1
|
Cheng Y, Lin G, Xie Y, Xuan B, He S, Shang Z, Yan M, Lin J, Wei L, Peng J, Shen A. Baicalin ameliorates angiotensin II-induced cardiac hypertrophy and mitogen-activated protein kinase signaling pathway activation: A target-based network pharmacology approach. Eur J Pharmacol 2024; 981:176876. [PMID: 39127302 DOI: 10.1016/j.ejphar.2024.176876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 07/20/2024] [Accepted: 08/08/2024] [Indexed: 08/12/2024]
Abstract
Baicalin, a flavonoid glycoside from Scutellaria baicalensis Georgi., exerts anti-hypertensive effects. The present study aimed to assess the cardioprotective role of baicalin and explore its potential mechanisms. Network pharmacology analysis pointed out a total of 477 potential targets of baicalin were obtained from the PharmMapper and SwissTargetPrediction databases, while 11,280 targets were identified associating with hypertensive heart disease from GeneCards database. Based on the above 382 common targets, Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway analyses revealed enrichment in the regulation of cardiac hypertrophy, cardiac contraction, cardiac relaxation, as well as the mitogen-activated protein kinase (MAPK) and other signaling pathways. Moreover, baicalin treatment exhibited the amelioration of increased cardiac index and pathological alterations in angiotensin II (Ang II)-infused C57BL/6 mice. Furthermore, baicalin treatment demonstrated a reduction in cell surface area and a down-regulation of hypertrophy markers (including atrial natriuretic peptide and brain natriuretic peptide) in vivo and in vitro. In addition, baicalin treatment led to a decrease in the expression of phosphorylated c-Jun N-terminal kinase (p-JNK)/JNK, phosphorylated p38 (p-p38)/p38, and phosphorylated extracellular signal-regulated kinase (p-ERK)/ERK in the cardiac tissues of Ang II-infused mice and Ang II-stimulated H9c2 cells. These findings highlight the cardioprotective effects of baicalin, as it alleviates hypertensive cardiac injury, cardiac hypertrophy, and the activation of the MAPK pathway.
Collapse
Affiliation(s)
- Ying Cheng
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China; Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China; Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou, Fujian, 350122, China
| | - Guosheng Lin
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China; Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China; Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou, Fujian, 350122, China
| | - Yi Xie
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China; Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China; Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou, Fujian, 350122, China
| | - Bihan Xuan
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China; Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China; Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou, Fujian, 350122, China
| | - Shuyu He
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China; Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China; Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou, Fujian, 350122, China
| | - Zucheng Shang
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China; Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China; Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou, Fujian, 350122, China
| | - Mengchao Yan
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China; Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China; Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou, Fujian, 350122, China
| | - Jing Lin
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China; Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China; Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou, Fujian, 350122, China
| | - Lihui Wei
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China; Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China; Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou, Fujian, 350122, China; Innovation and Transformation Center, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China.
| | - Jun Peng
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China; Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China; Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou, Fujian, 350122, China.
| | - Aling Shen
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China; Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China; Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou, Fujian, 350122, China; Innovation and Transformation Center, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China.
| |
Collapse
|
2
|
Zhang M, Liu Q, Meng H, Duan H, Liu X, Wu J, Gao F, Wang S, Tan R, Yuan J. Ischemia-reperfusion injury: molecular mechanisms and therapeutic targets. Signal Transduct Target Ther 2024; 9:12. [PMID: 38185705 PMCID: PMC10772178 DOI: 10.1038/s41392-023-01688-x] [Citation(s) in RCA: 114] [Impact Index Per Article: 114.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 08/29/2023] [Accepted: 10/18/2023] [Indexed: 01/09/2024] Open
Abstract
Ischemia-reperfusion (I/R) injury paradoxically occurs during reperfusion following ischemia, exacerbating the initial tissue damage. The limited understanding of the intricate mechanisms underlying I/R injury hinders the development of effective therapeutic interventions. The Wnt signaling pathway exhibits extensive crosstalk with various other pathways, forming a network system of signaling pathways involved in I/R injury. This review article elucidates the underlying mechanisms involved in Wnt signaling, as well as the complex interplay between Wnt and other pathways, including Notch, phosphatidylinositol 3-kinase/protein kinase B, transforming growth factor-β, nuclear factor kappa, bone morphogenetic protein, N-methyl-D-aspartic acid receptor-Ca2+-Activin A, Hippo-Yes-associated protein, toll-like receptor 4/toll-interleukine-1 receptor domain-containing adapter-inducing interferon-β, and hepatocyte growth factor/mesenchymal-epithelial transition factor. In particular, we delve into their respective contributions to key pathological processes, including apoptosis, the inflammatory response, oxidative stress, extracellular matrix remodeling, angiogenesis, cell hypertrophy, fibrosis, ferroptosis, neurogenesis, and blood-brain barrier damage during I/R injury. Our comprehensive analysis of the mechanisms involved in Wnt signaling during I/R reveals that activation of the canonical Wnt pathway promotes organ recovery, while activation of the non-canonical Wnt pathways exacerbates injury. Moreover, we explore novel therapeutic approaches based on these mechanistic findings, incorporating evidence from animal experiments, current standards, and clinical trials. The objective of this review is to provide deeper insights into the roles of Wnt and its crosstalk signaling pathways in I/R-mediated processes and organ dysfunction, to facilitate the development of innovative therapeutic agents for I/R injury.
Collapse
Affiliation(s)
- Meng Zhang
- The Collaborative Innovation Center, Jining Medical University, Jining, Shandong, 272067, China
| | - Qian Liu
- Clinical Medical College, Jining Medical University, Jining, Shandong, 272067, China
| | - Hui Meng
- Clinical Medical College, Jining Medical University, Jining, Shandong, 272067, China
| | - Hongxia Duan
- Clinical Medical College, Jining Medical University, Jining, Shandong, 272067, China
| | - Xin Liu
- Second Clinical Medical College, Jining Medical University, Jining, Shandong, 272067, China
| | - Jian Wu
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Fei Gao
- The Collaborative Innovation Center, Jining Medical University, Jining, Shandong, 272067, China
- Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Shijun Wang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China.
| | - Rubin Tan
- Department of Physiology, Basic medical school, Xuzhou Medical University, Xuzhou, 221004, China.
| | - Jinxiang Yuan
- The Collaborative Innovation Center, Jining Medical University, Jining, Shandong, 272067, China.
| |
Collapse
|
3
|
Martin-Puig S, Menendez-Montes I. Cardiac Metabolism. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1441:365-396. [PMID: 38884721 DOI: 10.1007/978-3-031-44087-8_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Abstract
The heart is composed of a heterogeneous mixture of cellular components perfectly intermingled and able to integrate common environmental signals to ensure proper cardiac function and performance. Metabolism defines a cell context-dependent signature that plays a critical role in survival, proliferation, or differentiation, being a recognized master piece of organ biology, modulating homeostasis, disease progression, and adaptation to tissue damage. The heart is a highly demanding organ, and adult cardiomyocytes require large amount of energy to fulfill adequate contractility. However, functioning under oxidative mitochondrial metabolism is accompanied with a concomitant elevation of harmful reactive oxygen species that indeed contributes to the progression of several cardiovascular pathologies and hampers the regenerative capacity of the mammalian heart. Cardiac metabolism is dynamic along embryonic development and substantially changes as cardiomyocytes mature and differentiate within the first days after birth. During early stages of cardiogenesis, anaerobic glycolysis is the main energetic program, while a progressive switch toward oxidative phosphorylation is a hallmark of myocardium differentiation. In response to cardiac injury, different signaling pathways participate in a metabolic rewiring to reactivate embryonic bioenergetic programs or the utilization of alternative substrates, reflecting the flexibility of heart metabolism and its central role in organ adaptation to external factors. Despite the well-established metabolic pattern of fetal, neonatal, and adult cardiomyocytes, our knowledge about the bioenergetics of other cardiac populations like endothelial cells, cardiac fibroblasts, or immune cells is limited. Considering the close intercellular communication and the influence of nonautonomous cues during heart development and after cardiac damage, it will be fundamental to better understand the metabolic programs in different cardiac cells in order to develop novel interventional opportunities based on metabolic rewiring to prevent heart failure and improve the limited regenerative capacity of the mammalian heart.
Collapse
Affiliation(s)
- Silvia Martin-Puig
- Department of Metabolic and Immune Diseases, Institute for Biomedical Research "Sols-Morreale", National Spanish Research Council, CSIC, Madrid, Spain.
- Cardiac Regeneration Program, National Center for Cardiovascular Research, CNIC, Madrid, Spain.
| | - Ivan Menendez-Montes
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
4
|
Ke H, Chen Z, Zhao X, Yang C, Luo T, Ou W, Wang L, Liu H. Research progress on activation transcription factor 3: A promising cardioprotective molecule. Life Sci 2023:121869. [PMID: 37355225 DOI: 10.1016/j.lfs.2023.121869] [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: 03/31/2023] [Revised: 06/05/2023] [Accepted: 06/15/2023] [Indexed: 06/26/2023]
Abstract
Activation transcription factor 3 (ATF3), a member of the ATF/cyclic adenosine monophosphate response element binding family, can be induced by a variety of stresses. Numerous studies have indicated that ATF3 plays multiple roles in the development and progression of cardiovascular diseases, including atherosclerosis, hypertrophy, fibrosis, myocardial ischemia-reperfusion, cardiomyopathy, and other cardiac dysfunctions. In past decades, ATF3 has been demonstrated to be detrimental to some cardiac diseases. Current studies have indicated that ATF3 can function as a cardioprotective molecule in antioxidative stress, lipid metabolic metabolism, energy metabolic regulation, and cell death modulation. To unveil the potential therapeutic role of ATF3 in cardiovascular diseases, we organized this review to explore the protective effects and mechanisms of ATF3 on cardiac dysfunction, which might provide rational evidence for the prevention and cure of cardiovascular diseases.
Collapse
Affiliation(s)
- Haoteng Ke
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou 510280, China; Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Zexing Chen
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou 510280, China; Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Xuanbin Zhao
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou 510280, China; Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Chaobo Yang
- Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Tao Luo
- Department of Pathophysiology, Zhuhai Campus of Zunyi Medical University, Zhuhai, China
| | - Wen Ou
- Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Lizi Wang
- Department of Health Management, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Haiqiong Liu
- Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China; Department of Health Management, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China.
| |
Collapse
|
5
|
Bischof C, Mirtschink P, Yuan T, Wu M, Zhu C, Kaur J, Pham MD, Gonzalez-Gonoggia S, Hammer M, Rogg EM, Sharma R, Bottermann K, Gercken B, Hagag E, Berthonneche C, Sossalla S, Stehr SN, Maxeiner J, Duda MA, Latreille M, Zamboni N, Martelli F, Pedrazzini T, Dimmeler S, Krishnan J. Mitochondrial-cell cycle cross-talk drives endoreplication in heart disease. Sci Transl Med 2021; 13:eabi7964. [PMID: 34878823 DOI: 10.1126/scitranslmed.abi7964] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Corinne Bischof
- MRC Clinical Sciences Centre, Imperial College London, London W12 0NN, UK.,Institute of Cardiovascular Regeneration, Centre for Molecular Medicine, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Peter Mirtschink
- Institute of Clinical Chemistry and Laboratory Medicine, Department of Clinical Pathobiochemistry, University Hospital Dresden, Fetscherstasse 74, 01307 Dresden, Germany
| | - Ting Yuan
- Institute of Cardiovascular Regeneration, Centre for Molecular Medicine, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany.,Department of Medicine III, Division of Cardiology/Nephrology/Angiology, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Meiqian Wu
- Institute of Cardiovascular Regeneration, Centre for Molecular Medicine, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany.,Department of Medicine III, Division of Cardiology/Nephrology/Angiology, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Chaonan Zhu
- Institute of Cardiovascular Regeneration, Centre for Molecular Medicine, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany.,Department of Medicine III, Division of Cardiology/Nephrology/Angiology, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Jaskiran Kaur
- Institute of Cardiovascular Regeneration, Centre for Molecular Medicine, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany.,Department of Medicine III, Division of Cardiology/Nephrology/Angiology, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Minh Duc Pham
- Institute of Cardiovascular Regeneration, Centre for Molecular Medicine, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany.,Genome Biologics, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | | | - Marie Hammer
- Institute of Cardiovascular Regeneration, Centre for Molecular Medicine, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Eva-Maria Rogg
- Institute of Cardiovascular Regeneration, Centre for Molecular Medicine, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Rahul Sharma
- Institute of Cardiovascular Regeneration, Centre for Molecular Medicine, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Katharina Bottermann
- Institute of Cardiovascular Regeneration, Centre for Molecular Medicine, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Bettina Gercken
- Institute of Clinical Chemistry and Laboratory Medicine, Department of Clinical Pathobiochemistry, University Hospital Dresden, Fetscherstasse 74, 01307 Dresden, Germany
| | - Eman Hagag
- Institute of Clinical Chemistry and Laboratory Medicine, Department of Clinical Pathobiochemistry, University Hospital Dresden, Fetscherstasse 74, 01307 Dresden, Germany
| | - Corinne Berthonneche
- Cardiovascular Assessment Facility, University of Lausanne, CHUV, CH-1011 Lausanne, Switzerland
| | - Samuel Sossalla
- Department of Internal Medicine II, University Medical Center Regensburg, 93053 Regensburg, Germany.,Klinik für Kardiologie und Pneumologie, Georg-August-Universität Goettingen, DZHK (German Centre for Cardiovascular Research), Robert-Koch Str. 40, D-37075 Goettingen, Germany
| | - Sebastian N Stehr
- Department of Anesthesiology and Critical Care Medicine, University Hospital Leipzig, Liebigstrasse 20, D-04103 Leipzig, Germany
| | - Joachim Maxeiner
- Genome Biologics, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Maria Anna Duda
- Genome Biologics, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Mathieu Latreille
- MRC Clinical Sciences Centre, Imperial College London, London W12 0NN, UK
| | - Nicola Zamboni
- Institute of Molecular Systems Biology, ETH Zurich, Zurich 8093, Switzerland
| | - Fabio Martelli
- Molecular Cardiology Laboratory, IRCCS-Policlinico San Donato, 20097, San Donato Milanese, Milan, Italy
| | - Thierry Pedrazzini
- Department of Medicine, University of Lausanne Medical School, CHUV, MP14-220, 1011 Lausanne, Switzerland
| | - Stefanie Dimmeler
- Institute of Cardiovascular Regeneration, Centre for Molecular Medicine, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany.,DZHK Partner Site RheinMain, Mainz, Germany.,Cardio-Pulmonary Institute, Giessen, Germany
| | - Jaya Krishnan
- MRC Clinical Sciences Centre, Imperial College London, London W12 0NN, UK.,Institute of Cardiovascular Regeneration, Centre for Molecular Medicine, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany.,Department of Medicine III, Division of Cardiology/Nephrology/Angiology, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany.,Cardio-Pulmonary Institute, Giessen, Germany
| |
Collapse
|
6
|
Ip JE, Xu L, Dai J, Steegborn C, Jaffré F, Evans T, Cheung JW, Basson CT, Panaghie G, Krogh-Madsen T, Abbott GW, Lerman BB. Constitutively Activating GNAS Somatic Mutation in Right Ventricular Outflow Tract Tachycardia. Circ Arrhythm Electrophysiol 2021; 14:e010082. [PMID: 34587755 PMCID: PMC8569928 DOI: 10.1161/circep.121.010082] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- James E. Ip
- Division of Cardiology, Department of Medicine, Cornell University Medical Center, New York, NY
| | - Linna Xu
- Division of Cardiology, Department of Medicine, Cornell University Medical Center, New York, NY
| | - Jie Dai
- Division of Cardiology, Department of Medicine, Cornell University Medical Center, New York, NY
| | - Clemens Steegborn
- Department of Biochemistry, Weill-Cornell Medical College, New York, NY
- Present Address: Department of Biochemistry, University of Bayreuth, Germany
| | - Fabrice Jaffré
- Department of Surgery, Weill-Cornell Medical College, New York, NY
| | - Todd Evans
- Department of Surgery, Weill-Cornell Medical College, New York, NY
| | - Jim W. Cheung
- Division of Cardiology, Department of Medicine, Cornell University Medical Center, New York, NY
| | - Craig T. Basson
- Division of Cardiology, Department of Medicine, Cornell University Medical Center, New York, NY
- Present Address: Boston Pharmaceuticals, Cambridge, MA
| | - Gianina Panaghie
- Division of Cardiology, Department of Medicine, Cornell University Medical Center, New York, NY
| | - Trine Krogh-Madsen
- Division of Cardiology, Department of Medicine, Cornell University Medical Center, New York, NY
| | - Geoffrey W. Abbott
- Division of Cardiology, Department of Medicine, Cornell University Medical Center, New York, NY
- Present Address: Department of Physiology & Biophysics, University of California, Irvine, CA
| | - Bruce B. Lerman
- Division of Cardiology, Department of Medicine, Cornell University Medical Center, New York, NY
| |
Collapse
|
7
|
Dudek J, Kutschka I, Maack C. Metabolic and Redox Regulation of Cardiovascular Stem Cell Biology and Pathology. Antioxid Redox Signal 2021; 35:163-181. [PMID: 33121253 DOI: 10.1089/ars.2020.8201] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Significance: Cardiovascular stem cells are important for regeneration and repair of damaged tissue. Recent Advances: Pluripotent stem cells have a unique metabolism, which is adopted for their energetic and biosynthetic demand as rapidly proliferating cells. Stem cell differentiation requires an exceptional metabolic flexibility allowing for metabolic remodeling between glycolysis and oxidative phosphorylation. Critical Issues: Respiration is associated with the generation of reactive oxygen species (ROS) by the mitochondrial respiratory chain. But also the membrane-bound protein nicotinamide adenine dinucleotide phosphate oxidase (NADPH oxidase, NOX) contributes to ROS levels. ROS not only play a significant role in stem cell differentiation and tissue renewal but also cause senescence and contribute to tissue aging. Future Directions: For utilization of stem cells in therapeutic approaches, a deep understanding of the molecular mechanisms how metabolism and the cellular redox state regulate stem cell differentiation is required. Modulating the redox state of stem cells using antioxidative agents may be suitable to enhance activity of endothelial progenitor cells. Antioxid. Redox Signal. 35, 163-181.
