1
|
Small GR. Myocardial extracellular volume by CT: Assessing the state of the union. J Cardiovasc Comput Tomogr 2024; 18:465-466. [PMID: 39098511 DOI: 10.1016/j.jcct.2024.07.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 07/30/2024] [Indexed: 08/06/2024]
Affiliation(s)
- Gary R Small
- Division of Cardiology, Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada.
| |
Collapse
|
2
|
Qi X, Hatami S, Bozso S, Wang X, Saleme B, Nagendran J, Michelakis E, Sutendra G, Freed DH. The Effects of Oxygen-Derived Free-Radical Scavengers During Normothermic Ex-Situ Heart Perfusion. ASAIO J 2024; 70:741-749. [PMID: 38457627 DOI: 10.1097/mat.0000000000002176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/10/2024] Open
Abstract
Oxidative stress occurs during ex-situ heart perfusion (ESHP) and may negatively affect functional preservation of the heart. We sought to assess the status of key antioxidant enzymes during ESHP, and the effects of augmenting these antioxidants on the attenuation of oxidative stress and improvement of myocardial and endothelial preservation in ESHP. Porcine hearts were perfused for 6 hours with oxygen-derived free-radical scavengers polyethylene glycol (PEG)-catalase or PEG-superoxide dismutase (SOD) or with naive perfusate (control). The oxidative stress-related modifications were determined in the myocardium and coronary vasculature, and contractile function, injury, and endothelial integrity were compared between the groups. The activity of key antioxidant enzymes decreased and adding catalase and SOD restored the enzyme activity. Cardiac function and endothelial integrity were preserved better with restored catalase activity. Catalase and SOD both decreased myocardial injury and catalase reduced ROS production and oxidative modification of proteins in the myocardium and coronary vasculature. The activity of antioxidant enzymes decrease in ESHP. Catalase may improve the preservation of cardiac function and endothelial integrity during ESHP. While catalase and SOD may both exert cardioprotective effects, unbalanced SOD and catalase activity may paradoxically increase the production of reactive species during ESHP.
Collapse
Affiliation(s)
- Xiao Qi
- From the Department of Surgery, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Sanaz Hatami
- Department of Medicine, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Sabin Bozso
- From the Department of Surgery, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Xiuhua Wang
- From the Department of Surgery, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Bruno Saleme
- Department of Medicine, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Jayan Nagendran
- From the Department of Surgery, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta, Canada
- Alberta Transplant Institute, Edmonton, Alberta, Canada
- Canadian Donation and Transplantation Research Program, Edmonton, Alberta, Canada
| | - Evangelos Michelakis
- Department of Medicine, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Gopinath Sutendra
- Department of Medicine, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Darren H Freed
- From the Department of Surgery, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta, Canada
- Alberta Transplant Institute, Edmonton, Alberta, Canada
- Canadian Donation and Transplantation Research Program, Edmonton, Alberta, Canada
- Department of Physiology, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta, Canada
- Department of Biomedical Engineering, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
3
|
Ferreira JP, Packer M, Sattar N, Butler J, Pocock SJ, Anker SD, Maldonado SG, Panova-Noeva M, Sumin M, Masson S, Zannad F, Januzzi JL. Carbohydrate antigen 125 concentrations across the ejection fraction spectrum in chronic heart failure: The EMPEROR programme. Eur J Heart Fail 2024; 26:788-802. [PMID: 38439582 DOI: 10.1002/ejhf.3166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 01/30/2024] [Accepted: 02/01/2024] [Indexed: 03/06/2024] Open
Abstract
AIM Vascular congestion may lead to an increase of carbohydrate antigen 125 (CA-125). The role of CA-125 as a biomarker of congestion or for prognosis across the full ejection fraction (EF) spectrum of chronic heart failure (HF) remains unknown. METHODS AND RESULTS Serum CA-125 was measured in 1111 study participants from the EMPEROR-Reduced and EMPEROR-Preserved trials. Congestive signs and symptoms were evaluated across CA-125 tertiles. Cox regression was used to study the association with outcomes. The primary outcome was a composite of first HF hospitalization or cardiovascular (CV) death. No significant association was present between baseline CA-125 levels and congestive signs or symptoms. In the overall population, higher CA-125 levels were not associated with an increased risk of primary outcome (tertile 3 vs. tertile 1: hazard ratio [HR] 1.34; 95% confidence interval [CI] 0.91-1.96; p-trend = 0.11). However, higher CA-125 levels were associated with an increased risk of primary outcome in patients with HF and reduced EF (HFrEF; tertile 3 vs. tertile 1: HR 2.25 [95% CI 1.30-3.89]), but not among patients with preserved EF (HFpEF; tertile 3 vs. tertile 1: HR 0.68 [95% CI 0.38-1.21]); interaction-p = 0.02). Patients in the upper CA-125 tertile also showed the steepest estimated glomerular filtration rate decline over time (p-trend = 0.03). The effect of empagliflozin to reduce the risk of CV death or HF hospitalization appeared to be attenuated in those with lower baseline CA-125 levels (interaction-p-trend = 0.09). CONCLUSION Across the range of EF in patients with chronic HF enrolled in the EMPEROR trials, the majority of whom did not have clinical evidence of congestion, CA-125 concentrations were not significantly associated with congestive signs or symptoms. CA-125 concentrations may predict HF hospitalization/CV death in patients with HFrEF, but not those with HFpEF. CLINICAL TRIAL REGISTRATION EMPEROR-Reduced (NCT03057977), EMPEROR-Preserved (NCT03057951).
Collapse
Affiliation(s)
- João Pedro Ferreira
- Department of Surgery and Physiology, Faculty of Medicine of the University of Porto, Cardiovascular Research and Development Center (UnIC@RISE), Porto, Portugal
- Heart Failure Clinic, Internal Medicine Department, Centro Hospitalar de Vila Nova de Gaia/Espinho, Gaia, Portugal
- Centre d'Investigations Cliniques Plurithématique 14-33, Inserm U1116, CHRU, F-CRIN INI-CRCT (Cardiovascular and Renal Clinical Trialists), Cardiovascular Research and Development Center, Université de Lorraine, Nancy, France
| | - Milton Packer
- Imperial College London, London, UK
- Baylor Heart and Vascular Institute, Baylor University Medical Center, Dallas, TX, USA
| | - Naveed Sattar
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Javed Butler
- Baylor Scott and White Research Institute, Dallas, TX, USA
- University of Mississippi Medical Center, Jackson, MS, USA
| | | | - Stefan D Anker
- Center for Regenerative Therapies, Berlin Institute of Health, Department of Cardiology, German Centre for Cardiovascular Research, partner site Berlin, Charité Universitätsmedizin Berlin, Berlin, Germany
| | | | | | - Mikhail Sumin
- Boehringer Ingelheim International GmbH, Ingelheim, Germany
| | - Serge Masson
- Roche Diagnostics International Ltd, Rotkreuz, Switzerland
| | - Faiez Zannad
- Centre d'Investigations Cliniques Plurithématique 1433, INSERM, Université de Lorraine, Nancy, France
| | - James L Januzzi
- Massachusetts General Hospital and Baim Institute for Clinical Research, Boston, MA, USA
| |
Collapse
|
4
|
Guan D, Tian L, Li W, Gao H. Using LDDMM and a kinematic cardiac growth model to quantify growth and remodelling in rat hearts under PAH. Comput Biol Med 2024; 171:108218. [PMID: 38428098 DOI: 10.1016/j.compbiomed.2024.108218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 01/20/2024] [Accepted: 02/25/2024] [Indexed: 03/03/2024]
Abstract
Pulmonary arterial hypertension (PAH) is a rapidly progressive and fatal disease, with right ventricular failure being the primary cause of death in patients with PAH. This study aims to determine the mechanical stimuli that may initiate heart growth and remodelling (G&R). To achieve this, two bi-ventricular models were constructed: one for a control rat heart and another for a rat heart with PAH. The growth of the diseased heart was estimated by warping it to the control heart using an improved large deformation diffeomorphic metric mapping (LDDMM) framework. Correlation analysis was then performed between mechanical cues (stress and strain) and growth tensors, which revealed that principal strains may serve as a triggering stimulus for myocardial growth and remodelling under PAH. The growth tensors, estimated from in vivo images, could explain 84.3% of the observed geometrical changes in the diseased heart with PAH by using a kinematic cardiac growth model. Our approach has the potential to quantify G&R using sparse in vivo images and to provide insights into the underlying mechanism of triggering right heart failure from a biomechanical perspective.
Collapse
Affiliation(s)
- Debao Guan
- School of Control Science and Engineering, Shandong University, China; School of Mathematics and Statistics, University of Glasgow, UK
| | - Lian Tian
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, UK
| | - Wei Li
- School of Control Science and Engineering, Shandong University, China
| | - Hao Gao
- School of Mathematics and Statistics, University of Glasgow, UK.
| |
Collapse
|
5
|
Yi B, Xu Y, Wang X, Wang G, Li S, Xu R, Liu X, Zhou Q. Overview of Injectable Hydrogels for the Treatment of Myocardial Infarction. CARDIOVASCULAR INNOVATIONS AND APPLICATIONS 2024; 9. [DOI: 10.15212/cvia.2024.0019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025] Open
Abstract
Myocardial infarction (MI) triggers adverse remodeling mechanisms, thus leading to heart failure. Since the application of biomaterial-based scaffolds emerged as a viable approach for providing mechanical support and promoting cell growth, injectable hydrogels have garnered substantial attention in MI treatment because of their minimally invasive administration through injection and diminished risk of infection. To fully understand the interplay between injectable hydrogels and infarcted myocardium repair, this review provides an overview of recent advances in injectable hydrogel-mediated MI therapy, including: I) material designs for repairing the infarcted myocardium, considering the pathophysiological mechanism of MI and design principles for biomaterials in MI treatment; II) the development of injectable functional hydrogels for MI treatment, including conductive, self-healing, drug-loaded, and stimulus-responsive hydrogels; and III) research progress in using injectable hydrogels to restore cardiac function in infarcted myocardium by promoting neovascularization, enhancing cardiomyocyte proliferation, decreasing myocardial fibrosis, and inhibiting excessive inflammation. Overall, this review presents the current state of injectable hydrogel research in MI treatment, offering valuable information to facilitate interdisciplinary knowledge transfer and enable the development of prognostic markers for suitable injectable materials.
Collapse
|
6
|
Elkrief D, Matusovsky O, Cheng YS, Rassier DE. From amino-acid to disease: the effects of oxidation on actin-myosin interactions in muscle. J Muscle Res Cell Motil 2023; 44:225-254. [PMID: 37805961 DOI: 10.1007/s10974-023-09658-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 08/15/2023] [Indexed: 10/10/2023]
Abstract
Actin-myosin interactions form the basis of the force-producing contraction cycle within the sarcomere, serving as the primary mechanism for muscle contraction. Post-translational modifications, such as oxidation, have a considerable impact on the mechanics of these interactions. Considering their widespread occurrence, the explicit contributions of these modifications to muscle function remain an active field of research. In this review, we aim to provide a comprehensive overview of the basic mechanics of the actin-myosin complex and elucidate the extent to which oxidation influences the contractile cycle and various mechanical characteristics of this complex at the single-molecule, myofibrillar and whole-muscle levels. We place particular focus on amino acids shown to be vulnerable to oxidation in actin, myosin, and some of their binding partners. Additionally, we highlight the differences between in vitro environments, where oxidation is controlled and limited to actin and myosin and myofibrillar or whole muscle environments, to foster a better understanding of oxidative modification in muscle. Thus, this review seeks to encompass a broad range of studies, aiming to lay out the multi layered effects of oxidation in in vitro and in vivo environments, with brief mention of clinical muscular disorders associated with oxidative stress.
Collapse
Affiliation(s)
- Daren Elkrief
- Department of Physiology, McGill University, Montreal, QC, Canada
| | - Oleg Matusovsky
- Department of Kinesiology and Physical Education, McGill University, Montreal, QC, Canada
| | - Yu-Shu Cheng
- Department of Kinesiology and Physical Education, McGill University, Montreal, QC, Canada
| | - Dilson E Rassier
- Department of Physiology, McGill University, Montreal, QC, Canada.
