1
|
Souza NSC, Barenco-Marins T, Ferraz AP, Barbosa RAQ, Maciel L, Ponte CG, Seara FAC, Olivares EL, Nascimento JHM. Low Thyroid Hormones Level Attenuates Mitochondrial Dysfunction and Right Ventricular Failure in Pulmonary Hypertensive Rats. Cardiovasc Drugs Ther 2024:10.1007/s10557-024-07618-5. [PMID: 39215901 DOI: 10.1007/s10557-024-07618-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/13/2024] [Indexed: 09/04/2024]
Abstract
PURPOSE This study is to investigate the repercussions of hypothyroidism in the pathophysiological progression of pulmonary arterial hypertension (PAH). METHODS While the control (CTL, n = 5) male Wistar rats received vehicle, PAH was induced with monocrotaline (MCT group, n = 15). Hypothyroidism was induced in a subset of rats by methimazole 3 weeks prior to the MCT injection (MMZ + MCT group, n = 15). Plasma thyroid hormones were measured by radioimmunoassay. Electrocardiographic, echocardiographic, and hemodynamic analyses were performed to evaluate the progression of PAH. Gene expression of antioxidant enzymes and cardiac hypertrophy markers were assessed by qPCR. Mitochondrial respiration, ATP levels, and ROS production were measured in right ventricular (RV) samples. RESULTS Plasma T3 and T4 decreased in both MCT and MMZ + MCT groups (p < 0.05). Right ventricular systolic pressure (RVSP) increased, and RV - dP/dt, + dP/dt, and contractility index decreased in the MCT versus the CTL group and remained within control levels in the MMZ + MCT group (p < 0.05). Relative RV weight, RV wall thickness, RV diastolic area, and relative lung weight were augmented in the MCT versus the CTL group, whereas all parameters were improved to the CTL levels in the MMZ + MCT group (p < 0.05). Only the MCT group exhibited an increased duration of QTc interval compared to the baseline period (p < 0.05). ADP-induced mitochondrial respiration and ATP levels were decreased, and ROS production was increased in MCT versus the CTL group (p < 0.05), while the MMZ + MCT group exhibited increased mitochondrial respiration versus the MCT group (p < 0.05). CONCLUSION Hypothyroidism attenuated the RV mitochondrial dysfunction and the pathophysiological progression of MCT-induced PAH.
Collapse
Affiliation(s)
- Natalia Soares Carvalho Souza
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, 373 Carlos Chagas Filho Avenue, Rio de Janeiro, Brazil
| | - Thais Barenco-Marins
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, 373 Carlos Chagas Filho Avenue, Rio de Janeiro, Brazil
| | - Ana Paula Ferraz
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, 373 Carlos Chagas Filho Avenue, Rio de Janeiro, Brazil
| | - Raiana Andrade Quintanilha Barbosa
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, 373 Carlos Chagas Filho Avenue, Rio de Janeiro, Brazil
| | - Leonardo Maciel
- Campus Professor Geraldo Cidade, Universidade Federal do Rio de Janeiro, Duque de Caxias, Brazil
| | | | - Fernando Azevedo Cruz Seara
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, 373 Carlos Chagas Filho Avenue, Rio de Janeiro, Brazil.
| | - Emerson Lopes Olivares
- Instituto de Ciências Biológicas e da Saúde, Universidade Federal Rural do Rio de Janeiro, Seropédica, Brazil
- Sociedade Brasileira de Fisiologia, Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas, São Paulo, Brazil
| | - Jose Hamilton Matheus Nascimento
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, 373 Carlos Chagas Filho Avenue, Rio de Janeiro, Brazil
| |
Collapse
|
2
|
Mably JD, Wang DZ. Long non-coding RNAs in cardiac hypertrophy and heart failure: functions, mechanisms and clinical prospects. Nat Rev Cardiol 2024; 21:326-345. [PMID: 37985696 PMCID: PMC11031336 DOI: 10.1038/s41569-023-00952-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/16/2023] [Indexed: 11/22/2023]
Abstract
The surge in reports describing non-coding RNAs (ncRNAs) has focused attention on their possible biological roles and effects on development and disease. ncRNAs have been touted as previously uncharacterized regulators of gene expression and cellular processes, possibly working to fine-tune these functions. The sheer number of ncRNAs identified has outpaced the capacity to characterize each molecule thoroughly and to reliably establish its clinical relevance; it has, nonetheless, created excitement about their potential as molecular targets for novel therapeutic approaches to treat human disease. In this Review, we focus on one category of ncRNAs - long non-coding RNAs - and their expression, functions and molecular mechanisms in cardiac hypertrophy and heart failure. We further discuss the prospects for this specific class of ncRNAs as novel targets for the diagnosis and treatment of these conditions.
Collapse
Affiliation(s)
- John D Mably
- Center for Regenerative Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
- USF Health Heart Institute, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Da-Zhi Wang
- Center for Regenerative Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, USA.
- USF Health Heart Institute, Morsani College of Medicine, University of South Florida, Tampa, FL, USA.
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, USA.
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA.
| |
Collapse
|
3
|
Julovi SM, Trinh K, Robertson H, Xu C, Minhas N, Viswanathan S, Patrick E, Horowitz JD, Meijles DN, Rogers NM. Thrombospondin-1 Drives Cardiac Remodeling in Chronic Kidney Disease. JACC Basic Transl Sci 2024; 9:607-627. [PMID: 38984053 PMCID: PMC11228122 DOI: 10.1016/j.jacbts.2024.01.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 01/29/2024] [Accepted: 01/29/2024] [Indexed: 07/11/2024]
Abstract
Patients with chronic kidney disease (CKD) face a high risk of cardiovascular disease. Previous studies reported that endogenous thrombospondin 1 (TSP1) involves right ventricular remodeling and dysfunction. Here we show that a murine model of CKD increased myocardial TSP1 expression and produced left ventricular hypertrophy, fibrosis, and dysfunction. TSP1 knockout mice were protected from these features. In vitro, indoxyl sulfate is driving deleterious changes in cardiomyocyte through the TSP1. In patients with CKD, TSP1 and aryl hydrocarbon receptor were both differentially expressed in the myocardium. Our findings summon large clinical studies to confirm the translational role of TSP1 in patients with CKD.
Collapse
Affiliation(s)
- Sohel M Julovi
- Kidney Injury Group, Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Camperdown, New South Wales, Australia
| | - Katie Trinh
- Kidney Injury Group, Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, New South Wales, Australia
| | - Harry Robertson
- Kidney Injury Group, Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Camperdown, New South Wales, Australia
- Sydney Precision Data Science Centre, University of Sydney, New South Wales, Australia
| | - Cuicui Xu
- Kidney Injury Group, Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, New South Wales, Australia
| | - Nikita Minhas
- Kidney Injury Group, Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, New South Wales, Australia
| | - Seethalakshmi Viswanathan
- Faculty of Medicine and Health, The University of Sydney, Camperdown, New South Wales, Australia
- Tissue Pathology and Diagnostic Oncology, Institute of Clinical Pathology and Medical Research, Westmead Hospital, Westmead, New South Wales, Australia
| | - Ellis Patrick
- Kidney Injury Group, Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- Sydney Precision Data Science Centre, University of Sydney, New South Wales, Australia
- School of Mathematics, University of Sydney, New South Wales, Australia
- Laboratory of Data Discovery for Health Limited (D24H), Science Park, Hong Kong Special Administrative Region, China
| | - John D Horowitz
- Molecular and Clinical Sciences Research Institute, St George's University of London, London, United Kingdom
- Cardiovascular Pathophysiology and Therapeutics Research Group, Basil Hetzel Institute, Woodville, South Australia, Australia
- Department of Medicine, The University of Adelaide, Adelaide, South Australia, Australia
| | - Daniel N Meijles
- Molecular and Clinical Sciences Research Institute, St George's University of London, London, United Kingdom
| | - Natasha M Rogers
- Kidney Injury Group, Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Camperdown, New South Wales, Australia
- Renal and Transplantation Unit, Westmead Hospital, New South Wales, Australia
| |
Collapse
|
4
|
Rishiq A, Islam O, Golomb E, Gilon D, Smith Y, Savchenko I, Eliaz R, Foo RS, Razin E, Tshori S. The Role Played by Transcription Factor E3 in Modulating Cardiac Hypertrophy. Int Heart J 2021; 62:1358-1368. [PMID: 34744144 DOI: 10.1536/ihj.21-088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Transcription factor E3 (TFE3), which is a key regulator of cellular adaptation, is expressed in most tissues, including the heart, and is reportedly overexpressed during cardiac hypertrophy. In this study, TFE3's role in cardiac hypertrophy was investigated. To understand TFE3's physiological importance in cardiac hypertrophy, pressure-overload cardiac hypertrophy was induced through transverse aortic constriction (TAC) in both wild-type (WT) and TFE3 knockout mice (TFE3-/-). Eleven weeks after TAC induction, cardiac hypertrophy was observed in both WT and TFE3-/- mice. However, significant reductions in ejection fraction and fractional shortening were observed in WT mice compared to TFE3-/- mice. To understand the mechanism, we found that myosin heavy chain (Myh7), which increases during hemodynamic overload, was lower in TFE3-/- TAC mice than in WT TAC mice, whereas extracellular signal-regulated protein kinases (ERK) phosphorylation, which confers cardioprotection, was lower in the left ventricles of WT mice than in TFE3-/- mice. We also found high expressions of TFE3, histone, and MYH7 and low expression of pERK in the normal human heart compared to the hypertensive heart. In the H9c2 cell line, we found that ERK inhibition caused TFE3 nuclear localization. In addition, we found that MYH7 was associated with TFE3, and during TFE3 knockdown, MYH7 and histone were downregulated. Therefore, we showed that TFE3 expression was increased in the mouse model of cardiac hypertrophy and tissues from human hypertensive hearts, whereas pERK was decreased reversibly, which suggested that TFE3 is involved in cardiac hypertrophy through TFE3-histone-MYH7-pERK signaling.
Collapse
Affiliation(s)
- Ahmed Rishiq
- Department of Biochemistry and Molecular Biology, Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School
| | - Omedul Islam
- NUS-HUJ-CREATE Cellular & Molecular Mechanisms of Inflammation Program, Department of Microbiology and Immunology
| | - Eliahu Golomb
- Department of Pathology, Shaare Zedek Medical Center
| | - Dan Gilon
- Heart Institute, Hadassah Hebrew University Medical Center
| | - Yoav Smith
- Unit of Genomic Data Analysis, The Hebrew University-Hadassah Medical School
| | | | - Ran Eliaz
- Heart Institute, Hadassah Hebrew University Medical Center
| | - Roger Sy Foo
- Cardiovascular Research institute, Center of Translational Medicine
| | - Ehud Razin
- Department of Biochemistry and Molecular Biology, Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School
- NUS-HUJ-CREATE Cellular & Molecular Mechanisms of Inflammation Program, Department of Microbiology and Immunology
| | - Sagi Tshori
- Department of Biochemistry and Molecular Biology, Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School
- Cardiac Research Laboratory, Kaplan Medical Center
| |
Collapse
|
5
|
Bao J, Lu Y, She Q, Dou W, Tang R, Xu X, Zhang M, Zhu L, Zhou Q, Li H, Zhou G, Yang Z, Shi S, Liu Z, Zheng C. MicroRNA-30 regulates left ventricular hypertrophy in chronic kidney disease. JCI Insight 2021; 6:138027. [PMID: 33848263 PMCID: PMC8262338 DOI: 10.1172/jci.insight.138027] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 04/07/2021] [Indexed: 12/04/2022] Open
Abstract
Left ventricular hypertrophy (LVH) is a primary feature of cardiovascular complications in patients with chronic kidney disease (CKD). miRNA-30 is an important posttranscriptional regulator of LVH, but it is unknown whether miRNA-30 participates in the process of CKD-induced LVH. In the present study, we found that CKD not only resulted in LVH but also suppressed miRNA-30 expression in the myocardium. Rescue of cardiomyocyte-specific miRNA-30 attenuated LVH in CKD rats without altering CKD progression. Importantly, in vivo and in vitro knockdown of miRNA-30 in cardiomyocytes led to cardiomyocyte hypertrophy by upregulating the calcineurin signaling directly. Furthermore, CKD-related detrimental factors, such as fibroblast growth factor-23, uremic toxin, angiotensin II, and transforming growth factor–β, suppressed cardiac miRNA-30 expression, while miRNA-30 supplementation blunted cardiomyocyte hypertrophy induced by such factors. These results uncover a potentially novel mechanism of CKD-induced LVH and provide a potential therapeutic target for CKD patients with LVH. Downregulation of myocardial miRNA-30 is involved in chronic kidney disease–induced left ventricular hypertrophy, whereas exogenous miRNA-30 rescue inhibits this process.
Collapse
Affiliation(s)
- Jingfu Bao
- National Clinical Research Center of Kidney Diseases, and
| | - Yinghui Lu
- National Clinical Research Center of Kidney Diseases, and
| | - Qinying She
- National Clinical Research Center of Kidney Diseases, and
| | - Weijuan Dou
- National Clinical Research Center of Kidney Diseases, and
| | - Rong Tang
- National Clinical Research Center of Kidney Diseases, and
| | - Xiaodong Xu
- National Clinical Research Center of Kidney Diseases, and
| | - Mingchao Zhang
- National Clinical Research Center of Kidney Diseases, and
| | - Ling Zhu
- National Clinical Research Center of Kidney Diseases, and
| | - Qing Zhou
- Department of Pharmacology, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Hui Li
- Department of Pharmacology, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Guohua Zhou
- Department of Pharmacology, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Zhongzhou Yang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Cardiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University School of Medicine, and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Nanjing, China
| | - Shaolin Shi
- National Clinical Research Center of Kidney Diseases, and
| | - Zhihong Liu
- National Clinical Research Center of Kidney Diseases, and
| | - Chunxia Zheng
- National Clinical Research Center of Kidney Diseases, and
| |
Collapse
|
6
|
Ovics P, Regev D, Baskin P, Davidor M, Shemer Y, Neeman S, Ben-Haim Y, Binah O. Drug Development and the Use of Induced Pluripotent Stem Cell-Derived Cardiomyocytes for Disease Modeling and Drug Toxicity Screening. Int J Mol Sci 2020; 21:E7320. [PMID: 33023024 PMCID: PMC7582587 DOI: 10.3390/ijms21197320] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 09/23/2020] [Accepted: 09/27/2020] [Indexed: 12/19/2022] Open
Abstract
: Over the years, numerous groups have employed human induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) as a superb human-compatible model for investigating the function and dysfunction of cardiomyocytes, drug screening and toxicity, disease modeling and for the development of novel drugs for heart diseases. In this review, we discuss the broad use of iPSC-CMs for drug development and disease modeling, in two related themes. In the first theme-drug development, adverse drug reactions, mechanisms of cardiotoxicity and the need for efficient drug screening protocols-we discuss the critical need to screen old and new drugs, the process of drug development, marketing and Adverse Drug reactions (ADRs), drug-induced cardiotoxicity, safety screening during drug development, drug development and patient-specific effect and different mechanisms of ADRs. In the second theme-using iPSC-CMs for disease modeling and developing novel drugs for heart diseases-we discuss the rationale for using iPSC-CMs and modeling acquired and inherited heart diseases with iPSC-CMs.
