51
|
Lörchner H, Pöling J, Gajawada P, Hou Y, Polyakova V, Kostin S, Adrian-Segarra JM, Boettger T, Wietelmann A, Warnecke H, Richter M, Kubin T, Braun T. Myocardial healing requires Reg3β-dependent accumulation of macrophages in the ischemic heart. Nat Med 2015; 21:353-62. [PMID: 25751817 DOI: 10.1038/nm.3816] [Citation(s) in RCA: 130] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2014] [Accepted: 02/06/2015] [Indexed: 12/14/2022]
Abstract
Cardiac healing after myocardial ischemia depends on the recruitment and local expansion of myeloid cells, particularly macrophages. Here we identify Reg3β as an essential regulator of macrophage trafficking to the damaged heart. Using mass spectrometry-based secretome analysis, we found that dedifferentiating cardiomyocytes release Reg3β in response to the cytokine OSM, which signals through Jak1 and Stat3. Loss of Reg3β led to a large decrease in the number of macrophages in the ischemic heart, accompanied by increased ventricular dilatation and insufficient removal of neutrophils. This defect in neutrophil removal in turn caused enhanced matrix degradation, delayed collagen deposition and increased susceptibility to cardiac rupture. Our data indicate that OSM, acting through distinct intracellular pathways, regulates both cardiomyocyte dedifferentiation and cardiomyocyte-dependent regulation of macrophage trafficking. Release of OSM from infiltrating neutrophils and macrophages initiates a positive feedback loop in which OSM-induced production of Reg3β in cardiomyocytes attracts additional OSM-secreting macrophages. The activity of the feedback loop controls the degree of macrophage accumulation in the heart, which is instrumental in myocardial healing.
Collapse
Affiliation(s)
- Holger Lörchner
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Jochen Pöling
- 1] Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany. [2] Department of Cardiac Surgery, Schüchtermann-Clinic, Bad Rothenfelde, Germany
| | - Praveen Gajawada
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Yunlong Hou
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Viktoria Polyakova
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Sawa Kostin
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Juan M Adrian-Segarra
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Thomas Boettger
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Astrid Wietelmann
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Henning Warnecke
- Department of Cardiac Surgery, Schüchtermann-Clinic, Bad Rothenfelde, Germany
| | - Manfred Richter
- Department of Cardiac Surgery, Kerckhoff-Klinik, Bad Nauheim, Germany
| | - Thomas Kubin
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Thomas Braun
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| |
Collapse
|
52
|
Drenckhahn JD, Strasen J, Heinecke K, Langner P, Yin KV, Skole F, Hennig M, Spallek B, Fischer R, Blaschke F, Heuser A, Cox TC, Black MJ, Thierfelder L. Impaired myocardial development resulting in neonatal cardiac hypoplasia alters postnatal growth and stress response in the heart. Cardiovasc Res 2015; 106:43-54. [PMID: 25661081 DOI: 10.1093/cvr/cvv028] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
AIMS Foetal growth has been proposed to influence cardiovascular health in adulthood, a process referred to as foetal programming. Indeed, intrauterine growth restriction in animal models alters heart size and cardiomyocyte number in the perinatal period, yet the consequences for the adult or challenged heart are largely unknown. The aim of this study was to elucidate postnatal myocardial growth pattern, left ventricular function, and stress response in the adult heart after neonatal cardiac hypoplasia in mice. METHODS AND RESULTS Utilizing a new mouse model of impaired cardiac development leading to fully functional but hypoplastic hearts at birth, we show that myocardial mass is normalized until early adulthood by accelerated physiological cardiomyocyte hypertrophy. Compensatory hypertrophy, however, cannot be maintained upon ageing, resulting in reduced organ size without maladaptive myocardial remodelling. Angiotensin II stress revealed aberrant cardiomyocyte growth kinetics in adult hearts after neonatal hypoplasia compared with normally developed controls, characterized by reversible overshooting hypertrophy. This exaggerated growth mainly depends on STAT3, whose inhibition during angiotensin II treatment reduces left ventricular mass in both groups but causes contractile dysfunction in developmentally impaired hearts only. Whereas JAK/STAT3 inhibition reduces cardiomyocyte cross-sectional area in the latter, it prevents fibrosis in control hearts, indicating fundamentally different mechanisms of action. CONCLUSION Impaired prenatal development leading to neonatal cardiac hypoplasia alters postnatal cardiac growth and stress response in vivo, thereby linking foetal programming to organ size control in the heart.
Collapse
Affiliation(s)
- Jörg-Detlef Drenckhahn
- Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Straße 10, Berlin 13125, Germany
| | - Jette Strasen
- Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Straße 10, Berlin 13125, Germany
| | - Kirsten Heinecke
- Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Straße 10, Berlin 13125, Germany
| | - Patrick Langner
- Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Straße 10, Berlin 13125, Germany
| | - Kom Voy Yin
- Department of Anatomy and Developmental Biology, Monash University, Melbourne, Australia
| | - Friederike Skole
- Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Straße 10, Berlin 13125, Germany
| | - Maria Hennig
- Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Straße 10, Berlin 13125, Germany
| | - Bastian Spallek
- Experimental and Clinical Research Center, Charité Medical Faculty, Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | - Robert Fischer
- Experimental and Clinical Research Center, Charité Medical Faculty, Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | - Florian Blaschke
- Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Straße 10, Berlin 13125, Germany Charité Universitätsmedizin Berlin, Medizinische Klinik mit Schwerpunkt Kardiologie, Berlin, Germany
| | - Arnd Heuser
- Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Straße 10, Berlin 13125, Germany
| | - Timothy C Cox
- Department of Anatomy and Developmental Biology, Monash University, Melbourne, Australia Department of Pediatrics, University of Washington, Seattle, USA Center of Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, USA
| | - Mary Jane Black
- Department of Anatomy and Developmental Biology, Monash University, Melbourne, Australia
| | - Ludwig Thierfelder
- Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Straße 10, Berlin 13125, Germany
| |
Collapse
|
53
|
Akhmedov A, Montecucco F, Braunersreuther V, Camici GG, Jakob P, Reiner MF, Glanzmann M, Burger F, Paneni F, Galan K, Pelli G, Vuilleumier N, Belin A, Vallée JP, Mach F, Lüscher TF. Genetic deletion of the adaptor protein p66Shc increases susceptibility to short-term ischaemic myocardial injury via intracellular salvage pathways. Eur Heart J 2014; 36:516-26a. [PMID: 25336219 DOI: 10.1093/eurheartj/ehu400] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
AIMS Several intracellular mediators have been implicated as new therapeutic targets against myocardial ischaemia and reperfusion injury. However, clinically effective salvage pathways remain undiscovered. Here, we focused on the potential role of the adaptor protein p66(Shc) as a regulator of myocardial injury in a mouse model of cardiac ischaemia and reperfusion. METHODS AND RESULTS Adult male p66(Shc) deficient (p66(Shc) (-/-)) and C57Bl/6 wild-type (WT) mice were exposed to 30, 45, or 60 min of ischaemia and reperfusion (5, 15 min, or 24 h). Infarct size, systemic and intracardiac inflammation and oxidants, as well as cytosolic and mitochondrial apoptotic pathways were investigated. Following 30, but not 45 or 60 min of ischaemia, genetic p66(Shc) deficiency was associated with larger infarcts. In WT mice, in vivo p66(Shc) knock down by siRNA with transient protein deficiency confirmed these findings. P66(Shc) inhibition was not associated with any modification in post-infarction inflammation, oxidative burst nor cardiac vessel density or structure. However, in p66(Shc) (-/-) mice activation of the protective and anti-apoptotic Reperfusion Injury Salvage Kinases and Survivor Activating Factor Enhancement pathways were blunted and mitochondrial swelling and cellular apoptosis via the caspase-3 pathway increased compared with WT. CONCLUSIONS Genetic deletion of p66(Shc) increased susceptibility to myocardial injury in response to short-term ischaemia and reperfusion in mice. Still, additional studies are needed for assessing the role of this pathway in acute coronary syndrome patients.