- Department of Kinesiology and Physical Education, McGill University, Montreal, QC, Canada.
- Simon Fraser University, Burnaby, BC, Canada.
| |
Collapse
|
7
|
Guan D, Zhuan X, Luo X, Gao H. An updated Lagrangian constrained mixture model of pathological cardiac growth and remodelling. Acta Biomater 2023; 166:375-399. [PMID: 37201740 DOI: 10.1016/j.actbio.2023.05.022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 05/03/2023] [Accepted: 05/10/2023] [Indexed: 05/20/2023]
Abstract
Progressive left ventricular (LV) growth and remodelling (G&R) is often induced by volume and pressure overload, characterized by structural and functional adaptation through myocyte hypertrophy and extracellular matrix remodelling, which are dynamically regulated by biomechanical factors, inflammation, neurohormonal pathways, etc. When prolonged, it can eventually lead to irreversible heart failure. In this study, we have developed a new framework for modelling pathological cardiac G&R based on constrained mixture theory using an updated reference configuration, which is triggered by altered biomechanical factors to restore biomechanical homeostasis. Eccentric and concentric growth, and their combination have been explored in a patient-specific human LV model under volume and pressure overload. Eccentric growth is triggered by overstretching of myofibres due to volume overload, i.e. mitral regurgitation, whilst concentric growth is driven by excessive contractile stress due to pressure overload, i.e. aortic stenosis. Different biological constituent's adaptations under pathological conditions are integrated together, which are the ground matrix, myofibres and collagen network. We have shown that this constrained mixture-motivated G&R model can capture different phenotypes of maladaptive LV G&R, such as chamber dilation and wall thinning under volume overload, wall thickening under pressure overload, and more complex patterns under both pressure and volume overload. We have further demonstrated how collagen G&R would affect LV structural and functional adaption by providing mechanistic insight on anti-fibrotic interventions. This updated Lagrangian constrained mixture based myocardial G&R model has the potential to understand the turnover processes of myocytes and collagen due to altered local mechanical stimuli in heart diseases, and in providing mechanistic links between biomechanical factors and biological adaption at both the organ and cellular levels. Once calibrated with patient data, it can be used for assessing heart failure risk and designing optimal treatment therapies. STATEMENT OF SIGNIFICANCE: Computational modelling of cardiac G&R has shown high promise to provide insight into heart disease management when mechanistic understandings are quantified between biomechanical factors and underlying cellular adaptation processes. The kinematic growth theory has been dominantly used to phenomenologically describe the biological G&R process but neglecting underlying cellular mechanisms. We have developed a constrained mixture based G&R model with updated reference by taking into account different mechanobiological processes in the ground matrix, myocytes and collagen fibres. This G&R model can serve as a basis for developing more advanced myocardial G&R models further informed by patient data to assess heart failure risk, predict disease progression, select the optimal treatment by hypothesis testing, and eventually towards a truly precision cardiology using in-silico models.
Collapse
Affiliation(s)
- Debao Guan
- School of Mathematics and Statistics, University of Glasgow, Glasgow G12 8QQ, UK
| | - Xin Zhuan
- School of Mathematics and Statistics, University of Glasgow, Glasgow G12 8QQ, UK
| | - Xiaoyu Luo
- School of Mathematics and Statistics, University of Glasgow, Glasgow G12 8QQ, UK
| | - Hao Gao
- School of Mathematics and Statistics, University of Glasgow, Glasgow G12 8QQ, UK.
| |
Collapse
|
8
|
Al-U’datt DGF, Tranchant CC, Al-Husein B, Hiram R, Al-Dwairi A, AlQudah M, Al-shboul O, Jaradat S, Alqbelat J, Almajwal A. Involvement and possible role of transglutaminases 1 and 2 in mediating fibrotic signalling, collagen cross-linking and cell proliferation in neonatal rat ventricular fibroblasts. PLoS One 2023; 18:e0281320. [PMID: 36848364 PMCID: PMC9970086 DOI: 10.1371/journal.pone.0281320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 01/19/2023] [Indexed: 03/01/2023] Open
Abstract
Transglutaminase (TG) isoforms control diverse normal and pathophysiologic processes through their capacity to cross-link extracellular matrix (ECM) proteins. Their functional and signalling roles in cardiac fibrosis remain poorly understood, despite some evidence of TG2 involvement in abnormal ECM remodelling in heart diseases. In this study, we investigated the role of TG1 and TG2 in mediating fibrotic signalling, collagen cross-linking, and cell proliferation in healthy fibroblasts by siRNA-mediated knockdown. siRNA for TG1, TG2 or negative control was transfected into cultured neonatal rat ventricular fibroblasts and cardiomyocytes. mRNA expression of TGs and profibrotic, proliferation and apoptotic markers was assessed by qPCR. Cell proliferation and soluble and insoluble collagen were determined by ELISA and LC-MS/MS, respectively. TG1 and TG2 were both expressed in neonatal rat cardiomyocytes and fibroblasts before transfection. Other TGs were not detected before and after transfection. TG2 was predominantly expressed and more effectively silenced than TG1. Knocking down TG1 or TG2 significantly modified profibrotic markers mRNA expression in fibroblasts, decreasing connective tissue growth factor (CTGF) and increasing transforming growth factor-β1 compared to the negative siRNA control. Reduced expression of collagen 3A1 was found upon TG1 knockdown, while TG2 knockdown raised α-smooth muscle actin expression. TG2 knockdown further increased fibroblast proliferation and the expression of proliferation marker cyclin D1. Lower insoluble collagen content and collagen cross-linking were evidenced upon silencing TG1 or TG2. Transcript levels of collagen 1A1, fibronectin 1, matrix metalloproteinase-2, cyclin E2, and BCL-2-associated X protein/B-cell lymphoma 2 ratio were strongly correlated with TG1 mRNA expression, whereas TG2 expression correlated strongly with CTGF mRNA abundance. These findings support a functional and signalling role for TG1 and TG2 from fibroblasts in regulating key processes underlying myocardial ECM homeostasis and dysregulation, suggesting that these isoforms could be potential and promising targets for the development of cardiac fibrosis therapies.
Collapse
Affiliation(s)
- Doa’a G. F. Al-U’datt
- Department of Physiology and Biochemistry, Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - Carole C. Tranchant
- School of Food Science, Nutrition and Family Studies, Faculty of Health Sciences and Community Services, Université de Moncton, New Brunswick, Canada
| | - Belal Al-Husein
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| | - Roddy Hiram
- Montreal Heart Institute, Université de Montréal, Montreal, Quebec, Canada
- Department of Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Ahmed Al-Dwairi
- Department of Physiology and Biochemistry, Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - Mohammad AlQudah
- Department of Physiology and Biochemistry, Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
- Physiology Department, Arabian Gulf University, Manama, Bahrain
| | - Othman Al-shboul
- Department of Physiology and Biochemistry, Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - Saied Jaradat
- Princess Haya Biotechnology Center, Jordan University of Science and Technology, Irbid, Jordan
| | - Jenan Alqbelat
- Department of Physiology and Biochemistry, Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - Ali Almajwal
- Department of Community Health Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
9
|
Cavalcante JS, Borges da Silva WRG, de Oliveira LA, Brito IMC, Muller KS, J Vidal IS, Dos Santos LD, Jorge RJB, Almeida C, de Lima Bicho C. Blood plasma proteome alteration after local tissue damage induced by Bothrops erythromelas snake venom in mice. J Proteomics 2022; 269:104742. [PMID: 36174952 DOI: 10.1016/j.jprot.2022.104742] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 09/03/2022] [Accepted: 09/22/2022] [Indexed: 11/26/2022]
Abstract
Snakes of the genus Bothrops are responsible the most snakebites in the Brazil, causing a diverse and complex pathophysiological condition. Bothrops erythromelas is the main specie of medical relevance found in the Caatinga from the Brazilian Northeast region. The pathophysiological effects involving B. erythromelas snakebite as well as the organism reaction in response to this envenomation are not so explored. Thus, edema was induced in mice paws using 2.5 μg or 5.0 μg of B. erythromelas venom, and the percentage of edema was measured. Plasma was collected 30 minutes after the envenomation-induced in mice and analyzed by mass spectrometry. It was identified a total of 112 common plasma proteins differentially abundant among experimental groups, which are involved with the complement system and coagulation cascades, oxidative stress, neutrophil degranulation, platelets degranulation and inflammatory response. Apolipoprotein A1 (Apoa), serum amyloid protein A-4 (Saa4), adiponectin (Adipoq) showed up-regulated in mice plasma after injection of venom, while fibulin (Fbln1), factor XII (F12) and vitamin K-dependent protein Z (Proz) showed down-regulated. The results indicate a protein pattern of thrombo-inflammation to the B. erythromelas snakebite, evidencing potential biomarkers for monitoring this snakebite, new therapeutic targets and its correlations with the degree of envenomation once showed modulations in the abundance among the different groups according to the amount of venom injected into the mice.
Collapse
Affiliation(s)
- Joeliton S Cavalcante
- Graduate Program in Tropical Diseases, Botucatu Medical School (FMB), São Paulo University (UNESP - Univ Estadual Paulista), Botucatu, São Paulo, Brazil.
| | - Weslley Ruan G Borges da Silva
- Department of Biology, Center of Biological and Health Sciences, Paraíba State University (UEPB), Campina Grande, Paraíba, Brazil
| | - Laudicéia Alves de Oliveira
- Graduate Program in Tropical Diseases, Botucatu Medical School (FMB), São Paulo University (UNESP - Univ Estadual Paulista), Botucatu, São Paulo, Brazil
| | - Ingrid Mayara C Brito
- Graduate Program in Tropical Diseases, Botucatu Medical School (FMB), São Paulo University (UNESP - Univ Estadual Paulista), Botucatu, São Paulo, Brazil
| | - Kevin S Muller
- Institute of Biosciences, São Paulo University (UNESP - Univ Estadual Paulista), Botucatu, São Paulo, Brazil
| | - Ivynna Suellen J Vidal
- Graduate Program in Translational Medicine, Drug Research and Development Center, Federal University of Ceará (UFC), Fortaleza, Ceará, Brazil
| | - Lucilene Delazari Dos Santos
- Graduate Program in Tropical Diseases, Botucatu Medical School (FMB), São Paulo University (UNESP - Univ Estadual Paulista), Botucatu, São Paulo, Brazil; Biotechnology Institute (IBTEC), São Paulo University (UNESP - Univ Estadual Paulista), Botucatu, São Paulo, Brazil
| | - Roberta Jeane Bezerra Jorge
- Drug Research and Development Center, Federal University of Ceará (UFC), Fortaleza, Ceará, Brazil; Department of Physiology and Pharmacology, School of Medicine, Federal University of Ceará (UFC), Fortaleza, Ceará, Brazil
| | - Cayo Almeida
- Center of Mathematics, Computing Sciences and Cognition, Federal University of ABC, São Paulo, SP, Brazil
| | - Carla de Lima Bicho
- Department of Biology, Center of Biological and Health Sciences, Paraíba State University (UEPB), Campina Grande, Paraíba, Brazil
| |
Collapse
|
10
|
Gonçalves PR, Nascimento LD, Gerlach RF, Rodrigues KE, Prado AF. Matrix Metalloproteinase 2 as a Pharmacological Target in Heart Failure. Pharmaceuticals (Basel) 2022; 15:ph15080920. [PMID: 35893744 PMCID: PMC9331741 DOI: 10.3390/ph15080920] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 07/11/2022] [Accepted: 07/11/2022] [Indexed: 12/18/2022] Open
Abstract
Heart failure (HF) is an acute or chronic clinical syndrome that results in a decrease in cardiac output and an increase in intracardiac pressure at rest or upon exertion. The pathophysiology of HF is heterogeneous and results from an initial harmful event in the heart that promotes neurohormonal changes such as autonomic dysfunction and activation of the renin-angiotensin-aldosterone system, endothelial dysfunction, and inflammation. Cardiac remodeling occurs, which is associated with degradation and disorganized synthesis of extracellular matrix (ECM) components that are controlled by ECM metalloproteinases (MMPs). MMP-2 is part of this group of proteases, which are classified as gelatinases and are constituents of the heart. MMP-2 is considered a biomarker of patients with HF with reduced ejection fraction (HFrEF) or preserved ejection fraction (HFpEF). The role of MMP-2 in the development of cardiac injury and dysfunction has clearly been demonstrated in animal models of cardiac ischemia, transgenic models that overexpress MMP-2, and knockout models for this protease. New research to minimize cardiac structural and functional alterations using non-selective and selective inhibitors for MMP-2 demonstrates that this protease could be used as a possible pharmacological target in the treatment of HF.
