, St. Cloud, MN (J.A.S.); Cincinnati Children's Hospital Medical Center, OH (A.M.); Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt (M.A.S.); and Center for Stem Cell Biology, Vanderbilt University School of Medicine, Nashville, TN (M.A.M.)
| | - Thomas B Thompson
- From the Division of Cardiovascular Medicine, Department of Medicine (L.N.S., J.A.S., A.M., R.J.G., A.K.H.), Department of Cell and Developmental Biology (L.N.S., A.K.H.), Division of Clinical Pharmacology, Department of Medicine (M.A.S., W.G.M., D.G.H.), and Division of General Surgery, Department of Surgery (W.G.M.), Vanderbilt University Medical Center, Nashville, TN; Maine Medical Center Research Institute, Scarborough (S.R.); Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati, OH (K.N., T.B.T.); CentraCare Health, St. Cloud, MN (J.A.S.); Cincinnati Children's Hospital Medical Center, OH (A.M.); Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt (M.A.S.); and Center for Stem Cell Biology, Vanderbilt University School of Medicine, Nashville, TN (M.A.M.)
| | - Mark A Magnuson
- From the Division of Cardiovascular Medicine, Department of Medicine (L.N.S., J.A.S., A.M., R.J.G., A.K.H.), Department of Cell and Developmental Biology (L.N.S., A.K.H.), Division of Clinical Pharmacology, Department of Medicine (M.A.S., W.G.M., D.G.H.), and Division of General Surgery, Department of Surgery (W.G.M.), Vanderbilt University Medical Center, Nashville, TN; Maine Medical Center Research Institute, Scarborough (S.R.); Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati, OH (K.N., T.B.T.); CentraCare Health, St. Cloud, MN (J.A.S.); Cincinnati Children's Hospital Medical Center, OH (A.M.); Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt (M.A.S.); and Center for Stem Cell Biology, Vanderbilt University School of Medicine, Nashville, TN (M.A.M.)
| | - David G Harrison
- From the Division of Cardiovascular Medicine, Department of Medicine (L.N.S., J.A.S., A.M., R.J.G., A.K.H.), Department of Cell and Developmental Biology (L.N.S., A.K.H.), Division of Clinical Pharmacology, Department of Medicine (M.A.S., W.G.M., D.G.H.), and Division of General Surgery, Department of Surgery (W.G.M.), Vanderbilt University Medical Center, Nashville, TN; Maine Medical Center Research Institute, Scarborough (S.R.); Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati, OH (K.N., T.B.T.); CentraCare Health, St. Cloud, MN (J.A.S.); Cincinnati Children's Hospital Medical Center, OH (A.M.); Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt (M.A.S.); and Center for Stem Cell Biology, Vanderbilt University School of Medicine, Nashville, TN (M.A.M.)
| | - Antonis K Hatzopoulos
- From the Division of Cardiovascular Medicine, Department of Medicine (L.N.S., J.A.S., A.M., R.J.G., A.K.H.), Department of Cell and Developmental Biology (L.N.S., A.K.H.), Division of Clinical Pharmacology, Department of Medicine (M.A.S., W.G.M., D.G.H.), and Division of General Surgery, Department of Surgery (W.G.M.), Vanderbilt University Medical Center, Nashville, TN; Maine Medical Center Research Institute, Scarborough (S.R.); Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati, OH (K.N., T.B.T.); CentraCare Health, St. Cloud, MN (J.A.S.); Cincinnati Children's Hospital Medical Center, OH (A.M.); Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt (M.A.S.); and Center for Stem Cell Biology, Vanderbilt University School of Medicine, Nashville, TN (M.A.M.).
| |
Collapse
|
969
|
Dufner MC, Andre F, Stiepak J, Zelniker T, Chorianopoulos E, Preusch M, Katus HA, Leuschner F. Therapeutic hypothermia impacts leukocyte kinetics after cardiac arrest. Cardiovasc Diagn Ther 2016; 6:199-207. [PMID: 27280083 DOI: 10.21037/cdt.2016.02.06] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
BACKGROUND Patients admitted to the hospital after primarily successful cardiopulmonary resuscitation (CPR) are at a very high risk for neurologic deficits and death. Targeted temperature management (TTM) for mild therapeutic hypothermia has been shown to improve survival compared to standard treatment. Acute cardiovascular events, such as myocardial infarction (MI), are a major cause for cardiac arrest (CA) in patients who undergo CPR. Recent findings have demonstrated the importance and impact of the leukocyte response following acute MI. METHODS In this retrospective, single center study we enrolled 169 patients with CA due to non-traumatic causes and primarily successful CPR. A total of 111 subjects (66%) underwent TTM aiming for a target temperature of 32-34 °C. RESULTS Analysis of 30 day follow up showed a significantly improved survival of all patients who received TTM compared to patients without hypothermia (P=0.0001). Furthermore TTM was an independent variable of good neurological outcome after 6 months (P=0.0030). Therapeutic hypothermia was found to be beneficial independent of differences in age and sex between both groups. While a higher rate of pneumonia was observed with TTM, this diagnosis had no additional impact on survival or neurological outcome. The beneficial effect on mortality remained significant in patients with the diagnosis of an acute cardiac event (P=0.0145). Next, we evaluated the kinetics of leukocytes in this group over the course of 7 days after CA. At presentation, patients showed a mean level of 16.5±6.7 of leukocytes per microliter. While this level stayed stable in the group of patients without hypothermia, patients who received TTM showed a significant decline of leukocyte levels resulting in significantly lower numbers of leukocytes on days 3 and 5 after CPR. Interestingly, these differences in leukocyte counts remained beyond the time period of TTM while C-reactive protein (CRP) levels were suppressed only during ongoing cooling, but differences between the groups were diminished after TTM was terminated (from day 3 on, P>0.2). Finally, patients who received TTM and showed a leukocyte count of less than 12.7/µL on day 3 had an improved survival (P=0.0214) and neurological outcome (P=0.0049) compared to patients above that level. CONCLUSIONS Our data underline the beneficial effects of TTM and demonstrate an impact of hypothermia on leukocyte counts after CA.
Collapse
Affiliation(s)
- Matthias C Dufner
- Department of Internal Medicine III, University of Heidelberg, 69120 Heidelberg, Germany ; DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Heidelberg, Germany
| | - Florian Andre
- Department of Internal Medicine III, University of Heidelberg, 69120 Heidelberg, Germany ; DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Heidelberg, Germany
| | - Jan Stiepak
- Department of Internal Medicine III, University of Heidelberg, 69120 Heidelberg, Germany ; DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Heidelberg, Germany
| | - Thomas Zelniker
- Department of Internal Medicine III, University of Heidelberg, 69120 Heidelberg, Germany ; DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Heidelberg, Germany
| | - Emmanuel Chorianopoulos
- Department of Internal Medicine III, University of Heidelberg, 69120 Heidelberg, Germany ; DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Heidelberg, Germany
| | - Michael Preusch
- Department of Internal Medicine III, University of Heidelberg, 69120 Heidelberg, Germany ; DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Heidelberg, Germany
| | - Hugo A Katus
- Department of Internal Medicine III, University of Heidelberg, 69120 Heidelberg, Germany ; DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Heidelberg, Germany
| | - Florian Leuschner
- Department of Internal Medicine III, University of Heidelberg, 69120 Heidelberg, Germany ; DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Heidelberg, Germany
| |
Collapse
|
970
|
Lin X, Bernloehr C, Hildebrandt T, Stadler FJ, Doods H, Wu D. Kinin B1 receptor blockade and ACE inhibition attenuate cardiac postinfarction remodeling and heart failure in rats. Toxicol Appl Pharmacol 2016; 305:153-160. [PMID: 27288733 DOI: 10.1016/j.taap.2016.06.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Revised: 05/18/2016] [Accepted: 06/06/2016] [Indexed: 10/21/2022]
Abstract
INTRODUCTION The aim of the present study was to evaluate the effects of the novel kinin B1 receptor antagonist BI113823 on postinfarction cardiac remodeling and heart failure, and to determine whether B1 receptor blockade alters the cardiovascular effects of an angiotensin 1 converting enzyme (ACE) inhibitor in rats. METHODS AND RESULTS Sprague Dawley rats were subjected to permanent occlusion of the left coronary artery. Cardiovascular function was determined at 6weeks postinfarction. Treatment with either B1 receptor antagonist (BI113823) or an ACE inhibitor (lisinopril) alone or in combination significantly reduced the heart weight-to-body weight and lung weight-to-body weight ratios, and improved postinfarction cardiac function as evidenced by greater cardiac output, the maximum rate of left ventricular pressure rise (±dP/dtmax), left ventricle ejection fraction, fractional shorting, better wall motion, and attenuation of elevated left ventricular end diastolic pressure (LVEDP). Furthermore, all three treatment groups exhibited significant reduction in cardiac interstitial fibrosis, collagen deposition, CD68 positive macrophages, neutrophils, and proinflammatory cytokine production (TNF-α and IL-1β), compared to vehicle controls. CONCLUSION The present study shows that treatment with the novel kinin B1 receptor antagonist, BI113823, reduces postinfarction cardiac remodeling and heart failure, and does not influence the cardiovascular effects of the ACE inhibitor.