Collapse
Affiliation(s)
- Alexander Akhmedov
- Center for Molecular Cardiology, Schlieren Campus, University of Zurich, Zurich, Switzerland Department of Cardiology, University Heart Center, Center for Molecular Cardiology, University Hospital and University of Zurich, Rämistrasse 100, CH-8091 Zurich, Switzerland Zurich Center of Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Fabrizio Montecucco
- Division of Cardiology, Foundation for Medical Researches, University of Geneva, Geneva, Switzerland First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa School of Medicine, Genoa, Italy IRCCS Azienda Ospedaliera Universitaria San Martino-IST Istituto Nazionale per la Ricerca sul Cancro, Genoa, Italy
| | - Vincent Braunersreuther
- Division of Cardiology, Foundation for Medical Researches, University of Geneva, Geneva, Switzerland
| | - Giovanni G Camici
- Center for Molecular Cardiology, Schlieren Campus, University of Zurich, Zurich, Switzerland Department of Cardiology, University Heart Center, Center for Molecular Cardiology, University Hospital and University of Zurich, Rämistrasse 100, CH-8091 Zurich, Switzerland Zurich Center of Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Philipp Jakob
- Center for Molecular Cardiology, Schlieren Campus, University of Zurich, Zurich, Switzerland Department of Cardiology, University Heart Center, Center for Molecular Cardiology, University Hospital and University of Zurich, Rämistrasse 100, CH-8091 Zurich, Switzerland Zurich Center of Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Martin F Reiner
- Center for Molecular Cardiology, Schlieren Campus, University of Zurich, Zurich, Switzerland Department of Cardiology, University Heart Center, Center for Molecular Cardiology, University Hospital and University of Zurich, Rämistrasse 100, CH-8091 Zurich, Switzerland Zurich Center of Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Martina Glanzmann
- Center for Molecular Cardiology, Schlieren Campus, University of Zurich, Zurich, Switzerland Department of Cardiology, University Heart Center, Center for Molecular Cardiology, University Hospital and University of Zurich, Rämistrasse 100, CH-8091 Zurich, Switzerland Zurich Center of Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Fabienne Burger
- Division of Cardiology, Foundation for Medical Researches, University of Geneva, Geneva, Switzerland First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa School of Medicine, Genoa, Italy IRCCS Azienda Ospedaliera Universitaria San Martino-IST Istituto Nazionale per la Ricerca sul Cancro, Genoa, Italy
| | - Francesco Paneni
- Center for Molecular Cardiology, Schlieren Campus, University of Zurich, Zurich, Switzerland Department of Cardiology, University Heart Center, Center for Molecular Cardiology, University Hospital and University of Zurich, Rämistrasse 100, CH-8091 Zurich, Switzerland Zurich Center of Integrative Human Physiology, University of Zurich, Zurich, Switzerland Cardiology Unit, Department of Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Katia Galan
- Division of Cardiology, Foundation for Medical Researches, University of Geneva, Geneva, Switzerland
| | - Graziano Pelli
- Division of Cardiology, Foundation for Medical Researches, University of Geneva, Geneva, Switzerland
| | - Nicolas Vuilleumier
- Division of Laboratory Medicine, Department of Genetics and Laboratory Medicine, Geneva University Hospitals, Switzerland Department of Human Protein Science, Geneva Faculty of Medicine, Geneva, Switzerland
| | - Alexandre Belin
- Department of Radiology, CIBM, Geneva University Hospital, Geneva, Switzerland
| | - Jean-Paul Vallée
- Department of Radiology, CIBM, Geneva University Hospital, Geneva, Switzerland
| | - Francois Mach
- Division of Cardiology, Foundation for Medical Researches, University of Geneva, Geneva, Switzerland
| | - Thomas F Lüscher
- Center for Molecular Cardiology, Schlieren Campus, University of Zurich, Zurich, Switzerland Department of Cardiology, University Heart Center, Center for Molecular Cardiology, University Hospital and University of Zurich, Rämistrasse 100, CH-8091 Zurich, Switzerland Zurich Center of Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| |
Collapse
|
54
|
Calduch-Giner JA, Echasseriau Y, Crespo D, Baron D, Planas JV, Prunet P, Pérez-Sánchez J. Transcriptional assessment by microarray analysis and large-scale meta-analysis of the metabolic capacity of cardiac and skeletal muscle tissues to cope with reduced nutrient availability in Gilthead Sea Bream (Sparus aurata L.). MARINE BIOTECHNOLOGY (NEW YORK, N.Y.) 2014; 16:423-435. [PMID: 24626932 DOI: 10.1007/s10126-014-9562-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2013] [Accepted: 01/06/2014] [Indexed: 06/03/2023]
Abstract
The effects of nutrient availability on the transcriptome of cardiac and skeletal muscle tissues were assessed in juvenile gilthead sea bream fed with a standard diet at two feeding levels: (1) full ration size and (2) 70 % satiation followed by a finishing phase at the maintenance ration. Microarray analysis evidenced a characteristic transcriptomic profile for each muscle tissue following changes in oxidative capacity (heart > red skeletal muscle > white skeletal muscle). The transcriptome of heart and secondly that of red skeletal muscle were highly responsive to nutritional changes, whereas that of glycolytic white skeletal muscle showed less ability to respond. The highly expressed and nutritionally regulated genes of heart were mainly related to signal transduction and transcriptional regulation. In contrast, those of white muscle were enriched in gene ontology (GO) terms related to proteolysis and protein ubiquitination. Microarray meta-analysis using the bioinformatic tool Fish and Chips ( http://fishandchips.genouest.org/index.php ) showed the close association of a representative cluster of white skeletal muscle with some of cardiac and red skeletal muscle, and many GO terms related to mitochondrial function appeared to be common links between them. A second round of cluster comparisons revealed that mitochondria-related GOs also linked differentially expressed genes of heart with those of liver from cortisol-treated gilthead sea bream. These results show that mitochondria are among the first responders to environmental and nutritional stress stimuli in gilthead sea bream, and functional phenotyping of this cellular organelle is highly promising to obtain reliable markers of growth performance and well-being in this fish species.
Collapse
Affiliation(s)
- Josep A Calduch-Giner
- Nutrigenomics and Fish Growth Endocrinology Group, Instituto de Acuicultura Torre de la Sal (IATS-CSIC), Castellón, Spain
| | | | | | | | | | | | | |
Collapse
|
55
|
Fiaschi T, Magherini F, Gamberi T, Lucchese G, Faggian G, Modesti A, Modesti PA. Hyperglycemia and angiotensin II cooperate to enhance collagen I deposition by cardiac fibroblasts through a ROS-STAT3-dependent mechanism. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2014; 1843:2603-10. [PMID: 25072659 DOI: 10.1016/j.bbamcr.2014.07.009] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Revised: 07/18/2014] [Accepted: 07/21/2014] [Indexed: 01/09/2023]
Abstract
Cardiac fibroblasts significantly contribute to diabetes-induced structural and functional changes in the myocardium. The objective of the present study was to determine the effects of high glucose (alone or supplemented with angiotensin II) in the activation of the JAK2/STAT3 pathway and its involvement in collagen I production by cardiac fibroblasts. We observed that the diabetic environment 1) enhanced tyrosine phosphorylation of JAK2 and STAT3; 2) induced nuclear localization of tyrosine phosphorylated STAT3 through a reactive oxygen species-mediated mechanism, with angiotensin II stimulation further enhancing STAT3 nuclear accumulation; and 3) stimulated collagen I production. The effects were inhibited by depletion of reactive oxygen species or silencing of STAT3 in high glucose alone or supplemented with exogenous angiotensin II. Combined, our data demonstrate that increased collagen I deposition in the setting of high glucose occurred through a reactive oxygen species- and STAT3-dependent mechanism. Our results reveal a novel role for STAT3 as a key signaling molecule of collagen I production in cardiac fibroblasts exposed to a diabetic environment.
