1
|
Zervou S, McAndrew DJ, Lake HA, Kuznecova E, Preece C, Davies B, Neubauer S, Lygate CA. Cardiac function and energetics in mice with combined genetic augmentation of creatine and creatine kinase activity. J Mol Cell Cardiol 2024; 196:105-114. [PMID: 39276853 DOI: 10.1016/j.yjmcc.2024.09.007] [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: 07/12/2024] [Revised: 08/23/2024] [Accepted: 09/10/2024] [Indexed: 09/17/2024]
Abstract
Improving energy provision in the failing heart by augmenting the creatine kinase (CK) system is a desirable therapeutic target. However, over-expression of the creatine transporter (CrT-OE) has shown that very high creatine levels result in cardiac hypertrophy and dysfunction. We hypothesise this is due to insufficient endogenous CK activity to maintain thermodynamically favourable metabolite ratios. If correct, then double transgenic mice (dTg) overexpressing both CrT and the muscle isoform of CK (CKM-OE) would rescue the adverse phenotype. In Study 1, overexpressing lines were crossed and cardiac function assessed by invasive haemodynamics and echocardiography. This demonstrated that CKM-OE was safe, but too few hearts had creatine in the toxic range. In Study 2, a novel CrT-OE line was generated with higher, homogeneous, creatine levels and phenotyped as before. Myocardial creatine was 4-fold higher in CrT-OE and dTg hearts compared to wildtype and was associated with hypertrophy and contractile dysfunction. The inability of dTg hearts to rescue this phenotype was attributed to downregulation of CK activity, as occurs in the failing heart. Nevertheless, combining both studies in a linear regression analysis suggests a modest positive effect of CKM over a range of creatine concentrations. In conclusion, we confirm that moderate elevation of creatine is well tolerated, but very high levels are detrimental. Correlation analysis lends support to the theory that this may be a consequence of limited CK activity. Future studies should focus on preventing CKM downregulation to unlock the potential synergy of augmenting both creatine and CK in the heart.
Collapse
Affiliation(s)
- Sevasti Zervou
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, British Heart Foundation Centre for Research Excellence, University of Oxford, Oxford, UK; Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Debra J McAndrew
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, British Heart Foundation Centre for Research Excellence, University of Oxford, Oxford, UK; Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Hannah A Lake
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, British Heart Foundation Centre for Research Excellence, University of Oxford, Oxford, UK; Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Elina Kuznecova
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, British Heart Foundation Centre for Research Excellence, University of Oxford, Oxford, UK; Centre for Human Genetics, University of Oxford, Oxford, UK
| | | | | | - Stefan Neubauer
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, British Heart Foundation Centre for Research Excellence, University of Oxford, Oxford, UK
| | - Craig A Lygate
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, British Heart Foundation Centre for Research Excellence, University of Oxford, Oxford, UK; Centre for Human Genetics, University of Oxford, Oxford, UK; School of Cardiovascular and Metabolic Health, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK.
| |
Collapse
|
2
|
Davenport A, Kessinger CW, Pfeiffer RD, Shah N, Xu R, Abel ED, Tucker NR, Lin Z. Comparative analysis of two independent Myh6-Cre transgenic mouse lines. JOURNAL OF MOLECULAR AND CELLULAR CARDIOLOGY PLUS 2024; 9:100081. [PMID: 39323506 PMCID: PMC11423776 DOI: 10.1016/j.jmccpl.2024.100081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/27/2024]
Abstract
We have previously shown that the Myh6 promoter drives Cre expression in a subset of male germ line cells in three independent Myh6-Cre mouse lines, including two transgenic lines and one knock-in allele. In this study, we further compared the tissue-specificity of the two Myh6-Cre transgenic mouse lines, MDS Myh6-Cre and AUTR Myh6-Cre, through examining the expression of tdTomato (tdTom) red fluorescence protein in multiple internal organs, including the heart, brain, liver, lung, pancreas and brown adipose tissue. Our results show that MDS Myh6-Cre mainly activates tdTom reporter in the heart, whereas AUTR Myh6-Cre activates tdTom expression significantly in the heart, and in the cells of liver, pancreas and brain. In the heart, similar to MDS Myh6-Cre, AUTR Myh6-Cre activates tdTom in most cardiomyocytes. In the other organs, AUTR Myh6-Cre not only mosaically activates tdTom in some parenchymal cells, such as hepatocytes in the liver and neurons in the brain, but also turns on tdTom in some interstitial cells of unknown identity.
Collapse
Affiliation(s)
- Amanda Davenport
- Department of Biomedical Research and Translational Medicine, Masonic Medical Research Institute, 2150 Bleecker Street, Utica, NY 13501, United States of America
| | - Chase W. Kessinger
- Department of Biomedical Research and Translational Medicine, Masonic Medical Research Institute, 2150 Bleecker Street, Utica, NY 13501, United States of America
| | - Ryan D. Pfeiffer
- Department of Biomedical Research and Translational Medicine, Masonic Medical Research Institute, 2150 Bleecker Street, Utica, NY 13501, United States of America
| | - Nikita Shah
- Department of Biomedical Research and Translational Medicine, Masonic Medical Research Institute, 2150 Bleecker Street, Utica, NY 13501, United States of America
- College of Arts and Sciences, SUNY Polytechnic Institute, Utica, NY 13502, United States of America
| | - Richard Xu
- Department of Biomedical Research and Translational Medicine, Masonic Medical Research Institute, 2150 Bleecker Street, Utica, NY 13501, United States of America
| | - E. Dale Abel
- Department of Medicine David Geffen School of Medicine and UCLA Health, United States of America
| | - Nathan R. Tucker
- Department of Biomedical Research and Translational Medicine, Masonic Medical Research Institute, 2150 Bleecker Street, Utica, NY 13501, United States of America
| | - Zhiqiang Lin
- Department of Biomedical Research and Translational Medicine, Masonic Medical Research Institute, 2150 Bleecker Street, Utica, NY 13501, United States of America
| |
Collapse
|
3
|
Han YS, Pakkam M, Fogarty MJ, Sieck GC, Brozovich FV. Alterations in cardiac contractile and regulatory proteins contribute to age-related cardiac dysfunction in male rats. Physiol Rep 2024; 12:e70012. [PMID: 39169429 PMCID: PMC11338742 DOI: 10.14814/phy2.70012] [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: 03/26/2024] [Revised: 08/06/2024] [Accepted: 08/07/2024] [Indexed: 08/23/2024] Open
Abstract
Aging is associated with cardiac contractile abnormalities, but the etiology of these contractile deficits is unclear. We hypothesized that cardiac contractile and regulatory protein expression is altered during aging. To investigate this possibility, left ventricular (LV) lysates were prepared from young (6 months) and old (24 months) Fischer344 rats. There are no age-related changes in SERCA2 expression or phospholamban phosphorylation. Additionally, neither titin isoform expression nor phosphorylation differed. However, there is a significant increase in β-isoform of the myosin heavy chain (MyHC) expression and phosphorylation of TnI and MyBP-C during aging. In permeabilized strips of papillary muscle, force and Ca2+ sensitivity are reduced during aging, consistent with the increase in β-MyHC expression and TnI phosphorylation. However, the increase in MyBP-C phosphorylation during aging may represent a mechanism to compensate for age-related contractile deficits. In isolated cardiomyocytes loaded with Fura-2, the peak of the Ca2+ transient is reduced, but the kinetics of the Ca2+ transient are not altered. Furthermore, the extent of shortening and the rates of both sarcomere shortening and re-lengthening are reduced. These results demonstrate that aging is associated with changes in contractile and regulatory protein expression and phosphorylation, which affect the mechanical properties of cardiac muscle.
Collapse
Affiliation(s)
- Young Soo Han
- Department of Physiology & Biomedical EngineeringMayo ClinicRochesterMinnesotaUSA
| | - Madona Pakkam
- Department of Physiology & Biomedical EngineeringMayo ClinicRochesterMinnesotaUSA
| | - Matthew J. Fogarty
- Department of Physiology & Biomedical EngineeringMayo ClinicRochesterMinnesotaUSA
| | - Gary C. Sieck
- Department of Physiology & Biomedical EngineeringMayo ClinicRochesterMinnesotaUSA
| | - Frank V. Brozovich
- Department of Physiology & Biomedical EngineeringMayo ClinicRochesterMinnesotaUSA
- Department of Cardiovascular DiseasesMayo ClinicRochesterMinnesotaUSA
| |
Collapse
|
4
|
Wu Q, Zou S, Liu W, Liang M, Chen Y, Chang J, Liu Y, Yu X. A novel onco-cardiological mouse model of lung cancer-induced cardiac dysfunction and its application in identifying potential roles of tRNA-derived small RNAs. Biomed Pharmacother 2023; 165:115117. [PMID: 37406509 DOI: 10.1016/j.biopha.2023.115117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 06/28/2023] [Accepted: 07/01/2023] [Indexed: 07/07/2023] Open
Abstract
An increasing body of research suggests cancer-induced cardiovascular diseases, leading to the appearance of an interdisciplinary study known as onco-cardiology. Lung cancer has the highest incidence and mortality. Cardiac dysfunction constitutes a major cause of death in lung cancer patients. However, its mechanism has not been elucidated because suitable animal models that adequately mimic clinical features are lacking. Here, we established a novel chemically induced lung cancer mouse model using benzo[a]pyrene and urethane to recapitulate the general characteristics of cardiac dysfunction caused by lung cancer, the cardiac disorders in the context of the progression of lung cancer were evaluated using echocardiographic and histological approaches. The pathological changes included myocardial ischaemia, pericarditis, cardiac pre-cachexia, and pulmonary artery hypertension. We performed sequencing to detect the tRNA-derived fragments and tRNA-derived stress-induced RNAs (tRFs/tiRNAs) expressions in mouse heart tissue. 22 upregulated and 16 downregulated tRFs/tiRNAs were identified. Subsequently, the top 10 significant results of Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis were presented. The in vitro model was established by exposing neonatal rat cardiomyocytes and myocardial fibroblasts to lung tumour cell-conditioned medium, respectively. Western blotting revealed significant changes in cardiac failure markers (atrial natriuretic peptide and α-myosin heavy chain) and cardiac fibrosis markers (Collagen-1 and Collagen-3). Our model adequately reflects the pathological features of lung cancer-induced cardiac dysfunction. Furthermore, the altered tRF/tiRNA profiles showed great promise as novel targets for therapies. These results might pave the way for research on therapeutic targets in onco-cardiology.
Collapse
Affiliation(s)
- Qian Wu
- Department of Pharmacology, the Municipal & Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, National Medical Products Administration & State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, PR China
| | - Shiting Zou
- Department of Pharmacology, the Municipal & Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, National Medical Products Administration & State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, PR China
| | - Wanjie Liu
- Department of Pharmacology, the Municipal & Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, National Medical Products Administration & State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, PR China
| | - Miao Liang
- Department of Pharmacology, the Municipal & Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, National Medical Products Administration & State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, PR China
| | - Yuling Chen
- Department of Pharmacology, the Municipal & Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, National Medical Products Administration & State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, PR China
| | - Jishuo Chang
- Department of Pharmacology, the Municipal & Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, National Medical Products Administration & State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, PR China
| | - Yinghua Liu
- Department of Pharmacology, the Municipal & Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, National Medical Products Administration & State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, PR China.
| | - Xiyong Yu
- Department of Pharmacology, the Municipal & Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, National Medical Products Administration & State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, PR China.
| |
Collapse
|
5
|
Petersen M, Schmiedel N, Dierck F, Hille S, Remes A, Senger F, Schmidt I, Lüllmann-Rauch R, Müller OJ, Frank D, Rangrez AY, Frey N, Kuhn C. Fibin regulates cardiomyocyte hypertrophy and causes protein-aggregate-associated cardiomyopathy in vivo. Front Mol Biosci 2023; 10:1169658. [PMID: 37342207 PMCID: PMC10278231 DOI: 10.3389/fmolb.2023.1169658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 05/16/2023] [Indexed: 06/22/2023] Open
Abstract
Despite the identification of numerous molecular pathways modulating cardiac hypertrophy its pathogenesis is not completely understood. In this study we define an unexpected role for Fibin ("fin bud initiation factor homolog") in cardiomyocyte hypertrophy. Via gene expression profiling in hypertrophic murine hearts after transverse aortic constriction we found a significant induction of Fibin. Moreover, Fibin was upregulated in another mouse model of cardiac hypertrophy (calcineurin-transgenics) as well as in patients with dilated cardiomyopathy. Immunoflourescence microscopy revealed subcellular localization of Fibin at the sarcomeric z-disc. Overexpression of Fibin in neonatal rat ventricular cardiomyocytes revealed a strong anti-hypertrophic effect through inhibiting both, NFAT- and SRF-dependent signalling. In contrast, transgenic mice with cardiac-restricted overexpression of Fibin developed dilated cardiomyopathy, accompanied by induction of hypertrophy-associated genes. Moreover, Fibin overexpression accelerated the progression to heart failure in the presence of prohypertrophic stimuli such as pressure overload and calcineurin overexpression. Histological and ultrastructural analyses surprisingly showed large protein aggregates containing Fibin. On the molecular level, aggregate formation was accompanied by an induction of the unfolded protein response subsequent UPR-mediated apoptosis and autophagy. Taken together, we identified Fibin as a novel potent negative regulator of cardiomyocyte hypertrophy in vitro. Yet, heart-specific Fibin overexpression in vivo causes development of a protein-aggregate-associated cardiomyopathy. Because of close similarities to myofibrillar myopathies, Fibin represents a candidate gene for cardiomyopathy and Fibin transgenic mice may provide additional mechanistic insight into aggregate formation in these diseases.
