1
|
Osman EEA, Neamati N. Ironing Out the Mechanism of gp130 Signaling. Pharmacol Rev 2024; 76:1399-1443. [PMID: 39414364 DOI: 10.1124/pharmrev.124.001245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 08/15/2024] [Accepted: 08/16/2024] [Indexed: 10/18/2024] Open
Abstract
gp130 functions as a shared signal-transducing subunit not only for interleukin (IL)-6 but also for eight other human cytokine receptor complexes. The IL-6 signaling pathway mediated through gp130 encompasses classical, trans, or cluster signaling, intricately regulated by a diverse array of modulators affecting IL-6, its receptor, and gp130. Currently, only a limited number of small molecule antagonists and agonists for gp130 are known. This review aims to comprehensively examine the current knowledge of these modulators and provide insights into their pharmacological properties, particularly in the context of cancer and other diseases. Notably, the prominent gp130 modulators SC144, bazedoxifene, and raloxifene are discussed in detail, with a specific focus on the discovery of SC144's iron-chelating properties. This adds a new dimension to the understanding of its pharmacological effects and therapeutic potential in conditions where iron homeostasis is significant. Our bioinformatic analysis of gp130 and genes related to iron homeostasis reveals insightful correlations, implicating the role of iron in the gp130 signaling pathway. Overall, this review contributes to the evolving understanding of gp130 modulation and its potential therapeutic applications in various disease contexts. SIGNIFICANCE STATEMENT: This perspective provides a timely and comprehensive analysis of advancements in gp130 signaling research, emphasizing the therapeutic implications of the currently available modulators. Bioinformatic analysis demonstrates potential interplay between gp130 and genes that regulate iron homeostasis, suggesting new therapeutic avenues. By combining original research findings with a broader discussion of gp130's therapeutic potential, this perspective significantly contributes to the field.
Collapse
Affiliation(s)
- Essam Eldin A Osman
- Department of Medicinal Chemistry, College of Pharmacy, and Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan (E.E.A.O., N.N.) and Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt (E.E.A.O.)
| | - Nouri Neamati
- Department of Medicinal Chemistry, College of Pharmacy, and Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan (E.E.A.O., N.N.) and Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt (E.E.A.O.)
| |
Collapse
|
2
|
Cugudda A, La Manna S, Marasco D. Are peptidomimetics the compounds of choice for developing new modulators of the JAK-STAT pathway? Front Immunol 2024; 15:1406886. [PMID: 38983855 PMCID: PMC11232365 DOI: 10.3389/fimmu.2024.1406886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 06/12/2024] [Indexed: 07/11/2024] Open
Abstract
Protein-protein interactions (PPIs) play critical roles in a wide range of biological processes including the dysregulation of cellular pathways leading to the loss of cell function, which in turn leads to diseases. The dysfunction of several signaling pathways is linked to the insurgence of pathological processes such as inflammation, cancer development and neurodegeneration. Thus, there is an urgent need for novel chemical modulators of dysregulated PPIs to drive progress in targeted therapies. Several PPIs have been targeted by bioactive compounds, and, often, to properly cover interacting protein regions and improve the biological activities of modulators, a particular focus concerns the employment of macrocycles as proteomimetics. Indeed, for their physicochemical properties, they occupy an intermediate space between small organic molecules and macromolecular proteins and are prominent in the drug discovery process. Peptide macrocycles can modulate fundamental biological mechanisms and here we will focus on peptidomimetics active on the Janus kinase/signal transducers and activators of transcription (JAK-STAT) pathways.
Collapse
Affiliation(s)
| | | | - Daniela Marasco
- Department of Pharmacy, University of Naples “Federico II”, Naples, Italy
| |
Collapse
|
3
|
Mann DL. The Emerging Field of Cardioimmunology: Past, Present and Foreseeable Future. Circ Res 2024; 134:1663-1680. [PMID: 38843286 PMCID: PMC11160976 DOI: 10.1161/circresaha.123.323656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 02/08/2024] [Indexed: 06/09/2024]
Abstract
Over the past 30 years, the field of cardioimmunology has moved from being dismissed as a field that was chasing an epiphenomenon of little biological consequence to a scientific discipline that is providing important new insights into the immunologic basis for hypertension, atherosclerosis, myocarditis, pericarditis, autoimmune heart disease, and heart failure. In this article, we will review the conceptual insights and technical breakthroughs that have allowed the field to move forward, as well as the clinical trials in the cardioimmunology space, to provide a historical context for the articles that will appear in the compendium that is focused on the interface between cardioimmunology, myocardial function, and disease.
Collapse
Affiliation(s)
- Douglas L Mann
- Cardiovascular Division, Center for Cardiovascular Research, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
4
|
Sun M, Mao S, Wu C, Zhao X, Guo C, Hu J, Xu S, Zheng F, Zhu G, Tao H, He S, Hu J, Zhang Y. Piezo1-Mediated Neurogenic Inflammatory Cascade Exacerbates Ventricular Remodeling After Myocardial Infarction. Circulation 2024; 149:1516-1533. [PMID: 38235590 DOI: 10.1161/circulationaha.123.065390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 12/19/2023] [Indexed: 01/19/2024]
Abstract
BACKGROUND Heart failure is associated with a high rate of mortality and morbidity, and ventricular remodeling invariably precedes heart failure. Ventricular remodeling is fundamentally driven by mechanotransduction that is regulated by both the nervous system and the immune system. However, it remains unknown which key molecular factors govern the neuro/immune/cardio axis that underlies mechanotransduction during ventricular remodeling. Here, we investigated whether the mechanosensitive Piezo cation channel-mediated neurogenic inflammatory cascade underlies ventricular remodeling-related mechanotransduction. METHODS By ligating the left coronary artery of rats to establish an in vivo model of chronic myocardial infarction (MI), lentivirus-mediated thoracic dorsal root ganglion (TDRG)-specific Piezo1 knockdown rats and adeno-associated virus-PHP.S-mediated TDRG neuron-specific Piezo1 knockout mice were used to investigate whether Piezo1 in the TDRG plays a functional role during ventricular remodeling. Subsequently, neutralizing antibody-mediated TDRG IL-6 (interleukin-6) inhibition rats and adeno-associated virus-PHP.S-mediated TDRG neuron-specific IL-6 knockdown mice were used to determine the mechanism underlying neurogenic inflammation. Primary TDRG neurons were used to evaluate Piezo1 function in vitro. RESULTS Expression of Piezo1 and IL-6 was increased, and these factors were functionally activated in TDRG neurons at 4 weeks after MI. Both knockdown of TDRG-specific Piezo1 and deletion of TDRG neuron-specific Piezo1 lessened the severity of ventricular remodeling at 4 weeks after MI and decreased the level of IL-6 in the TDRG or heart. Furthermore, inhibition of TDRG IL-6 or knockdown of TDRG neuron-specific IL-6 also ameliorated ventricular remodeling and suppressed the IL-6 cascade in the heart, whereas the Piezo1 level in the TDRG was not affected. In addition, enhanced Piezo1 function, as reflected by abundant calcium influx induced by Yoda1 (a selective agonist of Piezo1), led to increased release of IL-6 from TDRG neurons in mice 4 weeks after MI. CONCLUSIONS Our findings point to a critical role for Piezo1 in ventricular remodeling at 4 weeks after MI and reveal a neurogenic inflammatory cascade as a previously unknown facet of the neuronal immune signaling axis underlying mechanotransduction.
Collapse
Affiliation(s)
- Meiyan Sun
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China (M.S., S.M., C.W., C.G., J.H., S.X., H.T., S.H., Y.Z.)
- Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China (M.S., S.M., C.W., C.G., J.H., S.X., H.T., S.H., Y.Z.)
- Laboratory of Anesthesia and Critical Care Medicine in Colleges and Universities of Shandong Province, School of Anesthesiology, Weifang Medical University, China (M.S.)
| | - Sui Mao
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China (M.S., S.M., C.W., C.G., J.H., S.X., H.T., S.H., Y.Z.)
- Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China (M.S., S.M., C.W., C.G., J.H., S.X., H.T., S.H., Y.Z.)
| | - Chao Wu
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China (M.S., S.M., C.W., C.G., J.H., S.X., H.T., S.H., Y.Z.)
- Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China (M.S., S.M., C.W., C.G., J.H., S.X., H.T., S.H., Y.Z.)
| | - Xiaoyong Zhao
- Department of Anesthesiology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China (X.Z.)
| | - Chengxiao Guo
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China (M.S., S.M., C.W., C.G., J.H., S.X., H.T., S.H., Y.Z.)
- Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China (M.S., S.M., C.W., C.G., J.H., S.X., H.T., S.H., Y.Z.)
| | - Jun Hu
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China (M.S., S.M., C.W., C.G., J.H., S.X., H.T., S.H., Y.Z.)
- Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China (M.S., S.M., C.W., C.G., J.H., S.X., H.T., S.H., Y.Z.)
| | - Shijin Xu
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China (M.S., S.M., C.W., C.G., J.H., S.X., H.T., S.H., Y.Z.)
- Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China (M.S., S.M., C.W., C.G., J.H., S.X., H.T., S.H., Y.Z.)
| | - Fen Zheng
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, and Department of Physiology, Nanjing Medical University, China (F.Z., G.Z.)
| | - Guoqing Zhu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, and Department of Physiology, Nanjing Medical University, China (F.Z., G.Z.)
| | - Hui Tao
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China (M.S., S.M., C.W., C.G., J.H., S.X., H.T., S.H., Y.Z.)
- Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China (M.S., S.M., C.W., C.G., J.H., S.X., H.T., S.H., Y.Z.)
| | - Shufang He
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China (M.S., S.M., C.W., C.G., J.H., S.X., H.T., S.H., Y.Z.)
- Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China (M.S., S.M., C.W., C.G., J.H., S.X., H.T., S.H., Y.Z.)
| | - Ji Hu
- Laboratory of Stress Neurobiology, School of Life Science and Technology, ShanghaiTech University, China (J.H.)
| | - Ye Zhang
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China (M.S., S.M., C.W., C.G., J.H., S.X., H.T., S.H., Y.Z.)
- Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China (M.S., S.M., C.W., C.G., J.H., S.X., H.T., S.H., Y.Z.)
| |
Collapse
|
5
|
Schelemei P, Wagner E, Picard FSR, Winkels H. Macrophage mediators and mechanisms in cardiovascular disease. FASEB J 2024; 38:e23424. [PMID: 38275140 DOI: 10.1096/fj.202302001r] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 12/21/2023] [Accepted: 12/29/2023] [Indexed: 01/27/2024]
Abstract
Macrophages are major players in myocardial infarction (MI) and atherosclerosis, two major cardiovascular diseases (CVD). Atherosclerosis is caused by the buildup of cholesterol-rich lipoproteins in blood vessels, causing inflammation, vascular injury, and plaque formation. Plaque rupture or erosion can cause thrombus formation resulting in inadequate blood flow to the heart muscle and MI. Inflammation, particularly driven by macrophages, plays a central role in both atherosclerosis and MI. Recent integrative approaches of single-cell analysis-based classifications in both murine and human atherosclerosis as well as experimental MI showed overlap in origin, diversity, and function of macrophages in the aorta and the heart. We here discuss differences and communalities between macrophages in the heart and aorta at steady state and in atherosclerosis or upon MI. We focus on markers, mediators, and functional states of macrophage subpopulations. Recent trials testing anti-inflammatory agents show a major benefit in reducing the inflammatory burden of CVD patients, but highlight a necessity for a broader understanding of immune cell ontogeny and heterogeneity in CVD. The novel insights into macrophage biology in CVD represent exciting opportunities for the development of novel treatment strategies against CVD.
Collapse
Affiliation(s)
- Patrik Schelemei
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Clinic III for Internal Medicine, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Elena Wagner
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Clinic III for Internal Medicine, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Felix Simon Ruben Picard
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Clinic III for Internal Medicine, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Holger Winkels
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Clinic III for Internal Medicine, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| |
Collapse
|
6
|
Sheng SY, Li JM, Hu XY, Wang Y. Regulated cell death pathways in cardiomyopathy. Acta Pharmacol Sin 2023; 44:1521-1535. [PMID: 36914852 PMCID: PMC10374591 DOI: 10.1038/s41401-023-01068-9] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 02/20/2023] [Indexed: 03/16/2023]
Abstract
Heart disease is a worldwide health menace. Both intractable primary and secondary cardiomyopathies contribute to malignant cardiac dysfunction and mortality. One of the key cellular processes associated with cardiomyopathy is cardiomyocyte death. Cardiomyocytes are terminally differentiated cells with very limited regenerative capacity. Various insults can lead to irreversible damage of cardiomyocytes, contributing to progression of cardiac dysfunction. Accumulating evidence indicates that majority of cardiomyocyte death is executed by regulating molecular pathways, including apoptosis, ferroptosis, autophagy, pyroptosis, and necroptosis. Importantly, these forms of regulated cell death (RCD) are cardinal features in the pathogenesis of various cardiomyopathies, including dilated cardiomyopathy, diabetic cardiomyopathy, sepsis-induced cardiomyopathy, and drug-induced cardiomyopathy. The relevance between abnormity of RCD with adverse outcome of cardiomyopathy has been unequivocally evident. Therefore, there is an urgent need to uncover the molecular and cellular mechanisms for RCD in order to better understand the pathogenesis of cardiomyopathies. In this review, we summarize the latest progress from studies on RCD pathways in cardiomyocytes in context of the pathogenesis of cardiomyopathies, with particular emphasis on apoptosis, necroptosis, ferroptosis, autophagy, and pyroptosis. We also elaborate the crosstalk among various forms of RCD in pathologically stressed myocardium and the prospects of therapeutic applications targeted to various cell death pathways.
Collapse
Affiliation(s)
- Shu-Yuan Sheng
- Department of Cardiology, Zhejiang University School of Medicine, Second Affiliated Hospital, Hangzhou, 310009, China
| | - Jia-Min Li
- Department of Cardiology, Zhejiang University School of Medicine, Second Affiliated Hospital, Hangzhou, 310009, China
| | - Xin-Yang Hu
- Department of Cardiology, Zhejiang University School of Medicine, Second Affiliated Hospital, Hangzhou, 310009, China
| | - Yibin Wang
- Department of Cardiology, Zhejiang University School of Medicine, Second Affiliated Hospital, Hangzhou, 310009, China.