Collapse
Affiliation(s)
- Jan Dudek
- Department of Translational Research, Comprehensive Heart Failure Center (CHFC), University Clinic Würzburg, Würzburg, Germany
| | - Ilona Kutschka
- Department of Translational Research, Comprehensive Heart Failure Center (CHFC), University Clinic Würzburg, Würzburg, Germany
| | - Christoph Maack
- Department of Translational Research, Comprehensive Heart Failure Center (CHFC), University Clinic Würzburg, Würzburg, Germany.,Department of Internal Medicine I, University Hospital Würzburg, Würzburg, Germany
| |
Collapse
|
8
|
de Oliveira Camargo R, Abual'anaz B, Rattan SG, Filomeno KL, Dixon IMC. Novel factors that activate and deactivate cardiac fibroblasts: A new perspective for treatment of cardiac fibrosis. Wound Repair Regen 2021; 29:667-677. [PMID: 34076932 DOI: 10.1111/wrr.12947] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 04/06/2021] [Accepted: 04/13/2021] [Indexed: 12/12/2022]
Abstract
Heart disease with attendant cardiac fibrosis kills more patients in developed countries than any other disease, including cancer. We highlight the recent literature on factors that activate and also deactivate cardiac fibroblasts. Activation of cardiac fibroblasts results in myofibroblasts phenotype which incorporates aSMA to stress fibres, express ED-A fibronectin, elevated PDGFRα and are hypersecretory ECM components. These cells facilitate both acute wound healing (infarct site) and chronic cardiac fibrosis. Quiescent fibroblasts are associated with normal myocardial tissue and provide relatively slow turnover of the ECM. Deactivation of activated myofibroblasts is a much less studied phenomenon. In this context, SKI is a known negative regulator of TGFb1 /Smad signalling, and thus may share functional similarity to PPARγ activation. The discovery of SKI's potent anti-fibrotic role, and its ability to deactivate and/or myofibroblasts is featured and contrasted with PPARγ. While myofibroblasts are typically recruited from pools of potential precursor cells in a variety of organs, the importance of activation of resident cardiac fibroblasts has been recently emphasised. Myofibroblasts deposit ECM components at an elevated rate and contribute to both systolic and diastolic dysfunction with attendant cardiac fibrosis. A major knowledge gap exists as to specific proteins that may signal for fibroblast deactivation. As SKI may be a functionally pluripotent protein, we suggest that it serves as a scaffold to proteins other than R-Smads and associated Smad signal proteins, and thus its anti-fibrotic effects may extend beyond binding R-Smads. While cardiac fibrosis is causal to heart failure, the treatment of cardiac fibrosis is hampered by the lack of availability of effective pharmacological anti-fibrotic agents. The current review will provide an overview of work highlighting novel factors which cause fibroblast activation and deactivation to underscore putative therapeutic avenues for improving disease outcomes in cardiac patients with fibrosed hearts.
Collapse
Affiliation(s)
- Rebeca de Oliveira Camargo
- Institute of Cardiovascular Sciences, Albrechtsen Research Centre, Winnipeg, Canada.,Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Canada.,Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, Canada
| | - Besher Abual'anaz
- Institute of Cardiovascular Sciences, Albrechtsen Research Centre, Winnipeg, Canada.,Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Canada.,Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, Canada
| | - Sunil G Rattan
- Institute of Cardiovascular Sciences, Albrechtsen Research Centre, Winnipeg, Canada.,Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Canada.,Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, Canada
| | - Krista L Filomeno
- Institute of Cardiovascular Sciences, Albrechtsen Research Centre, Winnipeg, Canada
| | - Ian M C Dixon
- Institute of Cardiovascular Sciences, Albrechtsen Research Centre, Winnipeg, Canada.,Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Canada.,Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, Canada
| |
Collapse
|
9
|
da Rocha AL, Rovina RL, Pinto AP, Marafon BB, da Silva LECM, Simabuco FM, Frantz FG, Pauli JR, de Moura LP, Cintra DE, Ropelle ER, Filho HT, de Freitas EC, Rivas DA, da Silva ASR. Interleukin-6 ablation does not alter morphofunctional heart characteristics but modulates physiological and inflammatory markers after strenuous exercise. Cytokine 2021; 142:155494. [PMID: 33765652 DOI: 10.1016/j.cyto.2021.155494] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 02/19/2021] [Accepted: 03/04/2021] [Indexed: 12/17/2022]
Abstract
Interleukin-6 (IL-6) is associated with pathological cardiac hypertrophy and can be dramatically increased in serum after an acute strenuous exercise session. However, IL-6 is also associated with the increased production and release of anti-inflammatory cytokines and the inhibition of tumor necrosis factor-alpha (TNF-α) after chronic moderate exercise. To elucidate the relevance of IL-6 in inflammatory and hypertrophic signaling in the heart in response to an acute strenuous exercise session, we combined transcriptome analysis using the BXD mice database and exercised IL-6 knockout mice (IL-6KO). Bioinformatic analysis demonstrated that low or high-levels of Il6 mRNA in the heart did not change the inflammation- and hypertrophy-related genes in BXD mice strains. On the other hand, bioinformatic analysis revealed a strong positive correlation between Il6 gene expression in skeletal muscle with inflammation-related genes in cardiac tissue in several BXD mouse strains, suggesting that skeletal muscle-derived IL-6 could alter the heart's intracellular signals, particularly the inflammatory signaling. As expected, an acute strenuous exercise session increased IL-6 levels in wild-type, but not in IL-6KO mice. Despite not showing morphofunctional differences in the heart at rest, the IL-6KO group presented a reduction in physical performance and attenuated IL-6, TNF-α, and IL-1beta kinetics in serum, as well as lower p38MAPK phosphorylation, Ampkalpha expression, and higher Acta1 and Tnf gene expressions in the left ventricle in the basal condition. In response to strenuous exercise, IL-6 ablation was linked to a reduction in the pro-inflammatory response and higher activation of classical physiological cardiac hypertrophy proteins.
Collapse
Affiliation(s)
- Alisson L da Rocha
- Postgraduate Program in Rehabilitation and Functional Performance, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, São Paulo, Brazil.
| | - Rafael L Rovina
- School of Physical Education and Sport of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, São Paulo, Brazil
| | - Ana P Pinto
- Postgraduate Program in Rehabilitation and Functional Performance, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, São Paulo, Brazil
| | - Bruno B Marafon
- School of Physical Education and Sport of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, São Paulo, Brazil
| | - Lilian E C M da Silva
- Department of Ophthalmology, Otorhinolaryngology, and Head and Neck Surgery School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Fernando M Simabuco
- Laboratory of Molecular Biology of Exercise (LaBMEx), School of Applied Sciences, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Fabiani G Frantz
- Faculty of Pharmaceutical Sciences of Ribeirão Preto, Department of Clinical, Toxicological, and Bromatological Analysis, University of São Paulo (USP), Ribeirão Preto, São Paulo, Brazil
| | - José R Pauli
- Laboratory of Molecular Biology of Exercise (LaBMEx), School of Applied Sciences, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Leandro P de Moura
- Laboratory of Molecular Biology of Exercise (LaBMEx), School of Applied Sciences, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Dennys E Cintra
- Laboratory of Molecular Biology of Exercise (LaBMEx), School of Applied Sciences, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Eduardo R Ropelle
- Laboratory of Molecular Biology of Exercise (LaBMEx), School of Applied Sciences, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Hugo T Filho
- School of Physical Education and Sport of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, São Paulo, Brazil
| | - Ellen C de Freitas
- School of Physical Education and Sport of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, São Paulo, Brazil
| | - Donato A Rivas
- Nutrition, Exercise Physiology and Sarcopenia Laboratory, United States, Tufts University, Boston, Massachusetts 02111, USA
| | - Adelino S R da Silva
- Postgraduate Program in Rehabilitation and Functional Performance, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, São Paulo, Brazil; School of Physical Education and Sport of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, São Paulo, Brazil
| |
Collapse
|
10
|
Salah SM, Meisenheimer JD, Rao R, Peda JD, Wallace DP, Foster D, Li X, Li X, Zhou X, Vallejo JA, Wacker MJ, Fields TA, Swenson-Fields KI. MCP-1 promotes detrimental cardiac physiology, pulmonary edema, and death in the cpk model of polycystic kidney disease. Am J Physiol Renal Physiol 2019; 317:F343-F360. [PMID: 31091126 PMCID: PMC6732452 DOI: 10.1152/ajprenal.00240.2018] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 05/13/2019] [Accepted: 05/14/2019] [Indexed: 01/05/2023] Open
Abstract
Polycystic kidney disease (PKD) is characterized by slowly expanding renal cysts that damage the kidney, typically resulting in renal failure by the fifth decade. The most common cause of death in these patients, however, is cardiovascular disease. Expanding cysts in PKD induce chronic kidney injury that is accompanied by immune cell infiltration, including macrophages, which we and others have shown can promote disease progression in PKD mouse models. Here, we show that monocyte chemoattractant protein-1 [MCP-1/chemokine (C-C motif) ligand 2 (CCL2)] is responsible for the majority of monocyte chemoattractant activity produced by renal PKD cells from both mice and humans. To test whether the absence of MCP-1 lowers renal macrophage concentration and slows disease progression, we generated genetic knockout (KO) of MCP-1 in a mouse model of PKD [congenital polycystic kidney (cpk) mice]. Cpk mice are born with rapidly expanding renal cysts, accompanied by a decline in kidney function and death by postnatal day 21. Here, we report that KO of MCP-1 in these mice increased survival, with some mice living past 3 mo. Surprisingly, however, there was no significant difference in renal macrophage concentration, nor was there improvement in cystic disease or kidney function. Examination of mice revealed cardiac hypertrophy in cpk mice, and measurement of cardiac electrical activity via ECG revealed repolarization abnormalities. MCP-1 KO did not affect the number of cardiac macrophages, nor did it alleviate the cardiac aberrancies. However, MCP-1 KO did prevent the development of pulmonary edema, which occurred in cpk mice, and promoted decreased resting heart rate and increased heart rate variability in both cpk and noncystic mice. These data suggest that in this mouse model of PKD, MCP-1 altered cardiac/pulmonary function and promoted death outside of its role as a macrophage chemoattractant.