Collapse
Affiliation(s)
- Matthias Petersen
- Department of Internal Medicine III, University Medical Center of Schleswig-Holstein, Campus Kiel, Kiel, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Kiel, Germany
| | - Nesrin Schmiedel
- Department of Internal Medicine III, University Medical Center of Schleswig-Holstein, Campus Kiel, Kiel, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Kiel, Germany
| | - Franziska Dierck
- Department of Internal Medicine III, University Medical Center of Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Susanne Hille
- Department of Internal Medicine III, University Medical Center of Schleswig-Holstein, Campus Kiel, Kiel, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Kiel, Germany
| | - Anca Remes
- Department of Internal Medicine III, University Medical Center of Schleswig-Holstein, Campus Kiel, Kiel, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Kiel, Germany
| | - Frauke Senger
- Department of Internal Medicine III, University Medical Center of Schleswig-Holstein, Campus Kiel, Kiel, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Kiel, Germany
| | - Inga Schmidt
- Department of Internal Medicine III, University Medical Center of Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | | | - Oliver J. Müller
- Department of Internal Medicine III, University Medical Center of Schleswig-Holstein, Campus Kiel, Kiel, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Kiel, Germany
| | - Derk Frank
- Department of Internal Medicine III, University Medical Center of Schleswig-Holstein, Campus Kiel, Kiel, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Kiel, Germany
| | - Ashraf Y. Rangrez
- Department of Cardiology, Angiology and Pneumology, Heidelberg University Hospital, Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Heidelberg, Germany
| | - Norbert Frey
- Department of Cardiology, Angiology and Pneumology, Heidelberg University Hospital, Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Heidelberg, Germany
| | - Christian Kuhn
- Department of Internal Medicine III, University Medical Center of Schleswig-Holstein, Campus Kiel, Kiel, Germany
| |
Collapse
|
6
|
Martin M, Gähwiler EKN, Generali M, Hoerstrup SP, Emmert MY. Advances in 3D Organoid Models for Stem Cell-Based Cardiac Regeneration. Int J Mol Sci 2023; 24:ijms24065188. [PMID: 36982261 PMCID: PMC10049446 DOI: 10.3390/ijms24065188] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/03/2023] [Accepted: 03/06/2023] [Indexed: 03/11/2023] Open
Abstract
The adult human heart cannot regain complete cardiac function following tissue injury, making cardiac regeneration a current clinical unmet need. There are a number of clinical procedures aimed at reducing ischemic damage following injury; however, it has not yet been possible to stimulate adult cardiomyocytes to recover and proliferate. The emergence of pluripotent stem cell technologies and 3D culture systems has revolutionized the field. Specifically, 3D culture systems have enhanced precision medicine through obtaining a more accurate human microenvironmental condition to model disease and/or drug interactions in vitro. In this study, we cover current advances and limitations in stem cell-based cardiac regenerative medicine. Specifically, we discuss the clinical implementation and limitations of stem cell-based technologies and ongoing clinical trials. We then address the advent of 3D culture systems to produce cardiac organoids that may better represent the human heart microenvironment for disease modeling and genetic screening. Finally, we delve into the insights gained from cardiac organoids in relation to cardiac regeneration and further discuss the implications for clinical translation.
Collapse
Affiliation(s)
- Marcy Martin
- Institute for Regenerative Medicine (IREM), University of Zurich, 8952 Schlieren, Switzerland
| | - Eric K. N. Gähwiler
- Institute for Regenerative Medicine (IREM), University of Zurich, 8952 Schlieren, Switzerland
| | - Melanie Generali
- Institute for Regenerative Medicine (IREM), University of Zurich, 8952 Schlieren, Switzerland
| | - Simon P. Hoerstrup
- Institute for Regenerative Medicine (IREM), University of Zurich, 8952 Schlieren, Switzerland
- Wyss Zurich Translational Center, University of Zurich and ETH Zurich, 8092 Zurich, Switzerland
| | - Maximilian Y. Emmert
- Institute for Regenerative Medicine (IREM), University of Zurich, 8952 Schlieren, Switzerland
- Wyss Zurich Translational Center, University of Zurich and ETH Zurich, 8092 Zurich, Switzerland
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), 13353 Berlin, Germany
- Department of Cardiovascular Surgery, Charité Universitätsmedizin Berlin, 10117 Berlin, Germany
- Correspondence: ; Tel.: +41-44-634-5610
| |
Collapse
|
7
|
Sadayappan S, Kranias EG. In Memoriam Tribute to Jeffrey Robbins, PhD (1950–2022). Circ Res 2022; 131:870-872. [DOI: 10.1161/circresaha.122.322105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Sakthivel Sadayappan
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, (S.S.), University of Cincinnati College of Medicine, Cincinnati, OH
| | - Evangelia G. Kranias
- Department of Pharmacology and Systems Physiology (E.G.K.), University of Cincinnati College of Medicine, Cincinnati, OH
| |
Collapse
|
8
|
Hart CC, Lee YI, Hammers DW, Sweeney HL. Evaluation of the DBA/2J mouse as a potential background strain for genetic models of cardiomyopathy. JOURNAL OF MOLECULAR AND CELLULAR CARDIOLOGY PLUS 2022; 1:100012. [PMID: 37206988 PMCID: PMC10195103 DOI: 10.1016/j.jmccpl.2022.100012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
The potential use of the D2.mdx mouse (the mdx mutation on the DBA/2J genetic background) as a preclinical model of the cardiac aspects of Duchenne muscular dystrophy (DMD) has been criticized based on speculation that the DBA/2J genetic background displays an inherent hypertrophic cardiomyopathy (HCM) phenotype. Accordingly, the goal of the current study was to further examine the cardiac status of this mouse strain over a 12-month period to determine if observable signs of HCM develop, including histopathology and pathological enlargement of the myocardium. Previous reports have documented heightened TGFβ signaling in the DBA2/J striated muscles, as compared to the C57 background, which, as expected, is manifested as increased cardiomyocyte size, wall thickness, and heart mass as compared to the C57 background. While normalized heart mass is larger in the DBA/2J mice, compared to age-matched C57/BL10 mice, both strains similarly increase in size from 4 to 12 months of age. We also report that DBA/2J mice contain equivalent amounts of left ventricular collagen as healthy canine and human samples. In a longitudinal echocardiography study, neither sedentary nor exercised DBA/2J mice demonstrated left ventricular wall thickening or cardiac functional deficits. In summary, we find no evidence of HCM, nor any other cardiac pathology, and thus propose that it is an appropriate background strain for genetic modeling of cardiac diseases, including the cardiomyopathy associated with DMD.
Collapse
Affiliation(s)
| | | | | | - H. Lee Sweeney
- Corresponding author at: 1200 Newell, Dr. ARB R5-216, Gainesville, FL 32610-0267, United States of America. (H.L. Sweeney)
| |
Collapse
|
9
|
Rashbrook VS, Brash JT, Ruhrberg C. Cre toxicity in mouse models of cardiovascular physiology and disease. NATURE CARDIOVASCULAR RESEARCH 2022; 1:806-816. [PMID: 37692772 PMCID: PMC7615056 DOI: 10.1038/s44161-022-00125-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 07/27/2022] [Indexed: 09/12/2023]
Abstract
The Cre-LoxP system provides a widely used method for studying gene requirements in the mouse as the main mammalian genetic model organism. To define the molecular and cellular mechanisms that underlie cardiovascular development, function and disease, various mouse strains have been engineered that allow Cre-LoxP-mediated gene targeting within specific cell types of the cardiovascular system. Despite the usefulness of this system, evidence is accumulating that Cre activity can have toxic effects in cells, independently of its ability to recombine pairs of engineered LoxP sites in target genes. Here, we have gathered published evidence for Cre toxicity in cells and tissues relevant to cardiovascular biology and provide an overview of mechanisms proposed to underlie Cre toxicity. Based on this knowledge, we propose that each study utilising the Cre-LoxP system to investigate gene function in the cardiovascular system should incorporate appropriate controls to account for Cre toxicity.
Collapse
Affiliation(s)
- Victoria S. Rashbrook
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London EC1V 9EL, UK
| | - James T. Brash
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London EC1V 9EL, UK
| | - Christiana Ruhrberg
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London EC1V 9EL, UK
| |
Collapse
|
10
|
Zink M, Seewald A, Rohrbach M, Brodehl A, Liedtke D, Williams T, Childs SJ, Gerull B. Altered Expression of TMEM43 Causes Abnormal Cardiac Structure and Function in Zebrafish. Int J Mol Sci 2022; 23:9530. [PMID: 36076925 PMCID: PMC9455580 DOI: 10.3390/ijms23179530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 08/17/2022] [Accepted: 08/19/2022] [Indexed: 11/21/2022] Open
Abstract
Arrhythmogenic cardiomyopathy (ACM) is an inherited heart muscle disease caused by heterozygous missense mutations within the gene encoding for the nuclear envelope protein transmembrane protein 43 (TMEM43). The disease is characterized by myocyte loss and fibro-fatty replacement, leading to life-threatening ventricular arrhythmias and sudden cardiac death. However, the role of TMEM43 in the pathogenesis of ACM remains poorly understood. In this study, we generated cardiomyocyte-restricted transgenic zebrafish lines that overexpress eGFP-linked full-length human wild-type (WT) TMEM43 and two genetic variants (c.1073C>T, p.S358L; c.332C>T, p.P111L) using the Tol2-system. Overexpression of WT and p.P111L-mutant TMEM43 was associated with transcriptional activation of the mTOR pathway and ribosome biogenesis, and resulted in enlarged hearts with cardiomyocyte hypertrophy. Intriguingly, mutant p.S358L TMEM43 was found to be unstable and partially redistributed into the cytoplasm in embryonic and adult hearts. Moreover, both TMEM43 variants displayed cardiac morphological defects at juvenile stages and ultrastructural changes within the myocardium, accompanied by dysregulated gene expression profiles in adulthood. Finally, CRISPR/Cas9 mutants demonstrated an age-dependent cardiac phenotype characterized by heart enlargement in adulthood. In conclusion, our findings suggest ultrastructural remodeling and transcriptomic alterations underlying the development of structural and functional cardiac defects in TMEM43-associated cardiomyopathy.
Collapse
Affiliation(s)
- Miriam Zink
- Comprehensive Heart Failure Center, Department of Internal Medicine I, University Hospital Würzburg, 97080 Würzburg, Germany
| | - Anne Seewald
- Comprehensive Heart Failure Center, Department of Internal Medicine I, University Hospital Würzburg, 97080 Würzburg, Germany
| | - Mareike Rohrbach
- Comprehensive Heart Failure Center, Department of Internal Medicine I, University Hospital Würzburg, 97080 Würzburg, Germany
| | - Andreas Brodehl
- Erich and Hanna Klessmann Institute, Heart and Diabetes Center NRW, University Hospital of the Ruhr-University Bochum, 32545 Bad Oeynhausen, Germany
| | - Daniel Liedtke
- Institute for Human Genetics, Biocenter, Julius-Maximilians-University Würzburg, 97074 Würzburg, Germany
| | - Tatjana Williams
- Comprehensive Heart Failure Center, Department of Internal Medicine I, University Hospital Würzburg, 97080 Würzburg, Germany
| | - Sarah J. Childs
- Department of Biochemistry and Molecular Biology, Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Brenda Gerull
- Comprehensive Heart Failure Center, Department of Internal Medicine I, University Hospital Würzburg, 97080 Würzburg, Germany
| |
Collapse
|
11
|
Lin J, Chen Z, Yang L, Liu L, Yue P, Sun Y, Zhao M, Guo X, Hu X, Zhang Y, Zhang H, Li Y, Guo Y, Dong E. Cas9/AAV9-Mediated Somatic Mutagenesis Uncovered the Cell-Autonomous Role of Sarcoplasmic/Endoplasmic Reticulum Calcium ATPase 2 in Murine Cardiomyocyte Maturation. Front Cell Dev Biol 2022; 10:864516. [PMID: 35433671 PMCID: PMC9012521 DOI: 10.3389/fcell.2022.864516] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 03/03/2022] [Indexed: 11/24/2022] Open
Abstract
Sarcoplasmic/endoplasmic reticulum calcium ATPase 2 (SERCA2) is a key player in cardiomyocyte calcium handling and also a classic target in the gene therapy for heart failure. SERCA2 expression dramatically increases during cardiomyocyte maturation in the postnatal phase of heart development, which is essential for the heart to acquire its full function in adults. However, whether and how SERCA2 regulates cardiomyocyte maturation remains unclear. Here, we performed Cas9/AAV9-mediated somatic mutagenesis (CASAAV) in mice and achieved cardiomyocyte-specific knockout of Atp2a2, the gene coding SERCA2. Through a cardiac genetic mosaic analysis, we demonstrated the cell-autonomous role of SERCA2 in building key ultrastructures of mature ventricular cardiomyocytes, including transverse-tubules and sarcomeres. SERCA2 also exerts a profound impact on oxidative respiration gene expression and sarcomere isoform switching from Myh7/Tnni1 to Myh6/Tnni3, which are transcriptional hallmarks of cardiomyocyte maturation. Together, this study uncovered a pivotal role of SERCA2 in heart development and provided new insights about SERCA2-based cardiac gene therapy.
Collapse
Affiliation(s)
- Junsen Lin
- Peking University Health Science Center, School of Basic Medical Sciences, The Institute of Cardiovascular Sciences, Key Laboratory of Molecular Cardiovascular Science of Ministry of Education, Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, China
| | - Zhan Chen
- Peking University Health Science Center, School of Basic Medical Sciences, The Institute of Cardiovascular Sciences, Key Laboratory of Molecular Cardiovascular Science of Ministry of Education, Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, China
| | - Luzi Yang
- Peking University Health Science Center, School of Basic Medical Sciences, The Institute of Cardiovascular Sciences, Key Laboratory of Molecular Cardiovascular Science of Ministry of Education, Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, China
| | - Lei Liu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education (MOE), Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Peng Yue
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education (MOE), Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Yueshen Sun
- State Key Laboratory of Complex Severe and Rare Diseases, Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
- Department of Medical Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Mingming Zhao
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, National Health Commission of China (NHC) Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Cardiovascular Receptors Research. Beijing, China
| | - Xiaoling Guo
- Basic Medical Research Center, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiaomin Hu
- State Key Laboratory of Complex Severe and Rare Diseases, Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
- Department of Medical Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Yan Zhang
- Peking University Health Science Center, School of Basic Medical Sciences, The Institute of Cardiovascular Sciences, Key Laboratory of Molecular Cardiovascular Science of Ministry of Education, Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, China
| | - Hong Zhang
- Peking University Health Science Center, School of Basic Medical Sciences, The Institute of Cardiovascular Sciences, Key Laboratory of Molecular Cardiovascular Science of Ministry of Education, Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, China
| | - Yifei Li
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education (MOE), Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Yuxuan Guo
- Peking University Health Science Center, School of Basic Medical Sciences, The Institute of Cardiovascular Sciences, Key Laboratory of Molecular Cardiovascular Science of Ministry of Education, Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, China
- *Correspondence: Yuxuan Guo,
| | - Erdan Dong
- Peking University Health Science Center, School of Basic Medical Sciences, The Institute of Cardiovascular Sciences, Key Laboratory of Molecular Cardiovascular Science of Ministry of Education, Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, China
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, National Health Commission of China (NHC) Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Cardiovascular Receptors Research. Beijing, China
| |
Collapse
|
12
|
Shi J, Wei L. Rho Kinases in Embryonic Development and Stem Cell Research. Arch Immunol Ther Exp (Warsz) 2022; 70:4. [PMID: 35043239 PMCID: PMC8766376 DOI: 10.1007/s00005-022-00642-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 12/14/2021] [Indexed: 12/12/2022]
Abstract
The Rho-associated coiled-coil containing kinases (ROCKs or Rho kinases) belong to the AGC (PKA/PKG/PKC) family of serine/threonine kinases and are major downstream effectors of small GTPase RhoA, a key regulator of actin-cytoskeleton reorganization. The ROCK family contains two members, ROCK1 and ROCK2, which share 65% overall identity and 92% identity in kinase domain. ROCK1 and ROCK2 were assumed to be functionally redundant, based largely on their major common activators, their high degree kinase domain homology, and study results from overexpression with kinase constructs or chemical inhibitors. ROCK signaling research has expanded to all areas of biology and medicine since its discovery in 1996. The rapid advance is befitting ROCK’s versatile functions in modulating various cell behavior, such as contraction, adhesion, migration, proliferation, polarity, cytokinesis, and differentiation. The rapid advance is noticeably driven by an extensive linking with clinical medicine, including cardiovascular abnormalities, aberrant immune responsive, and cancer development and metastasis. The rapid advance during the past decade is further powered by novel biotechnologies including CRISPR-Cas and single cell omics. Current consensus, derived mainly from gene targeting and RNA interference approaches, is that the two ROCK isoforms have overlapping and distinct cellular, physiological and pathophysiology roles. In this review, we present an overview of the milestone discoveries in ROCK research. We then focus on the current understanding of ROCK signaling in embryonic development, current research status using knockout and knockin mouse models, and stem cell research.