Collapse
Affiliation(s)
- Tania Fiaschi
- Department of Biomedical, Experimental and Clinical Sciences, University of Florence, Florence, Italy
| | - Francesca Magherini
- Department of Biomedical, Experimental and Clinical Sciences, University of Florence, Florence, Italy
| | - Tania Gamberi
- Department of Biomedical, Experimental and Clinical Sciences, University of Florence, Florence, Italy
| | - Gianluca Lucchese
- Institute of Thoracic and Cardiovascular Surgery, University of Verona, Verona, Italy
| | - Giuseppe Faggian
- Institute of Thoracic and Cardiovascular Surgery, University of Verona, Verona, Italy
| | - Alessandra Modesti
- Department of Biomedical, Experimental and Clinical Sciences, University of Florence, Florence, Italy.
| | - Pietro Amedeo Modesti
- Department of Clinical and Experimental Medicine, University of Florence, School of Medicine, Florence, Italy.
| |
Collapse
|
56
|
Haghikia A, Hoch M, Stapel B, Hilfiker-Kleiner D. STAT3 regulation of and by microRNAs in development and disease. JAKSTAT 2014; 1:143-50. [PMID: 24058763 PMCID: PMC3670237 DOI: 10.4161/jkst.19573] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2011] [Revised: 02/01/2012] [Accepted: 02/02/2012] [Indexed: 01/19/2023] Open
Abstract
MicroRNAs (miRNAs) are endogenously expressed small non-coding RNAs acting at the post-transcriptional level where they promote mRNA degradation and block protein translation. Recent findings suggest that complex transcriptional and post-transcriptional circuits control miRNAs. STAT3 has emerged as an important regulator of their expression and biogenesis and, in turn, STAT3 signaling pathways are controlled by distinct miRNAs. We summarize the current knowledge on STAT3 mediated processing of individual miRNAs and contrariwise, the modulation of the STAT3 pathway by miRNAs in development and in pathophysiological conditions such as immune processes, infection, cancer, cardiovascular disease and pulmonary hypertension.
Collapse
Affiliation(s)
- Arash Haghikia
- Department of Cardiology and Angiology; Medical School Hannover; Hannover, Germany
| | | | | | | |
Collapse
|
57
|
Peters T, Schroen B. Missing links in cardiology: long non-coding RNAs enter the arena. Pflugers Arch 2014; 466:1177-87. [PMID: 24619481 DOI: 10.1007/s00424-014-1479-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Revised: 02/07/2014] [Accepted: 02/08/2014] [Indexed: 11/25/2022]
Abstract
Heart failure as a consequence of ischemic, hypertensive, infectious, or hereditary heart disease is a major challenge in cardiology and topic of intense research. Recently, new players appeared in this field and promise deeper insights into cardiac development, function, and disease. Long non-coding RNAs are a novel class of transcripts that can regulate gene expression and may have many more functions inside the cell. Here, we present examples on long non-coding RNA (lncRNA) function in cardiac development and give suggestions on how lncRNAs may be involved in cardiomyocyte dysfunction, myocardial fibrosis, and inflammation, three hallmarks of the failing heart. Above that, we point out opportunities as well as challenges that should be considered in the endeavor to investigate cardiac lncRNAs.
Collapse
Affiliation(s)
- Tim Peters
- Experimental Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, The Netherlands
| | | |
Collapse
|
58
|
Haghikia A, Ricke-Hoch M, Stapel B, Gorst I, Hilfiker-Kleiner D. STAT3, a key regulator of cell-to-cell communication in the heart. Cardiovasc Res 2014; 102:281-9. [PMID: 24518140 DOI: 10.1093/cvr/cvu034] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The signal transducer and activator of transcription 3 (STAT3) is fundamental for physiological homeostasis and stress-induced remodelling of the heart as deregulated STAT3 circuits are sufficient to induce dilated and peripartum cardiomyopathy and adverse remodelling after myocardial infarction. STAT3 activity depends on multiple post-translational modifications (phosphorylation, acetylation, and dimerization). It is regulated by multiple receptor systems, which are coupled to positive and negative feedback loops to ensure physiological and beneficial action. Its intracellular functions are diverse as it acts as a signalling protein, a transcription factor but also participates in mitochondria energy production and protection. STAT3 modulates proliferation, differentiation, survival, oxidative stress, and/or metabolism in cardiomyocytes, fibroblasts, endothelial cells, progenitor cells, and various inflammatory cells. By regulating the secretome of these cardiac cells, STAT3 influences a broad range of intercellular communication systems. It thereby impacts on the communication between cardiomyocytes, the plasticity of the cardiac microenvironment, the vasculature, the extracellular matrix, and the inflammation in response to physiological and pathophysiological stress. Here, we sum up current knowledge on STAT3-mediated intra- and intercellular communication within the heterogeneous cellular network of the myocardium to co-ordinate complex biological processes and discuss STAT3-dependent targets as novel therapeutic concepts to treat various forms of heart disease.
Collapse
Affiliation(s)
- Arash Haghikia
- Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg Str. 1, 30625 Hannover, Germany
| | | | | | | | | |
Collapse
|
59
|
Lin J, Peng Y, Zhou B, Dou Q, Li Y, Yang H, Zhang L, Rao L. Genetic association of IL-21 polymorphisms with dilated cardiomyopathy in a Han Chinese population. Herz 2014; 40:534-41. [PMID: 24445858 DOI: 10.1007/s00059-013-4039-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2013] [Accepted: 12/08/2013] [Indexed: 02/05/2023]
Abstract
BACKGROUND Autoimmune abnormalities appear to be major predisposing factors for dilated cardiomyopathy (DCM). Interleukin-21 (IL-21) gene polymorphisms have been previously found to be associated with autoimmune diseases. This study aimed to assess the role of IL-21 in DCM in a Han Chinese population. PATIENTS AND METHODS A total of 364 independent DCM patients and 384 unrelated healthy controls were recruited for this case-control association study. rs2055979 and rs12508721 were genotyped by PCR-RFLP. IL-21 plasma levels in samples from DCM and control individuals were evaluated by ELISA. The association between the SNPs and overall survival (OS) was evaluated by Kaplan-Meier analysis. Hazard ratios and 95 % confidence intervals (CIs) were assessed in a Cox regression analysis with adjustment for sex and age. RESULTS The T allele frequencies of both SNPs were higher in DCM patients than in controls (p < 0.001). The genotypic frequencies of rs2055979 G > T and rs12508721 C > T were associated with DCM in the codominant, dominant, and recessive models (p < 0.05). IL-21 plasma levels in patients were higher than those of the control subjects (p = 0.009). The TT genotypes of both SNPs were associated with significantly higher plasma levels (prs2055979 = 0.03, prs12508721 < 0.001). Kaplan-Meier analysis showed that the genotypic frequencies of both SNPs were associated with OS in the dominant and the recessive models (p < 0.001). The TT genotypes of both SNPs were associated with the worst OS (p < 0.001). CONCLUSION Our findings suggest that theIL-21 gene plays an important role in susceptibility to DCM as well as in the clinical outcome of this ailment in the Han Chinese population.