Collapse
Affiliation(s)
- Sally M Salah
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, Kansas
- Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas
| | - James D Meisenheimer
- Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas
| | - Reena Rao
- Department of Internal Medicine-Division of Nephrology and Hypertension, University of Kansas Medical Center, Kansas City, Kansas
- Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas
| | - Jacqueline D Peda
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas
- Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas
| | - Darren P Wallace
- Department of Internal Medicine-Division of Nephrology and Hypertension, University of Kansas Medical Center, Kansas City, Kansas
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas
- Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas
| | - Dawson Foster
- Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas
| | - Xiaogang Li
- Department of Internal Medicine-Division of Nephrology and Hypertension, University of Kansas Medical Center, Kansas City, Kansas
- Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, Kansas
| | - Xiaoyan Li
- Department of Internal Medicine-Division of Nephrology and Hypertension, University of Kansas Medical Center, Kansas City, Kansas
- Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas
| | - Xia Zhou
- Department of Internal Medicine-Division of Nephrology and Hypertension, University of Kansas Medical Center, Kansas City, Kansas
- Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas
| | - Julian A Vallejo
- Department of Biomedical Sciences, School of Medicine, University of Missouri-Kansas City, Kansas City, Missouri
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri-Kansas City, Kansas City, Missouri
| | - Michael J Wacker
- Department of Biomedical Sciences, School of Medicine, University of Missouri-Kansas City, Kansas City, Missouri
| | - Timothy A Fields
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas
- Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas
| | - Katherine I Swenson-Fields
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, Kansas
- Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas
| |
Collapse
|
11
|
ERK: A Key Player in the Pathophysiology of Cardiac Hypertrophy. Int J Mol Sci 2019; 20:ijms20092164. [PMID: 31052420 PMCID: PMC6539093 DOI: 10.3390/ijms20092164] [Citation(s) in RCA: 186] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 04/26/2019] [Accepted: 04/29/2019] [Indexed: 12/17/2022] Open
Abstract
Cardiac hypertrophy is an adaptive and compensatory mechanism preserving cardiac output during detrimental stimuli. Nevertheless, long-term stimuli incite chronic hypertrophy and may lead to heart failure. In this review, we analyze the recent literature regarding the role of ERK (extracellular signal-regulated kinase) activity in cardiac hypertrophy. ERK signaling produces beneficial effects during the early phase of chronic pressure overload in response to G protein-coupled receptors (GPCRs) and integrin stimulation. These functions comprise (i) adaptive concentric hypertrophy and (ii) cell death prevention. On the other hand, ERK participates in maladaptive hypertrophy during hypertension and chemotherapy-mediated cardiac side effects. Specific ERK-associated scaffold proteins are implicated in either cardioprotective or detrimental hypertrophic functions. Interestingly, ERK phosphorylated at threonine 188 and activated ERK5 (the big MAPK 1) are associated with pathological forms of hypertrophy. Finally, we examine the connection between ERK activation and hypertrophy in (i) transgenic mice overexpressing constitutively activated RTKs (receptor tyrosine kinases), (ii) animal models with mutated sarcomeric proteins characteristic of inherited hypertrophic cardiomyopathies (HCMs), and (iii) mice reproducing syndromic genetic RASopathies. Overall, the scientific literature suggests that during cardiac hypertrophy, ERK could be a “good” player to be stimulated or a “bad” actor to be mitigated, depending on the pathophysiological context.
Collapse
|
12
|
Sousa Fialho MDL, Abd Jamil AH, Stannard GA, Heather LC. Hypoxia-inducible factor 1 signalling, metabolism and its therapeutic potential in cardiovascular disease. Biochim Biophys Acta Mol Basis Dis 2019; 1865:831-843. [DOI: 10.1016/j.bbadis.2018.09.024] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 08/24/2018] [Accepted: 09/18/2018] [Indexed: 12/20/2022]
|
13
|
Nomura S, Satoh M, Fujita T, Higo T, Sumida T, Ko T, Yamaguchi T, Tobita T, Naito AT, Ito M, Fujita K, Harada M, Toko H, Kobayashi Y, Ito K, Takimoto E, Akazawa H, Morita H, Aburatani H, Komuro I. Cardiomyocyte gene programs encoding morphological and functional signatures in cardiac hypertrophy and failure. Nat Commun 2018; 9:4435. [PMID: 30375404 PMCID: PMC6207673 DOI: 10.1038/s41467-018-06639-7] [Citation(s) in RCA: 176] [Impact Index Per Article: 25.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2017] [Accepted: 09/18/2018] [Indexed: 11/09/2022] Open
Abstract
Pressure overload induces a transition from cardiac hypertrophy to heart failure, but its underlying mechanisms remain elusive. Here we reconstruct a trajectory of cardiomyocyte remodeling and clarify distinct cardiomyocyte gene programs encoding morphological and functional signatures in cardiac hypertrophy and failure, by integrating single-cardiomyocyte transcriptome with cell morphology, epigenomic state and heart function. During early hypertrophy, cardiomyocytes activate mitochondrial translation/metabolism genes, whose expression is correlated with cell size and linked to ERK1/2 and NRF1/2 transcriptional networks. Persistent overload leads to a bifurcation into adaptive and failing cardiomyocytes, and p53 signaling is specifically activated in late hypertrophy. Cardiomyocyte-specific p53 deletion shows that cardiomyocyte remodeling is initiated by p53-independent mitochondrial activation and morphological hypertrophy, followed by p53-dependent mitochondrial inhibition, morphological elongation, and heart failure gene program activation. Human single-cardiomyocyte analysis validates the conservation of the pathogenic transcriptional signatures. Collectively, cardiomyocyte identity is encoded in transcriptional programs that orchestrate morphological and functional phenotypes.
Collapse
Affiliation(s)
- Seitaro Nomura
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-8655, Japan
- Genome Science Division, Research Center for Advanced Science and Technologies, The University of Tokyo, Tokyo, 153-0041, Japan
| | - Masahiro Satoh
- Genome Science Division, Research Center for Advanced Science and Technologies, The University of Tokyo, Tokyo, 153-0041, Japan
- Department of Cardiovascular Medicine, Chiba University Graduate School of Medicine, Chiba, 260-8670, Japan
| | - Takanori Fujita
- Genome Science Division, Research Center for Advanced Science and Technologies, The University of Tokyo, Tokyo, 153-0041, Japan
| | - Tomoaki Higo
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Osaka, 565-0871, Japan
| | - Tomokazu Sumida
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-8655, Japan
| | - Toshiyuki Ko
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-8655, Japan
| | - Toshihiro Yamaguchi
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-8655, Japan
| | - Takashige Tobita
- Department of Cardiology, Tokyo Women's Medical University, Tokyo, 162-8666, Japan
| | - Atsuhiko T Naito
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-8655, Japan
| | - Masamichi Ito
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-8655, Japan
| | - Kanna Fujita
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-8655, Japan
| | - Mutsuo Harada
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-8655, Japan
| | - Haruhiro Toko
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-8655, Japan
| | - Yoshio Kobayashi
- Department of Cardiovascular Medicine, Chiba University Graduate School of Medicine, Chiba, 260-8670, Japan
| | - Kaoru Ito
- Laboratory for Cardiovascular Diseases, RIKEN Center for Integrative Medical Sciences, Kanagawa, 230-0045, Japan
| | - Eiki Takimoto
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-8655, Japan
| | - Hiroshi Akazawa
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-8655, Japan
| | - Hiroyuki Morita
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-8655, Japan
| | - Hiroyuki Aburatani
- Genome Science Division, Research Center for Advanced Science and Technologies, The University of Tokyo, Tokyo, 153-0041, Japan.
| | - Issei Komuro
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-8655, Japan.
| |
Collapse
|
14
|
Gao J, Zhu M, Liu RF, Zhang JS, Xu M. Cardiac Hypertrophy is Positively Regulated by MicroRNA‑24 in Rats. Chin Med J (Engl) 2018; 131:1333-1341. [PMID: 29786048 PMCID: PMC5987506 DOI: 10.4103/0366-6999.232793] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Background: MicroRNA-24 (miR-24) plays an important role in heart failure by reducing the efficiency of myocardial excitation-contraction coupling. Prolonged cardiac hypertrophy may lead to heart failure, but little is known about the role of miR-24 in cardiac hypertrophy. This study aimed to preliminarily investigate the function of miR-24 and its mechanisms in cardiac hypertrophy. Methods: Twelve Sprague-Dawley rats with a body weight of 50 ± 5 g were recruited and randomly divided into two groups: a transverse aortic constriction (TAC) group and a sham surgery group. Hypertrophy index was measured and calculated by echocardiography and hematoxylin and eosin staining. TargetScans algorithm-based prediction was used to search for the targets of miR-24, which was subsequently confirmed by a real-time polymerase chain reaction and luciferase assay. Immunofluorescence labeling was used to measure the cell surface area, and 3H-leucine incorporation was used to detect the synthesis of total protein in neonatal rat cardiac myocytes (NRCMs) with the overexpression of miR-24. In addition, flow cytometry was performed to observe the alteration in the cell cycle. Statistical analysis was carried out with GraphPad Prism v5.0 and SPSS 19.0. A two-sided P < 0.05 was considered as the threshold for significance. Results: The expression of miR-24 was abnormally increased in TAC rat cardiac tissue (t = −2.938, P < 0.05). TargetScans algorithm-based prediction demonstrated that CDKN1B (p27, Kip1), a cell cycle regulator, was a putative target of miR-24, and was confirmed by luciferase assay. The expression of p27 was decreased in TAC rat cardiac tissue (t = 2.896, P < 0.05). The overexpression of miR-24 in NRCMs led to the decreased expression of p27 (t = 4.400, P < 0.01), and decreased G0/G1 arrest in cell cycle and cardiomyocyte hypertrophy. Conclusion: MiR-24 promotes cardiac hypertrophy partly by affecting the cell cycle through down-regulation of p27 expression.
Collapse
Affiliation(s)
- Juan Gao
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital; Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education; Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing 100191, China
| | - Min Zhu
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital; Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education; Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing 100191, China
| | - Rui-Feng Liu
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital; Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education; Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing 100191, China
| | - Jian-Shu Zhang
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital; Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education; Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing 100191, China
| | - Ming Xu
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital; Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education; Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing 100191, China
| |
Collapse
|
15
|
Park D, Lee HS, Kang JH, Kim SM, Gong JR, Cho KH. Attractor landscape analysis of the cardiac signaling network reveals mechanism-based therapeutic strategies for heart failure. J Mol Cell Biol 2018; 10:180-194. [PMID: 29579284 DOI: 10.1093/jmcb/mjy019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Accepted: 03/19/2018] [Indexed: 01/03/2025] Open
Abstract
Apoptosis and hypertrophy of cardiomyocytes are the primary causes of heart failure (HF), a global leading cause of death, and are regulated through the complicated intracellular signaling network, limiting the development of effective treatments due to its complexity. To identify effective therapeutic strategies for HF at a system level, we develop a large-scale comprehensive mathematical model of the cardiac signaling network by integrating all available experimental evidence. Attractor landscape analysis of the network model identifies distinct sets of control nodes that effectively suppress apoptosis and hypertrophy of cardiomyocytes under ischemic or pressure overload-induced HF, the two major types of HF. Intriguingly, our system-level analysis suggests that intervention of these control nodes may increase the efficacy of clinical drugs for HF and, of most importance, different combinations of control nodes are suggested as potentially effective candidate drug targets depending on the types of HF. Our study provides a systematic way of developing mechanism-based therapeutic strategies for HF.
Collapse
Affiliation(s)
- Daebeom Park
- Laboratory for Systems Biology and Bio-inspired Engineering, Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Ho-Sung Lee
- Laboratory for Systems Biology and Bio-inspired Engineering, Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
- Graduate School of Medical Science and Engineering, KAIST, Daejeon, Republic of Korea
| | - Jun Hyuk Kang
- Laboratory for Systems Biology and Bio-inspired Engineering, Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
- Graduate School of Medical Science and Engineering, KAIST, Daejeon, Republic of Korea
| | - Seon-Myeong Kim
- Laboratory for Systems Biology and Bio-inspired Engineering, Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Jeong-Ryeol Gong
- Laboratory for Systems Biology and Bio-inspired Engineering, Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Kwang-Hyun Cho
- Laboratory for Systems Biology and Bio-inspired Engineering, Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
- Graduate School of Medical Science and Engineering, KAIST, Daejeon, Republic of Korea
| |
Collapse
|
16
|
Abstract
Cardiovascular disease (CVD) accounts for more deaths globally than any other single disease. There are on average 1.5 million episodes of myocardial infarction (heart attack) each year in the United States alone with roughly one-third resulting in death. There is therefore a major need for developing new and effective strategies to promote cardiac repair. Intramyocardial transplantation of mesenchymal stem cells (MSCs) has emerged as a leading contender in the pursuit of clinical intervention and therapy. MSCs are potent mediators of cardiac repair and are therefore an attractive tool in the development of preclinical and clinical trials. MSCs are capable of secreting a large array of soluble factors, which have had demonstrated effects on pathogenic cardiac remolding, fibrosis, immune activation, and cardiac stem cell proliferation within the damaged heart. MSCs are also capable of differentiation into cardiomyocytes, endothelial cells, and vascular smooth muscle cells, although the relative contribution of trilineage differentiation and paracrine effectors on cardiac repair remains the subject of active investigation.
Collapse
|
17
|
Torán JL, Aguilar S, López JA, Torroja C, Quintana JA, Santiago C, Abad JL, Gomes-Alves P, Gonzalez A, Bernal JA, Jiménez-Borreguero LJ, Alves PM, R-Borlado L, Vázquez J, Bernad A. CXCL6 is an important paracrine factor in the pro-angiogenic human cardiac progenitor-like cell secretome. Sci Rep 2017; 7:12490. [PMID: 28970523 PMCID: PMC5624898 DOI: 10.1038/s41598-017-11976-6] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Accepted: 08/29/2017] [Indexed: 12/22/2022] Open
Abstract
Studies in recent years have established that the principal effects in cardiac cell therapy are associated with paracrine/autocrine factors. We combined several complementary techniques to define human cardiac progenitor cell (CPC) secretome constituted by 914 proteins/genes; 51% of these are associated with the exosomal compartment. To define the set of proteins specifically or highly differentially secreted by CPC, we compared human mesenchymal stem cells and dermal fibroblasts; the study defined a group of growth factors, cytokines and chemokines expressed at high to medium levels by CPC. Among them, IL-1, GROa (CXCL1), CXCL6 (GCP2) and IL-8 are examples whose expression was confirmed by most techniques used. ELISA showed that CXCL6 is significantly overexpressed in CPC conditioned medium (CM) (18- to 26-fold) and western blot confirmed expression of its receptors CXCR1 and CXCR2. Addition of anti-CXCL6 completely abolished migration in CPC-CM compared with anti-CXCR2, which promoted partial inhibition, and anti-CXCR1, which was inefficient. Anti-CXCL6 also significantly inhibited CPC CM angiogenic activity. In vivo evaluation also supported a relevant role for angiogenesis. Altogether, these results suggest a notable angiogenic potential in CPC-CM and identify CXCL6 as an important paracrine factor for CPC that signals mainly through CXCR2.
