1
|
Farboud SP, Fathi E, Valipour B, Farahzadi R. Toward the latest advancements in cardiac regeneration using induced pluripotent stem cells (iPSCs) technology: approaches and challenges. J Transl Med 2024; 22:783. [PMID: 39175068 PMCID: PMC11342568 DOI: 10.1186/s12967-024-05499-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 07/10/2024] [Indexed: 08/24/2024] Open
Abstract
A novel approach to treating heart failures was developed with the introduction of iPSC technology. Knowledge in regenerative medicine, developmental biology, and the identification of illnesses at the cellular level has exploded since the discovery of iPSCs. One of the most frequent causes of mortality associated with cardiovascular disease is the loss of cardiomyocytes (CMs), followed by heart failure. A possible treatment for heart failure involves restoring cardiac function and replacing damaged tissue with healthy, regenerated CMs. Significant strides in stem cell biology during the last ten years have transformed the in vitro study of human illness and enhanced our knowledge of the molecular pathways underlying human disease, regenerative medicine, and drug development. We seek to examine iPSC advancements in disease modeling, drug discovery, iPSC-Based cell treatments, and purification methods in this article.
Collapse
Affiliation(s)
- Seyedeh Parya Farboud
- Department of Clinical Sciences, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Ezzatollah Fathi
- Department of Clinical Sciences, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran.
| | - Behnaz Valipour
- Department of Anatomical Sciences, Sarab Faculty of Medical Sciences, Sarab, Iran
- Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Raheleh Farahzadi
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
2
|
Pardon G, Vander Roest AS, Chirikian O, Birnbaum F, Lewis H, Castillo EA, Wilson R, Denisin AK, Blair CA, Holbrook C, Koleckar K, Chang ACY, Blau HM, Pruitt BL. Tracking single hiPSC-derived cardiomyocyte contractile function using CONTRAX an efficient pipeline for traction force measurement. Nat Commun 2024; 15:5427. [PMID: 38926342 PMCID: PMC11208611 DOI: 10.1038/s41467-024-49755-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 06/18/2024] [Indexed: 06/28/2024] Open
Abstract
Cardiomyocytes derived from human induced pluripotent stem cells (hiPSC-CMs) are powerful in vitro models to study the mechanisms underlying cardiomyopathies and cardiotoxicity. Quantification of the contractile function in single hiPSC-CMs at high-throughput and over time is essential to disentangle how cellular mechanisms affect heart function. Here, we present CONTRAX, an open-access, versatile, and streamlined pipeline for quantitative tracking of the contractile dynamics of single hiPSC-CMs over time. Three software modules enable: parameter-based identification of single hiPSC-CMs; automated video acquisition of >200 cells/hour; and contractility measurements via traction force microscopy. We analyze >4,500 hiPSC-CMs over time in the same cells under orthogonal conditions of culture media and substrate stiffnesses; +/- drug treatment; +/- cardiac mutations. Using undirected clustering, we reveal converging maturation patterns, quantifiable drug response to Mavacamten and significant deficiencies in hiPSC-CMs with disease mutations. CONTRAX empowers researchers with a potent quantitative approach to develop cardiac therapies.
Collapse
Grants
- K99 HL153679 NHLBI NIH HHS
- RM1 GM131981 NIGMS NIH HHS
- 20POST35211011 American Heart Association (American Heart Association, Inc.)
- 17CSA33590101 American Heart Association (American Heart Association, Inc.)
- 18CDA34110411 American Heart Association (American Heart Association, Inc.)
- 1R21HL13099301 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- 18POST34080160 American Heart Association (American Heart Association, Inc.)
- 1F31HL158227 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- F31 HL158227 NHLBI NIH HHS
- 201411MFE-338745-169197 Gouvernement du Canada | Canadian Institutes of Health Research (Instituts de Recherche en Santé du Canada)
- P2SKP2_164954 Schweizerischer Nationalfonds zur Förderung der Wissenschaftlichen Forschung (Swiss National Science Foundation)
- 13POST14480004 American Heart Association (American Heart Association, Inc.)
- RM1GM131981 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- 82070248 National Natural Science Foundation of China (National Science Foundation of China)
- P400PM_180825 Schweizerischer Nationalfonds zur Förderung der Wissenschaftlichen Forschung (Swiss National Science Foundation)
- U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- Shanghai Pujiang Program 19PJ1407000 Program for Professor of Special Appointment (Eastern Scholar) at Shanghai Institutions of Higher Learning 0900000024 to A.C.Y.C. Innovative Research Team of High-Level Local Universities in Shanghai (A.C.Y.C.)
- the Baxter Foundation, Li Ka Shing Foundation and The Stanford Cardiovascular Institute
Collapse
Affiliation(s)
- Gaspard Pardon
- Departments of Mechanical Engineering and of Bioengineering, Stanford University, School of Engineering and School of Medicine, Stanford, CA, USA
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Departments of Bioengineering and Mechanical Engineering, University of California, Santa Barbara, CA, USA
- School of Life Sciences, EPFL École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Alison S Vander Roest
- Departments of Mechanical Engineering and of Bioengineering, Stanford University, School of Engineering and School of Medicine, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pediatrics (Cardiology), Stanford University School of Medicine, Stanford, CA, USA
- Department of Biomedical Engineering, Michigan Engineering, University of Michigan Ann Arbor, MI, USA
| | - Orlando Chirikian
- Biomolecular Science and Engineering Program, University of California, Santa Barbara, CA, USA
| | - Foster Birnbaum
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Henry Lewis
- Departments of Mechanical Engineering and of Bioengineering, Stanford University, School of Engineering and School of Medicine, Stanford, CA, USA
| | - Erica A Castillo
- Departments of Mechanical Engineering and of Bioengineering, Stanford University, School of Engineering and School of Medicine, Stanford, CA, USA
- Departments of Bioengineering and Mechanical Engineering, University of California, Santa Barbara, CA, USA
| | - Robin Wilson
- Departments of Mechanical Engineering and of Bioengineering, Stanford University, School of Engineering and School of Medicine, Stanford, CA, USA
| | - Aleksandra K Denisin
- Departments of Mechanical Engineering and of Bioengineering, Stanford University, School of Engineering and School of Medicine, Stanford, CA, USA
| | - Cheavar A Blair
- Departments of Mechanical Engineering and of Bioengineering, Stanford University, School of Engineering and School of Medicine, Stanford, CA, USA
- Departments of Bioengineering and Mechanical Engineering, University of California, Santa Barbara, CA, USA
- Department of Physiology, University of Kentucky, Lexington, KY, USA
| | - Colin Holbrook
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Kassie Koleckar
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Alex C Y Chang
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Shanghai Institute of Precision Medicine and Department of Cardiology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, China
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Helen M Blau
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Beth L Pruitt
- Departments of Mechanical Engineering and of Bioengineering, Stanford University, School of Engineering and School of Medicine, Stanford, CA, USA.
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA.
- Departments of Bioengineering and Mechanical Engineering, University of California, Santa Barbara, CA, USA.
- Biomolecular Science and Engineering Program, University of California, Santa Barbara, CA, USA.
| |
Collapse
|
3
|
Salaun E, Drory S, Coté M, Tremblay V, Bédard E, Steinberg C, Paré D, O'Connor K, Cieza T, Coté N, Poirier P, Douville P, Blais J, Desmeules P, Kalavrouziotis D, Mohammadi S, Voisine P, Bernier M, Pibarot P, Thériault S. Role of Antitroponin Antibodies and Macrotroponin in the Clinical Interpretation of Cardiac Troponin. J Am Heart Assoc 2024; 13:e035128. [PMID: 38879450 PMCID: PMC11255741 DOI: 10.1161/jaha.123.035128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/19/2024]
Abstract
Cardiac troponin is extensively used as a biomarker in modern medicine due to its diagnostic capability for myocardial injury, as well as its predictive and prognostic value for cardiac diseases. However, heterophile antibodies, antitroponin antibodies, and macrotroponin complexes can be observed both in seemingly healthy individuals and patients with cardiac diseases, potentially leading to false positive or disproportionate elevation of cTn (cardiac troponin) assay results and introducing discrepancies in clinical interpretations with impact on medical management. In this review article, we describe the possible mechanisms of cTn release and the sources of variations in the assessment of circulating cTn levels. We also explore the pathophysiological mechanisms underlying antitroponin antibody development and discuss the influence exerted by macrotroponin complexes on the results of immunoassays. Additionally, we explore approaches to detect these complexes by presenting various clinical scenarios encountered in routine clinical practice. Finally, unsolved questions about the development, prevalence, and clinical significance of cardiac autoantibodies are discussed.
Collapse
Affiliation(s)
- Erwan Salaun
- Institut Universitaire de Cardiologie et de Pneumologie de QuébecUniversité LavalQuébecCanada
| | - Samuel Drory
- Institut Universitaire de Cardiologie et de Pneumologie de QuébecUniversité LavalQuébecCanada
| | - Marc‐André Coté
- Institut Universitaire de Cardiologie et de Pneumologie de QuébecUniversité LavalQuébecCanada
| | - Veronic Tremblay
- Institut Universitaire de Cardiologie et de Pneumologie de QuébecUniversité LavalQuébecCanada
| | - Elisabeth Bédard
- Institut Universitaire de Cardiologie et de Pneumologie de QuébecUniversité LavalQuébecCanada
| | - Christian Steinberg
- Institut Universitaire de Cardiologie et de Pneumologie de QuébecUniversité LavalQuébecCanada
| | - David Paré
- Institut Universitaire de Cardiologie et de Pneumologie de QuébecUniversité LavalQuébecCanada
| | - Kim O'Connor
- Institut Universitaire de Cardiologie et de Pneumologie de QuébecUniversité LavalQuébecCanada
| | - Tomas Cieza
- Institut Universitaire de Cardiologie et de Pneumologie de QuébecUniversité LavalQuébecCanada
| | - Nancy Coté
- Institut Universitaire de Cardiologie et de Pneumologie de QuébecUniversité LavalQuébecCanada
| | - Paul Poirier
- Institut Universitaire de Cardiologie et de Pneumologie de QuébecUniversité LavalQuébecCanada
- Faculty of pharmacyUniversité LavalQuébecCanada
| | - Pierre Douville
- Centre Hospitalier Universitaire de QuébecUniversité LavalQuébecCanada
| | - Jonatan Blais
- Centre Hospitalier Universitaire de QuébecUniversité LavalQuébecCanada
| | - Philippe Desmeules
- Institut Universitaire de Cardiologie et de Pneumologie de QuébecUniversité LavalQuébecCanada
- Centre Hospitalier Universitaire de QuébecUniversité LavalQuébecCanada
| | - Dimitris Kalavrouziotis
- Institut Universitaire de Cardiologie et de Pneumologie de QuébecUniversité LavalQuébecCanada
| | - Siamak Mohammadi
- Institut Universitaire de Cardiologie et de Pneumologie de QuébecUniversité LavalQuébecCanada
| | - Pierre Voisine
- Institut Universitaire de Cardiologie et de Pneumologie de QuébecUniversité LavalQuébecCanada
- Division of Cardiac SurgeryUniversity of Ottawa Heart InstituteOttawaOntarioCanada
| | - Mathieu Bernier
- Institut Universitaire de Cardiologie et de Pneumologie de QuébecUniversité LavalQuébecCanada
| | - Philippe Pibarot
- Institut Universitaire de Cardiologie et de Pneumologie de QuébecUniversité LavalQuébecCanada
| | - Sébastien Thériault
- Institut Universitaire de Cardiologie et de Pneumologie de QuébecUniversité LavalQuébecCanada
| |
Collapse
|
4
|
Chirikian O, Faynus MA, Merk M, Singh Z, Muray C, Pham J, Chialastri A, Vander Roest A, Goldstein A, Pyle T, Lane KV, Roberts B, Smith JE, Gunawardane RN, Sniadecki NJ, Mack DL, Davis J, Bernstein D, Streichan SJ, Clegg DO, Dey SS, Wilson MZ, Pruitt BL. YAP dysregulation triggers hypertrophy by CCN2 secretion and TGFβ uptake in human pluripotent stem cell-derived cardiomyocytes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.03.597045. [PMID: 38895282 PMCID: PMC11185505 DOI: 10.1101/2024.06.03.597045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Hypertrophy Cardiomyopathy (HCM) is the most prevalent hereditary cardiovascular disease - affecting >1:500 individuals. Advanced forms of HCM clinically present with hypercontractility, hypertrophy and fibrosis. Several single-point mutations in b-myosin heavy chain (MYH7) have been associated with HCM and increased contractility at the organ level. Different MYH7 mutations have resulted in increased, decreased, or unchanged force production at the molecular level. Yet, how these molecular kinetics link to cell and tissue pathogenesis remains unclear. The Hippo Pathway, specifically its effector molecule YAP, has been demonstrated to be reactivated in pathological hypertrophic growth. We hypothesized that changes in force production (intrinsically or extrinsically) directly alter the homeostatic mechano-signaling of the Hippo pathway through changes in stresses on the nucleus. Using human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs), we asked whether homeostatic mechanical signaling through the canonical growth regulator, YAP, is altered 1) by changes in the biomechanics of HCM mutant cardiomyocytes and 2) by alterations in the mechanical environment. We use genetically edited hiPSC-CM with point mutations in MYH7 associated with HCM, and their matched controls, combined with micropatterned traction force microscopy substrates to confirm the hypercontractile phenotype in MYH7 mutants. We next modulate contractility in healthy and disease hiPSC-CMs by treatment with positive and negative inotropic drugs and demonstrate a correlative relationship between contractility and YAP activity. We further demonstrate the activation of YAP in both HCM mutants and healthy hiPSC-CMs treated with contractility modulators is through enhanced nuclear deformation. We conclude that the overactivation of YAP, possibly initiated and driven by hypercontractility, correlates with excessive CCN2 secretion (connective tissue growth factor), enhancing cardiac fibroblast/myofibroblast transition and production of known hypertrophic signaling molecule TGFβ. Our study suggests YAP being an indirect player in the initiation of hypertrophic growth and fibrosis in HCM. Our results provide new insights into HCM progression and bring forth a testbed for therapeutic options in treating HCM.
Collapse
|
5
|
Yao J, Chen Y, Huang Y, Sun X, Shi X. The role of cardiac microenvironment in cardiovascular diseases: implications for therapy. Hum Cell 2024; 37:607-624. [PMID: 38498133 DOI: 10.1007/s13577-024-01052-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 02/25/2024] [Indexed: 03/20/2024]
Abstract
Due to aging populations and changes in lifestyle, cardiovascular diseases including cardiomyopathy, hypertension, and atherosclerosis, are the leading causes of death worldwide. The heart is a complicated organ composed of multicellular types, including cardiomyocytes, fibroblasts, endothelial cells, vascular smooth muscle cells, and immune cells. Cellular specialization and complex interplay between different cell types are crucial for the cardiac tissue homeostasis and coordinated function of the heart. Mounting studies have demonstrated that dysfunctional cells and disordered cardiac microenvironment are closely associated with the pathogenesis of various cardiovascular diseases. In this paper, we discuss the composition and the homeostasis of cardiac tissues, and focus on the role of cardiac environment and underlying molecular mechanisms in various cardiovascular diseases. Besides, we elucidate the novel treatment for cardiovascular diseases, including stem cell therapy and targeted therapy. Clarification of these issues may provide novel insights into the prevention and potential targets for cardiovascular diseases.
Collapse
Affiliation(s)
- Jiayu Yao
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China
| | - Yuejun Chen
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China
| | - Yuqing Huang
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China
| | - Xiaoou Sun
- Institute of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, China.
| | - Xingjuan Shi
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China.
| |
Collapse
|
6
|
Neumann S, Siegert S. Investigation of α-Klotho Concentrations in Serum of Cats Affected by Hypertrophic Cardiomyopathy. Vet Sci 2024; 11:184. [PMID: 38787156 PMCID: PMC11125955 DOI: 10.3390/vetsci11050184] [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: 03/22/2024] [Revised: 04/11/2024] [Accepted: 04/19/2024] [Indexed: 05/25/2024] Open
Abstract
Being involved in various physiological and pathophysiological mechanisms (ageing, kidney damage, cardiovascular diseases, etc.), Klotho is a parameter of increasing interest. Studies in veterinary medicine are still rare, but it is exciting to find out whether the findings obtained can be transferred to animals. The aim of this study was therefore to investigate Klotho in cats. This study addressed α-Klotho concentrations in the serum of two groups of cats: one diseased group affected by hypertrophic cardiomyopathy (n = 27) and one healthy control group (n = 35). α-Klotho concentrations in serum were measured using an ELISA. The results were evaluated in the context of several echocardiographic measurement parameters in the diseased group. No significant difference between α-Klotho concentrations in the two groups was found. A slight negative correlation was found between α-Klotho concentrations and the relation of left atrium/aorta (La/Ao) in the diseased group. Gaining initial information on α-Klotho in cats, it was not possible to draw definite conclusions concerning cardiomyopathies in this species. The assessment of Klotho should be considered in terms of its broad implications in disease processes, but it is also recommended to focus on specific disease features. Both approaches might be promising as possible applications of Klotho in veterinary medicine.