Collapse
Affiliation(s)
- Xinchun Lin
- Department of Research, Mount Sinai Medical Center, Miami Beach, FL 33140, USA
| | | | | | - Florian J Stadler
- Shenzhen Engineering Laboratory for Advanced Technology of Ceramics, Shenzhen 518060, PR China.
| | - Henri Doods
- Boehringer Ingelheim Pharma GmbH & Co.KG, Biberach, Germany
| | - Dongmei Wu
- Department of Research, Mount Sinai Medical Center, Miami Beach, FL 33140, USA; Department of BIN Convergence Technology, Chonbuk National University, South Korea.
| |
Collapse
|
971
|
Ferrari R, Malagù M, Biscaglia S, Fucili A, Rizzo P. Remodelling after an Infarct: Crosstalk between Life and Death. Cardiology 2016; 135:68-76. [DOI: 10.1159/000445882] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 03/29/2016] [Indexed: 11/19/2022]
|
972
|
Hervas A, Ruiz-Sauri A, Gavara J, Monmeneu JV, de Dios E, Rios-Navarro C, Perez-Sole N, Perez I, Monleon D, Morales JM, Minana G, Nunez J, Bonanad C, Diaz A, Vila JM, Chorro FJ, Bodi V. A Multidisciplinary Assessment of Remote Myocardial Fibrosis After Reperfused Myocardial Infarction in Swine and Patients. J Cardiovasc Transl Res 2016; 9:321-33. [DOI: 10.1007/s12265-016-9698-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 05/16/2016] [Indexed: 12/20/2022]
|
973
|
Cheow ESH, Cheng WC, Lee CN, de Kleijn D, Sorokin V, Sze SK. Plasma-derived Extracellular Vesicles Contain Predictive Biomarkers and Potential Therapeutic Targets for Myocardial Ischemic (MI) Injury. Mol Cell Proteomics 2016; 15:2628-40. [PMID: 27234505 DOI: 10.1074/mcp.m115.055731] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Indexed: 12/21/2022] Open
Abstract
Myocardial infarction (MI) triggers a potent inflammatory response via the release of circulatory mediators, including extracellular vesicles (EVs) by damaged cardiac cells, necessary for myocardial healing. Timely repression of inflammatory response are critical to prevent and minimize cardiac tissue injuries, nonetheless, progression in this aspect remains challenging. The ability of EVs to trigger a functional response upon delivery of carried bioactive cargos, have made them clinically attractive diagnostic biomarkers and vectors for therapeutic interventions. Using label-free quantitative proteomics approach, we compared the protein cargo of plasma EVs between patients with MI and from patients with stable angina (NMI). We report, for the first time, the proteomics profiling on 252 EV proteins that were modulated with >1.2-fold after MI. We identified six up-regulated biomarkers with potential for clinical applications; these reflected post-infarct pathways of complement activation (Complement C1q subcomponent subunit A (C1QA), 3.23-fold change, p = 0.012; Complement C5 (C5), 1.27-fold change, p = 0.087), lipoprotein metabolism (Apoliporotein D (APOD), 1.86-fold change, p = 0.033; Apolipoprotein C-III (APOCC3), 2.63-fold change, p = 0.029) and platelet activation (Platelet glycoprotein Ib alpha chain (GP1BA), 9.18-fold change, p < 0.0001; Platelet basic protein (PPBP), 4.72-fold change, p = 0.027). The data have been deposited to the ProteomeXchange with identifier PXD002950. This novel biomarker panel was validated in 43 patients using antibody-based assays (C1QA (p = 0.005); C5 (p = 0.0047), APOD (p = 0.0267); APOC3 (p = 0.0064); GP1BA (p = 0.0031); PPBP (p = 0.0465)). We further present that EV-derived fibrinogen components were paradoxically down-regulated in MI, suggesting that a compensatory mechanism may suppress post-infarct coagulation pathways, indicating potential for therapeutic targeting of this mechanism in MI. Taken together, these data demonstrated that plasma EVs contain novel diagnostic biomarkers and therapeutic targets that can be further developed for clinical use to benefit patients with coronary artery diseases (CADs).
Collapse
Affiliation(s)
- Esther Sok Hwee Cheow
- From the ‡School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551
| | - Woo Chin Cheng
- §Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, and Cardiovascular Research Institute, Singapore 119228
| | - Chuen Neng Lee
- §Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, and Cardiovascular Research Institute, Singapore 119228; ¶National University Heart Centre, Department of Cardiac, Thoracic & Vascular Surgery, Singapore 119228; ‖Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228
| | - Dominique de Kleijn
- §Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, and Cardiovascular Research Institute, Singapore 119228; **Experimental Cardiology Laboratory, Cardiology, University Medical Center Utrecht, the Netherlands & Interuniversity Cardiovascular Institute of the Netherlands, Utrecht, the Netherlands
| | - Vitaly Sorokin
- §Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, and Cardiovascular Research Institute, Singapore 119228; ¶National University Heart Centre, Department of Cardiac, Thoracic & Vascular Surgery, Singapore 119228
| | - Siu Kwan Sze
- From the ‡School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551;
| |
Collapse
|
974
|
Abstract
The immune system is responsible for defending an organism against the myriad of microbial invaders it constantly confronts. It has become increasingly clear that the immune system has a second major function: the maintenance of organismal homeostasis. Foxp3(+)CD4(+) regulatory T cells (Tregs) are important contributors to both of these critical activities, defense being the primary purview of Tregs circulating through lymphoid organs, and homeostasis ensured mainly by their counterparts residing in parenchymal tissues. This review focuses on so-called tissue Tregs. We first survey existing information on the phenotype, function, sustaining factors, and human equivalents of the three best-characterized tissue-Treg populations-those operating in visceral adipose tissue, skeletal muscle, and the colonic lamina propria. We then attempt to distill general principles from this body of work-as concerns the provenance, local adaptation, molecular sustenance, and targets of action of tissue Tregs, in particular.
Collapse
Affiliation(s)
- Marisella Panduro
- Division of Immunology, Department of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts 02115; , ,
- Evergrande Center for Immunologic Diseases, Harvard Medical School, Boston, Massachusetts 02115
- Brigham and Women's Hospital, Boston, Massachusetts 02115
| | - Christophe Benoist
- Division of Immunology, Department of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts 02115; , ,
- Evergrande Center for Immunologic Diseases, Harvard Medical School, Boston, Massachusetts 02115
- Brigham and Women's Hospital, Boston, Massachusetts 02115
| | - Diane Mathis
- Division of Immunology, Department of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts 02115; , ,
- Evergrande Center for Immunologic Diseases, Harvard Medical School, Boston, Massachusetts 02115
- Brigham and Women's Hospital, Boston, Massachusetts 02115
| |
Collapse
|
975
|
Extracellular mtDNA activates NF-κB via toll-like receptor 9 and induces cell death in cardiomyocytes. Basic Res Cardiol 2016; 111:42. [PMID: 27164906 DOI: 10.1007/s00395-016-0553-6] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Accepted: 04/05/2016] [Indexed: 12/31/2022]
Abstract
Acute myocardial infarction (AMI) causes sterile inflammation, which exacerbates tissue injury. Elevated levels of circulating mitochondrial DNA (mtDNA) have been associated with AMI. We hypothesized that mtDNA triggers an innate immune response via TLR9 and NF-κB activation, causing cardiomyocyte injury. Murine cardiomyocytes express TLR9 mRNA and protein and were able to internalize fluorescently labeled mouse mtDNA. Incubation of human embryonic kidney cells with serum from AMI patients containing naturally elevated levels of mtDNA induced TLR9-dependent NF-κB activity. This effect was mimicked by isolated mtDNA. mtDNA activated NF-κB in reporter mice both in vivo and in isolated cardiomyocytes. Moreover, incubation of isolated cardiomyocytes with mtDNA induced cell death after 4 and 24 h. Laser confocal microscopy showed that incubation of cardiomyocytes with mtDNA accelerated mitochondrial depolarization induced by reactive oxygen species. In contrast to mtDNA, isolated total DNA did not activate NF-κB nor induce cell death. In conclusion, mtDNA can induce TLR9-dependent NF-κB activation in reporter cells and activate NF-κB in cardiomyocytes. In cardiomyocytes, mtDNA causes mitochondrial dysfunction and death. Endogenous mtDNA in the extracellular space is a danger signal with direct detrimental effects on cardiomyocytes.
Collapse
|
976
|
Yang Q, Cui J, Wang P, Du X, Wang W, Zhang T, Chen Y. Changes in interconnected pathways implicating microRNAs are associated with the activity of apocynin in attenuating myocardial fibrogenesis. Eur J Pharmacol 2016; 784:22-32. [PMID: 27174579 DOI: 10.1016/j.ejphar.2016.05.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Revised: 05/03/2016] [Accepted: 05/04/2016] [Indexed: 12/11/2022]
Abstract
Myocardial fibrosis is the endpoint pathology common to many cardiovascular disorders. We have previously shown that apocynin (APO), a naturally occurring NADPH oxidase inhibitor, significantly prevents the development of isoproterenol (ISO)-induced myocardial injury and fibrogenesis. The current study investigated the changes in microRNAs (miRNAs) and their potential implication in the cardioprotective effects of APO. Integrative analyses of whole-genome miRNA and gene expression profiles were first performed, revealing that altered expression of miRNAs likely contributed to dysregulated expression of genes associated with multiple interconnected fibrogenic signaling pathways. Importantly, APO treatment exhibited a broad impact on these signaling pathways, which could in part be mediated through miRNA-mediated gene expression regulation. The expression of differentially expressed miRNAs was further validated by real-time PCR analyses. Consistent with the data from miRNA array, compared to that from vehicle-treated normal controls, significantly decreased expression of miR-10b, miR-29c*, miR-30c-1*, miR-30e*, miR-148b, miR-181d, miR-218 and miR-3107* was observed in ISO-challenged vehicle-treated mouse hearts. In contrast, significantly increased expression of these miRNAs was observed in ISO-challenged APO-treated hearts compared to that from ISO-challenged vehicle-treated mice. Moreover, increased expression of miR-21 was observed as a result of ISO administration, which was significantly reduced by APO treatment. Altered protein levels of Col1, TIMP1, Rac2 and gp91(phox) were also validated. Lastly, APO treatment was shown to attenuate pre-established myocardial fibrosis induced by ISO. The results therefore demonstrated for the first time that complex changes in miRNA-mRNA interactome network are associated with the protective effects of APO against ISO-induced myocardial injury and fibrogenesis.
Collapse
Affiliation(s)
- Qinbo Yang
- Clinical Research Institute of Integrative Medicine and Yueyang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Jingang Cui
- Clinical Research Institute of Integrative Medicine and Yueyang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Peiwei Wang
- Clinical Research Institute of Integrative Medicine and Yueyang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Xiaoye Du
- Clinical Research Institute of Integrative Medicine and Yueyang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Wenjian Wang
- Clinical Research Institute of Integrative Medicine and Yueyang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Teng Zhang
- Clinical Research Institute of Integrative Medicine and Yueyang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China.
| | - Yu Chen
- Clinical Research Institute of Integrative Medicine and Yueyang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China.
| |
Collapse
|
977
|
Fukushima S, Miyagawa S, Sakai Y, Sawa Y. A sustained-release drug-delivery system of synthetic prostacyclin agonist, ONO-1301SR: a new reagent to enhance cardiac tissue salvage and/or regeneration in the damaged heart. Heart Fail Rev 2016; 20:401-13. [PMID: 25708182 PMCID: PMC4464640 DOI: 10.1007/s10741-015-9477-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Cardiac failure is a major cause of mortality and morbidity worldwide, since the standard treatment for cardiac failure in the clinical practice is chiefly to focus on removal of insults against the heart or minimisation of additional factors to exacerbate cardiac failure, but not on regeneration of the damaged cardiac tissue. A synthetic prostacyclin agonist, ONO-1301, has been developed as a long-acting drug for acute and chronic pathologies related to regional ischaemia, inflammation and/or interstitial fibrosis by pre-clinical studies. In addition, poly-lactic co-glycolic acid-polymerised form of ONO-1301, ONO-1301SR, was generated to achieve a further sustained release of this drug into the targeted region. This unique reagent has been shown to act on fibroblasts, vascular smooth muscle cells and endothelial cells in the tissue via the prostaglandin IP receptor to exert paracrinal release of multiple protective factors, such as hepatocyte growth factor, vascular endothelial growth factor or stromal cell-derived factor-1, into the adjacent damaged tissue, which is salvaged and/or regenerated as a result. Our laboratory developed a new surgical approach to treat acute and chronic cardiac failure using a variety of animal models, in which ONO-1301SR is directly placed over the cardiac surface to maximise the therapeutic effects and minimise the systemic complications. This review summarises basic and pre-clinical information of ONO-1301 and ONO-1301SR as a new reagent to enhance tissue salvage and/or regeneration, with a particular focus on the therapeutic effects on acute and chronic cardiac failure and underlying mechanisms, to explore a potential in launching the clinical study.