Collapse
Affiliation(s)
- Paz Ovics
- Department of Physiology, Biophysics and Systems Biology, The Rappaport Institute, Ruth & Bruce Rappaport Faculty of Medicine, Technion, Haifa 31096, Israel; (P.O.); (D.R.); (P.B.); (M.D.); (Y.S.); (S.N.)
| | - Danielle Regev
- Department of Physiology, Biophysics and Systems Biology, The Rappaport Institute, Ruth & Bruce Rappaport Faculty of Medicine, Technion, Haifa 31096, Israel; (P.O.); (D.R.); (P.B.); (M.D.); (Y.S.); (S.N.)
| | - Polina Baskin
- Department of Physiology, Biophysics and Systems Biology, The Rappaport Institute, Ruth & Bruce Rappaport Faculty of Medicine, Technion, Haifa 31096, Israel; (P.O.); (D.R.); (P.B.); (M.D.); (Y.S.); (S.N.)
| | - Mor Davidor
- Department of Physiology, Biophysics and Systems Biology, The Rappaport Institute, Ruth & Bruce Rappaport Faculty of Medicine, Technion, Haifa 31096, Israel; (P.O.); (D.R.); (P.B.); (M.D.); (Y.S.); (S.N.)
| | - Yuval Shemer
- Department of Physiology, Biophysics and Systems Biology, The Rappaport Institute, Ruth & Bruce Rappaport Faculty of Medicine, Technion, Haifa 31096, Israel; (P.O.); (D.R.); (P.B.); (M.D.); (Y.S.); (S.N.)
| | - Shunit Neeman
- Department of Physiology, Biophysics and Systems Biology, The Rappaport Institute, Ruth & Bruce Rappaport Faculty of Medicine, Technion, Haifa 31096, Israel; (P.O.); (D.R.); (P.B.); (M.D.); (Y.S.); (S.N.)
| | - Yael Ben-Haim
- Institute of Molecular and Clinical Sciences, St. George’s University of London, London SW17 0RE, UK;
- Cardiology Clinical Academic Group, St. George’s University Hospitals NHS Foundation Trust, London SW17 0QT, UK
| | - Ofer Binah
- Department of Physiology, Biophysics and Systems Biology, The Rappaport Institute, Ruth & Bruce Rappaport Faculty of Medicine, Technion, Haifa 31096, Israel; (P.O.); (D.R.); (P.B.); (M.D.); (Y.S.); (S.N.)
| |
Collapse
|
7
|
Bjorkman KK, Guess MG, Harrison BC, Polmear MM, Peter AK, Leinwand LA. miR-206 enforces a slow muscle phenotype. J Cell Sci 2020; 133:jcs243162. [PMID: 32620696 PMCID: PMC7438006 DOI: 10.1242/jcs.243162] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Accepted: 06/25/2020] [Indexed: 12/21/2022] Open
Abstract
Striated muscle is a highly specialized collection of tissues with contractile properties that vary according to functional needs. Although muscle fiber types are established postnatally, lifelong plasticity facilitates stimulus-dependent adaptation. Functional adaptation requires molecular adaptation, which is partially provided by miRNA-mediated post-transcriptional regulation. miR-206 is a muscle-specific miRNA enriched in slow muscles. We investigated whether miR-206 drives the slow muscle phenotype or is merely an outcome. We found that miR-206 expression increases in both physiological (including female sex and endurance exercise) and pathological conditions (muscular dystrophy and adrenergic agonism) that promote a slow phenotype. Consistent with that observation, the slow soleus muscle of male miR-206-knockout mice displays a faster phenotype than wild-type mice. Moreover, left ventricles of male miR-206 knockout mice have a faster myosin profile, accompanied by dilation and systolic dysfunction. Thus, miR-206 appears to be necessary to enforce a slow skeletal and cardiac muscle phenotype and to play a key role in muscle sexual dimorphisms.
Collapse
Affiliation(s)
- Kristen K Bjorkman
- Department of Molecular, Cellular, and Developmental Biology, BioFrontiers Institute, University of Colorado Boulder, 3415 Colorado Ave., UCB596, Boulder, CO 80303, USA
| | - Martin G Guess
- Department of Molecular, Cellular, and Developmental Biology, BioFrontiers Institute, University of Colorado Boulder, 3415 Colorado Ave., UCB596, Boulder, CO 80303, USA
| | - Brooke C Harrison
- Department of Molecular, Cellular, and Developmental Biology, BioFrontiers Institute, University of Colorado Boulder, 3415 Colorado Ave., UCB596, Boulder, CO 80303, USA
| | - Michael M Polmear
- Department of Molecular, Cellular, and Developmental Biology, BioFrontiers Institute, University of Colorado Boulder, 3415 Colorado Ave., UCB596, Boulder, CO 80303, USA
| | - Angela K Peter
- Department of Molecular, Cellular, and Developmental Biology, BioFrontiers Institute, University of Colorado Boulder, 3415 Colorado Ave., UCB596, Boulder, CO 80303, USA
| | - Leslie A Leinwand
- Department of Molecular, Cellular, and Developmental Biology, BioFrontiers Institute, University of Colorado Boulder, 3415 Colorado Ave., UCB596, Boulder, CO 80303, USA
| |
Collapse
|
8
|
Seara FAC, Barbosa RAQ, Santos MVN, Domingos AE, Monnerat G, Carvalho AB, Olivares EL, Mill JG, Nascimento JHM, Campos de Carvalho AC. Paradoxical effect of testosterone supplementation therapy on cardiac ischemia/reperfusion injury in aged rats. J Steroid Biochem Mol Biol 2019; 191:105335. [PMID: 30930218 DOI: 10.1016/j.jsbmb.2019.03.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 03/18/2019] [Accepted: 03/19/2019] [Indexed: 02/08/2023]
Abstract
Aging is followed by numerous physiological limitations that reduce health span, particularly cardiovascular and metabolic disorders. Testosterone supplementation therapy (TST) has been widely used in the treatment of aging dysfunctions in either adult or aged patients, although recent evidence have suggested that the incidence of myocardial infarction might be increased in elderly patients. So far, though, the effects of TST in the progression of cardiac ischemia/reperfusion (IR) injury in aged hearts remain unclear. Male aged (23-24 months old) and adult (6 months old) Wistar rats were treated with placebo (Old + Placebo n = 5 / Adult + Placebo n = 5) or TST (Old + TST n = 7 / Adult + TST n = 5) for 30 days. After euthanasia, artificially-perfused isolated rat hearts were submitted to IR. Cardiac expression levels of genes encoding α and β myosin heavy chain (MHC), ryanodine receptor (RyR), brain-natriuretic peptide (BNP), sarcoplasmic reticulum Ca2+ ATPase 2a (SERCA2a), glucose-regulated protein 78 kDa (GRP78), eukaryotic initiation factor 2α (eIF2α), C/EBP-homologous protein (CHOP), caspase 3 and B cell lymphoma 2 (Bcl-2) were accessed by qRT-PCR. Protein levels of CHOP, p-Akt, and p-glycogen synthase kinase 3β (p-GSK-3β) were measured by Western Blot. Compared to placebo-treated aged rats, Old + TST group exhibited increased heart weight and up-regulation of αMHC mRNA expression levels, whereas βMHC mRNA expression (p < 0.05). During reperfusion, left ventricular developed pressure, dP/dt+, dP/dt-, and cardiac contractile function index were increased in Old + TST rat hearts (p < 0.05), whereas infarct size was increased (p < 0.05) in comparison with Old + Placebo group. p-Akt levels of Old + TST rat hearts were decreased when compared to Old + Placebo group. Conversely, TST did not promote significant effects in adult rat hearts. Taken together, these findings suggest that myocardial stunning and infarct size of aged hearts were distinctly affected by TST.
Collapse
Affiliation(s)
- Fernando A C Seara
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil; Department of Physiological Sciences, Federal Rural University of Rio de Janeiro, Seropedica, RJ, Brazil.
| | - Raiana A Q Barbosa
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Marcus V N Santos
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Ainá E Domingos
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Gustavo Monnerat
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Adriana B Carvalho
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Emerson L Olivares
- Department of Physiological Sciences, Federal Rural University of Rio de Janeiro, Seropedica, RJ, Brazil
| | - José G Mill
- Department of Physiological Sciences, Federal University of Espirito Santo, Vitoria, ES, Brazil
| | - Jose H M Nascimento
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Antonio C Campos de Carvalho
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil; National Center for Structural Biology and Bioimaging, Federal University of Rio de Janeiro, RJ, Brazil
| |
Collapse
|
9
|
K N H, Okabe J, Mathiyalagan P, Khan AW, Jadaan SA, Sarila G, Ziemann M, Khurana I, Maxwell SS, Du XJ, El-Osta A. Sex-Based Mhrt Methylation Chromatinizes MeCP2 in the Heart. iScience 2019; 17:288-301. [PMID: 31323475 PMCID: PMC6639684 DOI: 10.1016/j.isci.2019.06.031] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 05/13/2019] [Accepted: 06/20/2019] [Indexed: 01/15/2023] Open
Abstract
In the heart, primary microRNA-208b (pri-miR-208b) and Myheart (Mhrt) are long non-coding RNAs (lncRNAs) encoded by the cardiac myosin heavy chain genes. Although preclinical studies have shown that lncRNAs regulate gene expression and are protective for pathological hypertrophy, the mechanism underlying sex-based differences remains poorly understood. In this study, we examined DNA- and RNA-methylation-dependent regulation of pri-miR-208b and Mhrt. Expression of pri-miR-208b is elevated in the left ventricle of the female heart. Despite indistinguishable DNA methylation between sexes, the interaction of MeCP2 on chromatin is subject to RNase digestion, highlighting that affinity of the methyl-CG reader is broader than previously thought. A specialized procedure to isolate RNA from soluble cardiac chromatin emphasizes sex-based affinity of an MeCP2 co-repressor complex with Rest and Hdac2. Sex-specific Mhrt methylation chromatinizes MeCP2 at the pri-miR-208b promoter and extends the functional relevance of default transcriptional suppression in the heart. Mechanisms underlying sex-based gene expression are poorly understood Expression of primary miR-208b is independent of DNA methylation in the heart Sex-specific methylation of the long non-coding RNA Mhrt distinguishes MeCP2 Procedures assessing soluble chromatin emphasize RNA-dependent affinities
Collapse
Affiliation(s)
- Harikrishnan K N
- Epigenetics in Human Health and Disease, Central Clinical School, Faculty of Medicine, Monash University, Melbourne, VIC 3004, Australia; Baker Heart and Diabetes Institute, The Alfred Medical Research and Education Precinct, Melbourne, VIC 3004, Australia; Department of Clinical Pathology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Jun Okabe
- Epigenetics in Human Health and Disease, Central Clinical School, Faculty of Medicine, Monash University, Melbourne, VIC 3004, Australia; Baker Heart and Diabetes Institute, The Alfred Medical Research and Education Precinct, Melbourne, VIC 3004, Australia
| | - Prabhu Mathiyalagan
- Baker Heart and Diabetes Institute, The Alfred Medical Research and Education Precinct, Melbourne, VIC 3004, Australia
| | - Abdul Waheed Khan
- Epigenetics in Human Health and Disease, Central Clinical School, Faculty of Medicine, Monash University, Melbourne, VIC 3004, Australia; Baker Heart and Diabetes Institute, The Alfred Medical Research and Education Precinct, Melbourne, VIC 3004, Australia; Department of Clinical Pathology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Sameer A Jadaan
- Epigenetics in Human Health and Disease, Central Clinical School, Faculty of Medicine, Monash University, Melbourne, VIC 3004, Australia; Baker Heart and Diabetes Institute, The Alfred Medical Research and Education Precinct, Melbourne, VIC 3004, Australia; Department of Clinical Pathology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Gulcan Sarila
- Epigenetics in Human Health and Disease, Central Clinical School, Faculty of Medicine, Monash University, Melbourne, VIC 3004, Australia; Baker Heart and Diabetes Institute, The Alfred Medical Research and Education Precinct, Melbourne, VIC 3004, Australia
| | - Mark Ziemann
- Epigenetics in Human Health and Disease, Central Clinical School, Faculty of Medicine, Monash University, Melbourne, VIC 3004, Australia; Baker Heart and Diabetes Institute, The Alfred Medical Research and Education Precinct, Melbourne, VIC 3004, Australia
| | - Ishant Khurana
- Epigenetics in Human Health and Disease, Central Clinical School, Faculty of Medicine, Monash University, Melbourne, VIC 3004, Australia; Baker Heart and Diabetes Institute, The Alfred Medical Research and Education Precinct, Melbourne, VIC 3004, Australia
| | - Scott S Maxwell
- Epigenetics in Human Health and Disease, Central Clinical School, Faculty of Medicine, Monash University, Melbourne, VIC 3004, Australia; Baker Heart and Diabetes Institute, The Alfred Medical Research and Education Precinct, Melbourne, VIC 3004, Australia
| | - Xiao-Jun Du
- Baker Heart and Diabetes Institute, The Alfred Medical Research and Education Precinct, Melbourne, VIC 3004, Australia
| | - Assam El-Osta
- Epigenetics in Human Health and Disease, Central Clinical School, Faculty of Medicine, Monash University, Melbourne, VIC 3004, Australia; Baker Heart and Diabetes Institute, The Alfred Medical Research and Education Precinct, Melbourne, VIC 3004, Australia; Department of Clinical Pathology, The University of Melbourne, Parkville, VIC 3010, Australia; Hong Kong Institute of Diabetes and Obesity, Prince of Wales Hospital, The Chinese University of Hong Kong, 3/F Lui Che Woo Clinical Sciences Building, 30-32 Ngan Shing Street, Sha Tin, Hong Kong SAR; University College Copenhagen, Faculty of Health, Department of Technology, Biomedical Laboratory Science, Copenhagen, Denmark.
| |
Collapse
|
10
|
McCormack K. The cardioprotective effect of dexrazoxane (Cardioxane) is consistent with sequestration of poly(ADP-ribose) by self-assembly and not depletion of topoisomerase 2B. Ecancermedicalscience 2018; 12:889. [PMID: 30792806 PMCID: PMC6351063 DOI: 10.3332/ecancer.2018.889] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Indexed: 01/12/2023] Open
Abstract
Following systematic scrutiny of the evidence in support of the hypothesis that the cardioprotective mechanism of action of dexrazoxane is mediated by a 'depletion' or 'downregulation' of Top2β protein levels in heart tissue, the author concludes that this hypothesis is untenable. In seeking to understand how dexrazoxane protects the heart, the outcomes of a customised association rule learning algorithm incorporating the use of antecedent surrogate variables (CEME, 2017 McCormack Pharma) reveal a previously unknown relationship between dexrazoxane and poly(ADP-ribose) (PAR) polymer. The author shows how this previously unknown relationship explains both acute and long-term cardioprotection in patients receiving anthracyclines. In addition, as a direct inhibitor of PAR dexrazoxane has access to the epigenome and this offers a new insight into protection by dexrazoxane against doxorubicin-induced late-onset damage [McCormack K, manuscript in preparation]. Notably, through this review article, the author illustrates the practical application of probing natural language text using an association rule learning algorithm for the discovery of new and interesting associations that, otherwise, would remain lost. Historically, the use of CEME enabled the first report of the capacity of a small molecule to catalyse the hybrid self-assembly of a nucleic acid biopolymer via canonical and non-canonical, non-covalent interactions analogous to Watson Crick and Hoogsteen base pairing, respectively.