Collapse
Affiliation(s)
- Stephan Neumann
- Institute of Veterinary Medicine, Georg-August-University of Goettingen, Burckhardtweg 2, 37077 Goettingen, Germany;
| | | |
Collapse
|
7
|
Kelly CM, Martin JL, Previs MJ. Myosin folding boosts solubility in cardiac muscle sarcomeres. JCI Insight 2024; 9:e178131. [PMID: 38483507 PMCID: PMC11141871 DOI: 10.1172/jci.insight.178131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 03/05/2024] [Indexed: 04/23/2024] Open
Abstract
The polymerization of myosin molecules into thick filaments in muscle sarcomeres is essential for cardiac contractility, with the attenuation of interactions between the heads of myosin molecules within the filaments being proposed to result in hypercontractility, as observed in hypertrophic cardiomyopathy (HCM). However, experimental evidence demonstrates that the structure of these giant macromolecular complexes is highly dynamic, with molecules exchanging between the filaments and a pool of soluble molecules on the minute timescale. Therefore, we sought to test the hypothesis that the enhancement of interactions between the heads of myosin molecules within thick filaments limits the mobility of myosin by taking advantage of mavacamten, a small molecule approved for the treatment of HCM. Myosin molecules were labeled in vivo with a green fluorescent protein (GFP) and imaged in intact hearts using multiphoton microscopy. Treatment of the intact hearts with mavacamten resulted in an unexpected > 5-fold enhancement in GFP-myosin mobility within the sarcomere. In vitro biochemical assays suggested that mavacamten enhanced the mobility of GFP-myosin by increasing the solubility of myosin molecules, through the stabilization of a compact/folded conformation of the molecules, once disassociated from the thick filaments. These findings provide alternative insight into the mechanisms by which molecules exchange into and out of thick filaments and have implications for how mavacamten may affect cardiac contractility.
Collapse
Affiliation(s)
- Colleen M Kelly
- Molecular Physiology and Biophysics Department, Larner College of Medicine, University of Vermont, Burlington, Vermont, USA
| | - Jody L Martin
- Department of Pharmacology, University of California, Davis, Davis, California, USA
| | - Michael J Previs
- Molecular Physiology and Biophysics Department, Larner College of Medicine, University of Vermont, Burlington, Vermont, USA
| |
Collapse
|
8
|
Masarone D, Kittleson MM, D'Onofrio A, Falco L, Fumarulo I, Massetti M, Crea F, Aspromonte N, Pacileo G. Basic science of cardiac contractility modulation therapy: Molecular and electrophysiological mechanisms. Heart Rhythm 2024; 21:82-88. [PMID: 37769793 DOI: 10.1016/j.hrthm.2023.09.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 09/21/2023] [Accepted: 09/21/2023] [Indexed: 10/03/2023]
Abstract
In heart failure with reduced ejection fraction and heart failure with preserved ejection fraction, profound cellular and molecular changes have recently been documented in the failing myocardium. These changes include altered calcium handling and metabolic efficiency of the cardiac myocyte, reactivation of the fetal gene program, changes in the electrophysiological properties of the heart, and accumulation of collagen (fibrosis) at the interstitial level. Cardiac contractility modulation therapy is an innovative device-based therapy currently approved for heart failure with reduced ejection fraction in patients with narrow QRS complex and under investigation for the treatment of heart failure with preserved ejection fraction. This therapy is based on the delivery of high-voltage biphasic electrical signals to the septal wall of the right ventricle during the absolute refractory period of the myocardium. At the cellular level, in patients with heart failure with reduced ejection fraction, cardiac contractility modulation therapy has been shown to restore calcium handling and improve the metabolic status of cardiac myocytes, reverse the heart failure-associated fetal gene program, and reduce the extent of interstitial fibrosis. This review summarizes the preclinical literature on the use of cardiac contractility modulation therapy in heart failure with reduced and preserved ejection fraction, correlating the molecular and electrophysiological effects with the clinical benefits demonstrated by this therapy.
Collapse
Affiliation(s)
- Daniele Masarone
- Heart Failure Unit, Department of Cardiology, AORN dei Colli/Monaldi Hospital, Naples, Italy.
| | - Michelle M Kittleson
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Antonio D'Onofrio
- Electrophysiology Unit, Department of Cardiology, AORN dei Colli/Monaldi Hospital, Naples, Italy
| | - Luigi Falco
- Heart Failure Unit, Department of Cardiology, AORN dei Colli/Monaldi Hospital, Naples, Italy
| | | | - Massimo Massetti
- Catholic University of the Sacred Heart, Rome, Italy; Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Filippo Crea
- Catholic University of the Sacred Heart, Rome, Italy; Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Nadia Aspromonte
- Catholic University of the Sacred Heart, Rome, Italy; Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Giuseppe Pacileo
- Heart Failure Unit, Department of Cardiology, AORN dei Colli/Monaldi Hospital, Naples, Italy
| |
Collapse
|
9
|
Guo G, Wang L, Li X, Fu W, Cao J, Zhang J, Liu Y, Liu M, Wang M, Zhao G, Zhao X, Zhou Y, Niu S, Liu G, Zhang Y, Dong J, Tao H, Zhao X. Enhanced myofilament calcium sensitivity aggravates abnormal calcium handling and diastolic dysfunction in patient-specific induced pluripotent stem cell-derived cardiomyocytes with MYH7 mutation. Cell Calcium 2024; 117:102822. [PMID: 38101154 DOI: 10.1016/j.ceca.2023.102822] [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: 06/05/2023] [Revised: 10/04/2023] [Accepted: 11/06/2023] [Indexed: 12/17/2023]
Abstract
Hypertrophic cardiomyopathy (HCM), the most common inherited heart disease, is frequently caused by mutations in the β-cardiac myosin heavy chain gene (MYH7). Abnormal calcium handling and diastolic dysfunction are archetypical features of HCM caused by MYH7 gene mutations. However, the mechanism of how MYH7 mutations leads to these features remains unclear, which inhibits the development of effective therapies. Initially, cardiomyocytes were generated from induced pluripotent stem cells from an eight-year-old girl diagnosed with HCM carrying a MYH7(C.1063 G>A) heterozygous mutation(mutant-iPSC-CMs) and mutation-corrected isogenic iPSCs(control-iPSC-CMs) in the present study. Next, we compared phenotype of mutant-iPSC-CMs to that of control-iPSC-CMs, by assessing their morphology, hypertrophy-related genes expression, calcium handling, diastolic function and myofilament calcium sensitivity at days 15 and 40 respectively. Finally, to better understand increased myofilament Ca2+ sensitivity as a central mechanism of central pathogenicity in HCM, inhibition of calcium sensitivity with mavacamten can improveed cardiomyocyte hypertrophy. Mutant-iPSC-CMs exhibited enlarged areas, increased sarcomere disarray, enhanced expression of hypertrophy-related genes proteins, abnormal calcium handling, diastolic dysfunction and increased myofilament calcium sensitivity at day 40, but only significant increase in calcium sensitivity and mild diastolic dysfunction at day 15. Increased calcium sensitivity by levosimendan aggravates cardiomyocyte hypertrophy phenotypes such as expression of hypertrophy-related genes, abnormal calcium handling and diastolic dysfunction, while inhibition of calcium sensitivity significantly improves cardiomyocyte hypertrophy phenotypes in mutant-iPSC-CMs, suggesting increased myofilament calcium sensitivity is the primary mechanisms for MYH7 mutations pathogenesis. Our studies have uncovered a pathogenic mechanism of HCM caused by MYH7 gene mutations through which enhanced myofilament calcium sensitivity aggravates abnormal calcium handling and diastolic dysfunction. Correction of the myofilament calcium sensitivity was found to be an effective method for treating the development of HCM phenotype in vitro.
Collapse
Affiliation(s)
- Guangli Guo
- Department of Cardiology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China
| | - Lu Wang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Xiaowei Li
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China; Henan Key Laboratory of Hereditary Cardiovascular Diseases, Zhengzhou, 450052, China
| | - Wanrong Fu
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Jinhua Cao
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Jianchao Zhang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Yangyang Liu
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China; Henan Key Laboratory of Hereditary Cardiovascular Diseases, Zhengzhou, 450052, China
| | - Mengduan Liu
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China; Henan Key Laboratory of Hereditary Cardiovascular Diseases, Zhengzhou, 450052, China
| | - Mengyu Wang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China; Henan Key Laboratory of Hereditary Cardiovascular Diseases, Zhengzhou, 450052, China
| | - Guojun Zhao
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Xi Zhao
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China; Henan Key Laboratory of Hereditary Cardiovascular Diseases, Zhengzhou, 450052, China
| | - Yangfan Zhou
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China; Henan Key Laboratory of Hereditary Cardiovascular Diseases, Zhengzhou, 450052, China
| | - Shaohui Niu
- Department of Cardiology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China
| | - Gangqiong Liu
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Yanzhou Zhang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Jianzeng Dong
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China; Henan Key Laboratory of Hereditary Cardiovascular Diseases, Zhengzhou, 450052, China; Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, National Clinical Research Centre for Cardiovascular Diseases, No. 2 Beijing Anzhen Road, Chaoyang District, Beijing 100029, China.
| | - Hailong Tao
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| | - Xiaoyan Zhao
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China; Henan Key Laboratory of Hereditary Cardiovascular Diseases, Zhengzhou, 450052, China.
| |
Collapse
|
10
|
Mhanna M, Singhal A, Panos A, Firchau DJ, Mansour S, Isom N, Briasoulis A, Duque ER. Cardiogenic Shock in Non-Ischemic Cardiomyopathy: Dynamic Mechanical Circulatory Support and Pathophysiology Illustration. Curr Probl Cardiol 2024; 49:102126. [PMID: 37802166 DOI: 10.1016/j.cpcardiol.2023.102126] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Accepted: 09/30/2023] [Indexed: 10/08/2023]
Abstract
Nonischemic cardiomyopathy (NICM) is a significant cause of cardiogenic shock (CS). We present a case of a 56-year-old previously healthy man who arrived with vague abdominal symptoms, over 2 weeks. Subsequently, the patient's condition rapidly deteriorated over 12 hours, leading to cardiogenic shock categorized as Society for Cardiovascular Angiography and Interventions (SCAI) stage D. Echocardiography and right heart catheterization confirmed multiorgan failure secondary to severe cardiac dysfunction. Mechanical circulatory support was initiated using an Impella CP device 20 hours after admission due to ongoing deterioration. Considering refractory cardiogenic shock and within 24 hours, the patient received combined veno-arterial extracorporeal membrane oxygenation (VA-ECMO) and Impella CP support (ECPElla). With gradual improvement in the patient's clinical status and organ function, successful weaning from VA ECMO to Impella 5.5 was achieved. Ultimately, the patient underwent a successful orthotopic heart and kidney transplantation, marking a significant milestone in his recovery. The case underscores the importance of promptly identifying and responding to cardiogenic shock through invasive hemodynamic assessment. Collaborative decision-making involving a multidisciplinary team played a crucial role in the initiation, escalation, and eventual weaning of mechanical circulatory support, culminating in the successful bridging to a dual organ transplantation for this patient with CS secondary to NICM.
Collapse
Affiliation(s)
- Mohammed Mhanna
- Division of Cardiology, Department of Medicine, University of Iowa, Iowa City, IA.
| | - Arun Singhal
- Division of Cardiothoracic Surgery, Department of Surgery, University of Iowa, Iowa City, IA
| | - Anthony Panos
- Division of Cardiothoracic Surgery, Department of Surgery, University of Iowa, Iowa City, IA
| | - Dennis J Firchau
- Division of Pathology, Department of Medicine, University of Iowa, Iowa City, IA
| | - Shareef Mansour
- Division of Cardiology, Department of Medicine, University of Iowa, Iowa City, IA
| | - Nicholas Isom
- Division of Cardiology, Department of Medicine, University of Iowa, Iowa City, IA
| | | | - Ernesto Ruiz Duque
- Division of Cardiology, Department of Medicine, University of Iowa, Iowa City, IA.
| |
Collapse
|
11
|
Orgil BO, Purevjav E. Molecular Pathways and Animal Models of Cardiomyopathies. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1441:991-1019. [PMID: 38884766 DOI: 10.1007/978-3-031-44087-8_64] [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
Cardiomyopathies are a heterogeneous group of disorders of the heart muscle that ultimately result in congestive heart failure. Rapid progress in genetics, molecular and cellular biology with breakthrough innovative genetic-engineering techniques, such as next-generation sequencing and multiomics platforms, stem cell reprogramming, as well as novel groundbreaking gene-editing systems over the past 25 years has greatly improved the understanding of pathogenic signaling pathways in inherited cardiomyopathies. This chapter will focus on intracellular and intercellular molecular signaling pathways that are activated by a genetic insult in cardiomyocytes to maintain tissue and organ level regulation and resultant cardiac remodeling in certain forms of cardiomyopathies. In addition, animal models of different clinical forms of human cardiomyopathies with their summaries of triggered key molecules and signaling pathways will be described.
Collapse
Affiliation(s)
- Buyan-Ochir Orgil
- Department of Pediatrics, The Heart Institute, Division of Cardiology, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Enkhsaikhan Purevjav
- Department of Pediatrics, The Heart Institute, Division of Cardiology, University of Tennessee Health Science Center, Memphis, TN, USA.
| |
Collapse
|
12
|
Ahmed DW, Eiken MK, DePalma SJ, Helms AS, Zemans RL, Spence JR, Baker BM, Loebel C. Integrating mechanical cues with engineered platforms to explore cardiopulmonary development and disease. iScience 2023; 26:108472. [PMID: 38077130 PMCID: PMC10698280 DOI: 10.1016/j.isci.2023.108472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/12/2024] Open
Abstract
Mechanical forces provide critical biological signals to cells during healthy and aberrant organ development as well as during disease processes in adults. Within the cardiopulmonary system, mechanical forces, such as shear, compressive, and tensile forces, act across various length scales, and dysregulated forces are often a leading cause of disease initiation and progression such as in bronchopulmonary dysplasia and cardiomyopathies. Engineered in vitro models have supported studies of mechanical forces in a number of tissue and disease-specific contexts, thus enabling new mechanistic insights into cardiopulmonary development and disease. This review first provides fundamental examples where mechanical forces operate at multiple length scales to ensure precise lung and heart function. Next, we survey recent engineering platforms and tools that have provided new means to probe and modulate mechanical forces across in vitro and in vivo settings. Finally, the potential for interdisciplinary collaborations to inform novel therapeutic approaches for a number of cardiopulmonary diseases are discussed.
Collapse
Affiliation(s)
- Donia W. Ahmed
- Department of Biomedical Engineering, University of Michigan, Lurie Biomedical Engineering Building, 1101 Beal Avenue, Ann Arbor, MI 48109, USA
| | - Madeline K. Eiken
- Department of Biomedical Engineering, University of Michigan, Lurie Biomedical Engineering Building, 1101 Beal Avenue, Ann Arbor, MI 48109, USA
| | - Samuel J. DePalma
- Department of Biomedical Engineering, University of Michigan, Lurie Biomedical Engineering Building, 1101 Beal Avenue, Ann Arbor, MI 48109, USA
| | - Adam S. Helms
- Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Rachel L. Zemans
- Department of Internal Medicine, Division of Pulmonary Sciences and Critical Care Medicine – Gastroenterology, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI 48109, USA
| | - Jason R. Spence
- Department of Internal Medicine – Gastroenterology, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI 48109, USA
| | - Brendon M. Baker
- Department of Biomedical Engineering, University of Michigan, Lurie Biomedical Engineering Building, 1101 Beal Avenue, Ann Arbor, MI 48109, USA
| | - Claudia Loebel
- Department of Biomedical Engineering, University of Michigan, Lurie Biomedical Engineering Building, 1101 Beal Avenue, Ann Arbor, MI 48109, USA
- Department of Materials Science & Engineering, University of Michigan, North Campus Research Complex, 2800 Plymouth Road, Ann Arbor, MI 48109, USA
| |
Collapse
|
13
|
Osten F, Weber N, Wendland M, Holler T, Piep B, Kröhn S, Teske J, Bodenschatz AK, Devadas SB, Menge KS, Chatterjee S, Schwanke K, Kosanke M, Montag J, Thum T, Zweigerdt R, Kraft T, Iorga B, Meissner JD. Myosin expression and contractile function are altered by replating stem cell-derived cardiomyocytes. J Gen Physiol 2023; 155:e202313377. [PMID: 37656049 PMCID: PMC10473967 DOI: 10.1085/jgp.202313377] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 06/16/2023] [Accepted: 07/19/2023] [Indexed: 09/02/2023] Open
Abstract
Myosin heavy chain (MyHC) is the main determinant of contractile function. Human ventricular cardiomyocytes (CMs) predominantly express the β-isoform. We previously demonstrated that ∼80% of human embryonic stem cell-derived cardiomyocytes (hESC-CMs) express exclusively β-MyHC after long-term culture on laminin-coated glass coverslips. Here, we investigated the impact of enzymatically detaching hESC-CMs after long-term culture and subsequently replating them for characterization of cellular function. We observed that force-related kinetic parameters, as measured in a micromechanical setup, resembled α- rather than β-MyHC-expressing myofibrils, as well as changes in calcium transients. Single-cell immunofluorescence analysis revealed that replating hESC-CMs led to rapid upregulation of α-MyHC, as indicated by increases in exclusively α-MyHC- and in mixed α/β-MyHC-expressing hESC-CMs. A comparable increase in heterogeneity of MyHC isoform expression was also found among individual human induced pluripotent stem cell (hiPSC)-derived CMs after replating. Changes in MyHC isoform expression and cardiomyocyte function induced by replating were reversible in the course of the second week after replating. Gene enrichment analysis based on RNA-sequencing data revealed changes in the expression profile of mechanosensation/-transduction-related genes and pathways, especially integrin-associated signaling. Accordingly, the integrin downstream mediator focal adhesion kinase (FAK) promoted β-MyHC expression on a stiff matrix, further validating gene enrichment analysis. To conclude, detachment and replating induced substantial changes in gene expression, MyHC isoform composition, and function of long-term cultivated human stem cell-derived CMs, thus inducing alterations in mechanosensation/-transduction, that need to be considered, particularly for downstream in vitro assays.