Collapse
Affiliation(s)
- Pricila Rodrigues Gonçalves
- Cardiovascular System Pharmacology and Toxicology Laboratory, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, PA, Brazil; (P.R.G.); (L.D.N.); (K.E.R.)
| | - Lisandra Duarte Nascimento
- Cardiovascular System Pharmacology and Toxicology Laboratory, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, PA, Brazil; (P.R.G.); (L.D.N.); (K.E.R.)
| | - Raquel Fernanda Gerlach
- Department of Basic and Oral Biology, Faculty of Dentistry of Ribeirao Preto, University of Sao Paulo (FORP/USP), Ribeirao Preto 14040-904, SP, Brazil;
| | - Keuri Eleutério Rodrigues
- Cardiovascular System Pharmacology and Toxicology Laboratory, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, PA, Brazil; (P.R.G.); (L.D.N.); (K.E.R.)
| | - Alejandro Ferraz Prado
- Cardiovascular System Pharmacology and Toxicology Laboratory, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, PA, Brazil; (P.R.G.); (L.D.N.); (K.E.R.)
- Correspondence:
| |
Collapse
|
11
|
Corporan D, Saadeh M, Yoldas A, Mudigonda J, Lane BA, Padala M. Passive mechanical properties of the left ventricular myocardium and extracellular matrix in hearts with chronic volume overload from mitral regurgitation. Physiol Rep 2022; 10:e15305. [PMID: 35871778 PMCID: PMC9309441 DOI: 10.14814/phy2.15305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 04/05/2022] [Accepted: 04/14/2022] [Indexed: 06/15/2023] Open
Abstract
Cardiac volume overload from mitral regurgitation (MR) is a trigger for left ventricular dilatation, remodeling, and ultimate failure. While the functional and structural adaptations to this overload are known, the adaptation of myocardial mechanical properties remains unknown. Using a rodent model of MR, in this study, we discern changes in the passive material properties of the intact and decellularized myocardium. Eighty Sprague-Dawley rats (350-400 g) were assigned to two groups: (1) MR (n = 40) and (2) control (n = 40). MR was induced in the beating heart by perforating the mitral leaflet with a 23G needle, and rats were terminated at 2, 10, 20, or 40 weeks (n = 10/time-point). Echocardiography was performed at baseline and termination, and explanted hearts were used for equibiaxial mechanical testing of the intact myocardium and after decellularization. Two weeks after inducing severe MR, the myocardium was more extensible compared to control, however, stiffness and extensibility of the extracellular matrix did not differ from control at this timepoint. By 20 weeks, the myocardium was stiffer with a higher elastic modulus of 1920 ± 246 kPa, and a parallel rise in extracellular matrix stiffness. Despite some matrix stiffening, it only contributed to 31% and 36% of the elastic modulus of the intact tissue in the circumferential and longitudinal directions. At 40 weeks, similar trends of increasing stiffness were observed, but the contribution of extracellular matrix remained relatively low. Chronic MR induces ventricular myocardial stiffening, which seems to be driven by the myocyte compartment of the muscle, and not the extracellular matrix.
Collapse
Affiliation(s)
- Daniella Corporan
- Structural Heart Research and Innovation LaboratoryCarlyle Fraser Heart CenterEmory University Hospital MidtownAtlantaGeorgiaUSA
- Division of Cardiothoracic SurgeryDepartment of SurgeryEmory University School of MedicineAtlantaGeorgiaUSA
| | - Maher Saadeh
- Structural Heart Research and Innovation LaboratoryCarlyle Fraser Heart CenterEmory University Hospital MidtownAtlantaGeorgiaUSA
| | - Alessandra Yoldas
- Structural Heart Research and Innovation LaboratoryCarlyle Fraser Heart CenterEmory University Hospital MidtownAtlantaGeorgiaUSA
| | - Jahnavi Mudigonda
- Structural Heart Research and Innovation LaboratoryCarlyle Fraser Heart CenterEmory University Hospital MidtownAtlantaGeorgiaUSA
- Division of Cardiothoracic SurgeryDepartment of SurgeryEmory University School of MedicineAtlantaGeorgiaUSA
| | - Brooks Alexander Lane
- Structural Heart Research and Innovation LaboratoryCarlyle Fraser Heart CenterEmory University Hospital MidtownAtlantaGeorgiaUSA
- Division of Cardiothoracic SurgeryDepartment of SurgeryEmory University School of MedicineAtlantaGeorgiaUSA
| | - Muralidhar Padala
- Structural Heart Research and Innovation LaboratoryCarlyle Fraser Heart CenterEmory University Hospital MidtownAtlantaGeorgiaUSA
- Division of Cardiothoracic SurgeryDepartment of SurgeryEmory University School of MedicineAtlantaGeorgiaUSA
| |
Collapse
|
12
|
The Bioinformatical Identification of Potential Biomarkers in Heart Failure Diagnosis and Treatment. Genet Res (Camb) 2022; 2022:8727566. [PMID: 35645616 PMCID: PMC9126668 DOI: 10.1155/2022/8727566] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 04/11/2022] [Accepted: 04/15/2022] [Indexed: 12/20/2022] Open
Abstract
Background Heart failure (HF) is defined as the inability of the heart's systolic and diastolic function to properly discharge blood flow from the veins to the heart. The goal of our research is to look into the possible mechanism that causes HF. Methods The GSE5406 database was used for screening the differentially expressed genes (DEGs). Gene ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and Protein-Protein Interaction (PPI) network were applied to analyze DEGs. Besides, cell counting Kit-8 (CCK-8) was conducted to observe the knockdown effect of hub genes on cell proliferation. Results Finally, 377 upregulated and 461 downregulated DEGs came out, enriched in the extracellular matrix organization and gap junction. According to GSEA results, Hoft cd4 positive alpha beta memory t cell bcg vaccine age 18–45 yo id 7 dy top 100 deg ex vivo up, Sobolev t cell pandemrix age 18–64 yo 7 dy dn, and so on were significantly related to gene set GSE5406. 7 hub genes, such as COL1A1, UBB, COL3A1, HSP90AA1, MYC, STAT3 and MAPK1, were selected from PPI networks. CCK-8 indicated silencing of STAT3 promoted the proliferation of H9C2 cells and silencing of UBB inhibited the proliferation of H9C2 cells. Conclusion Our analysis reveals that COL1A1, UBB, COL3A1, HSP90AA1, MYC, STAT3, and MAPK1 might promote the progression of HF and become the biomarkers for diagnosis and treatment of HF.
Collapse
|
13
|
Khomtchouk BB, Lee YS, Khan ML, Sun P, Mero D, Davidson MH. Targeting the cytoskeleton and extracellular matrix in cardiovascular disease drug discovery. Expert Opin Drug Discov 2022; 17:443-460. [PMID: 35258387 PMCID: PMC9050939 DOI: 10.1080/17460441.2022.2047645] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 02/24/2022] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Currently, cardiovascular disease (CVD) drug discovery has focused primarily on addressing the inflammation and immunopathology aspects inherent to various CVD phenotypes such as cardiac fibrosis and coronary artery disease. However, recent findings suggest new biological pathways for cytoskeletal and extracellular matrix (ECM) regulation across diverse CVDs, such as the roles of matricellular proteins (e.g. tenascin-C) in regulating the cellular microenvironment. The success of anti-inflammatory drugs like colchicine, which targets microtubule polymerization, further suggests that the cardiac cytoskeleton and ECM provide prospective therapeutic opportunities. AREAS COVERED Potential therapeutic targets include proteins such as gelsolin and calponin 2, which play pivotal roles in plaque development. This review focuses on the dynamic role that the cytoskeleton and ECM play in CVD pathophysiology, highlighting how novel target discovery in cytoskeletal and ECM-related genes may enable therapeutics development to alter the regulation of cellular architecture in plaque formation and rupture, cardiac contractility, and other molecular mechanisms. EXPERT OPINION Further research into the cardiac cytoskeleton and its associated ECM proteins is an area ripe for novel target discovery. Furthermore, the structural connection between the cytoskeleton and the ECM provides an opportunity to evaluate both entities as sources of potential therapeutic targets for CVDs.
Collapse
Affiliation(s)
- Bohdan B. Khomtchouk
- University of Chicago, Department of Medicine, Section of Computational Biomedicine and Biomedical Data Science, Institute for Genomics and Systems Biology, Chicago, IL USA
| | - Yoon Seo Lee
- The College of the University of Chicago, Chicago, IL USA
| | - Maha L. Khan
- The College of the University of Chicago, Chicago, IL USA
| | - Patrick Sun
- The College of the University of Chicago, Chicago, IL USA
| | | | - Michael H. Davidson
- University of Chicago, Department of Medicine, Section of Cardiology, Chicago, IL USA
| |
Collapse
|
14
|
Pype LL, Bertrand PB, Paelinck BP, Heidbuchel H, Van Craenenbroeck EM, Van De Heyning CM. Left Ventricular Remodeling in Non-syndromic Mitral Valve Prolapse: Volume Overload or Concomitant Cardiomyopathy? Front Cardiovasc Med 2022; 9:862044. [PMID: 35498019 PMCID: PMC9039519 DOI: 10.3389/fcvm.2022.862044] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 03/07/2022] [Indexed: 01/11/2023] Open
Abstract
Mitral valve prolapse (MVP) is a common valvular disorder that can be associated with mitral regurgitation (MR), heart failure, ventricular arrhythmias and sudden cardiac death. Given the prognostic impact of these conditions, it is important to evaluate not only mitral valve morphology and regurgitation, but also the presence of left ventricular (LV) function and remodeling. To date, several possible hypotheses have been proposed regarding the underlying mechanisms of LV remodeling in the context of non-syndromic MVP, but the exact pathophysiological explanation remains elusive. Overall, volume overload related to severe MR is considered the main cause of LV dilatation in MVP. However, significant LV remodeling has been observed in patients with MVP and no/mild MR, particularly in patients with bileaflet MVP or Barlow's disease, generating several new hypotheses. Recently, the concept of "prolapse volume" was introduced, adding a significant volume load to the LV on top of the transvalvular MR volume. Another possible hypothesis is the existence of a concomitant cardiomyopathy, supported by the link between MVP and myocardial fibrosis. The origin of this cardiomyopathy could be either genetic, a second hit (e.g., on top of genetic predisposition) and/or frequent ventricular ectopic beats. This review provides an overview of the different mechanisms and remaining questions regarding LV remodeling in non-syndromic MVP. Since technical specifications of imaging modalities impact the evaluation of MR severity and LV remodeling, and therefore might influence clinical decision making in these patients, this review will also discuss assessment of MVP using different imaging modalities.