Collapse
MESH Headings
- Animals
- Antibodies, Neutralizing/pharmacology
- Cell Movement
- Chemokine CXCL1/genetics
- Chemokine CXCL1/metabolism
- Chemokine CXCL6/antagonists & inhibitors
- Chemokine CXCL6/genetics
- Chemokine CXCL6/metabolism
- Culture Media, Conditioned/chemistry
- Culture Media, Conditioned/metabolism
- Fibroblasts/cytology
- Fibroblasts/drug effects
- Fibroblasts/metabolism
- Gene Expression Regulation
- Human Umbilical Vein Endothelial Cells/cytology
- Human Umbilical Vein Endothelial Cells/drug effects
- Human Umbilical Vein Endothelial Cells/metabolism
- Humans
- Interleukin-1/genetics
- Interleukin-1/metabolism
- Interleukin-8/genetics
- Interleukin-8/metabolism
- Male
- Mesenchymal Stem Cells/cytology
- Mesenchymal Stem Cells/drug effects
- Mesenchymal Stem Cells/metabolism
- Mice
- Mice, Inbred C57BL
- Myocardium/cytology
- Myocardium/metabolism
- Neovascularization, Physiologic/genetics
- Paracrine Communication/genetics
- Proteome/genetics
- Proteome/metabolism
- Receptors, Interleukin-8A/antagonists & inhibitors
- Receptors, Interleukin-8A/genetics
- Receptors, Interleukin-8A/metabolism
- Receptors, Interleukin-8B/antagonists & inhibitors
- Receptors, Interleukin-8B/genetics
- Receptors, Interleukin-8B/metabolism
- Signal Transduction
- Stem Cells/cytology
- Stem Cells/drug effects
- Stem Cells/metabolism
Collapse
Affiliation(s)
- José Luis Torán
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB-CSIC), Campus Universidad Autónoma de Madrid, 28049, Madrid, Spain
- Cardiovascular Development and Repair Department, Spanish National Cardiovascular Research Center (CNIC), Melchor Fernández Almagro 3, 28029, Madrid, Spain
| | - Susana Aguilar
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB-CSIC), Campus Universidad Autónoma de Madrid, 28049, Madrid, Spain
- Cardiovascular Development and Repair Department, Spanish National Cardiovascular Research Center (CNIC), Melchor Fernández Almagro 3, 28029, Madrid, Spain
| | - Juan Antonio López
- Cardiovascular Proteomics Laboratory, Spanish National Cardiovascular Research Center (CNIC), Melchor Fernaández Almagro 3, 28029, Madrid, Spain
| | - Carlos Torroja
- Bioinformatics Unit, Spanish National Cardiovascular Research Center (CNIC), Melchor Fernández Almagro 3, 28029, Madrid, Spain
| | - Juan Antonio Quintana
- Cardiovascular Development and Repair Department, Spanish National Cardiovascular Research Center (CNIC), Melchor Fernández Almagro 3, 28029, Madrid, Spain
- Cell and Developmental Biology, Spanish National Cardiovascular Research Center (CNIC), Melchor Fernández Almagro 3, 28029, Madrid, Spain
| | - Cesar Santiago
- Department of Macromolecular Structures, Centro Nacional de Biotecnología (CNB-CSIC), Campus Universidad Autónoma de Madrid, 28049, Madrid, Spain
| | - José Luis Abad
- Coretherapix SLU, Santiago Grisolia 2, 28769, Tres Cantos, Madrid, Spain
| | - Patricia Gomes-Alves
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157, Oeiras, Portugal
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901, Oeiras, Portugal
| | - Andrés Gonzalez
- Myocardial pathophysiology, Spanish National Cardiovascular Research Center (CNIC), Melchor Fernández Almagro 3, 28029, Madrid, Spain
| | - Juan Antonio Bernal
- Myocardial pathophysiology, Spanish National Cardiovascular Research Center (CNIC), Melchor Fernández Almagro 3, 28029, Madrid, Spain
| | - Luis Jesús Jiménez-Borreguero
- Cell and Developmental Biology, Spanish National Cardiovascular Research Center (CNIC), Melchor Fernández Almagro 3, 28029, Madrid, Spain
- Hospital de la Princesa, Diego de León 62, 28006, Madrid, Spain
| | - Paula Marques Alves
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157, Oeiras, Portugal
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901, Oeiras, Portugal
| | - Luis R-Borlado
- Coretherapix SLU, Santiago Grisolia 2, 28769, Tres Cantos, Madrid, Spain
| | - Jesús Vázquez
- Cardiovascular Proteomics Laboratory, Spanish National Cardiovascular Research Center (CNIC), Melchor Fernaández Almagro 3, 28029, Madrid, Spain
| | - Antonio Bernad
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB-CSIC), Campus Universidad Autónoma de Madrid, 28049, Madrid, Spain.
- Cardiovascular Development and Repair Department, Spanish National Cardiovascular Research Center (CNIC), Melchor Fernández Almagro 3, 28029, Madrid, Spain.
| |
Collapse
|
18
|
Li S, Li H, Yuan Z, Zhang B, Chen A, Zhou M, Liu J, Cai J, Wang Z, Ye X, Zhao Q. Off-pump coronary artery bypass surgery outcomes in patients with ischaemic left ventricular systolic dysfunction with or without detected viable myocardium. Interact Cardiovasc Thorac Surg 2017; 25:218-224. [PMID: 28475697 DOI: 10.1093/icvts/ivx064] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Accepted: 02/07/2017] [Indexed: 11/14/2022] Open
Abstract
OBJECTIVES The prognostic value of myocardial viability before coronary bypass grafting remains controversial. The present study evaluated the effects of off-pump coronary artery bypass (OPCAB) grafting on patients with coronary artery disease (CAD) with or without viable myocardium (VM) preoperatively detected via nuclear imaging. METHODS A total of 115 consecutive patients with 3-vessel disease and impaired left ventricular ejection fraction (LVEF ≤ 45%) who underwent OPCAB grafting were recruited in this prospective study. The patients were divided into 2 groups based on myocardial viability, the non-viable myocardium (NVM, 55 patients) and VM (60 patients) groups. Positron emission tomography and radionuclide imaging examination were applied to evaluate the myocardium viability. A Kaplan-Meier analysis was conducted to evaluate the 1-year survival rate. RESULTS The preoperative data were similar between groups. An improvement in the LVEF was observed in both groups 12 months after OPCAB grafting (P < 0.05). A binary logistic regression revealed that NVM was an independent predictor of a 5% improvement in LVEF at 6 months (P = 0.012). The rate of main adverse cardiovascular and cerebrovascular events (MACCEs) rate at 1 year was similar between the 2 groups (P = 0.06). At 1 year, the death rates were 14.5% in the NVM group and 5% in the VM group (P = 0.17). A Cox regression analysis revealed that NVM and age were independent predictors of mortality [the hazard ratio for death associated with NVM and age were 1.62, 95% confidence interval (CI) = 1.16-2.89, P = 0.036 and 1.05, 95% CI = 0.98-1.12, P =0.025, respectively]. CONCLUSIONS The MACCEs and mortality rates of the NVM group were higher than those of the VM group. However, OPCAB surgery improved LVEF, regardless of myocardium status. Therefore, the assessment of myocardial viability might not be the sole deciding factor in decision-making process regarding OPCAB surgery.
Collapse
Affiliation(s)
- Sen Li
- Department of Cardiac Surgery, Ruijin Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, China
| | - Haiqing Li
- Department of Cardiac Surgery, Ruijin Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, China
| | - Zhize Yuan
- Department of Cardiac Surgery, Ruijin Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, China
| | - Baoli Zhang
- Department of Hypertension, Ruijin Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, China
| | - Anqing Chen
- Department of Cardiac Surgery, Ruijin Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, China
| | - Mi Zhou
- Department of Cardiac Surgery, Ruijin Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, China
| | - Jun Liu
- Department of Cardiac Surgery, Ruijin Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, China
| | - Junfeng Cai
- Department of Cardiac Surgery, Ruijin Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, China
| | - Zhe Wang
- Department of Cardiac Surgery, Ruijin Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, China
| | - Xiaofeng Ye
- Department of Cardiac Surgery, Ruijin Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, China
| | - Qiang Zhao
- Department of Cardiac Surgery, Ruijin Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, China
| |
Collapse
|
19
|
Ceccarelli G, Benedetti L, Arcari ML, Carubbi C, Galli D. Muscle Stem Cell and Physical Activity: What Point is the Debate at? Open Med (Wars) 2017; 12:144-156. [PMID: 28765836 PMCID: PMC5529938 DOI: 10.1515/med-2017-0022] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Accepted: 04/21/2017] [Indexed: 12/11/2022] Open
Abstract
In the last 15 years, it emerged that the practice of regular physical activity reduces the risks of many diseases (cardiovascular diseases, diabetes, etc.) and it is fundamental in weight control and energy consuming to contrast obesity. Different groups proposed many molecular mechanisms as responsible for the positive effects of physical activity in healthy life. However, many points remain to be clarified. In this mini-review we reported the latest observations on the effects of physical exercise on healthy skeletal and cardiac muscle focusing on muscle stem cells. The last ones represent the fundamental elements for muscle regeneration post injury, but also for healthy muscle homeostasis. Interestingly, in both muscle tissues the morphological consequence of physical activity is a physiological hypertrophy that depends on different phenomena both in differentiated cells and stem cells. The signaling pathways for physical exercise effects present common elements in skeletal and cardiac muscle, like activation of specific transcription factors, proliferative pathways, and cytokines. More recently, post translational (miRNAs) or epigenetic (DNA methylation) modifications have been demonstrated. However, several points remain unresolved thus requiring new research on the effect of exercise on muscle stem cells.
Collapse
Affiliation(s)
- Gabriele Ceccarelli
- Department of Public Health, Experimental Medicine and Forensic, University of Pavia, Pavia, Italy.,Center of Health Technologies (CHT), University of Pavia, Pavia, Italy
| | - Laura Benedetti
- Department of Public Health, Experimental Medicine and Forensic, University of Pavia, Pavia, Italy.,Center of Health Technologies (CHT), University of Pavia, Pavia, Italy
| | - Maria Luisa Arcari
- Department of Medicine and Surgery, S.Bi.Bi.T. Unit, University of Parma, Parma, Italy
| | - Cecilia Carubbi
- Department of Medicine and Surgery, S.Bi.Bi.T. Unit, University of Parma, Parma, Italy
| | - Daniela Galli
- Department of Medicine and Surgery, S.Bi.Bi.T. Unit and Sport and Exercise Medicine Center (SEM)., University of Parma c/o Ospedale Maggiore, Via Gramsci, 14, 43126, Tel: +39-0521-036306, , Parma, Italy.,Department of Medicine and Surgery, S.Bi.Bi.T. Unit, University of Parma, Parma, Italy
| |
Collapse
|
20
|
Troupes CD, Wallner M, Borghetti G, Zhang C, Mohsin S, von Lewinski D, Berretta RM, Kubo H, Chen X, Soboloff J, Houser S. Role of STIM1 (Stromal Interaction Molecule 1) in Hypertrophy-Related Contractile Dysfunction. Circ Res 2017; 121:125-136. [PMID: 28592415 DOI: 10.1161/circresaha.117.311094] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 06/02/2017] [Accepted: 06/07/2017] [Indexed: 12/20/2022]
Abstract
RATIONALE Pathological increases in cardiac afterload result in myocyte hypertrophy with changes in myocyte electrical and mechanical phenotype. Remodeling of contractile and signaling Ca2+ occurs in pathological hypertrophy and is central to myocyte remodeling. STIM1 (stromal interaction molecule 1) regulates Ca2+ signaling in many cell types by sensing low endoplasmic reticular Ca2+ levels and then coupling to plasma membrane Orai channels to induce a Ca2+ influx pathway. Previous reports suggest that STIM1 may play a role in cardiac hypertrophy, but its role in electrical and mechanical phenotypic alterations is not well understood. OBJECTIVE To define the contributions of STIM1-mediated Ca2+ influx on electrical and mechanical properties of normal and diseased myocytes, and to determine whether Orai channels are obligatory partners for STIM1 in these processes using a clinically relevant large animal model of hypertrophy. METHODS AND RESULTS Cardiac hypertrophy was induced by slow progressive pressure overload in adult cats. Hypertrophied myocytes had increased STIM1 expression and activity, which correlated with altered Ca2+-handling and action potential (AP) prolongation. Exposure of hypertrophied myocytes to the Orai channel blocker BTP2 caused a reduction of AP duration and reduced diastolic Ca2+ spark rate. BTP2 had no effect on normal myocytes. Forced expression of STIM1 in cultured adult feline ventricular myocytes increased diastolic spark rate and prolonged AP duration. STIM1 expression produced an increase in the amount of Ca2+ stored within the sarcoplasmic reticulum and activated Ca2+/calmodulin-dependent protein kinase II. STIM1 expression also increased spark rates and induced spontaneous APs. STIM1 effects were eliminated by either BTP2 or by coexpression of a dominant negative Orai construct. CONCLUSIONS STIM1 can associate with Orai in cardiac myocytes to produce a Ca2+ influx pathway that can prolong the AP duration and load the sarcoplasmic reticulum and likely contributes to the altered electromechanical properties of the hypertrophied heart.