Collapse
Affiliation(s)
- Keith McCormack
- McCormack Pharma, a division of McCormack Ltd, Stirling House, 9 Burroughs Gardens, London NW4 4AU, UK
| |
Collapse
|
11
|
Ovchinnikova E, Hoes M, Ustyantsev K, Bomer N, de Jong TV, van der Mei H, Berezikov E, van der Meer P. Modeling Human Cardiac Hypertrophy in Stem Cell-Derived Cardiomyocytes. Stem Cell Reports 2018; 10:794-807. [PMID: 29456183 PMCID: PMC5918264 DOI: 10.1016/j.stemcr.2018.01.016] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2017] [Revised: 01/15/2018] [Accepted: 01/15/2018] [Indexed: 12/17/2022] Open
Abstract
Cardiac hypertrophy accompanies many forms of cardiovascular diseases. The mechanisms behind the development and regulation of cardiac hypertrophy in the human setting are poorly understood, which can be partially attributed to the lack of a human cardiomyocyte-based preclinical test system recapitulating features of diseased myocardium. The objective of our study is to determine whether human embryonic stem cell-derived cardiomyocytes (hESC-CMs) subjected to mechanical stretch can be used as an adequate in vitro model for studying molecular mechanisms of cardiac hypertrophy. We show that hESC-CMs subjected to cyclic stretch, which mimics mechanical overload, exhibit essential features of a hypertrophic state on structural, functional, and gene expression levels. The presented hESC-CM stretch approach provides insight into molecular mechanisms behind mechanotransduction and cardiac hypertrophy and lays groundwork for the development of pharmacological approaches as well as for discovering potential circulating biomarkers of cardiac dysfunction.
Collapse
Affiliation(s)
- Ekaterina Ovchinnikova
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, PO Box 30.001, Groningen, the Netherlands; European Research Institute for the Biology of Ageing, University of Groningen, University Medical Center Groningen, Antonius Deusinglaan, 1, PO Box 196, Groningen, the Netherlands
| | - Martijn Hoes
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, PO Box 30.001, Groningen, the Netherlands
| | - Kirill Ustyantsev
- Laboratory of Molecular Genetic Systems, Institute of Cytology and Genetics, Novosibirsk, 630090, Russia
| | - Nils Bomer
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, PO Box 30.001, Groningen, the Netherlands
| | - Tristan V de Jong
- European Research Institute for the Biology of Ageing, University of Groningen, University Medical Center Groningen, Antonius Deusinglaan, 1, PO Box 196, Groningen, the Netherlands
| | - Henny van der Mei
- University of Groningen, University Medical Center Groningen, Biomedical Engineering Department, Groningen, 9713AV, the Netherlands
| | - Eugene Berezikov
- European Research Institute for the Biology of Ageing, University of Groningen, University Medical Center Groningen, Antonius Deusinglaan, 1, PO Box 196, Groningen, the Netherlands.
| | - Peter van der Meer
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, PO Box 30.001, Groningen, the Netherlands.
| |
Collapse
|
12
|
Puddighinu G, D'Amario D, Foglio E, Manchi M, Siracusano A, Pontemezzo E, Cordella M, Facchiano F, Pellegrini L, Mangoni A, Tafani M, Crea F, Germani A, Russo MA, Limana F. Molecular mechanisms of cardioprotective effects mediated by transplanted cardiac ckit + cells through the activation of an inflammatory hypoxia-dependent reparative response. Oncotarget 2017; 9:937-957. [PMID: 29416668 PMCID: PMC5787525 DOI: 10.18632/oncotarget.22946] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 11/12/2017] [Indexed: 12/16/2022] Open
Abstract
The regenerative effects of cardiac ckit+ stem cells (ckit+CSCs) in acute myocardial infarction (MI) have been studied extensively, but how these cells exert a protective effect on cardiomyocytes is not well known. Growing evidences suggest that in adult stem cells injury triggers inflammatory signaling pathways which control tissue repair and regeneration. Aim of the present study was to determine the mechanisms underlying the cardioprotective effects of ckit+CSCs following transplantation in a murine model of MI. Following isolation and in vitro expansion, cardiac ckit+CSCs were subjected to normoxic and hypoxic conditions and assessed at different time points. These cells adapted to hypoxia as showed by the activation of HIF-1α and the expression of a number of genes, such as VEGF, GLUT1, EPO, HKII and, importantly, of alarmin receptors, such as RAGE, P2X7R, TLR2 and TLR4. Activation of these receptors determined an NFkB-dependent inflammatory and reparative gene response (IRR). Importantly, hypoxic ckit+CSCs increased the secretion of the survival growth factors IGF-1 and HGF. To verify whether activation of the IRR in a hypoxic microenvironment could exert a beneficial effect in vivo, autologous ckit+CSCs were transplanted into mouse heart following MI. Interestingly, transplantation of ckit+CSCs lowered apoptotic rates and induced autophagy in the peri-infarct area; further, it reduced hypertrophy and fibrosis and, most importantly, improved cardiac function. ckit+CSCs are able to adapt to a hypoxic environment and activate an inflammatory and reparative response that could account, at least in part, for a protective effect on stressed cardiomyocytes following transplantation in the infarcted heart.
Collapse
Affiliation(s)
- Giovanni Puddighinu
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Domenico D'Amario
- Department of Cardiovascular Sciences, Catholic University of The Sacred Heart, Rome, Italy
| | - Eleonora Foglio
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Melissa Manchi
- Department of Cardiovascular Sciences, Catholic University of The Sacred Heart, Rome, Italy
| | - Andrea Siracusano
- Department of Cardiovascular Sciences, Catholic University of The Sacred Heart, Rome, Italy
| | - Elena Pontemezzo
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Martina Cordella
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Francesco Facchiano
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Laura Pellegrini
- Department of Neurorehabilitation Sciences, Casa Cura Policlinico (CCP), Milan, Italy
| | - Antonella Mangoni
- Department of Pathological Anatomy, Catholic University of The Sacred Heart, Rome, Italy
| | - Marco Tafani
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Filippo Crea
- Department of Cardiovascular Sciences, Catholic University of The Sacred Heart, Rome, Italy
| | - Antonia Germani
- Laboratory of Vascular Pathology, Istituto Dermopatico dell'Immacolata, IDI-IRCCS, Fondazione Luigi Maria Monti, Rome, Italy
| | - Matteo Antonio Russo
- IRCCS San Raffaele Pisana, Rome, Italy.,MEBIC Consortium, San Raffaele Roma Open University, Rome, Italy
| | - Federica Limana
- IRCCS San Raffaele Pisana, Rome, Italy.,San Raffaele Roma Open University, Rome, Italy
| |
Collapse
|
13
|
Shirpoor A, Zerehpoosh M, Ansari MHK, Kheradmand F, Rasmi Y. Ginger extract mitigates ethanol-induced changes of alpha and beta - myosin heavy chain isoforms gene expression and oxidative stress in the heart of male wistar rats. DNA Repair (Amst) 2017; 57:45-49. [PMID: 28654806 DOI: 10.1016/j.dnarep.2017.06.023] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2016] [Revised: 04/28/2017] [Accepted: 06/15/2017] [Indexed: 10/19/2022]
Abstract
The association between ethanol consumption and heart abnormalities, such as chamber dilation, myocyte damage, ventricular hypertrophy, and hypertension is well known. However, underlying molecular mediators involved in ethanol-induced heart abnormalities remain elusive. The aim of this study was to investigate the effect of chronic ethanol exposure on alpha and beta - myosin heavy chain (MHC) isoforms gene expression transition and oxidative stress in rats' heart. It was also planned to find out whether ginger extract mitigated the abnormalities induced by ethanol in rats' heart. Male wistar rats were divided into three groups of eight animals as follows: Control, ethanol, and ginger extract treated ethanolic (GETE) groups. After six weeks of treatment, the results revealed a significant increase in the β-MHC gene expression, 8- OHdG amount, and NADPH oxidase level. Furthermore, a significant decrease in the ratio of α-MHC/β-MHC gene expression to the amount of paraoxonase enzyme in the ethanol group compared to the control group was found. The consumption of Ginger extract along with ethanol ameliorated the changes in MHC isoforms gene expression and reduced the elevated amount of 8-OHdG and NADPH oxidase. Moreover, compared to the consumption of ethanol alone, it increased the paraoxonase level significantly. These findings indicate that ethanol-induced heart abnormalities may in part be associated with MHC isoforms changes mediated by oxidative stress, and that these effects can be alleviated by using ginger extract as an antioxidant molecule.
Collapse
Affiliation(s)
- Alireza Shirpoor
- Department of Physiology, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran.
| | - Mitra Zerehpoosh
- Department of Biochemistry, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | | | - Fatemeh Kheradmand
- Department of Biochemistry, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Yousef Rasmi
- Department of Biochemistry, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| |
Collapse
|
14
|
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
|
15
|
Ceccarelli G, Benedetti L, Arcari ML, Carubbi C, Galli D. Muscle Stem Cell and Physical Activity: What Point is the Debate at? Open Med (Wars) 2017; 12:144-156. [PMID: 28765836 PMCID: PMC5529938 DOI: 10.1515/med-2017-0022] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Accepted: 04/21/2017] [Indexed: 12/11/2022] Open
Abstract
In the last 15 years, it emerged that the practice of regular physical activity reduces the risks of many diseases (cardiovascular diseases, diabetes, etc.) and it is fundamental in weight control and energy consuming to contrast obesity. Different groups proposed many molecular mechanisms as responsible for the positive effects of physical activity in healthy life. However, many points remain to be clarified. In this mini-review we reported the latest observations on the effects of physical exercise on healthy skeletal and cardiac muscle focusing on muscle stem cells. The last ones represent the fundamental elements for muscle regeneration post injury, but also for healthy muscle homeostasis. Interestingly, in both muscle tissues the morphological consequence of physical activity is a physiological hypertrophy that depends on different phenomena both in differentiated cells and stem cells. The signaling pathways for physical exercise effects present common elements in skeletal and cardiac muscle, like activation of specific transcription factors, proliferative pathways, and cytokines. More recently, post translational (miRNAs) or epigenetic (DNA methylation) modifications have been demonstrated. However, several points remain unresolved thus requiring new research on the effect of exercise on muscle stem cells.
Collapse
Affiliation(s)
- Gabriele Ceccarelli
- Department of Public Health, Experimental Medicine and Forensic, University of Pavia, Pavia, Italy.,Center of Health Technologies (CHT), University of Pavia, Pavia, Italy
| | - Laura Benedetti
- Department of Public Health, Experimental Medicine and Forensic, University of Pavia, Pavia, Italy.,Center of Health Technologies (CHT), University of Pavia, Pavia, Italy
| | - Maria Luisa Arcari
- Department of Medicine and Surgery, S.Bi.Bi.T. Unit, University of Parma, Parma, Italy
| | - Cecilia Carubbi
- Department of Medicine and Surgery, S.Bi.Bi.T. Unit, University of Parma, Parma, Italy
| | - Daniela Galli
- Department of Medicine and Surgery, S.Bi.Bi.T. Unit and Sport and Exercise Medicine Center (SEM)., University of Parma c/o Ospedale Maggiore, Via Gramsci, 14, 43126, Tel: +39-0521-036306, , Parma, Italy.,Department of Medicine and Surgery, S.Bi.Bi.T. Unit, University of Parma, Parma, Italy
| |
Collapse
|
16
|
Sukumaran A, Chang J, Han M, Mintri S, Khaw BA, Kim J. Iron overload exacerbates age-associated cardiac hypertrophy in a mouse model of hemochromatosis. Sci Rep 2017; 7:5756. [PMID: 28720890 PMCID: PMC5516030 DOI: 10.1038/s41598-017-05810-2] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Accepted: 06/02/2017] [Indexed: 12/12/2022] Open
Abstract
Cardiac damage associated with iron overload is the most common cause of morbidity and mortality in patients with hereditary hemochromatosis, but the precise mechanisms leading to disease progression are largely unexplored. Here we investigated the effects of iron overload and age on cardiac hypertrophy using 1-, 5- and 12-month old Hfe-deficient mice, an animal model of hemochromatosis in humans. Cardiac iron levels increased progressively with age, which was exacerbated in Hfe-deficient mice. The heart/body weight ratios were greater in Hfe-deficient mice at 5- and 12-month old, compared with their age-matched wild-type controls. Cardiac hypertrophy in 12-month old Hfe-deficient mice was consistent with decreased alpha myosin and increased beta myosin heavy chains, suggesting an alpha-to-beta conversion with age. This was accompanied by cardiac fibrosis and up-regulation of NFAT-c2, reflecting increased calcineurin/NFAT signaling in myocyte hypertrophy. Moreover, there was an age-dependent increase in the cardiac isoprostane levels in Hfe-deficient mice, indicating elevated oxidative stress. Also, rats fed high-iron diet demonstrated increased heart-to-body weight ratios, alpha myosin heavy chain and cardiac isoprostane levels, suggesting that iron overload promotes oxidative stress and cardiac hypertrophy. Our findings provide a molecular basis for the progression of age-dependent cardiac stress exacerbated by iron overload hemochromatosis.
Collapse
Affiliation(s)
- Abitha Sukumaran
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, USA
| | - JuOae Chang
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, USA
| | - Murui Han
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, USA
| | - Shrutika Mintri
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, USA
| | - Ban-An Khaw
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, USA
| | - Jonghan Kim
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, USA.
| |
Collapse
|
17
|
Can the Drosophila model help in paving the way for translational medicine in heart failure? Biochem Soc Trans 2017; 44:1549-1560. [PMID: 27911738 DOI: 10.1042/bst20160017c] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 08/02/2016] [Accepted: 08/15/2016] [Indexed: 01/09/2023]
Abstract
Chronic heart failure is a common consequence of various heart diseases. Mechanical force is known to play a key role in heart failure development through regulating cardiomyocyte hypertrophy. In order to understand the complex disease mechanism, this article discussed a multi-disciplinary approach that may aid the illustration of heart failure molecular process.
Collapse
|
18
|
Seara FDAC, Barbosa RAQ, de Oliveira DF, Gran da Silva DLS, Carvalho AB, Freitas Ferreira AC, Matheus Nascimento JH, Olivares EL. Administration of anabolic steroid during adolescence induces long-term cardiac hypertrophy and increases susceptibility to ischemia/reperfusion injury in adult Wistar rats. J Steroid Biochem Mol Biol 2017; 171:34-42. [PMID: 28179209 DOI: 10.1016/j.jsbmb.2017.01.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Revised: 01/09/2017] [Accepted: 01/19/2017] [Indexed: 01/27/2023]
Abstract
Chronic administration of anabolic androgenic steroids (AAS) in adult rats results in cardiac hypertrophy and increased susceptibility to myocardial ischemia/reperfusion (IR) injury. Molecular analyses demonstrated that hyperactivation of type 1 angiotensin II (AT1) receptor mediates cardiac hypertrophy induced by AAS and also induces down-regulation of myocardial ATP-sensitive potassium channel (KATP), resulting in loss of exercise-induced cardioprotection. Exposure to AAS during adolescence promoted long-term cardiovascular dysfunctions, such as dysautonomia. We tested the hypothesis that chronic AAS exposure in the pre/pubertal phase increases the susceptibility to myocardial ischemia/reperfusion (IR) injury in adult rats. Male Wistar rats (26day old) were treated with vehicle (Control, n=12) or testosterone propionate (TP) (AAS, 5mgkg-1 n=12) 5 times/week during 5 weeks. At the end of AAS exposure, rats underwent 23days of washout period and were submitted to euthanasia. Langendorff-perfused hearts were submitted to IR injury and evaluated for mechanical dysfunctions and infarct size. Molecular analysis was performed by mRNA levels of α-myosin heavy chain (MHC), βMHC and brain-derived natriuretic peptide (BNP), ryanodine receptor (RyR2) and sarcoplasmic reticulum calcium ATPase 2a (SERCA2a) by quantitative RT-PCR (qRT-PCR). The expression of AT1 receptor and KATP channel subunits (Kir6.1 and SURa) was analyzed by qRT-PCR and Western Blot. NADPH oxidase (Nox)-related reactive oxygen species generation was assessed by spectrofluorimetry. The expression of antioxidant enzymes was measured by qRT-PCR in order to address a potential role of redox unbalance. AAS exposure promoted long-term cardiac hypertrophy characterized by increased expression of βMHC and βMHC/αMHC ratio. Baseline derivative of pressure (dP/dt) was impaired by AAS exposure. Postischemic recovery of mechanical properties was impaired (decreased left ventricle [LV] developed pressure and maximal dP/dt; increased LV end-diastolic pressure and minimal dP/dt) and infarct size was larger in the AAS group. Catalase mRNA expression was significantly decreased in the AAS group. In conclusion, chronic administration of AAS during adolescence promoted long-term pathological cardiac hypertrophy and persistent increase in the susceptibility to myocardial IR injury possible due to disturbances on catalase expression.