Collapse
Affiliation(s)
- Lobke L. Pype
- Department of Cardiology, Antwerp University Hospital, Antwerp, Belgium
- Genetics, Pharmacology and Physiopathology of Heart, Vasculature and Skeleton (GENCOR) Research Group, University of Antwerp, Antwerp, Belgium
| | - Philippe B. Bertrand
- Department of Cardiology, Ziekenhuis Oost-Limburg, Genk, Belgium
- Cardio and Organ Systems (COST) Resarch Group, Hasselt University, Hasselt, Belgium
| | - Bernard P. Paelinck
- Genetics, Pharmacology and Physiopathology of Heart, Vasculature and Skeleton (GENCOR) Research Group, University of Antwerp, Antwerp, Belgium
- Department of Cardiac Surgery, Antwerp University Hospital, Antwerp, Belgium
| | - Hein Heidbuchel
- Department of Cardiology, Antwerp University Hospital, Antwerp, Belgium
- Genetics, Pharmacology and Physiopathology of Heart, Vasculature and Skeleton (GENCOR) Research Group, University of Antwerp, Antwerp, Belgium
| | - Emeline M. Van Craenenbroeck
- Department of Cardiology, Antwerp University Hospital, Antwerp, Belgium
- Genetics, Pharmacology and Physiopathology of Heart, Vasculature and Skeleton (GENCOR) Research Group, University of Antwerp, Antwerp, Belgium
| | - Caroline M. Van De Heyning
- Department of Cardiology, Antwerp University Hospital, Antwerp, Belgium
- Genetics, Pharmacology and Physiopathology of Heart, Vasculature and Skeleton (GENCOR) Research Group, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
15
|
Zelko IN, Dassanayaka S, Malovichko MV, Howard CM, Garrett LF, Uchida S, Brittian KR, Conklin DJ, Jones SP, Srivastava S. Chronic Benzene Exposure Aggravates Pressure Overload-Induced Cardiac Dysfunction. Toxicol Sci 2021; 185:64-76. [PMID: 34718823 DOI: 10.1093/toxsci/kfab125] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Benzene is a ubiquitous environmental pollutant abundant in household products, petrochemicals and cigarette smoke. Benzene is a well-known carcinogen in humans and experimental animals; however, little is known about the cardiovascular toxicity of benzene. Recent population-based studies indicate that benzene exposure is associated with an increased risk for heart failure. Nonetheless, it is unclear whether benzene exposure is sufficient to induce and/or exacerbate heart failure. We examined the effects of benzene (50 ppm, 6 h/day, 5 days/week, 6 weeks) or HEPA-filtered air exposure on transverse aortic constriction (TAC)-induced pressure overload in male C57BL/6J mice. Our data show that benzene exposure had no effect on cardiac function in the Sham group; however, it significantly compromised cardiac function as depicted by a significant decrease in fractional shortening and ejection fraction, as compared with TAC/Air-exposed mice. RNA-seq analysis of the cardiac tissue from the TAC/benzene-exposed mice showed a significant increase in several genes associated with adhesion molecules, cell-cell adhesion, inflammation, and stress response. In particular, neutrophils were implicated in our unbiased analyses. Indeed, immunofluorescence studies showed that TAC/benzene exposure promotes infiltration of CD11b+/S100A8+/myeloperoxidase+-positive neutrophils in the hearts by 3-fold. In vitro, the benzene metabolites, hydroquinone and catechol, induced the expression of P-selectin in cardiac microvascular endothelial cells by 5-fold and increased the adhesion of neutrophils to these endothelial cells by 1.5-2.0-fold. Benzene metabolite-induced adhesion of neutrophils to the endothelial cells was attenuated by anti-P-selectin antibody. Together, these data suggest that benzene exacerbates heart failure by promoting endothelial activation and neutrophil recruitment.
Collapse
Affiliation(s)
- Igor N Zelko
- University of Louisville Superfund Research Center.,Diabetes and Obesity Center.,Envirome Institute.,Department of Medicine, Division of Environmental Medicine, University of Louisville, Louisville, KY 40202
| | - Sujith Dassanayaka
- Diabetes and Obesity Center.,Department of Medicine, Division of Environmental Medicine, University of Louisville, Louisville, KY 40202
| | - Marina V Malovichko
- University of Louisville Superfund Research Center.,Diabetes and Obesity Center.,Envirome Institute.,Department of Medicine, Division of Environmental Medicine, University of Louisville, Louisville, KY 40202
| | - Caitlin M Howard
- Diabetes and Obesity Center.,Envirome Institute.,Department of Medicine, Division of Environmental Medicine, University of Louisville, Louisville, KY 40202
| | - Lauren F Garrett
- Diabetes and Obesity Center.,Envirome Institute.,Department of Medicine, Division of Environmental Medicine, University of Louisville, Louisville, KY 40202
| | - Shizuka Uchida
- Center for RNA Medicine, Department of Clinical Medicine, Aalborg University, Copenhagen SV, Denmark
| | - Kenneth R Brittian
- Diabetes and Obesity Center.,Envirome Institute.,Department of Medicine, Division of Environmental Medicine, University of Louisville, Louisville, KY 40202
| | - Daniel J Conklin
- University of Louisville Superfund Research Center.,Diabetes and Obesity Center.,Envirome Institute.,Department of Medicine, Division of Environmental Medicine, University of Louisville, Louisville, KY 40202
| | - Steven P Jones
- Diabetes and Obesity Center.,Envirome Institute.,Department of Medicine, Division of Environmental Medicine, University of Louisville, Louisville, KY 40202
| | - Sanjay Srivastava
- University of Louisville Superfund Research Center.,Diabetes and Obesity Center.,Envirome Institute.,Department of Medicine, Division of Environmental Medicine, University of Louisville, Louisville, KY 40202
| |
Collapse
|
16
|
Zhang S, Ou K, Huang J, Fang L, Wang C, Wang Q. Prenatal EGCG exposure-induced heart mass reduction in adult male mice and underlying mechanisms. Food Chem Toxicol 2021; 157:112588. [PMID: 34600025 DOI: 10.1016/j.fct.2021.112588] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 09/24/2021] [Accepted: 09/28/2021] [Indexed: 12/17/2022]
Abstract
Epigallocatechin-3-gallate (EGCG), which is a major polyphenol in tea, has an unclear effect on cardiac development. In the present study, mice (C57BL/6) were exposed in utero to EGCG dissolved in drinking water (3 μg/ml) for 16 days. A significant decrease in the heart/body weight ratio was observed in adult males but not in adult females. The protein expression levels of TGF-β1 and its downstream transcription factors SMAD3 and SMAD4 were significantly decreased in male hearts. The PI3K/AKT signaling pathway was inhibited, the expression of proapoptotic proteins, such as BAX, Cleaved Caspase3 and Cleaved Caspase9, was elevated, and the level of antiapoptotic proteins, such as BCL-2, was decreased. A reduced heart/body weight ratio may be associated with the loss of cardiac fibers and an increase in myocardial apoptosis. The cardiac levels of aromatic hydrocarbon receptor and androgen receptor were elevated only in males, which may explain the sexual dimorphism in the effects. The promoter methylation levels of pik3r1, tgf-β, smad4 were elevated, and those of ahr were reduced, explaining the mechanism underlying the cardiac histological alteration caused by prenatal exposure to EGCG. The results suggest that ingestion of EGCG during pregnancy may be a risk factor for cardiac development in offspring.
Collapse
Affiliation(s)
- Shenli Zhang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, PR China
| | - Kunlin Ou
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, PR China
| | - Jie Huang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, PR China
| | - Lu Fang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, PR China
| | - Chonggang Wang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, PR China
| | - Qin Wang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, PR China.
| |
Collapse
|
17
|
Childers RC, Trask AJ, Liu J, Lucchesi PA, Gooch KJ. Paired Pressure-Volume Loop Analysis and Biaxial Mechanical Testing Characterize Differences in Left Ventricular Tissue Stiffness of Volume Overload and Angiotensin-Induced Pressure Overload Hearts. J Biomech Eng 2021; 143:081003. [PMID: 33729495 PMCID: PMC10782875 DOI: 10.1115/1.4050541] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 01/29/2021] [Indexed: 12/18/2022]
Abstract
Pressure overload (PO) and volume overload (VO) of the heart result in distinctive changes to geometry, due to compensatory structural remodeling. This remodeling potentially leads to changes in tissue mechanical properties. Understanding such changes is important, as tissue modulus has an impact on cardiac performance, disease progression, and influences on cell phenotype. Pressure-volume (PV) loop analysis, a clinically relevant method for measuring left ventricular (LV) chamber stiffness, was performed in vivo on control rat hearts and rats subjected to either chronic PO through Angiotensin-II infusion (4-weeks) or VO (8-weeks). Immediately following PV loops, biaxial testing was performed on LV free wall tissue to directly measure tissue mechanical properties. The β coefficient, an index of chamber stiffness calculated from the PV loop analysis, increased 98% in PO (n = 4) and decreased 38% in VO (n = 5) compared to control (n = 6). Material constants of LV walls obtained from ex vivo biaxial testing (n = 9-10) were not changed in Angiotensin-II induced PO and decreased by about half in VO compared to control (47% in the circumferential and 57% the longitudinal direction). PV loop analysis showed the expected increase in chamber stiffness of PO and expected decrease in chamber stiffness of VO. Biaxial testing showed a decreased modulus of the myocardium of the VO model, but no changes in the PO model, this suggests the increased chamber stiffness in PO, as shown in the PV loop analysis, may be secondary to changes in tissue mass and/or geometry but not an increase in passive tissue mechanical properties.
Collapse
Affiliation(s)
- Rachel C. Childers
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210
| | - Aaron J. Trask
- Center for Cardiovascular Research and The Heart Center, Department of Pediatrics, Nationwide Children's Hospital, The Ohio State University, Columbus, OH 43205
| | - Jun Liu
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210
| | - Pamela A. Lucchesi
- Departments of Pharmacology and Physiology, New York Medical College, Valhalla, NY 10595
| | - Keith J. Gooch
- Institute Frick Center for Heart Failure, Department of Biomedical Engineering, Davis Heart Lung Research, The Ohio State University Fontana Labs, 140 W 19th Avenue, Columbus, OH 43210
| |
Collapse
|
18
|
Childers RC, Lucchesi PA, Gooch KJ. Decreased Substrate Stiffness Promotes a Hypofibrotic Phenotype in Cardiac Fibroblasts. Int J Mol Sci 2021; 22:ijms22126231. [PMID: 34207723 PMCID: PMC8230133 DOI: 10.3390/ijms22126231] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/20/2021] [Accepted: 05/26/2021] [Indexed: 11/16/2022] Open
Abstract
A hypofibrotic phenotype has been observed in cardiac fibroblasts (CFs) isolated from a volume overload heart failure model, aortocaval fistula (ACF). This paradoxical phenotype results in decreased ECM synthesis despite increased TGF-β presence. Since ACF results in decreased tissue stiffness relative to control (sham) hearts, this study investigates whether the effects of substrate stiffness could account for the observed hypofibrotic phenotype in CFs isolated from ACF. CFs isolated from ACF and sham hearts were plated on polyacrylamide gels of a range of stiffness (2 kPa to 50 kPa). Markers related to cytoskeletal and fibrotic proteins were measured. Aspects of the hypofibrotic phenotype observed in ACF CFs were recapitulated by sham CFs on soft substrates. For instance, sham CFs on the softest gels compared to ACF CFs on the stiffest gels results in similar CTGF (0.80 vs. 0.76) and transgelin (0.44 vs. 0.57) mRNA expression. The changes due to stiffness may be explained by the observed decreased nuclear translocation of transcriptional regulators, MRTF-A and YAP. ACF CFs appear to have a mechanical memory of a softer environment, supported by a hypofibrotic phenotype overall compared to sham with less YAP detected in the nucleus, and less CTGF and transgelin on all stiffnesses.
Collapse
Affiliation(s)
- Rachel C. Childers
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA;
| | - Pamela A. Lucchesi
- Department of Pharmacology, New York Medical College, Valhalla, NY 10595, USA
- Correspondence: (P.A.L.); (K.J.G.)
| | - Keith J. Gooch
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA;
- Correspondence: (P.A.L.); (K.J.G.)
| |
Collapse
|
19
|
Lai J, Chen C. The Role of Epoxyeicosatrienoic Acids in Cardiac Remodeling. Front Physiol 2021; 12:642470. [PMID: 33716791 PMCID: PMC7943617 DOI: 10.3389/fphys.2021.642470] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 01/25/2021] [Indexed: 12/12/2022] Open
Abstract
Epoxyeicosatrienoic acids (EETs) are metabolites of arachidonic acid by cytochrome P450 (CYP) epoxygenases, which include four regioisomers: 5,6-EET, 8,9-EET, 11,12-EET, and 14,15-EET. Each of them possesses beneficial effects against inflammation, fibrosis, and apoptosis, which could combat cardiovascular diseases. Numerous studies have demonstrated that elevation of EETs by overexpression of CYP2J2, inhibition of sEH, or treatment with EET analogs showed protective effects in various cardiovascular diseases, including hypertension, myocardial infarction, and heart failure. As is known to all, cardiac remodeling is the major pathogenesis of cardiovascular diseases. This review will begin with the introduction of EETs and their protective effects in cardiovascular diseases. In the following, the roles of EETs in cardiac remodeling, with a particular emphasis on myocardial hypertrophy, apoptosis, fibrosis, inflammation, and angiogenesis, will be summarized. Finally, it is suggested that upregulation of EETs is a potential therapeutic strategy for cardiovascular diseases. The EET-related drug development against cardiac remodeling is also discussed, including the overexpression of CYP2J2, inhibition of sEH, and the analogs of EET.