- Signature Program in Cardiovascular and Metabolic Diseases, DukeNUS Medical School and National Heart Center of Singapore, Singapore, Singapore.
| |
Collapse
|
7
|
Bazgir F, Nau J, Nakhaei-Rad S, Amin E, Wolf MJ, Saucerman JJ, Lorenz K, Ahmadian MR. The Microenvironment of the Pathogenesis of Cardiac Hypertrophy. Cells 2023; 12:1780. [PMID: 37443814 PMCID: PMC10341218 DOI: 10.3390/cells12131780] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 06/22/2023] [Accepted: 06/29/2023] [Indexed: 07/15/2023] Open
Abstract
Pathological cardiac hypertrophy is a key risk factor for the development of heart failure and predisposes individuals to cardiac arrhythmia and sudden death. While physiological cardiac hypertrophy is adaptive, hypertrophy resulting from conditions comprising hypertension, aortic stenosis, or genetic mutations, such as hypertrophic cardiomyopathy, is maladaptive. Here, we highlight the essential role and reciprocal interactions involving both cardiomyocytes and non-myocardial cells in response to pathological conditions. Prolonged cardiovascular stress causes cardiomyocytes and non-myocardial cells to enter an activated state releasing numerous pro-hypertrophic, pro-fibrotic, and pro-inflammatory mediators such as vasoactive hormones, growth factors, and cytokines, i.e., commencing signaling events that collectively cause cardiac hypertrophy. Fibrotic remodeling is mediated by cardiac fibroblasts as the central players, but also endothelial cells and resident and infiltrating immune cells enhance these processes. Many of these hypertrophic mediators are now being integrated into computational models that provide system-level insights and will help to translate our knowledge into new pharmacological targets. This perspective article summarizes the last decades' advances in cardiac hypertrophy research and discusses the herein-involved complex myocardial microenvironment and signaling components.
Collapse
Affiliation(s)
- Farhad Bazgir
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (F.B.); (J.N.)
| | - Julia Nau
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (F.B.); (J.N.)
| | - Saeideh Nakhaei-Rad
- Stem Cell Biology, and Regenerative Medicine Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad 91779-48974, Iran;
| | - Ehsan Amin
- Institute of Neural and Sensory Physiology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany;
| | - Matthew J. Wolf
- Department of Medicine and Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA 22908, USA;
| | - Jeffry J. Saucerman
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA;
| | - Kristina Lorenz
- Institute of Pharmacology and Toxicology, University of Würzburg, Leibniz Institute for Analytical Sciences, 97078 Würzburg, Germany;
| | - Mohammad Reza Ahmadian
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (F.B.); (J.N.)
| |
Collapse
|
8
|
Moustafa A, Hashemi S, Brar G, Grigull J, Ng SHS, Williams D, Schmitt-Ulms G, McDermott JC. The MEF2A transcription factor interactome in cardiomyocytes. Cell Death Dis 2023; 14:240. [PMID: 37019881 PMCID: PMC10076289 DOI: 10.1038/s41419-023-05665-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 12/02/2022] [Accepted: 02/08/2023] [Indexed: 04/07/2023]
Abstract
Transcriptional regulators encoded by the Myocyte Enhancer Factor 2 (MEF2) gene family play a fundamental role in cardiac development, homeostasis and pathology. Previous studies indicate that MEF2A protein-protein interactions serve as a network hub in several cardiomyocyte cellular processes. Based on the idea that interactions with regulatory protein partners underly the diverse roles of MEF2A in cardiomyocyte gene expression, we undertook a systematic unbiased screen of the MEF2A protein interactome in primary cardiomyocytes using an affinity purification-based quantitative mass spectrometry approach. Bioinformatic processing of the MEF2A interactome revealed protein networks involved in the regulation of programmed cell death, inflammatory responses, actin dynamics and stress signaling in primary cardiomyocytes. Further biochemical and functional confirmation of specific protein-protein interactions documented a dynamic interaction between MEF2A and STAT3 proteins. Integration of transcriptome level data from MEF2A and STAT3-depleted cardiomyocytes reveals that the balance between MEF2A and STAT3 activity exerts a level of executive control over the inflammatory response and cardiomyocyte cell survival and experimentally ameliorates Phenylephrine induced cardiomyocyte hypertrophy. Lastly, we identified several MEF2A/STAT3 co-regulated genes, including the MMP9 gene. Herein, we document the cardiomyocyte MEF2A interactome, which furthers our understanding of protein networks involved in the hierarchical control of normal and pathophysiological cardiomyocyte gene expression in the mammalian heart.
Collapse
Affiliation(s)
- Amira Moustafa
- Department of Biology, York University, Toronto, ON, M3J 1P3, Canada
- Muscle Health Research Centre (MHRC), York University, Toronto, ON, M3J 1P3, Canada
- Centre for Research in Biomolecular Interactions (CRBI), York University, Toronto, ON, M3J 1P3, Canada
| | - Sara Hashemi
- Analytical Sciences, Sanofi, Toronto, ON, M2R 3T4, Canada
- Seneca College, School of Health Sciences, King City, ON, L7B 1B3, Canada
| | - Gurnoor Brar
- Department of Biology, York University, Toronto, ON, M3J 1P3, Canada
- Muscle Health Research Centre (MHRC), York University, Toronto, ON, M3J 1P3, Canada
- Centre for Research in Biomolecular Interactions (CRBI), York University, Toronto, ON, M3J 1P3, Canada
| | - Jörg Grigull
- Department of Mathematics and Statistics, York University, Toronto, ON, M3J1P3, Canada
| | - Siemon H S Ng
- Analytical Sciences, Sanofi, Toronto, ON, M2R 3T4, Canada
- Analytical Development, Notch Therapeutics, Toronto, ON, M5G 1M1, Canada
| | - Declan Williams
- Tanz Centre for Research in Neurodegenerative Diseases, and Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5T 0S8, Canada
| | - Gerold Schmitt-Ulms
- Tanz Centre for Research in Neurodegenerative Diseases, and Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5T 0S8, Canada
| | - John C McDermott
- Department of Biology, York University, Toronto, ON, M3J 1P3, Canada.
- Muscle Health Research Centre (MHRC), York University, Toronto, ON, M3J 1P3, Canada.
- Centre for Research in Biomolecular Interactions (CRBI), York University, Toronto, ON, M3J 1P3, Canada.
| |
Collapse
|
9
|
Prameswari HS, Putra ICS, Raffaello WM, Nathaniel M, Suhendro AS, Khalid AF, Pranata R. Managing Covid-19 in patients with heart failure: current status and future prospects. Expert Rev Cardiovasc Ther 2022; 20:807-828. [PMID: 36185009 DOI: 10.1080/14779072.2022.2132230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 09/30/2022] [Indexed: 10/17/2022]
Abstract
INTRODUCTION COVID-19 may contribute to decompensation of previously stable chronic HF or cause a de-novo heart failure, which may come from the hyperinflammatory response and subsequent increase in metabolic demand. AREAS COVERED Two independent investigators searched MEDLINE (via PubMed), Europe PMC, and ScienceDirect databases with the following search terms: COVID-19, heart failure, COVID-19 drugs, heart failure drugs, and device therapy. All of the included full-text articles were rigorously evaluated by both authors in case there was disagreement about whether research should be included or not. In total, 157 studies were included and underwent extensive reading by the authors. EXPERT OPINION The World Health Organization (WHO) and the National Institute of Health (NIH) have published COVID-19 drug recommendations, although recommendations for HF-specific drug choices in COVID-19 are still lacking. We hope that this review can answer the void of comprehensive research data regarding the management options of HF in the COVID-19 condition so that clinicians can at least choose a more beneficial therapy or avoid combination therapies that have a high burden of side effects on HF; thus, morbidity and mortality in COVID-19 patients with HF may be reduced.
Collapse
Affiliation(s)
- Hawani Sasmaya Prameswari
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
| | - Iwan Cahyo Santosa Putra
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
| | | | - Michael Nathaniel
- School of Medicine and Health Sciences Atma Jaya Catholic University of Indonesia, Jakarta, Indonesia
| | - Adrian Sebastian Suhendro
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
| | - Achmad Fitrah Khalid
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
| | - Raymond Pranata
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
| |
Collapse
|
10
|
Liu X, Xie X, Luo M, Zhao Y, Li M, Peng F, Peng C. The synergistic compatibility mechanisms of fuzi against chronic heart failure in animals: A systematic review and meta-analysis. Front Pharmacol 2022; 13:954253. [PMID: 36188581 PMCID: PMC9515783 DOI: 10.3389/fphar.2022.954253] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 08/05/2022] [Indexed: 12/09/2022] Open
Abstract
Background: Fuzi’s compatibilities with other medicines are effective treatments for chronic heart failure. Pre-clinical animal experiments have indicated many possible synergistic compatibility mechanisms of it, but the results were not reliable and reproducible enough. Therefore, we performed this systematic review and meta-analysis of pre-clinical animal studies to integrate evidence, conducted both qualitative and quantitative evaluations of the compatibility and summarized potential synergistic mechanisms. Method: An exhaustive search was conducted for potentially relevant studies in nine online databases. The selection criteria were based on the Participants, Interventions, Control, Outcomes, and Study designs strategy. The SYRCLE risk of bias tool for animal trials was used to perform the methodological quality assessment. RevMan V.5.3 and STATA/SE 15.1 were used to perform the meta-analysis following the Cochrane Handbook for Systematic Reviews of Interventions. Result: 24 studies were included in the systematic review and meta-analysis. 12 outcomes were evaluated in the meta-analysis, including BNP, HR, HWI, ALD, LVEDP, LVSP, EF, FS, +dP/dtmax, −dP/dtmax, TNF-α and the activity of Na + -K + -ATPase. Subgroup analyses were performed depending on the modeling methods and duration. Conclusion: The synergistic Fuzi compatibility therapeutic effects against CHF animals were superior to those of Fuzi alone, as shown by improvements in cardiac function, resistance to ventricular remodeling and cardiac damage, regulation of myocardial energy metabolism disorder and RAAS, alleviation of inflammation, the metabolic process in vivo, and inhibition of cardiomyocyte apoptosis. Variations in CHF modeling methods and medication duration brought out possible model–effect and time-effect relationships.
Collapse
Affiliation(s)
- Xingyu Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaofang Xie
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- *Correspondence: Xiaofang Xie, ; Cheng Peng,
| | - Maozhu Luo
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yuting Zhao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Mengting Li
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Department of Pharmacy, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Fu Peng
- West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Cheng Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- *Correspondence: Xiaofang Xie, ; Cheng Peng,
| |
Collapse
|
11
|
Li W, Qu X, Kang X, Zhang H, Zhang X, Hu H, Yao L, Zhang L, Zheng J, Zheng Y, Zhang J, Xu Y. Silibinin eliminates mitochondrial ROS and restores autophagy through IL6ST/JAK2/STAT3 signaling pathway to protect cardiomyocytes from doxorubicin-induced injury. Eur J Pharmacol 2022; 929:175153. [PMID: 35839932 DOI: 10.1016/j.ejphar.2022.175153] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 06/26/2022] [Accepted: 07/08/2022] [Indexed: 11/26/2022]
Abstract
Growing evidence indicates that silibinin (SLB), a main component extracted from Chinese herb Silybum marianum, can effectively antagonize doxorubicin (DOX) induced myocardial injury (DIMI), but the specific molecular mechanism is still unelucidated. Herein, DOX induced human AC16 cardiomyocyte injury model and Network Pharmacology are used to predict and verify the potential mechanism. The analysis results of the core PPI network of SLB against DIMI show that JAK/STAT signaling pathway and autophagy are significantly enriched. Molecular docking results indicate that SLB has stronger binding ability to signaling key proteins IL6ST, JAK2 and STAT3 (affinity ≤ -7.0 kcal/mol). The detection results of pathway activation and autophagy level demonstrate that SLB significantly alleviates DOX induced IL6ST/JAK2/STAT3 signaling pathway inhibition and autophagy inhibition, reduces the death rate of cardiomyocytes. This protective effect of SLB is eliminated when key pathway proteins (IL6ST, JAK2, STAT3) are knocked down or autophagy is inhibited (3-MA or Beclin1 knockdown). These results suggest that the regulation of IL6ST/JAK2/STAT3 signaling pathway and autophagy may be important mechanism for SLB's protective effect on DOX injured cardiomyocytes. Further experimental results prove that knockdown of IL6ST, JAK2 and STAT3 eliminate the mitochondrial ROS scavenging effect and autophagy promoting effect of SLB. In sum, SLB can decrease the mitochondrial ROS and restore autophagy to antagonize DOX-induced cardiomyocyte injury by activating IL6ST/JAK2/STAT3 signaling pathway.