Collapse
Affiliation(s)
- J Lin
- Department of Cardiology, West China Hospital, Sichuan University, 610041, Chengdu, P.R. China
| | | | | | | | | | | | | | | |
Collapse
|
60
|
LI YONGGUANG, ZHU WEI, LI JINGBO, LIU MINGYA, WEI MENG. Resveratrol suppresses the STAT3 signaling pathway and inhibits proliferation of high glucose-exposed HepG2 cells partly through SIRT1. Oncol Rep 2013; 30:2820-8. [DOI: 10.3892/or.2013.2748] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Accepted: 08/21/2013] [Indexed: 11/06/2022] Open
|
61
|
Zhang Q, Raje V, Yakovlev VA, Yacoub A, Szczepanek K, Meier J, Derecka M, Chen Q, Hu Y, Sisler J, Hamed H, Lesnefsky EJ, Valerie K, Dent P, Larner AC. Mitochondrial localized Stat3 promotes breast cancer growth via phosphorylation of serine 727. J Biol Chem 2013; 288:31280-8. [PMID: 24019511 DOI: 10.1074/jbc.m113.505057] [Citation(s) in RCA: 136] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Signal transducer and activator of transcription 3 (Stat3) is a key mediator in the development of many cancers. For 20 years, it has been assumed that Stat3 mediates its biological activities as a nuclear localized transcription factor activated by many cytokines. However, recent studies from this laboratory and others indicate that Stat3 has an independent function in the mitochondria (mitoStat3) where it controls the activity of the electron transport chain (ETC) and mediates Ras-induced transformation of mouse embryo fibroblasts. The actions of mitoStat3 in controlling respiration and Ras transformation are mediated by the phosphorylation state of serine 727. To address the role of mitoStat3 in the pathogenesis of cells that are transformed, we used 4T1 breast cancer cells, which form tumors that metastasize in immunocompetent mice. Substitution of Ser-727 for an alanine or aspartate in Stat3 that has a mitochondrial localization sequence, MLS-Stat3, has profound effects on tumor growth, complex I activity of the ETC, and accumulation of reactive oxygen species (ROS). Cells expressing MLS-Stat3(S727A) display slower tumor growth, decreased complex I activity of the ETC, and increased ROS accumulation under hypoxia compared with cells expressing MLS-Stat3. In contrast, cells expressing MLS-Stat3(S727D) show enhanced tumor growth and complex I activity and decreased production of ROS. These results highlight the importance of serine 727 of mitoStat3 in breast cancer and suggest a novel role for mitoStat3 in regulation of ROS concentrations through its action on the ETC.
Collapse
Affiliation(s)
- Qifang Zhang
- From the Department of Biochemistry and Molecular Biology, and Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia 23298
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
62
|
Liu M, Wilson NO, Hibbert JM, Stiles JK. STAT3 regulates MMP3 in heme-induced endothelial cell apoptosis. PLoS One 2013; 8:e71366. [PMID: 23967200 PMCID: PMC3742773 DOI: 10.1371/journal.pone.0071366] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Accepted: 06/30/2013] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND We have previously reported that free Heme generated during experimental cerebral malaria (ECM) in mice, is central to the pathogenesis of fatal ECM. Heme-induced up-regulation of STAT3 and CXCL10 promotes whereas up-regulation of HO-1 prevents brain tissue damage in ECM. We have previously demonstrated that Heme is involved in the induction of apoptosis in vascular endothelial cells. In the present study, we further tested the hypothesis that Heme reduces blood-brain barrier integrity during ECM by induction of apoptosis of brain vascular endothelial cells through STAT3 and its target gene matrix metalloproteinase three (MMP3) signaling. METHODS Genes associated with the JAK/STAT3 signaling pathway induced upon stimulation by Heme treatment, were assessed using real time RT(2) Profile PCR arrays. A human MMP3 promoter was cloned into a luciferase reporter plasmid, pMMP3, and its activity was examined following exposure to Heme treatment by a luciferase reporter gene assay. In order to determine whether activated nuclear protein STAT3 binds to the MMP3 promoter and regulates MMP3 gene, we conducted a ChIP analysis using Heme-treated and untreated human brain microvascular endothelial cells (HBVEC), and determined mRNA and protein expression levels of MMP3 using qRT-PCR and Western blot. Apoptosis in HBVEC treated with Heme was evaluated by MTT and TUNEL assay. RESULTS The results show that (1) Heme activates a variety of JAK/STAT3 downstream pathways in HBVEC. STAT3 targeted genes such as MMP3 and C/EBPb (Apoptosis-related genes), are up regulated in HBVEC treated with Heme. (2) Heme-induced HBVEC apoptosis via activation of STAT3 as well as its downstream signaling molecule MMP3 and upregulation of CXCL10 and HO-1 expressions. (3) Phosphorylated STAT3 binds to the MMP3 promoter in HBVEC cells, STAT3 transcribed MMP3 and induced MMP3 protein expression in HBVEC cells. CONCLUSIONS Activated STAT3 binds to the MMP3 promoter region and regulates MMP3 in Heme-induced endothelial cell apoptosis.
Collapse
Affiliation(s)
- Mingli Liu
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, Georgia, United States of America
- * E-mail: (ML); (JKS)
| | - Nana O. Wilson
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, Georgia, United States of America
| | - Jacqueline M. Hibbert
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, Georgia, United States of America
| | - Jonathan K. Stiles
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, Georgia, United States of America
- * E-mail: (ML); (JKS)
| |
Collapse
|
63
|
Zgheib C, Zouein FA, Kurdi M, Booz GW. Differential STAT3 signaling in the heart: Impact of concurrent signals and oxidative stress. JAKSTAT 2013; 1:101-10. [PMID: 23904970 PMCID: PMC3670289 DOI: 10.4161/jkst.19776] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Multiple lines of evidence suggest that the transcription factor STAT3 is linked to a protective and reparative response in the heart. Thus, increasing duration or intensity of STAT3 activation ought to minimize damage and improve heart function under conditions of stress. Two recent studies using genetic mouse models, however, report findings that appear to refute this proposition. Unfortunately, studies often approach the question of the role of STAT3 in the heart from the perspective that all STAT3 signaling is equivalent, particularly when it comes to signaling by IL-6 type cytokines, which share the gp130 signaling protein. Moreover, STAT3 activation is typically equated with phosphorylation of a critical tyrosine residue. Yet, STAT3 transcriptional behavior is subject to modulation by serine phosphorylation, acetylation, and redox status of the cell. Unphosphorylated STAT3 is implicated in gene induction as well. Thus, how STAT3 is activated and also what other signaling events are occurring at the same time is likely to impact on the outcome ultimately linked to STAT3. Notably STAT3 may serve as a scaffold protein allowing it to interact with other singling pathways. In this context, canonical gp130 cytokine signaling may function to integrate STAT3 signaling with a protective PI3K/AKT signaling network via mutual involvement of JAK tyrosine kinases. Differences in the extent of integration may occur between those cytokines that signal through gp130 homodimers and those through heterodimers of gp130 with a receptor α chain. Signal integration may have importance not only for deciding the particular gene profile linked to STAT3, but for the newly described mitochondrial stabilization role of STAT3 as well. In addition, disruption of integrated gp130-related STAT3 signaling may occur under conditions of oxidative stress, which negatively impacts on JAK catalytic activity. For these reasons, understanding the importance of STAT3 signaling to heart function requires a greater appreciation of the plasticity of this transcription factor in the context in which it is investigated.