Collapse
Affiliation(s)
- Satsuki Fukushima
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan,
| | | | | | | |
Collapse
|
978
|
de Almeida Salles T, Zogbi C, de Lima TM, de Godoi Carneiro C, Garcez AT, Barbeiro HV, Antonio EL, Dos Santos L, da Costa Pereira A, Tucci PJF, de Paula Faria D, Soriano FG, Girardi ACC. The contributions of dipeptidyl peptidase IV to inflammation in heart failure. Am J Physiol Heart Circ Physiol 2016; 310:H1760-72. [PMID: 27199127 DOI: 10.1152/ajpheart.00735.2015] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 04/12/2016] [Indexed: 01/14/2023]
Abstract
Circulating dipeptidyl peptidase IV (DPPIV) activity correlates with cardiac dysfunction in humans and experimental heart failure (HF) models. Similarly, inflammatory markers are associated with poorer outcomes in HF patients. However, the contributions of DPPIV to inflammation in HF remain elusive. Therefore, this study aimed to investigate whether the cardioprotective effects of DPPIV inhibition after myocardial injury are accompanied by reduced cardiac inflammation, whether circulating DPPIV activity correlates with the levels of systemic inflammatory markers in HF patients, and whether leukocytes and/or splenocytes may be one of the sources of circulating DPPIV in HF. Experimental HF was induced in male Wistar rats by left ventricular myocardial injury after radiofrequency catheter ablation. The rats were divided into three groups: sham, HF, and HF + DPPIV inhibitor (sitagliptin). Six weeks after surgery, cardiac function, perfusion and inflammatory status were evaluated. Sitagliptin treatment improved cardiac function and perfusion, reduced macrophage infiltration, and diminished the levels of inflammatory biomarkers including TNF-α, IL-1β, and CCL2. In HF patients, serum DPPIV activity correlated with CCL2, suggesting that leukocytes may be the source of circulating DPPIV in HF. Unexpectedly, DPPIV release was higher in splenocytes from HF rats and similar in HF circulating mononuclear cells compared with those from sham, suggesting an organ-specific modulation of DPPIV in HF. Collectively, our data provide new evidence that the cardioprotective effects of DPPIV inhibition in HF may be due to suppression of inflammatory cytokines. Moreover, they suggest that a vicious circle between DPPIV and inflammation may contribute to HF development and progression.
Collapse
Affiliation(s)
| | - Camila Zogbi
- Heart Institute (InCor), University of São Paulo Medical School, São Paulo, Brazil
| | - Thais Martins de Lima
- Department of Clinical Medicine,University of São Paulo Medical School, São Paulo, Brazil
| | - Camila de Godoi Carneiro
- Laboratory of Nuclear Medicine (LIM 43), University of São Paulo Medical School, São Paulo, Brazil
| | - Alexandre Teles Garcez
- Laboratory of Nuclear Medicine (LIM 43), University of São Paulo Medical School, São Paulo, Brazil
| | - Hermes Vieira Barbeiro
- Department of Clinical Medicine,University of São Paulo Medical School, São Paulo, Brazil
| | - Ednei Luiz Antonio
- Department of Physiology, Federal University of São Paulo, São Paulo, Brazil; and
| | - Leonardo Dos Santos
- Department of Physiological Sciences, Federal University of Espírito Santo, Vitoria, Espírito Santo, Brazil
| | | | | | - Daniele de Paula Faria
- Laboratory of Nuclear Medicine (LIM 43), University of São Paulo Medical School, São Paulo, Brazil
| | | | | |
Collapse
|
979
|
Kaur H, Takefuji M, Ngai CY, Carvalho J, Bayer J, Wietelmann A, Poetsch A, Hoelper S, Conway SJ, Möllmann H, Looso M, Troidl C, Offermanns S, Wettschureck N. Targeted Ablation of Periostin-Expressing Activated Fibroblasts Prevents Adverse Cardiac Remodeling in Mice. Circ Res 2016; 118:1906-17. [PMID: 27140435 DOI: 10.1161/circresaha.116.308643] [Citation(s) in RCA: 187] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
RATIONALE Activated cardiac fibroblasts (CF) are crucial players in the cardiac damage response; excess fibrosis, however, may result in myocardial stiffening and heart failure development. Inhibition of activated CF has been suggested as a therapeutic strategy in cardiac disease, but whether this truly improves cardiac function is unclear. OBJECTIVE To study the effect of CF ablation on cardiac remodeling. METHODS AND RESULTS We characterized subgroups of murine CF by single-cell expression analysis and identified periostin as the marker showing the highest correlation to an activated CF phenotype. We generated bacterial artificial chromosome-transgenic mice allowing tamoxifen-inducible Cre expression in periostin-positive cells as well as their diphtheria toxin-mediated ablation. In the healthy heart, periostin expression was restricted to valvular fibroblasts; ablation of this population did not affect cardiac function. After chronic angiotensin II exposure, ablation of activated CF resulted in significantly reduced cardiac fibrosis and improved cardiac function. After myocardial infarction, ablation of periostin-expressing CF resulted in reduced fibrosis without compromising scar stability, and cardiac function was significantly improved. Single-cell transcriptional analysis revealed reduced CF activation but increased expression of prohypertrophic factors in cardiac macrophages and cardiomyocytes, resulting in localized cardiomyocyte hypertrophy. CONCLUSIONS Modulation of the activated CF population is a promising approach to prevent adverse cardiac remodeling in response to angiotensin II and after myocardial infarction.
Collapse
Affiliation(s)
- Harmandeep Kaur
- From the Department of Pharmacology (H.K., C.Y.N., J.C., S.O., N.W.), Bioinformatics Facility (J.B., M.L.), Nuclear Magnetic Resonance Imaging Facility (A.W.), and Mass Spectrometry Group (A.P., S.H.), Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan (M.T.); Department of Pediatrics, Indiana University School of Medicine, Indianapolis (S.J.C.); Department of Cardiology, Kerckhoff Heart and Thorax Center, Bad Nauheim, Germany (H.M., C.T.); and Medical Faculty, J.W. Goethe University Frankfurt, Frankfurt, Germany (S.O., N.W.)
| | - Mikito Takefuji
- From the Department of Pharmacology (H.K., C.Y.N., J.C., S.O., N.W.), Bioinformatics Facility (J.B., M.L.), Nuclear Magnetic Resonance Imaging Facility (A.W.), and Mass Spectrometry Group (A.P., S.H.), Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan (M.T.); Department of Pediatrics, Indiana University School of Medicine, Indianapolis (S.J.C.); Department of Cardiology, Kerckhoff Heart and Thorax Center, Bad Nauheim, Germany (H.M., C.T.); and Medical Faculty, J.W. Goethe University Frankfurt, Frankfurt, Germany (S.O., N.W.)
| | - C Y Ngai
- From the Department of Pharmacology (H.K., C.Y.N., J.C., S.O., N.W.), Bioinformatics Facility (J.B., M.L.), Nuclear Magnetic Resonance Imaging Facility (A.W.), and Mass Spectrometry Group (A.P., S.H.), Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan (M.T.); Department of Pediatrics, Indiana University School of Medicine, Indianapolis (S.J.C.); Department of Cardiology, Kerckhoff Heart and Thorax Center, Bad Nauheim, Germany (H.M., C.T.); and Medical Faculty, J.W. Goethe University Frankfurt, Frankfurt, Germany (S.O., N.W.)
| | - Jorge Carvalho
- From the Department of Pharmacology (H.K., C.Y.N., J.C., S.O., N.W.), Bioinformatics Facility (J.B., M.L.), Nuclear Magnetic Resonance Imaging Facility (A.W.), and Mass Spectrometry Group (A.P., S.H.), Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan (M.T.); Department of Pediatrics, Indiana University School of Medicine, Indianapolis (S.J.C.); Department of Cardiology, Kerckhoff Heart and Thorax Center, Bad Nauheim, Germany (H.M., C.T.); and Medical Faculty, J.W. Goethe University Frankfurt, Frankfurt, Germany (S.O., N.W.)
| | - Julia Bayer
- From the Department of Pharmacology (H.K., C.Y.N., J.C., S.O., N.W.), Bioinformatics Facility (J.B., M.L.), Nuclear Magnetic Resonance Imaging Facility (A.W.), and Mass Spectrometry Group (A.P., S.H.), Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan (M.T.); Department of Pediatrics, Indiana University School of Medicine, Indianapolis (S.J.C.); Department of Cardiology, Kerckhoff Heart and Thorax Center, Bad Nauheim, Germany (H.M., C.T.); and Medical Faculty, J.W. Goethe University Frankfurt, Frankfurt, Germany (S.O., N.W.)
| | - Astrid Wietelmann
- From the Department of Pharmacology (H.K., C.Y.N., J.C., S.O., N.W.), Bioinformatics Facility (J.B., M.L.), Nuclear Magnetic Resonance Imaging Facility (A.W.), and Mass Spectrometry Group (A.P., S.H.), Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan (M.T.); Department of Pediatrics, Indiana University School of Medicine, Indianapolis (S.J.C.); Department of Cardiology, Kerckhoff Heart and Thorax Center, Bad Nauheim, Germany (H.M., C.T.); and Medical Faculty, J.W. Goethe University Frankfurt, Frankfurt, Germany (S.O., N.W.)