Collapse
Affiliation(s)
- Jianjian Shi
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, School of Medicine, Indiana University, 1044 West Walnut Street, R4-370, Indianapolis, IN, 46202-5225, USA.
| | - Lei Wei
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, School of Medicine, Indiana University, 1044 West Walnut Street, R4-370, Indianapolis, IN, 46202-5225, USA.
| |
Collapse
|
13
|
Sakabe M, Thompson M, Chen N, Verba M, Hassan A, Lu R, Xin M. Inhibition of β1-AR/Gαs signaling promotes cardiomyocyte proliferation in juvenile mice through activation of RhoA-YAP axis. eLife 2022; 11:74576. [PMID: 36479975 PMCID: PMC9767473 DOI: 10.7554/elife.74576] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Accepted: 12/07/2022] [Indexed: 12/14/2022] Open
Abstract
The regeneration potential of the mammalian heart is incredibly limited, as cardiomyocyte proliferation ceases shortly after birth. β-adrenergic receptor (β-AR) blockade has been shown to improve heart functions in response to injury; however, the underlying mechanisms remain poorly understood. Here, we inhibited β-AR signaling in the heart using metoprolol, a cardio-selective β blocker for β1-adrenergic receptor (β1-AR) to examine its role in heart maturation and regeneration in postnatal mice. We found that metoprolol enhanced cardiomyocyte proliferation and promoted cardiac regeneration post myocardial infarction, resulting in reduced scar formation and improved cardiac function. Moreover, the increased cardiomyocyte proliferation was also induced by the genetic deletion of Gnas, the gene encoding G protein alpha subunit (Gαs), a downstream effector of β-AR. Genome wide transcriptome analysis revealed that the Hippo-effector YAP, which is associated with immature cardiomyocyte proliferation, was upregulated in the cardiomyocytes of β-blocker treated and Gnas cKO hearts. Moreover, the increased YAP activity is modulated by RhoA signaling. Our pharmacological and genetic studies reveal that β1-AR-Gαs-YAP signaling axis is involved in regulating postnatal cardiomyocyte proliferation. These results suggest that inhibiting β-AR-Gαs signaling promotes the regenerative capacity and extends the cardiac regenerative window in juvenile mice by activating YAP-mediated transcriptional programs.
Collapse
Affiliation(s)
- Masahide Sakabe
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical CenterCincinnatiUnited States,Department of Pediatrics, College of Medicine, University of CincinnatiCincinnatiUnited States
| | - Michael Thompson
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical CenterCincinnatiUnited States,Department of Pediatrics, College of Medicine, University of CincinnatiCincinnatiUnited States
| | - Nong Chen
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical CenterCincinnatiUnited States,Department of Pediatrics, College of Medicine, University of CincinnatiCincinnatiUnited States
| | - Mark Verba
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical CenterCincinnatiUnited States,Department of Pediatrics, College of Medicine, University of CincinnatiCincinnatiUnited States
| | - Aishlin Hassan
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical CenterCincinnatiUnited States,Department of Pediatrics, College of Medicine, University of CincinnatiCincinnatiUnited States
| | - Richard Lu
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical CenterCincinnatiUnited States,Department of Pediatrics, College of Medicine, University of CincinnatiCincinnatiUnited States
| | - Mei Xin
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical CenterCincinnatiUnited States,Department of Pediatrics, College of Medicine, University of CincinnatiCincinnatiUnited States
| |
Collapse
|
14
|
Moore J, Emili A. Mass-Spectrometry-Based Functional Proteomic and Phosphoproteomic Technologies and Their Application for Analyzing Ex Vivo and In Vitro Models of Hypertrophic Cardiomyopathy. Int J Mol Sci 2021; 22:13644. [PMID: 34948439 PMCID: PMC8709159 DOI: 10.3390/ijms222413644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/10/2021] [Accepted: 12/15/2021] [Indexed: 11/25/2022] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is an autosomal dominant disease thought to be principally caused by mutations in sarcomeric proteins. Despite extensive genetic analysis, there are no comprehensive molecular frameworks for how single mutations in contractile proteins result in the diverse assortment of cellular, phenotypic, and pathobiological cascades seen in HCM. Molecular profiling and system biology approaches are powerful tools for elucidating, quantifying, and interpreting dynamic signaling pathways and differential macromolecule expression profiles for a wide range of sample types, including cardiomyopathy. Cutting-edge approaches combine high-performance analytical instrumentation (e.g., mass spectrometry) with computational methods (e.g., bioinformatics) to study the comparative activity of biochemical pathways based on relative abundances of functionally linked proteins of interest. Cardiac research is poised to benefit enormously from the application of this toolkit to cardiac tissue models, which recapitulate key aspects of pathogenesis. In this review, we evaluate state-of-the-art mass-spectrometry-based proteomic and phosphoproteomic technologies and their application to in vitro and ex vivo models of HCM for global mapping of macromolecular alterations driving disease progression, emphasizing their potential for defining the components of basic biological systems, the fundamental mechanistic basis of HCM pathogenesis, and treating the ensuing varied clinical outcomes seen among affected patient cohorts.
Collapse
Affiliation(s)
- Jarrod Moore
- Center for Network Systems Biology, Boston University School of Medicine, Boston, MA 02118, USA;
- Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA
- MD-PhD Program, Boston University School of Medicine, Boston, MA 02118, USA
| | - Andrew Emili
- Center for Network Systems Biology, Boston University School of Medicine, Boston, MA 02118, USA;
- Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA
| |
Collapse
|
15
|
Generation and characterization of a Myh6-driven Cre knockin mouse line. Transgenic Res 2021; 30:821-835. [PMID: 34542814 PMCID: PMC8580938 DOI: 10.1007/s11248-021-00285-4] [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: 08/02/2021] [Accepted: 09/10/2021] [Indexed: 10/25/2022]
Abstract
Gene deletion by the Cre-Loxp system has facilitated functional studies of many critical genes in mice, offering important insights and allowing deeper understanding on the mechanisms underlying organ development and diseases, such as heart development and diseases. In this study, we generated a Myh6-Cre knockin mouse model by inserting the IRES-Cre-wpre-polyA cassette between the translational stop codon and the 3' untranslated region of the endogenous Myh6 gene. By crossing knockin mice with the Rosa26 reporter lines, we found that Myh6-Cre targeted cardiomyocytes at the embryonic and postnatal stages. In addition, we were able to inactivate the desmosome gene Desmoplakin (Dsp) by breeding Myh6-Cre mice with a conditional Dspflox knockout mouse line, which resulted in embryonic lethality during the mid-term pregnancy. These results suggest that the new Myh6-Cre mouse line can serve as a robust tool to dissect the distinct roles of genes involved in heart development and function.
Collapse
|
16
|
Gu X, Zhou F, Mu J. Recent Advances in Maturation of Pluripotent Stem Cell-Derived Cardiomyocytes Promoted by Mechanical Stretch. Med Sci Monit 2021; 27:e931063. [PMID: 34381009 PMCID: PMC8369941 DOI: 10.12659/msm.931063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Stem cells have significant potential use in tissue regeneration, especially for treating cardiac diseases because of their multi-directional differentiation capability. By mimicking the in vivo physiological environment of native cardiomyocytes during their development and maturation, researchers have been able to induce pluripotent stem cell-derived cardiomyocytes (PSC-CMs) at high purity. However, the phenotype of these PSC-CMs is immature compared with that of adult cardiomyocytes. Various strategies have been explored to improve the maturity of PSC-CMs, such as long-term culturing, mechanical stimuli, chemical stimuli, and combinations of these strategies. Among these strategies, mechanical stretch as a key mechanical stimulus plays an important role in PSC-CM maturation. In this review, the optimal parameters of mechanical stretch, the effects of mechanical stretch on maturation of PSC-CMs, underlying molecular mechanisms as well as existing problems are discussed. Mechanical stretch is a powerful approach to promote the maturation of SC-CMs in terms of morphology, structure, and functionality. Nonetheless, further research efforts are needed to reach a satisfactory standard for clinical applications of PSC-CMs in treating cardiac diseases.
Collapse
Affiliation(s)
- Xingwang Gu
- Capital Medical University, Beijing, China (mainland)
| | - Fan Zhou
- Department of Ultrasound, Third Medical Center of Chinese People's Liberation Army (PLA) General Hospital, Beijing, China (mainland)
| | - Junsheng Mu
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Beijing, China (mainland)
| |
Collapse
|
17
|
ERRγ enhances cardiac maturation with T-tubule formation in human iPSC-derived cardiomyocytes. Nat Commun 2021; 12:3596. [PMID: 34155205 PMCID: PMC8217550 DOI: 10.1038/s41467-021-23816-3] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 05/17/2021] [Indexed: 01/17/2023] Open
Abstract
One of the earliest maturation steps in cardiomyocytes (CMs) is the sarcomere protein isoform switch between TNNI1 and TNNI3 (fetal and neonatal/adult troponin I). Here, we generate human induced pluripotent stem cells (hiPSCs) carrying a TNNI1EmGFP and TNNI3mCherry double reporter to monitor and isolate mature sub-populations during cardiac differentiation. Extensive drug screening identifies two compounds, an estrogen-related receptor gamma (ERRγ) agonist and an S-phase kinase-associated protein 2 inhibitor, that enhances cardiac maturation and a significant change to TNNI3 expression. Expression, morphological, functional, and molecular analyses indicate that hiPSC-CMs treated with the ERRγ agonist show a larger cell size, longer sarcomere length, the presence of transverse tubules, and enhanced metabolic function and contractile and electrical properties. Here, we show that ERRγ-treated hiPSC-CMs have a mature cellular property consistent with neonatal CMs and are useful for disease modeling and regenerative medicine. Cardiomyocytes (CMs) derived from human induced pluripotent stem cells (hiPSCs) suffer from limited maturation. Here the authors identify ERRγ agonist as a factor that enhances cardiac morphological, metabolic, contractile and electrical maturation of hiPSC-derived CMs with T-tubule formation.
Collapse
|
18
|
Guo S, Okyere AD, McEachern E, Strong JL, Carter RL, Patwa VC, Thomas TP, Landy M, Song J, Lucchese AM, Martin TG, Gao E, Rajan S, Kirk JA, Koch WJ, Cheung JY, Tilley DG. Epidermal growth factor receptor-dependent maintenance of cardiac contractility. Cardiovasc Res 2021; 118:1276-1288. [PMID: 33892492 DOI: 10.1093/cvr/cvab149] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 02/16/2021] [Accepted: 04/21/2021] [Indexed: 01/22/2023] Open
Abstract
AIMS Epidermal growth factor receptor (EGFR) is essential to the development of multiple tissues and organs and is a target of cancer therapeutics. Due to the embryonic lethality of global EGFR deletion and conflicting reports of cardiac-overexpressed EGFR mutants, its specific impact on the adult heart, normally or in response to chronic stress, has not been established. Using complimentary genetic strategies to modulate cardiomyocyte-specific EGFR expression, we aim to define its role in the regulation of cardiac function and remodeling. METHODS AND RESULTS A floxed EGFR mouse model with α-myosin heavy chain-Cre-mediated cardiomyocyte-specific EGFR downregulation (CM-EGFR-KD mice) developed contractile dysfunction by 9 weeks of age, marked by impaired diastolic relaxation, as monitored via echocardiographic, hemodynamic and isolated cardiomyocyte contractility analyses. This contractile defect was maintained over time without overt cardiac remodeling until 10 months of age, after which the mice ultimately developed severe heart failure and reduced lifespan. Acute downregulation of EGFR in adult floxed EGFR mice with adeno-associated virus 9 (AAV9)-encoded Cre with a cardiac troponin T promoter (AAV9-cTnT-Cre) recapitulated the CM-EGFR-KD phenotype, while AAV9-cTnT-EGFR treatment of adult CM-EGFR-KD mice rescued the phenotype. Notably, chronic administration of the β-adrenergic receptor (βAR) agonist isoproterenol effectively and reversibly compensated for the contractile dysfunction in the absence of cardiomyocyte hypertrophy in CM-EGFR-KD mice. Mechanistically, EGFR downregulation reduced the expression of protein phosphatase 2 A (PP2A) regulatory subunit Ppp2r3a/PR72, which was associated with decreased phosphorylation of phospholamban (PLB) and Ca2+ clearance, and whose re-expression via AAV9-cTnT-PR72 rescued the CM-EGFR-KD phenotype. CONCLUSIONS Altogether our study highlights a previously unrecognized role for EGFR in maintaining contractile homeostasis under physiologic conditions in the adult heart via regulation of PR72 expression. TRANSLATIONAL PERSPECTIVE Our study highlights a previously unrecognized role for EGFR in maintaining contractile homeostasis under physiologic conditions in the adult heart via regulation of PR72, a PP2A regulatory subunit with an unknown impact on cardiac function. Further, we have shown that cardiomyocyte-expressed EGFR is required for the promotion of cardiac hypertrophy under conditions of chronic catecholamine stress. Altogether, our study provides new insight into the dynamic nature of cardiomyocyte-specific EGFR.