Collapse
Affiliation(s)
- Jinsheng Lai
- Division of Cardiology, Tongji Hospital, Tongji Medical College and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan, China
| | - Chen Chen
- Division of Cardiology, Tongji Hospital, Tongji Medical College and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
20
|
Progression of left ventricular diastolic function in the neonate and early childhood from transmitral color M-mode filling analysis. Pediatr Res 2021; 89:987-995. [PMID: 32570271 DOI: 10.1038/s41390-020-1011-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 05/22/2020] [Accepted: 06/03/2020] [Indexed: 11/08/2022]
Abstract
BACKGROUND We implemented sophisticated color M-mode analysis to assess age-dependent progression of left ventricular (LV) diastolic function. METHODS Normal infants were prospectively enrolled for serial echocardiograms at 1 week, 1 month, 6 months, 1 year, and 2 years. From color M-mode scans, propagation velocity (VP), strength of filling (VS), and intraventricular pressure difference (IVPD) in 3 segments along apex-to-mitral valve scan line were measured. RESULTS Age-wise comparisons of diastolic filling from 121 echocardiograms in 31 infants showed VP (cm/s), VS (cm2/s), and E-wave IVPD (mmHg) at 1 week to be 66.2 ± 11.9, 75.3 ± 19.9, and 1.5 ± 0.4, respectively, while VP, VS, and E-wave IVPD at 1 month were 80.3 ± 14.4, 101.2 ± 28.3, and 2.42 ± 1.1, respectively. There were significant differences in VP and segmental IVPD between first week and first month (p < 0.005) and IVPD between the age groups (p < 0.001). CONCLUSIONS Comprehensive analysis of transmitral color M-mode data is feasible in infants, enabling calculation of pressure drop between the LV base and apex and strength of propagation from two distinct slopes. Profound changes very early followed by relatively constant filling mechanics in later infancy indicate significant LV maturation occurring during the first month of life. IMPACT We implemented sophisticated analytic methods for color M-mode echocardiography in infants to assess age- and dimension-dependent changes in left ventricular diastolic function. Comprehensive characterization of transmitral color M-mode flow was feasible, enabling calculation of pressure drop between left ventricular base and apex and strength of propagation. Left ventricular diastolic filling function has predictable maturational progression, with significant differences in the intraventricular pressure between infants from birth to 2 years. This study forms the basis for future studies to examine alteration of early diastolic filling in congenital heart disease.
Collapse
|
21
|
Role of FAK signaling in chagasic cardiac hypertrophy. Braz J Infect Dis 2020; 24:386-397. [PMID: 32931757 PMCID: PMC9392126 DOI: 10.1016/j.bjid.2020.08.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 08/03/2020] [Accepted: 08/16/2020] [Indexed: 12/27/2022] Open
Abstract
Cardiac hypertrophy and dysfunction are a significant complication of chronic Chagas disease, with heart failure, stroke, and sudden death related to disease progression. Thus, understanding the signaling pathways involved in the chagasic cardiac hypertrophy may provide potential targets for pharmacological therapy. Herein, we investigated the implication of focal adhesion kinase (FAK) signaling pathway in triggering hypertrophic phenotype during acute and chronic T. cruzi infection. C57BL/6 mice infected with T. cruzi (Brazil strain) were evaluated for electrocardiographic (ECG) changes, plasma levels of endothelin-1 (ET-1) and activation of signaling pathways involved in cardiac hypertrophy, including FAK and ERK1/2, as well as expression of hypertrophy marker and components of the extracellular matrix in the different stages of T. cruzi infection (60-210 dpi). Heart dysfunction, evidenced by prolonged PR interval and decrease in heart rates in ECG tracing, was associated with high plasma ET-1 level, extracellular matrix remodeling and FAK signaling activation. Upregulation of both FAK tyrosine 397 (FAK-Y397) and serine 910 (FAK-S910) residues phosphorylation as well as ERK1/2 activation, lead to an enhancement of atrial natriuretic peptide gene expression in chronic infection. Our findings highlight FAK-ERK1/2 signaling as a regulator of cardiac hypertrophy in Trypanosoma cruzi infection. Both mechanical stress, induced by cardiac extracellular matrix (ECM) augment and cardiac overload, and ET-1 stimuli orchestrated FAK signaling activation with subsequent activation of the fetal cardiac gene program in the chronic phase of infection, highlighting FAK as an attractive target for Chagas disease therapy.
Collapse
|
22
|
Brayson D, Holohan S, Bardswell SC, Arno M, Lu H, Jensen HK, Tran PK, Barallobre‐Barreiro J, Mayr M, dos Remedios CG, Tsang VT, Frigiola A, Kentish JC. Right Ventricle Has Normal Myofilament Function But Shows Perturbations in the Expression of Extracellular Matrix Genes in Patients With Tetralogy of Fallot Undergoing Pulmonary Valve Replacement. J Am Heart Assoc 2020; 9:e015342. [PMID: 32805183 PMCID: PMC7660801 DOI: 10.1161/jaha.119.015342] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 05/28/2020] [Indexed: 02/06/2023]
Abstract
Background Patients with repair of tetralogy of Fallot (rToF) who are approaching adulthood often exhibit pulmonary valve regurgitation, leading to right ventricle (RV) dilatation and dysfunction. The regurgitation can be corrected by pulmonary valve replacement (PVR), but the optimal surgical timing remains under debate, mainly because of the poorly understood nature of RV remodeling in patients with rToF. The goal of this study was to probe for pathologic molecular, cellular, and tissue changes in the myocardium of patients with rToF at the time of PVR. Methods and Results We measured contractile function of permeabilized myocytes, collagen content of tissue samples, and the expression of mRNA and selected proteins in RV tissue samples from patients with rToF undergoing PVR for severe pulmonary valve regurgitation. The data were compared with nondiseased RV tissue from unused donor hearts. Contractile performance and passive stiffness of the myofilaments in permeabilized myocytes were similar in rToF-PVR and RV donor samples, as was collagen content and cross-linking. The patients with rToF undergoing PVR had enhanced mRNA expression of genes associated with connective tissue diseases and tissue remodeling, including the small leucine-rich proteoglycans ASPN (asporin), LUM (lumican), and OGN (osteoglycin), although their protein levels were not significantly increased. Conclusions RV myofilaments from patients with rToF undergoing PVR showed no functional impairment, but the changes in extracellular matrix gene expression may indicate the early stages of remodeling. Our study found no evidence of major damage at the cellular and tissue levels in the RV of patients with rToF who underwent PVR according to current clinical criteria.
Collapse
Affiliation(s)
- Daniel Brayson
- School of Cardiovascular Medicine and SciencesKing's College London BHF Centre for Research ExcellenceLondonUnited Kingdom
| | - So‐Jin Holohan
- School of Cardiovascular Medicine and SciencesKing's College London BHF Centre for Research ExcellenceLondonUnited Kingdom
| | - Sonya C. Bardswell
- School of Cardiovascular Medicine and SciencesKing's College London BHF Centre for Research ExcellenceLondonUnited Kingdom
| | - Matthew Arno
- Genomics CentreFaculty of Life Sciences and MedicineKing’s College LondonLondonUnited Kingdom
| | - Han Lu
- Genomics CentreFaculty of Life Sciences and MedicineKing’s College LondonLondonUnited Kingdom
| | | | | | - Javier Barallobre‐Barreiro
- School of Cardiovascular Medicine and SciencesKing's College London BHF Centre for Research ExcellenceLondonUnited Kingdom
| | - Manuel Mayr
- School of Cardiovascular Medicine and SciencesKing's College London BHF Centre for Research ExcellenceLondonUnited Kingdom
| | | | | | - Alessandra Frigiola
- Great Ormond Street HospitalLondonUnited Kingdom
- Guys and St Thomas’ NHS Foundation TrustSt Thomas’ HospitalLondonUnited Kingdom
- School of Biomedical Engineering and Imaging SciencesKings CollegeLondonUnited Kingdom
| | - Jonathan C. Kentish
- School of Cardiovascular Medicine and SciencesKing's College London BHF Centre for Research ExcellenceLondonUnited Kingdom
| |
Collapse
|
23
|
Bartko PE, Arfsten H, Heitzinger G, Pavo N, Winter MP, Toma A, Strunk G, Hengstenberg C, Hülsmann M, Goliasch G. Natural history of bivalvular functional regurgitation. Eur Heart J Cardiovasc Imaging 2020; 20:565-573. [PMID: 30508183 DOI: 10.1093/ehjci/jey178] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 10/29/2018] [Indexed: 01/07/2023] Open
Abstract
AIMS Bivalvular functional regurgitation (BVFR) defined as concomitant mitral and tricuspid insufficiency has not been described or systematically assessed before. Therefore, this study sought to define incidence, impact and natural history of BVFR in heart failure with reduced ejection fraction (HFrEF) to provide the foundation for risk assessment and directions for potential treatment strategies. METHODS AND RESULTS We enrolled 1021 consecutive patients with HFrEF under guideline-directed medical therapy and performed comprehensive echocardiographic and neurohumoral profiling. All-cause mortality during a 5 years of follow-up served as the primary endpoint. Thirty percent of patients suffered from moderate or severe BVFR. Long-term mortality increased with the presence and severity of functional regurgitation (FR) with severe BVFR representing the highest risk-subset (P < 0.001). Severe BVFR patients were more symptomatic and displayed an adverse remodelling and neurohumoral activation pattern (all P < 0.05). Severe BVFR was associated with excess mortality independently of clinical [adjusted hazard ratio (HR) 1.52, 95% confidence interval (CI) 1.39-1.84; P < 0.001] and echocardiographic (adjusted HR 1.31, 95% CI 1.11-1.54; P = 0.001) confounders, guideline-directed medical therapy (adjusted HR 1.55, 95% CI 1.35-1.79; P < 0.001) and neurohumoral activation (adjusted HR 1.31, 95% CI 1.07-1.59; P = 0.009). Moderate BVFR (n = 99) comprised equal baseline characteristics and similar risk as isolated severe FR (HR 0.95, 95% CI 0.69-1.30; P = 0.73). CONCLUSION This long-term outcome study shows the multi-faceted nature of FR and defines BVFR as an important clinical entity associated with impaired functional class, adverse cardiac remodelling, and excess risk of mortality. Moderate BVFR conveys similar risk as isolated severe FR reflecting the deleterious impact of the global regurgitant load on the failing heart and the need of an integrated understanding for risk-assessment.