| | - Ansgar Poetsch
- From the Department of Pharmacology (H.K., C.Y.N., J.C., S.O., N.W.), Bioinformatics Facility (J.B., M.L.), Nuclear Magnetic Resonance Imaging Facility (A.W.), and Mass Spectrometry Group (A.P., S.H.), Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan (M.T.); Department of Pediatrics, Indiana University School of Medicine, Indianapolis (S.J.C.); Department of Cardiology, Kerckhoff Heart and Thorax Center, Bad Nauheim, Germany (H.M., C.T.); and Medical Faculty, J.W. Goethe University Frankfurt, Frankfurt, Germany (S.O., N.W.)
| | - Soraya Hoelper
- From the Department of Pharmacology (H.K., C.Y.N., J.C., S.O., N.W.), Bioinformatics Facility (J.B., M.L.), Nuclear Magnetic Resonance Imaging Facility (A.W.), and Mass Spectrometry Group (A.P., S.H.), Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan (M.T.); Department of Pediatrics, Indiana University School of Medicine, Indianapolis (S.J.C.); Department of Cardiology, Kerckhoff Heart and Thorax Center, Bad Nauheim, Germany (H.M., C.T.); and Medical Faculty, J.W. Goethe University Frankfurt, Frankfurt, Germany (S.O., N.W.)
| | - Simon J Conway
- From the Department of Pharmacology (H.K., C.Y.N., J.C., S.O., N.W.), Bioinformatics Facility (J.B., M.L.), Nuclear Magnetic Resonance Imaging Facility (A.W.), and Mass Spectrometry Group (A.P., S.H.), Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan (M.T.); Department of Pediatrics, Indiana University School of Medicine, Indianapolis (S.J.C.); Department of Cardiology, Kerckhoff Heart and Thorax Center, Bad Nauheim, Germany (H.M., C.T.); and Medical Faculty, J.W. Goethe University Frankfurt, Frankfurt, Germany (S.O., N.W.)
| | - Helge Möllmann
- From the Department of Pharmacology (H.K., C.Y.N., J.C., S.O., N.W.), Bioinformatics Facility (J.B., M.L.), Nuclear Magnetic Resonance Imaging Facility (A.W.), and Mass Spectrometry Group (A.P., S.H.), Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan (M.T.); Department of Pediatrics, Indiana University School of Medicine, Indianapolis (S.J.C.); Department of Cardiology, Kerckhoff Heart and Thorax Center, Bad Nauheim, Germany (H.M., C.T.); and Medical Faculty, J.W. Goethe University Frankfurt, Frankfurt, Germany (S.O., N.W.)
| | - Mario Looso
- From the Department of Pharmacology (H.K., C.Y.N., J.C., S.O., N.W.), Bioinformatics Facility (J.B., M.L.), Nuclear Magnetic Resonance Imaging Facility (A.W.), and Mass Spectrometry Group (A.P., S.H.), Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan (M.T.); Department of Pediatrics, Indiana University School of Medicine, Indianapolis (S.J.C.); Department of Cardiology, Kerckhoff Heart and Thorax Center, Bad Nauheim, Germany (H.M., C.T.); and Medical Faculty, J.W. Goethe University Frankfurt, Frankfurt, Germany (S.O., N.W.)
| | - Christian Troidl
- From the Department of Pharmacology (H.K., C.Y.N., J.C., S.O., N.W.), Bioinformatics Facility (J.B., M.L.), Nuclear Magnetic Resonance Imaging Facility (A.W.), and Mass Spectrometry Group (A.P., S.H.), Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan (M.T.); Department of Pediatrics, Indiana University School of Medicine, Indianapolis (S.J.C.); Department of Cardiology, Kerckhoff Heart and Thorax Center, Bad Nauheim, Germany (H.M., C.T.); and Medical Faculty, J.W. Goethe University Frankfurt, Frankfurt, Germany (S.O., N.W.)
| | - Stefan Offermanns
- From the Department of Pharmacology (H.K., C.Y.N., J.C., S.O., N.W.), Bioinformatics Facility (J.B., M.L.), Nuclear Magnetic Resonance Imaging Facility (A.W.), and Mass Spectrometry Group (A.P., S.H.), Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan (M.T.); Department of Pediatrics, Indiana University School of Medicine, Indianapolis (S.J.C.); Department of Cardiology, Kerckhoff Heart and Thorax Center, Bad Nauheim, Germany (H.M., C.T.); and Medical Faculty, J.W. Goethe University Frankfurt, Frankfurt, Germany (S.O., N.W.)
| | - Nina Wettschureck
- From the Department of Pharmacology (H.K., C.Y.N., J.C., S.O., N.W.), Bioinformatics Facility (J.B., M.L.), Nuclear Magnetic Resonance Imaging Facility (A.W.), and Mass Spectrometry Group (A.P., S.H.), Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan (M.T.); Department of Pediatrics, Indiana University School of Medicine, Indianapolis (S.J.C.); Department of Cardiology, Kerckhoff Heart and Thorax Center, Bad Nauheim, Germany (H.M., C.T.); and Medical Faculty, J.W. Goethe University Frankfurt, Frankfurt, Germany (S.O., N.W.).
| |
Collapse
|
980
|
Peng Y, Pan W, Ou Y, Xu W, Kaelber S, Borlongan CV, Sun M, Yu G. Extracardiac-Lodged Mesenchymal Stromal Cells Propel an Inflammatory Response against Myocardial Infarction via Paracrine Effects. Cell Transplant 2016; 25:929-35. [PMID: 26498018 DOI: 10.3727/096368915x689758] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Transplantation of stem cells, including mesenchymal stromal cells (MSCs), improves the recovery of cardiac function after myocardial infarction (MI) in experimental studies using animal models and in patients. However, the improvement of cardiac function following MSC transplantation remains suboptimal in both preclinical and clinical studies. Understanding the mechanism of cell therapy may improve its therapeutic outcomes, but the mode of action mediating stem cell promotion of cardiac repair is complex and not fully understood. Recent studies suggest that the immunomodulatory effects of MSCs on the macrophage M1/M2 subtype transition allow the transplanted stem cells to inhibit inflammation-induced injury and promote cardiac repair in acute MI. However, equally compelling evidence shows that there is poor survival and minimal graft persistence of transplanted MSCs within the infarcted heart tissues, negating the view that graft survival per se is required for the observed high rate and long duration of the transition from proinflammatory M1 to reparative M2 macrophages in the infarcted myocardium. Therefore, we raised a novel hypothesis that the therapeutic effects of MSC transplantation for acute MI depends not primarily on the grafted cells in infarct myocardium, but that MSCs migrating to and being lodged in the extracardiac organs, demonstrating good graft survival and persistence, may render the therapeutic effects in MI. More specifically, MSC transplantation promotes the transition from M1 to M2 in extracardiac organs, such as spleen and bone marrow, and therapeutic effects are conferred to the infarcted myocardium via paracrine effects. In MSC transplantation, the conversion from proinflammatory M1 to anti-inflammatory M2 monocytes may occur remotely from the heart and may serve as one of the major pathways in regulating the dual effects of inflammation. This hypothesis, if proven valid, may represent an important new mechanism of action to be considered for the future of MSC transplantation in the treatment of MI.
Collapse
Affiliation(s)
- Yi Peng
- Department of Cardiology, Xiangya Hospital, Central Southern University, Changsha, Hunan, China
| | - Wei Pan
- Department of Cardiology, Xiangya Hospital, Central Southern University, Changsha, Hunan, China
| | - Yali Ou
- Department of Cardiology, Xiangya Hospital, Central Southern University, Changsha, Hunan, China
| | - Weifang Xu
- Department of Cardiology, Xiangya Hospital, Central Southern University, Changsha, Hunan, China
| | - Sussannah Kaelber
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida, Morsani College of Medicine, Tampa, FL, USA
| | - Cesario V. Borlongan
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida, Morsani College of Medicine, Tampa, FL, USA
| | - Meiqin Sun
- Department of Cardiology, Xiangya Hospital, Central Southern University, Changsha, Hunan, China
| | - Guolong Yu
- Department of Cardiology, Xiangya Hospital, Central Southern University, Changsha, Hunan, China
| |
Collapse
|
981
|
Westman PC, Lipinski MJ, Luger D, Waksman R, Bonow RO, Wu E, Epstein SE. Inflammation as a Driver of Adverse Left Ventricular Remodeling After Acute Myocardial Infarction. J Am Coll Cardiol 2016; 67:2050-60. [DOI: 10.1016/j.jacc.2016.01.073] [Citation(s) in RCA: 218] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Revised: 01/20/2016] [Accepted: 01/26/2016] [Indexed: 12/18/2022]
|
982
|
Zhou X, Liu XL, Ji WJ, Liu JX, Guo ZZ, Ren D, Ma YQ, Zeng S, Xu ZW, Li HX, Wang PP, Zhang Z, Li YM, Benefield BC, Zawada AM, Thorp EB, Lee DC, Heine GH. The Kinetics of Circulating Monocyte Subsets and Monocyte-Platelet Aggregates in the Acute Phase of ST-Elevation Myocardial Infarction: Associations with 2-Year Cardiovascular Events. Medicine (Baltimore) 2016; 95:e3466. [PMID: 27149446 PMCID: PMC4863763 DOI: 10.1097/md.0000000000003466] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
In experimental myocardial infarction (MI), a rise in cell counts of circulating monocyte subsets contributes to impaired myocardial healing and to atherosclerotic plaque destabilization. In humans, the prognostic role of monocyte subsets in patients suffering ST-elevation MI (STEMI) is still unclear. In the present study, we aimed to determine the kinetics of the 3 monocyte subsets (classical CD14++CD16-, intermediate CD14++CD16+, and nonclassical CD14+CD16++ monocytes), as well as the subset-specific monocyte-platelet aggregates (MPA), in acute STEMI followed by primary percutaneous coronary intervention (PCI), and their relationships with cardiovascular outcomes during a 2-year follow-up.Monocyte subsets and MPA were measured in 100 STEMI patients receiving primary PCI on days 1, 2, 3, 5, and 7 of symptom onset, which were compared with 60 stable coronary heart disease patients and 35 healthy volunteers. From day 1 to day 7, significant increases in the counts of CD14++CD16+ monocytes and CD14++CD16+ MPA were observed, with peak levels on day 2. During a median follow-up of 2.0 years, 28 first cardiovascular events (defined as cardiovascular death, nonfatal ischemic stroke, recurrent MI, need for emergency or repeat revascularization, and rehospitalization for heart failure) were recorded. After adjustment for confounders, CD14++CD16+ monocytosis (day 1 [HR: 3.428; 95% CI: 1.597-7.358; P = 0.002], day 2 [HR: 4.835; 95% CI: 1.106-21.13; P = 0.04], day 3 [HR: 2.734; 95% CI: 1.138-6.564; P = 0.02], and day 7 [HR: 2.647; 95% CI: 1.196-5.861; P = 0.02]), as well as increased levels of CD14++CD16+ MPA measured on all time points (days 1, 2, 3, 5, and 7), had predictive values for adverse cardiovascular events.In conclusion, our data show the expansion of the CD14++CD16+ monocyte subset during acute phase of STEMI has predictive values for 2-year adverse cardiovascular outcomes in patients treated with primary PCI. Future studies will be warranted to elucidate whether CD14++CD16+ monocytes may become a target cell population for new therapeutic strategies after STEMI.