Collapse
Affiliation(s)
- Felix Osten
- Institute of Molecular and Cell Physiology, Hannover Medical School, Hannover, Germany
| | - Natalie Weber
- Institute of Molecular and Cell Physiology, Hannover Medical School, Hannover, Germany
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| | - Meike Wendland
- Institute of Molecular and Cell Physiology, Hannover Medical School, Hannover, Germany
| | - Tim Holler
- Institute of Molecular and Cell Physiology, Hannover Medical School, Hannover, Germany
| | - Birgit Piep
- Institute of Molecular and Cell Physiology, Hannover Medical School, Hannover, Germany
| | - Simon Kröhn
- Institute of Molecular and Cell Physiology, Hannover Medical School, Hannover, Germany
| | - Jana Teske
- Department of Cardiothoracic, Transplantation and Vascular Surgery, Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Hannover Medical School, Hannover, Germany
| | - Alea K. Bodenschatz
- Institute of Molecular and Cell Physiology, Hannover Medical School, Hannover, Germany
| | - Santoshi Biswanath Devadas
- Department of Cardiothoracic, Transplantation and Vascular Surgery, Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Hannover Medical School, Hannover, Germany
| | - Kaja S. Menge
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| | - Shambhabi Chatterjee
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| | - Kristin Schwanke
- Department of Cardiothoracic, Transplantation and Vascular Surgery, Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Hannover Medical School, Hannover, Germany
| | - Maike Kosanke
- Research Core Unit Genomics, Hannover Medical School, Hannover, Germany
| | - Judith Montag
- Institute of Molecular and Cell Physiology, Hannover Medical School, Hannover, Germany
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
- REBIRTH Center for Translational Regenerative Therapies, Hannover Medical School, Hannover, Germany
- Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
| | - Robert Zweigerdt
- Department of Cardiothoracic, Transplantation and Vascular Surgery, Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Hannover Medical School, Hannover, Germany
| | - Theresia Kraft
- Institute of Molecular and Cell Physiology, Hannover Medical School, Hannover, Germany
| | - Bogdan Iorga
- Institute of Molecular and Cell Physiology, Hannover Medical School, Hannover, Germany
- Department of Analytical Chemistry and Physical Chemistry, Faculty of Chemistry, University of Bucharest, Bucharest, Romania
| | - Joachim D. Meissner
- Institute of Molecular and Cell Physiology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
14
|
Neininger-Castro AC, Hayes JB, Sanchez ZC, Taneja N, Fenix AM, Moparthi S, Vassilopoulos S, Burnette DT. Independent regulation of Z-lines and M-lines during sarcomere assembly in cardiac myocytes revealed by the automatic image analysis software sarcApp. eLife 2023; 12:RP87065. [PMID: 37921850 PMCID: PMC10624428 DOI: 10.7554/elife.87065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2023] Open
Abstract
Sarcomeres are the basic contractile units within cardiac myocytes, and the collective shortening of sarcomeres aligned along myofibrils generates the force driving the heartbeat. The alignment of the individual sarcomeres is important for proper force generation, and misaligned sarcomeres are associated with diseases, including cardiomyopathies and COVID-19. The actin bundling protein, α-actinin-2, localizes to the 'Z-Bodies" of sarcomere precursors and the 'Z-Lines' of sarcomeres, and has been used previously to assess sarcomere assembly and maintenance. Previous measurements of α-actinin-2 organization have been largely accomplished manually, which is time-consuming and has hampered research progress. Here, we introduce sarcApp, an image analysis tool that quantifies several components of the cardiac sarcomere and their alignment in muscle cells and tissue. We first developed sarcApp to utilize deep learning-based segmentation and real space quantification to measure α-actinin-2 structures and determine the organization of both precursors and sarcomeres/myofibrils. We then expanded sarcApp to analyze 'M-Lines' using the localization of myomesin and a protein that connects the Z-Lines to the M-Line (titin). sarcApp produces 33 distinct measurements per cell and 24 per myofibril that allow for precise quantification of changes in sarcomeres, myofibrils, and their precursors. We validated this system with perturbations to sarcomere assembly. We found perturbations that affected Z-Lines and M-Lines differently, suggesting that they may be regulated independently during sarcomere assembly.
Collapse
Affiliation(s)
- Abigail C Neininger-Castro
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine Basic SciencesNashvilleUnited States
| | - James B Hayes
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine Basic SciencesNashvilleUnited States
| | - Zachary C Sanchez
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine Basic SciencesNashvilleUnited States
| | - Nilay Taneja
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine Basic SciencesNashvilleUnited States
| | - Aidan M Fenix
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine Basic SciencesNashvilleUnited States
| | - Satish Moparthi
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en MyologieParisFrance
| | - Stéphane Vassilopoulos
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en MyologieParisFrance
| | - Dylan Tyler Burnette
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine Basic SciencesNashvilleUnited States
| |
Collapse
|
15
|
DePalma SJ, Jillberto J, Stis AE, Huang DD, Lo J, Davidson CD, Chowdhury A, Jewett ME, Kobeissi H, Chen CS, Lejeune E, Helms AS, Nordsletten DA, Baker BM. Matrix architecture and mechanics regulate myofibril organization, costamere assembly, and contractility of engineered myocardial microtissues. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.20.563346. [PMID: 37961415 PMCID: PMC10634701 DOI: 10.1101/2023.10.20.563346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
The mechanical function of the myocardium is defined by cardiomyocyte contractility and the biomechanics of the extracellular matrix (ECM). Understanding this relationship remains an important unmet challenge due to limitations in existing approaches for engineering myocardial tissue. Here, we established arrays of cardiac microtissues with tunable mechanics and architecture by integrating ECM-mimetic synthetic, fiber matrices and induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs), enabling real-time contractility readouts, in-depth structural assessment, and tissue-specific computational modeling. We find that the stiffness and alignment of matrix fibers distinctly affect the structural development and contractile function of pure iPSC-CM tissues. Further examination into the impact of fibrous matrix stiffness enabled by computational models and quantitative immunofluorescence implicates cell-ECM interactions in myofibril assembly and notably costamere assembly, which correlates with improved contractile function of tissues. These results highlight how iPSC-CM tissue models with controllable architecture and mechanics can inform the design of translatable regenerative cardiac therapies.
Collapse
|
16
|
Neininger-Castro AC, Hayes JB, Sanchez ZC, Taneja N, Fenix AM, Moparthi S, Vassilopoulos S, Burnette DT. Independent regulation of Z-lines and M-lines during sarcomere assembly in cardiac myocytes revealed by the automatic image analysis software sarcApp. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.11.523681. [PMID: 36711995 PMCID: PMC9882215 DOI: 10.1101/2023.01.11.523681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Sarcomeres are the basic contractile units within cardiac myocytes, and the collective shortening of sarcomeres aligned along myofibrils generates the force driving the heartbeat. The alignment of the individual sarcomeres is important for proper force generation, and misaligned sarcomeres are associated with diseases including cardiomyopathies and COVID-19. The actin bundling protein, α-actinin-2, localizes to the "Z-Bodies" of sarcomere precursors and the "Z-Lines" of sarcomeres, and has been used previously to assess sarcomere assembly and maintenance. Previous measurements of α-actinin-2 organization have been largely accomplished manually, which is time-consuming and has hampered research progress. Here, we introduce sarcApp, an image analysis tool that quantifies several components of the cardiac sarcomere and their alignment in muscle cells and tissue. We first developed sarcApp to utilize deep learning-based segmentation and real space quantification to measure α-actinin-2 structures and determine the organization of both precursors and sarcomeres/myofibrils. We then expanded sarcApp to analyze "M-Lines" using the localization of myomesin and a protein that connects the Z-Lines to the M-Line (titin). sarcApp produces 33 distinct measurements per cell and 24 per myofibril that allow for precise quantification of changes in sarcomeres, myofibrils, and their precursors. We validated this system with perturbations to sarcomere assembly. We found perturbations that affected Z-Lines and M-Lines differently, suggesting that they may be regulated independently during sarcomere assembly.
Collapse
Affiliation(s)
- Abigail C. Neininger-Castro
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine Basic Sciences, Nashville, TN
| | - James B. Hayes
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine Basic Sciences, Nashville, TN
| | - Zachary C. Sanchez
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine Basic Sciences, Nashville, TN
| | - Nilay Taneja
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine Basic Sciences, Nashville, TN
| | - Aidan M. Fenix
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine Basic Sciences, Nashville, TN
| | - Satish Moparthi
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, Paris, France
| | - Stéphane Vassilopoulos
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, Paris, France
| | - Dylan T. Burnette
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine Basic Sciences, Nashville, TN
| |
Collapse
|
17
|
Jiang H, Fang T, Cheng Z. Mechanism of heart failure after myocardial infarction. J Int Med Res 2023; 51:3000605231202573. [PMID: 37818767 PMCID: PMC10566288 DOI: 10.1177/03000605231202573] [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: 07/02/2023] [Accepted: 08/14/2023] [Indexed: 10/13/2023] Open
Abstract
Despite the widespread use of early revascularization and drugs to regulate the neuroendocrine system, the impact of such measures on alleviating the development of heart failure (HF) after myocardial infarction (MI) remains limited. Therefore, it is important to discuss the development of new therapeutic strategies to prevent or reverse HF after MI. This requires a better understanding of the potential mechanisms involved. HF after MI is the result of complex pathophysiological processes, with adverse ventricular remodeling playing a major role. Adverse ventricular remodeling refers to the heart's adaptation in terms of changes in ventricular size, shape, and function under the influence of various regulatory factors, including the mechanical, neurohormonal, and cardiac inflammatory immune environments; ischemia/reperfusion injury; energy metabolism; and genetic correlation factors. Additionally, unique right ventricular dysfunction can occur secondary to ischemic shock in the surviving myocardium. HF after MI may also be influenced by other factors. This review summarizes the main pathophysiological mechanisms of HF after MI and highlights sex-related differences in the prognosis of patients with acute MI. These findings provide new insights for guiding the development of targeted treatments to delay the progression of HF after MI and offering incremental benefits to existing therapies.
Collapse
Affiliation(s)
- Huaiyu Jiang
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tingting Fang
- Department of Cardiology, The Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, China
| | - Zeyi Cheng
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
18
|
Braczko A, Harasim G, Kawecka A, Walczak I, Kapusta M, Narajczyk M, Stawarska K, Smoleński RT, Kutryb-Zając B. Blocking cholesterol formation and turnover improves cellular and mitochondria function in murine heart microvascular endothelial cells and cardiomyocytes. Front Physiol 2023; 14:1216267. [PMID: 37745244 PMCID: PMC10512729 DOI: 10.3389/fphys.2023.1216267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 08/29/2023] [Indexed: 09/26/2023] Open
Abstract
Background: Statins and proprotein convertase subtilisin/kexin type 9 inhibitors (PCSK9i) are cornerstones of therapy to prevent cardiovascular disease, acting by lowering lipid concentrations and only partially identified pleiotropic effects. This study aimed to analyze impacts of atorvastatin and synthetic peptide PCSK9i on bioenergetics and function of microvascular endothelial cells and cardiomyocytes. Methods: Mitochondrial function and abundance as well as intracellular nucleotides, membrane potential, cytoskeleton structure, and cell proliferation rate were evaluated in mouse heart microvascular endothelial cells (H5V) and cardiomyocytes (HL-1) under normal and hypoxia-mimicking conditions (CoCl2 exposure). Results: In normal conditions PCSK9i, unlike atorvastatin, enhanced mitochondrial respiratory parameters, increased nucleotide levels, prevented actin cytoskeleton disturbances and stimulated endothelial cell proliferation. Under hypoxia-mimicking conditions both atorvastatin and PCSK9i improved the mitochondrial respiration and membrane potential in both cell types. Conclusion: This study demonstrated that both treatments benefited the endothelial cell and cardiomyocyte bioenergetics, but the effects of PCSK9i were superior.
Collapse
Affiliation(s)
- Alicja Braczko
- Department of Biochemistry, Medical University of Gdansk, Gdańsk, Poland
| | - Gabriela Harasim
- Department of Biochemistry, Medical University of Gdansk, Gdańsk, Poland
| | - Ada Kawecka
- Department of Biochemistry, Medical University of Gdansk, Gdańsk, Poland
| | - Iga Walczak
- Department of Biochemistry, Medical University of Gdansk, Gdańsk, Poland
| | | | | | - Klaudia Stawarska
- Department of Biochemistry, Medical University of Gdansk, Gdańsk, Poland
| | | | | |
Collapse
|
19
|
Darby JR, Zhang S, Holman SL, Muhlhausler BS, McMillen IC, Morrison JL. Cardiac growth and metabolism of the fetal sheep are not vulnerable to a 10 day increase in fetal glucose and insulin concentrations during late gestation. Heliyon 2023; 9:e18292. [PMID: 37519661 PMCID: PMC10372399 DOI: 10.1016/j.heliyon.2023.e18292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 07/06/2023] [Accepted: 07/13/2023] [Indexed: 08/01/2023] Open
Abstract
Aims To evaluate the effects of fetal glucose infusion in late gestation on the mRNA expression and protein abundance of molecules involved in the regulation of cardiac growth and metabolism. Main methods Either saline or glucose was infused into fetal sheep from 130 to 140 days (d) gestation (term, 150 d). At 140 d gestation, left ventricle tissue samples were collected. Quantitative real-time RT-PCR and Western blot were used to determine the mRNA expression and protein abundance of key signalling molecules within the left ventricle of the fetal heart. Key findings Although intra-fetal glucose infusion increased fetal plasma glucose and insulin concentrations, there was no change in the expression of molecules within the signalling pathways that regulate proliferation, hypertrophy, apoptosis or fibrosis in the fetal heart. Cardiac Solute carrier family 2 member 1 (SLC2A1) mRNA expression was decreased by glucose infusion. Glucose infusion increased cardiac mRNA expression of both Peroxisome proliferator activated receptor alpha (PPARA) and peroxisome proliferator activated receptor gamma (PPARG). However, there was no change in the mRNA expression of PPAR cofactors or molecules with PPAR response elements. Furthermore, glucose infusion did not impact the protein abundance of the 5 oxidative phosphorylation complexes of the electron transport chain. Significance Despite a 10-day doubling of fetal plasma glucose and insulin concentrations, the present study suggests that within the fetal left ventricle, the mRNA and protein expression of the signalling molecules involved in cardiac growth, development and metabolism are relatively unaffected.