Collapse
Affiliation(s)
- Fernando de Azevedo Cruz Seara
- Laboratory of Cardiovascular Physiology and Pharmacology, Department of Physiological Sciences, Institute of Biology, Federal Rural University of Rio de Janeiro, 23890-000 Seropedica, RJ, Brazil; Laboratory of Cardiac Electrophysiology, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, 21941-902 RJ, Brazil
| | - Raiana Andrade Quintanilha Barbosa
- Laboratory of Cellular and Molecular Cardiology, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro, 21941-902 RJ, Brazil
| | - Dahienne Ferreira de Oliveira
- Laboratory of Cardiac Electrophysiology, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, 21941-902 RJ, Brazil
| | - Diorney Luiz Souza Gran da Silva
- Laboratory of Endocrine Physiology, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro, 21941-902 RJ, Brazil
| | - Adriana Bastos Carvalho
- Laboratory of Cellular and Molecular Cardiology, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro, 21941-902 RJ, Brazil
| | - Andrea Claudia Freitas Ferreira
- Laboratory of Endocrine Physiology, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro, 21941-902 RJ, Brazil; NUMPEX-Bio, Pólo de Xerém, Federal University of Rio de Janeiro, Rio de Janeiro, 21941-902 RJ, Brazil
| | - José Hamilton Matheus Nascimento
- Laboratory of Cardiac Electrophysiology, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, 21941-902 RJ, Brazil
| | - Emerson Lopes Olivares
- Laboratory of Cardiovascular Physiology and Pharmacology, Department of Physiological Sciences, Institute of Biology, Federal Rural University of Rio de Janeiro, 23890-000 Seropedica, RJ, Brazil.
| |
Collapse
|
19
|
Cheraghi M, Namdari M, Daraee H, Negahdari B. Cardioprotective effect of magnetic hydrogel nanocomposite loaded N,α-L-rhamnopyranosyl vincosamide isolated fromMoringa oleiferaleaves against doxorubicin-induced cardiac toxicity in rats:in vitroandin vivostudies. J Microencapsul 2017; 34:335-341. [DOI: 10.1080/02652048.2017.1311955] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Mostafa Cheraghi
- Department of Cardiology, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Mehrdad Namdari
- Department of Cardiology, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Hadis Daraee
- Department of Cardiology, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Babak Negahdari
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical sciences, Tehran, Iran
| |
Collapse
|
20
|
Jin Y, Wei L, Cai W, Lin Z, Wu Z, Peng Y, Kohmoto T, Moss RL, Ge Y. Complete Characterization of Cardiac Myosin Heavy Chain (223 kDa) Enabled by Size-Exclusion Chromatography and Middle-Down Mass Spectrometry. Anal Chem 2017; 89:4922-4930. [PMID: 28366003 PMCID: PMC5526197 DOI: 10.1021/acs.analchem.7b00113] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Myosin heavy chain (MHC), the major component of the myosin motor molecule, plays an essential role in force production during muscle contraction. However, a comprehensive analysis of MHC proteoforms arising from sequence variations and post-translational modifications (PTMs) remains challenging due to the difficulties in purifying MHC (∼223 kDa) and achieving complete sequence coverage. Herein, we have established a strategy to effectively purify and comprehensively characterize MHC from heart tissue by combining size-exclusion chromatography (SEC) and middle-down mass spectrometry (MS). First, we have developed a MS-compatible SEC method for purifying MHC from heart tissue with high efficiency. Next, we have optimized the Glu-C, Asp-N, and trypsin limited digestion conditions for middle-down MS. Subsequently, we have applied this strategy with optimized conditions to comprehensively characterize human MHC and identified β-MHC as the predominant isoform in human left ventricular tissue. Full sequence coverage based on highly accurate mass measurements has been achieved using middle-down MS combining 1 Glu-C, 1 Asp-N, and 1 trypsin digestion. Three different PTMs: acetylation, methylation, and trimethylation were identified in human β-MHC and the corresponding sites were localized to the N-terminal Gly, Lys34, and Lys129, respectively, by electron capture dissociation (ECD). Taken together, we have demonstrated this strategy is highly efficient for purification and characterization of MHC, which can be further applied to studies of the role of MHC proteoforms in muscle-related diseases. We also envision that this integrated SEC/middle-down MS strategy can be extended for the characterization of other large proteins over 200 kDa.
Collapse
Affiliation(s)
- Yutong Jin
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Liming Wei
- Institutes of Biomedical Sciences, Fudan University, Shanghai, P. R. China
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Wenxuan Cai
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Molecular and Cellular Pharmacology Training Program, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Ziqing Lin
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Human Proteomics Program, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Zhijie Wu
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Ying Peng
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Human Proteomics Program, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Takushi Kohmoto
- Institutes of Biomedical Sciences, Fudan University, Shanghai, P. R. China
| | - Richard L. Moss
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Human Proteomics Program, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Ying Ge
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Human Proteomics Program, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
| |
Collapse
|
21
|
Yamaoka T, Hirata M, Dan T, Yamashita A, Otaka A, Nakaoki T, Miskon A, Kakinoki S, Mahara A. Individual evaluation of cardiac marker expression and self-beating during cardiac differentiation of P19CL6 cells on different culture substrates. J Biomed Mater Res A 2017; 105:1166-1174. [DOI: 10.1002/jbm.a.35977] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 10/13/2016] [Accepted: 12/06/2016] [Indexed: 12/18/2022]
Affiliation(s)
- Tetsuji Yamaoka
- Department of Biomedical Engineering; National Cerebral and Cardiovascular Center Research Institute; 5-7-1 Fujishirodai Suita Osaka 565-8565 Japan
- JST-CREST; 5 Sanbancho Chiyoda-ku Tokyo 102-0075 Japan
| | - Mitsuhi Hirata
- Department of Biomedical Engineering; National Cerebral and Cardiovascular Center Research Institute; 5-7-1 Fujishirodai Suita Osaka 565-8565 Japan
- JST-CREST; 5 Sanbancho Chiyoda-ku Tokyo 102-0075 Japan
| | - Takaaki Dan
- Department of Biomedical Engineering; National Cerebral and Cardiovascular Center Research Institute; 5-7-1 Fujishirodai Suita Osaka 565-8565 Japan
- Department of Materials Chemistry; Ryukoku University; 1-5 Seta Otsu Shiga 520-2194 Japan
| | - Atsushi Yamashita
- Department of Biomedical Engineering; National Cerebral and Cardiovascular Center Research Institute; 5-7-1 Fujishirodai Suita Osaka 565-8565 Japan
- JST-CREST; 5 Sanbancho Chiyoda-ku Tokyo 102-0075 Japan
| | - Akihisa Otaka
- Department of Biomedical Engineering; National Cerebral and Cardiovascular Center Research Institute; 5-7-1 Fujishirodai Suita Osaka 565-8565 Japan
| | - Takahiko Nakaoki
- Department of Materials Chemistry; Ryukoku University; 1-5 Seta Otsu Shiga 520-2194 Japan
| | - Azizi Miskon
- Department of Biomedical Engineering; National Cerebral and Cardiovascular Center Research Institute; 5-7-1 Fujishirodai Suita Osaka 565-8565 Japan
| | - Sachiro Kakinoki
- Department of Biomedical Engineering; National Cerebral and Cardiovascular Center Research Institute; 5-7-1 Fujishirodai Suita Osaka 565-8565 Japan
- JST-CREST; 5 Sanbancho Chiyoda-ku Tokyo 102-0075 Japan
| | - Atsushi Mahara
- Department of Biomedical Engineering; National Cerebral and Cardiovascular Center Research Institute; 5-7-1 Fujishirodai Suita Osaka 565-8565 Japan
| |
Collapse
|
22
|
Klos M, Mundada L, Banerjee I, Morgenstern S, Myers S, Leone M, Kleid M, Herron T, Devaney E. Altered myocyte contractility and calcium homeostasis in alpha-myosin heavy chain point mutations linked to familial dilated cardiomyopathy. Arch Biochem Biophys 2017; 615:53-60. [PMID: 28088328 DOI: 10.1016/j.abb.2016.12.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Revised: 12/06/2016] [Accepted: 12/17/2016] [Indexed: 01/19/2023]
Abstract
Mutations in the human cardiac motor protein beta-myosin heavy chain (βMHC) have been long recognized as a cause of familial hypertrophic cardiomyopathy. Recently, mutations (P830L and A1004S) in the less abundant but faster isoform alpha-myosin heavy chain (αMHC) have been linked to dilated cardiomyopathy (DCM). In this study, we sought to determine the cellular contractile phenotype associated with these point mutations. Ventricular myocytes were isolated from 2 month male Sprague Dawley rats. Cells were cultured in M199 media and infected with recombinant adenovirus containing the P830L or the A1004S mutant human αMHC at a MOI of 500 for 18 h. Uninfected cells (UI), human βMHC (MOI 500, 18 h), and human αMHC (MOI 500, 18 h) were used as controls. Cells were loaded with fura-2 (1 μM, 15 min) after 48 h. Sarcomere shortening and calcium transients were recorded in CO2 buffered M199 media (36°±1 C) with and without 10 nM isoproterenol (Iso). The A1004S mutation resulted in decreased peak sarcomere shortening while P830L demonstrated near normal shortening kinetics at baseline. In the presence of Iso, the A1004S sarcomere shortening was identical to the βMHC shortening while the P830L was identical to the αMHC control. All experimental groups had identical calcium transients. Despite a shared association with DCM, the P830L and A1004S αMHC mutations alter myocyte contractility in completely different ways while at the same preserving peak intracellular calcium.
Collapse
Affiliation(s)
- Matthew Klos
- Department of Pediatric Cardiac Surgery, UH Hospitals Cleveland, Cleveland, OH 44106, USA
| | - Lakshmi Mundada
- Department of Internal Medicine, Cardiovascular Medicine, Center for Arrhythmia Research, University of Michigan, Ann Arbor, MI 48108, USA
| | - Indroneal Banerjee
- Department of Medicine, University of California, San Diego, CA 92103, USA
| | - Sherry Morgenstern
- Department of Pediatric Cardiac Surgery, UH Hospitals Cleveland, Cleveland, OH 44106, USA
| | - Stephanie Myers
- Department of Medicine, University of California, San Diego, CA 92103, USA
| | - Michael Leone
- Department of Medicine, University of California, San Diego, CA 92103, USA
| | - Mark Kleid
- Department of Medicine, University of California, San Diego, CA 92103, USA
| | - Todd Herron
- Department of Internal Medicine, Cardiovascular Medicine, Center for Arrhythmia Research, University of Michigan, Ann Arbor, MI 48108, USA
| | - Eric Devaney
- Department of Pediatric Cardiac Surgery, UH Hospitals Cleveland, Cleveland, OH 44106, USA.
| |
Collapse
|
23
|
Namdari M, Eatemadi A. Cardioprotective effects of curcumin-loaded magnetic hydrogel nanocomposite (nanocurcumin) against doxorubicin-induced cardiac toxicity in rat cardiomyocyte cell lines. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2016; 45:731-739. [PMID: 27924631 DOI: 10.1080/21691401.2016.1261033] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Curcumin, is a yellow substance extracted from Curcuma longa rhizomes, it is a crystalline compound that has been traditionally applied in culinary practices and medicines in India. The aim of our study is to demonstrate the efficacy of curcumin-loaded magnetic hydrogel nanocomposite in the treatment of heart hypertrophy. 10 rats weighing 150-200 g each were induced with heart failure using 2.5 mg/kg doxorubicin for 2 weeks. The test groups were treated with curcumin-loaded magnetic hydrogel nanocomposite while the control was treated with curcumin alone. malondialdehyde (MDA) levels, superoxide dismutase (SOD), and glutathione peroxidase (GPX) enzymes activities were monitored after two weeks of last the dose. In addition, the expression of three heart failure markers; atrial natriuretic peptide (ANP), B type natriuretic peptide (BNP), and beta major histocompatibility complex (β-MHC) were observed, it was found that the expression of these markers decreases with an increase in the concentration of curcumin (P < 0.05). Curcumin elevated the decreased level of GPX and SOD, and reduced the elevated level of MDA in cardiac tissue. We suggest this combination to be a potent therapy for heart failure and hypertension in the nearest future.
Collapse
Affiliation(s)
- Mehrdad Namdari
- a Department of Cardiology , Lorestan University of Medical Sciences , Khoramabad , Iran
| | - Ali Eatemadi
- b Department of Medical Biotechnology, School of Advanced Technologies in Medicine , Tehran University of Medical sciences , Tehran , Iran
| |
Collapse
|
24
|
Németh BT, Mátyás C, Oláh A, Lux Á, Hidi L, Ruppert M, Kellermayer D, Kökény G, Szabó G, Merkely B, Radovits T. Cinaciguat prevents the development of pathologic hypertrophy in a rat model of left ventricular pressure overload. Sci Rep 2016; 6:37166. [PMID: 27853261 PMCID: PMC5112572 DOI: 10.1038/srep37166] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 10/25/2016] [Indexed: 01/19/2023] Open
Abstract
Pathologic myocardial hypertrophy develops when the heart is chronically pressure-overloaded. Elevated intracellular cGMP-levels have been reported to prevent the development of pathologic myocardial hypertrophy, therefore we investigated the effects of chronic activation of the cGMP producing enzyme, soluble guanylate cyclase by Cinaciguat in a rat model of pressure overload-induced cardiac hypertrophy. Abdominal aortic banding (AAB) was used to evoke pressure overload-induced cardiac hypertrophy in male Wistar rats. Sham operated animals served as controls. Experimental and control groups were treated with 10 mg/kg/day Cinaciguat (Cin) or placebo (Co) p.o. for six weeks, respectively. Pathologic myocardial hypertrophy was present in the AABCo group following 6 weeks of pressure overload of the heart, evidenced by increased relative heart weight, average cardiomyocyte diameter, collagen content and apoptosis. Cinaciguat did not significantly alter blood pressure, but effectively attenuated all features of pathologic myocardial hypertrophy, and normalized functional changes, such as the increase in contractility following AAB. Our results demonstrate that chronic enhancement of cGMP signalling by pharmacological activation of sGC might be a novel therapeutic approach in the prevention of pathologic myocardial hypertrophy.