Collapse
Affiliation(s)
- Xin Zhou
- From the Tianjin Key Laboratory of Cardiovascular Remodeling and Target Organ Injury (XZ, X-LL, W-JJ, J-XL, Z-ZG, DR, Y-QM, SZ, Z-WX, H-XL, Y-ML), Pingjin Hospital Heart Center, Logistics University of Chinese People's Armed Police Forces, Tianjin, China; Division of Community Health and Humanities (PPW), Faculty of Medicine, Memorial University of Newfoundland, Newfoundland and Labrador, Canada; Department of Radiology (ZZ); Feinberg Cardiovascular Research Institute (BCB, DCL); Department of Pathology (EBT), Northwestern University Feinberg School of Medicine, Chicago, IL, USA; and Department of Internal Medicine IV (AMZ and GHH), Nephrology and Hypertension, Saarland University Medical Center, Homburg, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
983
|
Anagnostopoulos CD, Georgakopoulos A, Pianou N, Tsirebolos G, Nekolla SG, Danias PG, Rizos IA. PET/CT and CMR imaging in a patient with chest pain and unobstructed coronary vessels. J Nucl Cardiol 2016; 23:326-30. [PMID: 26153262 DOI: 10.1007/s12350-015-0191-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Accepted: 04/29/2015] [Indexed: 10/23/2022]
Affiliation(s)
- Constantinos D Anagnostopoulos
- Center for Clinical Research, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, 4 Soranou Ephessiou Street, 11527, Athens, Greece.
| | - Alexandros Georgakopoulos
- Center for Clinical Research, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, 4 Soranou Ephessiou Street, 11527, Athens, Greece
| | - Nikoletta Pianou
- Center for Clinical Research, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, 4 Soranou Ephessiou Street, 11527, Athens, Greece
| | - George Tsirebolos
- Second Department of Cardiology, Medical School, National and Kapodistrian University of Athens, General University Hospital "ATTIKON", Athens, Greece
| | - Stephan G Nekolla
- Nuklearmedizinische Klinik und Poliklinik, Klinikum rechts der Isar der Technischen Universitaet München, Munich, Germany
| | - Peter G Danias
- Cardiac MR Center, Hygeia Hospital, Athens, Greece
- Tufts University School of Medicine, Boston, MA, USA
| | - Ioannis-Antonios Rizos
- Second Department of Cardiology, Medical School, National and Kapodistrian University of Athens, General University Hospital "ATTIKON", Athens, Greece
| |
Collapse
|
984
|
Reduced acute myocardial ischemia-reperfusion injury in IL-6-deficient mice employing a closed-chest model. Inflamm Res 2016; 65:489-99. [PMID: 26935770 PMCID: PMC4841857 DOI: 10.1007/s00011-016-0931-4] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 02/18/2016] [Accepted: 02/18/2016] [Indexed: 01/04/2023] Open
Abstract
Objective and design We examined the role of IL-6 in the temporal development of cardiac ischemia–reperfusion injury employing a closed-chest I/R model. Materials/methods Infarction, local and systemic inflammation, neutrophil infiltration, coagulation and ST elevation/resolution were compared between wild-type (WT) and IL-6-deficient (IL-6−/−) mice after 1 h ischemia and 0, ½, 3, and 24 h reperfusion. Results IL-6 deficiency reduced infarct size at 3 h reperfusion (28.8 ± 4.5 % WT vs 17.6 ± 2.5 % IL-6−/−), which reduction persisted and remained similar at 24 h reperfusion (25.1 ± 3.0 % WT vs 14.6 ± 4.4 % IL-6−/−). Serum Amyloid A was reduced at 24 h reperfusion only (57.5 ± 4.9 WT vs 24.8 ± 5.6 ug/ml IL-6−/− mice). Cardiac cytokines (IL-6, IL-1β and TNFα) peaked at 3 h reperfusion, but IL-1β and TNFα levels were unaffected by IL-6 deficiency. Significant neutrophil influx was only detected at 24 h reperfusion and was similar for WT and IL-6−/−. Tissue factor peaked at 24 h reperfusion, whereas fibrin/fibrinogen peaked at 3 h reperfusion and was completely resolved at 24 h reperfusion; both coagulation factors were unaltered by IL-6 deficiency. Prolonged ST elevation was observed during ischemia that completely resolved for both genotypes at early reperfusion. Conclusions The data suggest that, in the absence of major surgical intervention, IL-6 contributes to the development of infarct size in the early phase of reperfusion; this contribution did not depend on neutrophil influx, IL-1β and TNFα, tissue factor and fibrin.
Collapse
|
985
|
Zuo D, Subjeck J, Wang XY. Unfolding the Role of Large Heat Shock Proteins: New Insights and Therapeutic Implications. Front Immunol 2016; 7:75. [PMID: 26973652 PMCID: PMC4771732 DOI: 10.3389/fimmu.2016.00075] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Accepted: 02/15/2016] [Indexed: 11/13/2022] Open
Abstract
Heat shock proteins (HSPs) of eukaryotes are evolutionarily conserved molecules present in all the major intracellular organelles. They mainly function as molecular chaperones and participate in maintenance of protein homeostasis in physiological state and under stressful conditions. Despite their relative abundance, the large HSPs, i.e., Hsp110 and glucose-regulated protein 170 (Grp170), have received less attention compared to other conventional HSPs. These proteins are distantly related to the Hsp70 and belong to Hsp70 superfamily. Increased sizes of Hsp110 and Grp170, due to the presence of a loop structure, result in their exceptional capability in binding to polypeptide substrates or non-protein ligands, such as pathogen-associated molecules. These interactions that occur in the extracellular environment during tissue injury or microbial infection may lead to amplification of an immune response engaging both innate and adaptive immune components. Here, we review the current advances in understanding these large HSPs as molecular chaperones in proteostasis control and immune modulation as well as their therapeutic implications in treatment of cancer and neurodegeneration. Given their unique immunoregulatory activities, we also discuss the emerging evidence of their potential involvement in inflammatory and immune-related diseases.
Collapse
Affiliation(s)
- Daming Zuo
- Department of Immunology, Southern Medical University, Guangzhou, China; State Key Laboratory of Organ Failure Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - John Subjeck
- Department of Cellular Stress Biology, Roswell Park Cancer Institute , Buffalo, NY , USA
| | - Xiang-Yang Wang
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, USA; VCU Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA; VCU Institute of Molecular Medicine, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
986
|
Tao H, Yang JJ, Shi KH, Li J. Wnt signaling pathway in cardiac fibrosis: New insights and directions. Metabolism 2016; 65:30-40. [PMID: 26773927 DOI: 10.1016/j.metabol.2015.10.013] [Citation(s) in RCA: 93] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Revised: 09/19/2015] [Accepted: 10/01/2015] [Indexed: 12/15/2022]
Abstract
OBJECTIVE Wnt signaling pathway significantly participates in cardiac fibrosis and CFs activation. Therefore, we reviewed current evidence on the new perspectives and biological association between Wnt signaling pathway and cardiac fibrosis. DESIGN AND METHODS A PubMed database search was performed for studies of Wnt signaling pathway in cardiac fibrosis and CFs activation. RESULTS Numerous studies have shown that the Wnt signaling pathway significantly participates in cardiac fibrosis pathogenesis. The aim of this review is to describe the present knowledge about the Wnt signaling pathway significantly participating in cardiac fibrosis and CFs activation, and look ahead on new perspectives of Wnt signaling pathway research. Moreover, we will discuss the different insights that interact with the Wnt signaling pathway-regulated cardiac fibrosis. The Wnt proteins are glycoproteins that bind to the Fz receptors on the cell surface, which lead to several important biological functions, such as cell differentiation and proliferation. There are several signals among the characterized pathways of cardiac fibrosis, including Wnt/β-catenin signaling. In this review, new insight into the Wnt signaling pathway in cardiac fibrosis pathogenesis is discussed, with special emphasis on Wnt/β-catenin. CONCLUSION It seems reasonable to suggest the potential targets of Wnt signaling pathway and it can be developed as a therapeutic target for cardiac fibrosis.
Collapse
Affiliation(s)
- Hui Tao
- Department of Cardiothoracic Surgery, The Second Hospital of Anhui Medical University, Hefei, China 230601; Cardiovascular Research Center, Anhui Medical University, Hefei, China 230601
| | - Jing-Jing Yang
- Department of Pharmacology, The Second Hospital of Anhui Medical University, Hefei, China 230601.
| | - Kai-Hu Shi
- Department of Cardiothoracic Surgery, The Second Hospital of Anhui Medical University, Hefei, China 230601; Cardiovascular Research Center, Anhui Medical University, Hefei, China 230601.
| | - Jun Li
- School of pharmacy, Anhui Medical University, Hefei, China 230032
| |
Collapse
|
987
|
Kan X, Wu Y, Ma Y, Zhang C, Li P, Wu L, Zhang S, Li Y, Du J. Deficiency of IL-12p35 improves cardiac repair after myocardial infarction by promoting angiogenesis. Cardiovasc Res 2016; 109:249-259. [DOI: 10.1093/cvr/cvv255] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/30/2023] Open
|
988
|
Reversible cardiac hypertrophy induced by PEG-coated gold nanoparticles in mice. Sci Rep 2016; 6:20203. [PMID: 26830764 PMCID: PMC4735330 DOI: 10.1038/srep20203] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 12/23/2015] [Indexed: 11/29/2022] Open
Abstract
Gold nanoparticles (GNPs) are attracting more and more attention for their great potential value in biomedical application. Currently, no study has been reported on the chronic cardiac toxicity of GNPs after repeated administration. Here we carried out a comprehensive evaluation of the chronic cardiac toxicity of GNPs to the heart. Polyethylene glycol (PEG) -coated GNPs at three different sizes (10, 30 and 50 nm) or PBS was administrated to mice via tail vein for 14 consecutive days. Then the mice were euthanized at 2 weeks, 4 weeks or 12 weeks after the first injection. The accumulation of GNPs in the mouse heart and their effects on cardiac function, structure, fibrosis and inflammation were analysized. GNPs with smaller size showed higher accumulation and faster elimination. None of the three sizes of GNPs affected cardiac systolic function. The LVIDd (left ventricular end-diastolicinner-dimension), LVMass (left ventricular mass) and HW/BW (heart weight/body weight) were significantly increased in the mice receiving 10 nm PEG-GNPs for 2 weeks, but not for 4 weeks or 12 weeks. These results indicated that the accumulation of small size GNPs can induce reversible cardiac hypertrophy. Our results provide the basis for the further biomedical applications of GNPs in cardiac diseases.