Collapse
Affiliation(s)
- Carlos Zgheib
- Department of Pharmacology and Toxicology; School of Medicine; and the Center for Excellence in Cardiovascular-Renal Research; The University of Mississippi Medical Center; Jackson, MS USA
| | | | | | | |
Collapse
|
64
|
Zhang M, Wei J, Shan H, Wang H, Zhu Y, Xue J, Lin L, Yan R. Calreticulin-STAT3 signaling pathway modulates mitochondrial function in a rat model of furazolidone-induced dilated cardiomyopathy. PLoS One 2013; 8:e66779. [PMID: 23818963 PMCID: PMC3688564 DOI: 10.1371/journal.pone.0066779] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2012] [Accepted: 05/10/2013] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Calreticulin is a Ca(2+)-binding chaperone of the endoplasmic reticulum which regulates the signal transducer and activator of transcription 3 (STAT3). The effects of the calreticulin-STAT3 signaling pathway on cardiac mitochondria and on the progress of dilated cardiomyopathy (DCM) are still unclear. METHODS AND RESULTS The DCM model was generated in rats by the daily oral administration of furazolidone. Echocardiographic and hemodynamic studies demonstrated enlarged LV dimensions and reduced systolic and diastolic functions at thirty weeks after the first furazolidone administration. Morphometric analysis showed significant myocardial degeneration, interstitial fibrosis, and mitochondrial swelling with fractured or dissolved cristae in the model group. Compared with the control group, the mitochondrial membrane potential (MMP) level of the freshly isolated cardiac mitochondria and the enzyme activities of cytochrome c oxidase and succinate dehydrogenase in the model group were significantly decreased (P<0.05). Real-time PCR and western-blot revealed the increased expression of calreticulin associated with decreased activity of STAT3 in the model group. When cultured neonatal rat cardiomyocytes were exposed to furazolidone, a dose-dependent decrease in cell viability and MMP, and the increase of apoptosis rate were observed. The mRNA and protein expression of CRT gradually increased with the increase of furazolidone concentration, associated with a gradual decrease of the STAT3 phosphorylation level both in the whole cell and mitochondrial fraction. When calreticulin was knocked down with siRNA in cardiomyocytes, these changes of cardiomyocytes and mitochondria induced by furazolidone were significantly attenuated. CONCLUSIONS A rat model of DCM induced by furazolidone is successfully established. The calreticulin-STAT3 signaling pathway is involved in cardiac mitochondrial injury and the progress of furazolidone induced DCM.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Apoptosis/drug effects
- Blotting, Western
- Calreticulin/genetics
- Calreticulin/metabolism
- Cardiomyopathy, Dilated/chemically induced
- Cardiomyopathy, Dilated/metabolism
- Cardiomyopathy, Dilated/physiopathology
- Cell Survival/drug effects
- Cells, Cultured
- Disease Models, Animal
- Dose-Response Relationship, Drug
- Electron Transport Complex IV/metabolism
- Furazolidone/toxicity
- Humans
- Male
- Membrane Potential, Mitochondrial/drug effects
- Membrane Potential, Mitochondrial/physiology
- Mitochondria, Heart/metabolism
- Mitochondria, Heart/physiology
- Mitochondrial Swelling/drug effects
- Mitochondrial Swelling/physiology
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/physiology
- RNA Interference
- Rats
- Rats, Sprague-Dawley
- Reverse Transcriptase Polymerase Chain Reaction
- STAT3 Transcription Factor/genetics
- STAT3 Transcription Factor/metabolism
- Signal Transduction
- Succinate Dehydrogenase/metabolism
Collapse
Affiliation(s)
- Ming Zhang
- Department of Cardiology, The Second Affiliated Hospital, Xi’an Jiaotong University School of Medicine, Xi’an, Shaanxi, China
| | - Jin Wei
- Department of Cardiology, The Second Affiliated Hospital, Xi’an Jiaotong University School of Medicine, Xi’an, Shaanxi, China
- * E-mail:
| | - Hu Shan
- Department of Cardiology, The Second Affiliated Hospital, Xi’an Jiaotong University School of Medicine, Xi’an, Shaanxi, China
| | - Hao Wang
- Department of Anesthesiology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, United States of America
| | - Yanhe Zhu
- Department of Cardiology, The Second Affiliated Hospital, Xi’an Jiaotong University School of Medicine, Xi’an, Shaanxi, China
| | - Jiahong Xue
- Department of Cardiology, The Second Affiliated Hospital, Xi’an Jiaotong University School of Medicine, Xi’an, Shaanxi, China
| | - Lin Lin
- Department of Cardiology, The Second Affiliated Hospital, Xi’an Jiaotong University School of Medicine, Xi’an, Shaanxi, China
| | - Rui Yan
- Department of Cardiology, The Second Affiliated Hospital, Xi’an Jiaotong University School of Medicine, Xi’an, Shaanxi, China
| |
Collapse
|
65
|
Zouein FA, Kurdi M, Booz GW. Dancing rhinos in stilettos: The amazing saga of the genomic and nongenomic actions of STAT3 in the heart. JAKSTAT 2013; 2:e24352. [PMID: 24069556 PMCID: PMC3772108 DOI: 10.4161/jkst.24352] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Revised: 03/18/2013] [Accepted: 03/18/2013] [Indexed: 01/15/2023] Open
Abstract
A substantial body of evidence has shown that signal transducer and activator of transcription 3 (STAT3) has an important role in the heart in protecting the myocardium from ischemia and oxidative stress. These actions are attributed to STAT3 functioning as a transcription factor in upregulating cardioprotective genes. Loss of STAT3 has been implicated as well in the pathogenesis of heart failure and, in that context and in addition to the loss of a cardioprotective gene program, nuclear STAT3 has been identified as a transcriptional repressor important for the normal functioning of the ubiquitin-proteasome system for protein degradation. The later finding establishes a genomic role for STAT3 in controlling cellular homeostasis in cardiac myocytes independent of stress. Surprisingly, although a well-studied area, very few downstream gene targets of STAT3 in the heart have been definitively identified. In addition, STAT3 is now known to induce gene expression by noncanonical means that are not well characterized in the heart. On the other hand, recent evidence has shown that STAT3 has important nongenomic actions in cardiac myocytes that affect microtubule stability, mitochondrial respiration, and autophagy. These extranuclear actions of STAT3 involve protein–protein interactions that are incompletely understood, as is their regulation in both the healthy and injured heart. Moreover, how the diverse genomic and nongenomic actions of STAT3 crosstalk with each other is unchartered territory. Here we present an overview of what is and is not known about both the genomic and nongenomic actions of STAT3 in the heart from a structure-function perspective that focuses on the impact of posttranslational modifications and oxidative stress in regulating the actions and interactions of STAT3. Even though we have learnt a great deal about the role played by STAT3 in the heart, much more awaits to be discovered.
Collapse
Affiliation(s)
- Fouad A Zouein
- Department of Pharmacology and Toxicology; School of Medicine; and The Jackson Center for Heart Research at UMMC; The Cardiovascular-Renal Research Center; The University of Mississippi Medical Center; Jackson, MS USA
| | | | | |
Collapse
|
66
|
Ansley DM, Wang B. Oxidative stress and myocardial injury in the diabetic heart. J Pathol 2013; 229:232-41. [PMID: 23011912 DOI: 10.1002/path.4113] [Citation(s) in RCA: 171] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2012] [Revised: 09/13/2012] [Accepted: 09/14/2012] [Indexed: 12/14/2022]
Abstract
Reactive oxygen or nitrogen species play an integral role in both myocardial injury and repair. This dichotomy is differentiated at the level of species type, amount and duration of free radical generated. Homeostatic mechanisms designed to prevent free radical generation in the first instance, scavenge, or enzymatically convert them to less toxic forms and water, playing crucial roles in the maintenance of cellular structure and function. The outcome between functional recovery and dysfunction is dependent upon the inherent ability of these homeostatic antioxidant defences to withstand acute free radical generation, in the order of seconds to minutes. Alternatively, pre-existent antioxidant capacity (from intracellular and extracellular sources) may regulate the degree of free radical generation. This converts reactive oxygen and nitrogen species to the role of second messenger involved in cell signalling. The adaptive capacity of the cell is altered by the balance between death or survival signal converging at the level of the mitochondria, with distinct pathophysiological consequences that extends the period of injury from hours to days and weeks. Hyperglycaemia, hyperlipidaemia and insulin resistance enhance oxidative stress in the diabetic myocardium that cannot adapt to ischaemia-reperfusion. Altered glucose flux, mitochondrial derangements and nitric oxide synthase uncoupling in the presence of decreased antioxidant defence and impaired prosurvival cell signalling may render the diabetic myocardium more vulnerable to injury, remodelling and heart failure.