Collapse
Affiliation(s)
- Constantine D Troupes
- From the Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA (C.D.T., M.W., G.B., C.Z., S.M., R.M.B., H.K., X.C., S.H.); Department of Cardiology, Medical University of Graz, Austria (D.v.L.); and Fels Institute for Cancer Research and Molecular Biology, Lewis Katz School of Medicine, Temple University School of Medicine, Philadelphia, PA (J.S.)
| | - Markus Wallner
- From the Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA (C.D.T., M.W., G.B., C.Z., S.M., R.M.B., H.K., X.C., S.H.); Department of Cardiology, Medical University of Graz, Austria (D.v.L.); and Fels Institute for Cancer Research and Molecular Biology, Lewis Katz School of Medicine, Temple University School of Medicine, Philadelphia, PA (J.S.)
| | - Giulia Borghetti
- From the Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA (C.D.T., M.W., G.B., C.Z., S.M., R.M.B., H.K., X.C., S.H.); Department of Cardiology, Medical University of Graz, Austria (D.v.L.); and Fels Institute for Cancer Research and Molecular Biology, Lewis Katz School of Medicine, Temple University School of Medicine, Philadelphia, PA (J.S.)
| | - Chen Zhang
- From the Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA (C.D.T., M.W., G.B., C.Z., S.M., R.M.B., H.K., X.C., S.H.); Department of Cardiology, Medical University of Graz, Austria (D.v.L.); and Fels Institute for Cancer Research and Molecular Biology, Lewis Katz School of Medicine, Temple University School of Medicine, Philadelphia, PA (J.S.)
| | - Sadia Mohsin
- From the Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA (C.D.T., M.W., G.B., C.Z., S.M., R.M.B., H.K., X.C., S.H.); Department of Cardiology, Medical University of Graz, Austria (D.v.L.); and Fels Institute for Cancer Research and Molecular Biology, Lewis Katz School of Medicine, Temple University School of Medicine, Philadelphia, PA (J.S.)
| | - Dirk von Lewinski
- From the Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA (C.D.T., M.W., G.B., C.Z., S.M., R.M.B., H.K., X.C., S.H.); Department of Cardiology, Medical University of Graz, Austria (D.v.L.); and Fels Institute for Cancer Research and Molecular Biology, Lewis Katz School of Medicine, Temple University School of Medicine, Philadelphia, PA (J.S.)
| | - Remus M Berretta
- From the Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA (C.D.T., M.W., G.B., C.Z., S.M., R.M.B., H.K., X.C., S.H.); Department of Cardiology, Medical University of Graz, Austria (D.v.L.); and Fels Institute for Cancer Research and Molecular Biology, Lewis Katz School of Medicine, Temple University School of Medicine, Philadelphia, PA (J.S.)
| | - Hajime Kubo
- From the Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA (C.D.T., M.W., G.B., C.Z., S.M., R.M.B., H.K., X.C., S.H.); Department of Cardiology, Medical University of Graz, Austria (D.v.L.); and Fels Institute for Cancer Research and Molecular Biology, Lewis Katz School of Medicine, Temple University School of Medicine, Philadelphia, PA (J.S.)
| | - Xiongwen Chen
- From the Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA (C.D.T., M.W., G.B., C.Z., S.M., R.M.B., H.K., X.C., S.H.); Department of Cardiology, Medical University of Graz, Austria (D.v.L.); and Fels Institute for Cancer Research and Molecular Biology, Lewis Katz School of Medicine, Temple University School of Medicine, Philadelphia, PA (J.S.)
| | - Jonathan Soboloff
- From the Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA (C.D.T., M.W., G.B., C.Z., S.M., R.M.B., H.K., X.C., S.H.); Department of Cardiology, Medical University of Graz, Austria (D.v.L.); and Fels Institute for Cancer Research and Molecular Biology, Lewis Katz School of Medicine, Temple University School of Medicine, Philadelphia, PA (J.S.)
| | - Steven Houser
- From the Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA (C.D.T., M.W., G.B., C.Z., S.M., R.M.B., H.K., X.C., S.H.); Department of Cardiology, Medical University of Graz, Austria (D.v.L.); and Fels Institute for Cancer Research and Molecular Biology, Lewis Katz School of Medicine, Temple University School of Medicine, Philadelphia, PA (J.S.).
| |
Collapse
|
21
|
17β-Estradiol enhances sulforaphane cardioprotection against oxidative stress. J Nutr Biochem 2017; 42:26-36. [DOI: 10.1016/j.jnutbio.2016.12.017] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 12/06/2016] [Accepted: 12/28/2016] [Indexed: 11/19/2022]
|
22
|
Abstract
The tumor suppressor Trp53 (p53) inhibits cell growth after acute stress by regulating gene transcription. The mammalian genome contains hundreds of p53-binding sites. However, whether p53 participates in the regulation of cardiac tissue homeostasis under normal conditions is not known. To examine the physiologic role of p53 in adult cardiomyocytes in vivo, Cre-loxP-mediated conditional gene targeting in adult mice was used. Genome-wide transcriptome analyses of conditional heart-specific p53 knockout mice were performed. Genome-wide annotation and pathway analyses of >5,000 differentially expressed transcripts identified many p53-regulated gene clusters. Correlative analyses identified >20 gene sets containing more than 1,000 genes relevant to cardiac architecture and function. These transcriptomic changes orchestrate cardiac architecture, excitation-contraction coupling, mitochondrial biogenesis, and oxidative phosphorylation capacity. Interestingly, the gene expression signature in p53-deficient hearts confers resistance to acute biomechanical stress. The data presented here demonstrate a role for p53, a previously unrecognized master regulator of the cardiac transcriptome. The complex contributions of p53 define a biological paradigm for the p53 regulator network in the heart under physiological conditions.
Collapse
|
23
|
Weeks KL, Bernardo BC, Ooi JYY, Patterson NL, McMullen JR. The IGF1-PI3K-Akt Signaling Pathway in Mediating Exercise-Induced Cardiac Hypertrophy and Protection. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1000:187-210. [PMID: 29098623 DOI: 10.1007/978-981-10-4304-8_12] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Regular physical activity or exercise training can lead to heart enlargement known as cardiac hypertrophy. Cardiac hypertrophy is broadly defined as an increase in heart mass. In adults, cardiac hypertrophy is often considered a poor prognostic sign because it often progresses to heart failure. Heart enlargement in a setting of cardiac disease is referred to as pathological cardiac hypertrophy and is typically characterized by cell death and depressed cardiac function. By contrast, physiological cardiac hypertrophy, as occurs in response to chronic exercise training (i.e. the 'athlete's heart'), is associated with normal or enhanced cardiac function. The following chapter describes the morphologically distinct types of heart growth, and the key role of the insulin-like growth factor 1 (IGF1) - phosphoinositide 3-kinase (PI3K)-Akt signaling pathway in regulating exercise-induced physiological cardiac hypertrophy and cardiac protection. Finally we summarize therapeutic approaches that target the IGF1-PI3K-Akt signaling pathway which are showing promise in preclinical models of heart disease.
Collapse
Affiliation(s)
- Kate L Weeks
- Baker Heart & Diabetes Institute, P.O. Box 6492, Melbourne, VIC, 3004, Australia.
| | - Bianca C Bernardo
- Baker Heart & Diabetes Institute, P.O. Box 6492, Melbourne, VIC, 3004, Australia
| | - Jenny Y Y Ooi
- Baker Heart & Diabetes Institute, P.O. Box 6492, Melbourne, VIC, 3004, Australia
| | - Natalie L Patterson
- Baker Heart & Diabetes Institute, P.O. Box 6492, Melbourne, VIC, 3004, Australia
| | - Julie R McMullen
- Baker Heart & Diabetes Institute, P.O. Box 6492, Melbourne, VIC, 3004, Australia.
| |
Collapse
|
24
|
An CI, Ichihashi Y, Peng J, Sinha NR, Hagiwara N. Transcriptome Dynamics and Potential Roles of Sox6 in the Postnatal Heart. PLoS One 2016; 11:e0166574. [PMID: 27832192 PMCID: PMC5104335 DOI: 10.1371/journal.pone.0166574] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2016] [Accepted: 10/31/2016] [Indexed: 01/20/2023] Open
Abstract
The postnatal heart undergoes highly coordinated developmental processes culminating in the complex physiologic properties of the adult heart. The molecular mechanisms of postnatal heart development remain largely unexplored despite their important clinical implications. To gain an integrated view of the dynamic changes in gene expression during postnatal heart development at the organ level, time-series transcriptome analyses of the postnatal hearts of neonatal through adult mice (P1, P7, P14, P30, and P60) were performed using a newly developed bioinformatics pipeline. We identified functional gene clusters by principal component analysis with self-organizing map clustering which revealed organized, discrete gene expression patterns corresponding to biological functions associated with the neonatal, juvenile and adult stages of postnatal heart development. Using weighted gene co-expression network analysis with bootstrap inference for each of these functional gene clusters, highly robust hub genes were identified which likely play key roles in regulating expression of co-expressed, functionally linked genes. Additionally, motivated by the role of the transcription factor Sox6 in the functional maturation of skeletal muscle, the role of Sox6 in the postnatal maturation of cardiac muscle was investigated. Differentially expressed transcriptome analyses between Sox6 knockout (KO) and control hearts uncovered significant upregulation of genes involved in cell proliferation at postnatal day 7 (P7) in the Sox6 KO heart. This result was validated by detecting mitotically active cells in the P7 Sox6 KO heart. The current report provides a framework for the complex molecular processes of postnatal heart development, thus enabling systematic dissection of the developmental regression observed in the stressed and failing adult heart.
Collapse
Affiliation(s)
- Chung-Il An
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California Davis, Davis, California, United States of America
- * E-mail: (CA); (YI); (NH)
| | - Yasunori Ichihashi
- Department of Plant Biology, University of California Davis, Davis, California, United States of America
- * E-mail: (CA); (YI); (NH)
| | - Jie Peng
- Department of Statistics, University of California Davis, Davis, California, United States of America
| | - Neelima R. Sinha
- Department of Plant Biology, University of California Davis, Davis, California, United States of America
| | - Nobuko Hagiwara
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California Davis, Davis, California, United States of America
- * E-mail: (CA); (YI); (NH)
| |
Collapse
|
25
|
Sala V, Gallo S, Gatti S, Medico E, Vigna E, Cantarella D, Fontani L, Natale M, Cimino J, Morello M, Comoglio PM, Ponzetto A, Crepaldi T. Cardiac concentric hypertrophy promoted by activated Met receptor is mitigated in vivo by inhibition of Erk1,2 signalling with Pimasertib. J Mol Cell Cardiol 2016; 93:84-97. [PMID: 26924269 DOI: 10.1016/j.yjmcc.2016.02.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 02/08/2016] [Accepted: 02/22/2016] [Indexed: 12/25/2022]
Abstract
Cardiac hypertrophy is a major risk factor for heart failure. Hence, its attenuation represents an important clinical goal. Erk1,2 signalling is pivotal in the cardiac response to stress, suggesting that its inhibition may be a good strategy to revert heart hypertrophy. In this work, we unveiled the events associated with cardiac hypertrophy by means of a transgenic model expressing activated Met receptor. c-Met proto-oncogene encodes for the tyrosine kinase receptor of Hepatocyte growth factor and is a strong inducer of Ras-Raf-Mek-Erk1,2 pathway. We showed that three weeks after the induction of activated Met, the heart presents a remarkable concentric hypertrophy, with no signs of congestive failure and preserved contractility. Cardiac enlargement is accompanied by upregulation of growth-regulating transcription factors, natriuretic peptides, cytoskeletal proteins, and Extracellular Matrix remodelling factors (Timp1 and Pai1). At a later stage, cardiac hypertrophic remodelling results into heart failure with preserved systolic function. Prevention trial by suppressing activated Met showed that cardiac hypertrophy is reversible, and progression to heart failure is prevented. Notably, treatment with Pimasertib, Mek1 inhibitor, attenuates cardiac hypertrophy and remodelling. Our results suggest that modulation of Erk1.2 signalling may constitute a new therapeutic approach for treating cardiac hypertrophies.
Collapse
Affiliation(s)
- Valentina Sala
- Department of Medical Sciences, University of Turin, 10126 Turin, Italy; Department of Molecular Biotechnology and Health Sciences, University of Turin, 10126 Turin, Italy
| | - Simona Gallo
- Department of Oncology, University of Turin, 10126 Turin, Italy
| | - Stefano Gatti
- Department of Oncology, University of Turin, 10126 Turin, Italy
| | - Enzo Medico
- Department of Oncology, University of Turin, 10126 Turin, Italy; FPO-IRCCS, 10060 Candiolo, TO, Italy
| | - Elisa Vigna
- Department of Oncology, University of Turin, 10126 Turin, Italy; FPO-IRCCS, 10060 Candiolo, TO, Italy
| | | | | | | | - James Cimino
- Department of Molecular Biotechnology and Health Sciences, University of Turin, 10126 Turin, Italy
| | - Mara Morello
- Department of Medical Sciences, University of Turin, 10126 Turin, Italy
| | - Paolo Maria Comoglio
- Department of Oncology, University of Turin, 10126 Turin, Italy; FPO-IRCCS, 10060 Candiolo, TO, Italy
| | - Antonio Ponzetto
- Department of Medical Sciences, University of Turin, 10126 Turin, Italy
| | - Tiziana Crepaldi
- Department of Oncology, University of Turin, 10126 Turin, Italy.
| |
Collapse
|
26
|
van Deel E, Ridwan Y, van Vliet JN, Belenkov S, Essers J. In Vivo Quantitative Assessment of Myocardial Structure, Function, Perfusion and Viability Using Cardiac Micro-computed Tomography. J Vis Exp 2016:53603. [PMID: 26967592 PMCID: PMC4828165 DOI: 10.3791/53603] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
The use of Micro-Computed Tomography (MicroCT) for in vivo studies of small animals as models of human disease has risen tremendously due to the fact that MicroCT provides quantitative high-resolution three-dimensional (3D) anatomical data non-destructively and longitudinally. Most importantly, with the development of a novel preclinical iodinated contrast agent called eXIA160, functional and metabolic assessment of the heart became possible. However, prior to the advent of commercial MicroCT scanners equipped with X-ray flat-panel detector technology and easy-to-use cardio-respiratory gating, preclinical studies of cardiovascular disease (CVD) in small animals required a MicroCT technologist with advanced skills, and thus were impractical for widespread implementation. The goal of this work is to provide a practical guide to the use of the high-speed Quantum FX MicroCT system for comprehensive determination of myocardial global and regional function along with assessment of myocardial perfusion, metabolism and viability in healthy mice and in a cardiac ischemia mouse model induced by permanent occlusion of the left anterior descending coronary artery (LAD).
Collapse
Affiliation(s)
- Elza van Deel
- Department of Genetics, Erasmus MC, Rotterdam; Department of Experimental Cardiology, Erasmus MC, Rotterdam
| | | | | | | | - Jeroen Essers
- Department of Genetics, Erasmus MC, Rotterdam; Department of Vascular Surgery, Erasmus MC, Rotterdam; Department of Radiation Oncology, Erasmus MC, Rotterdam;
| |
Collapse
|
27
|
Ladd AN. New Insights Into the Role of RNA-Binding Proteins in the Regulation of Heart Development. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2016; 324:125-85. [PMID: 27017008 DOI: 10.1016/bs.ircmb.2015.12.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The regulation of gene expression during development takes place both at the transcriptional and posttranscriptional levels. RNA-binding proteins (RBPs) regulate pre-mRNA processing, mRNA localization, stability, and translation. Many RBPs are expressed in the heart and have been implicated in heart development, function, or disease. This chapter will review the current knowledge about RBPs in the developing heart, focusing on those that regulate posttranscriptional gene expression. The involvement of RBPs at each stage of heart development will be considered in turn, including the establishment of specific cardiac cell types and formation of the primitive heart tube, cardiac morphogenesis, and postnatal maturation and aging. The contributions of RBPs to cardiac birth defects and heart disease will also be considered in these contexts. Finally, the interplay between RBPs and other regulatory factors in the developing heart, such as transcription factors and miRNAs, will be discussed.