Collapse
|
989
|
Lighthouse JK, Small EM. Transcriptional control of cardiac fibroblast plasticity. J Mol Cell Cardiol 2016; 91:52-60. [PMID: 26721596 PMCID: PMC4764462 DOI: 10.1016/j.yjmcc.2015.12.016] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Revised: 12/15/2015] [Accepted: 12/20/2015] [Indexed: 12/11/2022]
Abstract
Cardiac fibroblasts help maintain the normal architecture of the healthy heart and are responsible for scar formation and the healing response to pathological insults. Various genetic, biomechanical, or humoral factors stimulate fibroblasts to become contractile smooth muscle-like cells called myofibroblasts that secrete large amounts of extracellular matrix. Unfortunately, unchecked myofibroblast activation in heart disease leads to pathological fibrosis, which is a major risk factor for the development of cardiac arrhythmias and heart failure. A better understanding of the molecular mechanisms that control fibroblast plasticity and myofibroblast activation is essential to develop novel strategies to specifically target pathological cardiac fibrosis without disrupting the adaptive healing response. This review highlights the major transcriptional mediators of fibroblast origin and function in development and disease. The contribution of the fetal epicardial gene program will be discussed in the context of fibroblast origin in development and following injury, primarily focusing on Tcf21 and C/EBP. We will also highlight the major transcriptional regulatory axes that control fibroblast plasticity in the adult heart, including transforming growth factor β (TGFβ)/Smad signaling, the Rho/myocardin-related transcription factor (MRTF)/serum response factor (SRF) axis, and Calcineurin/transient receptor potential channel (TRP)/nuclear factor of activated T-Cell (NFAT) signaling. Finally, we will discuss recent strategies to divert the fibroblast transcriptional program in an effort to promote cardiomyocyte regeneration. This article is a part of a Special Issue entitled "Fibrosis and Myocardial Remodeling".
Collapse
Affiliation(s)
- Janet K Lighthouse
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY 14624, USA
| | - Eric M Small
- Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14624, USA; Department of Pharmacology and Physiology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14624, USA; Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY 14624, USA.
| |
Collapse
|
990
|
Ramos G, Hofmann U, Frantz S. Myocardial fibrosis seen through the lenses of T-cell biology. J Mol Cell Cardiol 2016; 92:41-5. [PMID: 26804387 DOI: 10.1016/j.yjmcc.2016.01.018] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 01/20/2016] [Accepted: 01/20/2016] [Indexed: 01/07/2023]
Abstract
Lymphocytes came recently into focus as modulators of non-infectious myocardial diseases. Several lines of experimental evidence now indicate that CD4(+) T-cells can influence the healing and scarring processes that follow a myocardial infarction episode. Furthermore, such heart-directed T-cell activity has also been implicated in the pathogenesis cardiac remodeling that develops in response to chronic pressure-overload conditions. Mechanistically, different T-cell subsets can secrete several mediators and growth factors that influence the myocardial molecular milieu and directly interfere with the macrophages' and fibroblasts' activity. Therefore, the present review summarizes the current experimental evidence on the role of T-cells in myocardial scar formation after infarction and myocardial fibrosis as central mechanism of ventricular remodeling.
Collapse
Affiliation(s)
- Gustavo Ramos
- University Clinic Halle, Department of Internal Medicine III, D-06120 Halle, Germany
| | - Ulrich Hofmann
- University Clinic Halle, Department of Internal Medicine III, D-06120 Halle, Germany; Comprehensive Heart Failure Center, Würzburg, D-97078 Würzburg, Germany
| | - Stefan Frantz
- University Clinic Halle, Department of Internal Medicine III, D-06120 Halle, Germany.
| |
Collapse
|
991
|
Ridker PM. From C-Reactive Protein to Interleukin-6 to Interleukin-1: Moving Upstream To Identify Novel Targets for Atheroprotection. Circ Res 2016; 118:145-56. [PMID: 26837745 PMCID: PMC4793711 DOI: 10.1161/circresaha.115.306656] [Citation(s) in RCA: 627] [Impact Index Per Article: 78.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Accepted: 10/01/2015] [Indexed: 12/23/2022]
Abstract
Plasma levels of the inflammatory biomarker high-sensitivity C-reactive protein (hsCRP) predict vascular risk with an effect estimate as large as that of total or high-density lipoprotein cholesterol. Further, randomized trial data addressing hsCRP have been central to understanding the anti-inflammatory effects of statin therapy and have consistently demonstrated on-treatment hsCRP levels to be as powerful a predictor of residual cardiovascular risk as on-treatment levels of low-density lipoprotein cholesterol. Yet, although hsCRP is clinically useful as a biomarker for risk prediction, most mechanistic studies suggest that CRP itself is unlikely to be a target for intervention. Moving upstream in the inflammatory cascade from CRP to interleukin (IL)-6 to IL-1 provides novel therapeutic opportunities for atheroprotection that focus on the central IL-6 signaling system and ultimately on inhibition of the IL-1β-producing NOD-like receptor family pyrin domain containing 3 inflammasome. Cholesterol crystals, neutrophil extracellular traps, atheroprone flow, and local tissue hypoxia activate the NOD-like receptor family pyrin domain containing 3 inflammasome. As such, a unifying concept of hsCRP as a downstream surrogate biomarker for upstream IL-1β activity has emerged. From a therapeutic perspective, small ischemia studies show reductions in acute-phase hsCRP production with the IL-1 receptor antagonist anakinra and the IL-6 receptor blocker tocilizumab. A phase IIb study conducted among diabetic patients at high vascular risk indicates that canakinumab, a human monoclonal antibody that targets IL-1β, markedly reduces plasma levels of IL-6, hsCRP, and fibrinogen with little change in atherogenic lipids. Canakinumab in now being tested as a method to prevent recurrent cardiovascular events in a randomized trial of 10 065 post-myocardial infarction patients with elevated hsCRP that is fully enrolled and due to complete in 2017. Clinical trials using alternative anti-inflammatory agents active against the CRP/IL-6/IL-1 axis, including low-dose methotrexate and colchicine, are being explored. If successful, these trials will close the loop on the inflammatory hypothesis of atherosclerosis and serve as examples of how fundamental biologic principles can be translated into personalized medical practice.
Collapse
Affiliation(s)
- Paul M Ridker
- From the Center for Cardiovascular Disease Prevention, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA.
| |
Collapse
|
992
|
Saxena A, Russo I, Frangogiannis NG. Inflammation as a therapeutic target in myocardial infarction: learning from past failures to meet future challenges. Transl Res 2016; 167:152-66. [PMID: 26241027 PMCID: PMC4684426 DOI: 10.1016/j.trsl.2015.07.002] [Citation(s) in RCA: 113] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Revised: 07/08/2015] [Accepted: 07/09/2015] [Indexed: 12/14/2022]
Abstract
In the infarcted myocardium, necrotic cardiomyocytes release danger signals, activating an intense inflammatory response. Inflammatory pathways play a crucial role in regulation of a wide range of cellular processes involved in injury, repair, and remodeling of the infarcted heart. Proinflammatory cytokines, such as tumor necrosis factor α and interleukin 1, are markedly upregulated in the infarcted myocardium and promote adhesive interactions between endothelial cells and leukocytes by stimulating chemokine and adhesion molecule expression. Distinct pairs of chemokines and chemokine receptors are implicated in recruitment of various leukocyte subpopulations in the infarcted myocardium. For more than the past 30 years, extensive experimental work has explored the role of inflammatory signals and the contributions of leukocyte subpopulations in myocardial infarction. Robust evidence derived from experimental models of myocardial infarction has identified inflammatory targets that may attenuate cardiomyocyte injury or protect from adverse remodeling. Unfortunately, attempts to translate the promising experimental findings to clinical therapy have failed. This review article discusses the biology of the inflammatory response after myocardial infarction, attempts to identify the causes for the translational failures of the past, and proposes promising new therapeutic directions. Because of their potential involvement in injurious, reparative, and regenerative responses, inflammatory cells may hold the key for design of new therapies in myocardial infarction.
Collapse
Affiliation(s)
- Amit Saxena
- Department of Medicine (Cardiology), The Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY
| | - Ilaria Russo
- Department of Medicine (Cardiology), The Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY
| | - Nikolaos G Frangogiannis
- Department of Medicine (Cardiology), The Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY.
| |
Collapse
|
993
|
Miranda B, Barrabés JA, Figueras J, Pineda V, Rodríguez-Palomares J, Lidón RM, Sambola A, Bañeras J, Otaegui I, García-Dorado D. Plasma bilirubin values on admission and ventricular remodeling after a first anterior ST-segment elevation acute myocardial infarction. Ann Med 2016; 48:1-9. [PMID: 26631587 DOI: 10.3109/07853890.2015.1112027] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
INTRODUCTION Bilirubin may elicit cardiovascular protection and heme oxygenase-1 overexpression attenuated post-infarction ventricular remodeling in experimental animals, but the association between bilirubin levels and post-infarction remodeling is unknown. MATERIALS AND METHODS In 145 patients with a first anterior ST-segment elevation acute myocardial infarction (STEMI), we assessed whether plasma bilirubin on admission predicted adverse remodeling (left ventricular end-diastolic volume [LVEDV] increase ≥20% between discharge and 6 months, estimated by magnetic resonance imaging). RESULTS Patients' baseline characteristics and management were comparable among bilirubin tertiles. LVEDV increased at 6 months (P < 0.001) with respect to the initial exam, but the magnitude of this increase was similar across increasing bilirubin tertiles (10.8 [30.2], 10.1 [22.9], and 12.7 [24.3]%, P = 0.500). Median (25-75 percentile) bilirubin values in patients with and without adverse remodeling were 0.75 (0.60-0.93) and 0.73 (0.60-0.92) mg/dL (P = 0.693). Absence of final TIMI flow grade 3 (odds ratio 3.92, 95% CI 1.12-13.66) and a history of hypertension (2.04, 0.93-4.50), but not admission bilirubin, were independently associated with adverse remodeling. Bilirubin also did not predict the increase in ejection fraction at 6 months. CONCLUSIONS Admission bilirubin values are not related to LVEDV or ejection fraction progression after a first anterior STEMI and do not predict adverse ventricular remodeling. Key messages Bilirubin levels are inversely related to cardiovascular disease, and overexpression of heme oxygenase-1 (the enzyme that determines bilirubin production) has prevented post-infarction ventricular remodeling in experimental animals, but the association between bilirubin levels and the progression of ventricular volumes and function in patients with acute myocardial infarction remained unexplored. In this cohort of patients with a first acute anterior ST-segment elevation myocardial infarction receiving contemporary management, bilirubin levels on admission were not predictive of the changes in left ventricular volumes or ejection fraction at 6 months measured by serial cardiac magnetic resonance imaging. The data are contrary to a significant protective effect of bilirubin against post-infarction ventricular remodeling.