Collapse
Affiliation(s)
- Philipp E Bartko
- Department of Internal Medicine II, Medical University of Vienna, Waehringer Guertel 18-20, A Vienna, Austria
| | - Henrike Arfsten
- Department of Internal Medicine II, Medical University of Vienna, Waehringer Guertel 18-20, A Vienna, Austria
| | - Gregor Heitzinger
- Department of Internal Medicine II, Medical University of Vienna, Waehringer Guertel 18-20, A Vienna, Austria
| | - Noemi Pavo
- Department of Internal Medicine II, Medical University of Vienna, Waehringer Guertel 18-20, A Vienna, Austria
| | - Max-Paul Winter
- Department of Internal Medicine II, Medical University of Vienna, Waehringer Guertel 18-20, A Vienna, Austria
| | - Aurel Toma
- Department of Internal Medicine II, Medical University of Vienna, Waehringer Guertel 18-20, A Vienna, Austria
| | - Guido Strunk
- FH Campus Vienna and Complexity Research, Favoritenstraße 226, A Vienna, Austria
| | - Christian Hengstenberg
- Department of Internal Medicine II, Medical University of Vienna, Waehringer Guertel 18-20, A Vienna, Austria
| | - Martin Hülsmann
- Department of Internal Medicine II, Medical University of Vienna, Waehringer Guertel 18-20, A Vienna, Austria
| | - Georg Goliasch
- Department of Internal Medicine II, Medical University of Vienna, Waehringer Guertel 18-20, A Vienna, Austria
| |
Collapse
|
24
|
Tayal U, Wage R, Newsome S, Manivarmane R, Izgi C, Muthumala A, Dungu JN, Assomull R, Hatipoglu S, Halliday BP, Lota AS, Ware JS, Gregson J, Frenneaux M, Cook SA, Pennell DJ, Scott AD, Cleland JG, Prasad SK. Predictors of left ventricular remodelling in patients with dilated cardiomyopathy – a cardiovascular magnetic resonance study. Eur J Heart Fail 2020; 22:1160-1170. [DOI: 10.1002/ejhf.1734] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 07/05/2019] [Accepted: 11/29/2019] [Indexed: 01/28/2023] Open
Affiliation(s)
- Upasana Tayal
- National Heart Lung Institute Imperial College London UK
- Cardiovascular Magnetic Resonance Unit Royal Brompton Hospital London UK
| | - Ricardo Wage
- Cardiovascular Magnetic Resonance Unit Royal Brompton Hospital London UK
| | - Simon Newsome
- Department of Medical Statistics London School of Hygiene and Tropical Medicine London UK
| | | | - Cemil Izgi
- Cardiovascular Magnetic Resonance Unit Royal Brompton Hospital London UK
| | - Amal Muthumala
- North Middlesex University Hospital and St Bartholomew's Hospital London UK
| | | | | | - Suzan Hatipoglu
- National Heart Lung Institute Imperial College London UK
- Cardiovascular Magnetic Resonance Unit Royal Brompton Hospital London UK
| | - Brian P. Halliday
- National Heart Lung Institute Imperial College London UK
- Cardiovascular Magnetic Resonance Unit Royal Brompton Hospital London UK
| | - Amrit S. Lota
- National Heart Lung Institute Imperial College London UK
- Cardiovascular Magnetic Resonance Unit Royal Brompton Hospital London UK
| | - James S. Ware
- National Heart Lung Institute Imperial College London UK
- Cardiovascular Magnetic Resonance Unit Royal Brompton Hospital London UK
- MRC London Institute of Medical Sciences London UK
| | - John Gregson
- Department of Medical Statistics London School of Hygiene and Tropical Medicine London UK
| | - Michael Frenneaux
- National Heart Lung Institute Imperial College London UK
- University of East Anglia Norwich UK
| | | | - Dudley J. Pennell
- National Heart Lung Institute Imperial College London UK
- Cardiovascular Magnetic Resonance Unit Royal Brompton Hospital London UK
| | - Andrew D. Scott
- National Heart Lung Institute Imperial College London UK
- Cardiovascular Magnetic Resonance Unit Royal Brompton Hospital London UK
| | - John G.F. Cleland
- National Heart Lung Institute Imperial College London UK
- Cardiovascular Magnetic Resonance Unit Royal Brompton Hospital London UK
| | - Sanjay K. Prasad
- National Heart Lung Institute Imperial College London UK
- Cardiovascular Magnetic Resonance Unit Royal Brompton Hospital London UK
| |
Collapse
|
25
|
McCutcheon K, Dickens C, van Pelt J, Dix-Peek T, Grinter S, McCutcheon L, Patel A, Hale M, Tsabedze N, Vachiat A, Zachariah D, Duarte R, Janssens S, Manga P. Dynamic Changes in the Molecular Signature of Adverse Left Ventricular Remodeling in Patients With Compensated and Decompensated Chronic Primary Mitral Regurgitation. Circ Heart Fail 2019; 12:e005974. [PMID: 31510777 DOI: 10.1161/circheartfailure.119.005974] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
BACKGROUND There is no proven medical therapy that attenuates adverse left ventricular remodeling in patients with chronic primary mitral regurgitation (CPMR). Identification of molecular pathways important in the progression of left ventricular remodeling in patients with CPMR may lead to development of new therapeutic strategies. METHODS AND RESULTS We performed baseline echocardiographic, cardiac catheterization, and serum NT-pro-BNP analysis in patients with severe CPMR awaiting mitral valve surgery and stratified the study population into compensated or decompensated CPMR. We obtained left ventricular endomyocardial biopsies (n=12) for mRNA expression analysis, and compared baseline transcript levels of 109 genes important in volume-overload left ventricular remodeling with levels in normal hearts (n=5) and between patients with compensated (n=6) versus decompensated (n=6) CPMR. Patients were then randomized to treatment with and without carvedilol and followed until the time of surgery (mean follow-up 8.3 months) when repeat endomyocardial biopsies were obtained to correlate transcriptional dynamics with indices of adverse remodeling. CPMR was associated with increased NPPA expression levels (21.6-fold, P=0.004), decreased transcripts of genes important in cell survival, and enrichment of extracellular matrix genes. Decompensated CPMR was associated with downregulation of SERCA2 (0.77-fold, P=0.009) and mitochondrial gene expression levels and upregulation of genes related to inflammation, the extracellular matrix, and apoptosis, which were refractory to carvedilol therapy. CONCLUSIONS Transition to decompensated CPMR is associated with calcium dysregulation, increased expression of inflammatory, extracellular matrix and apoptotic genes, and downregulation of genes important in bioenergetics. These changes are not attenuated by carvedilol therapy and highlight the need for development of specific combinatorial therapies, targeting myocardial inflammation and apoptosis, together with urgent surgical or percutaneous valve interventions.
Collapse
Affiliation(s)
- Keir McCutcheon
- Division of Cardiology, Department of Internal Medicine (K.M., S.G., L.M., N.T., A.V., D.Z., P.M.), Charlotte Maxeke Johannesburg Academic Hospital & University of the Witwatersrand, Johannesburg, South Africa.,Department of Cardiovascular Diseases, University Hospitals Leuven, Belgium (K.M., S.J.)
| | - Caroline Dickens
- Molecular Biology Laboratory, Department of Internal Medicine (C.D., T.D.-P., R.D.), University of the Witwatersrand, Johannesburg, South Africa
| | - Jos van Pelt
- Department of Clinical Digestive Oncology, Faculty of Medicine, Katholieke Universiteit, Leuven and Leuven Cancer Institute, Leuven, Belgium (J.v.P.)
| | - Therese Dix-Peek
- Molecular Biology Laboratory, Department of Internal Medicine (C.D., T.D.-P., R.D.), University of the Witwatersrand, Johannesburg, South Africa
| | - Sacha Grinter
- Division of Cardiology, Department of Internal Medicine (K.M., S.G., L.M., N.T., A.V., D.Z., P.M.), Charlotte Maxeke Johannesburg Academic Hospital & University of the Witwatersrand, Johannesburg, South Africa
| | - Lindsay McCutcheon
- Division of Cardiology, Department of Internal Medicine (K.M., S.G., L.M., N.T., A.V., D.Z., P.M.), Charlotte Maxeke Johannesburg Academic Hospital & University of the Witwatersrand, Johannesburg, South Africa
| | - Atulkumar Patel
- Department of Cardiothoracic Surgery (A.P.), Charlotte Maxeke Johannesburg Academic Hospital & University of the Witwatersrand, Johannesburg, South Africa
| | - Martin Hale
- Department of Anatomical Pathology (M.H.), University of the Witwatersrand, Johannesburg, South Africa
| | - Nqoba Tsabedze
- Division of Cardiology, Department of Internal Medicine (K.M., S.G., L.M., N.T., A.V., D.Z., P.M.), Charlotte Maxeke Johannesburg Academic Hospital & University of the Witwatersrand, Johannesburg, South Africa
| | - Ahmed Vachiat
- Division of Cardiology, Department of Internal Medicine (K.M., S.G., L.M., N.T., A.V., D.Z., P.M.), Charlotte Maxeke Johannesburg Academic Hospital & University of the Witwatersrand, Johannesburg, South Africa
| | - Don Zachariah
- Division of Cardiology, Department of Internal Medicine (K.M., S.G., L.M., N.T., A.V., D.Z., P.M.), Charlotte Maxeke Johannesburg Academic Hospital & University of the Witwatersrand, Johannesburg, South Africa
| | - Raquel Duarte
- Molecular Biology Laboratory, Department of Internal Medicine (C.D., T.D.-P., R.D.), University of the Witwatersrand, Johannesburg, South Africa
| | - Stefan Janssens
- Department of Cardiovascular Diseases, University Hospitals Leuven, Belgium (K.M., S.J.).,Department of Cardiovascular Sciences, Katholieke Universiteit, Leuven, Belgium (S.J.)
| | - Pravin Manga
- Division of Cardiology, Department of Internal Medicine (K.M., S.G., L.M., N.T., A.V., D.Z., P.M.), Charlotte Maxeke Johannesburg Academic Hospital & University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
26
|
Dubes V, Benoist D, Roubertie F, Gilbert SH, Constantin M, Charron S, Elbes D, Vieillot D, Quesson B, Cochet H, Haïssaguerre M, Rooryck C, Bordachar P, Thambo JB, Bernus O. Arrhythmogenic Remodeling of the Left Ventricle in a Porcine Model of Repaired Tetralogy of Fallot. Circ Arrhythm Electrophysiol 2019; 11:e006059. [PMID: 30354410 PMCID: PMC6553519 DOI: 10.1161/circep.117.006059] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Supplemental Digital Content is available in the text. Background Ventricular arrhythmias are frequent in patients with repaired tetralogy of Fallot (rTOF), but their origin and underlying mechanisms remain unclear. In this study, the involvement of left ventricular (LV) electrical and structural remodeling was assessed in an animal model mimicking rTOF sequelae. Methods Piglets underwent a tetralogy of Fallot repair–like surgery (n=6) or were sham operated (Sham, n=5). Twenty-three weeks post-surgery, cardiac function was assessed in vivo by magnetic resonance imaging. Electrophysiological properties were characterized by optical mapping. LV fibrosis and connexin-43 localization were assessed on histological sections and protein expression assessed by Western Blot. Results Right ventricular dysfunction was evident, whereas LV function remained unaltered in rTOF pigs. Optical mapping showed longer action potential duration on the rTOF LV epicardium and endocardium. Epicardial conduction velocity was significantly reduced in the longitudinal direction in rTOF LVs but not in the transverse direction compared with Sham. An elevated collagen content was found in LV basal and apical sections from rTOF pigs. Moreover, a trend for connexin-43 lateralization with no change in protein expression was found in the LV of rTOFs. Finally, rTOF LVs had a lower threshold for arrhythmia induction using incremental pacing protocols. Conclusions We found an arrhythmogenic substrate with prolonged heterogeneous action potential duration and reduced conduction velocity in the LV of rTOF pigs. This remodeling precedes LV dysfunction and is likely to contribute to ventricular arrhythmias and sudden cardiac death in patients with rTOF.