Collapse
Affiliation(s)
| | | | | | | | | | - Janna L. Morrison
- Corresponding author. Australian Research Council Future Fellow, Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, GPO Box 2471, Adelaide, SA, 5001, Australia,
| |
Collapse
|
20
|
Korover N, Etzion S, Cherniak A, Rabinski T, Levitas A, Etzion Y, Ofir R, Parvari R, Cohen S. Functional defects in hiPSCs-derived cardiomyocytes from patients with a PLEKHM2-mutation associated with dilated cardiomyopathy and left ventricular non-compaction. Biol Res 2023; 56:34. [PMID: 37349842 PMCID: PMC10288792 DOI: 10.1186/s40659-023-00442-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 05/17/2023] [Indexed: 06/24/2023] Open
Abstract
Dilated cardiomyopathy (DCM) is a primary myocardial disease, leading to heart failure and excessive risk of sudden cardiac death with rather poorly understood pathophysiology. In 2015, Parvari's group identified a recessive mutation in the autophagy regulator, PLEKHM2 gene, in a family with severe recessive DCM and left ventricular non-compaction (LVNC). Fibroblasts isolated from these patients exhibited abnormal subcellular distribution of endosomes, Golgi apparatus, lysosomes and had impaired autophagy flux. To better understand the effect of mutated PLEKHM2 on cardiac tissue, we generated and characterized induced pluripotent stem cells-derived cardiomyocytes (iPSC-CMs) from two patients and a healthy control from the same family. The patient iPSC-CMs showed low expression levels of genes encoding for contractile functional proteins (α and β-myosin heavy chains and 2v and 2a-myosin light chains), structural proteins integral to heart contraction (Troponin C, T and I) and proteins participating in Ca2+ pumping action (SERCA2 and Calsequestrin 2) compared to their levels in control iPSC-derived CMs. Furthermore, the sarcomeres of the patient iPSC-CMs were less oriented and aligned compared to control cells and generated slowly beating foci with lower intracellular calcium amplitude and abnormal calcium transient kinetics, measured by IonOptix system and MuscleMotion software. Autophagy in patient's iPSC-CMs was impaired as determined from a decrease in the accumulation of autophagosomes in response to chloroquine and rapamycin treatment, compared to control iPSC-CMs. Impairment in autophagy together with the deficiency in the expression of NKX2.5, MHC, MLC, Troponins and CASQ2 genes, which are related to contraction-relaxation coupling and intracellular Ca2+ signaling, may contribute to the defective function of the patient CMs and possibly affect cell maturation and cardiac failure with time.
Collapse
Affiliation(s)
- Nataly Korover
- Avram and Stella Goldstein-Goren Department of Biotechnology Engineering, Ben-Gurion University of the Negev, 84105, Beer-Sheva, Israel.
| | - Sharon Etzion
- Regenerative Medicine & Stem Cell Research Center, Ben-Gurion University of the Negev, 84105, Beer-Sheva, Israel
| | - Alexander Cherniak
- Avram and Stella Goldstein-Goren Department of Biotechnology Engineering, Ben-Gurion University of the Negev, 84105, Beer-Sheva, Israel
| | - Tatiana Rabinski
- Regenerative Medicine & Stem Cell Research Center, Ben-Gurion University of the Negev, 84105, Beer-Sheva, Israel
| | - Aviva Levitas
- Department of Pediatric Cardiology, Soroka University Medical Center and Faculty of Health Sciences, Ben-Gurion University of the Negev, 84105, Beer-Sheva, Israel
| | - Yoram Etzion
- Regenerative Medicine & Stem Cell Research Center, Ben-Gurion University of the Negev, 84105, Beer-Sheva, Israel
- Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, 84105, Beer-Sheva, Israel
| | - Rivka Ofir
- Regenerative Medicine & Stem Cell Research Center, Ben-Gurion University of the Negev, 84105, Beer-Sheva, Israel
- Dead Sea & Arava Science Center, 8691000, Masada, Israel
| | - Ruti Parvari
- Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, 84105, Beer-Sheva, Israel
- National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Smadar Cohen
- Avram and Stella Goldstein-Goren Department of Biotechnology Engineering, Ben-Gurion University of the Negev, 84105, Beer-Sheva, Israel
- Regenerative Medicine & Stem Cell Research Center, Ben-Gurion University of the Negev, 84105, Beer-Sheva, Israel
- Ilse Katz Institute for Nanoscale Science and Technology, Ben-Gurion University of the Negev, 84105, Beer-Sheva, Israel
| |
Collapse
|
21
|
Belhaj A, Dewachter L, Monier A, Vegh G, Rorive S, Remmelink M, Closset M, Melot C, Creteur J, Salmon I, Rondelet B. Beneficial Effects of Tacrolimus on Brain-Death-Associated Right Ventricular Dysfunction in Pigs. Int J Mol Sci 2023; 24:10439. [PMID: 37445625 PMCID: PMC10341891 DOI: 10.3390/ijms241310439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 06/16/2023] [Accepted: 06/19/2023] [Indexed: 07/15/2023] Open
Abstract
BACKGROUND Right ventricular (RV) dysfunction remains a major problem after heart transplantation and may be associated with brain death (BD) in a donor. A calcineurin inhibitor tacrolimus was recently found to have beneficial effects on heart function. Here, we examined whether tacrolimus might prevent BD-induced RV dysfunction and the associated pathobiological changes. METHODS After randomized tacrolimus (n = 8; 0.05 mg·kg-1·day-1) or placebo (n = 9) pretreatment, pigs were assigned to a BD procedure and hemodynamically investigated 1, 3, 5, and 7 h after the Cushing reflex. After euthanasia, myocardial tissue was sampled for pathobiological evaluation. Seven pigs were used as controls. RESULTS Calcineurin inhibition prevented increases in pulmonary vascular resistance and RV-arterial decoupling induced by BD. BD was associated with an increased RV pro-apoptotic Bax-to-Bcl2 ratio and RV and LV apoptotic rates, which were prevented by tacrolimus. BD induced increased expression of the pro-inflammatory IL-6-to-IL-10 ratio, their related receptors, and vascular cell adhesion molecule-1 in both the RV and LV. These changes were prevented by tacrolimus. RV and LV neutrophil infiltration induced by BD was partly prevented by tacrolimus. BD was associated with decreased RV expression of the β-1 adrenergic receptor and sarcomere (myosin heavy chain [MYH]7-to-MYH6 ratio) components, while β-3 adrenergic receptor, nitric oxide-synthase 3, and glucose transporter 1 expression increased. These changes were prevented by tacrolimus. CONCLUSIONS Brain death was associated with isolated RV dysfunction. Tacrolimus prevented RV dysfunction induced by BD through the inhibition of apoptosis and inflammation activation.
Collapse
Affiliation(s)
- Asmae Belhaj
- Department of Cardio-Vascular, Thoracic Surgery and Lung Transplantation, CHU UCL Namur, UCLouvain, 5530 Yvoir, Belgium;
- Laboratory of Physiology and Pharmacology, Faculty of Medicine, Université Libre de Bruxelles, 1070 Brussels, Belgium; (L.D.); (A.M.); (G.V.); (C.M.)
| | - Laurence Dewachter
- Laboratory of Physiology and Pharmacology, Faculty of Medicine, Université Libre de Bruxelles, 1070 Brussels, Belgium; (L.D.); (A.M.); (G.V.); (C.M.)
| | - Astrid Monier
- Laboratory of Physiology and Pharmacology, Faculty of Medicine, Université Libre de Bruxelles, 1070 Brussels, Belgium; (L.D.); (A.M.); (G.V.); (C.M.)
| | - Gregory Vegh
- Laboratory of Physiology and Pharmacology, Faculty of Medicine, Université Libre de Bruxelles, 1070 Brussels, Belgium; (L.D.); (A.M.); (G.V.); (C.M.)
| | - Sandrine Rorive
- Department of Anatomopathology, Erasmus Academic Hospital, 1070 Brussels, Belgium; (S.R.); (M.R.); (I.S.)
| | - Myriam Remmelink
- Department of Anatomopathology, Erasmus Academic Hospital, 1070 Brussels, Belgium; (S.R.); (M.R.); (I.S.)
| | - Mélanie Closset
- Department of Laboratory Medicine, CHU UCL Namur, UCLouvain, 5530 Yvoir, Belgium;
| | - Christian Melot
- Laboratory of Physiology and Pharmacology, Faculty of Medicine, Université Libre de Bruxelles, 1070 Brussels, Belgium; (L.D.); (A.M.); (G.V.); (C.M.)
| | - Jacques Creteur
- Department of Critical Care, Erasmus Academic Hospital, 1070 Brussels, Belgium;
| | - Isabelle Salmon
- Department of Anatomopathology, Erasmus Academic Hospital, 1070 Brussels, Belgium; (S.R.); (M.R.); (I.S.)
| | - Benoît Rondelet
- Department of Cardio-Vascular, Thoracic Surgery and Lung Transplantation, CHU UCL Namur, UCLouvain, 5530 Yvoir, Belgium;
- Laboratory of Physiology and Pharmacology, Faculty of Medicine, Université Libre de Bruxelles, 1070 Brussels, Belgium; (L.D.); (A.M.); (G.V.); (C.M.)
| |
Collapse
|
22
|
Orgil BO, Munkhsaikhan U, Pierre JF, Li N, Xu F, Alberson NR, Johnson JN, Wetzel GT, Boukens BJD, Lu L, Towbin JA, Purevjav E. The TMEM43 S358L mutation affects cardiac, small intestine, and metabolic homeostasis in a knock-in mouse model. Am J Physiol Heart Circ Physiol 2023; 324:H866-H880. [PMID: 37083466 PMCID: PMC10190833 DOI: 10.1152/ajpheart.00712.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 03/23/2023] [Accepted: 03/23/2023] [Indexed: 04/22/2023]
Abstract
The transmembrane protein 43 (TMEM43/LUMA) p.S358L mutation causes arrhythmogenic cardiomyopathy named as ARVC5, a fully penetrant disease with high risk of ventricular arrhythmias, sudden death, and heart failure. Male gender and vigorous exercise independently predicted deleterious outcome. Our systems genetics analysis revealed the importance of Tmem43 for cardiac and metabolic pathways associated with elevated lipid absorption from small intestine. This study sought to delineate gender-specific cardiac, intestinal, and metabolic phenotypes in vivo and investigate underlying pathophysiological mechanisms of S358L mutation. Serial echocardiography, surface electrocardiography (ECG), treadmill running, and body EchoMRI have been used in knock-in heterozygous (Tmem43WT/S358L), homozygous (Tmem43S358L), and wildtype (Tmem43WT) littermate mice. Electron microscopy, histology, immunohistochemistry, transcriptome, and protein analysis have been performed in cardiac and intestinal tissues. Systolic dysfunction was apparent in 3-mo-old Tmem43S358L and 6-mo-old Tmem43WT/S358L mutants. Both mutant lines displayed intolerance to acute stress at 6 mo of age, arrhythmias, fibro-fatty infiltration, and subcellular abnormalities in the myocardium. Microarray analysis found significantly differentially expressed genes between left ventricular (LV) and right ventricular (RV) myocardium. Mutants displayed diminished PPARG activities and significantly reduced TMEM43 and β-catenin expression in the heart, whereas junctional plakoglobin (JUP) translocated into nuclei of mutant cardiomyocytes. Conversely, elongated villi, fatty infiltration, and overexpression of gut epithelial proliferation markers, β-catenin and Ki-67, were evident in small intestine of mutants. We defined Tmem43 S358L-induced pathological effects on cardiac and intestinal homeostasis via distinctly disturbed WNT-β-catenin and PPARG signaling thereby contributing to ARVC5 pathophysiology. Results suggest that cardiometabolic assessment in mutation carriers may be important for predictive and personalized care.NEW & NOTEWORTHY This manuscript describes the findings of our investigation of cardiac, small intestine, and metabolic features of Tmem43-S358L mouse model. By investigating interorgan pathologies, we uncovered multiple mechanisms of the S358L-induced disease, and these unique mechanisms likely appear to contribute to the disease pathogenesis. We hope our findings are important and novel and open new avenues in the hunting for additional diagnostic and therapeutic targets in subjects carrying TMEM43 mutation.
Collapse
Affiliation(s)
- Buyan-Ochir Orgil
- Department of Pediatrics, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, United States
- Children's Foundation Research Institute, Le Bonheur Children's Hospital Memphis, Memphis, Tennessee, United States
| | - Undral Munkhsaikhan
- Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee, United States
| | - Joseph F Pierre
- Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, Wisconsin, United States
| | - Ning Li
- Department of Pediatrics, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, United States
- Children's Foundation Research Institute, Le Bonheur Children's Hospital Memphis, Memphis, Tennessee, United States
- Department of Cardiology, Second Affiliated Hospital, Harbin Medical University, Harbin, People's Republic of China
| | - Fuyi Xu
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, Tennessee, United States
- School of Pharmacy, Binzhou Medical University, Yantai, People's Republic of China
| | - Neely R Alberson
- Department of Pediatrics, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, United States
- Children's Foundation Research Institute, Le Bonheur Children's Hospital Memphis, Memphis, Tennessee, United States
| | - Jason N Johnson
- Department of Pediatrics, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, United States
- Children's Foundation Research Institute, Le Bonheur Children's Hospital Memphis, Memphis, Tennessee, United States
| | - Glenn T Wetzel
- Department of Pediatrics, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, United States
- Children's Foundation Research Institute, Le Bonheur Children's Hospital Memphis, Memphis, Tennessee, United States
| | - Bastiaan J D Boukens
- Department of Medical Biology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Lu Lu
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, Tennessee, United States
| | - Jeffrey A Towbin
- Department of Pediatrics, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, United States
- Children's Foundation Research Institute, Le Bonheur Children's Hospital Memphis, Memphis, Tennessee, United States
- Pediatric Cardiology, St. Jude Children's Research Hospital, Memphis, Tennessee, United States
| | - Enkhsaikhan Purevjav
- Department of Pediatrics, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, United States
- Children's Foundation Research Institute, Le Bonheur Children's Hospital Memphis, Memphis, Tennessee, United States
| |
Collapse
|
23
|
Bonilla IM, Baine S, Pokrass A, Mariángelo JIE, Kalyanasundaram A, Bogdanov V, Mezache L, Sakuta G, Beard CM, Belevych A, Tikunova S, Terentyeva R, Terentyev D, Davis J, Veeraraghavan R, Carnes CA, Györke S. STIM1 ablation impairs exercise-induced physiological cardiac hypertrophy and dysregulates autophagy in mouse hearts. J Appl Physiol (1985) 2023; 134:1287-1299. [PMID: 36995910 PMCID: PMC10190841 DOI: 10.1152/japplphysiol.00363.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 03/13/2023] [Accepted: 03/21/2023] [Indexed: 03/31/2023] Open
Abstract
Cardiac stromal interaction molecule 1 (STIM1), a key mediator of store-operated Ca2+ entry (SOCE), is a known determinant of cardiomyocyte pathological growth in hypertrophic cardiomyopathy. We examined the role of STIM1 and SOCE in response to exercise-dependent physiological hypertrophy. Wild-type (WT) mice subjected to exercise training (WT-Ex) showed a significant increase in exercise capacity and heart weight compared with sedentary (WT-Sed) mice. Moreover, myocytes from WT-Ex hearts displayed an increase in length, but not width, compared with WT-Sed myocytes. Conversely, exercised cardiac-specific STIM1 knock-out mice (cSTIM1KO-Ex), although displaying significant increase in heart weight and cardiac dilation, evidenced no changes in myocyte size and displayed a decreased exercise capacity, impaired cardiac function, and premature death compared with sedentary cardiac-specific STIM1 knock-out mice (cSTIM1KO-Sed). Confocal Ca2+ imaging demonstrated enhanced SOCE in WT-Ex myocytes compared with WT-Sed myocytes with no measurable SOCE detected in cSTIM1KO myocytes. Exercise training induced a significant increase in cardiac phospho-Akt Ser473 in WT mice but not in cSTIM1KO mice. No differences were observed in phosphorylation of mammalian target of rapamycin (mTOR) and glycogen synthase kinase (GSK) in exercised versus sedentary cSTIM1KO mice hearts. cSTIM1KO-Sed mice showed increased basal MAPK phosphorylation compared with WT-Sed that was not altered by exercise training. Finally, histological analysis revealed exercise resulted in increased autophagy in cSTIM1KO but not in WT myocytes. Taken together, our results suggest that adaptive cardiac hypertrophy in response to exercise training involves STIM1-mediated SOCE. Our results demonstrate that STIM1 is involved in and essential for the myocyte longitudinal growth and mTOR activation in response to endurance exercise training.NEW & NOTEWORTHY Store-operated Ca2+ entry (SOCE) has been implicated in pathological cardiac hypertrophy; however, its role in physiological hypertrophy is unknown. Here we report that SOCE is also essential for physiological cardiac hypertrophy and functional adaptations in response to endurance exercise. These adaptations were associated with activation of AKT/mTOR pathway and curtailed cardiac autophagy and degeneration. Thus, SOCE is a common mechanism and an important bifurcation point for signaling paths involved in physiological and pathological hypertrophy.