Collapse
Affiliation(s)
- Berta Miranda
- a Servicio de Cardiología, Hospital Universitari Vall d'Hebron, VHIR, Universitat Autònoma de Barcelona , Barcelona , Spain
| | - José A Barrabés
- a Servicio de Cardiología, Hospital Universitari Vall d'Hebron, VHIR, Universitat Autònoma de Barcelona , Barcelona , Spain
| | - Jaume Figueras
- a Servicio de Cardiología, Hospital Universitari Vall d'Hebron, VHIR, Universitat Autònoma de Barcelona , Barcelona , Spain
| | - Victor Pineda
- b Servicio de Radiología, Hospital Universitari Vall d'Hebron, VHIR, Universitat Autònoma de Barcelona , Barcelona , Spain
| | - José Rodríguez-Palomares
- a Servicio de Cardiología, Hospital Universitari Vall d'Hebron, VHIR, Universitat Autònoma de Barcelona , Barcelona , Spain
| | - Rosa-Maria Lidón
- a Servicio de Cardiología, Hospital Universitari Vall d'Hebron, VHIR, Universitat Autònoma de Barcelona , Barcelona , Spain
| | - Antonia Sambola
- a Servicio de Cardiología, Hospital Universitari Vall d'Hebron, VHIR, Universitat Autònoma de Barcelona , Barcelona , Spain
| | - Jordi Bañeras
- a Servicio de Cardiología, Hospital Universitari Vall d'Hebron, VHIR, Universitat Autònoma de Barcelona , Barcelona , Spain
| | - Imanol Otaegui
- a Servicio de Cardiología, Hospital Universitari Vall d'Hebron, VHIR, Universitat Autònoma de Barcelona , Barcelona , Spain
| | - David García-Dorado
- a Servicio de Cardiología, Hospital Universitari Vall d'Hebron, VHIR, Universitat Autònoma de Barcelona , Barcelona , Spain
| |
Collapse
|
994
|
Abstract
PURPOSE OF REVIEW Cardiomyocyte necrosis activates an inflammatory response that serves to clear the injured myocardium from dead cells, and stimulates repair, but may also extend injury. This manuscript discusses new findings that advanced our understanding of the role of inflammation in cardiac injury and repair. RECENT FINDINGS Recently published studies have identified interleukin-1α and RNA released by necrotic cardiomyocytes as key danger signals that trigger the inflammatory response following infarction. Interleukin-1 promotes activation of a proinflammatory phenotype in leukocytes and fibroblasts, and delays myofibroblast transdifferentiation. Inhibitory lymphocytes play a crucial role in negative regulation of the postinfarction inflammatory response by modulating macrophage and fibroblast phenotype. Cardiac macrophages exhibit significant heterogeneity and phenotypic plasticity and may orchestrate the reparative response following infarction. In neonatal mice, resident embryonic macrophage subpopulations may promote a regenerative response. In contrast, in adult animals replacement of resident macrophage populations with monocyte-derived macrophages may induce inflammation while inhibiting cardiac regeneration. These exciting observations highlight the crucial role of macrophages in cardiac injury and repair, but should be interpreted with caution considering the limitations of murine models of neonatal myocardial injury. SUMMARY Design of novel strategies to reduce cardiac injury, improve repair and promote regeneration is dependent on understanding of the cell biology of the inflammatory response.
Collapse
|
995
|
The crossroads of inflammation, fibrosis, and arrhythmia following myocardial infarction. J Mol Cell Cardiol 2015; 91:114-22. [PMID: 26739214 DOI: 10.1016/j.yjmcc.2015.12.024] [Citation(s) in RCA: 172] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Revised: 12/23/2015] [Accepted: 12/26/2015] [Indexed: 01/08/2023]
Abstract
Optimal healing of damaged tissue following myocardial infarction (MI) requires a coordinated cellular response that can be divided into three phases: inflammatory, proliferative/reparative, and maturation. The inflammatory phase, characterized by rapid influx of cytokines, chemokines, and immune cells, is critical to the removal of damaged tissue. The onset of the proliferative/reparative phase is marked by increased proliferation of myofibroblasts and secretion of collagen to replace dead tissue. Lastly, crosslinking of collagen fibers and apoptosis of immune cells marks the maturation phase. Excessive inflammation or fibrosis has been linked to increased incidence of arrhythmia and other MI-related pathologies. This review describes the roles of inflammation and fibrosis in arrhythmogenesis and prospective therapies for anti-arrhythmic treatment.
Collapse
|
996
|
Alestalo K, Miettinen JA, Vuolteenaho O, Huikuri H, Lehenkari P. Bone Marrow Mononuclear Cell Transplantation Restores Inflammatory Balance of Cytokines after ST Segment Elevation Myocardial Infarction. PLoS One 2015; 10:e0145094. [PMID: 26690350 PMCID: PMC4687062 DOI: 10.1371/journal.pone.0145094] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Accepted: 11/29/2015] [Indexed: 01/10/2023] Open
Abstract
Background Acute myocardial infarction (AMI) launches an inflammatory response and a repair process to compensate cardiac function. During this process, the balance between proinflammatory and anti-inflammatory cytokines is important for optimal cardiac repair. Stem cell transplantation after AMI improves tissue repair and increases the ventricular ejection fraction. Here, we studied in detail the acute effect of bone marrow mononuclear cell (BMMNC) transplantation on proinflammatory and anti-inflammatory cytokines in patients with ST segment elevation myocardial infarction (STEMI). Methods Patients with STEMI treated with thrombolysis followed by percutaneous coronary intervention (PCI) were randomly assigned to receive either BMMNC or saline as an intracoronary injection. Cardiac function was evaluated by left ventricle angiogram during the PCI and again after 6 months. The concentrations of 27 cytokines were measured from plasma samples up to 4 days after the PCI and the intracoronary injection. Results Twenty-six patients (control group, n = 12; BMMNC group, n = 14) from the previously reported FINCELL study (n = 80) were included to this study. At day 2, the change in the proinflammatory cytokines correlated with the change in the anti-inflammatory cytokines in both groups (Kendall’s tau, control 0.6; BMMNC 0.7). At day 4, the correlation had completely disappeared in the control group but was preserved in the BMMNC group (Kendall’s tau, control 0.3; BMMNC 0.7). Conclusions BMMNC transplantation is associated with preserved balance between pro- and anti-inflammatory cytokines after STEMI in PCI-treated patients. This may partly explain the favorable effect of stem cell transplantation after AMI.
Collapse
Affiliation(s)
- Kirsi Alestalo
- Surgery Clinic, Medical Research Center, Oulu University Hospital, Oulu, Finland
- Department of Anatomy and Cell Biology, Medical Research Center, University of Oulu, Oulu, Finland
- * E-mail:
| | - Johanna A. Miettinen
- Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Olli Vuolteenaho
- Department of Physiology, Institute of Biomedicine, University of Oulu, Oulu, Finland
| | - Heikki Huikuri
- Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Petri Lehenkari
- Surgery Clinic, Medical Research Center, Oulu University Hospital, Oulu, Finland
- Department of Anatomy and Cell Biology, Medical Research Center, University of Oulu, Oulu, Finland
| |
Collapse
|
997
|
Russo I, Frangogiannis NG. Diabetes-associated cardiac fibrosis: Cellular effectors, molecular mechanisms and therapeutic opportunities. J Mol Cell Cardiol 2015; 90:84-93. [PMID: 26705059 DOI: 10.1016/j.yjmcc.2015.12.011] [Citation(s) in RCA: 326] [Impact Index Per Article: 36.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Revised: 12/13/2015] [Accepted: 12/14/2015] [Indexed: 02/07/2023]
Abstract
Both type 1 and type 2 diabetes are associated with cardiac fibrosis that may reduce myocardial compliance, contribute to the pathogenesis of heart failure, and trigger arrhythmic events. Diabetes-associated fibrosis is mediated by activated cardiac fibroblasts, but may also involve fibrogenic actions of macrophages, cardiomyocytes and vascular cells. The molecular basis responsible for cardiac fibrosis in diabetes remains poorly understood. Hyperglycemia directly activates a fibrogenic program, leading to accumulation of advanced glycation end-products (AGEs) that crosslink extracellular matrix proteins, and transduce fibrogenic signals through reactive oxygen species generation, or through activation of Receptor for AGEs (RAGE)-mediated pathways. Pro-inflammatory cytokines and chemokines may recruit fibrogenic leukocyte subsets in the cardiac interstitium. Activation of transforming growth factor-β/Smad signaling may activate fibroblasts inducing deposition of structural extracellular matrix proteins and matricellular macromolecules. Adipokines, endothelin-1 and the renin-angiotensin-aldosterone system have also been implicated in the diabetic myocardium. This manuscript reviews our current understanding of the cellular effectors and molecular pathways that mediate fibrosis in diabetes. Based on the pathophysiologic mechanism, we propose therapeutic interventions that may attenuate the diabetes-associated fibrotic response and discuss the challenges that may hamper clinical translation.