Collapse
Affiliation(s)
- Shuchi Guo
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| | - Ama Dedo Okyere
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| | - Erin McEachern
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| | - Joshua L Strong
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| | - Rhonda L Carter
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| | - Viren C Patwa
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| | - Toby P Thomas
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| | - Melissa Landy
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| | - Jianliang Song
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| | - Ana Maria Lucchese
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| | - Thomas G Martin
- Loyola University Chicago, Department of Cell and Molecular Physiology, Chicago, Illinois, USA
| | - Erhe Gao
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| | - Sudarsan Rajan
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| | - Jonathan A Kirk
- Loyola University Chicago, Department of Cell and Molecular Physiology, Chicago, Illinois, USA
| | - Walter J Koch
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| | - Joseph Y Cheung
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| | - Douglas G Tilley
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| |
Collapse
|
19
|
Knight WE, Ali HR, Nakano SJ, Wilson CE, Walker LA, Woulfe KC. Ex vivo Methods for Measuring Cardiac Muscle Mechanical Properties. Front Physiol 2021; 11:616996. [PMID: 33488406 PMCID: PMC7820907 DOI: 10.3389/fphys.2020.616996] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 12/10/2020] [Indexed: 11/13/2022] Open
Abstract
Cardiovascular disease continues to be the leading cause of morbidity and mortality in the United States and thousands of manuscripts each year are aimed at elucidating mechanisms underlying cardiac disease. The methods for quantifying cardiac performance are quite varied, with each technique assessing unique features of cardiac muscle mechanical properties. Accordingly, in this review, we discuss current ex vivo methods for quantifying cardiac muscle performance, highlighting what can be learned from each method, and how each technique can be used in conjunction to complement others for a more comprehensive understanding of cardiac function. Importantly, cardiac function can be assessed at several different levels, from the whole organ down to individual protein-protein interactions. Here, we take a reductionist view of methods that are commonly used to measure the distinct aspects of cardiac mechanical function, beginning with whole heart preparations and finishing with the in vitro motility assay. While each of the techniques are individually well-documented in the literature, there is a significant need for a comparison of the techniques, delineating the mechanical parameters that can are best measured with each technique, as well as the strengths and weaknesses inherent to each method. Additionally, we will consider complementary techniques and how these methods can be used in combination to improve our understanding of cardiac mechanical function. By presenting each of these methods, with their strengths and limitations, in a single manuscript, this review will assist cardiovascular biologists in understanding the existing literature on cardiac mechanical function, as well as designing future experiments.
Collapse
Affiliation(s)
- Walter E Knight
- Department of Medicine, Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Hadi R Ali
- Department of Medicine, Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Stephanie J Nakano
- Department of Pediatrics, Division of Cardiology, Children's Hospital, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Cortney E Wilson
- Department of Medicine, Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Lori A Walker
- Department of Medicine, Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Kathleen C Woulfe
- Department of Medicine, Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| |
Collapse
|
20
|
Hydrodynamics-Based Transplacental Delivery as a Useful Noninvasive Tool for Manipulating Fetal Genome. Cells 2020; 9:cells9071744. [PMID: 32708213 PMCID: PMC7409276 DOI: 10.3390/cells9071744] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 07/04/2020] [Accepted: 07/18/2020] [Indexed: 11/17/2022] Open
Abstract
We previously demonstrated that the injection of pregnant wild-type female mice (carrying enhanced green fluorescent protein (EGFP)-expressing transgenic fetuses) at embryonic day (E) 12.5 with an all-in-one plasmid conferring the expression of both Cas9 and guide RNA (targeted to the EGFP cDNA) complexed with the gene delivery reagent, resulted in some fetuses exhibiting reduced fluorescence in their hearts and gene insertion/deletion (indel) mutations. In this study, we examined whether the endogenous myosin heavy-chain α (MHCα) gene can be successfully genome-edited by this method in the absence of a gene delivery reagent with potential fetal toxicity. For this, we employed a hydrodynamics-based gene delivery (HGD) system with the aim of ensuring fetal gene delivery rates and biosafety. We also investigated which embryonic stages are suitable for the induction of genome editing in fetuses. Of the three pregnant females injected at E9.5, one had mutated fetuses: all examined fetuses carried exogenous plasmid DNA, and four of 10 (40%) exhibited mosaic indel mutations in MHCα. Gene delivery to fetuses at E12.5 and E15.5 did not cause mutations. Thus, the HGD-based transplacental delivery of a genome editing vector may be able to manipulate the fetal genomes of E9.5 fetuses.
Collapse
|
21
|
Anderson BR, Jensen ML, Hagedorn PH, Little SC, Olson RE, Ammar R, Kienzle B, Thompson J, McDonald I, Mercer S, Vikesaa J, Nordbo B, Iben L, Cao Y, Natale J, Dalton-Kay G, Cacace A, Hansen BR, Hedtjärn M, Koch T, Bristow LJ. Allele-Selective Knockdown of MYH7 Using Antisense Oligonucleotides. MOLECULAR THERAPY-NUCLEIC ACIDS 2020; 19:1290-1298. [PMID: 32092825 PMCID: PMC7033438 DOI: 10.1016/j.omtn.2020.01.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 01/02/2020] [Accepted: 01/02/2020] [Indexed: 01/09/2023]
Abstract
Hundreds of dominant-negative myosin mutations have been identified that lead to hypertrophic cardiomyopathy, and the biomechanical link between mutation and disease is heterogeneous across this patient population. To increase the therapeutic feasibility of treating this diverse genetic population, we investigated the ability of locked nucleic acid (LNA)-modified antisense oligonucleotides (ASOs) to selectively knock down mutant myosin transcripts by targeting single-nucleotide polymorphisms (SNPs) that were found to be common in the myosin heavy chain 7 (MYH7) gene. We identified three SNPs in MYH7 and designed ASO libraries to selectively target either the reference or alternate MYH7 sequence. We identified ASOs that selectively knocked down either the reference or alternate allele at all three SNP regions. We also show allele-selective knockdown in a mouse model that was humanized on one allele. These results suggest that SNP-targeting ASOs are a promising therapeutic modality for treating cardiac pathology.
Collapse
Affiliation(s)
- Brian R Anderson
- Bristol-Myers Squibb Research and Development, 3551 Lawrenceville Rd, Princeton, NJ 08540, USA.
| | - Marianne L Jensen
- RNA Therapeutics Research, Therapeutic Modalities, Roche Pharma Research and Early Development, Roche Innovation Center Copenhagen, Hørsholm, Denmark
| | - Peter H Hagedorn
- RNA Therapeutics Research, Therapeutic Modalities, Roche Pharma Research and Early Development, Roche Innovation Center Copenhagen, Hørsholm, Denmark
| | - Sean C Little
- Bristol-Myers Squibb Research and Development, 3551 Lawrenceville Rd, Princeton, NJ 08540, USA
| | - Richard E Olson
- Bristol-Myers Squibb Research and Development, 3551 Lawrenceville Rd, Princeton, NJ 08540, USA
| | - Ron Ammar
- Bristol-Myers Squibb Research and Development, 3551 Lawrenceville Rd, Princeton, NJ 08540, USA
| | - Bernadette Kienzle
- Bristol-Myers Squibb Research and Development, 3551 Lawrenceville Rd, Princeton, NJ 08540, USA
| | - John Thompson
- Bristol-Myers Squibb Research and Development, 3551 Lawrenceville Rd, Princeton, NJ 08540, USA
| | - Ivar McDonald
- Bristol-Myers Squibb Research and Development, 3551 Lawrenceville Rd, Princeton, NJ 08540, USA
| | - Stephen Mercer
- Bristol-Myers Squibb Research and Development, 3551 Lawrenceville Rd, Princeton, NJ 08540, USA
| | - Jonas Vikesaa
- RNA Therapeutics Research, Therapeutic Modalities, Roche Pharma Research and Early Development, Roche Innovation Center Copenhagen, Hørsholm, Denmark
| | - Bettina Nordbo
- RNA Therapeutics Research, Therapeutic Modalities, Roche Pharma Research and Early Development, Roche Innovation Center Copenhagen, Hørsholm, Denmark
| | - Larry Iben
- Bristol-Myers Squibb Research and Development, 3551 Lawrenceville Rd, Princeton, NJ 08540, USA
| | - Yang Cao
- Bristol-Myers Squibb Research and Development, 3551 Lawrenceville Rd, Princeton, NJ 08540, USA
| | - Joanne Natale
- Bristol-Myers Squibb Research and Development, 3551 Lawrenceville Rd, Princeton, NJ 08540, USA
| | - Greg Dalton-Kay
- Bristol-Myers Squibb Research and Development, 3551 Lawrenceville Rd, Princeton, NJ 08540, USA
| | - Angela Cacace
- Bristol-Myers Squibb Research and Development, 3551 Lawrenceville Rd, Princeton, NJ 08540, USA
| | - Bo R Hansen
- RNA Therapeutics Research, Therapeutic Modalities, Roche Pharma Research and Early Development, Roche Innovation Center Copenhagen, Hørsholm, Denmark
| | - Maj Hedtjärn
- RNA Therapeutics Research, Therapeutic Modalities, Roche Pharma Research and Early Development, Roche Innovation Center Copenhagen, Hørsholm, Denmark
| | - Troels Koch
- RNA Therapeutics Research, Therapeutic Modalities, Roche Pharma Research and Early Development, Roche Innovation Center Copenhagen, Hørsholm, Denmark
| | - Linda J Bristow
- Bristol-Myers Squibb Research and Development, 3551 Lawrenceville Rd, Princeton, NJ 08540, USA
| |
Collapse
|
22
|
Reduced hybrid/complex N-glycosylation disrupts cardiac electrical signaling and calcium handling in a model of dilated cardiomyopathy. J Mol Cell Cardiol 2019; 132:13-23. [PMID: 31071333 DOI: 10.1016/j.yjmcc.2019.05.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 04/12/2019] [Accepted: 05/01/2019] [Indexed: 12/19/2022]
Abstract
Dilated cardiomyopathy (DCM) is the third most common cause of heart failure, with ~70% of DCM cases considered idiopathic. We showed recently, through genetic ablation of the MGAT1 gene, which encodes an essential glycosyltransferase (GlcNAcT1), that prevention of cardiomyocyte hybrid/complex N-glycosylation was sufficient to cause DCM that led to heart failure and early death. Our findings are consistent with increasing evidence suggesting a link between aberrant glycosylation and heart diseases of acquired and congenital etiologies. However, the mechanisms by which changes in glycosylation contribute to disease onset and progression remain largely unknown. Activity and gating of voltage-gated Na+ and K+ channels (Nav and Kv respectively) play pivotal roles in the initiation, shaping and conduction of cardiomyocyte action potentials (APs) and aberrant channel activity was shown to contribute to cardiac disease. We and others showed that glycosylation can impact Nav and Kv function; therefore, here, we investigated the effects of reduced cardiomyocyte hybrid/complex N-glycosylation on channel activity to investigate whether chronic aberrant channel function can contribute to DCM. Ventricular cardiomyocytes from MGAT1 deficient (MGAT1KO) mice display prolonged APs and pacing-induced aberrant early re-activation that can be attributed to, at least in part, a significant reduction in Kv expression and activity that worsens over time suggesting heart disease-related remodeling. MGAT1KO Nav demonstrate no change in expression or maximal conductance but show depolarizing shifts in voltage-dependent gating. Together, the changes in MGAT1KO Nav and Kv function likely contribute to observed anomalous electrocardiograms and Ca2+ handling. These findings provide insight into mechanisms by which altered glycosylation contributes to DCM through changes in Nav and Kv activity that impact conduction, Ca2+ handling and contraction. The MGAT1KO can also serve as a useful model to study the effects of aberrant electrical signaling on cardiac function and the remodeling events that can occur with heart disease progression.
Collapse
|
23
|
Wang Z, Huang J. Apela Promotes Cardiomyocyte Differentiation from Transgenic Human Embryonic Stem Cell Lines. Appl Biochem Biotechnol 2019; 189:396-410. [PMID: 31025171 DOI: 10.1007/s12010-019-03012-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 03/27/2019] [Indexed: 12/22/2022]
Abstract
Although embryonic stem (ES) cells (ESCs) may be a promising donor source for the repair of infarcted or ischemic heart tissues, their successful application in regenerative medicine has been hampered by difficulties in enriching, identifying, and selecting cardiomyocytes from the differentiating cells. We established transgenic human ES cell lines by transcriptional control of the α-cardiac myosin heavy chain (α-MHC) promoter driving green fluorescent protein (GFP) expression. Differentiated GFP-expressing cells display the characteristics of cardiomyocytes (CMs). Apela, a recently identified short peptide, up-regulated the expression of the cardiac-restricted transcription factors Tbx5 and GATA4 as well as differentiated the cardiomyocyte markers α-MHC and β-MHC. Flow cytometric analysis showed that apela increased the percentage of GFP-expressing cells in the beating foci of the embryoid bodies. The percentage of cardiac troponin T (TNT)-positive cells and the protein expression of TNT were increased in the ES cell-derived CMs with apela treatment. Functionally, the contractile frequency of the ES-derived CMs responded appropriately to the vasoactive drugs isoprenaline and carbachol. Our work presented a protocol for specially labelling and enriching CMs by combining transgenic human ES cell lines and exogenous growth factor treatment.
Collapse
Affiliation(s)
- Zhi Wang
- Department of Cardiovascular Medicine, Nanjing Brain Hospital, Nanjing Medical University, Nanjing, People's Republic of China. .,Department of Cardiovascular Medicine, Nanjing Chest Hospital, Nanjing, People's Republic of China. .,Department of Cardiology, Nanjing Brain Hospital, Nanjing Medical University, No. 264 Guangzhou Road, Nanjing, 210029, People's Republic of China.
| | - Jin Huang
- Department of Cardiovascular Medicine, Nanjing Brain Hospital, Nanjing Medical University, Nanjing, People's Republic of China. .,Department of Cardiovascular Medicine, Nanjing Chest Hospital, Nanjing, People's Republic of China.
| |
Collapse
|
24
|
Wan TC, Tampo A, Kwok WM, Auchampach JA. Ability of CP-532,903 to protect mouse hearts from ischemia/reperfusion injury is dependent on expression of A 3 adenosine receptors in cardiomyoyctes. Biochem Pharmacol 2019; 163:21-31. [PMID: 30710517 DOI: 10.1016/j.bcp.2019.01.022] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Accepted: 01/28/2019] [Indexed: 10/27/2022]
Abstract
A3 adenosine receptor (A3AR) agonists are effective at limiting injury caused by ischemia/reperfusion injury of the heart in experimental animal models. However, understanding of their mechanism of action, which is likely multifactorial, remains incomplete. In prior studies, it has been demonstrated that A3AR-mediated ischemic protection is blocked by glibenclamide and is absent in Kir6.2 gene ablated mice that lack the pore-forming subunit of the ATP-sensitive potassium (KATP) channel, suggesting one contributing mechanism may involve accelerated activation of KATP channels. However, presence of A3ARs in the myocardium has yet to be established. Utilizing a whole-cell recording technique, in this study we confirm functional expression of the A3AR in adult mouse ventricular cardiomyocytes, coupled to activation of ATP-dependent potassium (KATP) channels via Gi inhibitory proteins. We further show that ischemic protection provided by the selective A3AR agonist CP-532,903 in an isolated, buffer-perfused heart model is lost completely in Adora3LoxP/LoxP;Myh6-Cre mice, which is a newly developed model developed and comprehensively described herein whereby the A3AR gene (Adora3) is deleted exclusively in cardiomyocytes. Our findings, taken together with previously published work, are consistent with the hypothesis that A3AR agonists provide ischemic tolerance, at least in part, by facilitating opening of myocardial KATP channels.