Collapse
Affiliation(s)
- Ingrid M Bonilla
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, United States
- Veterans Affairs Caribbean Healthcare System, San Juan, Puerto Rico, United States
- Department of Pharmacology and Toxicology, School of Medicine, University of Puerto Rico, San Juan, Puerto Rico, United States
| | - Stephen Baine
- Department of Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio, United States
| | - Anastasia Pokrass
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, United States
| | - Juan Ignacio Elio Mariángelo
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, United States
| | - Anuradha Kalyanasundaram
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, United States
- Bob and Corrine Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart & Lung Research Institute, Columbus, Ohio, United States
| | - Vladimir Bogdanov
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, United States
| | - Louisa Mezache
- Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus, Ohio, United States
| | - Galina Sakuta
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, United States
| | - Casey M Beard
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, United States
| | - Andriy Belevych
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, United States
| | - Svetlana Tikunova
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, United States
| | - Radmila Terentyeva
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, United States
| | - Dmitry Terentyev
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, United States
| | - Jonathan Davis
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, United States
| | - Rengasayee Veeraraghavan
- Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus, Ohio, United States
| | - Cynthia A Carnes
- Department of Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio, United States
| | - Sandor Györke
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, United States
| |
Collapse
|
24
|
Chakrabarti M, Raut GK, Jain N, Bhadra MP. Prohibitin1 maintains mitochondrial quality in isoproterenol-induced cardiac hypertrophy in H9C2 cells. Biol Cell 2023; 115:e2200094. [PMID: 36453777 DOI: 10.1111/boc.202200094] [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: 10/04/2022] [Revised: 11/21/2022] [Accepted: 11/24/2022] [Indexed: 12/03/2022]
Abstract
BACKGROUND INFORMATION Various types of stress initially induce a state of cardiac hypertrophy (CH) in the heart. But, persistent escalation of cardiac stress leads to progression from an adaptive physiological to a maladaptive pathological state. So, elucidating molecular mechanisms that can attenuate CH is imperative in developing cardiac therapies. Previously, we showed that Prohibitin1 (PHB1) has a protective role in CH-induced oxidative stress. Nevertheless, it is unclear how PHB1, a mitochondrial protein, has a protective role in CH. Therefore, we hypothesized that PHB1 maintains mitochondrial quality in CH. To test this hypothesis, we used Isoproterenol (ISO) to induce CH in H9C2 cells overexpressing PHB1 and elucidated mitochondrial quality control pathways. RESULTS We found that overexpressing PHB1 attenuates ISO-induced CH and restores mitochondrial morphology in H9C2 cells. In addition, PHB1 blocks the pro-hypertrophic IGF1R/AKT pathway and restores the mitochondrial membrane polarization in ISO-treated cells. We observed that overexpressing PHB1 promotes mitochondrial biogenesis, improves mitochondrial respiratory capacity, and triggers mitophagy. CONCLUSION We conclude that PHB1 maintains mitochondrial quality in ISO-induced CH in H9C2 cells. SIGNIFICANCE Based on our results, we suggest that small molecules that induce PHB1 in cardiac cells may prove beneficial in developing cardiac therapies.
Collapse
Affiliation(s)
- Moumita Chakrabarti
- Applied Biology Department, CSIR-Indian Institute of Chemical Technology, Hyderabad, Telangana, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Ganesh Kumar Raut
- Applied Biology Department, CSIR-Indian Institute of Chemical Technology, Hyderabad, Telangana, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Nishant Jain
- Applied Biology Department, CSIR-Indian Institute of Chemical Technology, Hyderabad, Telangana, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Manika Pal Bhadra
- Applied Biology Department, CSIR-Indian Institute of Chemical Technology, Hyderabad, Telangana, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| |
Collapse
|
25
|
Min S, Cho SW. Engineered human cardiac tissues for modeling heart diseases. BMB Rep 2023; 56:32-42. [PMID: 36443005 PMCID: PMC9887099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Indexed: 01/28/2023] Open
Abstract
Heart disease is one of the major life-threatening diseases with high mortality and incidence worldwide. Several model systems, such as primary cells and animals, have been used to understand heart diseases and establish appropriate treatments. However, they have limitations in accuracy and reproducibility in recapitulating disease pathophysiology and evaluating drug responses. In recent years, three-dimensional (3D) cardiac tissue models produced using tissue engineering technology and human cells have outperformed conventional models. In particular, the integration of cell reprogramming techniques with bioengineering platforms (e.g., microfluidics, scaffolds, bioprinting, and biophysical stimuli) has facilitated the development of heart-ona- chip, cardiac spheroid/organoid, and engineered heart tissue (EHT) to recapitulate the structural and functional features of the native human heart. These cardiac models have improved heart disease modeling and toxicological evaluation. In this review, we summarize the cell types for the fabrication of cardiac tissue models, introduce diverse 3D human cardiac tissue models, and discuss the strategies to enhance their complexity and maturity. Finally, recent studies in the modeling of various heart diseases are reviewed. [BMB Reports 2023; 56(1): 32-42].
Collapse
Affiliation(s)
- Sungjin Min
- Department of Biotechnology, Yonsei University, Seoul 03722, Korea
| | - Seung-Woo Cho
- Department of Biotechnology, Yonsei University, Seoul 03722, Korea,Center for Nanomedicine, Institute for Basic Science (IBS), Seoul 03722, Korea,Corresponding author. Tel: +82-2-2123-5662; Fax: +82-2-362-7265; E-mail:
| |
Collapse
|
26
|
Min S, Cho SW. Engineered human cardiac tissues for modeling heart diseases. BMB Rep 2023; 56:32-42. [PMID: 36443005 PMCID: PMC9887099 DOI: 10.5483/bmbrep.2022-0185] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 11/28/2022] [Accepted: 11/28/2022] [Indexed: 07/30/2023] Open
Abstract
Heart disease is one of the major life-threatening diseases with high mortality and incidence worldwide. Several model systems, such as primary cells and animals, have been used to understand heart diseases and establish appropriate treatments. However, they have limitations in accuracy and reproducibility in recapitulating disease pathophysiology and evaluating drug responses. In recent years, three-dimensional (3D) cardiac tissue models produced using tissue engineering technology and human cells have outperformed conventional models. In particular, the integration of cell reprogramming techniques with bioengineering platforms (e.g., microfluidics, scaffolds, bioprinting, and biophysical stimuli) has facilitated the development of heart-ona- chip, cardiac spheroid/organoid, and engineered heart tissue (EHT) to recapitulate the structural and functional features of the native human heart. These cardiac models have improved heart disease modeling and toxicological evaluation. In this review, we summarize the cell types for the fabrication of cardiac tissue models, introduce diverse 3D human cardiac tissue models, and discuss the strategies to enhance their complexity and maturity. Finally, recent studies in the modeling of various heart diseases are reviewed. [BMB Reports 2023; 56(1): 32-42].
Collapse
Affiliation(s)
- Sungjin Min
- Department of Biotechnology, Yonsei University, Seoul 03722, Korea
| | - Seung-Woo Cho
- Department of Biotechnology, Yonsei University, Seoul 03722, Korea
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul 03722, Korea
| |
Collapse
|
27
|
Lv S, Zhang W, Yuan P, Lu C, Dong J, Zhang J. QiShenYiQi pill for myocardial collagen metabolism and apoptosis in rats of autoimmune cardiomyopathy. PHARMACEUTICAL BIOLOGY 2022; 60:722-728. [PMID: 35361037 PMCID: PMC8979511 DOI: 10.1080/13880209.2022.2056206] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 01/14/2022] [Accepted: 03/16/2022] [Indexed: 06/14/2023]
Abstract
CONTEXT QiShenYiQi pill (QSYQ) is a traditional Chinese medicine with a myocardial protective effect. OBJECTIVE To explore the effect of QSYQ on myocardial collagen metabolism in rats with autoimmune cardiomyopathy and explore the underlying mechanism from the aspect of apoptosis. MATERIALS AND METHODS We established an autoimmune cardiomyopathy model using Lewis rats. The rats were then randomly divided into six groups (n = 8): control, model, 3-methyladenine (15 mg/kg, intraperitoneal injection), QSYQ low-dose (135 mg/kg, gavage), QSYQ medium dose (270 mg/kg, gavage), and QSYQ high-dose (540 mg/kg, gavage) for four weeks. Van Gieson staining was applied for myocardial pathological characteristics, TUNEL fluorescence for myocardial cell apoptosis, enzyme-linked immunosorbent assay (ELISA) for serum PICP, PIIINP, and CTX-I levels, and western blot analysis for type I/III myocardial collagen, Bcl-2, Bax, and caspase-3 proteins. RESULTS Results showed that QSYQ (135, 270, or 540 mg/kg) significantly reduced the expression of myocardial type I/III collagen, and concentrations of serum PICP, PIIINP, and CTX-I in rats. Moreover, QSYQ could alleviate myocardial fibrosis more effectively at a higher dose. QSYQ could also inhibit myocardial apoptosis via downregulating Bcl-2 expression, and upregulating Bax and caspase-3 expression levels. DISCUSSION AND CONCLUSIONS The QSYQ can improve myocardial collagen metabolism by inhibiting apoptosis, which provides a potential therapeutic approach for autoimmune cardiomyopathy.
Collapse
Affiliation(s)
- Shichao Lv
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine (National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion), Tianjin, China
- Tianjin Key Laboratory of Traditional Research of TCM Prescription and Syndrome, Tianjin, China
| | - Wanqin Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine (National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion), Tianjin, China
| | - Peng Yuan
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine (National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion), Tianjin, China
| | - Chunmiao Lu
- Jiashan Hospital of Traditional Chinese Medicine, Zhejiang, China
| | | | - Junping Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine (National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion), Tianjin, China
| |
Collapse
|
28
|
Kuwabara JT, Hara A, Bhutada S, Gojanovich GS, Chen J, Hokutan K, Shettigar V, Lee AY, DeAngelo LP, Heckl JR, Jahansooz JR, Tacdol DK, Ziolo MT, Apte SS, Tallquist MD. Consequences of PDGFRα + fibroblast reduction in adult murine hearts. eLife 2022; 11:e69854. [PMID: 36149056 PMCID: PMC9576271 DOI: 10.7554/elife.69854] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 09/22/2022] [Indexed: 12/01/2022] Open
Abstract
Fibroblasts produce the majority of collagen in the heart and are thought to regulate extracellular matrix (ECM) turnover. Although fibrosis accompanies many cardiac pathologies and is generally deleterious, the role of fibroblasts in maintaining the basal ECM network and in fibrosis in vivo is poorly understood. We genetically ablated fibroblasts in mice to evaluate the impact on homeostasis of adult ECM and cardiac function after injury. Fibroblast-ablated mice demonstrated a substantive reduction in cardiac fibroblasts, but fibrillar collagen and the ECM proteome were not overtly altered when evaluated by quantitative mass spectrometry and N-terminomics. However, the distribution and quantity of collagen VI, microfibrillar collagen that forms an open network with the basement membrane, was reduced. In fibroblast-ablated mice, cardiac function was better preserved following angiotensin II/phenylephrine (AngII/PE)-induced fibrosis and myocardial infarction (MI). Analysis of cardiomyocyte function demonstrated altered sarcomere shortening and slowed calcium decline in both uninjured and AngII/PE-infused fibroblast-ablated mice. After MI, the residual resident fibroblasts responded to injury, albeit with reduced proliferation and numbers immediately after injury. These results indicate that the adult mouse heart tolerates a significant degree of fibroblast loss with a potentially beneficial impact on cardiac function after injury. The cardioprotective effect of controlled fibroblast reduction may have therapeutic value in heart disease.
Collapse
Affiliation(s)
- Jill T Kuwabara
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii at ManoaHonoluluUnited States
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii at ManoaHonoluluUnited States
| | - Akitoshi Hara
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii at ManoaHonoluluUnited States
| | - Sumit Bhutada
- Department of Biomedical Engineering, Cleveland Clinic Lerner Research InstituteClevelandUnited States
| | - Greg S Gojanovich
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii at ManoaHonoluluUnited States
| | - Jasmine Chen
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii at ManoaHonoluluUnited States
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii at ManoaHonoluluUnited States
| | - Kanani Hokutan
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii at ManoaHonoluluUnited States
| | - Vikram Shettigar
- Dorothy M. Davis Heart and Lung Research Institute, Department of Physiology and Cell Biology, The Ohio State University Wexner Medical CenterColumbusUnited States
| | - Anson Y Lee
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii at ManoaHonoluluUnited States
| | - Lydia P DeAngelo
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii at ManoaHonoluluUnited States
| | - Jack R Heckl
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii at ManoaHonoluluUnited States
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii at ManoaHonoluluUnited States
| | - Julia R Jahansooz
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii at ManoaHonoluluUnited States
| | - Dillon K Tacdol
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii at ManoaHonoluluUnited States
| | - Mark T Ziolo
- Dorothy M. Davis Heart and Lung Research Institute, Department of Physiology and Cell Biology, The Ohio State University Wexner Medical CenterColumbusUnited States
| | - Suneel S Apte
- Department of Biomedical Engineering, Cleveland Clinic Lerner Research InstituteClevelandUnited States
| | - Michelle D Tallquist
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii at ManoaHonoluluUnited States
| |
Collapse
|
29
|
Valero-Muñoz M, Saw EL, Hekman RM, Blum BC, Hourani Z, Granzier H, Emili A, Sam F. Proteomic and phosphoproteomic profiling in heart failure with preserved ejection fraction (HFpEF). Front Cardiovasc Med 2022; 9:966968. [PMID: 36093146 PMCID: PMC9452734 DOI: 10.3389/fcvm.2022.966968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Accepted: 08/01/2022] [Indexed: 11/13/2022] Open
Abstract
Although the prevalence of heart failure with preserved ejection fraction (HFpEF) is increasing, evidence-based therapies for HFpEF remain limited, likely due to an incomplete understanding of this disease. This study sought to identify the cardiac-specific features of protein and phosphoprotein changes in a murine model of HFpEF using mass spectrometry. HFpEF mice demonstrated moderate hypertension, left ventricle (LV) hypertrophy, lung congestion and diastolic dysfunction. Proteomics analysis of the LV tissue showed that 897 proteins were differentially expressed between HFpEF and Sham mice. We observed abundant changes in sarcomeric proteins, mitochondrial-related proteins, and NAD-dependent protein deacetylase sirtuin-3 (SIRT3). Upregulated pathways by GSEA analysis were related to immune modulation and muscle contraction, while downregulated pathways were predominantly related to mitochondrial metabolism. Western blot analysis validated SIRT3 downregulated cardiac expression in HFpEF vs. Sham (0.8 ± 0.0 vs. 1.0 ± 0.0; P < 0.001). Phosphoproteomics analysis showed that 72 phosphosites were differentially regulated between HFpEF and Sham LV. Aberrant phosphorylation patterns mostly occurred in sarcomere proteins and nuclear-localized proteins associated with contractile dysfunction and cardiac hypertrophy. Seven aberrant phosphosites were observed at the z-disk binding region of titin. Additional agarose gel analysis showed that while total titin cardiac expression remained unaltered, its stiffer N2B isoform was significantly increased in HFpEF vs. Sham (0.144 ± 0.01 vs. 0.127 ± 0.01; P < 0.05). In summary, this study demonstrates marked changes in proteins related to mitochondrial metabolism and the cardiac contractile apparatus in HFpEF. We propose that SIRT3 may play a role in perpetuating these changes and may be a target for drug development in HFpEF.
Collapse
Affiliation(s)
- María Valero-Muñoz
- Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, United States
| | - Eng Leng Saw
- Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, United States
| | - Ryan M. Hekman
- Department of Biology, Boston University, Boston, MA, United States
- Department of Biochemistry, Cell Biology and Genomics, Boston University, Boston, MA, United States
| | - Benjamin C. Blum
- Department of Biochemistry, Cell Biology and Genomics, Boston University, Boston, MA, United States
- Center for Network Systems Biology, Boston University, Boston, MA, United States
| | - Zaynab Hourani
- Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, AZ, United States
| | - Henk Granzier
- Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, AZ, United States
| | - Andrew Emili
- Department of Biology, Boston University, Boston, MA, United States
- Department of Biochemistry, Cell Biology and Genomics, Boston University, Boston, MA, United States
| | - Flora Sam
- Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, United States
| |
Collapse
|
30
|
Abstract
Cardiac remodelling is characterized by abnormal changes in the function and morphological properties such as diameter, mass, normal diameter of cavities, heart shape, fibrosis, thickening of vessels and heart layers, cardiomyopathy, infiltration of inflammatory cells, and some others. These damages are associated with damage to systolic and diastolic abnormalities, damage to ventricular function, and vascular remodelling, which may lead to heart failure and death. Exposure of the heart to radiation or anti-cancer drugs including chemotherapy drugs such as doxorubicin, receptor tyrosine kinase inhibitors (RTKIs) such as imatinib, and immune checkpoint inhibitors (ICIs) can induce several abnormal changes in the heart structure and function through the induction of inflammation and fibrosis, vascular remodelling, hypertrophy, and some others. This review aims to explain the basic mechanisms behind cardiac remodelling following cancer therapy by different anti-cancer modalities.