Collapse
Affiliation(s)
- Wenbiao Li
- Department of Biochemistry, School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Xinni Qu
- Department of Biochemistry, School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Xiangping Kang
- Department of Biochemistry, School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Haiyin Zhang
- Department of Biochemistry, School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Xueli Zhang
- Department of Biochemistry, School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Haiyan Hu
- Department of Biochemistry, School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Lingai Yao
- Department of Biochemistry, School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Lina Zhang
- Department of Biochemistry, School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Jing Zheng
- Department of Biochemistry, School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yuejuan Zheng
- Center for Traditional Chinese Medicine and Immunology Research, School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Jianghong Zhang
- Department of Radiation Biology, Institute of Radiation Medicine, Fudan University, Shanghai, 200032, China.
| | - Yanwu Xu
- Department of Biochemistry, School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
12
|
Li R, Nie Y. Letter by Li and Nie Regarding Article, "Inflammatory Glycoprotein 130 Signaling Links Changes in Microtubules and Junctophilin-2 to Altered Mitochondrial Metabolism and Right Ventricular Contractility". Circ Heart Fail 2022; 15:e009531. [PMID: 35758045 DOI: 10.1161/circheartfailure.122.009531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Ruopu Li
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yu Nie
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
13
|
Yao J, Ma F, Zhang L, Zhu C, Jumabay M, Yao Z, Wang L, Cai X, Zhang D, Qiao X, Shivkumar K, Pellegrini M, Yao Y, Wu X, Boström KI. Single-Cell RNA-Seq Identifies Dynamic Cardiac Transition Program from Adipose Derived Cells Induced by Leukemia Inhibitory Factor. Stem Cells 2022; 40:932-948. [PMID: 35896368 PMCID: PMC9585902 DOI: 10.1093/stmcls/sxac048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Accepted: 06/22/2022] [Indexed: 11/13/2022]
Abstract
Adipose-derived cells (ADCs) from white adipose tissue (WAT) are promising stem cell candidates because of their large regenerative reserves and the potential for cardiac regeneration. However, given the heterogeneity of ADC and its unsolved mechanisms of cardiac acquisition, ADC-cardiac transition efficiency remains low. In this study, we explored the heterogeneity of ADCs and the cellular kinetics of 39,432 single-cell transcriptomes along the leukemia inhibitory factor (LIF) induced ADC-cardiac transition. We identified distinct ADC subpopulations that reacted differentially to LIF when entering the cardiomyogenic program, further demonstrating that ADC-myogenesis is time-dependent and initiates from transient changes in nuclear factor erythroid 2-related factor 2 (Nrf2) signaling. At later stages, pseudotime analysis of ADCs navigated a trajectory with two branches corresponding to activated myofibroblast or cardiomyocyte-like cells. Our findings offer a high-resolution dissection of ADC heterogeneity and cell fate during ADC-cardiac transition, thus providing new insights into potential cardiac stem cells.
Collapse
Affiliation(s)
- Jiayi Yao
- Division of Cardiology, David Geffen School of Medicine at UCLA
| | - Feiyang Ma
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA.,Chongqing International Institute for Immunology, Chongqing 401338, China
| | - Li Zhang
- Division of Cardiology, David Geffen School of Medicine at UCLA
| | - Ching Zhu
- Division of Cardiology, David Geffen School of Medicine at UCLA
| | - Medet Jumabay
- Division of Allergy, Immunology Center for Immunity, Infection, and Inflammation Pediatrics, Dept of Medicine, University of California, San Diego, San Diego, CA
| | - Zehao Yao
- Peking Union Medical College, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
| | - Lumin Wang
- Institute of Precision Medicine, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xinjiang Cai
- Division of Cardiology, David Geffen School of Medicine at UCLA
| | - Daoqin Zhang
- Division of Cardiology, David Geffen School of Medicine at UCLA
| | - Xiaojing Qiao
- Division of Cardiology, David Geffen School of Medicine at UCLA
| | | | - Matteo Pellegrini
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA.,Dept of Molecular, Cell and Developmental Biology, University of California, Los Angeles, CA
| | - Yucheng Yao
- Division of Cardiology, David Geffen School of Medicine at UCLA
| | - Xiuju Wu
- Division of Cardiology, David Geffen School of Medicine at UCLA
| | - Kristina I Boström
- Division of Cardiology, David Geffen School of Medicine at UCLA.,Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA
| |
Collapse
|
14
|
Obana M, Maeda M, Fujio Y. Glycoprotein 130 orchestrates signal transduction network to promote cardiomyocyte proliferation for normal growth. Am J Physiol Heart Circ Physiol 2022; 323:H125-H127. [PMID: 35657617 DOI: 10.1152/ajpheart.00268.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Masanori Obana
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan.,Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiative (OTRI), Osaka University, Osaka, Japan.,Global Center for Medical Engineering and Informatics (MEI), Osaka University, Osaka, Japan
| | - Makiko Maeda
- Laboratory of Clinical Pharmacology and Therapeutics, 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.,Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiative (OTRI), Osaka University, Osaka, Japan.,Laboratory of Clinical Pharmacology and Therapeutics, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| |
Collapse
|
15
|
Kawagishi H, Nakada T, Numaga-Tomita T, Larrañaga M, Guo A, Song LS, Yamada M. Cytokine receptor gp130 promotes postnatal proliferation of cardiomyocytes required for the normal functional development of the heart. Am J Physiol Heart Circ Physiol 2022; 323:H103-H120. [PMID: 35594067 DOI: 10.1152/ajpheart.00698.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Mammalian ventricular cardiomyocytes are premature at birth and exhibit substantial phenotypic changes before weaning. Mouse ventricular myocytes undergo cell division several times after birth; however, the regulatory mechanisms and roles of cardiomyocyte division in postnatal heart development remain unclear. Here, we investigated the physiological role of gp130, the main subunit of multifunctional receptors for the IL-6 family of cytokines, in postnatal cardiomyocyte proliferation. Pharmacological inhibition of gp130 within the first month after birth induced significant systolic dysfunction of the left ventricle in mice. Consistently, mice with postnatal cardiomyocyte-specific gp130 depletion exhibited impaired left ventricular contractility compared to control mice. In these mice, cardiomyocytes exhibited a moderately decreased size and dramatically inhibited proliferation in the left ventricle but not in the right ventricle. Stereological analysis revealed that this change significantly decreased the number of cardiomyocytes in the left ventricle. Furthermore, IL-6 was mainly responsible for promoting ventricular cardiomyocyte proliferation by activating the JAK/STAT3 pathway. Taken together, the IL-6/gp130/JAK/STAT3 axis plays a crucial role in the physiological postnatal proliferation and hypertrophy of left ventricular cardiomyocytes to ensure normal cardiac functional development.
Collapse
Affiliation(s)
- Hiroyuki Kawagishi
- Department of Biotechnology, Institute for Biomedical Sciences, Shinshu University, Nagano, Japan.,Department of Molecular Pharmacology, Shinshu University School of Medicine, Nagano, Japan
| | - Tsutomu Nakada
- Department of Instrumental Analysis, Research Center for Supports to Advanced Science, Shinshu University, Nagano, Japan
| | - Takuro Numaga-Tomita
- Department of Molecular Pharmacology, Shinshu University School of Medicine, Nagano, Japan
| | - Maite Larrañaga
- Basic Sciences Department, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya, Catalunya, Spain
| | - Ang Guo
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota, United States
| | - Long-Sheng Song
- Division of Cardiovascular Medicine, Department of Internal Medicine and François M. Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine,Iowa City, Iowa, United States
| | - Mitsuhiko Yamada
- Department of Molecular Pharmacology, Shinshu University School of Medicine, Nagano, Japan
| |
Collapse
|
16
|
Mann DL. Targeting Inflammation in Pulmonary Artery Hypertension: Is It Time to Forget About the Lungs? Circ Heart Fail 2022; 15:e009290. [PMID: 34923831 PMCID: PMC8766894 DOI: 10.1161/circheartfailure.121.009290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- Douglas L Mann
- Center for Cardiovascular Research, Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
17
|
McElvaney OJ, Curley GF, Rose-John S, McElvaney NG. Interleukin-6: obstacles to targeting a complex cytokine in critical illness. THE LANCET. RESPIRATORY MEDICINE 2021; 9:643-654. [PMID: 33872590 PMCID: PMC8051931 DOI: 10.1016/s2213-2600(21)00103-x] [Citation(s) in RCA: 135] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 02/13/2021] [Accepted: 02/15/2021] [Indexed: 02/07/2023]
Abstract
Circulating concentrations of the pleiotropic cytokine interleukin-6 (IL-6) are known to be increased in pro-inflammatory critical care syndromes, such as sepsis and acute respiratory distress syndrome. Elevations in serum IL-6 concentrations in patients with severe COVID-19 have led to renewed interest in the cytokine as a therapeutic target. However, although the pro-inflammatory properties of IL-6 are widely known, the cytokine also has a series of important physiological and anti-inflammatory functions. An adequate understanding of the complex processes by which IL-6 signalling occurs is crucial for the correct interpretation of IL-6 concentrations in the blood or lung, the use of IL-6 as a critical care biomarker, or the design of effective anti-IL-6 strategies. Here, we outline the role of IL-6 in health and disease, explain the different types of IL-6 signalling and their contribution to the net biological effect of the cytokine, describe the approaches to IL-6 inhibition that are currently available, and discuss implications for the future use of treatments such as tocilizumab in the critical care setting.
Collapse
Affiliation(s)
- Oliver J McElvaney
- Department of Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland; Beaumont Hospital, Dublin, Ireland
| | - Gerard F Curley
- Beaumont Hospital, Dublin, Ireland; Department of Anaesthesia and Critical Care, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Stefan Rose-John
- Biochemical Institute, Christian-Albrechts-Universität zu Kiel, Kiel, Germany
| | - Noel G McElvaney
- Department of Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland; Beaumont Hospital, Dublin, Ireland.
| |
Collapse
|
18
|
Collura KM, Niu J, Sanders SS, Montersino A, Holland SM, Thomas GM. The palmitoyl acyltransferases ZDHHC5 and ZDHHC8 are uniquely present in DRG axons and control retrograde signaling via the Gp130/JAK/STAT3 pathway. J Biol Chem 2020; 295:15427-15437. [PMID: 32958558 PMCID: PMC7667964 DOI: 10.1074/jbc.ra120.013815] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Revised: 09/11/2020] [Indexed: 12/14/2022] Open
Abstract
Palmitoylation, the modification of proteins with the lipid palmitate, is a key regulator of protein targeting and trafficking. However, knowledge of the roles of specific palmitoyl acyltransferases (PATs), which catalyze palmitoylation, is incomplete. For example, little is known about which PATs are present in neuronal axons, although long-distance trafficking of palmitoyl-proteins is important for axon integrity and for axon-to-soma retrograde signaling, a process critical for axon development and for responses to injury. Identifying axonally targeted PATs might thus provide insights into multiple aspects of axonal biology. We therefore comprehensively determined the subcellular distribution of mammalian PATs in dorsal root ganglion (DRG) neurons and, strikingly, found that only two PATs, ZDHHC5 and ZDHHC8, were enriched in DRG axons. Signals via the Gp130/JAK/STAT3 and DLK/JNK pathways are important for axonal injury responses, and we found that ZDHHC5 and ZDHHC8 were required for Gp130/JAK/STAT3, but not DLK/JNK, axon-to-soma signaling. ZDHHC5 and ZDHHC8 robustly palmitoylated Gp130 in cotransfected nonneuronal cells, supporting the possibility that Gp130 is a direct ZDHHC5/8 substrate. In DRG neurons, Zdhhc5/8 shRNA knockdown reduced Gp130 palmitoylation and even more markedly reduced Gp130 surface expression, potentially explaining the importance of these PATs for Gp130-dependent signaling. Together, these findings provide new insights into the subcellular distribution and roles of specific PATs and reveal a novel mechanism by which palmitoylation controls axonal retrograde signaling.
Collapse
Affiliation(s)
- Kaitlin M Collura
- Shriners Hospitals Pediatric Research Center (Center for Neurorehabilitation and Neural Repair), Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania, USA
| | - Jingwen Niu
- Shriners Hospitals Pediatric Research Center (Center for Neurorehabilitation and Neural Repair), Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania, USA
| | - Shaun S Sanders
- Shriners Hospitals Pediatric Research Center (Center for Neurorehabilitation and Neural Repair), Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania, USA
| | - Audrey Montersino
- Shriners Hospitals Pediatric Research Center (Center for Neurorehabilitation and Neural Repair), Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania, USA
| | - Sabrina M Holland
- Shriners Hospitals Pediatric Research Center (Center for Neurorehabilitation and Neural Repair), Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania, USA
| | - Gareth M Thomas
- Shriners Hospitals Pediatric Research Center (Center for Neurorehabilitation and Neural Repair), Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania, USA; Department of Anatomy and Cell Biology, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania, USA.
| |
Collapse
|
19
|
Wang Y, Zhu S, Wei W, Tu Y, Chen C, Song J, Li J, Wang C, Xu Z, Sun S. Interleukin-6 knockout reverses macrophage differentiation imbalance and alleviates cardiac dysfunction in aging mice. Aging (Albany NY) 2020; 12:20184-20197. [PMID: 33099539 PMCID: PMC7655174 DOI: 10.18632/aging.103749] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 06/01/2020] [Indexed: 12/18/2022]
Abstract
Several interleukins (ILs) have been shown to be involved in aging, but the effects of IL-6 on aging-related cardiac dysfunction remain unknown. In this study, the expression and sources of cardiac IL-6 in aging hearts were investigated for the first time. The results showed that cardiac IL-6 expression in mice gradually increased with age, and the expression at 16 months, 20 months and 25 months was higher than that at 3 months. In addition, cardiac macrophages (Møs) were shown to be the main sources of IL-6 in aging mice. IL-6 knockout (KO) significantly alleviated cardiac dysfunction, increased M2 macrophage (Mø2) differentiation, reduced M1 macrophage (Mø1) differentiation and protected against cardiomyocyte apoptosis in aging mice. IL-6 KO also reversed the stimulatory effect of doxorubicin (DOX) treatment on Mø1s and the inhibitory effect of DOX treatment on Mø2s in vitro. Furthermore, the mRNA expression of both aging markers and apoptosis-related markers was markedly inhibited by IL-6 KO. Our results suggest that aging can be significantly reversed by IL-6 KO and that the mechanisms of this effect are related to alleviation of Mø1/Mø2 imbalance and protection against apoptosis in cardiomyocytes.