Collapse
Affiliation(s)
- Ilaria Russo
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx, NY, USA
| | - Nikolaos G Frangogiannis
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
998
|
ter Horst EN, Hakimzadeh N, van der Laan AM, Krijnen PAJ, Niessen HWM, Piek JJ. Modulators of Macrophage Polarization Influence Healing of the Infarcted Myocardium. Int J Mol Sci 2015; 16:29583-91. [PMID: 26690421 PMCID: PMC4691130 DOI: 10.3390/ijms161226187] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Revised: 11/30/2015] [Accepted: 12/01/2015] [Indexed: 12/20/2022] Open
Abstract
To diminish heart failure development after acute myocardial infarction (AMI), several preclinical studies have focused on influencing the inflammatory processes in the healing response post-AMI. The initial purpose of this healing response is to clear cell debris of the injured cardiac tissue and to eventually resolve inflammation and support scar tissue formation. This is a well-balanced reaction. However, excess inflammation can lead to infarct expansion, adverse ventricular remodeling and thereby propagate heart failure development. Different macrophage subtypes are centrally involved in both the promotion and resolution phase of inflammation. Modulation of macrophage subset polarization has been described to greatly affect the quality and outcome of healing after AMI. Therefore, it is of great interest to reveal the process of macrophage polarization to support the development of therapeutic targets. The current review summarizes (pre)clinical studies that demonstrate essential molecules involved in macrophage polarization that can be modulated and influence cardiac healing after AMI.
Collapse
Affiliation(s)
- Ellis N ter Horst
- Department of Pathology, VU University Medical Center, De Boelelaan 1117, Amsterdam 1081 HV, The Netherlands.
- Department of Cardiology, Academic Medical Center, University of Amsterdam, Meibergdreef 9, Amsterdam 1105 AZ, The Netherlands.
- Interuniversity Cardiology Institute of the Netherlands, Netherlands Heart Institute, Moreelsepark 1, Utrecht 3511 EP, The Netherlands.
- Institute for Cardiovascular Research, VU University Medical Center, van der Boechorstraat 7, Amsterdam 1081 BT, The Netherlands.
| | - Nazanin Hakimzadeh
- Department of Biomedical Engineering and Physics, Academic Medical Center, University of Amsterdam, Meibergdreef 9, Amsterdam 1105 AZ, The Netherlands.
| | - Anja M van der Laan
- Department of Cardiology, Academic Medical Center, University of Amsterdam, Meibergdreef 9, Amsterdam 1105 AZ, The Netherlands.
| | - Paul A J Krijnen
- Department of Pathology, VU University Medical Center, De Boelelaan 1117, Amsterdam 1081 HV, The Netherlands.
- Institute for Cardiovascular Research, VU University Medical Center, van der Boechorstraat 7, Amsterdam 1081 BT, The Netherlands.
| | - Hans W M Niessen
- Department of Pathology, VU University Medical Center, De Boelelaan 1117, Amsterdam 1081 HV, The Netherlands.
- Institute for Cardiovascular Research, VU University Medical Center, van der Boechorstraat 7, Amsterdam 1081 BT, The Netherlands.
- Department of Cardiac Surgery, VU University Medical Center, de Boelelaan 1117, Amsterdam 1081 HV, The Netherlands.
| | - Jan J Piek
- Department of Cardiology, Academic Medical Center, University of Amsterdam, Meibergdreef 9, Amsterdam 1105 AZ, The Netherlands.
| |
Collapse
|
999
|
Role of inflammatory cells in fibroblast activation. J Mol Cell Cardiol 2015; 93:143-8. [PMID: 26593723 DOI: 10.1016/j.yjmcc.2015.11.016] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Revised: 11/12/2015] [Accepted: 11/14/2015] [Indexed: 11/21/2022]
Abstract
Although fibrosis is an essential response to acute cardiac tissue injury, prolonged myofibroblast activation and progressive fibrosis lead to further distortion of tissue architecture and worsened cardiac function. Thus, optimal tissue repair following injury requires tight control over myofibroblast activation. It is now recognized that inflammation plays a critical role in regulating fibrosis. In this review we will highlight how advances in the field of innate immunity have led to a better understanding of the role of inflammation in cardiovascular disease and, in particular, in the regulation of fibrosis. Specifically, we will discuss how the innate immune system recognizes tissue damage in settings of acute injury and chronic cardiovascular disease. We will also review the role of different cell populations in this response, particularly the unique role of different macrophage subsets and mast cells.
Collapse
|
1000
|
Sager HB, Heidt T, Hulsmans M, Dutta P, Courties G, Sebas M, Wojtkiewicz GR, Tricot B, Iwamoto Y, Sun Y, Weissleder R, Libby P, Swirski FK, Nahrendorf M. Targeting Interleukin-1β Reduces Leukocyte Production After Acute Myocardial Infarction. Circulation 2015; 132:1880-90. [PMID: 26358260 PMCID: PMC4651795 DOI: 10.1161/circulationaha.115.016160] [Citation(s) in RCA: 198] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2015] [Accepted: 09/02/2015] [Indexed: 02/04/2023]
Abstract
BACKGROUND Myocardial infarction (MI) is an ischemic wound that recruits millions of leukocytes. MI-associated blood leukocytosis correlates inversely with patient survival, yet the signals driving heightened leukocyte production after MI remain incompletely understood. METHODS AND RESULTS With the use of parabiosis surgery, this study shows that soluble danger signals, among them interleukin-1β, increase bone marrow hematopoietic stem cell proliferation after MI. Data obtained in bone marrow reconstitution experiments reveal that interleukin-1β enhances hematopoietic stem cell proliferation by both direct actions on hematopoietic cells and through modulation of the bone marrow's hematopoietic microenvironment. An antibody that neutralizes interleukin-1β suppresses these effects. Anti-interleukin-1β treatment dampens the post-MI increase in hematopoietic stem cell proliferation. Consequently, decreased leukocyte numbers in the blood and infarct reduce inflammation and diminish post-MI heart failure in ApoE(-/-) mice with atherosclerosis. CONCLUSIONS The presented insight into post-MI bone marrow activation identifies a mechanistic target for muting inflammation in the ischemically damaged heart.
Collapse
Affiliation(s)
- Hendrik B Sager
- From Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Simches Research Building, Boston, MA (H.B.S., T.H., M.H., P.D., G.C., M.S., G.R.W., B.T., Y.I., Y.S., R.W., F.K.S., M.N.); Department of Systems Biology, Harvard Medical School, Boston, MA (R.W.); and Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA (P.L.)
| | - Timo Heidt
- From Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Simches Research Building, Boston, MA (H.B.S., T.H., M.H., P.D., G.C., M.S., G.R.W., B.T., Y.I., Y.S., R.W., F.K.S., M.N.); Department of Systems Biology, Harvard Medical School, Boston, MA (R.W.); and Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA (P.L.)
| | - Maarten Hulsmans
- From Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Simches Research Building, Boston, MA (H.B.S., T.H., M.H., P.D., G.C., M.S., G.R.W., B.T., Y.I., Y.S., R.W., F.K.S., M.N.); Department of Systems Biology, Harvard Medical School, Boston, MA (R.W.); and Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA (P.L.)
| | - Partha Dutta
- From Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Simches Research Building, Boston, MA (H.B.S., T.H., M.H., P.D., G.C., M.S., G.R.W., B.T., Y.I., Y.S., R.W., F.K.S., M.N.); Department of Systems Biology, Harvard Medical School, Boston, MA (R.W.); and Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA (P.L.)
| | - Gabriel Courties
- From Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Simches Research Building, Boston, MA (H.B.S., T.H., M.H., P.D., G.C., M.S., G.R.W., B.T., Y.I., Y.S., R.W., F.K.S., M.N.); Department of Systems Biology, Harvard Medical School, Boston, MA (R.W.); and Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA (P.L.)
| | - Matthew Sebas
- From Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Simches Research Building, Boston, MA (H.B.S., T.H., M.H., P.D., G.C., M.S., G.R.W., B.T., Y.I., Y.S., R.W., F.K.S., M.N.); Department of Systems Biology, Harvard Medical School, Boston, MA (R.W.); and Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA (P.L.)
| | - Gregory R Wojtkiewicz
- From Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Simches Research Building, Boston, MA (H.B.S., T.H., M.H., P.D., G.C., M.S., G.R.W., B.T., Y.I., Y.S., R.W., F.K.S., M.N.); Department of Systems Biology, Harvard Medical School, Boston, MA (R.W.); and Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA (P.L.)
| | - Benoit Tricot
- From Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Simches Research Building, Boston, MA (H.B.S., T.H., M.H., P.D., G.C., M.S., G.R.W., B.T., Y.I., Y.S., R.W., F.K.S., M.N.); Department of Systems Biology, Harvard Medical School, Boston, MA (R.W.); and Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA (P.L.)
| | - Yoshiko Iwamoto
- From Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Simches Research Building, Boston, MA (H.B.S., T.H., M.H., P.D., G.C., M.S., G.R.W., B.T., Y.I., Y.S., R.W., F.K.S., M.N.); Department of Systems Biology, Harvard Medical School, Boston, MA (R.W.); and Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA (P.L.)
| | - Yuan Sun
- From Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Simches Research Building, Boston, MA (H.B.S., T.H., M.H., P.D., G.C., M.S., G.R.W., B.T., Y.I., Y.S., R.W., F.K.S., M.N.); Department of Systems Biology, Harvard Medical School, Boston, MA (R.W.); and Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA (P.L.)
| | - Ralph Weissleder
- From Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Simches Research Building, Boston, MA (H.B.S., T.H., M.H., P.D., G.C., M.S., G.R.W., B.T., Y.I., Y.S., R.W., F.K.S., M.N.); Department of Systems Biology, Harvard Medical School, Boston, MA (R.W.); and Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA (P.L.)
| | - Peter Libby
- From Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Simches Research Building, Boston, MA (H.B.S., T.H., M.H., P.D., G.C., M.S., G.R.W., B.T., Y.I., Y.S., R.W., F.K.S., M.N.); Department of Systems Biology, Harvard Medical School, Boston, MA (R.W.); and Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA (P.L.)
| | - Filip K Swirski
- From Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Simches Research Building, Boston, MA (H.B.S., T.H., M.H., P.D., G.C., M.S., G.R.W., B.T., Y.I., Y.S., R.W., F.K.S., M.N.); Department of Systems Biology, Harvard Medical School, Boston, MA (R.W.); and Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA (P.L.)
| | - Matthias Nahrendorf
- From Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Simches Research Building, Boston, MA (H.B.S., T.H., M.H., P.D., G.C., M.S., G.R.W., B.T., Y.I., Y.S., R.W., F.K.S., M.N.); Department of Systems Biology, Harvard Medical School, Boston, MA (R.W.); and Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA (P.L.).
| |
Collapse
|