Collapse
Affiliation(s)
- David M Ansley
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, BC, Canada.
| | | |
Collapse
|
67
|
Zouein FA, Zgheib C, Hamza S, Fuseler JW, Hall JE, Soljancic A, Lopez-Ruiz A, Kurdi M, Booz GW. Role of STAT3 in angiotensin II-induced hypertension and cardiac remodeling revealed by mice lacking STAT3 serine 727 phosphorylation. Hypertens Res 2013; 36:496-503. [PMID: 23364341 DOI: 10.1038/hr.2012.223] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
STAT3 is involved in protection of the heart provided by ischemic preconditioning. However, the role of this transcription factor in the heart in chronic stresses such as hypertension has not been defined. We assessed whether STAT3 is important in hypertension-induced cardiac remodeling using mice with reduced STAT3 activity due to a S727A mutation (SA/SA). Wild type (WT) and SA/SA mice received angiotensin (ANG) II or saline for 17 days. ANG II increased mean arterial and systolic pressure in SA/SA and WT mice, but cardiac levels of cytokines associated with heart failure were increased less in SA/SA mice. Unlike WT mice, hearts of SA/SA mice showed signs of developing systolic dysfunction as evidenced by reduction in ejection fraction and fractional shortening. In the left ventricle of both WT and SA/SA mice, ANG II induced fibrosis. However, fibrosis in SA/SA mice appeared more extensive and was associated with loss of myocytes. Cardiac hypertrophy as indexed by heart to body weight ratio and left ventricular anterior wall dimension during diastole was greater in WT mice. In WT+ANG II mice there was an increase in the mass of individual myofibrils. In contrast, cardiac myocytes of SA/SA+ANG II mice showed a loss in myofibrils and myofibrillar mass density was decreased during ANG II infusion. Our findings reveal that STAT3 transcriptional activity is important for normal cardiac myocyte myofibril morphology. Loss of STAT3 may impair cardiac function in the hypertensive heart due to defective myofibrillar structure and remodeling that may lead to heart failure.
Collapse
Affiliation(s)
- Fouad A Zouein
- Department of Pharmacology and Toxicology, and Center for Excellence in Cardiovascular-Renal Research, The University of Mississippi Medical Center, School of Medicine, Jackson, MS, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
68
|
Fahmi A, Smart N, Punn A, Jabr R, Marber M, Heads R. p42/p44-MAPK and PI3K are sufficient for IL-6 family cytokines/gp130 to signal to hypertrophy and survival in cardiomyocytes in the absence of JAK/STAT activation. Cell Signal 2012; 25:898-909. [PMID: 23268184 PMCID: PMC3627957 DOI: 10.1016/j.cellsig.2012.12.008] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2012] [Accepted: 12/18/2012] [Indexed: 01/24/2023]
Abstract
The effect of differential signalling by IL-6 and leukaemia inhibitory factor (LIF) which signal by gp130 homodimerisation or LIFRβ/gp130 heterodimerisation on survival and hypertrophy was studied in neonatal rat cardiomyocytes. Both LIF and IL-6 [in the absence of soluble IL-6 receptor (sIL-6Rα)] activated Erk1/2, JNK1/2, p38-MAPK and PI3K signalling peaking at 20 min and induced cytoprotection against simulated ischemia-reperfusion injury which was blocked by the MEK1/2 inhibitor PD98059 but not the p38-MAPK inhibitor SB203580. In the absence of sIL-6R, IL-6 did not induce STAT1/3 phosphorylation, whereas IL-6/sIL-6R and LIF induced STAT1 and STAT3 phosphorylation. Furthermore, IL-6/sIL-6R induced phosphorylation of STAT1 Tyr701 and STAT3 Tyr705 were enhanced by SB203580. IL-6 and pheneylephrine (PE), but not LIF, induced cardiomyocyte iNOS expression and nitric oxide (NO) production. IL-6, LIF and PE induced cardiomyocyte hypertrophy, but with phenotypic differences in ANF and SERCA2 expression and myofilament organisation with IL-6 more resembling PE than LIF. Transfection of cardiomyocytes with full length or truncated chimaeric gp130 cytoplasmic domain/Erythropoietin receptor (EpoR) extracellular domain fusion constructs showed that the membrane proximal Box 1 and Box 2 containing region of gp130 was necessary and sufficient for MAPK and PI3K activation; hypertrophy; SERCA2 expression and iNOS/NO induction in the absence of JAK/STAT activation. In conclusion, IL-6 can signal in cardiomyocytes independent of sIL-6R and STAT1/3 and furthermore, that Erk1/2 and PI3K activation by IL-6 are both necessary and sufficient for induced cardioprotection. In addition, p38-MAPK may act as a negative feedback regulator of JAK/STAT activation in cardiomyocytes.
Collapse
Affiliation(s)
- Ahmed Fahmi
- King's College London, British Heart Foundation Centre of Research Excellence, Cardiovascular Division, School of Medicine, UK
| | | | | | | | | | | |
Collapse
|
69
|
Obana M, Miyamoto K, Murasawa S, Iwakura T, Hayama A, Yamashita T, Shiragaki M, Kumagai S, Miyawaki A, Takewaki K, Matsumiya G, Maeda M, Yoshiyama M, Nakayama H, Fujio Y. Therapeutic administration of IL-11 exhibits the postconditioning effects against ischemia-reperfusion injury via STAT3 in the heart. Am J Physiol Heart Circ Physiol 2012; 303:H569-77. [DOI: 10.1152/ajpheart.00060.2012] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Activation of cardiac STAT3 by IL-6 cytokine family contributes to cardioprotection. Previously, we demonstrated that IL-11, an IL-6 cytokine family, has the therapeutic potential to prevent adverse cardiac remodeling after myocardial infarction; however, it remains to be elucidated whether IL-11 exhibits postconditioning effects. To address the possibility that IL-11 treatment improves clinical outcome of recanalization therapy against acute myocardial infarction, we examined its postconditioning effects on ischemia/reperfusion (I/R) injury. C57BL/6 mice were exposed to ischemia (30 min) and reperfusion (24 h), and IL-11 was intravenously administered at the start of reperfusion. I/R injury mediated the activation of STAT3, which was enhanced by IL-11 administration. IL-11 treatment reduced I/R injury, analyzed by triphenyl tetrazolium chloride staining [PBS, 46.7 ± 14.4%; IL-11 (20 μg/kg), 28.6 ± 7.5% in the ratio of infarct to risk area]. Moreover, echocardiographic and hemodynamic analyses clarified that IL-11 treatment preserved cardiac function after I/R. Terminal deoxynucleotide transferase-mediated dUTP nick-end labeling staining revealed that IL-11 reduced the frequency of apoptotic cardiomyocytes after I/R. Interestingly, IL-11 reduced superoxide production assessed by in situ dihydroethidium fluorescence analysis, accompanied by the increased expression of metallothionein 1 and 2, reactive oxygen species (ROS) scavengers. Importantly, with the use of cardiac-specific STAT3 conditional knockout (STAT3 CKO) mice, it was revealed that cardiac-specific ablation of STAT3 abrogated IL-11-mediated attenuation of I/R injury. Finally, IL-11 failed to suppress the ROS production after I/R in STAT3 CKO mice. IL-11 administration exhibits the postconditioning effects through cardiac STAT3 activation, suggesting that IL-11 has the clinical therapeutic potential to prevent I/R injury in heart.