Collapse
Affiliation(s)
- Balázs Tamás Németh
- Heart and Vascular Center, Semmelweis University, Városmajor u. 68., 1122 Budapest, Hungary
| | - Csaba Mátyás
- Heart and Vascular Center, Semmelweis University, Városmajor u. 68., 1122 Budapest, Hungary
| | - Attila Oláh
- Heart and Vascular Center, Semmelweis University, Városmajor u. 68., 1122 Budapest, Hungary
| | - Árpád Lux
- Heart and Vascular Center, Semmelweis University, Városmajor u. 68., 1122 Budapest, Hungary
| | - László Hidi
- Heart and Vascular Center, Semmelweis University, Városmajor u. 68., 1122 Budapest, Hungary
| | - Mihály Ruppert
- Heart and Vascular Center, Semmelweis University, Városmajor u. 68., 1122 Budapest, Hungary
| | - Dalma Kellermayer
- Heart and Vascular Center, Semmelweis University, Városmajor u. 68., 1122 Budapest, Hungary
| | - Gábor Kökény
- Institute of Pathophysiology, Semmelweis University, Nagyvárad tér 4., 1089 Budapest, Hungary
| | - Gábor Szabó
- Department of Cardiac Surgery, University of Heidelberg, Im Neuenheimer Feld 110., 69210 Heidelberg, Germany
| | - Béla Merkely
- Heart and Vascular Center, Semmelweis University, Városmajor u. 68., 1122 Budapest, Hungary
| | - Tamás Radovits
- Heart and Vascular Center, Semmelweis University, Városmajor u. 68., 1122 Budapest, Hungary
| |
Collapse
|
25
|
Toepfer CN, Sikkel MB, Caorsi V, Vydyanath A, Torre I, Copeland O, Lyon AR, Marston SB, Luther PK, Macleod KT, West TG, Ferenczi MA. A post-MI power struggle: adaptations in cardiac power occur at the sarcomere level alongside MyBP-C and RLC phosphorylation. Am J Physiol Heart Circ Physiol 2016; 311:H465-75. [PMID: 27233767 PMCID: PMC5005282 DOI: 10.1152/ajpheart.00899.2015] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 05/24/2016] [Indexed: 01/25/2023]
Abstract
Myocardial remodeling in response to chronic myocardial infarction (CMI) progresses through two phases, hypertrophic "compensation" and congestive "decompensation." Nothing is known about the ability of uninfarcted myocardium to produce force, velocity, and power during these clinical phases, even though adaptation in these regions likely drives progression of compensation. We hypothesized that enhanced cross-bridge-level contractility underlies mechanical compensation and is controlled in part by changes in the phosphorylation states of myosin regulatory proteins. We induced CMI in rats by left anterior descending coronary artery ligation. We then measured mechanical performance in permeabilized ventricular trabecula taken distant from the infarct zone and assayed myosin regulatory protein phosphorylation in each individual trabecula. During full activation, the compensated myocardium produced twice as much power and 31% greater isometric force compared with noninfarcted controls. Isometric force during submaximal activations was raised >2.4-fold, while power was 2-fold greater. Electron and confocal microscopy demonstrated that these mechanical changes were not a result of increased density of contractile protein and therefore not an effect of tissue hypertrophy. Hence, sarcomere-level contractile adaptations are key determinants of enhanced trabecular mechanics and of the overall cardiac compensatory response. Phosphorylation of myosin regulatory light chain (RLC) increased and remained elevated post-MI, while phosphorylation of myosin binding protein-C (MyBP-C) was initially depressed but then increased as the hearts became decompensated. These sensitivities to CMI are in accordance with phosphorylation-dependent regulatory roles for RLC and MyBP-C in crossbridge function and with compensatory adaptation in force and power that we observed in post-CMI trabeculae.
Collapse
Affiliation(s)
- Christopher N Toepfer
- Molecular Medicine Section, National Heart and Lung Institute, Imperial College London, London, United Kingdom; Laboratory of Molecular Physiology, National Heart and Lung Institute, National Institutes of Health, Bethesda, Maryland;
| | - Markus B Sikkel
- Department of Cardiac Medicine, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Valentina Caorsi
- Molecular Medicine Section, National Heart and Lung Institute, Imperial College London, London, United Kingdom; Laboratoire Physico-Chimie, UMR168, Institute Curie, Paris, France
| | - Anupama Vydyanath
- Molecular Medicine Section, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Iratxe Torre
- Molecular Medicine Section, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - O'Neal Copeland
- Department of Cardiac Medicine, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Alexander R Lyon
- Department of Cardiac Medicine, National Heart and Lung Institute, Imperial College London, London, United Kingdom; Nationa Institute of Health Research Cardiovascular Biomedical Research Unit, Royal Brompton Hospital, London, United Kingdom
| | - Steven B Marston
- Department of Cardiac Medicine, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Pradeep K Luther
- Molecular Medicine Section, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Kenneth T Macleod
- Department of Cardiac Medicine, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Timothy G West
- Royal Veterinary College London, Structure & Motion Laboratory, North Mymms, United Kingdom; and
| | - Michael A Ferenczi
- Molecular Medicine Section, National Heart and Lung Institute, Imperial College London, London, United Kingdom; Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| |
Collapse
|
26
|
Maayah ZH, Abdelhamid G, El-Kadi AOS. Development of cellular hypertrophy by 8-hydroxyeicosatetraenoic acid in the human ventricular cardiomyocyte, RL-14 cell line, is implicated by MAPK and NF-κB. Cell Biol Toxicol 2016; 31:241-59. [PMID: 26493311 DOI: 10.1007/s10565-015-9308-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Accepted: 10/14/2015] [Indexed: 01/17/2023]
Abstract
Recent studies have established the role of mid-chain hydroxyeicosatetraenoic acids (mid-chain HETEs) in the development of cardiovascular disease. Among these mid-chains, 8-HETE has been reported to have a proliferator and proinflammatory action. However, whether 8-HETE can induce cardiac hypertrophy has never been investigated before. Therefore, the overall objectives of the present study are to elucidate the potential hypertrophic effect of 8-HETE in the human ventricular cardiomyocytes, RL-14 cells, and to explore the mechanism(s) involved. Our results showed that 8-HETE induced cellular hypertrophy in RL-14 cells as evidenced by the induction of cardiac hypertrophy markers ANP, BNP, α-MHC, and β-MHC in a concentration- and time-dependent manner as well as the increase in cell surface area. Mechanistically, 8-HETE was able to induce the NF-κB activity as well as it significantly induced the phosphorylation of ERK1/2. The induction of cellular hypertrophy was associated with a proportional increase in the formation of dihydroxyeicosatrienoic acids (DHETs) parallel to the increase of soluble epoxide hydrolase (sEH) enzyme activity. Blocking the induction of NF-κB, ERK1/2, and sEH signaling pathways significantly inhibited 8-HETE-induced cellular hypertrophy. Our study provides the first evidence that 8-HETE induces cellular hypertrophy in RL-14 cells through MAPK- and NF-κB-dependent mechanism
Collapse
|
27
|
|
28
|
Kumar RR, Narasimhan M, Shanmugam G, Hong J, Devarajan A, Palaniappan S, Zhang J, Halade GV, Darley-Usmar VM, Hoidal JR, Rajasekaran NS. Abrogation of Nrf2 impairs antioxidant signaling and promotes atrial hypertrophy in response to high-intensity exercise stress. J Transl Med 2016; 14:86. [PMID: 27048381 PMCID: PMC4822244 DOI: 10.1186/s12967-016-0839-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 03/24/2016] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND Anomalies in myocardial structure involving myocyte growth, hypertrophy, differentiation, apoptosis, necrosis etc. affects its function and render cardiac tissue more vulnerable to the development of heart failure. Although oxidative stress has a well-established role in cardiac remodeling and dysfunction, the mechanisms linking redox state to atrial cardiomyocyte hypertrophic changes are poorly understood. Here, we investigated the role of nuclear erythroid-2 like factor-2 (Nrf2), a central transcriptional mediator, in redox signaling under high intensity exercise stress (HIES) in atria. METHODS Age and sex-matched wild-type (WT) and Nrf2(-/-) mice at >20 months of age were subjected to HIES for 6 weeks. Gene markers of hypertrophy and antioxidant enzymes were determined in the atria of WT and Nrf2(-/-) mice by real-time qPCR analyses. Detection and quantification of antioxidants, 4-hydroxy-nonenal (4-HNE), poly-ubiquitination and autophagy proteins in WT and Nrf2(-/-) mice were performed by immunofluorescence analysis. The level of oxidative stress was measured by microscopical examination of di-hydro-ethidium (DHE) fluorescence. RESULTS Under the sedentary state, Nrf2 abrogation resulted in a moderate down regulation of some of the atrial antioxidant gene expression (Gsr, Gclc, Gstα and Gstµ) despite having a normal redox state. In response to HIES, enlarged atrial myocytes along with significantly increased gene expression of cardiomyocyte hypertrophy markers (Anf, Bnf and β-Mhc) were observed in Nrf2(-/-) when compared to WT mice. Further, the transcript levels of Gclc, Gsr and Gstµ and protein levels of NQO1, catalase, GPX1 were profoundly downregulated along with GSH depletion and increased oxidative stress in Nrf2(-/-) mice when compared to its WT counterparts after HIES. Impaired antioxidant state and profound oxidative stress were associated with enhanced atrial expression of LC3 and ATG7 along with increased ubiquitination of ATG7 in Nrf2(-/-) mice subjected to HIES. CONCLUSIONS Loss of Nrf2 describes an altered biochemical phenotype associated with dysregulation in genes related to redox state, ubiquitination and autophagy in HIES that result in atrial hypertrophy. Therefore, our findings direct that preserving Nrf2-related antioxidant function would be one of the effective strategies to safeguard atrial health.
Collapse
Affiliation(s)
- Radhakrishnan Rajesh Kumar
- />Cardiac Aging & Redox Signaling Laboratory, Division of Molecular & Cellular Pathology, Department of Pathology, The University of Alabama at Birmingham, Birmingham, AL 35294-2180 USA
| | - Madhusudhanan Narasimhan
- />Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, 3601 4th Street, Lubbock, TX 79430 USA
| | - Gobinath Shanmugam
- />Cardiac Aging & Redox Signaling Laboratory, Division of Molecular & Cellular Pathology, Department of Pathology, The University of Alabama at Birmingham, Birmingham, AL 35294-2180 USA
| | - Jennifer Hong
- />Division of Cardiovascular Medicine, Department of Medicine, The University of Utah School of Medicine, Salt Lake City, UT 84132 USA
| | - Asokan Devarajan
- />Department of Obstetrics and Gynecology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095 USA
| | - Sethu Palaniappan
- />Department of Bio-Engineering, Comprehensive Cardiovascular Center, The University of Alabama at Birmingham, Birmingham, AL USA
| | - Jianhua Zhang
- />Center for Free Radical Biology, The University of Alabama at Birmingham, Birmingham, AL 35294-2180 USA
| | - Ganesh V. Halade
- />Department of Medicine, Comprehensive Cardiovascular Center, The University of Alabama at Birmingham, Birmingham, AL USA
| | - Victor M. Darley-Usmar
- />Center for Free Radical Biology, The University of Alabama at Birmingham, Birmingham, AL 35294-2180 USA
| | - John R. Hoidal
- />Division of Pulmonary Medicine, Department of Medicine, The University of Utah School of Medicine, Salt Lake City, UT 84132 USA
| | - Namakkal S. Rajasekaran
- />Cardiac Aging & Redox Signaling Laboratory, Division of Molecular & Cellular Pathology, Department of Pathology, The University of Alabama at Birmingham, Birmingham, AL 35294-2180 USA
- />Division of Cardiovascular Medicine, Department of Medicine, The University of Utah School of Medicine, Salt Lake City, UT 84132 USA
- />Center for Free Radical Biology, The University of Alabama at Birmingham, Birmingham, AL 35294-2180 USA
- />Department of Exercise Physiology, College of Health, The University of Utah School of Medicine, Salt Lake City, UT 84132 USA
| |
Collapse
|
29
|
Grabner A, Amaral AP, Schramm K, Singh S, Sloan A, Yanucil C, Li J, Shehadeh LA, Hare JM, David V, Martin A, Fornoni A, Di Marco GS, Kentrup D, Reuter S, Mayer AB, Pavenstädt H, Stypmann J, Kuhn C, Hille S, Frey N, Leifheit-Nestler M, Richter B, Haffner D, Abraham R, Bange J, Sperl B, Ullrich A, Brand M, Wolf M, Faul C. Activation of Cardiac Fibroblast Growth Factor Receptor 4 Causes Left Ventricular Hypertrophy. Cell Metab 2015; 22:1020-32. [PMID: 26437603 PMCID: PMC4670583 DOI: 10.1016/j.cmet.2015.09.002] [Citation(s) in RCA: 389] [Impact Index Per Article: 43.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Revised: 07/01/2015] [Accepted: 09/01/2015] [Indexed: 11/18/2022]
Abstract
Chronic kidney disease (CKD) is a worldwide public health threat that increases risk of death due to cardiovascular complications, including left ventricular hypertrophy (LVH). Novel therapeutic targets are needed to design treatments to alleviate the cardiovascular burden of CKD. Previously, we demonstrated that circulating concentrations of fibroblast growth factor (FGF) 23 rise progressively in CKD and induce LVH through an unknown FGF receptor (FGFR)-dependent mechanism. Here, we report that FGF23 exclusively activates FGFR4 on cardiac myocytes to stimulate phospholipase Cγ/calcineurin/nuclear factor of activated T cell signaling. A specific FGFR4-blocking antibody inhibits FGF23-induced hypertrophy of isolated cardiac myocytes and attenuates LVH in rats with CKD. Mice lacking FGFR4 do not develop LVH in response to elevated FGF23, whereas knockin mice carrying an FGFR4 gain-of-function mutation spontaneously develop LVH. Thus, FGF23 promotes LVH by activating FGFR4, thereby establishing FGFR4 as a pharmacological target for reducing cardiovascular risk in CKD.