Collapse
Affiliation(s)
- Tina C Wan
- Department of Pharmacology & Toxicology, Medical College of Wisconsin, Milwaukee, WI 53226, United States; Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, United States
| | - Akihito Tampo
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI 53226, United States; Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, United States
| | - Wai-Meng Kwok
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI 53226, United States; Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, United States
| | - John A Auchampach
- Department of Pharmacology & Toxicology, Medical College of Wisconsin, Milwaukee, WI 53226, United States; Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, United States.
| |
Collapse
|
25
|
Fenix AM, Neininger AC, Taneja N, Hyde K, Visetsouk MR, Garde RJ, Liu B, Nixon BR, Manalo AE, Becker JR, Crawley SW, Bader DM, Tyska MJ, Liu Q, Gutzman JH, Burnette DT. Muscle-specific stress fibers give rise to sarcomeres in cardiomyocytes. eLife 2018; 7:42144. [PMID: 30540249 PMCID: PMC6307863 DOI: 10.7554/elife.42144] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 12/11/2018] [Indexed: 11/13/2022] Open
Abstract
The sarcomere is the contractile unit within cardiomyocytes driving heart muscle contraction. We sought to test the mechanisms regulating actin and myosin filament assembly during sarcomere formation. Therefore, we developed an assay using human cardiomyocytes to monitor sarcomere assembly. We report a population of muscle stress fibers, similar to actin arcs in non-muscle cells, which are essential sarcomere precursors. We show sarcomeric actin filaments arise directly from muscle stress fibers. This requires formins (e.g., FHOD3), non-muscle myosin IIA and non-muscle myosin IIB. Furthermore, we show short cardiac myosin II filaments grow to form ~1.5 μm long filaments that then 'stitch' together to form the stack of filaments at the core of the sarcomere (i.e., the A-band). A-band assembly is dependent on the proper organization of actin filaments and, as such, is also dependent on FHOD3 and myosin IIB. We use this experimental paradigm to present evidence for a unifying model of sarcomere assembly.
Collapse
Affiliation(s)
- Aidan M Fenix
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, United States
| | - Abigail C Neininger
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, United States
| | - Nilay Taneja
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, United States
| | - Karren Hyde
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, United States
| | - Mike R Visetsouk
- Department of Biological Sciences, Cell and Molecular Biology, University of Wisconsin Milwaukee, Milwaukee, United States
| | - Ryan J Garde
- Department of Biological Sciences, Cell and Molecular Biology, University of Wisconsin Milwaukee, Milwaukee, United States
| | - Baohong Liu
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, United States
| | - Benjamin R Nixon
- Department of Medicine, Vanderbilt University Medical Center, Nashville, United States
| | - Annabelle E Manalo
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, United States
| | - Jason R Becker
- Department of Medicine, Vanderbilt University Medical Center, Nashville, United States
| | - Scott W Crawley
- Department of Biological Sciences, The University of Toledo, Toledo, United States
| | - David M Bader
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, United States
| | - Matthew J Tyska
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, United States
| | - Qi Liu
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, United States
| | - Jennifer H Gutzman
- Department of Biological Sciences, Cell and Molecular Biology, University of Wisconsin Milwaukee, Milwaukee, United States
| | - Dylan T Burnette
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, United States
| |
Collapse
|
26
|
Wang Q, Fan Y, Kurita H, Jiang M, Koch S, Rao MB, Rubinstein J, Puga A. Aryl Hydrocarbon Receptor Ablation in Cardiomyocytes Protects Male Mice From Heart Dysfunction Induced by NKX2.5 Haploinsufficiency. Toxicol Sci 2018; 160:74-82. [PMID: 28973413 DOI: 10.1093/toxsci/kfx164] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Epidemiological studies in humans and research in vertebrates indicates that developmental exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), a ubiquitous and biopersistent environmental toxicant, is associated with incidence of early congenital heart disease in the embryo and later in the adult. TCDD-mediated toxicity depends on the aryl hydrocarbon receptor (AHR) but the role of the TCDD-activated AHR in cardiac function is not well-defined. To characterize the mechanisms responsible for AHR-mediated disruption of heart function, we generated several mouse strains with cardiomyocyte-specific Ahr gene knockout. Here, we report results on one of these strains in which the Ahr gene was deleted by cre recombinase regulated by the promoter of the cardiomyocyte-specific Nkx2.5 gene. We crossed mice with loxP-targeted Ahrfx/fx alleles with Nkx2.5+/cre mice bearing a "knock-in" cre recombinase gene integrated into one of the Nkx2.5 alleles. In these mice, loss of one Nkx2.5 allele is associated with disrupted cardiac development. In males, Nkx2.5 hemizygosity resulted in cardiac haploinsufficiency characterized by hypertrophy, dilated cardiomyopathy, and impaired ejection fraction. Ahr ablation protected Nkx2.5+/cre haploinsufficient males from cardiac dysfunction while inducing a significant increase in body weight. These effects were absent or largely blunted in females. Starting at 3 months of age, mice were exposed by oral gavage to 1 μg/kg/week of TCDD or control vehicle for an additional 2 months. TCDD exposure restored cardiac physiology in aging males, appearing to compensate for the heart dysfunction caused by Nkx2.5 hemizygosity. Our findings underscore the conclusion that deletion of the Ahr gene in cardiomyocytes protects males from heart dysfunction due to NKX2.5 haploinsufficiency.
Collapse
Affiliation(s)
- Qin Wang
- Department of Environmental Health and Center for Environmental Genetics
| | - Yunxia Fan
- Department of Environmental Health and Center for Environmental Genetics
| | - Hisaka Kurita
- Department of Environmental Health and Center for Environmental Genetics
| | - Min Jiang
- Department of Internal Medicine Cardiology Division, University of Cincinnati College of Medicine, Cincinnati, Ohio 45267
| | - Sheryl Koch
- Department of Internal Medicine Cardiology Division, University of Cincinnati College of Medicine, Cincinnati, Ohio 45267
| | - Marepalli B Rao
- Department of Environmental Health and Center for Environmental Genetics
| | - Jack Rubinstein
- Department of Internal Medicine Cardiology Division, University of Cincinnati College of Medicine, Cincinnati, Ohio 45267
| | - Alvaro Puga
- Department of Environmental Health and Center for Environmental Genetics
| |
Collapse
|
27
|
Da Silva F, Massa F, Motamedi FJ, Vidal V, Rocha AS, Gregoire EP, Cai CL, Wagner KD, Schedl A. Myocardial-specific R-spondin3 drives proliferation of the coronary stems primarily through the Leucine Rich Repeat G Protein coupled receptor LGR4. Dev Biol 2018; 441:42-51. [PMID: 29859889 DOI: 10.1016/j.ydbio.2018.05.024] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 03/29/2018] [Accepted: 05/29/2018] [Indexed: 10/14/2022]
Abstract
Coronary artery anomalies are common congenital disorders with serious consequences in adult life. Coronary circulation begins when the coronary stems form connections between the aorta and the developing vascular plexus. We recently identified the WNT signaling modulator R-spondin 3 (Rspo3), as a crucial regulator of coronary stem proliferation. Using expression analysis and tissue-specific deletion we now demonstrate that Rspo3 is primarily produced by cardiomyocytes. Moreover, we have employed CRISPR/Cas9 technology to generate novel Lgr4-null alleles that showed a significant decrease in coronary stem proliferation and thus phenocopied the coronary artery defects seen in Rspo3 mutants. Interestingly, Lgr4 mutants displayed slightly hypomorphic right ventricles, an observation also made after myocardial specific deletion of Rspo3. These results shed new light on the role of Rspo3 in heart development and demonstrate that LGR4 is the principal R-spondin 3 receptor in the heart.
Collapse
Affiliation(s)
- Fabio Da Silva
- Université Côte d'Azur, Inserm, CNRS, iBV, Nice 06108, France
| | - Filippo Massa
- Université Côte d'Azur, Inserm, CNRS, iBV, Nice 06108, France
| | | | - Valerie Vidal
- Université Côte d'Azur, Inserm, CNRS, iBV, Nice 06108, France
| | - Ana Sofia Rocha
- Université Côte d'Azur, Inserm, CNRS, iBV, Nice 06108, France
| | | | - Chen-Leng Cai
- Department of Developmental and Regenerative Biology, The Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
| | | | - Andreas Schedl
- Université Côte d'Azur, Inserm, CNRS, iBV, Nice 06108, France.
| |
Collapse
|
28
|
Kraev A. Insertional Mutagenesis Confounds the Mechanism of the Morbid Phenotype of a PLN R9C Transgenic Mouse Line. J Card Fail 2018; 24:115-125. [PMID: 29325795 DOI: 10.1016/j.cardfail.2017.12.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Revised: 12/18/2017] [Accepted: 12/21/2017] [Indexed: 12/29/2022]
Abstract
BACKGROUND A mouse line with heterozygous transgenic expression of phospholamban carrying a substitution of cysteine for arginine 9 (TgPLNR9C) under the control of α-myosin heavy chain (αMHC) promoter features dilated cardiomyopathy, heart failure, and premature death. METHODS AND RESULTS Determination of transgene chromosomal localization by conventional methods shows that in this line the transgenic array of 13 PLNR9C expression cassettes, arranged in a head-to-tail tandem orientation, have integrated into the bidirectional promoter of the αMHC (Myh6) gene and the gene for the regulatory noncoding RNA Myheart (Mhrt), both of which are known to be involved in cardiac development and pathology. Expression of the noncoding RNA Mhrt in TgPLNR9C mice exhibits profound deregulation, despite the presence of the second, intact allele. CONCLUSIONS The TgPLNR9C mouse strain is, in the best case, a functionally ambiguous phenocopy of the human PLNR9C heterozygote, because a similar constellation of genetically programmed events can not occur in a patient. Publications featuring "cardiac-specific overexpression" are focused on the phenotype and typically forgo the definition of the transgene integration site or transgene temporal expression profile, so caution should be exercised in attributing clinical relevance to pathologic phenomena observed in αMHC-driven transgenes.
Collapse
Affiliation(s)
- Alexander Kraev
- University of Toronto, 27 King's College Circle, Toronto, Ontario M5S 1A1, Canada.
| |
Collapse
|
29
|
Kawai M, Johnston JR, Karam T, Wang L, Singh RK, Pinto JR. Myosin Rod Hypophosphorylation and CB Kinetics in Papillary Muscles from a TnC-A8V KI Mouse Model. Biophys J 2017; 112:1726-1736. [PMID: 28445763 DOI: 10.1016/j.bpj.2017.02.045] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 02/17/2017] [Accepted: 02/22/2017] [Indexed: 12/11/2022] Open
Abstract
The cardiac troponin C (TnC)-A8V mutation is associated with hypertrophic and restrictive cardiomyopathy (HCM and RCM) in human and mice. The residue affected lies in the N-helix, a region known to affect Ca2+-binding affinity to the N-terminal domain. Here we report on the functional effects of this mutation in skinned papillary muscle fibers from homozygous knock-in TnC-A8V mice. Muscle fibers from left ventricle were activated at 25°C under the ionic conditions of working cardiomyocytes. The pCa-tension relationship showed a 3× increase in Ca2+-sensitivity and a decrease (0.8×) in cooperativity (nH) in mutant fibers. The elementary steps of the cross-bridge (CB) cycle were investigated by sinusoidal analysis. The ATP study revealed that there is no significant change in the affinity of ATP (K1) for the myosin head. In TnC-A8V mutant fibers, the CB detachment rate (k2) and its equilibrium constant (K2) increased (1.5×). The phosphate study revealed that rate constant of the force-generation step (k4) decreased (0.5×), reversal step (k-4) increased (2×), and the phosphate-release step (1/K5) increased (2×). Pro-Q Diamond staining of the skinned fibers samples revealed no significant changes in total phosphorylation of multiple sarcomeric proteins. Further investigation using liquid chromatography-tandem mass spectrometry revealed hypophosphorylation of the rod domain of myosin heavy chain in TnC-A8V mutant fibers compared to wild-type. Immunoblotting confirmed the results observed in the mass spectrometry analysis. The results suggest perturbed CB kinetics-possibly caused by changes in the α-myosin heavy chain phosphorylation profile-as a novel mechanism, to our knowledge, by which a mutation in TnC can have rippling effects in the myofilament and contribute to the pathogenesis of HCM/RCM.
Collapse
Affiliation(s)
- Masataka Kawai
- Departments of Anatomy and Cell Biology, and Internal Medicine, College of Medicine, University of Iowa, Iowa City, Iowa.
| | - Jamie R Johnston
- Department of Biomedical Sciences, College of Medicine, The Florida State University, Tallahassee, Florida
| | - Tarek Karam
- Departments of Anatomy and Cell Biology, and Internal Medicine, College of Medicine, University of Iowa, Iowa City, Iowa
| | - Li Wang
- Departments of Anatomy and Cell Biology, and Internal Medicine, College of Medicine, University of Iowa, Iowa City, Iowa; School of Nursing, Soochow University, Suzhou, Jiangsu, China
| | - Rakesh K Singh
- Translational Science Laboratory, College of Medicine, The Florida State University, Tallahassee, Florida
| | - Jose R Pinto
- Department of Biomedical Sciences, College of Medicine, The Florida State University, Tallahassee, Florida
| |
Collapse
|
30
|
Novel large-particle FACS purification of adult ventricular myocytes reveals accumulation of myosin and actin disproportionate to cell size and proteome in normal post-weaning development. J Mol Cell Cardiol 2017; 111:114-122. [PMID: 28780067 DOI: 10.1016/j.yjmcc.2017.07.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2017] [Accepted: 07/25/2017] [Indexed: 12/21/2022]
Abstract
RATIONALE Quantifying cellular proteins in ventricular myocytes (MCs) is challenging due to tissue heterogeneity and the variety of cell sizes in the heart. In post-weaning cardiac ontogeny, rod-shaped MCs make up the majority of the cardiac mass while remaining a minority of cardiac cells in number. Current biochemical analyses of cardiac proteins do not correlate well the content of MC-specific proteins to cell type or size in normally developing tissue. OBJECTIVE To develop a new large-particle fluorescent-activated cell sorting (LP-FACS) strategy for the purification of adult rod-shaped MCs. This approach is developed to enable growth-scaled measurements per-cell of the MC proteome and sarcomeric proteins (i.e. myosin heavy chain (MyHC) and alpha-actin (α-actin)) content. METHODS AND RESULTS Individual cardiac cells were isolated from 21 to 94days old mice. An LP-FACS jet-in-air system with a 200-μm nozzle was defined for the first time to purify adult MCs. Cell-type specific immunophenotyping and sorting yielded ≥95% purity of adult MCs independently of cell morphology and size. This approach excluded other cell types and tissue contaminants from further analysis. MC proteome, MyHC and α-actin proteins were measured in linear biochemical assays normalized to cell numbers. Using the allometric coefficient α, we scaled the MC-specific rate of protein accumulation to growth post-weaning. MC-specific volumes (α=1.02) and global protein accumulation (α=0.94) were proportional (i.e. isometric) to body mass. In contrast, MyHC and α-actin accumulated at a much greater rate (i.e. hyperallometric) than body mass (α=1.79 and 2.19 respectively) and MC volumes (α=1.76 and 1.45 respectively). CONCLUSION Changes in MC proteome and cell volumes measured in LP-FACS purified MCs are proportional to body mass post-weaning. Oppositely, MyHC and α-actin are concentrated more rapidly than what would be expected from MC proteome accumulation, cell enlargement, or animal growth alone. LP-FACS provides a new standard for adult MC purification and an approach to scale the biochemical content of specific proteins or group of proteins per cell in enlarging MCs.