Collapse
|
31
|
Correale M, Mazzeo P, Tricarico L, Croella F, Fortunato M, Magnesa M, Amatruda M, Alfieri S, Ferrara S, Ceci V, Dattilo G, Mele M, Iacoviello M, Brunetti ND. Pharmacological Anti-Remodelling Effects of Disease-Modifying Drugs in Heart Failure with Reduced Ejection Fraction. Clin Drug Investig 2022; 42:567-579. [PMID: 35726047 DOI: 10.1007/s40261-022-01166-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/26/2022] [Indexed: 11/25/2022]
Abstract
Cardiac remodelling is an adverse phenomenon linked to heart failure progression and an important contributor to heart failure severity. Cardiac remodelling could represent the real therapeutic goal in the treatment of patients with heart failure with reduced ejection fraction, being potentially reversed through different pharmacotherapies. Currently, there are well-established drugs such as angiotensin-converting enzyme inhibitors/angiotensin II receptor blockers and β-blockers with anti-remodelling effects; recently, angiotensin receptor neprilysin inhibitor effects on inhibiting cardiac remodelling (improving N-terminal pro-B-type natriuretic peptide levels, echocardiographic parameters of reverse cardiac remodelling and right ventricular function in patients with heart failure with reduced ejection fraction) were demonstrated. More recently, hemodynamic consequences of gliflozins, reduced cardiac hydrostatic pressure as a possible cause of ventricular remodelling and hypertrophy were proposed to explain potential anti-remodelling effects of gliflozins. Gliflozins exert their cardioprotective effects by attenuating myofibroblast activity and collagen-mediated remodelling. Another postulated mechanism is represented by the reduction in sympathetic activity, through the reduction in renal afferent nervous activity and the suppression of central reflex mechanisms. Benefits of gliflozins on left ventricular hypertrophy, dilation, and systolic and diastolic function were also described. In this review, we aimed to provide a wide overview on cardiac remodelling with a particular focus on possible anti-remodelling effects of angiotensin receptor neprilysin inhibitors and gliflozins.
Collapse
Affiliation(s)
- Michele Correale
- Cardiothoracic Department, Policlinico Riuniti University Hospital, Viale Pinto 1, 71100, Foggia, Italy.
| | - Pietro Mazzeo
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Lucia Tricarico
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Francesca Croella
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Martino Fortunato
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Michele Magnesa
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Marco Amatruda
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Simona Alfieri
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Salvatore Ferrara
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Vincenzo Ceci
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Giuseppe Dattilo
- Cardiology Unit, Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Marco Mele
- Cardiothoracic Department, Policlinico Riuniti University Hospital, Viale Pinto 1, 71100, Foggia, Italy
| | - Massimo Iacoviello
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | | |
Collapse
|
32
|
Dong T, Zhao Y, Jin HF, Shen L, Lin Y, Si LL, Chen L, Liu JC. SNTA1-deficient human cardiomyocytes demonstrate hypertrophic phenotype and calcium handling disorder. Stem Cell Res Ther 2022; 13:288. [PMID: 35773684 PMCID: PMC9248201 DOI: 10.1186/s13287-022-02955-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 05/07/2022] [Indexed: 11/13/2022] Open
Abstract
Background α-1-syntrophin (SNTA1), a protein encoded by SNTA1, is highly expressed in human cardiomyocytes. Mutations in SNTA1 are associated with arrhythmia and cardiomyopathy. Previous research on SNTA1 has been based on non-human cardiomyocytes. This study was designed to identify the phenotype of SNTA1-deficiency using human cardiomyocytes. Methods SNTA1 was knocked out in the H9 embryonic stem cell line using the CRISPR-Cas9 system. H9SNTA1KO cells were then induced to differentiate into cardiomyocytes using small molecule inhibitors. The phenotypic discrepancies associated with SNTA1-deficient cardiomyocytes were investigated. Results SNTA1 was truncated at the 149th amino acid position of PH1 domain by a stop codon (TGA) using the CRISPR-Cas9 system. SNTA1-deficiency did not affect the pluripotency of H9SNTA1KO, and they retain their in vitro ability to differentiate into cardiomyocytes. However, H9SNTA1KO derived cardiomyocytes exhibited hypertrophic phenotype, lower cardiac contractility, weak calcium transient intensity, and lower level of calcium in the sarcoplasmic reticulum. Early treatment of SNTA1-deficient cardiomyocytes with ranolazine improved the calcium transient intensity and cardiac contractility. Conclusion SNTA1-deficient cardiomyocytes can be used to research the etiology, pathogenesis, and potential therapies for myocardial diseases. The SNTA1-deficient cardiomyocyte model suggests that the maintenance of cardiac calcium homeostasis is a key target in the treatment of myocardial-related diseases. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-022-02955-4.
Collapse
Affiliation(s)
- Tao Dong
- Basic Medicine School, Qiqihar Medical University, 333 Bukui Street, Qiqihar, 161006, Heilongjiang, China.
| | - Yan Zhao
- College of Life Science and Agroforestry, Qiqihar University, Qiqihar, 161006, Heilongjiang, China
| | - Hai-Feng Jin
- Basic Medicine School, Qiqihar Medical University, 333 Bukui Street, Qiqihar, 161006, Heilongjiang, China
| | - Lei Shen
- Basic Medicine School, Qiqihar Medical University, 333 Bukui Street, Qiqihar, 161006, Heilongjiang, China
| | - Yan Lin
- Basic Medicine School, Qiqihar Medical University, 333 Bukui Street, Qiqihar, 161006, Heilongjiang, China
| | - Long-Long Si
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Li Chen
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Ji-Cheng Liu
- Qiqihar Institute of Medical and Pharmaceutical Sciences, Qiqihar Medical University, 333 Bukui Street, Qiqihar, 161006, Heilongjiang, China.
| |
Collapse
|
33
|
Ramos-Mondragon R, Edokobi N, Hodges SL, Wang S, Bouza AA, Canugovi C, Scheuing C, Juratli L, Abel WR, Noujaim SF, Madamanchi NR, Runge MS, Lopez-Santiago LF, Isom LL. Neonatal Scn1b-null mice have sinoatrial node dysfunction, altered atrial structure, and atrial fibrillation. JCI Insight 2022; 7:152050. [PMID: 35603785 PMCID: PMC9220823 DOI: 10.1172/jci.insight.152050] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 04/12/2022] [Indexed: 11/17/2022] Open
Abstract
Loss-of-function (LOF) variants in SCN1B, encoding the voltage-gated sodium channel β1/β1B subunits, are linked to neurological and cardiovascular diseases. Scn1b-null mice have spontaneous seizures and ventricular arrhythmias and die by approximately 21 days after birth. β1/β1B Subunits play critical roles in regulating the excitability of ventricular cardiomyocytes and maintaining ventricular rhythmicity. However, whether they also regulate atrial excitability is unknown. We used neonatal Scn1b-null mice to model the effects of SCN1B LOF on atrial physiology in pediatric patients. Scn1b deletion resulted in altered expression of genes associated with atrial dysfunction. Scn1b-null hearts had a significant accumulation of atrial collagen, increased susceptibility to pacing induced atrial fibrillation (AF), sinoatrial node (SAN) dysfunction, and increased numbers of cholinergic neurons in ganglia that innervate the SAN. Atropine reduced the incidence of AF in null animals. Action potential duration was prolonged in null atrial myocytes, with increased late sodium current density and reduced L-type calcium current density. Scn1b LOF results in altered atrial structure and AF, demonstrating the critical role played by Scn1b in atrial physiology during early postnatal mouse development. Our results suggest that SCN1B LOF variants may significantly impact the developing pediatric heart.
Collapse
Affiliation(s)
| | | | | | | | | | - Chandrika Canugovi
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | | | | | | | - Sami F. Noujaim
- Department of Molecular Pharmacology & Physiology, University of South Florida College of Medicine, Tampa, Florida, USA
| | - Nageswara R. Madamanchi
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Marschall S. Runge
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | | | - Lori L. Isom
- Department of Pharmacology and
- Department of Neurology and
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| |
Collapse
|
34
|
Nelson VL, Eadie AL, Brunt KR. A step towards a unifying pre-clinical model of dilated cardiomyopathy for drug development strategies or target validation. Am J Physiol Heart Circ Physiol 2022; 322:H1028-H1031. [PMID: 35427176 DOI: 10.1152/ajpheart.00141.2022] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Victoria L Nelson
- Department of Pharmacology, Dalhousie University; IMPART investigator team Canada, Saint John, New Brunswick, Canada
| | - Ashley L Eadie
- Department of Pharmacology, Dalhousie University; IMPART investigator team Canada, Saint John, N.B., Canada
| | - Keith R Brunt
- Department of Pharmacology, Dalhousie University; IMPART investigator team Canada, Saint John, NB, Canada
| |
Collapse
|
35
|
Acharya A, Nemade H, Rajendra Prasad K, Khan K, Hescheler J, Blackburn N, Hemmersbach R, Papadopoulos S, Sachinidis A. Live-Cell Imaging of the Contractile Velocity and Transient Intracellular Ca 2+ Fluctuations in Human Stem Cell-Derived Cardiomyocytes. Cells 2022; 11:1280. [PMID: 35455960 PMCID: PMC9031802 DOI: 10.3390/cells11081280] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 03/25/2022] [Accepted: 04/06/2022] [Indexed: 02/01/2023] Open
Abstract
Live-cell imaging techniques are essential for acquiring vital physiological and pathophysiological knowledge to understand and treat heart disease. For live-cell imaging of transient alterations of [Ca2+]i in human cardiomyocytes, we engineered human-induced pluripotent stem cells carrying a genetically-encoded Ca2+-indicator (GECI). To monitor sarcomere shortening and relaxation in cardiomyocytes in real-time, we generated a α-cardiac actinin (ACTN2)-copepod (cop) green fluorescent protein (GFP+)-human-induced pluripotent stem cell line by using the CRISPR-Cas9 and a homology directed recombination approach. The engineered human-induced pluripotent stem cells were differentiated in transgenic GECI-enhanced GFP+-cardiomyocytes and ACTN2-copGFP+-cardiomyocytes, allowing real-time imaging of [Ca2+]i transients and live recordings of the sarcomere shortening velocity of ACTN2-copGFP+-cardiomyocytes. We developed a video analysis software tool to quantify various parameters of sarcoplasmic Ca2+ fluctuations recorded during contraction of cardiomyocytes and to calculate the contraction velocity of cardiomyocytes in the presence and absence of different drugs affecting cardiac function. Our cellular and software tool not only proved the positive and negative inotropic and lusitropic effects of the tested cardioactive drugs but also quantified the expected effects precisely. Our platform will offer a human-relevant in vitro alternative for high-throughput drug screenings, as well as a model to explore the underlying mechanisms of cardiac diseases.
Collapse
Affiliation(s)
- Aviseka Acharya
- Working Group Sachinidis, Center for Physiology, Faculty of Medicine and University Hospital Cologne, The University of Cologne, 50931 Cologne, Germany; (A.A.); (H.N.); (K.R.P.); (K.K.); (J.H.); (S.P.)
| | - Harshal Nemade
- Working Group Sachinidis, Center for Physiology, Faculty of Medicine and University Hospital Cologne, The University of Cologne, 50931 Cologne, Germany; (A.A.); (H.N.); (K.R.P.); (K.K.); (J.H.); (S.P.)
| | - Krishna Rajendra Prasad
- Working Group Sachinidis, Center for Physiology, Faculty of Medicine and University Hospital Cologne, The University of Cologne, 50931 Cologne, Germany; (A.A.); (H.N.); (K.R.P.); (K.K.); (J.H.); (S.P.)
| | - Khadija Khan
- Working Group Sachinidis, Center for Physiology, Faculty of Medicine and University Hospital Cologne, The University of Cologne, 50931 Cologne, Germany; (A.A.); (H.N.); (K.R.P.); (K.K.); (J.H.); (S.P.)
| | - Jürgen Hescheler
- Working Group Sachinidis, Center for Physiology, Faculty of Medicine and University Hospital Cologne, The University of Cologne, 50931 Cologne, Germany; (A.A.); (H.N.); (K.R.P.); (K.K.); (J.H.); (S.P.)
| | - Nick Blackburn
- Bioras Company, Kaarsbergsvej 2, 8400 Ebeltoft, Denmark;
| | - Ruth Hemmersbach
- German Aerospace Center, Institute of Aerospace Medicine, Gravitational Biology, Linder Hoehe, 51147 Cologne, Germany;
| | - Symeon Papadopoulos
- Working Group Sachinidis, Center for Physiology, Faculty of Medicine and University Hospital Cologne, The University of Cologne, 50931 Cologne, Germany; (A.A.); (H.N.); (K.R.P.); (K.K.); (J.H.); (S.P.)
| | - Agapios Sachinidis
- Working Group Sachinidis, Center for Physiology, Faculty of Medicine and University Hospital Cologne, The University of Cologne, 50931 Cologne, Germany; (A.A.); (H.N.); (K.R.P.); (K.K.); (J.H.); (S.P.)
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931 Cologne, Germany
| |
Collapse
|
36
|
Wu W, Du Z, Wu L. Dexmedetomidine attenuates hypoxia-induced cardiomyocyte injury by promoting telomere/telomerase activity: Possible involvement of ERK1/2-Nrf2 signaling pathway. Cell Biol Int 2022; 46:1036-1046. [PMID: 35312207 DOI: 10.1002/cbin.11799] [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: 10/22/2021] [Revised: 12/26/2021] [Accepted: 01/22/2022] [Indexed: 11/06/2022]
Abstract
Dexmedetomidine (Dex), an α2-adrenergic receptor (α2-AR) agonist, possesses cardioprotection against ischaemic/hypoxic injury, but the exact mechanism is not fully elucidated. Since telomere/telomerase dysfunction is involved in myocardial ischemic damage, the present study aimed to investigate whether Dex ameliorates cobalt chloride (CoCl2; a hypoxia mimic agent in vitro)-induced the damage of H9c2 cardiomyocytes by improving telomere/telomerase dysfunction and further explored the underlying mechanism focusing on ERK1/2-Nrf2 signaling pathway. Result showed that Dex increased cell viability, decreased apoptosis, and reduced cardiomyocyte hypertrophy as illustrated by the decreases in cell surface area and the biomarker levels for cardiac hypertrophy including atrial natriuretic peptide (ANP), brain natriuretic peptide (BNP), and myosin heavy chain β (β-MHC) mRNA and protein in CoCl2 -exposed H9c2 cells. Intriguingly, Dex increased the telomere length and telomerase activity as well as telomere reverse transcriptase (TERT) protein and mRNA levels in H9c2 cells exposed to CoCl2 , indicating that Dex promotes telomere/telomerase function under hypoxia. In addition, Dex remarkably diminished the ROS generation, reduced MDA content, and increased antioxidative signaling as evidenced by the increases in SOD and GSH-Px activities. Furthermore, Dex increased the ratio of P-ERK1/2/T-ERK1/2 and P-Nrf2/T-Nrf2 and enhanced Nrf2 nuclear translocation in CoCl2 -subjected H9c2 cells, suggesting that Dex promotes the activation of the ERK1/2-Nrf2 signaling pathway. These novel findings indicated that Dex attenuates myocardial ischemic damage and reduces myocardial hypertrophy by promoting telomere/telomerase function, which may be associated with the activation of the ERK1/2-Nrf2 signaling pathway in vitro. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Wei Wu
- Department of Anesthesiology, Hunan Children's Hospital, Changsha, China 410007, People's Republic of China
| | - Zhen Du
- Department of Anesthesiology, Hunan Children's Hospital, Changsha, China 410007, People's Republic of China
| | - Lei Wu
- Department of Anesthesiology, Hunan Children's Hospital, Changsha, China 410007, People's Republic of China
| |
Collapse
|
37
|
Jiang Y, Li X, Guo T, Lu WJ, Ma S, Chang Y, Song Y, Zhang S, Bai R, Wang H, Qi M, Jiang H, Zhang H, Lan F. Ranolazine rescues the heart failure phenotype of PLN-deficient human pluripotent stem cell-derived cardiomyocytes. Stem Cell Reports 2022; 17:804-819. [PMID: 35334215 PMCID: PMC9023809 DOI: 10.1016/j.stemcr.2022.02.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 02/22/2022] [Accepted: 02/22/2022] [Indexed: 11/28/2022] Open
Abstract
Phospholamban (PLN) is a key regulator that controls the function of the sarcoplasmic reticulum (SR) and is required for the regulation of cardiac contractile function. Although PLN-deficient mice demonstrated improved cardiac function, PLN loss in humans can result in dilated cardiomyopathy (DCM) or heart failure (HF). The CRISPR-Cas9 technology was used to create a PLN knockout human induced pluripotent stem cell (hiPSC) line in this study. PLN deletion hiPSCs-CMs had enhanced contractility at day 30, but proceeded to a cardiac failure phenotype at day 60, with decreased contractility, mitochondrial damage, increased ROS production, cellular energy metabolism imbalance, and poor Ca2+ handling. Furthermore, adding ranolazine to PLN knockout hiPSCs-CMs at day 60 can partially restore Ca2+ handling disorders and cellular energy metabolism, alleviating the PLN knockout phenotype of HF, implying that the disorder of intracellular Ca2+ transport and the imbalance of cellular energy metabolism are the primary mechanisms for PLN deficiency pathogenesis.