Collapse
Affiliation(s)
- Yuan Wang
- Department of Thyroid Breast Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Shan Zhu
- Department of Thyroid Breast Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Wen Wei
- Department of Thyroid Breast Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Yi Tu
- Department of Thyroid Breast Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Chuang Chen
- Department of Thyroid Breast Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Junlong Song
- Department of Thyroid Breast Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Juanjuan Li
- Department of Thyroid Breast Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Changhua Wang
- Basic Medical School of Wuhan University, Wuhan 430060, China
| | - Zhiliang Xu
- Department of Thyroid Breast Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Shengrong Sun
- Department of Thyroid Breast Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| |
Collapse
|
20
|
Metformin decreased myocardial fibrosis and apoptosis in hyperhomocysteinemia -induced cardiac hypertrophy. Curr Res Transl Med 2020; 69:103270. [PMID: 33268288 DOI: 10.1016/j.retram.2020.103270] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 08/23/2020] [Accepted: 10/04/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND Hyperhomocysteinemia (HHcy) is one of the major risk factors of cardiovascular diseases. Metformin acts as a cardioprotective role in several cardiovascular diseases, including ischemia/reperfusion, atherosclerosis, and myocardial infarction. However, whether metformin protects against HHcy-induced cardiac hypertrophy is unclear. METHODS AND RESULTS HHcy model was established in C57BL/6 mice with high L-methionine (L-MET) diet for 12 weeks. AC16 cells were exposed to homocysteine (Hcy) and then intervened with different concentrations of metformin in in vitro studies. The results showed that HHcy was able to induce cardiac hypertrophy, and metformin could abrogate this effect. HHcy increased the fibrosis area and induced apoptosis in the myocardium, whereas metformin could reverse the detrimental effects above. TUNEL assay showed that metformin was able to decrease Hcy-induced apoptosis in AC16 cells. Moreover, western blotting assay revealed that metformin could decrease Hcy-induced expression of Bax and cleaved caspase3, and increase the expression of Bcl-2. CONCLUSIONS This study demonstrates that metformin is able to attenuate HHcy-induced cardiac hypertrophy by decreasing myocardial fibrosis and apoptosis.
Collapse
|
21
|
Clinical Aspects of Janus Kinase (JAK) Inhibitors in the Cardiovascular System in Patients with Rheumatoid Arthritis. Int J Mol Sci 2020; 21:ijms21197390. [PMID: 33036382 PMCID: PMC7583966 DOI: 10.3390/ijms21197390] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 10/02/2020] [Accepted: 10/03/2020] [Indexed: 12/17/2022] Open
Abstract
Janus kinase (JAK) inhibitors, a novel class of targeted synthetic disease-modifying antirheumatic drugs (DMARDs), have shown their safety and efficacy in rheumatoid arthritis (RA) and are being intensively tested in other autoimmune and inflammatory disorders. Targeting several cytokines with a single small compound leads to blocking the physiological response of hundreds of genes, thereby providing the background to stabilize the immune response. Unfortunately, blocking many cytokines with a single drug may also bring some negative consequences. In this review, we focused on the activity of JAK inhibitors in the cardiovascular system of patients with RA. Special emphasis was put on the modification of heart performance, progression of atherosclerosis, lipid profile disturbance, and risk of thromboembolic complications. We also discussed potential pathophysiological mechanisms that may be responsible for such JAK inhibitor-associated side effects.
Collapse
|
22
|
Li Y, Feng J, Song S, Li H, Yang H, Zhou B, Li Y, Yue Z, Lian H, Liu L, Hu S, Nie Y. gp130 Controls Cardiomyocyte Proliferation and Heart Regeneration. Circulation 2020; 142:967-982. [PMID: 32600062 DOI: 10.1161/circulationaha.119.044484] [Citation(s) in RCA: 111] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Background:
A key cause of the high mortality of cardiovascular diseases is the cardiomyocyte inability to renew after cardiac injury. As a promising strategy to supplement functional myocytes for cardiac repair, there is a pressing need to understand the cellular and molecular mechanisms of heart regeneration.
Methods:
Seven genetic mouse lines were used: global OSM (oncostatin M) knockout, monocyte-/macrophage-specific OSM deletion, cardiomyocyte-specific lines, including OSM receptor deletion, gp130 (glycoprotein 130) deletion, gp130 activation, and Yap (yes-associated protein) ablation with gp130 activation mice. A series of molecular signaling experiments, including RNA sequencing, immunostaining, coimmunoprecipitation, and imaging flow cytometry, were conducted. Two models of cardiac injury, apical resection and myocardial infarction operation, were performed in neonatal, juvenile, and adult mice. Heart regeneration and cardiac function were evaluated by Masson staining and echocardiography, respectively. Gene recombinant adenovirus-associated virus was constructed and infected myocardial-infarcted mice as a gene therapy.
Results:
OSM was identified by RNA sequencing as a key upstream regulator of cardiomyocyte proliferation during neonatal heart regeneration in mice. Cardiomyocyte proliferation and heart regeneration were suspended in neonatal mice after cardiac injury when OSM was conditionally knockout in macrophages. The cardiomyocyte-specific deficiency of the OSM receptor heterodimers, OSM receptor and gp130, individually in cardiomyocytes reduced myocyte proliferation and neonatal heart regeneration. Conditional activation of gp130 in cardiomyocytes promoted cardiomyocyte proliferation and heart regeneration in juvenile and adult mice. Using RNA sequencing and functional screening, we found that Src mediated gp130-triggered cardiomyocyte proliferation by activating Yap (yes-associated protein) with Y357 phosphorylation independently of the Hippo pathway. Cardiomyocyte-specific deletion of Yap in
Myh6-gp130
ACT
mice blocked the effect of gp130 activation–induced heart regeneration in juvenile mice. Gene therapy with adenovirus-associated virus encoding constitutively activated gp130 promoted cardiomyocyte proliferation and heart regeneration in adult mice after myocardial infarction.
Conclusions:
Macrophage recruitment is essential for heart regeneration through the secretion of OSM, which promotes cardiomyocyte proliferation. As the coreceptor of OSM, gp130 activation is sufficient to promote cardiomyocyte proliferation by activating Yap through Src during heart regeneration. gp130 is a potential therapeutic target to improve heart regeneration after cardiac injury.
Collapse
Affiliation(s)
- Yandong Li
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing (Yandong Li, J.F., S.S., H. Li, H. Lian, L.L., S.H., Y.N.)
| | - Jie Feng
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing (Yandong Li, J.F., S.S., H. Li, H. Lian, L.L., S.H., Y.N.)
| | - Shen Song
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing (Yandong Li, J.F., S.S., H. Li, H. Lian, L.L., S.H., Y.N.)
| | - Haotong Li
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing (Yandong Li, J.F., S.S., H. Li, H. Lian, L.L., S.H., Y.N.)
| | - Huijun Yang
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, China (H.Y.)
| | - Bin Zhou
- State Key Laboratory of Cell Biology, Chinese Academy of Sciences (CAS) Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Shanghai (B.Z.)
| | - Yan Li
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, China (Yan Li)
| | - Zhang Yue
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (Z.Y.)
| | - Hong Lian
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing (Yandong Li, J.F., S.S., H. Li, H. Lian, L.L., S.H., Y.N.)
| | - Lihui Liu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing (Yandong Li, J.F., S.S., H. Li, H. Lian, L.L., S.H., Y.N.)
| | - Shengshou Hu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing (Yandong Li, J.F., S.S., H. Li, H. Lian, L.L., S.H., Y.N.)
| | - Yu Nie
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing (Yandong Li, J.F., S.S., H. Li, H. Lian, L.L., S.H., Y.N.)
| |
Collapse
|
23
|
Yan W, Dong ZC, Wang JJ, Zhang YL, Wang HX, Zhang B, Li HH. Deficiency of the Immunoproteasome LMP10 Subunit Attenuates Angiotensin II-Induced Cardiac Hypertrophic Remodeling via Autophagic Degradation of gp130 and IGF1R. Front Physiol 2020; 11:625. [PMID: 32581853 PMCID: PMC7296172 DOI: 10.3389/fphys.2020.00625] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 05/18/2020] [Indexed: 01/26/2023] Open
Abstract
Background/Aim Hypertensive cardiac hypertrophy is the leading cause of cardiac remodeling and heart failure. We recently demonstrated that the immunoproteasome, an inducible form of the constitutive proteasome, plays a critical role in regulating cardiovascular diseases. However, the role of the immunoproteasome LMP10 (β2i) catalytic subunit in the regulation of angiotensin II (Ang II)-induced cardiac hypertrophic remodeling remains unclear. Methods Wild-type (WT) and LMP10 knockout (KO) mice were infused with Ang II 1,000 ng/kg/min for 2 weeks. Blood pressure was measured using a tail-cuff system. Cardiac function and hypertrophic remodeling were examined by echocardiography and histological staining. The expression levels of genes and proteins were examined with quantitative real-time PCR and immunoblotting analysis, respectively. Results LMP10 mRNA and protein expression was significantly increased in Ang II-stimulated hearts and primary cardiomyocytes. Moreover, Ang II infusion for 2 weeks increased systolic blood pressure, abnormal cardiac function, hypertrophy, fibrosis, and inflammation in WT mice, which were significantly reversed in KO mice. Moreover, a marked reduction in the protein levels of insulin growth factor-1 receptor (IGF1R), glycoprotein 130 (gp130), and phosphorylated AKT, mTOR, STAT3, and ERK1/2 and an increase in the LC3II/I ratio were also observed in LMP10 KO mice compared with WT mice after Ang II infusion. In vitro culture experiments confirmed that LMP10 knockdown activated autophagy and increased IGF1R and gp130 degradation, leading to the inhibition of cardiomyocyte hypertrophy. However, inhibiting autophagy with chloroquine reversed this effect. Conclusion The results of this study indicate that LMP10 KO attenuates Ang II-induced cardiac hypertrophic remodeling via the autophagy-dependent degradation of IGF1R and gp130, and suggests that LMP10 may be a novel therapeutic target for hypertrophic heart diseases.
Collapse
Affiliation(s)
- Wen Yan
- Department of Emergency Medicine, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Zhi-Chao Dong
- Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jing-Jing Wang
- Department of Laboratory Animal Sciences, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Yun-Long Zhang
- Department of Emergency Medicine, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Hong-Xia Wang
- Department of Physiology and Physiopathology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Bo Zhang
- Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Hui-Hua Li
- Department of Emergency Medicine, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
24
|
Gozalo AS, Zerfas PM, Elkins WR, Gieseck RL. Retrospective Study of Intercalated Disk Defects Associated with Dilated Cardiomyopathy, Atrial Thrombosis, and Heart Failure in BALB/c Mice Deficient in IL4 Receptor α. Comp Med 2020; 70:266-276. [PMID: 32384942 PMCID: PMC7287387 DOI: 10.30802/aalas-cm-19-000059] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 07/01/2019] [Accepted: 10/22/2019] [Indexed: 12/21/2022]
Abstract
An increased incidence of dilated cardiomyopathy and atrial thrombosis was noted in a breeding colony of BALB/c mice deficient in IL4 receptor α. The condition affected mice of both sexes and of various ages, and extensive testing (microbiology, serology, histopathology) failed to ascertain the cause. Transmission electron microscopy of heart samples showed structural defects in the myocardial intercalated disks, characterized by unorganized and heavily convoluted arrangement with lower density and less prominent desmosomes and adherens junctions, widening of the intercellular space, myofibrillar lysis adjacent to intercalated disks, occasional sarcomere lysis with marked myofiber degeneration, vacuolation, accumulation of cell debris, and myelin figures. The intercalated disk contains cell adhesion molecules that form cell junctions, allowing contraction coupling of cardiomyocytes and the electrical and mechanical connection between cardiac fibers. Thus, defects at this level result in poor myocardial contraction, intracardiac blood stagnation, and consequently cardiac dilation with clinical signs of heart failure. The background strain or, potentially, the Cre-loxP-mediated recombination system used to create these mice may have contributed to the elevated incidence of cardiomyopathy and atrial thrombosis in this colony. Due to the backcrossing breeding scheme used, we cannot discount the emergence and colonywide dissemination of a spontaneous mutation that affects the intercalated disk. This report underscores the importance of carefully monitoring genetically modified mice colonies for unexpected phenotypes that may result from spontaneous or unintended mutations or enhanced strain background pathology.
Collapse
Affiliation(s)
- Alfonso S Gozalo
- Comparative Medicine Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland;,
| | - Patricia M Zerfas
- Pathology Service, Office of Research Services, National Institutes of Health, Bethesda, Maryland
| | - William R Elkins
- Comparative Medicine Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Richard L Gieseck
- Immunopathogenesis Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
25
|
Abstract
Heart failure (HF) is the leading cause of morbidity and mortality in developed countries, and it is the primary cause of mortality in the elderly worldwide. The processes of inflammatory response activation, production and release of pro-inflammatory cytokines, activation of the complement system, synthesis of autoantibodies, and overexpression of Class II major histocompatibility complex molecules contribute to the HF development and progression. High levels of circulating cytokines correlate with the severity of HF, measured with the use of New York Heart Association's classification, and prognosis of the disease. In HF, there is an imbalance between pro-inflammatory and anti-inflammatory cytokines. Concentrations of several interleukins are increased in HF, including IL-1β, IL-6, IL-8, IL-9, IL-10, IL-13, IL-17, and IL-18, whereas the levels of IL-5, IL-7, or IL-33 are down-regulated. Concentrations of inflammatory mediators are associated with cardiac function and can be HF markers and predictors of adverse outcomes or mortality. This review presents the role of interleukins, which contribute to the HF initiation and progression, the importance of their pathways in transition from myocardial injury to HF, and the role of interleukins as markers of disease severity and outcome predictors.
Collapse
|
26
|
Nakao S, Tsukamoto T, Ueyama T, Kawamura T. STAT3 for Cardiac Regenerative Medicine: Involvement in Stem Cell Biology, Pathophysiology, and Bioengineering. Int J Mol Sci 2020; 21:ijms21061937. [PMID: 32178385 PMCID: PMC7139789 DOI: 10.3390/ijms21061937] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 03/07/2020] [Accepted: 03/10/2020] [Indexed: 12/28/2022] Open
Abstract
Heart disease is the most common cause of death in developed countries, but the medical treatments for heart failure remain limited. In this context, the development of cardiac regeneration therapy for severe heart failure is important. Owing to their unique characteristics, including multiple differentiation and infinitive self-renewal, pluripotent stem cells can be considered as a novel source for regenerative medicine. Janus kinase/signal transducer and activator of transcription 3 (JAK/STAT3) signaling plays critical roles in the induction, maintenance, and differentiation of pluripotent stem cells. In the heart, JAK/STAT3 signaling has diverse cellular functions, including myocardial differentiation, cell cycle re-entry of matured myocyte after injury, and anti-apoptosis in pathological conditions. Therefore, regulating STAT3 activity has great potential as a strategy of cardiac regeneration therapy. In this review, we summarize the current understanding of STAT3, focusing on stem cell biology and pathophysiology, as they contribute to cardiac regeneration therapy. We also introduce a recently reported therapeutic strategy for myocardial regeneration that uses engineered artificial receptors that trigger endogenous STAT3 signal activation.