Collapse
Affiliation(s)
- A N Ladd
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States of America.
| |
Collapse
|
28
|
Valiente-Alandi I, Albo-Castellanos C, Herrero D, Arza E, Garcia-Gomez M, Segovia JC, Capecchi M, Bernad A. Cardiac Bmi1(+) cells contribute to myocardial renewal in the murine adult heart. Stem Cell Res Ther 2015; 6:205. [PMID: 26503423 PMCID: PMC4620653 DOI: 10.1186/s13287-015-0196-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Revised: 09/15/2015] [Accepted: 10/02/2015] [Indexed: 12/19/2022] Open
Abstract
INTRODUCTION The mammalian adult heart maintains a continuous, low cardiomyocyte turnover rate throughout life. Although many cardiac stem cell populations have been studied, the natural source for homeostatic repair has not yet been defined. The Polycomb protein BMI1 is the most representative marker of mouse adult stem cell systems. We have evaluated the relevance and role of cardiac Bmi1 (+) cells in cardiac physiological homeostasis. METHODS Bmi1 (CreER/+);Rosa26 (YFP/+) (Bmi1-YFP) mice were used for lineage tracing strategy. After tamoxifen (TM) induction, yellow fluorescent protein (YFP) is expressed under the control of Rosa26 regulatory sequences in Bmi1 (+) cells. These cells and their progeny were tracked by FACS, immunofluorescence and RT-qPCR techniques from 5 days to 1 year. RESULTS FACS analysis of non-cardiomyocyte compartment from TM-induced Bmi1-YFP mice showed a Bmi1 (+)-expressing cardiac progenitor cell (Bmi1-CPC: B-CPC) population, SCA-1 antigen-positive (95.9 ± 0.4 %) that expresses some stemness-associated genes. B-CPC were also able to differentiate in vitro to the three main cardiac lineages. Pulse-chase analysis showed that B-CPC remained quite stable for extended periods (up to 1 year), which suggests that this Bmi1 (+) population contains cardiac progenitors with substantial self-maintenance potential. Specific immunostaining of Bmi1-YFP hearts serial sections 5 days post-TM induction indicated broad distribution of B-CPC, which were detected in variably sized clusters, although no YFP(+) cardiomyocytes (CM) were detected at this time. Between 2 to 12 months after TM induction, YFP(+) CM were clearly identified (3 ± 0.6 % to 6.7 ± 1.3 %) by immunohistochemistry of serial sections and by flow cytometry of total freshly isolated CM. B-CPC also contributed to endothelial and smooth muscle (SM) lineages in vivo. CONCLUSIONS High Bmi1 expression identifies a non-cardiomyocyte resident cardiac population (B-CPC) that contributes to the main lineages of the heart in vitro and in vivo.
Collapse
Affiliation(s)
- Iñigo Valiente-Alandi
- Cardiovascular Development and Repair Department, Spanish National Cardiovascular Research Center (CNIC), Madrid, Spain.
- The Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
| | - Carmen Albo-Castellanos
- Cardiovascular Development and Repair Department, Spanish National Cardiovascular Research Center (CNIC), Madrid, Spain.
- Vivebiotech, San Sebastian, Spain.
| | - Diego Herrero
- Cardiovascular Development and Repair Department, Spanish National Cardiovascular Research Center (CNIC), Madrid, Spain.
- Immunology and Oncology Department, Spanish National Center for Biotechnology (CNB-CSIC), Madrid, Spain.
| | - Elvira Arza
- Microscopy Unit, Spanish National Cardiovascular Research Center (CNIC), Madrid, Spain.
| | - Maria Garcia-Gomez
- Hematopoietic Innovative Therapies Division, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT)- Centro de Investigaciones Biomédicas en Red de Enfermedades Raras (CIBERER), Madrid, Spain.
- Advanced Therapies Mixed Unit, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz (IIS-FJD, UAM), Madrid, Spain.
| | - José C Segovia
- Hematopoietic Innovative Therapies Division, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT)- Centro de Investigaciones Biomédicas en Red de Enfermedades Raras (CIBERER), Madrid, Spain.
- Advanced Therapies Mixed Unit, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz (IIS-FJD, UAM), Madrid, Spain.
| | - Mario Capecchi
- Howard Hughes Medical Institute University of Utah, Salt Lake City, UT, USA.
| | - Antonio Bernad
- Cardiovascular Development and Repair Department, Spanish National Cardiovascular Research Center (CNIC), Madrid, Spain.
- Immunology and Oncology Department, Spanish National Center for Biotechnology (CNB-CSIC), Madrid, Spain.
| |
Collapse
|
29
|
Gao W, Pan B, Liu L, Huang X, Liu Z, Tian J. Alcohol exposure increases the expression of cardiac transcription factors through ERK1/2-mediated histone3 hyperacetylation in H9c2 cells. Biochem Biophys Res Commun 2015; 466:670-5. [DOI: 10.1016/j.bbrc.2015.09.090] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 09/16/2015] [Indexed: 12/14/2022]
|
30
|
Clark AL, Naya FJ. MicroRNAs in the Myocyte Enhancer Factor 2 (MEF2)-regulated Gtl2-Dio3 Noncoding RNA Locus Promote Cardiomyocyte Proliferation by Targeting the Transcriptional Coactivator Cited2. J Biol Chem 2015; 290:23162-72. [PMID: 26240138 DOI: 10.1074/jbc.m115.672659] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Indexed: 01/04/2023] Open
Abstract
Understanding cell cycle regulation in postmitotic cardiomyocytes may lead to new therapeutic approaches to regenerate damaged cardiac tissue. We have demonstrated previously that microRNAs encoded by the Gtl2-Dio3 noncoding RNA locus function downstream of the MEF2A transcription factor in skeletal muscle regeneration. We have also reported expression of these miRNAs in the heart. Here we investigated the role of two Gtl2-Dio3 miRNAs, miR-410 and miR-495, in cardiac muscle. Overexpression of miR-410 and miR-495 robustly stimulated cardiomyocyte DNA synthesis and proliferation. Interestingly, unlike our findings in skeletal muscle, these miRNAs did not modulate the activity of the WNT signaling pathway. Instead, these miRNAs targeted Cited2, a coactivator required for proper cardiac development. Consistent with miR-410 and miR-495 overexpression, siRNA knockdown of Cited2 in neonatal cardiomyocytes resulted in robust proliferation. This phenotype was associated with reduced expression of Cdkn1c/p57/Kip2, a cell cycle inhibitor, and increased expression of VEGFA, a growth factor with proliferation-promoting effects. Therefore, miR-410 and miR-495 are among a growing number of miRNAs that have the ability to potently stimulate neonatal cardiomyocyte proliferation.
Collapse
Affiliation(s)
- Amanda L Clark
- From the Department of Biology, Program in Cell and Molecular Biology, Boston University, Boston, Massachusetts 02215
| | - Francisco J Naya
- From the Department of Biology, Program in Cell and Molecular Biology, Boston University, Boston, Massachusetts 02215
| |
Collapse
|
31
|
Cotecchia S, Del Vescovo CD, Colella M, Caso S, Diviani D. The alpha1-adrenergic receptors in cardiac hypertrophy: signaling mechanisms and functional implications. Cell Signal 2015; 27:1984-93. [PMID: 26169957 DOI: 10.1016/j.cellsig.2015.06.009] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Revised: 06/22/2015] [Accepted: 06/30/2015] [Indexed: 01/05/2023]
Abstract
Cardiac hypertrophy is a complex remodeling process of the heart induced by physiological or pathological stimuli resulting in increased cardiomyocyte size and myocardial mass. Whereas cardiac hypertrophy can be an adaptive mechanism to stressful conditions of the heart, prolonged hypertrophy can lead to heart failure which represents the primary cause of human morbidity and mortality. Among G protein-coupled receptors, the α1-adrenergic receptors (α1-ARs) play an important role in the development of cardiac hypertrophy as demonstrated by numerous studies in the past decades, both in primary cardiomyocyte cultures and genetically modified mice. The results of these studies have provided evidence of a large variety of α1-AR-induced signaling events contributing to the defining molecular and cellular features of cardiac hypertrophy. Recently, novel signaling mechanisms have been identified and new hypotheses have emerged concerning the functional role of the α1-adrenergic receptors in the heart. This review will summarize the main signaling pathways activated by the α1-AR in the heart and their functional implications in cardiac hypertrophy.
Collapse
Affiliation(s)
- Susanna Cotecchia
- Dipartimento di Bioscienze, Biotecnologie e Biofarmaceutica, Università di Bari, Via Orabona 4, 70125 Bari, Italy.
| | - Cosmo Damiano Del Vescovo
- Department de Pharmacologie et de de Toxicologie, Université de Lausanne, Rue du Bugnon 27, 1005, Lausanne, Switzerland
| | - Matilde Colella
- Dipartimento di Bioscienze, Biotecnologie e Biofarmaceutica, Università di Bari, Via Orabona 4, 70125 Bari, Italy
| | - Stefania Caso
- Dipartimento di Bioscienze, Biotecnologie e Biofarmaceutica, Università di Bari, Via Orabona 4, 70125 Bari, Italy; Department de Pharmacologie et de de Toxicologie, Université de Lausanne, Rue du Bugnon 27, 1005, Lausanne, Switzerland
| | - Dario Diviani
- Department de Pharmacologie et de de Toxicologie, Université de Lausanne, Rue du Bugnon 27, 1005, Lausanne, Switzerland
| |
Collapse
|
32
|
Liang D, Li J, Wu Y, Zhen L, Li C, Qi M, Wang L, Deng F, Huang J, Lv F, Liu Y, Ma X, Yu Z, Zhang Y, Chen YH. miRNA-204 drives cardiomyocyte proliferation via targeting Jarid2. Int J Cardiol 2015; 201:38-48. [PMID: 26298346 DOI: 10.1016/j.ijcard.2015.06.163] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Revised: 06/11/2015] [Accepted: 06/29/2015] [Indexed: 02/07/2023]
Abstract
OBJECTIVES In mammals, the heart grows by hypertrophy but not proliferation of cardiomyocytes after birth. The paucity of cardiomyocyte proliferation limits cardiac regeneration in a variety of heart diseases. To explore the efficient strategies that drive cardiomyocyte proliferation, we employed in vitro and in vivo models to investigate the function of miRNA-204, which was demonstrated to regulate the proliferation and differentiation of human cardiac progenitor cells in our previous study. METHODS AND RESULTS miRNA-204 overexpression markedly promoted cardiomyocyte proliferation in both neonatal and adult rat cardiomyocytes in vitro. Transgenic mice with the cardiac-specific overexpression of miRNA-204 exhibited excessive cardiomyocyte proliferation throughout the embryonic and adult stages, leading to a pronounced increase in ventricular mass. Accordingly, the cell cycle regulators, including Cyclin A, Cyclin B, Cyclin D2, Cyclin E, CDC2 and PCNA, were upregulated in miRNA-204 transgenic embryonic hearts. Furthermore, we demonstrated that miRNA-204 directly targeted Jarid2. Knockdown of Jarid2 mimicked the pro-proliferative effect of miRNA-204 overexpression on cultured rat cardiomyocytes, whereas enhanced expression of Jarid2 conferred the myocytes with substantial resistance to proliferation by miRNA-204 overexpression. CONCLUSION Our findings identify a conserved role for miRNA-204 in regulating cardiomyocyte proliferation by targeting the Jarid2 signaling pathway.
Collapse
Affiliation(s)
- Dandan Liang
- Key Laboratory of Arrhythmias of the Ministry of Education of China, East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Research Center for Translational Medicine, Tongji University School of Medicine, Shanghai 200120, China; Institute of Medical Genetics, Tongji University, Shanghai 200092, China
| | - Jun Li
- Key Laboratory of Arrhythmias of the Ministry of Education of China, East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Research Center for Translational Medicine, Tongji University School of Medicine, Shanghai 200120, China; Institute of Medical Genetics, Tongji University, Shanghai 200092, China
| | - Yahan Wu
- Key Laboratory of Arrhythmias of the Ministry of Education of China, East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Department of Cardiology, East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Department of Pathology and Pathophysiology, Tongji University School of Medicine, Shanghai 200092, China
| | - Lixiao Zhen
- Key Laboratory of Arrhythmias of the Ministry of Education of China, East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Department of Cardiology, East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Department of Pathology and Pathophysiology, Tongji University School of Medicine, Shanghai 200092, China
| | - Changming Li
- Key Laboratory of Arrhythmias of the Ministry of Education of China, East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Department of Cardiology, East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Department of Pathology and Pathophysiology, Tongji University School of Medicine, Shanghai 200092, China
| | - Man Qi
- Key Laboratory of Arrhythmias of the Ministry of Education of China, East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Department of Cardiology, East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Department of Pathology and Pathophysiology, Tongji University School of Medicine, Shanghai 200092, China
| | - Lijie Wang
- Key Laboratory of Arrhythmias of the Ministry of Education of China, East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Department of Cardiology, East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Department of Pathology and Pathophysiology, Tongji University School of Medicine, Shanghai 200092, China
| | - Fangfei Deng
- Key Laboratory of Arrhythmias of the Ministry of Education of China, East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Department of Cardiology, East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Department of Pathology and Pathophysiology, Tongji University School of Medicine, Shanghai 200092, China
| | - Jian Huang
- Key Laboratory of Arrhythmias of the Ministry of Education of China, East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Department of Cardiology, East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Department of Pathology and Pathophysiology, Tongji University School of Medicine, Shanghai 200092, China
| | - Fei Lv
- Key Laboratory of Arrhythmias of the Ministry of Education of China, East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Department of Cardiology, East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Department of Pathology and Pathophysiology, Tongji University School of Medicine, Shanghai 200092, China
| | - Yi Liu
- Key Laboratory of Arrhythmias of the Ministry of Education of China, East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Research Center for Translational Medicine, Tongji University School of Medicine, Shanghai 200120, China; Institute of Medical Genetics, Tongji University, Shanghai 200092, China
| | - Xiue Ma
- Key Laboratory of Arrhythmias of the Ministry of Education of China, East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Department of Cardiology, East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Department of Pathology and Pathophysiology, Tongji University School of Medicine, Shanghai 200092, China
| | - Zuoren Yu
- Key Laboratory of Arrhythmias of the Ministry of Education of China, East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Research Center for Translational Medicine, Tongji University School of Medicine, Shanghai 200120, China
| | - Yuzhen Zhang
- Key Laboratory of Arrhythmias of the Ministry of Education of China, East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Research Center for Translational Medicine, Tongji University School of Medicine, Shanghai 200120, China
| | - Yi-Han Chen
- Key Laboratory of Arrhythmias of the Ministry of Education of China, East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Research Center for Translational Medicine, Tongji University School of Medicine, Shanghai 200120, China; Institute of Medical Genetics, Tongji University, Shanghai 200092, China; Department of Cardiology, East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Department of Pathology and Pathophysiology, Tongji University School of Medicine, Shanghai 200092, China.
| |
Collapse
|
33
|
Tadevosyan A, Allen BG, Nattel S. Isolation and study of cardiac nuclei from canine myocardium and adult ventricular myocytes. Methods Mol Biol 2015; 1234:69-80. [PMID: 25304349 DOI: 10.1007/978-1-4939-1755-6_7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
The nuclear envelope encloses the genome as well as the molecular machinery responsible for both the replication and transcription of DNA as well as the maturation of nascent RNA. Recent studies ascribe a growing number of functions to the nuclear membrane, in addition to sequestering the DNA, through receptors and their effectors, ion channels, as well as ion pumps and transporters located within the nuclear membrane itself. Despite the obvious structural and functional importance of the nucleus, certain aspects remain poorly understood due to the challenges associated with its accessibility in vivo, as well as isolating nuclei intact and with sufficient purity from cardiac cells to permit studies in vitro. Here, we present a detailed protocol for isolation of intact nuclei from both myocardial tissue and freshly isolated adult ventricular cardiomyocytes. These methods are based on partial permeabilization of plasma membrane with digitonin and cell disruption, followed by differential and discontinuous sucrose density centrifugation. These preparations provide for rapid separation of nonnuclear membranes and cytosol from nuclei.