Collapse
|
31
|
|
32
|
Moyer SM, Larsson CA, Lozano G. Mdm proteins: critical regulators of embry ogenesis and homeostasis. J Mol Cell Biol 2017; 9:mjx004. [PMID: 28093454 PMCID: PMC5439424 DOI: 10.1093/jmcb/mjx004] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Revised: 12/13/2016] [Accepted: 01/10/2017] [Indexed: 12/31/2022] Open
Abstract
Mdm2 and Mdm4 are negative regulators of the tumor suppressor p53; hence, this relationship is the focus of many cancer related studies. A multitude of experiments across various developmental stages have been conducted to explore the tissue-specific roles of these proteins in the mouse. When Mdm2 or Mdm4 are deleted in the germline or specific tissues, they display different phenotypic defects, some of which lead to embryonic lethality. Mdm2 loss is often more deleterious than loss of its homolog Mdm4 All tissues experience activation of p53 target genes upon loss of Mdm2 or Mdm4; however, the degree to which the p53 pathway is perturbed is highly tissue-specific and does not correlate to the severity of the morphological phenotypes. Therefore, a need for further understanding of how these proteins regulate p53 activity is warranted, as therapeutic targeting of the p53 pathway is rapidly evolving and gaining attention in the field of cancer research. In this review, we discuss the tissue-specificity of Mdm proteins in regulating p53 and expose the need for investigation at the cell-specific level.
Collapse
Affiliation(s)
- Sydney M Moyer
- Department of Genetics, The University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030, USA
| | - Connie A Larsson
- Department of Genetics, The University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030, USA
| | - Guillermina Lozano
- Department of Genetics, The University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030, USA
| |
Collapse
|
33
|
Depletion of Tip60 from In Vivo Cardiomyocytes Increases Myocyte Density, Followed by Cardiac Dysfunction, Myocyte Fallout and Lethality. PLoS One 2016; 11:e0164855. [PMID: 27768769 PMCID: PMC5074524 DOI: 10.1371/journal.pone.0164855] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 09/15/2016] [Indexed: 12/20/2022] Open
Abstract
Tat-interactive protein 60 (Tip60), encoded by the Kat5 gene, is a member of the MYST family of acetyltransferases. Cancer biology studies have shown that Tip60 induces the DNA damage response, apoptosis, and cell-cycle inhibition. Although Tip60 is expressed in the myocardium, its role in cardiomyocytes (CMs) is unclear. Earlier studies here showed that application of cardiac stress to globally targeted Kat5+/—haploinsufficient mice resulted in inhibition of apoptosis and activation of the CM cell-cycle, despite only modest reduction of Tip60 protein levels. It was therefore of interest to ascertain the effects of specifically and substantially depleting Tip60 from CMs using Kat5LoxP/-;Myh6-Cre mice in the absence of stress. We report initial findings using this model, in which the effects of specifically depleting Tip60 protein from ventricular CMs, beginning at early neonatal stages, were assessed in 2–12 week-old mice. Although 5’-bromodeoxyuridine immunostaining indicated that CM proliferation was not altered at any of these stages, CM density was increased in 2 week-old ventricles, which persisted in 4 week-old hearts when TUNEL staining revealed inhibition of apoptosis. By week 4, levels of connexin-43 were depleted, and its patterning was dysmorphic, concomitant with an increase in cardiac hypertrophy marker expression and interstitial fibrosis. This was followed by systolic dysfunction at 8 weeks, after which extensive apoptosis and CM fallout occurred, followed by lethality as mice approached 12 weeks of age. In summary, chronic depletion of Tip60 from the ventricular myocardium beginning at early stages of neonatal heart development causes CM death after 8 weeks; hence, Tip60 protein has a crucial function in the heart.
Collapse
|
34
|
Yan J, Zhang L, Sultana N, Oh JG, Wu B, Hajjar RJ, Zhou B, Cai CL. A series of robust genetic indicators for definitive identification of cardiomyocytes. J Mol Cell Cardiol 2016; 97:278-85. [PMID: 27266388 DOI: 10.1016/j.yjmcc.2016.06.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 06/02/2016] [Indexed: 12/21/2022]
Abstract
Definitively identifying the cell type of newly generated cells in the heart and defining their origins are central questions in cardiac regenerative medicine. Currently, it is challenging to ascertain the myocardial identity and to track myocardial progeny during heart development and disease due to lack of proper genetic tools. This may lead to many misinterpretations of the findings in cardiac regenerative biology. In this study, we developed a set of novel mouse models by inserting double reporter genes nlacZ/H2B-GFP, mGFP/H2B-mCherry into the start codon of Tnnt2 and Myh6. nlacZ (nuclear lacZ) and mGFP (membrane GFP) are flanked by two LoxP sites in these animals. We found that the reporter genes faithfully recapitulated Tnnt2 and Myh6 cardiac expression from embryonic stage and adulthood. The reporter mice provide unprecedented robustness and fidelity for visualizing and tracing cardiomyocytes with nuclear or cell membrane localization signals. These animal models offer superior genetic tools to meet a critical need in studies of heart development, cardiac stem cell biology and cardiac regenerative medicine.
Collapse
Affiliation(s)
- Jianyun Yan
- Department of Developmental and Regenerative Biology, The Black Family Stem Cell Institute, and The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Lu Zhang
- Department of Developmental and Regenerative Biology, The Black Family Stem Cell Institute, and The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Nishat Sultana
- Department of Developmental and Regenerative Biology, The Black Family Stem Cell Institute, and The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jae Gyun Oh
- Department of Medicine, Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Bingruo Wu
- Departments of Genetics, Albert Einstein College of Medicine of Yeshiva University, Bronx, New York 10461, USA
| | - Roger J Hajjar
- Department of Medicine, Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Bin Zhou
- Departments of Genetics, Albert Einstein College of Medicine of Yeshiva University, Bronx, New York 10461, USA
| | - Chen-Leng Cai
- Department of Developmental and Regenerative Biology, The Black Family Stem Cell Institute, and The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
35
|
Hille S, Dierck F, Kühl C, Sosna J, Adam-Klages S, Adam D, Lüllmann-Rauch R, Frey N, Kuhn C. Dyrk1a regulates the cardiomyocyte cell cycle via D-cyclin-dependent Rb/E2f-signalling. Cardiovasc Res 2016; 110:381-94. [PMID: 27056896 DOI: 10.1093/cvr/cvw074] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 04/01/2016] [Indexed: 11/14/2022] Open
Abstract
AIMS Down syndrome-associated dual-specificity tyrosine-(Y)-phosphorylation-regulated kinase 1A (DYRK1A) is a ubiquitously expressed protein kinase. Up to date a variety of targets have been identified, establishing a key role for Dyrk1a in selected signalling pathways. In cardiomyocytes, Dyrk1a acts as a negative regulator of hypertrophy by phosphorylating transcription factors of the NFAT family, but its mechanistic function in the heart remains poorly understood. This study was designed to investigate a potential protective role of Dyrk1a in cardiac hypertrophy in vivo. METHODS AND RESULTS We generated transgenic mice with cardiac-specific overexpression of Dyrk1a. Counterintuitively, these mice developed severe dilated cardiomyopathy associated with congestive heart failure and premature death. In search for the cause of this unexpected phenotype, we found that Dyrk1a interacts with all members of the D-cyclin family and represses their protein levels in vitro and in vivo. Particularly, forced expression of Dyrk1a leads to increased phosphorylation of Ccnd2 on Thr280 and promotes its subsequent proteasomal degradation. Accordingly, cardiomyocytes overexpressing Dyrk1a display hypo-phosphorylated Rb1, suppression of Rb/E2f-signalling, and reduced expression of E2f-target genes, which ultimately results in impaired cell cycle progression. CONCLUSIONS We identified Dyrk1a as a novel negative regulator of D-cyclin-mediated Rb/E2f-signalling. As dysregulation of this pathway with impaired cardiomyocyte proliferation leads to cardiomyopathy, dose-specific Dyrk1a expression and activity appears to be critical for the hyperplastic and hypertrophic growth of the developing heart.
Collapse
MESH Headings
- Animals
- Cardiomegaly/enzymology
- Cardiomegaly/genetics
- Cardiomegaly/pathology
- Cardiomegaly/physiopathology
- Cardiomyopathy, Dilated/enzymology
- Cardiomyopathy, Dilated/genetics
- Cardiomyopathy, Dilated/pathology
- Cardiomyopathy, Dilated/physiopathology
- Cell Cycle
- Cell Proliferation
- Cyclin D/genetics
- Cyclin D/metabolism
- Disease Models, Animal
- E2F Transcription Factors/metabolism
- Gene Expression Regulation
- HEK293 Cells
- Heart Failure/enzymology
- Heart Failure/genetics
- Heart Failure/pathology
- Heart Failure/physiopathology
- Humans
- Mice, Inbred C57BL
- Mice, Transgenic
- Myocytes, Cardiac/enzymology
- Myocytes, Cardiac/pathology
- Phosphorylation
- Protein Binding
- Protein Serine-Threonine Kinases/genetics
- Protein Serine-Threonine Kinases/metabolism
- Protein-Tyrosine Kinases/genetics
- Protein-Tyrosine Kinases/metabolism
- Rats, Wistar
- Retinoblastoma/metabolism
- Signal Transduction
- Time Factors
- Transfection
- Dyrk Kinases
Collapse
Affiliation(s)
- Susanne Hille
- Department of Internal Medicine III, University Medical Center of Schleswig-Holstein, Campus Kiel, Arnold-Heller-Str. 3 (Building 6), 24105 Kiel, Germany DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Kiel, Germany
| | - Franziska Dierck
- Department of Internal Medicine III, University Medical Center of Schleswig-Holstein, Campus Kiel, Arnold-Heller-Str. 3 (Building 6), 24105 Kiel, Germany DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Kiel, Germany
| | - Constantin Kühl
- Department of Internal Medicine III, University Medical Center of Schleswig-Holstein, Campus Kiel, Arnold-Heller-Str. 3 (Building 6), 24105 Kiel, Germany DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Kiel, Germany
| | - Justyna Sosna
- Institute of Immunology, Christian-Albrechts-University Kiel, UKSH Campus Kiel, 24105 Kiel, Germany
| | - Sabine Adam-Klages
- Institute of Immunology, Christian-Albrechts-University Kiel, UKSH Campus Kiel, 24105 Kiel, Germany
| | - Dieter Adam
- Institute of Immunology, Christian-Albrechts-University Kiel, UKSH Campus Kiel, 24105 Kiel, Germany
| | | | - Norbert Frey
- Department of Internal Medicine III, University Medical Center of Schleswig-Holstein, Campus Kiel, Arnold-Heller-Str. 3 (Building 6), 24105 Kiel, Germany DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Kiel, Germany
| | - Christian Kuhn
- Department of Internal Medicine III, University Medical Center of Schleswig-Holstein, Campus Kiel, Arnold-Heller-Str. 3 (Building 6), 24105 Kiel, Germany DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Kiel, Germany
| |
Collapse
|
36
|
Bhandary B, Robbins J. Giving credence to controls: Avoiding the false phenotype. J Mol Cell Cardiol 2015; 86:136-7. [PMID: 26235056 DOI: 10.1016/j.yjmcc.2015.07.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 07/11/2015] [Indexed: 12/12/2022]
Affiliation(s)
- Bidur Bhandary
- The Heart Institute, Department of Pediatrics, The Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Jeffrey Robbins
- The Heart Institute, Department of Pediatrics, The Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.
| |
Collapse
|
37
|
Yan J, Zhang L, Sultana N, Park DS, Shekhar A, Bu L, Hu J, Razzaque S, Cai CL. A Murine Myh6MerCreMer Knock-In Allele Specifically Mediates Temporal Genetic Deletion in Cardiomyocytes after Tamoxifen Induction. PLoS One 2015. [PMID: 26204265 PMCID: PMC4512710 DOI: 10.1371/journal.pone.0133472] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
A mouse model that mediates temporal, specific, and efficient myocardial deletion with Cre-LoxP technology will be a valuable tool to determine the function of genes during heart formation. Mhy6 encodes a cardiac muscle specific protein: alpha-myosin heavy chain. Here, we generated a new Myh6-MerCreMer (Myh6(MerCreMer/+)) inducible Cre knock-in mouse by inserting a MerCreMer cassette into the Myh6 start codon. By crossing knock-in mice with Rosa26 reporter lines, we found the Myh6(MerCreMer/+) mice mediate complete Cre-LoxP recombination in cardiomyocytes after tamoxifen induction. X-gal staining and immunohistochemistry analysis revealed that Myh6-driven Cre recombinase was specifically activated in cardiomyocytes at embryonic and adult stages. Furthermore, echocardiography showed that Myh6(MerCreMer/+) mice maintained normal cardiac structure and function before and after tamoxifen administration. These results suggest that the new Myh6(MerCreMer/+) mouse can serve as a robust tool to dissect the roles of genes in heart development and function. Additionally, myocardial progeny during heart development and after cardiac injury can be traced using this mouse line.