Collapse
Affiliation(s)
- Shu Nakao
- Department of Biomedical Sciences, College of Life Sciences, Ritsumeikan University, Kusatsu 525-8577, Japan; (S.N.); (T.T.); (T.U.)
- Ritsumeikan Global Innovation Research Institute, Ritsumeikan University, Kusatsu 525-8577, Japan
| | - Tasuku Tsukamoto
- Department of Biomedical Sciences, College of Life Sciences, Ritsumeikan University, Kusatsu 525-8577, Japan; (S.N.); (T.T.); (T.U.)
- Ritsumeikan Global Innovation Research Institute, Ritsumeikan University, Kusatsu 525-8577, Japan
| | - Tomoe Ueyama
- Department of Biomedical Sciences, College of Life Sciences, Ritsumeikan University, Kusatsu 525-8577, Japan; (S.N.); (T.T.); (T.U.)
- Ritsumeikan Global Innovation Research Institute, Ritsumeikan University, Kusatsu 525-8577, Japan
| | - Teruhisa Kawamura
- Department of Biomedical Sciences, College of Life Sciences, Ritsumeikan University, Kusatsu 525-8577, Japan; (S.N.); (T.T.); (T.U.)
- Ritsumeikan Global Innovation Research Institute, Ritsumeikan University, Kusatsu 525-8577, Japan
- Correspondence: ; Tel.: +81-75-599-4327
| |
Collapse
|
27
|
Arshad S, Naveed M, Ullia M, Javed K, Butt A, Khawar M, Amjad F. Targeting STAT-3 signaling pathway in cancer for development of novel drugs: Advancements and challenges. Genet Mol Biol 2020; 43:e20180160. [PMID: 32167126 PMCID: PMC7198026 DOI: 10.1590/1678-4685-gmb-2018-0160] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2018] [Accepted: 10/20/2018] [Indexed: 12/25/2022] Open
Abstract
Signal transducers and activators of transcription 3 (STAT-3) is a transcription
factor that regulates the gene expression of several target genes. These factors
are activated by the binding of cytokines and growth factors with STAT-3
specific receptors on cell membrane. Few years ago, STAT-3 was considered an
acute phase response element having several cellular functions such as
inflammation, cell survival, invasion, metastasis and proliferation, genetic
alteration, and angiogenesis. STAT-3 is activated by several types of
inflammatory cytokines, carcinogens, viruses, growth factors, and oncogenes.
Thus, the STAT3 pathway is a potential target for cancer therapeutics. Abnormal
STAT-3 activity in tumor development and cellular transformation can be targeted
by several genomic and pharmacological methodologies. An extensive review of the
literature has been conducted to emphasize the role of STAT-3 as a unique cancer
drug target. This review article discusses in detail the wide range of STAT-3
inhibitors that show antitumor effects both in vitro and
in vivo. Thus, targeting constitutive STAT-3 signaling is a
remarkable therapeutic methodology for tumor progression. Finally, current
limitations, trials and future perspectives of STAT-3 inhibitors are also
critically discussed.
Collapse
Affiliation(s)
- Sundas Arshad
- University of Lahore, Department of Allied Health Sciences, Gujrat Campus, Pakistan
| | - Muhammad Naveed
- University of Central Punjab, Faculty of life sciences, Department of Biotechnology, Lahore, Pakistan
| | - Mahad Ullia
- University of Gujrat, Department of Biochemistry and Biotechnology Sialkot sub Campus, Pakistan
| | - Khadija Javed
- University of Gujrat, Department of Biochemistry and Biotechnology Sialkot sub Campus, Pakistan
| | - Ayesha Butt
- University of Gujrat, Department of Biochemistry and Biotechnology Sialkot sub Campus, Pakistan
| | - Masooma Khawar
- University of Gujrat, Department of Biochemistry and Biotechnology Sialkot sub Campus, Pakistan
| | - Fazeeha Amjad
- University of Gujrat, Department of Biochemistry and Biotechnology Sialkot sub Campus, Pakistan
| |
Collapse
|
28
|
Takahashi M, Kinugawa S, Takada S, Kakutani N, Furihata T, Sobirin MA, Fukushima A, Obata Y, Saito A, Ishimori N, Iwabuchi K, Tsutsui H. The disruption of invariant natural killer T cells exacerbates cardiac hypertrophy and failure caused by pressure overload in mice. Exp Physiol 2020; 105:489-501. [PMID: 31957919 DOI: 10.1113/ep087652] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 01/17/2020] [Indexed: 12/18/2022]
Abstract
NEW FINDINGS What is the central question of this study? We questioned whether the disruption of invariant natural killer T (iNKT) cells exacerbates left ventricular (LV) remodelling and heart failure after transverse aortic constriction in mice. What are the main findings and their importance? Pressure overload induced by transverse aortic constriction increased the infiltration of iNKT cells in mouse hearts. The disruption of iNKT cells exacerbated LV remodelling and hastened the transition from hypertrophy to heart failure, in association with the activation of mitogen-activated protein kinase signalling. Activation of iNKT cells modulated the immunological balance in this process and played a protective role against LV remodelling and failure. ABSTRACT Chronic inflammation is involved in the development of cardiac remodelling and heart failure (HF). Invariant natural killer T (iNKT) cells, a subset of T lymphocytes, have been shown to produce various cytokines and orchestrate tissue inflammation. The pathophysiological role of iNKT cells in HF caused by pressure overload has not been studied. In the present study, we investigated whether the disruption of iNKT cells affected this process in mice. Transverse aortic constriction (TAC) and a sham operation were performed in male C57BL/6J wild-type (WT) and iNKT cell-deficient Jα18 knockout (KO) mice. The infiltration of iNKT cells was increased after TAC. The disruption of iNKT cells exacerbated left ventricular (LV) remodelling and hastened the transition to HF after TAC. Histological examinations also revealed that the disruption of iNKT cells induced greater myocyte hypertrophy and a greater increase in interstitial fibrosis after TAC. The expressions of interleukin-10 and tumour necrosis factor-α mRNA and their ratio in the LV after TAC were decreased in the KO compared with WT mice, which might indicate that the disruption of iNKT cells leads to an imbalance between T-helper type 1 and type 2 cytokines. The phosphorylation of extracellular signal-regulated kinase was significantly increased in the KO mice. The disruption of iNKT cells exacerbated the development of cardiac remodelling and HF after TAC. The activation of iNKT cells might play a protective role against HF caused by pressure overload. Targeting the activation of iNKT cells might thus be a promising candidate as a new therapeutic strategy for HF.
Collapse
Affiliation(s)
- Masashige Takahashi
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Shintaro Kinugawa
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Shingo Takada
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Naoya Kakutani
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Takaaki Furihata
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | | | - Arata Fukushima
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Yoshikuni Obata
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Akimichi Saito
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Naoki Ishimori
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Kazuya Iwabuchi
- Department of Immunobiology, Kitasato University School of Medicine, Kanagawa, Japan
| | - Hiroyuki Tsutsui
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
29
|
Krueger W, Bender N, Haeusler M, Henneberg M. The role of mechanotransduction in heart failure pathobiology-a concise review. Heart Fail Rev 2020; 26:981-995. [PMID: 31965473 DOI: 10.1007/s10741-020-09915-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
This review evaluates the role of mechanotransduction (MT) in heart failure (HF) pathobiology. Cardiac functional and structural modifications are regulated by biomechanical forces. Exposing cardiomyocytes and the myocardial tissue to altered biomechanical stress precipitates changes in the end-diastolic wall stress (EDWS). Thereby various interconnected biomolecular pathways, essentially mediated and orchestrated by MT, are launched and jointly contribute to adapt and remodel the myocardium. This cardiac MT-mediated feedback decisively determines the primary cardiac cellular and tissue response, the sort (concentric or eccentric) of hypertrophy/remodeling, to mechanical and/or hemodynamic alterations. Moreover, the altered EDWS affects the diastolic myocardial properties independent of the systolic function, and elevated EDWS causes diastolic dysfunction. The close interconnection between MT pathways and the cell nucleus, the genetic endowment, principally allows for the wide variety of phenotypic appearances. However, demographic, environmental features, comorbidities, and also the genetic make-up may modulate the phenotypic result. Cardiac MT takes a fundamental and superordinate position in the myocardial adaptation and remodeling processes in all HF categories and phenotypes. Therefore, the effects of MT should be integrated in all our scientific, clinical, and therapeutic considerations.
Collapse
Affiliation(s)
- Wolfgang Krueger
- Institute of Evolutionary Medicine, University of Zurich, Zurich, Switzerland. .,Medical University Department, Kantonsspital Aarau, Aarau, Switzerland.
| | - Nicole Bender
- Institute of Evolutionary Medicine, University of Zurich, Zurich, Switzerland
| | - Martin Haeusler
- Institute of Evolutionary Medicine, University of Zurich, Zurich, Switzerland
| | - Maciej Henneberg
- Institute of Evolutionary Medicine, University of Zurich, Zurich, Switzerland.,Adelaide Medical School, The University of Adelaide, Adelaide, Australia
| |
Collapse
|
30
|
Zhou X, Xia N, Lv B, Tang T, Nie S, Zhang M, Jiao J, Liu J, Xu C, Hou G, Yang X, Hu Y, Liao Y, Cheng X. Interleukin 35 ameliorates myocardial ischemia‐reperfusion injury by activating the gp130‐STAT3 axis. FASEB J 2020; 34:3224-3238. [PMID: 31917470 DOI: 10.1096/fj.201901718rr] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 12/17/2019] [Accepted: 12/20/2019] [Indexed: 12/18/2022]
Affiliation(s)
- Xingdi Zhou
- Department of Cardiology Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
- Key Laboratory of Biological Targeted Therapy of Education Ministry and Hubei Province Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Ni Xia
- Department of Cardiology Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
- Key Laboratory of Biological Targeted Therapy of Education Ministry and Hubei Province Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Bingjie Lv
- Department of Cardiology Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
- Key Laboratory of Biological Targeted Therapy of Education Ministry and Hubei Province Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Tingting Tang
- Department of Cardiology Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
- Key Laboratory of Biological Targeted Therapy of Education Ministry and Hubei Province Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Shaofang Nie
- Department of Cardiology Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
- Key Laboratory of Biological Targeted Therapy of Education Ministry and Hubei Province Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Min Zhang
- Department of Cardiology Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
- Key Laboratory of Biological Targeted Therapy of Education Ministry and Hubei Province Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Jiao Jiao
- Department of Cardiology Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
- Key Laboratory of Biological Targeted Therapy of Education Ministry and Hubei Province Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Jianfeng Liu
- Sino‐France Laboratory of cellular signaling, Key Laboratory of Molecular Biophysics of Ministry of Education College of Life Science and Technology and Collaborative Innovation Center for Genetics and Development Huazhong University of Science and Technology Wuhan Hubei China
| | - Chanjuan Xu
- Sino‐France Laboratory of cellular signaling, Key Laboratory of Molecular Biophysics of Ministry of Education College of Life Science and Technology and Collaborative Innovation Center for Genetics and Development Huazhong University of Science and Technology Wuhan Hubei China
| | - Guofei Hou
- Sino‐France Laboratory of cellular signaling, Key Laboratory of Molecular Biophysics of Ministry of Education College of Life Science and Technology and Collaborative Innovation Center for Genetics and Development Huazhong University of Science and Technology Wuhan Hubei China
| | - Xiangping Yang
- School of Basic Medicine Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Yu Hu
- Key Laboratory of Biological Targeted Therapy of Education Ministry and Hubei Province Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
- Institute of Hematology Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Yuhua Liao
- Department of Cardiology Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
- Key Laboratory of Biological Targeted Therapy of Education Ministry and Hubei Province Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Xiang Cheng
- Department of Cardiology Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
- Key Laboratory of Biological Targeted Therapy of Education Ministry and Hubei Province Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| |
Collapse
|
31
|
Bertero E, Ameri P, Maack C. Bidirectional Relationship Between Cancer and Heart Failure: Old and New Issues in Cardio-oncology. Card Fail Rev 2019; 5:106-111. [PMID: 31179021 PMCID: PMC6546001 DOI: 10.15420/cfr.2019.1.2] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 02/14/2019] [Indexed: 02/07/2023] Open
Abstract
The main focus of cardio-oncology has been the prevention and treatment of the cardiac toxicity of chemotherapy and radiotherapy. Furthermore, several targeted therapies have been associated with unexpected cardiotoxic side-effects. Recently, epidemiological studies reported a higher incidence of cancer in patients with heart failure (HF) compared with individuals without HF. On this basis, it has been proposed that HF might represent an oncogenic condition. This hypothesis is supported by preclinical studies demonstrating that hyperactivation of the sympathetic nervous system and renin-angiotensin-aldosterone system, which is a hallmark of HF, promotes cancer growth and dissemination. Another intriguing possibility is that the co-occurrence of HF and cancer is promoted by a common pathological milieu characterised by a state of chronic low-grade inflammation, which predisposes to both diseases. In this review, we provide an overview of the mechanisms underlying the bidirectional relationship between HF and cancer.