Collapse
MESH Headings
- Animals
- Calcineurin/metabolism
- Cells, Cultured
- Disease Models, Animal
- Female
- Fibroblast Growth Factor-23
- Fibroblast Growth Factors/genetics
- Fibroblast Growth Factors/metabolism
- Gene Knock-In Techniques
- Glucuronidase/genetics
- Glucuronidase/metabolism
- HEK293 Cells
- Humans
- Hypertrophy, Left Ventricular/metabolism
- Hypertrophy, Left Ventricular/pathology
- Klotho Proteins
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mutagenesis, Site-Directed
- Myocytes, Cardiac/cytology
- Myocytes, Cardiac/metabolism
- NFATC Transcription Factors/metabolism
- Phospholipase C gamma/metabolism
- Rats
- Rats, Sprague-Dawley
- Receptor, Fibroblast Growth Factor, Type 4/deficiency
- Receptor, Fibroblast Growth Factor, Type 4/genetics
- Receptor, Fibroblast Growth Factor, Type 4/metabolism
- Renal Insufficiency, Chronic/metabolism
- Renal Insufficiency, Chronic/pathology
- Signal Transduction
Collapse
Affiliation(s)
- Alexander Grabner
- Katz Family Drug Discovery Center and Division of Nephrology and Hypertension, Department of Medicine, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA
| | - Ansel P Amaral
- Katz Family Drug Discovery Center and Division of Nephrology and Hypertension, Department of Medicine, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA; Department of Cell Biology and Anatomy, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA
| | - Karla Schramm
- Katz Family Drug Discovery Center and Division of Nephrology and Hypertension, Department of Medicine, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA; Department of Cell Biology and Anatomy, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA
| | - Saurav Singh
- Katz Family Drug Discovery Center and Division of Nephrology and Hypertension, Department of Medicine, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA; Department of Cell Biology and Anatomy, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA
| | - Alexis Sloan
- Katz Family Drug Discovery Center and Division of Nephrology and Hypertension, Department of Medicine, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA; Department of Cell Biology and Anatomy, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA
| | - Christopher Yanucil
- Katz Family Drug Discovery Center and Division of Nephrology and Hypertension, Department of Medicine, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA; Department of Cell Biology and Anatomy, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA
| | - Jihe Li
- Interdisciplinary Stem Cell Institute, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA; Division of Cardiology, Department of Medicine, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA
| | - Lina A Shehadeh
- Interdisciplinary Stem Cell Institute, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA; Division of Cardiology, Department of Medicine, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA; Vascular Biology Institute, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA
| | - Joshua M Hare
- Interdisciplinary Stem Cell Institute, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA; Division of Cardiology, Department of Medicine, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA
| | - Valentin David
- Katz Family Drug Discovery Center and Division of Nephrology and Hypertension, Department of Medicine, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA; Division of Nephrology and Hypertension, Department of Medicine and Center for Translational Metabolism and Health, Institute for Public Health and Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Aline Martin
- Katz Family Drug Discovery Center and Division of Nephrology and Hypertension, Department of Medicine, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA; Division of Nephrology and Hypertension, Department of Medicine and Center for Translational Metabolism and Health, Institute for Public Health and Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Alessia Fornoni
- Katz Family Drug Discovery Center and Division of Nephrology and Hypertension, Department of Medicine, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA
| | - Giovana Seno Di Marco
- Department of Internal Medicine D, University Hospital Münster, 48149 Münster, Germany
| | - Dominik Kentrup
- Department of Internal Medicine D, University Hospital Münster, 48149 Münster, Germany
| | - Stefan Reuter
- Department of Internal Medicine D, University Hospital Münster, 48149 Münster, Germany
| | - Anna B Mayer
- Department of Internal Medicine D, University Hospital Münster, 48149 Münster, Germany
| | - Hermann Pavenstädt
- Department of Internal Medicine D, University Hospital Münster, 48149 Münster, Germany
| | - Jörg Stypmann
- Division of Cardiology, Department of Cardiovascular Medicine, University Hospital Münster, 48149 Münster, Germany
| | - Christian Kuhn
- Department of Cardiology and Angiology, University Medical Center of Schleswig-Holstein, Campus Kiel, 24105 Kiel, Germany
| | - Susanne Hille
- Department of Cardiology and Angiology, University Medical Center of Schleswig-Holstein, Campus Kiel, 24105 Kiel, Germany
| | - Norbert Frey
- Department of Cardiology and Angiology, University Medical Center of Schleswig-Holstein, Campus Kiel, 24105 Kiel, Germany
| | - Maren Leifheit-Nestler
- Department of Pediatric Kidney, Liver and Metabolic Diseases, Hannover Medical School, 30625 Hannover, Germany
| | - Beatrice Richter
- Department of Pediatric Kidney, Liver and Metabolic Diseases, Hannover Medical School, 30625 Hannover, Germany
| | - Dieter Haffner
- Department of Pediatric Kidney, Liver and Metabolic Diseases, Hannover Medical School, 30625 Hannover, Germany
| | | | | | - Bianca Sperl
- Department of Molecular Biology, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany
| | - Axel Ullrich
- Department of Molecular Biology, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany
| | - Marcus Brand
- Department of Internal Medicine D, University Hospital Münster, 48149 Münster, Germany
| | - Myles Wolf
- Division of Nephrology and Hypertension, Department of Medicine and Center for Translational Metabolism and Health, Institute for Public Health and Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Christian Faul
- Katz Family Drug Discovery Center and Division of Nephrology and Hypertension, Department of Medicine, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA; Department of Cell Biology and Anatomy, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA.
| |
Collapse
|
30
|
Schwan J, Campbell SG. Prospects for In Vitro Myofilament Maturation in Stem Cell-Derived Cardiac Myocytes. Biomark Insights 2015; 10:91-103. [PMID: 26085788 PMCID: PMC4463797 DOI: 10.4137/bmi.s23912] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Revised: 03/09/2015] [Accepted: 03/11/2015] [Indexed: 12/20/2022] Open
Abstract
Cardiomyocytes derived from human stem cells are quickly becoming mainstays of cardiac regenerative medicine, in vitro disease modeling, and drug screening. Their suitability for such roles may seem obvious, but assessments of their contractile behavior suggest that they have not achieved a completely mature cardiac muscle phenotype. This could be explained in part by an incomplete transition from fetal to adult myofilament protein isoform expression. In this commentary, we review evidence that supports this hypothesis and discuss prospects for ultimately generating engineered heart tissue specimens that behave similarly to adult human myocardium. We suggest approaches to better characterize myofilament maturation level in these in vitro systems, and illustrate how new computational models could be used to better understand complex relationships between muscle contraction, myofilament protein isoform expression, and maturation.
Collapse
Affiliation(s)
- Jonas Schwan
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Stuart G Campbell
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| |
Collapse
|
31
|
Prendiville TW, Guo H, Lin Z, Zhou P, Stevens SM, He A, VanDusen N, Chen J, Zhong L, Wang DZ, Gao G, Pu WT. Novel Roles of GATA4/6 in the Postnatal Heart Identified through Temporally Controlled, Cardiomyocyte-Specific Gene Inactivation by Adeno-Associated Virus Delivery of Cre Recombinase. PLoS One 2015; 10:e0128105. [PMID: 26023924 PMCID: PMC4449121 DOI: 10.1371/journal.pone.0128105] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 04/23/2015] [Indexed: 12/23/2022] Open
Abstract
GATA4 and GATA6 are central cardiac transcriptional regulators. The postnatal, stage-specific function of the cardiac transcription factors GATA4 and GATA6 have not been evaluated. In part, this is because current Cre-loxP approaches to cardiac gene inactivation require time consuming and costly breeding of Cre-expressing and “floxed” mouse lines, often with limited control of the extent or timing of gene inactivation. We investigated the stage-specific functions of GATA4 and GATA6 in the postnatal heart by using adeno-associated virus serotype 9 to control the timing and extent of gene inactivation by Cre. Systemic delivery of recombinant, adeno-associated virus 9 (AAV9) expressing Cre from the cardiac specific Tnnt2 promoter was well tolerated and selectively and efficiently recombined floxed target genes in cardiomyocytes. AAV9:Tnnt2-Cre efficiently inactivated Gata4 and Gata6. Neonatal Gata4/6 inactivation caused severe, rapidly lethal systolic heart failure. In contrast, Gata4/6 inactivation in adult heart caused only mild systolic dysfunction but severe diastolic dysfunction. Reducing the dose of AAV9:Tnnt2-Cre generated mosaics in which scattered cardiomyocytes lacked Gata4/6. This mosaic knockout revealed that Gata4/6 are required cell autonomously for physiological cardiomyocyte growth. Our results define novel roles of GATA4 and GATA6 in the neonatal and adult heart. Furthermore, our data demonstrate that evaluation of gene function hinges on controlling the timing and extent of gene inactivation. AAV9:Tnnt2-Cre is a powerful tool for controlling these parameters.
Collapse
Affiliation(s)
- Terence W. Prendiville
- Department of Cardiology, Boston Children’s Hospital, Boston, Massachusetts, United States of America
| | - Haidong Guo
- Department of Cardiology, Boston Children’s Hospital, Boston, Massachusetts, United States of America
- Department of Anatomy, School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhiqiang Lin
- Department of Cardiology, Boston Children’s Hospital, Boston, Massachusetts, United States of America
| | - Pingzhu Zhou
- Department of Cardiology, Boston Children’s Hospital, Boston, Massachusetts, United States of America
| | - Sean M. Stevens
- Department of Cardiology, Boston Children’s Hospital, Boston, Massachusetts, United States of America
| | - Aibin He
- Department of Cardiology, Boston Children’s Hospital, Boston, Massachusetts, United States of America
- Institute of Molecular Medicine, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Nathan VanDusen
- Department of Cardiology, Boston Children’s Hospital, Boston, Massachusetts, United States of America
| | - Jinghai Chen
- Department of Cardiology, Boston Children’s Hospital, Boston, Massachusetts, United States of America
| | - Li Zhong
- Gene Therapy Center, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Da-Zhi Wang
- Department of Cardiology, Boston Children’s Hospital, Boston, Massachusetts, United States of America
- Harvard Stem Cell Institute, 1350 Massachusetts Ave, Cambridge, Massachusetts, United States of America
| | - Guangping Gao
- Gene Therapy Center, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - William T. Pu
- Department of Cardiology, Boston Children’s Hospital, Boston, Massachusetts, United States of America
- Gene Therapy Center, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
32
|
Chen YL, Sun CK, Tsai TH, Chang LT, Leu S, Zhen YY, Sheu JJ, Chua S, Yeh KH, Lu HI, Chang HW, Lee FY, Yip HK. Adipose-derived mesenchymal stem cells embedded in platelet-rich fibrin scaffolds promote angiogenesis, preserve heart function, and reduce left ventricular remodeling in rat acute myocardial infarction. Am J Transl Res 2015; 7:781-803. [PMID: 26175843 PMCID: PMC4494133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Accepted: 04/11/2015] [Indexed: 06/04/2023]
Abstract
OBJECTIVE This study tested the hypothesis that autologous adipose-derived mesenchymal stem cells (ADMSCs) embedded in platelet-rich fibrin (PRF) can significant promote myocardial regeneration and repair after acute myocardial infarction (AMI). SUMMARY BACKGROUND With avoiding the needle-related complications, PRF-embedded autologous ADMSCs graft provides a new effective stem cell-based therapeutic strategy for myocardial repair. METHODS Adult male Sprague-Dawley rats were equally divided (n = 8 per group) into group 1 (sham-operated), group 2 (AMI by ligating left coronary artery), group 3 (AMI+ PRF), and group 4 (AMI+PRF-embedded autologous ADMSCs). RPF with or without ADMSCs was patched on infarct area 1h after AMI induction. All animals were sacrificed on day 42 after echocardiography. RESULTS Left ventricular (LV) dimension and infarct/fibrotic areas were lowest in group 1, highest in group 2, in group 3 higher than in group 4, whereas LV performance and wall thickness exhibited a reversed pattern in all groups (all p < 0.001). Protein expressions of inflammatory (MMP-9, IL-1β), oxidative, apoptotic (Bax, cleaved PARP), fibrotic (Smad 3, TFG-β), hypertrophic (β-MHC), and heart failure (BNP) biomarkers displayed an identical pattern in infarct/fibrotic areas, whereas the protein expressions of anti-inflammatory (IL-10), anti-apoptotic (Bcl-2), anti-fibrotic (Smad1/5, BMP-2) biomarkers and α-MHC showed an opposite pattern (all p < 0.01). Angiogenic activities (c-Kit+, Sca-1+, CD31+, SDF-1α+, CXCR4+ cells; protein expressions of SDF-1α, CXCR4, VEGF) were highest in group 4 and lowest in group 1 (all p < 0.001). CONCLUSION ADMSCs embedded in PRF offered significant benefit in preserving LV function and limiting LV remodeling after AMI.
Collapse
Affiliation(s)
- Yung-Lung Chen
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of MedicineKaohsiung, 83301, Taiwan
| | - Cheuk-Kwan Sun
- Department of Emergency Medicine, E-Da Hospital, I-Shou University School of Medicine for International StudentsKaohsiung, 82245, Taiwan
| | - Tzu-Hsien Tsai
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of MedicineKaohsiung, 83301, Taiwan
| | - Li-Teh Chang
- Basic Science, Nursing Department, Meiho Institute of TechnologyPingtung, 91202, Taiwan
| | - Steve Leu
- Center for Translational Research in Biomedical Sciences, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of MedicineKaohsiung, 83301, Taiwan
| | - Yen-Yi Zhen
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of MedicineKaohsiung, 83301, Taiwan
| | - Jiunn-Jye Sheu
- Division of Thoracic and Cardiovascular Surgery, Department of Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of MedicineKaohsiung, 83301, Taiwan
| | - Sarah Chua
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of MedicineKaohsiung, 83301, Taiwan
| | - Kuo-Ho Yeh
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of MedicineKaohsiung, 83301, Taiwan
| | - Hung-I Lu
- Division of Thoracic and Cardiovascular Surgery, Department of Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of MedicineKaohsiung, 83301, Taiwan
| | - Hsueh-Wen Chang
- Department of Biological Sciences, National Sun Yat-Sen UniversityKaohsiung, 80424, Taiwan
| | - Fan-Yen Lee
- Division of Thoracic and Cardiovascular Surgery, Department of Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of MedicineKaohsiung, 83301, Taiwan
| | - Hon-Kan Yip
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of MedicineKaohsiung, 83301, Taiwan
- Center for Translational Research in Biomedical Sciences, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of MedicineKaohsiung, 83301, Taiwan
- Center for Shockwave Medicine and Tissue Engineering, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of MedicineKaohsiung, 83301, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical UniversityTaichung, 40402, Taiwan
| |
Collapse
|
33
|
Kralova E, Doka G, Pivackova L, Srankova J, Kuracinova K, Janega P, Babal P, Klimas J, Krenek P. l-Arginine Attenuates Cardiac Dysfunction, But Further Down-Regulates α-Myosin Heavy Chain Expression in Isoproterenol-Induced Cardiomyopathy. Basic Clin Pharmacol Toxicol 2015; 117:251-60. [DOI: 10.1111/bcpt.12405] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Accepted: 03/26/2015] [Indexed: 12/15/2022]
Affiliation(s)
- Eva Kralova
- Department of Pharmacology and Toxicology; Faculty of Pharmacy; Comenius University in Bratislava; Bratislava Slovak Republic
| | - Gabriel Doka
- Department of Pharmacology and Toxicology; Faculty of Pharmacy; Comenius University in Bratislava; Bratislava Slovak Republic
| | - Lenka Pivackova
- Department of Pharmacology and Toxicology; Faculty of Pharmacy; Comenius University in Bratislava; Bratislava Slovak Republic
| | - Jasna Srankova
- Department of Pharmacology and Toxicology; Faculty of Pharmacy; Comenius University in Bratislava; Bratislava Slovak Republic
| | - Kristina Kuracinova
- Department of Pathology; Faculty of Medicine; Comenius University in Bratislava; Bratislava Slovak Republic
| | - Pavol Janega
- Department of Pathology; Faculty of Medicine; Comenius University in Bratislava; Bratislava Slovak Republic
- Slovak Academy of Sciences; Institute of Normal and Pathological Anatomy; Bratislava Slovak Republic
| | - Pavel Babal
- Department of Pathology; Faculty of Medicine; Comenius University in Bratislava; Bratislava Slovak Republic
| | - Jan Klimas
- Department of Pharmacology and Toxicology; Faculty of Pharmacy; Comenius University in Bratislava; Bratislava Slovak Republic
| | - Peter Krenek
- Department of Pharmacology and Toxicology; Faculty of Pharmacy; Comenius University in Bratislava; Bratislava Slovak Republic
| |
Collapse
|
34
|
Reducing TRPC1 Expression through Liposome-Mediated siRNA Delivery Markedly Attenuates Hypoxia-Induced Pulmonary Arterial Hypertension in a Murine Model. Stem Cells Int 2014; 2014:316214. [PMID: 25587286 PMCID: PMC4281407 DOI: 10.1155/2014/316214] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Revised: 09/15/2014] [Accepted: 09/19/2014] [Indexed: 12/20/2022] Open
Abstract
We tested the hypothesis that Lipofectamine siRNA delivery to deplete transient receptor potential cation channel (TRPC) 1 protein expression can suppress hypoxia-induced pulmonary arterial hypertension (PAH) in mice. Adult male C57BL/6 mice were equally divided into group 1 (normal controls), group 2 (hypoxia), and group 3 (hypoxia + siRNA TRPC1). By day 28, right ventricular systolic pressure (RVSP), number of muscularized arteries, right ventricle (RV), and lung weights were increased in group 2 than in group 1 and reduced in group 3 compared with group 2. Pulmonary crowded score showed similar pattern, whereas number of alveolar sacs exhibited an opposite pattern compared to that of RVSP in all groups. Protein expressions of TRPCs, HIF-1α, Ku-70, apoptosis, and fibrosis and pulmonary mRNA expressions of inflammatory markers were similar pattern, whereas protein expressions of antifibrosis and VEGF were opposite to the pattern of RVSP. Cellular markers of pulmonary DNA damage, repair, and smooth muscle proliferation exhibited a pattern similar to that of RVSP. The mRNA expressions of proapoptotic and hypertrophy biomarkers displayed a similar pattern, whereas sarcomere length showed an opposite pattern compared to that of RVSP in all groups. Lipofectamine siRNA delivery effectively reduced TRPC1 expression, thereby attenuating PAH-associated RV and pulmonary arteriolar remodeling.