Collapse
Affiliation(s)
- Masanori Obana
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Kaori Miyamoto
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Shiho Murasawa
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Tomohiko Iwakura
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Akiko Hayama
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Tomomi Yamashita
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Momoko Shiragaki
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Shohei Kumagai
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Akimitsu Miyawaki
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Kana Takewaki
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Goro Matsumiya
- Department of Cardiovascular Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Makiko Maeda
- Department of Clinical Pharmacogenomics, School of Pharmacy, Hyogo University of Health Sciences
| | - Minoru Yoshiyama
- Department of Internal Medicine and Cardiology, Graduate School of Medicine, Osaka City University, Osaka, Japan
| | - Hiroyuki Nakayama
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Yasushi Fujio
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| |
Collapse
|
70
|
Zgheib C, Kurdi M, Zouein FA, Gunter BW, Stanley BA, Zgheib J, Romero DG, King SB, Paolocci N, Booz GW. Acyloxy nitroso compounds inhibit LIF signaling in endothelial cells and cardiac myocytes: evidence that STAT3 signaling is redox-sensitive. PLoS One 2012; 7:e43313. [PMID: 22905257 PMCID: PMC3419695 DOI: 10.1371/journal.pone.0043313] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2012] [Accepted: 07/19/2012] [Indexed: 12/31/2022] Open
Abstract
We previously showed that oxidative stress inhibits leukemia inhibitory factor (LIF) signaling by targeting JAK1, and the catalytic domains of JAK 1 and 2 have a cysteine-based redox switch. Thus, we postulated that the NO sibling and thiophylic compound, nitroxyl (HNO), would inhibit LIF-induced JAK-STAT3 activation. Pretreatment of human microvascular endothelial cells (HMEC-1) or neonatal rat cardiomyocytes with the HNO donors Angeli’s salt or nitrosocyclohexyl acetate (NCA) inhibited LIF-induced STAT3 activation. NCA pretreatment also blocked the induction of downstream inflammatory genes (e.g. intercellular adhesion molecule 1, CCAAT/enhancer binding protein delta). The related 1-nitrosocyclohexyl pivalate (NCP; not a nitroxyl donor) was equally effective in inhibiting STAT3 activation, suggesting that these compounds act as thiolate targeting electrophiles. The JAK1 redox switch is likely not a target of acyloxy nitroso compounds, as NCA had no effect on JAK1 catalytic activity and only modestly affected JAK1-induced phosphorylation of the LIF receptor. However, pretreatment of recombinant human STAT3 with NCA or NCP reduced labeling of free sulfhydryl residues. We show that NCP in the presence of diamide enhanced STAT3 glutathionylation and dimerization in adult mouse cardiac myocytes and altered STAT3 under non-reducing conditions. Finally, we show that monomeric STAT3 levels are decreased in the Gαq model of heart failure in a redox-sensitive manner. Altogether, our evidence indicates that STAT3 has redox-sensitive cysteines that regulate its activation and are targeted by HNO donors and acyloxy nitroso compounds. These findings raise the possibility of new therapeutic strategies to target STAT3 signaling via a redox-dependent manner, particularly in the context of cardiac and non-cardiac diseases with prominent pro-inflammatory signaling.
Collapse
Affiliation(s)
- Carlos Zgheib
- Departments of Pharmacology and Toxicology, School of Medicine, and Center for Excellence in Cardiovascular-Renal Research, The University of Mississippi Medical Center, Jackson, Mississippi, United States of America
| | - Mazen Kurdi
- Department of Chemistry and Biochemistry, Faculty of Sciences, Lebanese University, Rafic Hariri Educational Campus, Hadath, Lebanon
| | - Fouad A. Zouein
- Departments of Pharmacology and Toxicology, School of Medicine, and Center for Excellence in Cardiovascular-Renal Research, The University of Mississippi Medical Center, Jackson, Mississippi, United States of America
| | - Barak W. Gunter
- Departments of Pharmacology and Toxicology, School of Medicine, and Center for Excellence in Cardiovascular-Renal Research, The University of Mississippi Medical Center, Jackson, Mississippi, United States of America
| | - Brian A. Stanley
- Division of Cardiology, Johns Hopkins Medical Institutions, Baltimore, Maryland, United States of America
| | - Joe Zgheib
- Department of Medicine, Division of Cardiology, Centre Hospitalier Universitaire de Nancy, Brabois, France
| | - Damian G. Romero
- Department of Biochemistry, School of Medicine, and Center for Excellence in Cardiovascular-Renal Research, The University of Mississippi Medical Center, Jackson, Mississippi, United States of America
| | - S. Bruce King
- Department of Chemistry, Wake Forest University, Winston-Salem, North Carolina, United States of America
| | - Nazareno Paolocci
- Division of Cardiology, Johns Hopkins Medical Institutions, Baltimore, Maryland, United States of America
- Clinical Medicine Department, Section of General Pathology, University of Perugia, Perugia, Italy
| | - George W. Booz
- Departments of Pharmacology and Toxicology, School of Medicine, and Center for Excellence in Cardiovascular-Renal Research, The University of Mississippi Medical Center, Jackson, Mississippi, United States of America
- * E-mail:
| |
Collapse
|
71
|
Abstract
INTRODUCTION STAT3 is a key transcription factor for many regulatory factors that modulate gene transcription. Particularly important are cytokines and growth factors that maintain homeostasis by regulating immunocytes, stromal and epithelial cells. Dysregulation of STAT3 by constitutive activation plays an important role in the initiation of inflammation and cellular transformation in numerous cancers, especially of epithelial origin. This review focuses on STAT3 drive in gastric cancer initiation and progression, with emphasis on its activation by cytokines, and how targeting the primary drivers or gastric STAT3 therapeutically may prevent or slow stomach cancer development. AREAS COVERED This review will discuss the mechanics of STAT3 signalling, how constitutive STAT3 activation promotes gastric tumourigenesis in both human adenocarcinomas and mouse models, the nature of the upstream regulators of STAT3, and their association with chronic Helicobacter pylori infection, STAT3-activated genes that promote transformation and progression, and finally the development and use of STAT3 and upstream cytokine inhibitors as therapeutics. EXPERT OPINION Chronic STAT3 activation is a key event in gastric cancer induction and progression. Specific targeting of stomach epithelial STAT3 or blocking IL-11Rα/gp130 and/or EGFR signal transduction in chronic gastric inflammation and metaplasia may be therapeutically effective in preventing gastric carcinogenesis.
Collapse
Affiliation(s)
- Andrew S Giraud
- Murdoch Childrens Research Institute, Royal Childrens Hospital, Parkville, Australia.
| | | | | |
Collapse
|
72
|
Martinez-Abundis E, Rajapurohitam V, Haist JV, Gan XT, Karmazyn M. The obesity-related peptide leptin sensitizes cardiac mitochondria to calcium-induced permeability transition pore opening and apoptosis. PLoS One 2012; 7:e41612. [PMID: 22848545 PMCID: PMC3405002 DOI: 10.1371/journal.pone.0041612] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2012] [Accepted: 06/22/2012] [Indexed: 01/31/2023] Open
Abstract
The obesity-related 16 kDa peptide leptin is synthesized primarily in white adipocytes although its production has been reported in other tissues including the heart. There is emerging evidence that leptin may contribute to cardiac pathology especially that related to myocardial remodelling and heart failure. In view of the importance of mitochondria to these processes, the goal of the present study is to determine the effect of leptin on mitochondria permeability transition pore opening and the potential consequence in terms of development of apoptosis. Experiments were performed using neonatal rat ventricular myocytes exposed to 3.1 nM (50 ng/ml) leptin for 24 hours. Mitochondrial transition pore opening was analyzed as the capacity of mitochondria to retain the dye calcein-AM in presence of 200 µM CaCl2. Leptin significantly increased pore opening although the effect was markedly more pronounced in digitonin-permeabilized myocytes in the presence of calcium with both effects prevented by the transition pore inhibitor sanglifehrin A. These effects were associated with increased apoptosis as evidenced by increased TUNEL staining and caspase 3 activity, both of which were prevented by the transition pore inhibitor sanglifehrin A. Leptin enhanced Stat3 activation whereas a Stat 3 inhibitor peptide prevented leptin-induced mitochondrial transition pore opening as well as the hypertrophic and pro-apoptotic effects of the peptide. Inhibition of the RhoA/ROCK pathway prevented the hypertrophic response to leptin but had no effect on increased pore opening following leptin administration. We conclude that leptin can enhance calcium-mediated, Stat3-dependent pro-apoptotic effects as a result of increased mitochondrial transition pore opening and independently of its hypertrophic actions. Leptin may therefore contribute to mitochondrial dysfunction and the development of apoptosis in the diseased myocardium particularly under conditions of excessive intracellular calcium accumulation.