Collapse
Affiliation(s)
- Edoardo Bertero
- Comprehensive Heart Failure Center, University Clinic Würzburg Würzburg, Germany
| | - Pietro Ameri
- Cardiovascular Disease Unit, IRCCS Ospedale Policlinico San Martino - IRCCS Italian Cardiovascular Network Genova, Italy.,Department of Internal Medicine and Centre of Excellence for Biomedical Research, University of Genova Genova, Italy
| | - Christoph Maack
- Comprehensive Heart Failure Center, University Clinic Würzburg Würzburg, Germany
| |
Collapse
|
32
|
Zelt JG, Chaudhary KR, Cadete VJ, Mielniczuk LM, Stewart DJ. Medical Therapy for Heart Failure Associated With Pulmonary Hypertension. Circ Res 2019; 124:1551-1567. [DOI: 10.1161/circresaha.118.313650] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Jason G.E. Zelt
- From the Division of Cardiology, University of Ottawa Heart Institute (J.G.E.Z., L.M.M., D.J.S.), University of Ottawa, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine (J.G.E.Z., K.R.C., V.J.C., L.M.M., D.J.S.), University of Ottawa, Canada
| | - Ketul R. Chaudhary
- Department of Cellular and Molecular Medicine, Faculty of Medicine (J.G.E.Z., K.R.C., V.J.C., L.M.M., D.J.S.), University of Ottawa, Canada
- Sinclair Centre for Regenerative Medicine, Ottawa Hospital Research Institute, Canada (K.R.C., V.J.C., D.J.S.)
| | - Virgilio J. Cadete
- Department of Cellular and Molecular Medicine, Faculty of Medicine (J.G.E.Z., K.R.C., V.J.C., L.M.M., D.J.S.), University of Ottawa, Canada
- Sinclair Centre for Regenerative Medicine, Ottawa Hospital Research Institute, Canada (K.R.C., V.J.C., D.J.S.)
| | - Lisa M. Mielniczuk
- From the Division of Cardiology, University of Ottawa Heart Institute (J.G.E.Z., L.M.M., D.J.S.), University of Ottawa, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine (J.G.E.Z., K.R.C., V.J.C., L.M.M., D.J.S.), University of Ottawa, Canada
| | - Duncan J. Stewart
- From the Division of Cardiology, University of Ottawa Heart Institute (J.G.E.Z., L.M.M., D.J.S.), University of Ottawa, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine (J.G.E.Z., K.R.C., V.J.C., L.M.M., D.J.S.), University of Ottawa, Canada
- Sinclair Centre for Regenerative Medicine, Ottawa Hospital Research Institute, Canada (K.R.C., V.J.C., D.J.S.)
| |
Collapse
|
33
|
Galat A, Guellich A, Bodez D, Lipskaia L, Moutereau S, Bergoend E, Hüe S, Ternacle J, Mohty D, Monin JL, Derumeaux G, Radu C, Damy T. Causes and consequences of cardiac fibrosis in patients referred for surgical aortic valve replacement. ESC Heart Fail 2019; 6:649-657. [PMID: 31115164 PMCID: PMC6676299 DOI: 10.1002/ehf2.12451] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 03/02/2019] [Accepted: 04/21/2019] [Indexed: 12/25/2022] Open
Abstract
Aims Cardiac fibrosis is associated with left ventricular (LV) remodelling and contractile dysfunction in aortic stenosis (AS). The fibrotic process in this condition is still unclear. The aim of this study was to determine the role of both local and systemic inflammation as underlying mechanisms of LV fibrosis and contractile dysfunction. The diagnostic values of 2D‐strain echocardiography and serum biomarkers in the evaluation of cardiac fibrosis in this condition were assessed through correlation analyses. Methods and results Patients with AS referred for surgical valve replacement were prospectively and consecutively included. They all had a comprehensive echocardiography including 2D strain. Blood samples were collected to measure cytokines and inflammatory biomarkers using Luminex bead‐based assays. A per‐surgical myocardial biopsy of the basal antero‐septal segment (S1) was performed. Serial sections of each biopsy were stained with Sirius red. Digital image analysis was used to quantify fibrosis. Immunostainings using specific antibodies against macrophage, glycoprotein (gp) 130, and interleukin 6 (IL‐6) were also performed. Patients were divided into tertiles reflecting the severity of fibrosis: mild, moderate, and severe load (TF1 to TF3). The mean age of the 58 included patients was 73 ± 11 years. Twenty‐four (43%) were in New York Heart Association III–IV. Mean aortic valve area was 0.8 ± 0.2 cm2. Mean aortic stenosis peak velocity and mean gradient were respectively 4.5 ± 0.8 m/s and 54 ± 15 mmHg. The mean LV ejection fraction was 54 ± 12%, and the global LV longitudinal strain was −15 ± 4%. The mean S1 strain, corresponding to the biopsied region, was −10 ± 6% and was strongly correlated to fibrosis load (R = 0.83, P < 0.0001). TF3 was associated with higher mortality (P = 0.009), higher serum C‐reactive protein and IL‐6, and lower gp130 compared with the other tertiles (P < 0.05). IL‐6 and gp130 were expressed in the heart and respectively in the plasma membrane of macrophages and in the cytoplasm of both macrophages and cardiomyocytes. During follow‐up, three patients died and were all in the third fibrosis tertile. Conclusions We found a positive correlation between elevated inflammatory markers and degree of fibrosis load. These two parameters were associated with worse outcomes in patients with severe AS. Our results may be of interest especially in patients for whom a transcatheter aortic valve implantation is indicated and myocardial biopsy is not possible. Strategies aiming at preventing inflammation might be considered to decrease or limit the progression of cardiac fibrosis in patients followed for AS.
Collapse
Affiliation(s)
- Arnault Galat
- UPEC, AP-HP, Henri-Mondor Teaching Hospital, Créteil, France.,Department of Cardiology, AP-HP, Henri-Mondor Teaching Hospital, Créteil, France.,Département Hospitalo-Universitaire Ageing Thorax-Vessels-Blood (DHU ATVB), Créteil, France.,GRC Amyloid Research Institute, IMRB/INSERM U955, Créteil, France
| | - Aziz Guellich
- UPEC, AP-HP, Henri-Mondor Teaching Hospital, Créteil, France.,Department of Cardiology, AP-HP, Henri-Mondor Teaching Hospital, Créteil, France.,Département Hospitalo-Universitaire Ageing Thorax-Vessels-Blood (DHU ATVB), Créteil, France.,GRC Amyloid Research Institute, IMRB/INSERM U955, Créteil, France
| | - Diane Bodez
- UPEC, AP-HP, Henri-Mondor Teaching Hospital, Créteil, France.,Department of Cardiology, AP-HP, Henri-Mondor Teaching Hospital, Créteil, France.,Département Hospitalo-Universitaire Ageing Thorax-Vessels-Blood (DHU ATVB), Créteil, France.,GRC Amyloid Research Institute, IMRB/INSERM U955, Créteil, France
| | - Larissa Lipskaia
- UPEC, AP-HP, Henri-Mondor Teaching Hospital, Créteil, France.,Département Hospitalo-Universitaire Ageing Thorax-Vessels-Blood (DHU ATVB), Créteil, France.,GRC Amyloid Research Institute, IMRB/INSERM U955, Créteil, France
| | - Stéphane Moutereau
- UPEC, AP-HP, Henri-Mondor Teaching Hospital, Créteil, France.,Department of Biochemistry, AP-HP, Henri-Mondor Teaching Hospital, Créteil, France
| | - Eric Bergoend
- UPEC, AP-HP, Henri-Mondor Teaching Hospital, Créteil, France.,Département Hospitalo-Universitaire Ageing Thorax-Vessels-Blood (DHU ATVB), Créteil, France.,Department of Cardiovascular Surgery, AP-HP, Henri-Mondor Teaching Hospital, Créteil, France
| | - Sophie Hüe
- UPEC, AP-HP, Henri-Mondor Teaching Hospital, Créteil, France.,Department of Immunology, AP-HP, Henri-Mondor Teaching Hospital, Créteil, France
| | - Julien Ternacle
- UPEC, AP-HP, Henri-Mondor Teaching Hospital, Créteil, France.,Department of Cardiology, AP-HP, Henri-Mondor Teaching Hospital, Créteil, France.,Département Hospitalo-Universitaire Ageing Thorax-Vessels-Blood (DHU ATVB), Créteil, France.,GRC Amyloid Research Institute, IMRB/INSERM U955, Créteil, France
| | - Dania Mohty
- Department of Cardiology, Dupuytren Hospital, CHU Limoges, Pôle Cœur-Poumon-Rein, Limoges, France
| | - Jean-Luc Monin
- UPEC, AP-HP, Henri-Mondor Teaching Hospital, Créteil, France.,Department of Cardiology, AP-HP, Henri-Mondor Teaching Hospital, Créteil, France.,Département Hospitalo-Universitaire Ageing Thorax-Vessels-Blood (DHU ATVB), Créteil, France.,GRC Amyloid Research Institute, IMRB/INSERM U955, Créteil, France
| | - Geneviève Derumeaux
- UPEC, AP-HP, Henri-Mondor Teaching Hospital, Créteil, France.,Department of Cardiology, AP-HP, Henri-Mondor Teaching Hospital, Créteil, France.,Département Hospitalo-Universitaire Ageing Thorax-Vessels-Blood (DHU ATVB), Créteil, France.,GRC Amyloid Research Institute, IMRB/INSERM U955, Créteil, France
| | - Costin Radu
- UPEC, AP-HP, Henri-Mondor Teaching Hospital, Créteil, France.,Département Hospitalo-Universitaire Ageing Thorax-Vessels-Blood (DHU ATVB), Créteil, France.,Department of Cardiovascular Surgery, AP-HP, Henri-Mondor Teaching Hospital, Créteil, France
| | - Thibaud Damy
- UPEC, AP-HP, Henri-Mondor Teaching Hospital, Créteil, France.,Department of Cardiology, AP-HP, Henri-Mondor Teaching Hospital, Créteil, France.,Département Hospitalo-Universitaire Ageing Thorax-Vessels-Blood (DHU ATVB), Créteil, France.,GRC Amyloid Research Institute, IMRB/INSERM U955, Créteil, France.,Inserm, Clinical Investigation Centre 1430, AP-HP, Henri Mondor Teaching Hospital, Créteil, France
| |
Collapse
|
34
|
Murakami M, Kamimura D, Hirano T. Pleiotropy and Specificity: Insights from the Interleukin 6 Family of Cytokines. Immunity 2019; 50:812-831. [DOI: 10.1016/j.immuni.2019.03.027] [Citation(s) in RCA: 231] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 03/25/2019] [Accepted: 03/26/2019] [Indexed: 02/08/2023]
|
35
|
Chen D, Li Z, Bao P, Chen M, Zhang M, Yan F, Xu Y, Ji C, Hu X, Sanchis D, Zhang Y, Ye J. Nrf2 deficiency aggravates Angiotensin II-induced cardiac injury by increasing hypertrophy and enhancing IL-6/STAT3-dependent inflammation. Biochim Biophys Acta Mol Basis Dis 2019; 1865:1253-1264. [PMID: 30668979 DOI: 10.1016/j.bbadis.2019.01.020] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 01/07/2019] [Accepted: 01/16/2019] [Indexed: 12/17/2022]
Abstract
BACKGROUND NF-E2-related factor 2 (Nrf2) is a transcription factor playing cytoprotective effects in various pathological processes including oxidative stress and cardiac hypertrophy. Despite being a potential therapeutic target to treat several cardiomyopathies, the signaling underlying Nrf2-dependent cardioprotective action remains largely uncharacterized. AIM This study aimed to explore the signaling mediating the role of Nrf2 in the development of hypertensive cardiac pathogenesis by analyzing the response to Angiotensin II (Ang II) in the presence or absence of Nrf2 expression, both in vivo and in vitro. RESULTS Our results indicated that Nrf2 deficiency exacerbated cardiac damage triggered by Ang II infusion. Mechanistically, our study shows that Ang II-triggered hypertrophy and inflammation is exacerbated in the absence of Nrf2 expression and points to the involvement of the IL-6/STAT3 signaling pathway in this event. Indeed, our results show that IL-6 abundance triggered by Ang II is increased in the absence of Nrf2 and demonstrate the requirement of IL-6 in STAT3 activation and cardiac inflammation induced by Ang II. CONCLUSION Our results show that Nrf2 is important for the protection of the heart against Ang II-induced cardiac hypertrophy and inflammation by mechanisms involving the regulation of IL-6/STAT3-dependent signaling.
Collapse
Affiliation(s)
- Dandan Chen
- State Key Laboratory of Natural Medicines, Department of Biochemistry, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210006, China
| | - Zhe Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Cardiovascular research Institute, Wuhan University, Wuhan 430060, China; Hubei key Laboratory of Cardiology, Wuhan 430060, China
| | - Peiqing Bao
- State Key Laboratory of Natural Medicines, Department of Biochemistry, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210006, China
| | - Miao Chen
- State Key Laboratory of Natural Medicines, Department of Biochemistry, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210006, China
| | - Miao Zhang
- State Key Laboratory of Natural Medicines, Department of Biochemistry, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210006, China
| | - Fangrong Yan
- Research Center of Biostatistics and Computational Pharmacy, China Pharmaceutical University, Nanjing 210006, China
| | - Yitao Xu
- Division of Cancer, Department of Surgery and Cancer, Imperial College London, London W120NN, United Kingdom
| | - Caoyu Ji
- Research Center of Biostatistics and Computational Pharmacy, China Pharmaceutical University, Nanjing 210006, China
| | - Xinyue Hu
- State Key Laboratory of Natural Medicines, Department of Biochemistry, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210006, China
| | - Daniel Sanchis
- Institut de Recerca Biomedica de Lleida (IRBLLEIDA), Universitat de Lleida, Edifici Biomedicina-I. Av. Rovira Roure, 80, 25198 Lleida, Spain.
| | - Yubin Zhang
- State Key Laboratory of Natural Medicines, Department of Biochemistry, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210006, China.
| | - Junmei Ye
- State Key Laboratory of Natural Medicines, Department of Biochemistry, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210006, China.
| |
Collapse
|
36
|
Raso A, Dirkx E, Philippen LE, Fernandez-Celis A, De Majo F, Sampaio-Pinto V, Sansonetti M, Juni R, El Azzouzi H, Calore M, Bitsch N, Olieslagers S, Oerlemans MIFJ, Huibers MM, de Weger RA, Reckman YJ, Pinto YM, Zentilin L, Zacchigna S, Giacca M, da Costa Martins PA, López-Andrés N, De Windt LJ. Therapeutic Delivery of miR-148a Suppresses Ventricular Dilation in Heart Failure. Mol Ther 2018; 27:584-599. [PMID: 30559069 PMCID: PMC6403487 DOI: 10.1016/j.ymthe.2018.11.011] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 10/31/2018] [Accepted: 11/11/2018] [Indexed: 01/02/2023] Open
Abstract
Heart failure is preceded by ventricular remodeling, changes in left ventricular mass, and myocardial volume after alterations in loading conditions. Concentric hypertrophy arises after pressure overload, involves wall thickening, and forms a substrate for diastolic dysfunction. Eccentric hypertrophy develops in volume overload conditions and leads wall thinning, chamber dilation, and reduced ejection fraction. The molecular events underlying these distinct forms of cardiac remodeling are poorly understood. Here, we demonstrate that miR-148a expression changes dynamically in distinct subtypes of heart failure: while it is elevated in concentric hypertrophy, it decreased in dilated cardiomyopathy. In line, antagomir-mediated silencing of miR-148a caused wall thinning, chamber dilation, increased left ventricle volume, and reduced ejection fraction. Additionally, adeno-associated viral delivery of miR-148a protected the mouse heart from pressure-overload-induced systolic dysfunction by preventing the transition of concentric hypertrophic remodeling toward dilation. Mechanistically, miR-148a targets the cytokine co-receptor glycoprotein 130 (gp130) and connects cardiomyocyte responsiveness to extracellular cytokines by modulating the Stat3 signaling. These findings show the ability of miR-148a to prevent the transition of pressure-overload induced concentric hypertrophic remodeling toward eccentric hypertrophy and dilated cardiomyopathy and provide evidence for the existence of separate molecular programs inducing distinct forms of myocardial remodeling.