Collapse
Affiliation(s)
- Youxu Jiang
- Beijing Laboratory for Cardiovascular Precision Medicine, MOE Key Laboratory of Biomedical Engineering for Cardiovascular Disease Research, Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing 100029, China
| | - Xiaowei Li
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Tianwei Guo
- Beijing Laboratory for Cardiovascular Precision Medicine, MOE Key Laboratory of Biomedical Engineering for Cardiovascular Disease Research, Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing 100029, China
| | - Wen-Jing Lu
- Beijing Laboratory for Cardiovascular Precision Medicine, MOE Key Laboratory of Biomedical Engineering for Cardiovascular Disease Research, Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing 100029, China
| | - Shuhong Ma
- Beijing Laboratory for Cardiovascular Precision Medicine, MOE Key Laboratory of Biomedical Engineering for Cardiovascular Disease Research, Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing 100029, China
| | - Yun Chang
- Beijing Laboratory for Cardiovascular Precision Medicine, MOE Key Laboratory of Biomedical Engineering for Cardiovascular Disease Research, Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing 100029, China; Department of Cardiology, Peking University Third Hospital, Beijing, China
| | - Yuanxiu Song
- Department of Cardiology, Peking University Third Hospital, Beijing, China
| | - Siyao Zhang
- Beijing Laboratory for Cardiovascular Precision Medicine, MOE Key Laboratory of Biomedical Engineering for Cardiovascular Disease Research, Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing 100029, China
| | - Rui Bai
- Beijing Laboratory for Cardiovascular Precision Medicine, MOE Key Laboratory of Biomedical Engineering for Cardiovascular Disease Research, Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing 100029, China
| | - Hongyue Wang
- Beijing Laboratory for Cardiovascular Precision Medicine, MOE Key Laboratory of Biomedical Engineering for Cardiovascular Disease Research, Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing 100029, China
| | - Man Qi
- Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen, Shenzhen Key Laboratory of Cardiovascular Disease, State Key Laboratory of Cardiovascular Disease, Key Laboratory of Pluripotent Stem Cells in Cardiac Repair and Regeneration, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| | - Hongfeng Jiang
- Beijing Laboratory for Cardiovascular Precision Medicine, MOE Key Laboratory of Biomedical Engineering for Cardiovascular Disease Research, Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing 100029, China.
| | - Hongjia Zhang
- Beijing Laboratory for Cardiovascular Precision Medicine, MOE Key Laboratory of Biomedical Engineering for Cardiovascular Disease Research, Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing 100029, China.
| | - Feng Lan
- Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen, Shenzhen Key Laboratory of Cardiovascular Disease, State Key Laboratory of Cardiovascular Disease, Key Laboratory of Pluripotent Stem Cells in Cardiac Repair and Regeneration, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China.
| |
Collapse
|
38
|
Jiang Z, Cui X, Qu P, Shang C, Xiang M, Wang J. Roles and mechanisms of puerarin on cardiovascular disease:A review. Biomed Pharmacother 2022; 147:112655. [DOI: 10.1016/j.biopha.2022.112655] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Revised: 01/13/2022] [Accepted: 01/16/2022] [Indexed: 12/13/2022] Open
|
39
|
Ušaj M, Moretto L, Månsson A. Critical Evaluation of Current Hypotheses for the Pathogenesis of Hypertrophic Cardiomyopathy. Int J Mol Sci 2022; 23:2195. [PMID: 35216312 PMCID: PMC8880276 DOI: 10.3390/ijms23042195] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 02/07/2022] [Accepted: 02/14/2022] [Indexed: 02/04/2023] Open
Abstract
Hereditary hypertrophic cardiomyopathy (HCM), due to mutations in sarcomere proteins, occurs in more than 1/500 individuals and is the leading cause of sudden cardiac death in young people. The clinical course exhibits appreciable variability. However, typically, heart morphology and function are normal at birth, with pathological remodeling developing over years to decades, leading to a phenotype characterized by asymmetric ventricular hypertrophy, scattered fibrosis and myofibrillar/cellular disarray with ultimate mechanical heart failure and/or severe arrhythmias. The identity of the primary mutation-induced changes in sarcomere function and how they trigger debilitating remodeling are poorly understood. Support for the importance of mutation-induced hypercontractility, e.g., increased calcium sensitivity and/or increased power output, has been strengthened in recent years. However, other ideas that mutation-induced hypocontractility or non-uniformities with contractile instabilities, instead, constitute primary triggers cannot yet be discarded. Here, we review evidence for and criticism against the mentioned hypotheses. In this process, we find support for previous ideas that inefficient energy usage and a blunted Frank-Starling mechanism have central roles in pathogenesis, although presumably representing effects secondary to the primary mutation-induced changes. While first trying to reconcile apparently diverging evidence for the different hypotheses in one unified model, we also identify key remaining questions and suggest how experimental systems that are built around isolated primarily expressed proteins could be useful.
Collapse
Affiliation(s)
| | | | - Alf Månsson
- Department of Chemistry and Biomedical Sciences, Faculty of Health and Life Sciences, Linnaeus University, SE-39182 Kalmar, Sweden; (M.U.); (L.M.)
| |
Collapse
|
40
|
Heo JH, Lee SR, Jo SL, Yang H, Lee HW, Hong EJ. Letrozole Accelerates Metabolic Remodeling through Activation of Glycolysis in Cardiomyocytes: A Role beyond Hormone Regulation. Int J Mol Sci 2022; 23:ijms23010547. [PMID: 35008972 PMCID: PMC8745349 DOI: 10.3390/ijms23010547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/23/2021] [Accepted: 01/01/2022] [Indexed: 02/04/2023] Open
Abstract
Estrogen receptor-positive (ER+) breast cancer patients are recommended hormone therapy as a primary adjuvant treatment after surgery. Aromatase inhibitors (AIs) are widely administered to ER+ breast cancer patients as estrogen blockers; however, their safety remains controversial. The use of letrozole, an AI, has been reported to cause adverse cardiovascular effects. We aimed to elucidate the effects of letrozole on the cardiovascular system. Female rats exposed to letrozole for four weeks showed metabolic changes, i.e., decreased fatty acid oxidation, increased glycolysis, and hypertrophy in the left ventricle. Although lipid oxidation yields more ATP than carbohydrate metabolism, the latter predominates in the heart under pathological conditions. Reduced lipid metabolism is attributed to reduced β-oxidation due to low circulating estrogen levels. In letrozole-treated rats, glycolysis levels were found to be increased in the heart. Furthermore, the levels of glycolytic enzymes were increased (in a high glucose medium) and the glycolytic rate was increased in vitro (H9c2 cells); the same was not true in the case of estrogen treatment. Reduced lipid metabolism and increased glycolysis can lower energy supply to the heart, resulting in predisposition to heart failure. These data suggest that a letrozole-induced cardiac metabolic remodeling, i.e., a shift from β-oxidation to glycolysis, may induce cardiac structural remodeling.
Collapse
Affiliation(s)
- Jun H. Heo
- College of Veterinary Medicine, Chungnam National University, Daejeon 34134, Korea; (J.H.H.); (S.R.L.); (S.L.J.)
| | - Sang R. Lee
- College of Veterinary Medicine, Chungnam National University, Daejeon 34134, Korea; (J.H.H.); (S.R.L.); (S.L.J.)
| | - Seong Lae Jo
- College of Veterinary Medicine, Chungnam National University, Daejeon 34134, Korea; (J.H.H.); (S.R.L.); (S.L.J.)
| | - Hyun Yang
- KM Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, Korea; (H.Y.); (H.W.L.)
| | - Hye Won Lee
- KM Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, Korea; (H.Y.); (H.W.L.)
| | - Eui-Ju Hong
- College of Veterinary Medicine, Chungnam National University, Daejeon 34134, Korea; (J.H.H.); (S.R.L.); (S.L.J.)
- Correspondence:
| |
Collapse
|
41
|
Pereira JDJ, Ikegami RN, Kawakami JT, Garavelo SM, Reis MM, Palomino SAP, Mangini S, Moreno CR, de Barros SF, Souza AR, Higuchi MDL. Distinct Microbial Communities in Dilated Cardiomyopathy Explanted Hearts Are Associated With Different Myocardial Rejection Outcomes. Front Cell Infect Microbiol 2021; 11:732276. [PMID: 34912727 PMCID: PMC8668412 DOI: 10.3389/fcimb.2021.732276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 11/05/2021] [Indexed: 11/27/2022] Open
Abstract
Background Idiopathic dilated cardiomyopathy (IDCM) myocardial inflammation may be associated with external triggering factors such as infectious agents. Here, we searched if moderate/severe heart transplantation rejection is related to the presence of myocardial inflammation in IDCM explanted hearts, associated with microbial communities. Method Receptor myocardial samples from 18 explanted hearts were separated into groups according to post-transplant outcome: persistent moderate rejection (PMR; n = 6), moderate rejection (MR; n = 7) that regressed after pulse therapy, and no rejection (NR; n = 5)/light intensity rejection. Inflammation was quantified through immunohistochemistry (IHC), and infectious agents were evaluated by IHC, molecular biology, in situ hybridization technique, and transmission electron microscopy (TEM). Results NR presented lower numbers of macrophages, as well as B cells (p = 0.0001), and higher HLA class II expression (p ≤ 0.0001). PMR and MR showed higher levels of Mycoplasma pneumoniae (p = 0.003) and hepatitis B core (p = 0.0009) antigens. NR presented higher levels of parvovirus B19 (PVB19) and human herpes virus 6 (HHV6) and a positive correlation between Borrelia burgdorferi (Bb) and enterovirus genes. Molecular biology demonstrated the presence of M. pneumoniae, Bb, HHV6, and PVB19 genes in all studied groups. TEM revealed structures compatible with the cited microorganisms. Conclusions This initial study investigating on infectious agents and inflammation in the IDCM explanted hearts showed that the association between M. pneumoniae and hepatitis B core was associated with a worse outcome after HT, represented by MR and PMR, suggesting that different IDCM microbial communities may be contributing to post-transplant myocardial rejection.
Collapse
Affiliation(s)
- Jaqueline de Jesus Pereira
- Instituto do Coração (InCor), Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil.,Laboratório de Patologia Cardíaca, Departamento de Patologia, Instituto do Coração (InCor), Universidade de São Paulo, São Paulo, Brazil
| | - Renata Nishiyama Ikegami
- Instituto do Coração (InCor), Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil.,Laboratório de Patologia Cardíaca, Departamento de Patologia, Instituto do Coração (InCor), Universidade de São Paulo, São Paulo, Brazil
| | - Joyce Tiyeko Kawakami
- Instituto do Coração (InCor), Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil.,Laboratório de Patologia Cardíaca, Departamento de Patologia, Instituto do Coração (InCor), Universidade de São Paulo, São Paulo, Brazil
| | - Shérrira Menezes Garavelo
- Instituto do Coração (InCor), Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil.,Laboratório de Patologia Cardíaca, Departamento de Patologia, Instituto do Coração (InCor), Universidade de São Paulo, São Paulo, Brazil
| | - Marcia Martins Reis
- Instituto do Coração (InCor), Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil.,Laboratório de Patologia Cardíaca, Departamento de Patologia, Instituto do Coração (InCor), Universidade de São Paulo, São Paulo, Brazil
| | - Suely Aparecida Pinheiro Palomino
- Instituto do Coração (InCor), Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil.,Laboratório de Patologia Cardíaca, Departamento de Patologia, Instituto do Coração (InCor), Universidade de São Paulo, São Paulo, Brazil
| | - Sandrigo Mangini
- Instituto do Coração (InCor), Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Camila Rodrigues Moreno
- Instituto do Coração (InCor), Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil.,Laboratório de Patologia Cardíaca, Departamento de Patologia, Instituto do Coração (InCor), Universidade de São Paulo, São Paulo, Brazil
| | - Samar Freschi de Barros
- Instituto do Coração (InCor), Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Aline Rodrigues Souza
- Instituto do Coração (InCor), Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil.,Laboratório de Patologia Cardíaca, Departamento de Patologia, Instituto do Coração (InCor), Universidade de São Paulo, São Paulo, Brazil
| | - Maria de Lourdes Higuchi
- Instituto do Coração (InCor), Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil.,Laboratório de Patologia Cardíaca, Departamento de Patologia, Instituto do Coração (InCor), Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
42
|
Wang Y, Sun X, Sun X. The Functions of LncRNA H19 in the Heart. Heart Lung Circ 2021; 31:341-349. [PMID: 34840062 DOI: 10.1016/j.hlc.2021.10.022] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 10/20/2021] [Accepted: 10/27/2021] [Indexed: 10/19/2022]
Abstract
Cardiovascular diseases (CVDs) are major causes of morbidity and mortality worldwide. Great effort has been put into exploring early diagnostic biomarkers and innovative therapeutic strategies for preventing CVD progression over the last two decades. Long non-coding RNAs (lncRNAs) have been identified as novel regulators in cardiac development and cardiac pathogenesis. For example, lncRNA H19 (H19), also known as a fetal gene abundant in adult heart and skeletal muscles and evolutionarily conserved in humans and mice, has a regulatory role in aortic aneurysm, myocardial hypertrophy, extracellular matrix reconstitution, and coronary artery diseases. Yet, the exact function of H19 in the heart remains unknown. This review summarises the functions of H19 in the heart and discusses the challenges and possible strategies of H19 research for cardiovascular disease.
Collapse
Affiliation(s)
- Yao Wang
- Shandong Institute of Endocrine and Metabolic Diseases, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Xiaojing Sun
- Department of Geriatrics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Xianglan Sun
- Department of Geriatrics, Department of Geriatric Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China.
| |
Collapse
|
43
|
Barrick SK, Greenberg MJ. Cardiac myosin contraction and mechanotransduction in health and disease. J Biol Chem 2021; 297:101297. [PMID: 34634306 PMCID: PMC8559575 DOI: 10.1016/j.jbc.2021.101297] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 10/06/2021] [Accepted: 10/07/2021] [Indexed: 12/17/2022] Open
Abstract
Cardiac myosin is the molecular motor that powers heart contraction by converting chemical energy from ATP hydrolysis into mechanical force. The power output of the heart is tightly regulated to meet the physiological needs of the body. Recent multiscale studies spanning from molecules to tissues have revealed complex regulatory mechanisms that fine-tune cardiac contraction, in which myosin not only generates power output but also plays an active role in its regulation. Thus, myosin is both shaped by and actively involved in shaping its mechanical environment. Moreover, these studies have shown that cardiac myosin-generated tension affects physiological processes beyond muscle contraction. Here, we review these novel regulatory mechanisms, as well as the roles that myosin-based force generation and mechanotransduction play in development and disease. We describe how key intra- and intermolecular interactions contribute to the regulation of myosin-based contractility and the role of mechanical forces in tuning myosin function. We also discuss the emergence of cardiac myosin as a drug target for diseases including heart failure, leading to the discovery of therapeutics that directly tune myosin contractility. Finally, we highlight some of the outstanding questions that must be addressed to better understand myosin's functions and regulation, and we discuss prospects for translating these discoveries into precision medicine therapeutics targeting contractility and mechanotransduction.