Collapse
|
35
|
Direct implantation versus platelet-rich fibrin-embedded adipose-derived mesenchymal stem cells in treating rat acute myocardial infarction. Int J Cardiol 2014; 173:410-23. [PMID: 24685001 DOI: 10.1016/j.ijcard.2014.03.015] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2013] [Revised: 01/21/2014] [Accepted: 03/09/2014] [Indexed: 01/02/2023]
Abstract
BACKGROUND This study tested whether adipose-derived mesenchymal stem cells (ADMSC) embedded in platelet-rich fibrin (PRF) scaffold is superior to direct ADMSC implantation in improving left ventricular (LV) performance and reducing LV remodeling in a rat acute myocardial infarction (AMI) model of left anterior descending coronary artery (LAD) ligation. METHODS Twenty-eight male adult Sprague Dawley rats equally divided into group 1 [sham control], group 2 (AMI only), group 3 (AMI+direct ADMSC implantation), and group 4 (AMI+PRF-embedded autologous ADMSC) were sacrificed on day 42 after AMI. RESULTS LV systolic and diastolic dimensions and volumes, and infarct/fibrotic areas were highest in group 2, lowest in group 1 and significantly higher in group 3 than in group 4, whereas LV performance and LV fractional shortening exhibited a reversed pattern (p<0.005). Protein expressions of inflammation (oxidative stress, IL-1β, MMP-9), apoptosis (mitochondrial Bax, cleaved PARP), fibrosis (Smad3, TGF-β), and pressure-overload biomarkers (BNP, MHC-β) displayed a pattern similar to that of LV dimensions, whereas anti-inflammatory (IL-10), anti-apoptotic (Bcl-2), and anti-fibrotic (Smad1/5, BMP-2) indices showed a pattern similar to that of LV performance among the four groups (all p<0.05). Angiogenesis biomarkers at protein (CXCR4, SDF-1α, VEGF), cellular (CD31+, CXCR4+, SDF-1α+), and immunohistochemical (small vessels) levels, and cardiac stem cell markers (C-kit+, Sca-1+) in infarct myocardium were highest in group 4, lowest in group 1, and significantly higher in group 3 than in group 2 (all p<0.005). CONCLUSION PRF-embedded ADMSC is superior to direct ADMSC implantation in preserving LV function and attenuating LV remodeling.
Collapse
|
36
|
Songstad NT, Johansen D, How OJ, Kaaresen PI, Ytrehus K, Acharya G. Effect of transverse aortic constriction on cardiac structure, function and gene expression in pregnant rats. PLoS One 2014; 9:e89559. [PMID: 24586871 PMCID: PMC3930736 DOI: 10.1371/journal.pone.0089559] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Accepted: 01/21/2014] [Indexed: 01/08/2023] Open
Abstract
Background There is an increased risk of heart failure and pulmonary edema in pregnancies complicated by hypertensive disorders. However, in a previous study we found that pregnancy protects against fibrosis and preserves angiogenesis in a rat model of angiotensin II induced cardiac hypertrophy. In this study we test the hypothesis that pregnancy protects against negative effects of increased afterload. Methods Pregnant (gestational day 5.5–8.5) and non-pregnant Wistar rats were randomized to transverse aortic constriction (TAC) or sham surgery. After 14.2±0.14 days echocardiography was performed. Aortic blood pressure and left ventricular (LV) pressure-volume loops were obtained using a conductance catheter. LV collagen content and cardiomyocyte circumference were measured. Myocardial gene expression was assessed by real-time polymerase chain reaction. Results Heart weight was increased by TAC (p<0.001) but not by pregnancy. Cardiac myocyte circumference was larger in pregnant compared to non-pregnant rats independent of TAC (p = 0.01), however TAC per se did not affect this parameter. Collagen content in LV myocardium was not affected by pregnancy or TAC. TAC increased stroke work more in pregnant rats (34.1±2.4 vs 17.5±2.4 mmHg/mL, p<0.001) than in non-pregnant (28.2±1.7 vs 20.9±1.5 mmHg/mL, p = 0.06). However, it did not lead to overt heart failure in any group. In pregnant rats, α-MHC gene expression was reduced by TAC. Increased in the expression of β-MHC gene was higher in pregnant (5-fold) compared to non-pregnant rats (2-fold) after TAC (p = 0.001). Nine out of the 19 genes related to cardiac remodeling were affected by pregnancy independent of TAC. Conclusions This study did not support the hypothesis that pregnancy is cardioprotective against the negative effects of increased afterload. Some differences in cardiac structure, function and gene expression between pregnant and non-pregnant rats following TAC indicated that afterload increase is less tolerated in pregnancy.
Collapse
MESH Headings
- Animals
- Aorta, Thoracic/growth & development
- Aorta, Thoracic/metabolism
- Aorta, Thoracic/pathology
- Cardiomegaly/genetics
- Cardiomegaly/metabolism
- Cardiomegaly/pathology
- Cells, Cultured
- Constriction, Pathologic/genetics
- Constriction, Pathologic/metabolism
- Constriction, Pathologic/pathology
- Echocardiography
- Female
- Fibrosis/metabolism
- Fibrosis/pathology
- Gene Expression
- Heart/physiopathology
- Immunoenzyme Techniques
- Neovascularization, Pathologic/metabolism
- Neovascularization, Pathologic/pathology
- Pregnancy
- RNA, Messenger/genetics
- Rats
- Rats, Wistar
- Real-Time Polymerase Chain Reaction
- Reverse Transcriptase Polymerase Chain Reaction
Collapse
Affiliation(s)
- Nils Thomas Songstad
- Women's Health and Perinatology Research Group, Department of Clinical Medicine, Faculty of Health Sciences, University of Tromsø, Tromsø, Norway
- Department of Pediatrics, University Hospital of Northern Norway, Tromsø, Norway
- * E-mail:
| | - David Johansen
- Cardiovascular Research Group, Department of Medical Biology, Faculty of Health Sciences, University of Tromsø, Tromsø, Norway
| | - Ole-Jacob How
- Cardiovascular Research Group, Department of Medical Biology, Faculty of Health Sciences, University of Tromsø, Tromsø, Norway
| | - Per Ivar Kaaresen
- Department of Pediatrics, University Hospital of Northern Norway, Tromsø, Norway
- Pediatric Research Group, Department of Clinical Medicine, Faculty of Health Sciences, University of Tromsø, Tromsø, Norway
| | - Kirsti Ytrehus
- Cardiovascular Research Group, Department of Medical Biology, Faculty of Health Sciences, University of Tromsø, Tromsø, Norway
| | - Ganesh Acharya
- Women's Health and Perinatology Research Group, Department of Clinical Medicine, Faculty of Health Sciences, University of Tromsø, Tromsø, Norway
- Department of Obstetrics and Gynecology, University Hospital of Northern Norway, Tromsø, Norway
| |
Collapse
|
37
|
Fibroblast growth factor receptor 1 signaling in adult cardiomyocytes increases contractility and results in a hypertrophic cardiomyopathy. PLoS One 2013; 8:e82979. [PMID: 24349409 PMCID: PMC3859602 DOI: 10.1371/journal.pone.0082979] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2013] [Accepted: 10/29/2013] [Indexed: 11/19/2022] Open
Abstract
Fibroblast growth factors (FGFs) and their receptors are highly conserved signaling molecules that have been implicated in postnatal cardiac remodeling. However, it is not known whether cardiomyocyte-expressed FGF receptors are necessary or sufficient for ventricular remodeling in the adult heart. To determine whether cardiomyocytes were competent to respond to an activated FGF receptor, and to determine if this signal would result in the development of hypertrophy, we engineered a doxycycline (DOX)-inducible, cardiomyocyte-specific, constitutively active FGF receptor mouse model (αMHC-rtTA, TRE-caFgfr1-myc). Echocardiographic and hemodynamic analysis indicated that acute expression of caFGFR1 rapidly and directly increased cardiac contractility, while chronic expression resulted in significant hypertrophy with preservation of systolic function. Subsequent histologic analysis showed increased cardiomyocyte cross-sectional area and regions of myocyte disarray and fibrosis, classic features of hypertrophic cardiomyopathy (HCM). Analysis of downstream pathways revealed a lack of clear activation of classical FGF-mediated signaling pathways, but did demonstrate a reduction in Serca2 expression and troponin I phosphorylation. Isolated ventricular myocytes showed enhanced contractility and reduced relaxation, an effect that was partially reversed by inhibition of actin-myosin interactions. We conclude that adult cardiomyocytes are competent to transduce FGF signaling and that FGF signaling is sufficient to promote increased cardiomyocyte contractility in vitro and in vivo through enhanced intrinsic actin-myosin interactions. Long-term, FGFR overexpression results in HCM with a dynamic outflow tract obstruction, and may serve as a unique model of HCM.
Collapse
|
38
|
Kwon DH, Eom GH, Kee HJ, Nam YS, Cho YK, Kim DK, Koo JY, Kim HS, Nam KI, Kim KK, Lee IK, Park SB, Choi HS, Kook H. Estrogen-related receptor gamma induces cardiac hypertrophy by activating GATA4. J Mol Cell Cardiol 2013; 65:88-97. [DOI: 10.1016/j.yjmcc.2013.09.011] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Revised: 09/16/2013] [Accepted: 09/18/2013] [Indexed: 10/26/2022]
|
39
|
Nelson OL, Rourke BC. Increase in cardiac myosin heavy-chain (MyHC) alpha protein isoform in hibernating ground squirrels, with echocardiographic visualization of ventricular wall hypertrophy and prolonged contraction. ACTA ACUST UNITED AC 2013; 216:4678-90. [PMID: 24072796 DOI: 10.1242/jeb.088773] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Deep hibernators such as golden-mantled ground squirrels (Callospermophilus lateralis) have multiple challenges to cardiac function during low temperature torpor and subsequent arousals. As heart rates fall from over 300 beats min(-1) to less than 10, chamber dilation and reduced cardiac output could lead to congestive myopathy. We performed echocardiography on a cohort of individuals prior to and after several months of hibernation. The left ventricular chamber exhibited eccentric and concentric hypertrophy during hibernation and thus calculated ventricular mass was ~30% greater. Ventricular ejection fraction was mildly reduced during hibernation but stroke volumes were greater due to the eccentric hypertrophy and dramatically increased diastolic filling volumes. Globally, the systolic phase in hibernation was ~9.5 times longer, and the diastolic phase was 28× longer. Left atrial ejection generally was not observed during hibernation. Atrial ejection returned weakly during early arousal. Strain echocardiography assessed the velocity and total movement distance of contraction and relaxation for regional ventricular segments in active and early arousal states. Myocardial systolic strain during early arousal was significantly greater than the active state, indicating greater total contractile movement. This mirrored the increased ventricular ejection fraction noted with early arousal. However, strain rates were slower during early arousal than during the active period, particularly systolic strain, which was 33% of active, compared with the rate of diastolic strain, which was 67% of active. As heart rate rose during the arousal period, myocardial velocities and strain rates also increased; this was matched closely by cardiac output. Curiously, though heart rates were only 26% of active heart rates during early arousal, the cardiac output was nearly 40% of the active state, suggesting an efficient pumping system. We further analyzed proportions of cardiac myosin heavy-chain (MyHC) isoforms in a separate cohort of squirrels over 5 months, including time points before hibernation, during hibernation and just prior to emergence. Hibernating individuals were maintained in both a 4°C cold room and a 20°C warm room. Measured by SDS-PAGE, relative percentages of cardiac MyHC alpha were increased during hibernation, at both hibernacula temperatures. A potential increase in contractile speed, and power, from more abundant MyHC alpha may aid force generation at low temperature and at low heart rates. Unlike many models of cardiomyopathies where the alpha isoform is replaced by the beta isoform in order to reduce oxygen consumption, ground squirrels demonstrate a potential cardioprotective mechanism to maintain cardiac output during torpor.
Collapse
Affiliation(s)
- O Lynne Nelson
- College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA
| | | |
Collapse
|
40
|
Wan W, Xu X, Zhao W, Garza MA, Zhang JQ. Exercise training induced myosin heavy chain isoform alteration in the infarcted heart. Appl Physiol Nutr Metab 2013; 39:226-32. [PMID: 24476479 DOI: 10.1139/apnm-2013-0268] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The myosin heavy chain isoform MHC-α has 3-fold higher ATPase activity than MHC-β. After myocardial infarction (MI), MHC-α expression is profoundly downregulated and MHC-β expression is reciprocally upregulated. This shift, which is attributed to low thyroid hormone (TH), contributes to myocardial systolic dysfunction. We investigated the effect of post-MI exercise training on MHC isoforms, TH, and cardiac function. MI was surgically induced in 7-week-old rats by ligation of the coronary artery. The survivors were assigned to 3 groups (n = 10/group): Sham (no MI, no exercise), MISed (MI, no exercise), and MIEx (MI, exercise). Treadmill exercise training began 1 week post-MI and lasted for 8 weeks. Echocardiogram measurements were taken on the day prior to initiation of exercise training and at the end of exercise training. Tissue and blood samples were collected at the end of the experiment. MHC isoform gene and protein expression and TH were measured. Our results illustrated that MHC-α gene expression was higher and MHC-β gene expression was lower in the MIEx group than in the MISed group. Resting serum TH concentrations (T3 and T4) were similar between the 2 MI groups. The MIEx group had higher fractional shortening than the MISed group. In conclusion, post-MI exercise training beneficially altered MHC isoforms and improved cardiac function without changing TH.
Collapse
Affiliation(s)
- Wenhan Wan
- a Laboratory of Cardiovascular Research, University of Texas at San Antonio, 1 UTSA Circle, San Antonio, TX 78249, USA
| | | | | | | | | |
Collapse
|
41
|
Abstract
Both clinical and experimental findings at the molecular, cellular, tissue, organ and systematic levels have depicted the presence of a contemporary regulatory machinery namely compensation in various forms of cardiovascular diseases. Compensation is believed to be present and regulated within the scope of a biological entity and represents the initiation of dyshomeostasis. Compensation can be identified in multiple categories and organs in cardiovascular diseases at multiple levels. The capacity to reduce the unfavorable pathological compensation may be a criterion to evaluate the therapeutic effectiveness for cardiovascular diseases. This mini-review tries to take compensation into consideration in the understanding of onset and progression of cardiovascular diseases in particular, and thus, better or optimal therapeutic approaches may be achieved for the prevention and management of cardiovascular diseases.