Collapse
Affiliation(s)
- Andrea Raso
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, the Netherlands
| | - Ellen Dirkx
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, the Netherlands; International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - Leonne E Philippen
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, the Netherlands; Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA
| | - Amaya Fernandez-Celis
- Cardiovascular Translational Research, Navarrabiomed, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Federica De Majo
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, the Netherlands
| | - Vasco Sampaio-Pinto
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, the Netherlands; Instituto de Investigação e Inovação em Saúde (i3S), Porto, Portugal; Instituto Nacional de Engenharia Biomédica (INEB), Porto, Portugal; Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| | - Marida Sansonetti
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, the Netherlands
| | - Rio Juni
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, the Netherlands
| | - Hamid El Azzouzi
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, the Netherlands; Departments of Cardiology and Pathology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Martina Calore
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, the Netherlands
| | - Nicole Bitsch
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, the Netherlands
| | - Servé Olieslagers
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, the Netherlands
| | - Martinus I F J Oerlemans
- Departments of Cardiology and Pathology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Manon M Huibers
- Departments of Cardiology and Pathology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Roel A de Weger
- Departments of Cardiology and Pathology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Yolan J Reckman
- Department of Experimental Cardiology, Amsterdam UMC location AMC, Amsterdam, the Netherlands
| | - Yigal M Pinto
- Department of Experimental Cardiology, Amsterdam UMC location AMC, Amsterdam, the Netherlands
| | - Lorena Zentilin
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - Serena Zacchigna
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy; Department of Medicine, Surgery and Health Sciences, University of Trieste, Italy
| | - Mauro Giacca
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy; Department of Medicine, Surgery and Health Sciences, University of Trieste, Italy
| | - Paula A da Costa Martins
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, the Netherlands; Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Natalia López-Andrés
- Cardiovascular Translational Research, Navarrabiomed, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Leon J De Windt
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, the Netherlands.
| |
Collapse
|
37
|
Gomez I, Duval V, Silvestre JS. Cardiomyocytes and Macrophages Discourse on the Method to Govern Cardiac Repair. Front Cardiovasc Med 2018; 5:134. [PMID: 30333983 PMCID: PMC6175999 DOI: 10.3389/fcvm.2018.00134] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 09/05/2018] [Indexed: 12/11/2022] Open
Abstract
In response to pathophysiological stress, the cardiac tissue undergoes profound remodeling process that incorporates the elimination of dying resident cells, compensatory hypertrophy of functional cardiomyocytes, growth and remodeling of the vascular compartment and formation of a fibrotic scar. Accumulating evidences indicate that cardiac remodeling is, at least in part, controlled by a complex crosstalk between cardiomyocytes and macrophages. The strategic location of abundant macrophages to the proximity of cardiomyocytes suggest that they could regulate the fate of cardiomyocytes in the injured heart. As such, macrophages appear as critical support cells for cardiomyocytes and play central roles in cardiac hypertrophy, fibrosis and remodeling. Notably, the cardiac tissue expands heterogeneous population of cardiac macrophages through local proliferation of resident macrophage as well as recruitment and differentiation of blood-derived monocytes. It has also been suggested that cardiac-resident macrophages display distinct functional properties from that of monocyte-derived macrophages in cardiac tissue. Furthermore, macrophages are an overflowing source of biological entities with non-canonical roles on cardiac conduction or cardiomyocyte proliferation by regulating action potential diffusion or cardiac cell cycle reentry. Alternatively, stressed cardiomyocytes can trigger the release of a broad repertoire of instructive signals that can regulate macrophage number, skew their phenotype and therefore direct their beneficial or deleterious actions. In this review, we highlight recent discoveries describing how the intricate dialogue between cardiomyocytes and macrophages can shape the deleterious or healing signaling mechanisms in the injured cardiac tissue.
Collapse
Affiliation(s)
- Ingrid Gomez
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS-970, Paris Centre de Recherche Cardiovasculaire, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Vincent Duval
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS-970, Paris Centre de Recherche Cardiovasculaire, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Jean-Sébastien Silvestre
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS-970, Paris Centre de Recherche Cardiovasculaire, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| |
Collapse
|
38
|
Zhong W, Jin W, Xu S, Wu Y, Luo S, Liang M, Chen L. Pioglitazone Induces Cardiomyocyte Apoptosis and Inhibits Cardiomyocyte Hypertrophy Via VEGFR-2 Signaling Pathway. Arq Bras Cardiol 2018; 111:162-169. [PMID: 29972411 PMCID: PMC6122905 DOI: 10.5935/abc.20180108] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 03/14/2018] [Indexed: 01/02/2023] Open
Abstract
Background Pioglitazone has been widely used as an insulin-sensitizing agent for
improving glycemic control in patients with type 2 diabetes mellitus.
However, cardiovascular risk and protective effects of pioglitazone remain
controversial. Objectives In this study, we investigated whether pioglitazone affects cardiomyocyte
apoptosis and hypertrophy by regulating the VEGFR-2 signaling pathway. Methods Cardiomyocytes were enzymatically isolated from 1- to 3-day-old
Sprague-Dawley rat ventricles. Effects of pioglitazone and the
VEGFR-2-selective inhibitor apatinib on cardiomyocyte apoptotic rate was
determined using flow cytometry, and hypertrophy was evaluated using
[3H]-leucine incorporation. The protein expressions of
unphosphorylated and phosphorylated VEGFR-2, Akt, P53, and mTOR were
determined by Western-Blotting. Analysis of variance (ANOVA) was used to
assess the differences between groups. Results Pioglitazone and VEGFR-2-selective inhibitor apatinib reduced rat
cardiomyocyte viability and cardiomyocyte hypertrophy induced by angiotensin
II in vitro. Furthermore, in the same in vitro model, pioglitazone and
apatinib significantly increased the expression of Bax and phosphorylated
P53 and decreased the expression of phosphorylated VEGFR-2, Akt, and mTOR,
which promote cardiomyocyte hypertrophy. Conclusions These findings indicate that pioglitazone induces cardiomyocyte apoptosis and
inhibits cardiomyocyte hypertrophy by modulating the VEGFR-2 signaling
pathway.
Collapse
Affiliation(s)
- Wenliang Zhong
- Department of Cardiology, The First Hospital of Nanping City, affiliated to Fujian Medical University, Nanping, Fujian - China.,Department of Cardiology, Union Hospital, Fujian Medical University, Fuzhou, Fujian - China
| | - Wen Jin
- Cardiovascular Department, Guangdong N°.2 Provincial People's Hospital, Guangzhou, Guangdong - China
| | - Shanghua Xu
- Department of Cardiology, The First Hospital of Nanping City, affiliated to Fujian Medical University, Nanping, Fujian - China
| | - Yanqing Wu
- Department of Cardiology, The First Hospital of Nanping City, affiliated to Fujian Medical University, Nanping, Fujian - China
| | - Shunxiang Luo
- Department of Cardiology, The First Hospital of Nanping City, affiliated to Fujian Medical University, Nanping, Fujian - China
| | - Minlie Liang
- Department of Cardiology, The First Hospital of Nanping City, affiliated to Fujian Medical University, Nanping, Fujian - China
| | - Lianglong Chen
- Department of Cardiology, Union Hospital, Fujian Medical University, Fuzhou, Fujian - China
| |
Collapse
|
39
|
Abstract
Interleukin-6 (IL-6) is a pivotal cytokine with a diverse repertoire of physiological functions that include regulation of immune cell proliferation and differentiation. Dysregulation of IL-6 signalling is associated with inflammatory and lymphoproliferative disorders such as rheumatoid arthritis and Castleman disease, and several classes of therapeutics have been developed that target components of the IL-6 signalling pathway. So far, monoclonal antibodies against IL-6 or IL-6 receptor (IL-6R) and Janus kinases (JAK) inhibitors have been successfully developed for the treatment of autoimmune diseases such as rheumatoid arthritis. However, clinical trials of agents targeting IL-6 signalling have also raised questions about the diseases and patient populations for which such agents have an appropriate benefit-risk profile. Knowledge from clinical trials and advances in our understanding of the complexities of IL-6 signalling, including the potential to target an IL-6 trans-signalling pathway, are now indicating novel opportunities for therapeutic intervention. In this Review, we overview the roles of IL-6 in health and disease and analyse progress with several approaches of inhibiting IL-6-signalling, with the aim of illuminating when and how to apply IL-6 blockade.
Collapse
|
40
|
|
41
|
Affiliation(s)
- Thomas Eschenhagen
- Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Hamburg, Germany; and DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Germany
| |
Collapse
|
42
|
Chen F, Chen D, Zhang Y, Jin L, Zhang H, Wan M, Pan T, Wang X, Su Y, Xu Y, Ye J. Interleukin-6 deficiency attenuates angiotensin II-induced cardiac pathogenesis with increased myocyte hypertrophy. Biochem Biophys Res Commun 2017; 494:534-541. [DOI: 10.1016/j.bbrc.2017.10.119] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 10/23/2017] [Indexed: 11/25/2022]
|
43
|
Leukemia Inhibitory Factor Increases Survival of Pluripotent Stem Cell-Derived Cardiomyocytes. J Cardiovasc Transl Res 2017; 11:1-13. [PMID: 29019149 DOI: 10.1007/s12265-017-9769-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Accepted: 10/02/2017] [Indexed: 01/22/2023]
Abstract
Leukemia inhibitory factor (LIF) is a growth factor with pleiotropic biological functions. It has been reported that LIF acts at different stages during mesoderm development. Also, it has been shown that LIF has a cytoprotective effect on neonatal murine cardiomyocytes (CMs) in culture, but little is known about the role of LIF during human cardiogenesis. Thus, we analyzed the effects of LIF on human pluripotent stem cells (PSC) undergoing cardiac differentiation. We first showed that LIF is expressed in the human heart during early development. We found that the addition of LIF within a precise time window during the in vitro differentiation process significantly increased CMs viability. This finding was associated to a decrease in the expression of pro-apoptotic protein Bax, which coincides with a reduction of the apoptotic rate. Therefore, the addition of LIF may represent a promising strategy for increasing CMs survival derived from PSCs.
Collapse
|
44
|
DeMartini T, Nowell M, James J, Williamson L, Lahni P, Shen H, Kaplan JM. High fat diet-induced obesity increases myocardial injury and alters cardiac STAT3 signaling in mice after polymicrobial sepsis. Biochim Biophys Acta Mol Basis Dis 2017; 1863:2654-2660. [PMID: 28625915 PMCID: PMC5653424 DOI: 10.1016/j.bbadis.2017.06.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 06/06/2017] [Accepted: 06/10/2017] [Indexed: 11/24/2022]
Abstract
Little is known about how obesity affects the heart during sepsis and we sought to investigate the obesity-induced cardiac effects that occur during polymicrobial sepsis. Six-week old C57BL/6 mice were randomized to a high fat (HFD) (60% kcal fat) or normal diet (ND) (16% kcal fat). After 6weeks of feeding, mice were anesthetized with isoflurane and polymicrobial sepsis was induced by cecal ligation and puncture (CLP). Plasma and cardiac tissue were obtained for analysis. Echocardiography was performed on a separate cohort of mice at 0 and 18h after CLP. Following 6-weeks of dietary intervention, plasma cardiac troponin I was elevated in obese mice at baseline compared to non-obese mice but troponin increased only in non-obese septic mice. IL-17a expression was 27-fold higher in obese septic mice versus non-obese septic mice. Cardiac phosphorylation of STAT3 at Ser727 was increased at baseline in obese mice and increased further only in obese septic mice. Phosphorylation of STAT3 at Tyr705 was similar in both groups at baseline and increased after sepsis. SOCS3, a downstream protein and negative regulator of STAT3, was elevated in obese mice at baseline compared to non-obese mice. After sepsis non-obese mice had an increase in SOCS3 expression that was not observed in obese mice. Taken together, we show that obesity affects cardiac function and leads to cardiac injury. Furthermore, myocardial injury in obese mice during sepsis may occur through alteration of the STAT3 pathway.