Collapse
Affiliation(s)
- Artavazd Tadevosyan
- Montreal Heart Institute, 5000 Belanger Street, Montreal, QC, Canada, H1T 1C8
| | | | | |
Collapse
|
34
|
Pluess M, Daeubler G, Dos Remedios CG, Ehler E. Adaptations of cytoarchitecture in human dilated cardiomyopathy. Biophys Rev 2015; 7:25-32. [PMID: 28509975 PMCID: PMC4322184 DOI: 10.1007/s12551-014-0146-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Accepted: 11/10/2014] [Indexed: 12/30/2022] Open
Abstract
Hypertrophic cardiomyopathy is characterised by a histological phenotype of myocyte disarray, but heart tissue samples from patients with dilated cardiomyopathy (DCM) often look comparatively similar to those from healthy individuals apart from conspicuous regions of fibrosis and necrosis. We have previously investigated subcellular alterations in the cytoarchitecture of mouse models of dilated cardiomyopathy and found that both the organisation and composition of the intercalated disc, i.e. the specialised type of cell-cell contact in the heart, is altered. There is also is a change in the composition of the M-band of the sarcomere due to an expression shift towards the more extensible embryonic heart (EH)-myomesin isoform. Analysis of human samples from the Sydney Human Heart Tissue Bank have revealed similar structural findings and also provided evidence for a dramatic change in overall cardiomyocyte size control, which has also been seen in the mouse. Together these changes in cytoarchitecture probably contribute to the decreased functional output that is seen in DCM.
Collapse
Affiliation(s)
- Marlene Pluess
- Randall Division of Cell and Molecular Biophysics and Cardiovascular Division, British Heart Foundation Centre of Research Excellence, King's College London, New Hunt's House, Guy's Campus, London, SE1 1UL, UK
| | - Gregor Daeubler
- Randall Division of Cell and Molecular Biophysics and Cardiovascular Division, British Heart Foundation Centre of Research Excellence, King's College London, New Hunt's House, Guy's Campus, London, SE1 1UL, UK
| | | | - Elisabeth Ehler
- Randall Division of Cell and Molecular Biophysics and Cardiovascular Division, British Heart Foundation Centre of Research Excellence, King's College London, New Hunt's House, Guy's Campus, London, SE1 1UL, UK.
| |
Collapse
|
35
|
Zhao QD, Viswanadhapalli S, Williams P, Shi Q, Tan C, Yi X, Bhandari B, Abboud HE. NADPH oxidase 4 induces cardiac fibrosis and hypertrophy through activating Akt/mTOR and NFκB signaling pathways. Circulation 2015; 131:643-55. [PMID: 25589557 DOI: 10.1161/circulationaha.114.011079] [Citation(s) in RCA: 203] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
BACKGROUND NADPH oxidase 4 (Nox4) has been implicated in cardiac remodeling, but its precise role in cardiac injury remains controversial. Furthermore, little is known about the downstream effector signaling pathways activated by Nox4-derived reactive oxygen species in the myocardium. We investigated the role of Nox4 and Nox4-associated signaling pathways in the development of cardiac remodeling. METHODS AND RESULTS Cardiac-specific human Nox4 transgenic mice (c-hNox4Tg) were generated. Four groups of mice were studied: (1) control mice, littermates that are negative for hNox4 transgene but Cre positive; (2) c-hNox4 Tg mice; (3) angiotensin II (AngII)-infused control mice; and (4) c-hNox4Tg mice infused with AngII. The c-hNox4Tg mice exhibited an ≈10-fold increase in Nox4 protein expression and an 8-fold increase in the production of reactive oxygen species, and manifested cardiac interstitial fibrosis. AngII infusion to control mice increased cardiac Nox4 expression and induced fibrosis and hypertrophy. The Tg mice receiving AngII exhibited more advanced cardiac remodeling and robust elevation in Nox4 expression, indicating that AngII worsens cardiac injury, at least in part by enhancing Nox4 expression. Moreover, hNox4 transgene and AngII infusion induced the expression of cardiac fetal genes and activated the Akt-mTOR and NFκB signaling pathways. Treatment of AngII-infused c-hNox4Tg mice with GKT137831, a Nox4/Nox1 inhibitor, abolished the increase in oxidative stress, suppressed the Akt-mTOR and NFκB signaling pathways, and attenuated cardiac remodeling. CONCLUSIONS Upregulation of Nox4 in the myocardium causes cardiac remodeling through activating Akt-mTOR and NFκB signaling pathways. Inhibition of Nox4 has therapeutic potential to treat cardiac remodeling.
Collapse
Affiliation(s)
- Qingwei David Zhao
- From the Department of Medicine, University of Texas Health Science Center and South Texas Veterans Health Care System, San Antonio, TX.
| | - Suryavathi Viswanadhapalli
- From the Department of Medicine, University of Texas Health Science Center and South Texas Veterans Health Care System, San Antonio, TX
| | - Paul Williams
- From the Department of Medicine, University of Texas Health Science Center and South Texas Veterans Health Care System, San Antonio, TX
| | - Qian Shi
- From the Department of Medicine, University of Texas Health Science Center and South Texas Veterans Health Care System, San Antonio, TX
| | - Chunyan Tan
- From the Department of Medicine, University of Texas Health Science Center and South Texas Veterans Health Care System, San Antonio, TX
| | - Xiaolan Yi
- From the Department of Medicine, University of Texas Health Science Center and South Texas Veterans Health Care System, San Antonio, TX
| | - Basant Bhandari
- From the Department of Medicine, University of Texas Health Science Center and South Texas Veterans Health Care System, San Antonio, TX
| | - Hanna E Abboud
- From the Department of Medicine, University of Texas Health Science Center and South Texas Veterans Health Care System, San Antonio, TX
| |
Collapse
|
36
|
Abstract
The last decade has witnessed the publication of a large number of clinical trials, primarily using bone marrow-derived stem cells as the injected cell. Much has been learned through these "first-generation" clinical trials. The considerable advances in our understanding include (1) cell therapy is safe, (2) cell therapy has been modestly effective, (3) the recognition that in humans bone marrow-derived stem cells do not transdifferentiate into cardiomyocytes or new blood vessels (or at least in sufficient numbers to have any effect). The primary mechanism of action for cell therapy is now believed to be through paracrine effects that include the release of cytokines, chemokines, and growth factors that inhibit apoptosis and fibrosis, enhance contractility, and activate endogenous regenerative mechanisms through endogenous circulating or site-specific stem cells. The new direction for clinical trials includes the use of stem cells capable of cardiac lineage, such as endogenous cardiac stem cells.
Collapse
|
37
|
Cho GS, Fernandez L, Kwon C. Regenerative medicine for the heart: perspectives on stem-cell therapy. Antioxid Redox Signal 2014; 21:2018-31. [PMID: 25133793 PMCID: PMC4208610 DOI: 10.1089/ars.2014.6063] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
SIGNIFICANCE Despite decades of progress in cardiovascular biology and medicine, heart disease remains the leading cause of death, and there is no cure for the failing heart. Since heart failure is mostly caused by loss or dysfunction of cardiomyocytes (CMs), replacing dead or damaged CMs with new CMs might be an ideal way to reverse the disease. However, the adult heart is composed mainly of terminally differentiated CMs that have no significant self-regeneration capacity. RECENT ADVANCES Stem cells have tremendous regenerative potential and, thus, current cardiac regenerative research has focused on developing stem cell sources to repair damaged myocardium. CRITICAL ISSUES In this review, we examine the potential sources of cells that could be used for heart therapies, including embryonic stem cells and induced pluripotent stem cells, as well as alternative methods for activating the endogenous regenerative mechanisms of the heart via transdifferentiation and cell reprogramming. We also discuss the current state of knowledge of cell purification, delivery, and retention. FUTURE DIRECTIONS Efforts are underway to improve the current stem cell strategies and methodologies, which will accelerate the development of innovative stem-cell therapies for heart regeneration.
Collapse
Affiliation(s)
- Gun-Sik Cho
- Division of Cardiology, Department of Medicine, Institute for Cell Engineering, Johns Hopkins University , Baltimore, Maryland
| | | | | |
Collapse
|
38
|
The heart: mostly postmitotic or mostly premitotic? Myocyte cell cycle, senescence, and quiescence. Can J Cardiol 2014; 30:1270-8. [PMID: 25442430 DOI: 10.1016/j.cjca.2014.08.014] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Revised: 08/21/2014] [Accepted: 08/21/2014] [Indexed: 11/21/2022] Open
Abstract
The concept of myocyte division and myocyte-mediated regeneration has re-emerged in the past 5 years through development of sophisticated transgenic mice and carbon-dating of cells. Although recently, a couple of studies have been conducted as an attempt to intervene in myocyte division, the efficiency in adult animals remains discouragingly low. Re-enforcing myocyte division is a vision that has been desired for decades, leading to years of experience in myocyte resistance to proproliferative stimuli. Previous attempts have indeed provided a platform for basic knowledge on molecular players and signalling in myocytes. However, natural biological processes such as hypertrophy and binucleation provide layers of complexity in interpretation of previous and current findings. A major hurdle in mediating myocyte division is a lack of insight in the myocyte cell cycle. To date, no knowledge is gained on myoycte cell cycle progression and/or duration. This review will include an overview of previous and current literature on myocyte cell cycle and division. Furthermore, the limitations of current approaches and basic questions that might be essential in understanding myocardial resistance to division will be discussed.
Collapse
|
39
|
Hudson JE, Porrello ER. The non-coding road towards cardiac regeneration. J Cardiovasc Transl Res 2014; 6:909-23. [PMID: 23797382 DOI: 10.1007/s12265-013-9486-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2013] [Accepted: 06/05/2013] [Indexed: 12/31/2022]
Abstract
Our understanding of cardiovascular disease has evolved rapidly, leading to a number of treatments that have improved patient quality of life and mortality rates. However, there is still no cure for heart failure. This has led to the pursuit of cardiac regeneration to prevent, and ultimately cure, this debilitating condition. To this end, several approaches have been proposed, including activation of cardiomyocyte proliferation, activation of endogenous or exogenous stem/progenitor cells, delivery of de novo cardiomyocytes, and in situ direct reprogramming of cardiac fibroblasts. While these different methodologies are currently being intensely investigated, there are still a number of caveats limiting their application in the clinic. Given the emerging regulatory potential of non-coding RNAs for controlling diverse cellular processes, these molecules may offer potential solutions in this pursuit of cardiac regeneration. In this concise review, we discuss the potential role of non-coding RNAs in a variety of different cardiac regenerative approaches.
Collapse
|
40
|
Abstract
Although the adult mammalian heart was once believed to be a post-mitotic organ without any capacity for regeneration, recent findings have challenged this dogma. A modified view assigns to the mammalian heart a measurable capacity for regeneration throughout life. The ultimate goals of the cardiac regeneration field have been pursued by multiple strategies, including understanding the developmental biology of cardiomyocytes and cardiac stem and progenitor cells, applying chemical genetics, and engineering biomaterials and delivery methods that facilitate cell transplantation. Successful stimulation of endogenous regenerative capacity in injured adult mammalian hearts can benefit from studies of natural cardiac regeneration.
Collapse
Affiliation(s)
- Aurora Bernal
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), C/Melchor Fernández Almagro, 3, 28029 Madrid, Spain
| | - Beatriz G. Gálvez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), C/Melchor Fernández Almagro, 3, 28029 Madrid, Spain
| |
Collapse
|
41
|
Javadov S, Jang S, Agostini B. Crosstalk between mitogen-activated protein kinases and mitochondria in cardiac diseases: therapeutic perspectives. Pharmacol Ther 2014; 144:202-25. [PMID: 24924700 DOI: 10.1016/j.pharmthera.2014.05.013] [Citation(s) in RCA: 119] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Accepted: 05/30/2014] [Indexed: 02/07/2023]
Abstract
Cardiovascular diseases cause more mortality and morbidity worldwide than any other diseases. Although many intracellular signaling pathways influence cardiac physiology and pathology, the mitogen-activated protein kinase (MAPK) family has garnered significant attention because of its vast implications in signaling and crosstalk with other signaling networks. The extensively studied MAPKs ERK1/2, p38, JNK, and ERK5, demonstrate unique intracellular signaling mechanisms, responding to a myriad of mitogens and stressors and influencing the signaling of cardiac development, metabolism, performance, and pathogenesis. Definitive relationships between MAPK signaling and cardiac dysfunction remain elusive, despite 30 years of extensive clinical studies and basic research of various animal/cell models, severities of stress, and types of stimuli. Still, several studies have proven the importance of MAPK crosstalk with mitochondria, powerhouses of the cell that provide over 80% of ATP for normal cardiomyocyte function and play a crucial role in cell death. Although many questions remain unanswered, there exists enough evidence to consider the possibility of targeting MAPK-mitochondria interactions in the prevention and treatment of heart disease. The goal of this review is to integrate previous studies into a discussion of MAPKs and MAPK-mitochondria signaling in cardiac diseases, such as myocardial infarction (ischemia), hypertrophy and heart failure. A comprehensive understanding of relevant molecular mechanisms, as well as challenges for studies in this area, will facilitate the development of new pharmacological agents and genetic manipulations for therapy of cardiovascular diseases.