Collapse
Affiliation(s)
- Samantha K Barrick
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Michael J Greenberg
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, Missouri, USA.
| |
Collapse
|
44
|
Das A, Kelly C, Teh I, Nguyen C, Brown LAE, Chowdhary A, Jex N, Thirunavukarasu S, Sharrack N, Gorecka M, Swoboda PP, Greenwood JP, Kellman P, Moon JC, Davies RH, Lopes LR, Joy G, Plein S, Schneider JE, Dall'Armellina E. Phenotyping hypertrophic cardiomyopathy using cardiac diffusion magnetic resonance imaging: the relationship between microvascular dysfunction and microstructural changes. Eur Heart J Cardiovasc Imaging 2021; 23:352-362. [PMID: 34694365 PMCID: PMC8863073 DOI: 10.1093/ehjci/jeab210] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 10/16/2021] [Indexed: 01/05/2023] Open
Abstract
Aims Microvascular dysfunction in hypertrophic cardiomyopathy (HCM) is predictive of clinical decline, however underlying mechanisms remain unclear. Cardiac diffusion tensor imaging (cDTI) allows in vivo characterization of myocardial microstructure by quantifying mean diffusivity (MD), fractional anisotropy (FA) of diffusion, and secondary eigenvector angle (E2A). In this cardiac magnetic resonance (CMR) study, we examine associations between perfusion and cDTI parameters to understand the sequence of pathophysiology and the interrelation between vascular function and underlying microstructure. Methods and results Twenty HCM patients underwent 3.0T CMR which included: spin-echo cDTI, adenosine stress and rest perfusion mapping, cine-imaging, and late gadolinium enhancement (LGE). Ten controls underwent cDTI. Myocardial perfusion reserve (MPR), MD, FA, E2A, and wall thickness were calculated per segment and further divided into subendocardial (inner 50%) and subepicardial (outer 50%) regions. Segments with wall thickness ≤11 mm, MPR ≥2.2, and no visual LGE were classified as ‘normal’. Compared to controls, ‘normal’ HCM segments had increased MD (1.61 ± 0.09 vs. 1.46 ± 0.07 × 10−3 mm2/s, P = 0.02), increased E2A (60 ± 9° vs. 38 ± 12°, P < 0.001), and decreased FA (0.29 ± 0.04 vs. 0.35 ± 0.02, P = 0.002). Across all HCM segments, subendocardial regions had higher MD and lower MPR than subepicardial (MDendo 1.61 ± 0.08 × 10−3 mm2/s vs. MDepi 1.56 ± 0.18 × 10−3 mm2/s, P = 0.003, MPRendo 1.85 ± 0.83, MPRepi 2.28 ± 0.87, P < 0.0001). Conclusion In HCM patients, even in segments with normal wall thickness, normal perfusion, and no scar, diffusion is more isotropic than in controls, suggesting the presence of underlying cardiomyocyte disarray. Increased E2A suggests the myocardial sheetlets adopt hypercontracted angulation in systole. Increased MD, most notably in the subendocardium, is suggestive of regional remodelling which may explain the reduced subendocardial blood flow.
Collapse
Affiliation(s)
- Arka Das
- Biomedical Imaging Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds Teaching Hospitals NHS Trust, Leeds LS2 9JT, UK
| | - Christopher Kelly
- Biomedical Imaging Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds Teaching Hospitals NHS Trust, Leeds LS2 9JT, UK
| | - Irvin Teh
- Biomedical Imaging Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds Teaching Hospitals NHS Trust, Leeds LS2 9JT, UK
| | - Christopher Nguyen
- Cardiovascular Research Center, Massachusetts General Hospital, Charlestown, 55 Fruit St, Boston, MA 02114, USA.,A. A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, 55 Fruit St, Boston, MA 02114, USA.,Department of Medicine, Harvard Medical School, 25 Shattuck St, Boston, MA 02115, USA.,Biomedical Imaging Research Institute, Cedars-Sinai Medical Centre, 116 N Robertson Blvd, Los Angeles, CA 90048, USA
| | - Louise A E Brown
- Biomedical Imaging Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds Teaching Hospitals NHS Trust, Leeds LS2 9JT, UK
| | - Amrit Chowdhary
- Biomedical Imaging Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds Teaching Hospitals NHS Trust, Leeds LS2 9JT, UK
| | - Nicholas Jex
- Biomedical Imaging Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds Teaching Hospitals NHS Trust, Leeds LS2 9JT, UK
| | - Sharmaine Thirunavukarasu
- Biomedical Imaging Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds Teaching Hospitals NHS Trust, Leeds LS2 9JT, UK
| | - Noor Sharrack
- Biomedical Imaging Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds Teaching Hospitals NHS Trust, Leeds LS2 9JT, UK
| | - Miroslawa Gorecka
- Biomedical Imaging Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds Teaching Hospitals NHS Trust, Leeds LS2 9JT, UK
| | - Peter P Swoboda
- Biomedical Imaging Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds Teaching Hospitals NHS Trust, Leeds LS2 9JT, UK
| | - John P Greenwood
- Biomedical Imaging Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds Teaching Hospitals NHS Trust, Leeds LS2 9JT, UK
| | - Peter Kellman
- National Heart, Lung, and Blood Institute, National Institutes of Health, DHHS, 31 Center Dr, Bethesda, MD 20892, USA
| | - James C Moon
- Barts Heart Centre, The Cardiovascular Magnetic Resonance Imaging Unit and The Inherited Cardiovascular Diseases Unit, St Bartholomew's Hospital, West Smithfield, London EC1A 7BE, UK
| | - Rhodri H Davies
- Barts Heart Centre, The Cardiovascular Magnetic Resonance Imaging Unit and The Inherited Cardiovascular Diseases Unit, St Bartholomew's Hospital, West Smithfield, London EC1A 7BE, UK
| | - Luis R Lopes
- Barts Heart Centre, The Cardiovascular Magnetic Resonance Imaging Unit and The Inherited Cardiovascular Diseases Unit, St Bartholomew's Hospital, West Smithfield, London EC1A 7BE, UK.,Centre for Heart Muscle Disease, Institute of Cardiovascular Science, University College London, Gower St, London WC1E 6BT, UK
| | - George Joy
- Barts Heart Centre, The Cardiovascular Magnetic Resonance Imaging Unit and The Inherited Cardiovascular Diseases Unit, St Bartholomew's Hospital, West Smithfield, London EC1A 7BE, UK
| | - Sven Plein
- Biomedical Imaging Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds Teaching Hospitals NHS Trust, Leeds LS2 9JT, UK
| | - Jürgen E Schneider
- Biomedical Imaging Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds Teaching Hospitals NHS Trust, Leeds LS2 9JT, UK
| | - Erica Dall'Armellina
- Biomedical Imaging Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds Teaching Hospitals NHS Trust, Leeds LS2 9JT, UK
| |
Collapse
|
45
|
Crocini C, Gotthardt M. Cardiac sarcomere mechanics in health and disease. Biophys Rev 2021; 13:637-652. [PMID: 34745372 PMCID: PMC8553709 DOI: 10.1007/s12551-021-00840-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 08/27/2021] [Indexed: 12/23/2022] Open
Abstract
The sarcomere is the fundamental structural and functional unit of striated muscle and is directly responsible for most of its mechanical properties. The sarcomere generates active or contractile forces and determines the passive or elastic properties of striated muscle. In the heart, mutations in sarcomeric proteins are responsible for the majority of genetically inherited cardiomyopathies. Here, we review the major determinants of cardiac sarcomere mechanics including the key structural components that contribute to active and passive tension. We dissect the molecular and structural basis of active force generation, including sarcomere composition, structure, activation, and relaxation. We then explore the giant sarcomere-resident protein titin, the major contributor to cardiac passive tension. We discuss sarcomere dynamics exemplified by the regulation of titin-based stiffness and the titin life cycle. Finally, we provide an overview of therapeutic strategies that target the sarcomere to improve cardiac contraction and filling.
Collapse
Affiliation(s)
- Claudia Crocini
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Neuromuscular and Cardiovascular Cell Biology, Berlin, Germany
- German Center for Cardiovascular Research (DZHK) Partner Site Berlin, Berlin, Germany
- BioFrontiers Institute & Department of Molecular and Cellular Development, University of Colorado Boulder, Boulder, USA
| | - Michael Gotthardt
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Neuromuscular and Cardiovascular Cell Biology, Berlin, Germany
- German Center for Cardiovascular Research (DZHK) Partner Site Berlin, Berlin, Germany
- Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany
| |
Collapse
|
46
|
El-Sayed N, Mostafa YM, AboGresha NM, Ahmed AAM, Mahmoud IZ, El-Sayed NM. Dapagliflozin attenuates diabetic cardiomyopathy through erythropoietin up-regulation of AKT/JAK/MAPK pathways in streptozotocin-induced diabetic rats. Chem Biol Interact 2021; 347:109617. [PMID: 34391751 DOI: 10.1016/j.cbi.2021.109617] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 07/13/2021] [Accepted: 08/04/2021] [Indexed: 12/12/2022]
Abstract
PURPOSE This study was designed to investigate the mechanism of Dapagliflozin (Dapa) cardioprotection against diabetic cardiomyopathy (DCM). Structural and functional changes in the heart as well as decrease of erythropoietin (EPO) levels were reported in DCM. EPO simultaneously activates three pathways: the Janus-activated kinase-signal transducer and activator of transcription (JAK2/STAT5), phosphatidylinositol-3-kinase-Akt (PI3K/Akt), and extracellular signal-related kinase (ERK/MAPK) cascades, that result in proliferation and differentiation of cardiac cells. METHODS AND RESULTS DCM was induced by a high fat diet for 10 weeks followed by administration of streptozotocin. After confirmation of diabetes, rats were divided randomly to 5 groups: Group 1; normal control group, Group 2; untreated diabetic group and Groups (3-5); diabetic groups received Dapa daily (0.75 mg, 1.5 or 3 mg/Kg, p.o) respectively for a month. At the end of the experiment, full anaesthesia was induced in all rats using ether inhalation and ECG was recorded. Blood samples were collected then rats were sacrificed and their heart were dissected out and processed for biochemical and histopathological studies. Untreated diabetic rats showed abnormal ECG pattern, elevation of serum cardiac enzymes, decrease EPO levels, downregulation of P-Akt, P-JAK2 and pMAPK pathways, abnormal histological structure of the heart and increase immunostaining intensity of P53 and TNF α in the cardiomyocytes. Dapa in a dose dependent manner attenuated the alterations in the previously mentioned parameters. CONCLUSION The cardioprotective effect of Dapa could be mediated by increasing EPO levels and activation of P-Akt, P-JAK2 and pMAPK signalling cascades which in turn decrease apoptosis.
Collapse
Affiliation(s)
- Nora El-Sayed
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Sinai University, Kantra Branch, Ismailia, Egypt
| | - Yasser M Mostafa
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Suez Canal University, Ismailia, 41522, Egypt; Department of Pharmacology & Toxicology, Faculty of Pharmacy, Badr University, Badr, Egypt
| | - Noha M AboGresha
- Department of Physiology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Amal A M Ahmed
- Department of Cytology & Histology, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, Egypt
| | - Islam Z Mahmoud
- Department of Cardiovascular Medicine, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Norhan M El-Sayed
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Suez Canal University, Ismailia, 41522, Egypt.
| |
Collapse
|
47
|
Agarwal R, Paulo JA, Toepfer CN, Ewoldt JK, Sundaram S, Chopra A, Zhang Q, Gorham J, DePalma SR, Chen CS, Gygi SP, Seidman CE, Seidman JG. Filamin C Cardiomyopathy Variants Cause Protein and Lysosome Accumulation. Circ Res 2021; 129:751-766. [PMID: 34405687 PMCID: PMC9053646 DOI: 10.1161/circresaha.120.317076] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 08/17/2021] [Indexed: 01/02/2023]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Radhika Agarwal
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Joao A. Paulo
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Christopher N. Toepfer
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
- Radcliffe Department of Medicine, University of Oxford, OX3 9DU, UK
- Wellcome Centre for Human Genetics, University of Oxford, OX3 7BN, UK
| | - Jourdan K. Ewoldt
- Department of Biomedical Engineering, Boston University, Boston, MA 02115, USA
| | - Subramanian Sundaram
- Department of Biomedical Engineering, Boston University, Boston, MA 02115, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Anant Chopra
- Department of Biomedical Engineering, Boston University, Boston, MA 02115, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Qi Zhang
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Joshua Gorham
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Steven R. DePalma
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Christopher S. Chen
- Department of Biomedical Engineering, Boston University, Boston, MA 02115, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Steven P. Gygi
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Christine E. Seidman
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
- Division of Genetics, Department of Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - J. G. Seidman
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
48
|
Aung LHH, Jumbo JCC, Wang Y, Li P. Therapeutic potential and recent advances on targeting mitochondrial dynamics in cardiac hypertrophy: A concise review. MOLECULAR THERAPY. NUCLEIC ACIDS 2021; 25:416-443. [PMID: 34484866 PMCID: PMC8405900 DOI: 10.1016/j.omtn.2021.06.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Pathological cardiac hypertrophy begins as an adaptive response to increased workload; however, sustained hemodynamic stress will lead it to maladaptation and eventually cardiac failure. Mitochondria, being the powerhouse of the cells, can regulate cardiac hypertrophy in both adaptive and maladaptive phases; they are dynamic organelles that can adjust their number, size, and shape through a process called mitochondrial dynamics. Recently, several studies indicate that promoting mitochondrial fusion along with preventing mitochondrial fission could improve cardiac function during cardiac hypertrophy and avert its progression toward heart failure. However, some studies also indicate that either hyperfusion or hypo-fission could induce apoptosis and cardiac dysfunction. In this review, we summarize the recent knowledge regarding the effects of mitochondrial dynamics on the development and progression of cardiac hypertrophy with particular emphasis on the regulatory role of mitochondrial dynamics proteins through the genetic, epigenetic, and post-translational mechanisms, followed by discussing the novel therapeutic strategies targeting mitochondrial dynamic pathways.
Collapse
Affiliation(s)
- Lynn Htet Htet Aung
- Center for Molecular Genetics, Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China.,Center for Bioinformatics, Institute for Translational Medicine, School of Basic Science, College of Medicine, Qingdao University, Qingdao 266021, China
| | - Juan Carlos Cueva Jumbo
- School of Preclinical Medicine, Nanobody Research Center, Guangxi Medical University, Nanning 530021, China
| | - Yin Wang
- Center for Molecular Genetics, Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China
| | - Peifeng Li
- Center for Molecular Genetics, Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China.,Center for Bioinformatics, Institute for Translational Medicine, School of Basic Science, College of Medicine, Qingdao University, Qingdao 266021, China
| |
Collapse
|
49
|
Abstract
Cardiac hypertrophy, characterized by the enlargement of cardiomyocytes, is initially an adaptive response to physiological and pathological stimuli. Decompensated cardiac hypertrophy is related to fibrosis, inflammatory cytokine, maladaptive remodeling, and heart failure. Although pathological myocardial hypertrophy is the main cause of hypertrophy-related morbidity and mortality, our understanding of its mechanism is still poor. Long noncoding RNAs (lncRNAs) are noncoding RNAs that regulate various physiological and pathological processes through multiple molecular mechanisms. Recently, accumulating evidence has indicated that lncRNA-H19 is a potent regulator of the progression of cardiac hypertrophy. For the first time, this review summarizes the current studies about the role of lncRNA-H19 in cardiac hypertrophy, including its pathophysiological processes and underlying pathological mechanism, including calcium regulation, fibrosis, apoptosis, angiogenesis, inflammation, and methylation. The context within which lncRNA-H19 might be developed as a target for cardiac hypertrophy treatment is then discussed to gain better insight into the possible biological functions of lncRNA-H19 in cardiac hypertrophy.
Collapse
|
50
|
Mutation in FBXO32 causes dilated cardiomyopathy through up-regulation of ER-stress mediated apoptosis. Commun Biol 2021; 4:884. [PMID: 34272480 PMCID: PMC8285540 DOI: 10.1038/s42003-021-02391-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 06/23/2021] [Indexed: 02/06/2023] Open
Abstract
Endoplasmic reticulum (ER) stress induction of cell death is implicated in cardiovascular diseases. Sustained activation of ER-stress induces the unfolded protein response (UPR) pathways, which in turn activate three major effector proteins. We previously reported a missense homozygous mutation in FBXO32 (MAFbx, Atrogin-1) causing advanced heart failure by impairing autophagy. In the present study, we performed transcriptional profiling and biochemical assays, which unexpectedly revealed a reduced activation of UPR effectors in patient mutant hearts, while a strong up-regulation of the CHOP transcription factor and of its target genes are observed. Expression of mutant FBXO32 in cells is sufficient to induce CHOP-associated apoptosis, to increase the ATF2 transcription factor and to impair ATF2 ubiquitination. ATF2 protein interacts with FBXO32 in the human heart and its expression is especially high in FBXO32 mutant hearts. These findings provide a new underlying mechanism for FBXO32-mediated cardiomyopathy, implicating abnormal activation of CHOP. These results suggest alternative non-canonical pathways of CHOP activation that could be considered to develop new therapeutic targets for the treatment of FBXO32-associated DCM. Al-Yacoub et al. investigate the consequences of FBXO32 mutation on dilated cardiomyopathy. ER stress, abnormal CHOP activation and CHOP-induced apoptosis with no UPR effector activation are found to underlie the FBXO32 mutation induced cardiomyopathy, suggesting an alternative pathway that can be considered to develop new therapeutic targets for its treatment.
Collapse
|