Collapse
Affiliation(s)
- Eduardo Martinez-Abundis
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | | | | | | | | |
Collapse
|
73
|
Szczepanek K, Lesnefsky EJ, Larner AC. Multi-tasking: nuclear transcription factors with novel roles in the mitochondria. Trends Cell Biol 2012; 22:429-37. [PMID: 22705015 DOI: 10.1016/j.tcb.2012.05.001] [Citation(s) in RCA: 90] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2012] [Revised: 05/07/2012] [Accepted: 05/15/2012] [Indexed: 01/06/2023]
Abstract
Coordinated responses between the nucleus and mitochondria are essential for the maintenance of homeostasis. For over 15 years, pools of nuclear transcription factors (TFs), such as p53 and nuclear hormone receptors, have been observed in the mitochondria. The contribution of the mitochondrial pool of these TFs to their well-defined biological actions is in some cases clear and in others not well understood. Recently, a small mitochondrial pool of the TF signal transducer and activator of transcription factor 3 (STAT3) was shown to modulate the activity of the electron transport chain (ETC). The mitochondrial function of STAT3 encompasses both its biological actions in the heart as well as its oncogenic effects. This review highlights advances in our understanding of how mitochondrial pools of nuclear TFs may influence the function of this organelle.
Collapse
Affiliation(s)
- Karol Szczepanek
- Department of Biochemistry and Molecular Biology, and Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 23298, USA
| | | | | |
Collapse
|
74
|
Quijada P, Toko H, Fischer KM, Bailey B, Reilly P, Hunt KD, Gude NA, Avitabile D, Sussman MA. Preservation of myocardial structure is enhanced by pim-1 engineering of bone marrow cells. Circ Res 2012; 111:77-86. [PMID: 22619278 DOI: 10.1161/circresaha.112.265207] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
RATIONALE Bone marrow-derived cells to treat myocardial injury improve cardiac function and support beneficial cardiac remodeling. However, survival of stem cells is limited due to low proliferation of transferred cells. OBJECTIVE To demonstrate long-term potential of c-kit(+) bone marrow stem cells (BMCs) enhanced with Pim-1 kinase to promote positive cardiac remodeling. METHODS AND RESULTS Lentiviral modification of c-kit(+) BMCs to express Pim-1 (BMCeP) increases proliferation and expression of prosurvival proteins relative to BMCs expressing green fluorescent protein (BMCe). Intramyocardial delivery of BMCeP at time of infarction supports improvements in anterior wall dimensions and prevents left ventricle dilation compared with hearts treated with vehicle alone. Reduction of the akinetic left ventricular wall was observed in BMCeP-treated hearts at 4 and 12 weeks after infarction. Early recovery of cardiac function in BMCeP-injected hearts facilitated modest improvements in hemodynamic function up to 12 weeks after infarction between cell-treated groups. Persistence of BMCeP is improved relative to BMCe within the infarct together with increased recruitment of endogenous c-kit(+) cells. Delivery of BMC populations promotes cellular hypertrophy in the border and infarcted regions coupled with an upregulation of hypertrophic genes. Thus, BMCeP treatment yields improved structural remodeling of infarcted myocardium compared with control BMCs. CONCLUSIONS Genetic modification of BMCs with Pim-1 may serve as a therapeutic approach to promote recovery of myocardial structure. Future approaches may take advantage of salutary BMC actions in conjunction with other stem cell types to increase efficacy of cellular therapy and improve myocardial performance in the injured myocardium.
Collapse
|
75
|
Oba T, Yasukawa H, Hoshijima M, Sasaki KI, Futamata N, Fukui D, Mawatari K, Nagata T, Kyogoku S, Ohshima H, Minami T, Nakamura K, Kang D, Yajima T, Knowlton KU, Imaizumi T. Cardiac-specific deletion of SOCS-3 prevents development of left ventricular remodeling after acute myocardial infarction. J Am Coll Cardiol 2012; 59:838-52. [PMID: 22361405 DOI: 10.1016/j.jacc.2011.10.887] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2011] [Revised: 10/19/2011] [Accepted: 10/20/2011] [Indexed: 01/08/2023]
Abstract
OBJECTIVES The study investigated the role of myocardial suppressor of cytokine signaling-3 (SOCS3), an intrinsic negative feedback regulator of the janus kinase and signal transducer and activator of transcription (JAK-STAT) signaling pathway, in the development of left ventricular (LV) remodeling after acute myocardial infarction (AMI). BACKGROUND LV remodeling after AMI results in poor cardiac performance leading to heart failure. Although it has been shown that JAK-STAT-activating cytokines prevent LV remodeling after AMI in animals, little is known about the role of SOCS3 in this process. METHODS Cardiac-specific SOCS3 knockout mice (SOCS3-CKO) were generated and subjected to AMI induced by permanent ligation of the left anterior descending coronary artery. RESULTS Although the initial infarct size after coronary occlusion measured by triphenyltetrazolium chloride staining was comparable between SOCS3-CKO and control mice, the infarct size 14 days after AMI was remarkably inhibited in SOCS3-CKO, indicating that progression of LV remodeling after AMI was prevented in SOCS3-CKO hearts. Prompt and marked up-regulations of multiple JAK-STAT-activating cytokines including leukemia inhibitory factor and granulocyte colony-stimulating factor (G-CSF) were observed within the heart following AMI. Cardiac-specific SOCS3 deletion enhanced multiple cardioprotective signaling pathways including STAT3, AKT, and extracellular signal-regulated kinase (ERK)-1/2, while inhibiting myocardial apoptosis and fibrosis as well as augmenting antioxidant expression. CONCLUSIONS Enhanced activation of cardioprotective signaling pathways by inhibiting myocardial SOCS3 expression prevented LV remodeling after AMI. Our data suggest that myocardial SOCS3 may be a key molecule in the development of LV remodeling after AMI.
Collapse
Affiliation(s)
- Toyoharu Oba
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kurume University School of Medicine, 67 Asahi-machi, Kurume, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
76
|
Cui C, Shi Q, Zhang X, Liu X, Bai Y, Li J, Liu S, Hu S, Wei Y. CRP promotes MMP-10 expression via c-Raf/MEK/ERK and JAK1/ERK pathways in cardiomyocytes. Cell Signal 2011; 24:810-8. [PMID: 22142512 DOI: 10.1016/j.cellsig.2011.11.019] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2011] [Revised: 11/11/2011] [Accepted: 11/17/2011] [Indexed: 11/29/2022]
Abstract
C-reactive protein (CRP) was reported to be a predictor for left ventricular (LV) remodeling. Matrix metalloproteinase (MMP)-10 participates in the LV remodeling process. However, the intrinsic relationship between CRP and MMP-10 in cardiomyocytes remains unclear. The purpose of this study is to observe whether CRP may promote MMP-10 expression, and if so, to clarify signaling pathways to be involved in CRP-induced MMP-10 expression in cardiomyocytes. We observed in cultured cardiomyocytes that CRP at a dose of 5 μg/ml increased MMP-10 expression and activity in a time-dependent manner, as measured by real-time polymerase chain reaction (PCR), western blots, and casein zymography analysis. We hypothesized that signal pathways of mitogen-activated protein kinases (MAPKs) and Janus kinases (JAKs)/signal transducers and activators of transcription (STATs) might be involved in CRP-induced MMP-10 expression. Our results showed that CRP markedly activated c-Raf/MEK/ERK and JAK1/ERK signaling pathways but not JAK1/STAT3 signaling pathway by using the phosphor-specific antibodies against these pathways, and blockages of c-Raf/MEK/ERK and JAK1/ERK signaling pathways by the specific ERK1/2 inhibitor U0126 and JAK1 inhibitor piceatannol could significantly decrease CRP-induced MMP-10 expression. In addition, we demonstrated that the DNA binding sites of AP-1 and STAT3 in the nucleus of cardiomyocytes mediated CRP-induced MMP-10 expression. In conclusion, we demonstrated that CRP promoted MMP-10 expression and activity in cardiomyocytes, and clarified that c-Raf/MEK/ERK and JAK1/ERK signaling pathways were involved in MMP-10 expression regulation via activation of DNA binding sites for AP-1 and STAT3 in cardiomyocytes. Our findings suggest that CRP acts as a predictor for LV remodeling might be associated with its promotion effect on MMP-10 expression and activity.
Collapse
Affiliation(s)
- Chuanjue Cui
- State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Disease & Fuwai Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, PR China
| | | | | | | | | | | | | | | | | |
Collapse
|