Collapse
Affiliation(s)
- Virginie Dubes
- IHU LIRYC, L'Institut de Rythmologie et Modélisation Cardiaque, Fondation Bordeaux Université, Pessac, France (V.D., D.B., F.R., S.H.G., M.C., S.C., D.E., B.Q., H.C., M.H., C.R., P.B., J.-B.T., O.B.).,Inserm U1045, Centre de Recherche Cardio-Thoracique de Bordeaux, Université de Bordeaux, France. (V.D., D.B., F.R., S.H.G., M.C., S.C., D.E., B.Q., H.C., M.H., P.B., J.-B.T., O.B.)
| | - David Benoist
- IHU LIRYC, L'Institut de Rythmologie et Modélisation Cardiaque, Fondation Bordeaux Université, Pessac, France (V.D., D.B., F.R., S.H.G., M.C., S.C., D.E., B.Q., H.C., M.H., C.R., P.B., J.-B.T., O.B.).,Inserm U1045, Centre de Recherche Cardio-Thoracique de Bordeaux, Université de Bordeaux, France. (V.D., D.B., F.R., S.H.G., M.C., S.C., D.E., B.Q., H.C., M.H., P.B., J.-B.T., O.B.)
| | - François Roubertie
- IHU LIRYC, L'Institut de Rythmologie et Modélisation Cardiaque, Fondation Bordeaux Université, Pessac, France (V.D., D.B., F.R., S.H.G., M.C., S.C., D.E., B.Q., H.C., M.H., C.R., P.B., J.-B.T., O.B.).,Inserm U1045, Centre de Recherche Cardio-Thoracique de Bordeaux, Université de Bordeaux, France. (V.D., D.B., F.R., S.H.G., M.C., S.C., D.E., B.Q., H.C., M.H., P.B., J.-B.T., O.B.).,Centre Hospitalier Universitaire de Bordeaux, Hôpital Cardiologique du Haut-Lévêque, Pessac, France (F.R., H.C., M.H., C.R., P.B., J.-B.T.)
| | - Stephen H Gilbert
- IHU LIRYC, L'Institut de Rythmologie et Modélisation Cardiaque, Fondation Bordeaux Université, Pessac, France (V.D., D.B., F.R., S.H.G., M.C., S.C., D.E., B.Q., H.C., M.H., C.R., P.B., J.-B.T., O.B.).,Inserm U1045, Centre de Recherche Cardio-Thoracique de Bordeaux, Université de Bordeaux, France. (V.D., D.B., F.R., S.H.G., M.C., S.C., D.E., B.Q., H.C., M.H., P.B., J.-B.T., O.B.).,Max Delbröck Center for Molecular Medicine, Berlin, Germany (S.H.G.)
| | - Marion Constantin
- IHU LIRYC, L'Institut de Rythmologie et Modélisation Cardiaque, Fondation Bordeaux Université, Pessac, France (V.D., D.B., F.R., S.H.G., M.C., S.C., D.E., B.Q., H.C., M.H., C.R., P.B., J.-B.T., O.B.).,Inserm U1045, Centre de Recherche Cardio-Thoracique de Bordeaux, Université de Bordeaux, France. (V.D., D.B., F.R., S.H.G., M.C., S.C., D.E., B.Q., H.C., M.H., P.B., J.-B.T., O.B.)
| | - Sabine Charron
- IHU LIRYC, L'Institut de Rythmologie et Modélisation Cardiaque, Fondation Bordeaux Université, Pessac, France (V.D., D.B., F.R., S.H.G., M.C., S.C., D.E., B.Q., H.C., M.H., C.R., P.B., J.-B.T., O.B.).,Inserm U1045, Centre de Recherche Cardio-Thoracique de Bordeaux, Université de Bordeaux, France. (V.D., D.B., F.R., S.H.G., M.C., S.C., D.E., B.Q., H.C., M.H., P.B., J.-B.T., O.B.)
| | - Delphine Elbes
- IHU LIRYC, L'Institut de Rythmologie et Modélisation Cardiaque, Fondation Bordeaux Université, Pessac, France (V.D., D.B., F.R., S.H.G., M.C., S.C., D.E., B.Q., H.C., M.H., C.R., P.B., J.-B.T., O.B.).,Inserm U1045, Centre de Recherche Cardio-Thoracique de Bordeaux, Université de Bordeaux, France. (V.D., D.B., F.R., S.H.G., M.C., S.C., D.E., B.Q., H.C., M.H., P.B., J.-B.T., O.B.).,Institute of Biomedical Engineering, University of Oxford, United Kingdom (D.E.)
| | - Delphine Vieillot
- Plateforme Technologique d'Innovation Biomédicale, Université de Bordeaux, France. (D.V.)
| | - Bruno Quesson
- IHU LIRYC, L'Institut de Rythmologie et Modélisation Cardiaque, Fondation Bordeaux Université, Pessac, France (V.D., D.B., F.R., S.H.G., M.C., S.C., D.E., B.Q., H.C., M.H., C.R., P.B., J.-B.T., O.B.).,Inserm U1045, Centre de Recherche Cardio-Thoracique de Bordeaux, Université de Bordeaux, France. (V.D., D.B., F.R., S.H.G., M.C., S.C., D.E., B.Q., H.C., M.H., P.B., J.-B.T., O.B.)
| | - Hubert Cochet
- IHU LIRYC, L'Institut de Rythmologie et Modélisation Cardiaque, Fondation Bordeaux Université, Pessac, France (V.D., D.B., F.R., S.H.G., M.C., S.C., D.E., B.Q., H.C., M.H., C.R., P.B., J.-B.T., O.B.).,Inserm U1045, Centre de Recherche Cardio-Thoracique de Bordeaux, Université de Bordeaux, France. (V.D., D.B., F.R., S.H.G., M.C., S.C., D.E., B.Q., H.C., M.H., P.B., J.-B.T., O.B.).,Centre Hospitalier Universitaire de Bordeaux, Hôpital Cardiologique du Haut-Lévêque, Pessac, France (F.R., H.C., M.H., C.R., P.B., J.-B.T.)
| | - Michel Haïssaguerre
- IHU LIRYC, L'Institut de Rythmologie et Modélisation Cardiaque, Fondation Bordeaux Université, Pessac, France (V.D., D.B., F.R., S.H.G., M.C., S.C., D.E., B.Q., H.C., M.H., C.R., P.B., J.-B.T., O.B.).,Inserm U1045, Centre de Recherche Cardio-Thoracique de Bordeaux, Université de Bordeaux, France. (V.D., D.B., F.R., S.H.G., M.C., S.C., D.E., B.Q., H.C., M.H., P.B., J.-B.T., O.B.).,Centre Hospitalier Universitaire de Bordeaux, Hôpital Cardiologique du Haut-Lévêque, Pessac, France (F.R., H.C., M.H., C.R., P.B., J.-B.T.)
| | - Caroline Rooryck
- IHU LIRYC, L'Institut de Rythmologie et Modélisation Cardiaque, Fondation Bordeaux Université, Pessac, France (V.D., D.B., F.R., S.H.G., M.C., S.C., D.E., B.Q., H.C., M.H., C.R., P.B., J.-B.T., O.B.).,Inserm U1211, Maladies Rares: Génétique et Métabolisme, Université de Bordeaux, France. (C.R.).,Centre Hospitalier Universitaire de Bordeaux, Hôpital Cardiologique du Haut-Lévêque, Pessac, France (F.R., H.C., M.H., C.R., P.B., J.-B.T.)
| | - Pierre Bordachar
- IHU LIRYC, L'Institut de Rythmologie et Modélisation Cardiaque, Fondation Bordeaux Université, Pessac, France (V.D., D.B., F.R., S.H.G., M.C., S.C., D.E., B.Q., H.C., M.H., C.R., P.B., J.-B.T., O.B.).,Inserm U1045, Centre de Recherche Cardio-Thoracique de Bordeaux, Université de Bordeaux, France. (V.D., D.B., F.R., S.H.G., M.C., S.C., D.E., B.Q., H.C., M.H., P.B., J.-B.T., O.B.).,Centre Hospitalier Universitaire de Bordeaux, Hôpital Cardiologique du Haut-Lévêque, Pessac, France (F.R., H.C., M.H., C.R., P.B., J.-B.T.)
| | - Jean-Benoit Thambo
- IHU LIRYC, L'Institut de Rythmologie et Modélisation Cardiaque, Fondation Bordeaux Université, Pessac, France (V.D., D.B., F.R., S.H.G., M.C., S.C., D.E., B.Q., H.C., M.H., C.R., P.B., J.-B.T., O.B.).,Inserm U1045, Centre de Recherche Cardio-Thoracique de Bordeaux, Université de Bordeaux, France. (V.D., D.B., F.R., S.H.G., M.C., S.C., D.E., B.Q., H.C., M.H., P.B., J.-B.T., O.B.).,Centre Hospitalier Universitaire de Bordeaux, Hôpital Cardiologique du Haut-Lévêque, Pessac, France (F.R., H.C., M.H., C.R., P.B., J.-B.T.)
| | - Olivier Bernus
- IHU LIRYC, L'Institut de Rythmologie et Modélisation Cardiaque, Fondation Bordeaux Université, Pessac, France (V.D., D.B., F.R., S.H.G., M.C., S.C., D.E., B.Q., H.C., M.H., C.R., P.B., J.-B.T., O.B.).,Inserm U1045, Centre de Recherche Cardio-Thoracique de Bordeaux, Université de Bordeaux, France. (V.D., D.B., F.R., S.H.G., M.C., S.C., D.E., B.Q., H.C., M.H., P.B., J.-B.T., O.B.)
| |
Collapse
|
27
|
The orphan receptor GPRC5B modulates inflammatory and fibrotic pathways in cardiac fibroblasts and mice hearts. Biochem Biophys Res Commun 2019; 514:1198-1203. [DOI: 10.1016/j.bbrc.2019.05.038] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 05/04/2019] [Indexed: 12/26/2022]
|
28
|
Sahli Costabal F, Choy JS, Sack KL, Guccione JM, Kassab GS, Kuhl E. Multiscale characterization of heart failure. Acta Biomater 2019; 86:66-76. [PMID: 30630123 DOI: 10.1016/j.actbio.2018.12.053] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 12/28/2018] [Accepted: 12/31/2018] [Indexed: 12/27/2022]
Abstract
Dilated cardiomyopathy is a progressive irreversible disease associated with contractile dysfunction and heart failure. During dilated cardiomyopathy, elevated diastolic wall strains trigger mechanotransduction pathways that initiate the addition of sarcomeres in series and an overall increase in myocyte length. At the whole organ level, this results in a chronic dilation of the ventricles, an increase in end diastolic and end systolic volumes, and a decrease in ejection fraction. However, how exactly changes in sarcomere number translate into changes in myocyte morphology, and how these cellular changes translate into ventricular dilation remains incompletely understood. Here we combined a chronic animal study, continuum growth modeling, and machine learning to quantify correlations between sarcomere dynamics, myocyte morphology, and ventricular dilation. In an eight-week long volume overload study of six pigs, we found that the average sarcomere number increased by +3.8%/week, from 47 to 62, resulting in a myocyte lengthening of +3.3%/week, from 85 to 108 μm, while the sarcomere length and myocyte width remained unchanged. At the same time, the average end diastolic volume increased by +6.0%/week. Using continuum growth modeling and Bayesian inference, we correlated alterations on the subcellular, cellular, and organ scales and found that the serial sarcomere number explained 88% of myocyte lengthening, which, in turn, explained 54% of cardiac dilation. Our results demonstrate that sarcomere number and myocyte length are closely correlated and constitute the major determinants of dilated heart failure. We anticipate our study to be a starting point for more sophisticated multiscale models of heart failure. Our study suggests that altering sarcomere turnover-and with it myocyte morphology and ventricular dimensions-could be a potential therapeutic target to attenuate or reverse the progression of heart failure. STATEMENT OF SIGNIFICANCE: Heart failure is a significant global health problem that affects more than 25 million people worldwide and increases in prevalence as the population ages. Heart failure has been studied excessively at various scales; yet, there is no compelling concept to connect knowledge from the subcellular, cellular, and organ level across the scales. Here we combined a chronic animal study, continuum growth modeling, and machine learning to quantify correlations between sarcomere dynamics, myocyte morphology, and ventricular dilation. We found that the serial sarcomere number explained 88% of myocyte lengthening, which, in turn, explained 54% of cardiac dilation. Our results show that sarcomere number and myocyte length are closely correlated and constitute the major determinants of dilated heart failure. This suggests that altering the sarcomere turnover-and with it myocyte morphology and ventricular dimensions-could be a potential therapeutic target to attenuate or reverse heart failure.