Collapse
Affiliation(s)
- Theodore DeMartini
- Division of Critical Care Medicine, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Marchele Nowell
- Division of Critical Care Medicine, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Jeanne James
- Division of Molecular Cardiovascular Biology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, 3333 Burnet Avenue, MLC 2005, Cincinnati, OH 45229, USA
| | - Lauren Williamson
- Division of Critical Care Medicine, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Patrick Lahni
- Division of Critical Care Medicine, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Hui Shen
- Division of Critical Care Medicine, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Jennifer M Kaplan
- Division of Critical Care Medicine, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| |
Collapse
|
45
|
Zhang Y, Bauersachs J, Langer HF. Immune mechanisms in heart failure. Eur J Heart Fail 2017; 19:1379-1389. [DOI: 10.1002/ejhf.942] [Citation(s) in RCA: 115] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2017] [Revised: 05/26/2017] [Accepted: 06/19/2017] [Indexed: 12/15/2022] Open
Affiliation(s)
- Yingying Zhang
- University Hospital, Department of Cardiology and Cardiovascular Medicine; Eberhard Karls University Tuebingen; Tuebingen Germany
- Section for Cardioimmunology; Eberhard Karls University Tuebingen; Tübingen Germany
- Affiliated Hospital of Qingdao University, Department of Cardiology and Cardiovascular Medicine; Qingdao University; Qingdao China
| | - Johann Bauersachs
- Department of Cardiology and Angiology; Hannover Medical School; Hannover Germany
| | - Harald F. Langer
- University Hospital, Department of Cardiology and Cardiovascular Medicine; Eberhard Karls University Tuebingen; Tuebingen Germany
- Section for Cardioimmunology; Eberhard Karls University Tuebingen; Tübingen Germany
| |
Collapse
|
46
|
Abdul-Ghani M, Suen C, Jiang B, Deng Y, Weldrick JJ, Putinski C, Brunette S, Fernando P, Lee TT, Flynn P, Leenen FHH, Burgon PG, Stewart DJ, Megeney LA. Cardiotrophin 1 stimulates beneficial myogenic and vascular remodeling of the heart. Cell Res 2017; 27:1195-1215. [PMID: 28785017 PMCID: PMC5630684 DOI: 10.1038/cr.2017.87] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2016] [Revised: 03/06/2017] [Accepted: 06/21/2017] [Indexed: 12/12/2022] Open
Abstract
The post-natal heart adapts to stress and overload through hypertrophic growth, a process that may be pathologic or beneficial (physiologic hypertrophy). Physiologic hypertrophy improves cardiac performance in both healthy and diseased individuals, yet the mechanisms that propagate this favorable adaptation remain poorly defined. We identify the cytokine cardiotrophin 1 (CT1) as a factor capable of recapitulating the key features of physiologic growth of the heart including transient and reversible hypertrophy of the myocardium, and stimulation of cardiomyocyte-derived angiogenic signals leading to increased vascularity. The capacity of CT1 to induce physiologic hypertrophy originates from a CK2-mediated restraining of caspase activation, preventing the transition to unrestrained pathologic growth. Exogenous CT1 protein delivery attenuated pathology and restored contractile function in a severe model of right heart failure, suggesting a novel treatment option for this intractable cardiac disease.
Collapse
Affiliation(s)
- Mohammad Abdul-Ghani
- Sprott Centre for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa Hospital, Ottawa, Ontario K1H 8L6, Canada.,Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Colin Suen
- Sprott Centre for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa Hospital, Ottawa, Ontario K1H 8L6, Canada.,Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Baohua Jiang
- Sprott Centre for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa Hospital, Ottawa, Ontario K1H 8L6, Canada
| | - Yupu Deng
- Sprott Centre for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa Hospital, Ottawa, Ontario K1H 8L6, Canada
| | - Jonathan J Weldrick
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada.,University of Ottawa Heart Institute, Ottawa, Ontario K1Y 4W7, Canada
| | - Charis Putinski
- Sprott Centre for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa Hospital, Ottawa, Ontario K1H 8L6, Canada.,Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Steve Brunette
- Sprott Centre for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa Hospital, Ottawa, Ontario K1H 8L6, Canada
| | - Pasan Fernando
- Sprott Centre for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa Hospital, Ottawa, Ontario K1H 8L6, Canada.,Department of Biology, Carleton University, Ottawa, Ontario K1S 5B6, Canada
| | - Tom T Lee
- Fate Therapeutics Inc., 3535 General Atomics Court Suite 200, San Diego, CA 92121, USA
| | - Peter Flynn
- Fate Therapeutics Inc., 3535 General Atomics Court Suite 200, San Diego, CA 92121, USA
| | - Frans H H Leenen
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada.,Department of Medicine (Cardiology), Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada.,University of Ottawa Heart Institute, Ottawa, Ontario K1Y 4W7, Canada
| | - Patrick G Burgon
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada.,Department of Medicine (Cardiology), Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada.,University of Ottawa Heart Institute, Ottawa, Ontario K1Y 4W7, Canada
| | - Duncan J Stewart
- Sprott Centre for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa Hospital, Ottawa, Ontario K1H 8L6, Canada.,Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada.,Department of Medicine (Cardiology), Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Lynn A Megeney
- Sprott Centre for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa Hospital, Ottawa, Ontario K1H 8L6, Canada.,Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada.,Department of Medicine (Cardiology), Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| |
Collapse
|
47
|
Hesse J, Leberling S, Boden E, Friebe D, Schmidt T, Ding Z, Dieterich P, Deussen A, Roderigo C, Rose CR, Floss DM, Scheller J, Schrader J. CD73-derived adenosine and tenascin-C control cytokine production by epicardium-derived cells formed after myocardial infarction. FASEB J 2017; 31:3040-3053. [PMID: 28363952 DOI: 10.1096/fj.201601307r] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 03/13/2017] [Indexed: 01/29/2023]
Abstract
Epicardium-derived cells (EPDCs) play a fundamental role in embryonic cardiac development and are reactivated in the adult heart in response to myocardial infarction (MI). In this study, EPDCs from post-MI rat hearts highly expressed the ectoenzyme CD73 and secreted the profibrotic matricellular protein tenascin-C (TNC). CD73 on EPDCs extensively generated adenosine from both extracellular ATP and NAD. This in turn stimulated the release of additional nucleotides from a Brefeldin A-sensitive intracellular pool via adenosine-A2BR signaling, forming a positive-feedback loop. A2BR activation, in addition, strongly promoted the release of major regulatory cytokines, such as IL-6, IL-11, and VEGF. TNC was found to stimulate EPDC migration and, together with ATP-P2X7R signaling, to activate inflammasomes in EPDCs via TLR4. Our results demonstrate that EPDCs are an important source of various proinflammatory factors in the post-MI heart controlled by purinergic and TNC signaling.-Hesse, J., Leberling, S., Boden, E., Friebe, D., Schmidt, T., Ding, Z., Dieterich, P., Deussen, A., Roderigo, C., Rose, C. R., Floss, D. M., Scheller, J., Schrader, J. CD73-derived adenosine and tenascin-C control cytokine production by epicardium-derived cells formed after myocardial infarction.
Collapse
Affiliation(s)
- Julia Hesse
- Department of Molecular Cardiology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Stella Leberling
- Department of Molecular Cardiology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Elisabeth Boden
- Department of Molecular Cardiology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Daniela Friebe
- Department of Molecular Cardiology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Timo Schmidt
- Department of Molecular Cardiology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Zhaoping Ding
- Department of Molecular Cardiology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Peter Dieterich
- Institute of Physiology, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Andreas Deussen
- Institute of Physiology, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Claudia Roderigo
- Institute of Neurobiology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Christine R Rose
- Institute of Neurobiology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Doreen M Floss
- Institute of Biochemistry and Molecular Biology II, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Jürgen Scheller
- Institute of Biochemistry and Molecular Biology II, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Jürgen Schrader
- Department of Molecular Cardiology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany;
| |
Collapse
|
48
|
Ting WJ, Huang CY, Jiang CH, Lin YM, Chung LC, Shen CY, Pai P, Lin KH, Viswanadha VP, Liao SC. Treatment with 17β-Estradiol Reduced Body Weight and the Risk of Cardiovascular Disease in a High-Fat Diet-Induced Animal Model of Obesity. Int J Mol Sci 2017; 18:ijms18030629. [PMID: 28335423 PMCID: PMC5372642 DOI: 10.3390/ijms18030629] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 02/25/2017] [Accepted: 03/08/2017] [Indexed: 01/12/2023] Open
Abstract
Estrogen receptor α (ERα) and estrogen receptor β (ERβ) play important roles in cardiovascular disease (CVD) prevention. Recently, these estrogen receptors were reconsidered as an important treatment target of obesity leading to CVD. In this study, 17β-estradiol (17β-E) replacement therapy applied to high-fat diet-induced obese C57B male mice and ovariectomized (OVX) rats were evaluated, and the protective effects against high-fat diet-induced obesity were assessed in C57B mouse hearts. The results showed that 17β-E treatment activated both ERα and ERβ, and ERβ levels increased in a dose-dependent manner in high-fat diet C57B mouse cardiomyocytes following 17β-E treatment. Notably, an almost 16% reduction in body weight was observed in the 17β-E-treated (12 μg/kg/day for 60 days) high-fat diet-induced obese C57B male mice. These results suggested that 17β-E supplements may reduce CVD risk due to obesity.
Collapse
Affiliation(s)
- Wei-Jen Ting
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, B24 Yinquan South Road, Qingyuan 511518, China.
- Graduate Institute of Basic Medical Science, China Medical University, 91 Hsueh-Shih Road, Taichung 40402, Taiwan.
| | - Chih-Yang Huang
- Graduate Institute of Basic Medical Science, China Medical University, 91 Hsueh-Shih Road, Taichung 40402, Taiwan.
- Graduate Institute of Chinese Medical Science, China Medical University, 91 Hsueh-Shih Road, Taichung 40402, Taiwan.
- Department of Health and Nutrition Biotechnology, Asia University, 500 Lioufeng Road, Taichung 41354, Taiwan.
| | - Chong-He Jiang
- Department of Urology, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, B24 Yinquan South Road, Qingyuan 511518, China.
| | - Yueh-Min Lin
- Department of Pathology, Changhua Christian Hospital, 135 Nanxiao Street, Changhua 50006, Taiwan.
- Department of Medical Technology, Jen-Teh Junior College of Medicine, Nursing and Management, 79-9 Sha-Luen Hu, Hou-Loung Town, Miaoli 35664, Taiwan.
| | - Li-Chin Chung
- Department of Hospital and Health Care Administration, China Nan University of Pharmacy & Science, 60, Section 1, Erren Road, Rende District, Tainan 71710, Taiwan.
| | - Chia-Yao Shen
- Department of Nursing, Mei Ho University, 23 Pingguang Road, Pingtung 91202, Taiwan.
| | - Peiying Pai
- Division of Cardiology, China Medical University Hospital, 91 Hsueh-Shih Road, Taichung 40402, Taiwan.
| | - Kuan-Ho Lin
- Department of Emergency Medicine, China Medical University Hospital, 91 Hsueh-Shih Road, Taichung 40402, Taiwan.
| | | | - Shih-Chieh Liao
- School of Medicine, College of Medicine, China Medical University, 91 Hsueh-Shih Road, Taichung 40402, Taiwan.
| |
Collapse
|
49
|
Karra R, Poss KD. Redirecting cardiac growth mechanisms for therapeutic regeneration. J Clin Invest 2017; 127:427-436. [PMID: 28145902 DOI: 10.1172/jci89786] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Heart failure is a major source of morbidity and mortality. Replacing lost myocardium with new tissue is a major goal of regenerative medicine. Unlike adult mammals, zebrafish and neonatal mice are capable of heart regeneration following cardiac injury. In both contexts, the regenerative program echoes molecular and cellular events that occur during cardiac development and morphogenesis, notably muscle creation through division of cardiomyocytes. Based on studies over the past decade, it is now accepted that the adult mammalian heart undergoes a low grade of cardiomyocyte turnover. Recent data suggest that this cardiomyocyte turnover can be augmented in the adult mammalian heart by redeployment of developmental factors. These findings and others suggest that stimulating endogenous regenerative responses can emerge as a therapeutic strategy for human cardiovascular disease.
Collapse
|
50
|
Al-Naamani N, Chirinos JA, Zamani P, Ruthazer R, Paulus JK, Roberts KE, Barr RG, Lima JA, Bluemke DA, Kronmal R, Kawut SM. Association of Systemic Arterial Properties With Right Ventricular Morphology: The Multi-Ethnic Study of Atherosclerosis (MESA)-Right Ventricle Study. J Am Heart Assoc 2016; 5:e004162. [PMID: 27881423 PMCID: PMC5210393 DOI: 10.1161/jaha.116.004162] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Accepted: 10/17/2016] [Indexed: 12/14/2022]
Abstract
BACKGROUND Systemic arterial stiffness is recognized as a major contributor to development of left ventricular dysfunction and failure; however, the relationship of systemic arterial properties and the right ventricle (RV) is unknown. METHODS AND RESULTS The associations between systemic arterial measures (total arterial compliance [TAC], systemic vascular resistance [SVR], and aortic augmentation index [AI]) and RV morphology (mass, end-systolic [RVESV] and end-diastolic volume [RVEDV], and ejection fraction [RVEF]) were examined using data from the Multi-Ethnic Study of Atherosclerosis. All analyses were adjusted for anthropometric, demographic, and clinical variables and the corresponding left ventricular parameter. A total of 3842 subjects without clinical cardiovascular disease were included with a mean age of 61 years, 48% male, 39% non-Hispanic white, 25% Chinese-American, 23% Hispanic, and 13% black. RV measures were within normal range for age and sex. A 1-mL/mm Hg decrease in TAC was associated with 3.9-mL smaller RVESV, 7.6-mL smaller RVEDV, and 2.4-g lower RV mass. A 5-Wood-unit increase in SVR was associated with 0.6-mL decrease in RVESV, 1.7-mL decrease in RVEDV, and 0.4-g decrease in RV mass. A 1% increase in AI was associated with 0.2-mL decrease in RVEDV. We found significant effect modification by age, sex, and race for some of these relationships, with males, whites, and younger individuals having greater decreases in RV volumes and mass. CONCLUSIONS Markers of increased systemic arterial load were associated with smaller RV volumes and lower RV mass in a population of adults without clinical cardiovascular disease.
Collapse
Affiliation(s)
- Nadine Al-Naamani
- Department of Medicine, Tufts Medical Center, Boston, MA
- Clinical and Translational Science Institute, Tufts Medical Center, Boston, MA
| | - Julio A Chirinos
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Payman Zamani
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Robin Ruthazer
- Clinical and Translational Science Institute, Tufts Medical Center, Boston, MA
| | - Jessica K Paulus
- Clinical and Translational Science Institute, Tufts Medical Center, Boston, MA
| | - Kari E Roberts
- Department of Medicine, Tufts Medical Center, Boston, MA
| | - R Graham Barr
- Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY
| | - Joao A Lima
- Department of Medicine, Johns Hopkins University, Baltimore, MD
| | - David A Bluemke
- Radiology and Imaging Sciences, National Institutes of Health/Clinical Center, Bethesda, MD
| | - Richard Kronmal
- Department of Biostatistics, University of Washington, Seattle, WA
| | - Steven M Kawut
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
- Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|