Collapse
Affiliation(s)
- Jianyun Yan
- Department of Developmental and Regenerative Biology, The Black Family Stem Cell Institute, and The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - Lu Zhang
- Department of Developmental and Regenerative Biology, The Black Family Stem Cell Institute, and The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - Nishat Sultana
- Department of Developmental and Regenerative Biology, The Black Family Stem Cell Institute, and The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - David S. Park
- Leon H. Charney Division of Cardiology, New York University School of Medicine, New York, NY, United States of America
| | - Akshay Shekhar
- Leon H. Charney Division of Cardiology, New York University School of Medicine, New York, NY, United States of America
| | - Lei Bu
- Leon H. Charney Division of Cardiology, New York University School of Medicine, New York, NY, United States of America
| | - Jun Hu
- Department of Developmental and Regenerative Biology, The Black Family Stem Cell Institute, and The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - Shegufta Razzaque
- Department of Developmental and Regenerative Biology, The Black Family Stem Cell Institute, and The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - Chen-Leng Cai
- Department of Developmental and Regenerative Biology, The Black Family Stem Cell Institute, and The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
- * E-mail:
| |
Collapse
|
38
|
Analysis of cardiomyocyte movement in the developing murine heart. Biochem Biophys Res Commun 2015; 464:1000-1007. [PMID: 26168730 DOI: 10.1016/j.bbrc.2015.07.036] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 07/07/2015] [Indexed: 12/29/2022]
Abstract
The precise assemblage of several types of cardiac precursors controls heart organogenesis. The cardiac precursors show dynamic movement during early development and then form the complicated heart structure. However, cardiomyocyte movements inside the newly organized mammalian heart remain unclear. We previously established the method of ex vivo time-lapse imaging of the murine heart to study cardiomyocyte behavior by using the Fucci (fluorescent ubiquitination-based cell cycle indicator) system, which can effectively label individual G1, S/G2/M, and G1/S-transition phase nuclei in living cardiomyocytes as red, green, and yellow, respectively. Global analysis of gene expression in Fucci green positive ventricular cardiomyocytes confirmed that cell cycle regulatory genes expressed in G1/S, S, G2/M, and M phase transitions were upregulated. Interestingly, pathway analysis revealed that many genes related to the cell cycle were significantly upregulated in the Fucci green positive ventricular cardiomyocytes, while only a small number of genes related to cell motility were upregulated. Time-lapse imaging showed that murine proliferating cardiomyocytes did not exhibit dynamic movement inside the heart, but stayed on site after entering the cell cycle.
Collapse
|
39
|
Liang D, Li J, Wu Y, Zhen L, Li C, Qi M, Wang L, Deng F, Huang J, Lv F, Liu Y, Ma X, Yu Z, Zhang Y, Chen YH. miRNA-204 drives cardiomyocyte proliferation via targeting Jarid2. Int J Cardiol 2015; 201:38-48. [PMID: 26298346 DOI: 10.1016/j.ijcard.2015.06.163] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Revised: 06/11/2015] [Accepted: 06/29/2015] [Indexed: 02/07/2023]
Abstract
OBJECTIVES In mammals, the heart grows by hypertrophy but not proliferation of cardiomyocytes after birth. The paucity of cardiomyocyte proliferation limits cardiac regeneration in a variety of heart diseases. To explore the efficient strategies that drive cardiomyocyte proliferation, we employed in vitro and in vivo models to investigate the function of miRNA-204, which was demonstrated to regulate the proliferation and differentiation of human cardiac progenitor cells in our previous study. METHODS AND RESULTS miRNA-204 overexpression markedly promoted cardiomyocyte proliferation in both neonatal and adult rat cardiomyocytes in vitro. Transgenic mice with the cardiac-specific overexpression of miRNA-204 exhibited excessive cardiomyocyte proliferation throughout the embryonic and adult stages, leading to a pronounced increase in ventricular mass. Accordingly, the cell cycle regulators, including Cyclin A, Cyclin B, Cyclin D2, Cyclin E, CDC2 and PCNA, were upregulated in miRNA-204 transgenic embryonic hearts. Furthermore, we demonstrated that miRNA-204 directly targeted Jarid2. Knockdown of Jarid2 mimicked the pro-proliferative effect of miRNA-204 overexpression on cultured rat cardiomyocytes, whereas enhanced expression of Jarid2 conferred the myocytes with substantial resistance to proliferation by miRNA-204 overexpression. CONCLUSION Our findings identify a conserved role for miRNA-204 in regulating cardiomyocyte proliferation by targeting the Jarid2 signaling pathway.
Collapse
Affiliation(s)
- Dandan Liang
- Key Laboratory of Arrhythmias of the Ministry of Education of China, East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Research Center for Translational Medicine, Tongji University School of Medicine, Shanghai 200120, China; Institute of Medical Genetics, Tongji University, Shanghai 200092, China
| | - Jun Li
- Key Laboratory of Arrhythmias of the Ministry of Education of China, East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Research Center for Translational Medicine, Tongji University School of Medicine, Shanghai 200120, China; Institute of Medical Genetics, Tongji University, Shanghai 200092, China
| | - Yahan Wu
- Key Laboratory of Arrhythmias of the Ministry of Education of China, East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Department of Cardiology, East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Department of Pathology and Pathophysiology, Tongji University School of Medicine, Shanghai 200092, China
| | - Lixiao Zhen
- Key Laboratory of Arrhythmias of the Ministry of Education of China, East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Department of Cardiology, East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Department of Pathology and Pathophysiology, Tongji University School of Medicine, Shanghai 200092, China
| | - Changming Li
- Key Laboratory of Arrhythmias of the Ministry of Education of China, East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Department of Cardiology, East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Department of Pathology and Pathophysiology, Tongji University School of Medicine, Shanghai 200092, China
| | - Man Qi
- Key Laboratory of Arrhythmias of the Ministry of Education of China, East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Department of Cardiology, East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Department of Pathology and Pathophysiology, Tongji University School of Medicine, Shanghai 200092, China
| | - Lijie Wang
- Key Laboratory of Arrhythmias of the Ministry of Education of China, East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Department of Cardiology, East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Department of Pathology and Pathophysiology, Tongji University School of Medicine, Shanghai 200092, China
| | - Fangfei Deng
- Key Laboratory of Arrhythmias of the Ministry of Education of China, East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Department of Cardiology, East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Department of Pathology and Pathophysiology, Tongji University School of Medicine, Shanghai 200092, China
| | - Jian Huang
- Key Laboratory of Arrhythmias of the Ministry of Education of China, East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Department of Cardiology, East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Department of Pathology and Pathophysiology, Tongji University School of Medicine, Shanghai 200092, China
| | - Fei Lv
- Key Laboratory of Arrhythmias of the Ministry of Education of China, East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Department of Cardiology, East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Department of Pathology and Pathophysiology, Tongji University School of Medicine, Shanghai 200092, China
| | - Yi Liu
- Key Laboratory of Arrhythmias of the Ministry of Education of China, East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Research Center for Translational Medicine, Tongji University School of Medicine, Shanghai 200120, China; Institute of Medical Genetics, Tongji University, Shanghai 200092, China
| | - Xiue Ma
- Key Laboratory of Arrhythmias of the Ministry of Education of China, East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Department of Cardiology, East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Department of Pathology and Pathophysiology, Tongji University School of Medicine, Shanghai 200092, China
| | - Zuoren Yu
- Key Laboratory of Arrhythmias of the Ministry of Education of China, East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Research Center for Translational Medicine, Tongji University School of Medicine, Shanghai 200120, China
| | - Yuzhen Zhang
- Key Laboratory of Arrhythmias of the Ministry of Education of China, East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Research Center for Translational Medicine, Tongji University School of Medicine, Shanghai 200120, China
| | - Yi-Han Chen
- Key Laboratory of Arrhythmias of the Ministry of Education of China, East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Research Center for Translational Medicine, Tongji University School of Medicine, Shanghai 200120, China; Institute of Medical Genetics, Tongji University, Shanghai 200092, China; Department of Cardiology, East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Department of Pathology and Pathophysiology, Tongji University School of Medicine, Shanghai 200092, China.
| |
Collapse
|
40
|
Brodarac A, Šarić T, Oberwallner B, Mahmoodzadeh S, Neef K, Albrecht J, Burkert K, Oliverio M, Nguemo F, Choi YH, Neiss WF, Morano I, Hescheler J, Stamm C. Susceptibility of murine induced pluripotent stem cell-derived cardiomyocytes to hypoxia and nutrient deprivation. Stem Cell Res Ther 2015; 6:83. [PMID: 25900017 PMCID: PMC4445302 DOI: 10.1186/s13287-015-0057-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Revised: 05/23/2014] [Accepted: 03/19/2015] [Indexed: 01/06/2023] Open
Abstract
Introduction Induced pluripotent stem cell-derived cardiomyocytes (iPS-CMs) may be suitable for myocardial repair. While their functional and structural properties have been extensively investigated, their response to ischemia-like conditions has not yet been clearly defined. Methods iPS-CMs were differentiated and enriched from murine induced pluripotent stem cells expressing enhanced green fluorescent protein (eGFP) and puromycin resistance genes under the control of an α-myosin heavy chain (α-MHC) promoter. iPS-CMs maturity and function were characterized by microscopy, real-time PCR, calcium transient recordings, electrophysiology, and mitochondrial function assays, and compared to those from neonatal murine cardiomyocytes. iPS-CMs as well as neonatal murine cardiomyocytes were exposed for 3 hours to hypoxia (1% O2) and glucose/serum deprivation, and viability, apoptosis markers, reactive oxygen species, mitochondrial membrane potential and intracellular stress signaling cascades were investigated. Then, the iPS-CMs response to mesenchymal stromal cell-conditioned medium was determined. Results iPS-CMs displayed key morphological and functional properties that were comparable to those of neonatal cardiomyocytes, but several parameters indicated an earlier iPS-CMs maturation stage. During hypoxia and glucose/serum deprivation, iPS-CMs exhibited a significantly higher proportion of poly-caspase-active, 7-aminoactinomycin D-positive and TUNEL-positive cells than neonatal cardiomyocytes. The average mitochondrial membrane potential was reduced in “ischemic” iPS-CMs but remained unchanged in neonatal cardiomyocytes; reactive oxygen species production was only increased in “ischemic” iPS-CMs, and oxidoreductase activity in iPS-CMs dropped more rapidly than in neonatal cardiomyocytes. In iPS-CMs, hypoxia and glucose/serum deprivation led to upregulation of Hsp70 transcripts and decreased STAT3 phosphorylation and total PKCε protein expression. Treatment with mesenchymal stromal cell-conditioned medium preserved oxidoreductase activity and restored pSTAT3 and PKCε levels. Conclusion iPS-CMs appear to be particularly sensitive to hypoxia and nutrient deprivation. Counteracting the ischemic susceptibility of iPS-CMs with mesenchymal stromal cell-conditioned medium may help enhance their survival and efficacy in cell-based approaches for myocardial repair.
Collapse
Affiliation(s)
- Andreja Brodarac
- Berlin-Brandenburg Center for Regenerative Therapies, Föhrer Str.15, Berlin, 13353, Germany.
| | - Tomo Šarić
- Center for Physiology and Pathophysiology, Institute for Neurophysiology, Medical Faculty, University of Cologne, Cologne, Germany.
| | - Barbara Oberwallner
- Berlin-Brandenburg Center for Regenerative Therapies, Föhrer Str.15, Berlin, 13353, Germany.
| | | | - Klaus Neef
- Department of Cardiothoracic Surgery, Heart Center, University Hospital Cologne, Cologne, Germany.
| | - Julie Albrecht
- Center for Physiology and Pathophysiology, Institute for Neurophysiology, Medical Faculty, University of Cologne, Cologne, Germany.
| | - Karsten Burkert
- Center for Physiology and Pathophysiology, Institute for Neurophysiology, Medical Faculty, University of Cologne, Cologne, Germany.
| | - Matteo Oliverio
- Max-Planck-Institute for Metabolism Research, Cologne, Germany.
| | - Filomain Nguemo
- Center for Physiology and Pathophysiology, Institute for Neurophysiology, Medical Faculty, University of Cologne, Cologne, Germany.
| | - Yeong-Hoon Choi
- Department of Cardiothoracic Surgery, Heart Center, University Hospital Cologne, Cologne, Germany.
| | - Wolfram F Neiss
- Department of Anatomy I, Medical Faculty, University of Cologne, Cologne, Germany.
| | - Ingo Morano
- Max-Delbrueck-Center for Molecular Medicine, Berlin, Germany.
| | - Jürgen Hescheler
- Center for Physiology and Pathophysiology, Institute for Neurophysiology, Medical Faculty, University of Cologne, Cologne, Germany.
| | - Christof Stamm
- Berlin-Brandenburg Center for Regenerative Therapies, Föhrer Str.15, Berlin, 13353, Germany. .,Deutsches Herzzentrum Berlin, Berlin, Germany.
| |
Collapse
|
41
|
Cannon L, Yu ZY, Marciniec T, Waardenberg AJ, Iismaa SE, Nikolova-Krstevski V, Neist E, Ohanian M, Qiu MR, Rainer S, Harvey RP, Feneley MP, Graham RM, Fatkin D. Irreversible triggers for hypertrophic cardiomyopathy are established in the early postnatal period. J Am Coll Cardiol 2015; 65:560-9. [PMID: 25677315 DOI: 10.1016/j.jacc.2014.10.069] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Revised: 10/09/2014] [Accepted: 10/28/2014] [Indexed: 12/12/2022]
Abstract
BACKGROUND Hypertrophic cardiomyopathy (HCM) is caused by mutations in sarcomere protein genes, and left ventricular hypertrophy (LVH) develops as an adaptive response to sarcomere dysfunction. It remains unclear whether persistent expression of the mutant gene is required for LVH or whether early gene expression acts as an immutable inductive trigger. OBJECTIVES The aim of this study was to use a regulatable murine model of HCM to study the reversibility of pathological LVH. METHODS The authors generated a double-transgenic mouse model, tTAxαMHCR403Q, in which expression of the HCM-causing Arg403Gln mutation in the α-myosin heavy chain (MHC) gene is inhibited by doxycycline administration. Cardiac structure and function were evaluated in groups of mice that received doxycycline for varying periods from 0 to 40 weeks of age. RESULTS Untreated tTAxαMHCR403Q mice showed increased left ventricular (LV) mass, contractile dysfunction, myofibrillar disarray, and fibrosis. In contrast, mice treated with doxycycline from conception to 6 weeks had markedly less LVH and fibrosis at 40 weeks. Transgene inhibition from 6 weeks reduced fibrosis but did not prevent LVH or functional changes. There were no differences in LV parameters at 40 weeks between mice with transgene inhibition from 20 weeks and mice with continuous transgene expression. CONCLUSIONS These findings highlight the critical role of the early postnatal period in HCM pathogenesis and suggest that mutant sarcomeres manifest irreversible cardiomyocyte defects that induce LVH. In HCM, mutation-silencing therapies are likely to be ineffective for hypertrophy regression and would have to be administered very early in life to prevent hypertrophy development.