Collapse
Affiliation(s)
- Sabzali Javadov
- Department of Physiology, School of Medicine, University of Puerto Rico, PR, USA.
| | - Sehwan Jang
- Department of Physiology, School of Medicine, University of Puerto Rico, PR, USA
| | - Bryan Agostini
- Department of Physiology, School of Medicine, University of Puerto Rico, PR, USA
| |
Collapse
|
42
|
McGinley AL, Li Y, Deliu Z, Wang QT. Additional sex combs-likefamily genes are required for normal cardiovascular development. Genesis 2014; 52:671-86. [DOI: 10.1002/dvg.22793] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2014] [Revised: 05/14/2014] [Accepted: 05/20/2014] [Indexed: 01/23/2023]
Affiliation(s)
- Andrea L. McGinley
- Department of Biological Sciences; University of Illinois at Chicago; Chicago Illinois
| | - Yanyang Li
- Department of Biological Sciences; University of Illinois at Chicago; Chicago Illinois
| | - Zane Deliu
- Department of Biological Sciences; University of Illinois at Chicago; Chicago Illinois
| | - Q. Tian Wang
- Department of Biological Sciences; University of Illinois at Chicago; Chicago Illinois
| |
Collapse
|
43
|
Norris AW, Bahr TM, Scholz TD, Peterson ES, Volk KA, Segar JL. Angiotensin II-induced cardiovascular load regulates cardiac remodeling and related gene expression in late-gestation fetal sheep. Pediatr Res 2014; 75:689-696. [PMID: 24614802 PMCID: PMC4251591 DOI: 10.1038/pr.2014.37] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2013] [Accepted: 12/19/2013] [Indexed: 12/21/2022]
Abstract
BACKGROUND Angiotensin II (ANG II) stimulates fetal heart growth, although little is known regarding changes in cardiomyocyte endowment or the molecular pathways mediating the response. We measured cardiomyocyte proliferation and morphology in ANG II-treated fetal sheep and assessed transcriptional pathway responses in ANG II and losartan (an ANG II type 1 receptor antagonist) treated fetuses. METHODS In twin-gestation pregnant sheep, one fetus received ANG II (50 μg/kg/min i.v.) or losartan (20 mg/kg/d i.v.) for 7 d; noninstrumented twins served as controls. RESULTS ANG II produced increases in heart mass, cardiomyocyte area (left ventricle (LV) and right ventricle mononucleated and LV binucleated cells), and the percentage of Ki-67-positive mononucleated cells in the LV (all P < 0.05). ANG II and losartan produced generally opposing changes in gene expression, affecting an estimated 55% of the represented transcriptome. The most prominent significantly affected biological pathways included those involved in cytoskeletal remodeling and cell cycle activity. CONCLUSION ANG II produces an increase in fetal cardiac mass via cardiomyocyte hypertrophy and likely hyperplasia, involving transcriptional responses in cytoskeletal remodeling and cell cycle pathways.
Collapse
Affiliation(s)
- Andrew W. Norris
- Department of Pediatrics, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Timothy M. Bahr
- Department of Pediatrics, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Thomas D. Scholz
- Department of Pediatrics, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Emily S. Peterson
- Department of Pediatrics, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Ken A. Volk
- Department of Pediatrics, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Jeffrey L. Segar
- Department of Pediatrics, University of Iowa Carver College of Medicine, Iowa City, IA, USA,Corresponding Author: Jeffrey L. Segar, MD Professor, Department of Pediatrics University of Iowa Carver College of Medicine University of Iowa Children's Hospital 200 Hawkins Drive, Iowa City, IA 52242 319.356.7244 (phone) 319.356.4685 (facsimile)
| |
Collapse
|
44
|
Kwon YW, Yang HM, Cho HJ. Cell therapy for myocardial infarction. Int J Stem Cells 2014; 3:8-15. [PMID: 24855535 DOI: 10.15283/ijsc.2010.3.1.8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/09/2010] [Indexed: 12/27/2022] Open
Abstract
Ischemic heart disease, particularly acute myocardial infarction (MI), is the worldwide health care problem and the leading cause of morbidity and mortality. The fundamental treatment of MI remains a major unmet medical need. Although recent tremendous advances have been made in the treatment for acute MI such as percutaneous coronary intervention (PCI) and medical and surgical therapies, myocardial cell loss after ischemia and subsequent, adverse cardiac remodeling and heart failure are demanding for new therapeutic strategy. Since the first experimental studies of adult stem cell therapy into the ischemic heart were performed in the early 1990s, the identification and potential application of stem and/or progenitor cells has triggered attempts to regenerate damaged heart tissue and cell-based therapy is a promising option for treatment of MI. In this review, we would like to discuss the pathogenesis of acute MI, current standard treatments and their limitation, clinical results of recent stem or progenitor cell therapy which have shown a favorable safety profile with modest improvement in cardiac function, and putative mechanisms of benefits.
Collapse
Affiliation(s)
- Yoo-Wook Kwon
- Innovative Research Institute for Cell Therapy, Seoul National University Hospital, Seoul, Korea
| | - Han-Mo Yang
- Innovative Research Institute for Cell Therapy, Seoul National University Hospital, Seoul, Korea ; Cardiovascular Center, Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Hyun-Jai Cho
- Innovative Research Institute for Cell Therapy, Seoul National University Hospital, Seoul, Korea ; Cardiovascular Center, Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| |
Collapse
|
45
|
Liu R, Feng HZ, Jin JP. Physiological contractility of cardiomyocytes in the wall of mouse and rat azygos vein. Am J Physiol Cell Physiol 2014; 306:C697-704. [PMID: 24477237 PMCID: PMC3962596 DOI: 10.1152/ajpcell.00004.2014] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Accepted: 01/25/2014] [Indexed: 01/19/2023]
Abstract
We recently demonstrated the abundant presence of cardiomyocytes in the wall of thoracic veins of adult mouse and rat. The highly differentiated morphology and myofilament protein contents of the venous cardiomyocytes suggested contractile functions. Here we further investigated the contractility of mouse and rat azygos venous rings compared with that of atrial strips and ventricular papillary muscle. 5-Bromo-4-chloro-indolyl-galactopyranoside (X-gal) staining of transgenic mouse vessels expressing lacZ under a cloned cardiac troponin T promoter demonstrated that the venous cardiomyocytes are discontinuous from atrial myocardium and aligned in the wall of thoracic veins perpendicular to the vessel axis. Histological sections displayed sarcomeric striations in the venous cardiomyocytes, which indicate an encirclement orientation of myofibrils in the vessel wall. Mechanical studies found that the rings of mouse and rat azygos vein produce strong cardiac type twitch contractions when stimulated with electrical pacing in contrast to the weak and slow smooth muscle contractions induced using 90 mM KCl. The twitch contraction and relaxation of mouse azygos veins further exhibited a cardiac type of β-adrenergic responses. Quantitative comparison showed that the contractions of venous cardiomyocytes are slightly slower than those of atrium muscle but significantly faster than those of ventricular papillary muscle. These novel findings indicate that the cardiomyocytes abundant in the wall of rodent thoracic veins possess fully differentiated cardiac muscle phenotype despite their anatomical and functional segregations from the heart.
Collapse
Affiliation(s)
- Rong Liu
- Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan
| | | | | |
Collapse
|
46
|
Abstract
The last decade has witnessed the publication of a large number of clinical trials primarily using bone marrow-derived stem cells as the injected cell. These "first-generation" clinical trials have advanced our understanding and shown us that (1) cell therapy is safe, (2) cell therapy has been modestly effective, and (3) in humans, bone marrow-derived stem cells do not transdifferentiate into cardiomyocytes or new blood vessels (or at least in sufficient numbers to have any effect). The primary mechanism of action for cell therapy is now believed to be through paracrine effects that include the release of cytokines, chemokines, and growth factors that inhibit apoptosis and fibrosis, enhance contractility, and activate endogenous regenerative mechanisms through endogenous circulating or site-specific stem cells. The new direction for clinical trials includes the use of stem cells capable of cardiac lineage, such as endogenous cardiac stem cells.
Collapse
Affiliation(s)
- Robert E Michler
- Montefiore Medical Center, Albert Einstein College of Medicine, New York, New York
| |
Collapse
|
47
|
Michler R. Surgical options for the management of ischemic cardiomyopathy. CURRENT TREATMENT OPTIONS IN CARDIOVASCULAR MEDICINE 2013; 15:518-32. [PMID: 24018769 DOI: 10.1007/s11936-013-0261-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
OPINION STATEMENT Novel surgical alternatives and the refinement of conventional surgical therapies for the treatment of ischemic cardiomyopathy are in constant evolution. Current approaches involve the determined application of the appropriateness criteria for CABG surgery, the extension of mitral valve repair to complex patients with ischemic cardiomyopathy, finding appropriate patients who might benefit from surgical ventricular reconstruction, and surgical attempts to regenerate lost or damaged myocardium with transplanted stem cells. The refinement of surgical techniques and the medical optimization of candidates for surgery remain a cornerstone of management for patients with complex heart disease like ischemic cardiomyopathy. The horizon is bright for patients suffering from this condition and concentrated research efforts by groups such as the NHLBI-sponsored Cardiothoracic Surgery Network will have a major impact on the future of patients with heart disease.
Collapse
Affiliation(s)
- Robert Michler
- Montefiore Medical Center, Albert Einstein College of Medicine, New York City, NY, USA,
| |
Collapse
|
48
|
Blech-Hermoni Y, Ladd AN. RNA binding proteins in the regulation of heart development. Int J Biochem Cell Biol 2013; 45:2467-78. [PMID: 23973289 DOI: 10.1016/j.biocel.2013.08.008] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Revised: 08/09/2013] [Accepted: 08/13/2013] [Indexed: 11/28/2022]
Abstract
In vivo, RNA molecules are constantly accompanied by RNA binding proteins (RBPs), which are intimately involved in every step of RNA biology, including transcription, editing, splicing, transport and localization, stability, and translation. RBPs therefore have opportunities to shape gene expression at multiple levels. This capacity is particularly important during development, when dynamic chemical and physical changes give rise to complex organs and tissues. This review discusses RBPs in the context of heart development. Since the targets and functions of most RBPs--in the heart and at large--are not fully understood, this review focuses on the expression and roles of RBPs that have been implicated in specific stages of heart development or developmental pathology. RBPs are involved in nearly every stage of cardiogenesis, including the formation, morphogenesis, and maturation of the heart. A fuller understanding of the roles and substrates of these proteins could ultimately provide attractive targets for the design of therapies for congenital heart defects, cardiovascular disease, or cardiac tissue repair.
Collapse
Affiliation(s)
- Yotam Blech-Hermoni
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA; Program in Cell Biology, Department of Molecular Biology and Microbiology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | | |
Collapse
|
49
|
Anversa P, Leri A. Innate regeneration in the aging heart: healing from within. Mayo Clin Proc 2013; 88:871-83. [PMID: 23910414 PMCID: PMC3936323 DOI: 10.1016/j.mayocp.2013.04.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2013] [Revised: 03/29/2013] [Accepted: 04/01/2013] [Indexed: 12/31/2022]
Abstract
The concept of the heart as a terminally differentiated organ incapable of replacing damaged myocytes has been at the center of cardiovascular research and therapeutic development for the past 50 years. The progressive decline in myocyte number as a function of age and the formation of scarred tissue after myocardial infarction have been interpreted as irrefutable proofs of the postmitotic characteristic of the heart. However, emerging evidence supports a more dynamic view of the heart in which cell death and renewal are vital components of the remodeling process that governs cardiac homeostasis, aging, and disease. The identification of dividing myocytes in the adult and senescent heart raises the important question concerning the origin of these newly formed cells. In vitro and in vivo findings strongly suggest that replicating myocytes derive from lineage determination of resident primitive cells, supporting the notion that cardiomyogenesis is controlled by activation and differentiation of a stem cell compartment. It is the current view that the myocardium is an organ permissive of tissue regeneration mediated by exogenous and endogenous progenitor cells.
Collapse
Affiliation(s)
- Piero Anversa
- Department of Anesthesia, Department of Medicine, and Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA.
| | | |
Collapse
|
50
|
Gene expression deregulation in postnatal skeletal muscle of TK2 deficient mice reveals a lower pool of proliferating myogenic progenitor cells. PLoS One 2013; 8:e53698. [PMID: 23341978 PMCID: PMC3544874 DOI: 10.1371/journal.pone.0053698] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2012] [Accepted: 12/03/2012] [Indexed: 01/07/2023] Open
Abstract
Loss of thymidine kinase 2 (TK2) causes a heterogeneous myopathic form of mitochondrial DNA (mtDNA) depletion syndrome (MDS) in humans that predominantly affects skeletal muscle tissue. In mice, TK2 deficiency also affects several tissues in addition to skeletal muscle, including brain, heart, adipose tissue, kidneys and causes death about 3 weeks after birth. We analysed skeletal muscle and heart muscle tissues of Tk2 knockout mice at postnatal development phase and observed that TK2 deficient pups grew slower and their skeletal muscles appeared significantly underdeveloped, whereas heart was close to normal in size. Both tissues showed mtDNA depletion and mitochondria with altered ultrastructure, as revealed by transmission electron microscopy. Gene expression microarray analysis showed a strong down-regulation of genes involved in cell cycle and cell proliferation in both tissues, suggesting a lower pool of undifferentiated proliferating cells. Analysis of isolated primary myoblasts from Tk2 knockout mice showed slow proliferation, less ability to differentiate and signs of premature senescence, even in absence of mtDNA depletion. Our data demonstrate that TK2 deficiency disturbs myogenic progenitor cells function in postnatal skeletal muscle and we propose this as one of the causes of underdeveloped phenotype and myopathic characteristic of the TK2 deficient mice, in addition to the progressive mtDNA depletion, mitochondrial damage and respiratory chain deficiency in post-mitotic differentiated tissue.
Collapse
|