Collapse
Affiliation(s)
- Xiu-Juan Fan
- China Nepstar Chain Drugstore Ltd., Hangzhou 310003, Zhejiang, China.
| | | |
Collapse
|
42
|
Wang X, Wang J, Su M, Wang C, Chen J, Wang H, Song L, Zou Y, Zhang L, Zhang Y, Hui R. TNNI3K, a cardiac-specific kinase, promotes physiological cardiac hypertrophy in transgenic mice. PLoS One 2013; 8:e58570. [PMID: 23472207 PMCID: PMC3589374 DOI: 10.1371/journal.pone.0058570] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2012] [Accepted: 02/06/2013] [Indexed: 01/09/2023] Open
Abstract
Purpose Protein kinase plays an essential role in controlling cardiac growth and hypertrophic remodeling. The cardiac troponin I-interacting kinase (TNNI3K), a novel cardiac specific kinase, is associated with cardiomyocyte hypertrophy. However, the precise function of TNNI3K in regulating cardiac remodeling has remained controversial. Methods and Results In a rat model of cardiac hypertrophy generated by transverse aortic constriction, myocardial TNNI3K expression was significantly increased by 1.62 folds (P<0.05) after constriction for 15 days. To investigate the role of TNNI3K in cardiac hypertrophy, we generated transgenic mouse lines with overexpression of human TNNI3K specifically in the heart. At the age of 3 months, the high-copy-number TNNI3K transgenic mice demonstrated a phenotype of concentric hypertrophy with increased heart weight normalized to body weight (1.31 fold, P<0.01). Echocardiography and non-invasive hemodynamic assessments showed enhanced cardiac function. No necrosis or myocyte disarray was observed in the heart of TNNI3K transgenic mice. This concentric hypertrophy maintained up to 12 months of age without cardiac dysfunction. The phospho amino acid analysis revealed that TNNI3K is a protein-tyrosine kinase. The yeast two-hybrid screen and co-immunoprecipitation assay identified cTnI as a target for TNNI3K. Moreover, TNNI3K overexpression induced cTnI phosphorylation at Ser22/Ser23 in vivo and in vitro, suggesting that TNNI3K is a novel upstream regulator for cTnI phosphorylation. Conclusion TNNI3K promotes a concentric hypertrophy with enhancement of cardiac function via regulating the phosphorylation of cTnI. TNNI3K could be a potential therapeutic target for preventing from heart failure.
Collapse
Affiliation(s)
- Xiaojian Wang
- Sino-German Laboratory for Molecular Medicine, State Key Laboratory of Cardiovascular Disease, FuWai Hospital & Cardiovascular Institute, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, People's Republic of China
| | - Jizheng Wang
- Sino-German Laboratory for Molecular Medicine, State Key Laboratory of Cardiovascular Disease, FuWai Hospital & Cardiovascular Institute, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, People's Republic of China
| | - Ming Su
- Sino-German Laboratory for Molecular Medicine, State Key Laboratory of Cardiovascular Disease, FuWai Hospital & Cardiovascular Institute, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, People's Republic of China
| | - Changxin Wang
- Sino-German Laboratory for Molecular Medicine, State Key Laboratory of Cardiovascular Disease, FuWai Hospital & Cardiovascular Institute, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, People's Republic of China
| | - Jingzhou Chen
- Sino-German Laboratory for Molecular Medicine, State Key Laboratory of Cardiovascular Disease, FuWai Hospital & Cardiovascular Institute, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, People's Republic of China
| | - Hu Wang
- Sino-German Laboratory for Molecular Medicine, State Key Laboratory of Cardiovascular Disease, FuWai Hospital & Cardiovascular Institute, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, People's Republic of China
| | - Lei Song
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, FuWai Hospital & Cardiovascular Institute, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, People's Republic of China
| | - Yubao Zou
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, FuWai Hospital & Cardiovascular Institute, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, People's Republic of China
| | - Lianfeng Zhang
- Key Laboratory of Human Disease Comparative Medicine, Ministry of Health, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medical Center, Peking Union Medical College, Beijing, People's Republic of China
| | - Youyi Zhang
- Institute of Vascular Medicine, Peking University Third Hospital, Beijing, People's Republic of China
| | - Rutai Hui
- Sino-German Laboratory for Molecular Medicine, State Key Laboratory of Cardiovascular Disease, FuWai Hospital & Cardiovascular Institute, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, People's Republic of China
- * E-mail:
| |
Collapse
|
43
|
Patrizio M, Musumeci M, Piccone A, Raggi C, Mattei E, Marano G. Hormonal regulation of β-myosin heavy chain expression in the mouse left ventricle. J Endocrinol 2013. [PMID: 23179080 DOI: 10.1530/joe-12-0201] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
We investigated the influence of sex hormones on the expression of α- and β-cardiac myosin heavy chain isoforms (α-MHC and β-MHC) in C57bl/6 mice of both sexes under physiological and pathological conditions. In the left ventricles (LVs) of fertile female mice, β-MHC expression was tenfold higher compared with the age-matched males, whereas no difference was found in α-MHC expression. These differences disappeared after ovariectomy or in immature mice. We also found a sex-related difference in expression of β-adrenoceptors (β1-AR), as mRNA levels of this gene were 40% lower in fertile females compared with males of the same age but did not differ in prepubertal or ovariectomized animals. Interestingly, the deletion of both β1- and β2-ARs abolished sex difference of β-MHC expression, as mRNA levels in the LVs of knockout males were increased and reached values comparable to those of knockout females. Moreover, the β1-AR antagonist metoprolol induced about a threefold increase in β-MHC expression in adult male mice. The capability of gender to regulate β-MHC expression was also evaluated in the presence of hemodynamic overload. Thoracic aortic coarctation (TAC) produced cardiac hypertrophy along with a 12-fold increase in β-MHC and a 50% decrease in β1-AR expression in males but not in females, thus abolishing the gender difference observed in sham animals for such genes. By contrast, TAC did not change β2-AR expression. In conclusion, our results show that the expression of β-MHC and β1-AR in the LVs undergo gender-related and correlated changes under both physiological and pathological conditions and suggest a role of β1-AR-mediated signaling.
Collapse
Affiliation(s)
- Mario Patrizio
- Department of Drug Research and Evaluation, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy.
| | | | | | | | | | | |
Collapse
|
44
|
Soman S, Rajamanickam C, Rauf AA, Indira M. Beneficial effects of Psidium guajava leaf extract on diabetic myocardium. ACTA ACUST UNITED AC 2013; 65:91-5. [PMID: 21742475 DOI: 10.1016/j.etp.2011.06.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2010] [Revised: 05/27/2011] [Accepted: 06/07/2011] [Indexed: 11/30/2022]
Affiliation(s)
- Sowmya Soman
- Department of Biochemistry, University of Kerala, Kariavattom, Thiruvananthapuram 695 581, Kerala, India
| | | | | | | |
Collapse
|
45
|
Pinto JR, Gomes AV, Jones MA, Liang J, Nguyen S, Miller T, Parvatiyar MS, Potter JD. The functional properties of human slow skeletal troponin T isoforms in cardiac muscle regulation. J Biol Chem 2012; 287:37362-70. [PMID: 22977240 PMCID: PMC3481333 DOI: 10.1074/jbc.m112.364927] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2012] [Revised: 08/17/2012] [Indexed: 11/06/2022] Open
Abstract
Human slow skeletal troponin T (HSSTnT) shares a high degree of homology with cardiac TnT (CTnT). Although the presence of HSSTnT has not been confirmed in the heart at the protein level, detectable levels of HSSTnT mRNA have been found. Whether HSSTnT isoforms are expressed transiently remains unknown. Because transient re-expression of HSSTnT may be a potential mechanism of regulating function, we explored the effect of HSSTnT on the regulation of cardiac muscle. At least three HSSTnT isoforms have been found to exist in slow skeletal muscle: HSSTnT1 (+exons 5 and 12), HSSTnT2 (+exon 5, -exon 12), and HSSTnT3 (-exons 5 and 12). Another isoform, HSSTnT hypothetical (Hyp) (-exon 5, +exon 12), has only been found at the mRNA level. Compared with HCTnT3 (adult isoform), Tn complexes containing HSSTnT1, -2, and -3 did not alter the actomyosin ATPase activation and inhibition in the presence and absence of Ca(2+), respectively. HSSTnTHyp was not evaluated as it did not form a Tn complex under a variety of conditions. Porcine papillary skinned fibers displaced with HSSTnT1, -2, or -3 and reconstituted with human cardiac troponin I and troponin C (HCTnI·TnC) complex showed a decrease in the Ca(2+) sensitivity of force development and an increase in maximal recovered force (HSSTnT1 and -3) compared with HCTnT3. In contrast, HSSTnTHyp showed an increase in the Ca(2+) sensitivity of force development. This suggests that re- or overexpression of specific SSTnT isoforms might have therapeutic potential in the failing heart because they increase the maximal force of contraction. In addition, circular dichroism and proteolytic digestion experiments revealed structural differences between HSSTnT isoforms and HCTnT3 and that HSSTnT1 is more susceptible to calpain and trypsin proteolysis than the other HSSTnTs. Overall, HSSTnT isoforms despite being homologues of CTnT may display distinct functional properties in muscle regulation.
Collapse
Affiliation(s)
- Jose Renato Pinto
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, Florida 33136, USA.
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Morris RT, Fine DM, Lees SJ, Booth FW, Link CD, Ferrario CM, Stump CS, Sowers JR. Exercise training prevents development of cardiac contractile dysfunction in hypertensive TG (mREN-2)27 rats. ACTA ACUST UNITED AC 2012; 1:393-9. [PMID: 19050745 DOI: 10.1016/j.jash.2007.09.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Angiotensin-II (Ang-II) contributes to cardiac remodeling and left ventricular dysfunction. In contrast, exercise may have beneficial effects on left ventricular structure and function. METHODS AND RESULTS We investigated the effects of low-intensity exercise training (ET) on in vivo cardiac function in hypertensive TG (mREN-2)27 rats (Ren-2) which develop left ventricular hypertrophy and dysfunction. Ren-2 rats and Sprague Dawley (SD) controls (4-5 weeks) began treadmill exercise every day for 5-6 weeks. Cardiac function was evaluated by echocardiography. Cardiac output and stroke volume were increased by ET in both 8-wk-old SD and Ren-2. Slope of mitral deceleration time, a non-invasive measure of diastolic function, was lower in the Ren-2 rats, but not changed by ET. LV collagen deposition, as assessed by hydroxyproline assay, was not affected by rat strain or ET at 10-11 weeks of age. Left ventricular B-type natriuretic peptide mRNA levels were higher in the Ren-2 rats (100%), but not affected by ET. Both alpha (~14.5 fold) and beta (~2.5 fold) myosin heavy chain mRNA were higher in the LV of Ren-2 rats (p < 0.05), but were not changed by ET. CONCLUSION Low-intensity exercise training in Ren-2 rats, a model of Ang-II-mediated hypertension, maintains cardiac index and stroke volume in the presence of impaired diastolic function at 8 wks of age.
Collapse
Affiliation(s)
- R Tyler Morris
- Department of Medical Pharmacology and Physiology, School of Medicine
| | | | | | | | | | | | | | | |
Collapse
|
47
|
|
48
|
Martin TP, Robinson E, Harvey AP, MacDonald M, Grieve DJ, Paul A, Currie S. Surgical optimization and characterization of a minimally invasive aortic banding procedure to induce cardiac hypertrophy in mice. Exp Physiol 2012; 97:822-32. [PMID: 22447975 DOI: 10.1113/expphysiol.2012.065573] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Left ventricular pressure overload in response to aortic banding is an invaluable model for studying progression of cardiac hypertrophy and transition to heart failure. Traditional aortic banding has recently been superceded by minimally invasive transverse aortic banding (MTAB), which does not require ventilation so is less technically challenging. Although the MTAB approach is superior, few laboratories have documented success, and minimal information on the model is available. The aim of this study was to optimize conditions for MTAB and to characterize the development and progression of cardiac hypertrophy. Isofluorane proved the most suitable anaesthetic for MTAB surgery in mice, and 1 week after surgery the MTAB animals showed significant increases in systolic blood pressure (MTAB 110 ± 6 mmHg versus sham 78 ± 3 mmHg, n = 7, P < 0.0001) and heart weight to body weight ratio (MTAB 6.2 ± 0.2 versus sham 5.1 ± 0.1, n = 12, P < 0.001), together with systolic (e.g. fractional shortening, MTAB 31.7 ± 1% versus sham 36.6 ± 1.4%, P = 0.01) and diastolic dysfunction (e.g. left ventricular end-diastolic pressure, MTAB 12.7 ± 1.0 mmHg versus sham 6.7 ± 0.8 mmHg, P < 0.001). Leucocyte infiltration to the heart was evident after 1 week in MTAB hearts, signifying an inflammatory response. More pronounced remodelling was observed 4 weeks postsurgery (heart weight to body weight ratio, MTAB 9.1 ± 0.6 versus sham 4.6 ± 0.04, n = 10, P < 0.0001) and fractional shortening was further decreased (MTAB 24.3 ± 2.5% versus sham 43.6 ± 1.7%, n = 10, P = 0.003), together with a significant increase in cardiac fibrosis and further cardiac inflammation. Our findings demonstrate that MTAB is a relevant experimental model for studying development and progression of cardiac hypertrophy, which will be highly valuable for future studies examining potential novel therapeutic interventions in this setting.
Collapse
Affiliation(s)
- Tamara P Martin
- Strathclyde Institute of Pharmacy & Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | | | | | | | | | | | | |
Collapse
|
49
|
Nakagawa Y, Inoue H, Shinone K, Ikemura M, Nata M. Molecular biological analysis of cardiac effect of high temperature in rats. Leg Med (Tokyo) 2012; 14:63-8. [DOI: 10.1016/j.legalmed.2011.12.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2011] [Revised: 12/02/2011] [Accepted: 12/07/2011] [Indexed: 11/17/2022]
|
50
|
Abstract
Heart failure is one of the common end stages of cardiovascular diseases, the leading cause of death in developed countries. Molecular mechanisms underlying the development of heart failure remain elusive but there is a consistent observation of chronic immune activation and aberrant microRNA (miRNA) expression that is present in failing hearts. This review will focus on the interplay between the immune system and miRNAs as factors that play a role during the development of heart failure. Several studies have shown that heart failure patients can be characterized by a sustained innate immune activation. The role of inflammatory signaling is discussed and TLR4 signaling, IL-1β, TNFα and IL-6 expression appears to coincide with the development of heart failure. Furthermore, we describe the implication of the renin angiotensin aldosteron system in immunity and heart failure. In the past decade microRNAs (miRNAs), small non-coding RNAs that translationally repress protein synthesis by binding to partially complementary sequences of mRNA, have come to light as important regulators of several kinds of cardiovascular diseases including cardiac hypertrophy and heart failure. The involvement of differentially expressed miRNAs in the inflammation that occurs during the development of heart failure is still subject of investigation. Here, we summarize and comment on the first studies in this field and hypothesize on the putative involvement of certain miRNAs in heart failure. MicroRNAs have been shown to be critical regulators of cardiac function and inflammation. Future research will have to point out if dampening the immune response, and the miRNAs associated with it, during the development of heart failure is a therapeutically plausible route to follow.
Collapse
|