Collapse
Affiliation(s)
- Leah Cannon
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Darlinghurst, Australia
| | - Ze-Yan Yu
- Cardiac Physiology and Transplantation Division, Victor Chang Cardiac Research Institute, Darlinghurst, Australia
| | - Tadeusz Marciniec
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Darlinghurst, Australia
| | - Ashley J Waardenberg
- Cardiac Developmental and Stem Cell Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, Australia
| | - Siiri E Iismaa
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Darlinghurst, Australia; Faculty of Medicine, University of New South Wales, Kensington, Australia
| | - Vesna Nikolova-Krstevski
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Darlinghurst, Australia; Faculty of Medicine, University of New South Wales, Kensington, Australia
| | - Elysia Neist
- Cardiac Physiology and Transplantation Division, Victor Chang Cardiac Research Institute, Darlinghurst, Australia
| | - Monique Ohanian
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Darlinghurst, Australia
| | - Min Ru Qiu
- Anatomical Pathology Department, St. Vincent's Hospital, Darlinghurst, Australia
| | - Stephen Rainer
- Anatomical Pathology Department, St. Vincent's Hospital, Darlinghurst, Australia
| | - Richard P Harvey
- Cardiac Developmental and Stem Cell Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, Australia; Faculty of Medicine, University of New South Wales, Kensington, Australia; School of Biotechnology and Biomolecular Sciences, University of New South Wales, Kensington, Australia
| | - Michael P Feneley
- Cardiac Physiology and Transplantation Division, Victor Chang Cardiac Research Institute, Darlinghurst, Australia; Faculty of Medicine, University of New South Wales, Kensington, Australia; Cardiology Department, St. Vincent's Hospital, Darlinghurst, Australia
| | - Robert M Graham
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Darlinghurst, Australia; Faculty of Medicine, University of New South Wales, Kensington, Australia; School of Biotechnology and Biomolecular Sciences, University of New South Wales, Kensington, Australia; Cardiology Department, St. Vincent's Hospital, Darlinghurst, Australia.
| | - Diane Fatkin
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Darlinghurst, Australia; Faculty of Medicine, University of New South Wales, Kensington, Australia; Cardiology Department, St. Vincent's Hospital, Darlinghurst, Australia.
| |
Collapse
|
42
|
McLean BA, Zhabyeyev P, Patel VB, Basu R, Parajuli N, DesAulniers J, Murray AG, Kassiri Z, Vanhaesebroeck B, Oudit GY. PI3Kα is essential for the recovery from Cre/tamoxifen cardiotoxicity and in myocardial insulin signalling but is not required for normal myocardial contractility in the adult heart. Cardiovasc Res 2015; 105:292-303. [DOI: 10.1093/cvr/cvv016] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
|
43
|
Blackmore HL, Niu Y, Fernandez-Twinn DS, Tarry-Adkins JL, Giussani DA, Ozanne SE. Maternal diet-induced obesity programs cardiovascular dysfunction in adult male mouse offspring independent of current body weight. Endocrinology 2014; 155:3970-80. [PMID: 25051449 PMCID: PMC4255219 DOI: 10.1210/en.2014-1383] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Obese pregnancies are not only associated with adverse consequences for the mother but also the long-term health of her child. Human studies have shown that individuals from obese mothers are at increased risk of premature death from cardiovascular disease (CVD), but are unable to define causality. This study aimed to determine causality using a mouse model of maternal diet-induced obesity. Obesity was induced in female C57BL/6 mice by feeding a diet rich in simple sugars and saturated fat 6 weeks prior to pregnancy and throughout pregnancy and lactation. Control females were fed laboratory chow. Male offspring from both groups were weaned onto chow and studied at 3, 5, 8, and 12 weeks of age for gross cardiac morphometry using stereology, cardiomyocyte cell area by histology, and cardiac fetal gene expression using qRT-PCR. Cardiac function was assessed by isolated Langendorff technology at 12 weeks of age and hearts were analyzed at the protein level for the expression of the β1 adrenergic receptor, muscarinic type-2 acetylcholine receptor, and proteins involved in cardiac contraction. Offspring from obese mothers develop pathologic cardiac hypertrophy associated with re-expression of cardiac fetal genes. By young adulthood these offspring developed severe systolic and diastolic dysfunction and cardiac sympathetic dominance. Importantly, cardiac dysfunction occurred in the absence of any change in corresponding body weight and despite the offspring eating a healthy low-fat diet. These findings provide a causal link to explain human observations relating maternal obesity with premature death from CVD in her offspring.
Collapse
Affiliation(s)
- Heather L Blackmore
- University of Cambridge, Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science (H.L.B., D.S.F.-T., J.L.T.-A., S.E.O.), Addenbrookes Hospital, Cambridge CB2 0QQ, United Kingdom; and Department of Physiology, Development, and Neuroscience (Y.N., D.A.G.), University of Cambridge, Cambridge CB2 3EG, United Kingdom
| | | | | | | | | | | |
Collapse
|
44
|
MiRiad Roles for MicroRNAs in Cardiac Development and Regeneration. Cells 2014; 3:724-50. [PMID: 25055156 PMCID: PMC4197632 DOI: 10.3390/cells3030724] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Revised: 06/25/2014] [Accepted: 07/08/2014] [Indexed: 12/20/2022] Open
Abstract
Cardiac development is an exquisitely regulated process that is sensitive to perturbations in transcriptional activity and gene dosage. Accordingly, congenital heart abnormalities are prevalent worldwide, and are estimated to occur in approximately 1% of live births. Recently, small non-coding RNAs, known as microRNAs, have emerged as critical components of the cardiogenic regulatory network, and have been shown to play numerous roles in the growth, differentiation, and morphogenesis of the developing heart. Moreover, the importance of miRNA function in cardiac development has facilitated the identification of prospective therapeutic targets for patients with congenital and acquired cardiac diseases. Here, we discuss findings attesting to the critical role of miRNAs in cardiogenesis and cardiac regeneration, and present evidence regarding the therapeutic potential of miRNAs for cardiovascular diseases.
Collapse
|
45
|
Han Y, Zhang JP, Qian JQ, Hu CQ. Cardiotoxicity evaluation of anthracyclines in zebrafish (Danio rerio). J Appl Toxicol 2014; 35:241-52. [PMID: 24853142 DOI: 10.1002/jat.3007] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2013] [Revised: 01/12/2014] [Accepted: 02/10/2014] [Indexed: 12/14/2022]
Abstract
Drug-induced cardiotoxicity is a leading factor for drug withdrawals, and limits drug efficacy and clinical use. Therefore, new alternative animal models and methods for drug safety evaluation have been given great attention. Anthracyclines (ANTs) are widely prescribed anticancer agents that have a cumulative dose relationship with cardiotoxicity. We performed experiments to study the toxicity of ANTs in early developing zebrafish embryos, especially their effects on the heart. LC50 values for daunorubicin, pirarubicin, doxorubicin (DOX), epirubicin and DOX-liposome at 72 h post-fertilization were 122.7 μM, 111.9 μM, 31.2 μM, 108.3 μM and 55.8 μM, respectively. At the same time, zebrafish embryos were exposed to ANTs in three exposure stages and induced incomplete looping of the heart tube, pericardia edema and bradycardia in a dose-dependent manner, eventually leading to death. DOX caused the greatest heart defects in the treatment stages and its liposome reduced the effects on the heart, while daunorubicin produced the least toxicity. Genes and proteins related to heart development were also identified to be sensitive to ANT exposure and downregulated by ANTs. It revealed ANTs could disturb the heart formation and development. ANTs induced cardiotoxicity in zebrafish has similar effects in mammalian models, indicating that zebrafish may have a potential value for assessment of drug-induced developmental cardiotoxicity.
Collapse
Affiliation(s)
- Ying Han
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | | | | | | |
Collapse
|
46
|
Persistent overexpression of phosphoglycerate mutase, a glycolytic enzyme, modifies energy metabolism and reduces stress resistance of heart in mice. PLoS One 2013; 8:e72173. [PMID: 23951293 PMCID: PMC3741204 DOI: 10.1371/journal.pone.0072173] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2012] [Accepted: 07/10/2013] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Heart failure is associated with changes in cardiac energy metabolism. Glucose metabolism in particular is thought to be important in the pathogenesis of heart failure. We examined the effects of persistent overexpression of phosphoglycerate mutase 2 (Pgam2), a glycolytic enzyme, on cardiac energy metabolism and function. METHODS AND RESULTS Transgenic mice constitutively overexpressing Pgam2 in a heart-specific manner were generated, and cardiac energy metabolism and function were analyzed. Cardiac function at rest was normal. The uptake of analogs of glucose or fatty acids and the phosphocreatine/βATP ratio at rest were normal. A comprehensive metabolomic analysis revealed an increase in the levels of a few metabolites immediately upstream and downstream of Pgam2 in the glycolytic pathway, whereas the levels of metabolites in the initial few steps of glycolysis and lactate remained unchanged. The levels of metabolites in the tricarboxylic acid (TCA) cycle were altered. The capacity for respiration by isolated mitochondria in vitro was decreased, and that for the generation of reactive oxygen species (ROS) in vitro was increased. Impaired cardiac function was observed in response to dobutamine. Mice developed systolic dysfunction upon pressure overload. CONCLUSIONS Constitutive overexpression of Pgam2 modified energy metabolism and reduced stress resistance of heart in mice.
Collapse
|
47
|
Anene-Nzelu CG, Choudhury D, Li H, Fraiszudeen A, Peh KY, Toh YC, Ng SH, Leo HL, Yu H. Scalable cell alignment on optical media substrates. Biomaterials 2013; 34:5078-87. [PMID: 23601659 DOI: 10.1016/j.biomaterials.2013.03.070] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2013] [Accepted: 03/23/2013] [Indexed: 01/26/2023]
Abstract
Cell alignment by underlying topographical cues has been shown to affect important biological processes such as differentiation and functional maturation in vitro. However, the routine use of cell culture substrates with micro- or nano-topographies, such as grooves, is currently hampered by the high cost and specialized facilities required to produce these substrates. Here we present cost-effective commercially available optical media as substrates for aligning cells in culture. These optical media, including CD-R, DVD-R and optical grating, allow different cell types to attach and grow well on them. The physical dimension of the grooves in these optical media allowed cells to be aligned in confluent cell culture with maximal cell-cell interaction and these cell alignment affect the morphology and differentiation of cardiac (H9C2), skeletal muscle (C2C12) and neuronal (PC12) cell lines. The optical media is amenable to various chemical modifications with fibronectin, laminin and gelatin for culturing different cell types. These low-cost commercially available optical media can serve as scalable substrates for research or drug safety screening applications in industry scales.
Collapse
|
48
|
Crowder SW, Liang Y, Rath R, Park AM, Maltais S, Pintauro PN, Hofmeister W, Lim CC, Wang X, Sung HJ. Poly(ε-caprolactone)-carbon nanotube composite scaffolds for enhanced cardiac differentiation of human mesenchymal stem cells. Nanomedicine (Lond) 2013; 8:1763-76. [PMID: 23530764 DOI: 10.2217/nnm.12.204] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
AIM To evaluate the efficacy of electrically conductive, biocompatible composite scaffolds in modulating the cardiomyogenic differentiation of human mesenchymal stem cells (hMSCs). MATERIALS & METHODS Electrospun scaffolds of poly(ε-caprolactone) with or without carbon nanotubes were developed to promote the in vitro cardiac differentiation of hMSCs. RESULTS Results indicate that hMSC differentiation can be enhanced by either culturing in electrically conductive, carbon nanotube-containing composite scaffolds without electrical stimulation in the presence of 5-azacytidine, or extrinsic electrical stimulation in nonconductive poly(ε-caprolactone) scaffolds without carbon nanotube and azacytidine. CONCLUSION This study suggests a first step towards improving hMSC cardiomyogenic differentiation for local delivery into the infarcted myocardium.
Collapse
Affiliation(s)
- Spencer W Crowder
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Sontag DP, Wang J, Kardami E, Cattini PA. FGF-2 and FGF-16 Protect Isolated Perfused Mouse Hearts from Acute Doxorubicin-Induced Contractile Dysfunction. Cardiovasc Toxicol 2013; 13:244-53. [DOI: 10.1007/s12012-013-9203-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
50
|
Li M, Chen Y, Bi Y, Jiang W, Luo Q, He Y, Su Y, Liu X, Cui J, Zhang W, Li R, Kong Y, Zhang J, Wang J, Zhang H, Shui W, Wu N, Zhu J, Tian J, Yi QJ, Luu HH, Haydon RC, He TC, Zhu GH. Establishment and characterization of the reversibly immortalized mouse fetal heart progenitors. Int J Med Sci 2013; 10:1035-46. [PMID: 23801891 PMCID: PMC3691803 DOI: 10.7150/ijms.6639] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2013] [Accepted: 06/09/2013] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVE Progenitor cell-based cardiomyocyte regeneration holds great promise of repairing an injured heart. Although cardiomyogenic differentiation has been reported for a variety of progenitor cell types, the biological factors that regulate effective cardiomyogenesis remain largely undefined. Primary cardiomyogenic progenitors (CPs) have a limited life span in culture, hampering the CPs' in vitro and in vivo studies. The objective of this study is to investigate if primary CPs isolated from fetal mouse heart can be reversibly immortalized with SV40 large T and maintain long-term cell proliferation without compromising cardiomyogenic differentiation potential. METHODS Primary cardiomyocytes were isolated from mouse E15.5 fetal heart, and immortalized retrovirally with the expression of SV40 large T antigen flanked with loxP sites. Expression of cardiomyogenic markers were determined by quantitative RT-PCR and immunofluorescence staining. The immortalization phenotype was reversed by using an adenovirus-mediated expression of the Cre reconbinase. Cardiomyogenic differentiation induced by retinoids or dexamethasone was assessed by an α-myosin heavy chain (MyHC) promoter-driven reporter. RESULTS We demonstrate that the CPs derived from mouse E15.5 fetal heart can be efficiently immortalized by SV40 T antigen. The conditionally immortalized CPs (iCP15 clones) exhibit an increased proliferative activity and are able to maintain long-term proliferation, which can be reversed by Cre recombinase. The iCP15 cells express cardiomyogenic markers and retain differentiation potential as they can undergo terminal differentiate into cardiomyctes under appropriate differentiation conditions although the iCP15 clones represent a large repertoire of CPs at various differentiation stages. The removal of SV40 large T increases the iCPs' differentiation potential. Thus, the iCPs not only maintain long-term cell proliferative activity but also retain cardiomyogenic differentiation potential. CONCLUSIONS Our results suggest that the reported reversible SV40 T antigen-mediated immortalization represents an efficient approach for establishing long-term culture of primary cardiomyogenic progenitors for basic and translational research.
Collapse
Affiliation(s)
- Mi Li
- Stem Cell Biology and Therapy Laboratory, the Key Laboratory of Pediatrics Designated by Chinese Ministry of Education and Chongqing Bureau of Education, and the Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|