Collapse
Affiliation(s)
- F Sahli Costabal
- Departments of Mechanical Engineering & Bioengineering, Stanford University, CA, USA
| | - J S Choy
- California Medical Innovations Institute, Inc., San Diego, CA, USA
| | - K L Sack
- Department of Human Biology, University of Cape Town, Cape Town, South Africa
| | - J M Guccione
- Department of Surgery, University of California at San Francisco, San Francisco, CA, USA
| | - G S Kassab
- California Medical Innovations Institute, Inc., San Diego, CA, USA
| | - E Kuhl
- Departments of Mechanical Engineering & Bioengineering, Stanford University, CA, USA.
| |
Collapse
|
29
|
Sugiyama A, Mitsui A, Okada M, Yamawaki H. Cathepsin S degrades arresten and canstatin in infarcted area after myocardial infarction in rats. J Vet Med Sci 2019; 81:522-531. [PMID: 30726795 PMCID: PMC6483919 DOI: 10.1292/jvms.18-0674] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
The basement membrane surrounding cardiomyocytes is mainly composed of α1 and α2 chain of
type IV collagen. Arresten and canstatin are fragments of non-collagenous C-terminal
domain of α1 and α2 chain, respectively. We previously reported that the expression of
canstatin was decreased in infarcted area 2 weeks after myocardial infarction in rats. In
the present study, we investigated the regulatory mechanism for expression of arresten and
canstatin. Myocardial infarction model rats were produced by ligating left anterior
descending artery. Western blotting and immunohistochemical staining were performed to
determine the protein expression and distribution. Arresten and canstatin were highly
expressed in the heart. One day and three days after myocardial infarction, the expression
of arresten and canstatin in infarcted area was lower than that in non-infarcted area. The
expression of cathepsin S, which is known to degrade arresten and canstatin, was increased
in the infarcted area. A knockdown of cathepsin S gene using small interference RNA
suppressed the decline of arresten and canstatin in the infarcted area 3 days after
myocardial infarction. This study for the first time revealed that arresten and canstatin
are immediately degraded by cathepsin S in the infarcted area after myocardial infarction.
These findings present a novel fundamental insight into the pathogenesis of myocardial
infarction through the turnover of basement membrane-derived endogenous factors.
Collapse
Affiliation(s)
- Akira Sugiyama
- Laboratory of Veterinary Pharmacology, School of Veterinary Medicine, Kitasato University, Higashi 23 bancho 35-1, Towada, Aomori 034-8628, Japan
| | - Ayaka Mitsui
- Laboratory of Veterinary Pharmacology, School of Veterinary Medicine, Kitasato University, Higashi 23 bancho 35-1, Towada, Aomori 034-8628, Japan
| | - Muneyoshi Okada
- Laboratory of Veterinary Pharmacology, School of Veterinary Medicine, Kitasato University, Higashi 23 bancho 35-1, Towada, Aomori 034-8628, Japan
| | - Hideyuki Yamawaki
- Laboratory of Veterinary Pharmacology, School of Veterinary Medicine, Kitasato University, Higashi 23 bancho 35-1, Towada, Aomori 034-8628, Japan
| |
Collapse
|
30
|
McCutcheon K, Manga P. Left ventricular remodelling in chronic primary mitral regurgitation: implications for medical therapy. Cardiovasc J Afr 2019; 29:51-65. [PMID: 29582880 PMCID: PMC6002796 DOI: 10.5830/cvja-2017-009] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Accepted: 01/12/2017] [Indexed: 01/07/2023] Open
Abstract
Surgical repair or replacement of the mitral valve is currently the only recommended therapy for severe primary mitral regurgitation. The chronic elevation of wall stress caused by the resulting volume overload leads to structural remodelling of the muscular, vascular and extracellular matrix components of the myocardium. These changes are initially compensatory but in the long term have detrimental effects, which ultimately result in heart failure. Understanding the changes that occur in the myocardium due to volume overload at the molecular and cellular level may lead to medical interventions, which potentially could delay or prevent the adverse left ventricular remodelling associated with primary mitral regurgitation. The pathophysiological changes involved in left ventricular remodelling in response to chronic primary mitral regurgitation and the evidence for potential medical therapy, in particular beta-adrenergic blockers, are the focus of this review.
Collapse
Affiliation(s)
- Keir McCutcheon
- Division of Cardiology, Department of Internal Medicine, Charlotte Maxeke Johannesburg Academic Hospital and University of the Witwatersrand, Johannesburg, South Africa.
| | - Pravin Manga
- Division of Cardiology, Department of Internal Medicine, Charlotte Maxeke Johannesburg Academic Hospital and University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
31
|
van der Velden J, Stienen GJM. Cardiac Disorders and Pathophysiology of Sarcomeric Proteins. Physiol Rev 2019; 99:381-426. [PMID: 30379622 DOI: 10.1152/physrev.00040.2017] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The sarcomeric proteins represent the structural building blocks of heart muscle, which are essential for contraction and relaxation. During recent years, it has become evident that posttranslational modifications of sarcomeric proteins, in particular phosphorylation, tune cardiac pump function at rest and during exercise. This delicate, orchestrated interaction is also influenced by mutations, predominantly in sarcomeric proteins, which cause hypertrophic or dilated cardiomyopathy. In this review, we follow a bottom-up approach starting from a description of the basic components of cardiac muscle at the molecular level up to the various forms of cardiac disorders at the organ level. An overview is given of sarcomere changes in acquired and inherited forms of cardiac disease and the underlying disease mechanisms with particular reference to human tissue. A distinction will be made between the primary defect and maladaptive/adaptive secondary changes. Techniques used to unravel functional consequences of disease-induced protein changes are described, and an overview of current and future treatments targeted at sarcomeric proteins is given. The current evidence presented suggests that sarcomeres not only form the basis of cardiac muscle function but also represent a therapeutic target to combat cardiac disease.
Collapse
Affiliation(s)
- Jolanda van der Velden
- Amsterdam UMC, Vrije Universiteit Amsterdam, Physiology, Amsterdam Cardiovascular Sciences, Amsterdam , The Netherlands ; and Department of Physiology, Kilimanjaro Christian Medical University College, Moshi, Tanzania
| | - Ger J M Stienen
- Amsterdam UMC, Vrije Universiteit Amsterdam, Physiology, Amsterdam Cardiovascular Sciences, Amsterdam , The Netherlands ; and Department of Physiology, Kilimanjaro Christian Medical University College, Moshi, Tanzania
| |
Collapse
|
32
|
Corporan D, Onohara D, Hernandez-Merlo R, Sielicka A, Padala M. Temporal changes in myocardial collagen, matrix metalloproteinases, and their tissue inhibitors in the left ventricular myocardium in experimental chronic mitral regurgitation in rodents. Am J Physiol Heart Circ Physiol 2018; 315:H1269-H1278. [PMID: 30141979 PMCID: PMC6297825 DOI: 10.1152/ajpheart.00099.2018] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 07/27/2018] [Accepted: 07/27/2018] [Indexed: 12/19/2022]
Abstract
Mitral regurgitation (MR) imposes left ventricular volume overload, triggering rapid ventricular dilatation, increased myocardial compliance, and, ultimately, cardiac dysfunction. Breakdown of the extracellular matrix has been hypothesized to drive these rapid changes, partially from an imbalance in the matrix metalloproteinases (MMPs) and their tissue inhibitors [tissue inhibitors of metalloproteinase (TIMPs)]. In the present study, we developed a rat model of severe MR that mimics the human condition and investigated the temporal changes in extracellular matrix-related genes, collagen biosynthesis proteins, and proteolytic enzymes over a 20-wk period. Male Sprague-Dawley rats were anesthetized to a surgical plane with mechanical ventilation, and a thoracotomy was performed to expose the apex. Using transesophageal ultrasound guidance, a needle was inserted into the beating heart to perforate the anterior mitral leaflet and create severe MR. Animals were survived for 20 wk, with some animals terminated at 2, 10, and 20 wk for analysis of left ventricular tissue. A sham group that underwent the same surgery without mitral leaflet perforation and MR were used as controls. At 2 wk post-MR, increased collagen gene expression was measured, but protein levels of collagen did not corroborate this finding. In parallel, MMP-1-to-TIMP-4, MMP-2-to-TIMP-1, and MMP-2-to-TIMP-3 ratios were significantly elevated, indicating a proteolytic milieu in the myocardium, possibly causing collagen degradation. By 20 wk, many of the initial differences seen in the proteolytic ratios were not observed, with an increase in collagen compared with the 2-wk time point. Altogether, this data indicates that an imbalance in the MMP-to-TIMP ratio may occur early and potentially contribute to the early dilatation and compliance observed structurally. NEW & NOTEWORTHY In this rodent model of severe mitral regurgitation that mimics the human condition, eccentric left ventricular dilatation occurred rapidly and persisted over the 20-wk period with parallel changes in myocardial collagen and matrix metalloproteinases that may drive the extracellular matrix breakdown.
Collapse
Affiliation(s)
- Daniella Corporan
- Structural Heart Research and Innovation Laboratory, Carlyle Fraser Heart Center, Emory Hospital Midtown , Atlanta, Georgia
| | - Daisuke Onohara
- Structural Heart Research and Innovation Laboratory, Carlyle Fraser Heart Center, Emory Hospital Midtown , Atlanta, Georgia
| | - Roberto Hernandez-Merlo
- Structural Heart Research and Innovation Laboratory, Carlyle Fraser Heart Center, Emory Hospital Midtown , Atlanta, Georgia
| | - Alicja Sielicka
- Structural Heart Research and Innovation Laboratory, Carlyle Fraser Heart Center, Emory Hospital Midtown , Atlanta, Georgia
| | - Muralidhar Padala
- Structural Heart Research and Innovation Laboratory, Carlyle Fraser Heart Center, Emory Hospital Midtown , Atlanta, Georgia
- Division of Cardiothoracic Surgery, Joseph P. Whitehead Department of Surgery, Emory School of Medicine , Atlanta, Georgia
| |
Collapse
|
33
|
Fan Z, Gao S, Chen Y, Xu B, Yu C, Yue M, Tan X. Integrative analysis of competing endogenous RNA networks reveals the functional lncRNAs in heart failure. J Cell Mol Med 2018; 22:4818-4829. [PMID: 30019841 PMCID: PMC6156393 DOI: 10.1111/jcmm.13739] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2018] [Accepted: 05/20/2018] [Indexed: 02/05/2023] Open
Abstract
Heart failure has become one of the top causes of death worldwide. It is increasing evidence that lncRNAs play important roles in the pathology processes of multiple cardiovascular diseases. Additionally, lncRNAs can function as ceRNAs by sponging miRNAs to affect the expression level of mRNAs, implicating in numerous biological processes. However, the functional roles and regulatory mechanisms of lncRNAs in heart failure are still unclear. In our study, we constructed a heart failure-related lncRNA-mRNA network by integrating probe re-annotation pipeline and miRNA-target interactions. Firstly, some lncRNAs that had the central topological features were found in the heart failure-related lncRNA-mRNA network. Then, the lncRNA-associated functional modules were identified from the network, using bidirectional hierarchical clustering. Some lncRNAs that involved in modules were demonstrated to be enriched in many heart failure-related pathways. To investigate the role of lncRNA-associated ceRNA crosstalks in certain disease or physiological status, we further identified the lncRNA-associated dysregulated ceRNA interactions. And we also performed a random walk algorithm to identify more heart failure-related lncRNAs. All these lncRNAs were verified to show a strong diagnosis power for heart failure. These results will help us to understand the mechanism of lncRNAs in heart failure and provide novel lncRNAs as candidate diagnostic biomarkers or potential therapeutic targets.
Collapse
Affiliation(s)
- Zhimin Fan
- Department of CardiologyThe First Affiliated Hospital of Shantou University Medical CollegeShantouGuangdongChina
- Shantou University Medical CollegeShantouGuangdongChina
| | - Shanshan Gao
- Department of CardiologyThe First Affiliated Hospital of Shantou University Medical CollegeShantouGuangdongChina
- Shantou University Medical CollegeShantouGuangdongChina
| | - Yequn Chen
- Department of CardiologyThe First Affiliated Hospital of Shantou University Medical CollegeShantouGuangdongChina
- Shantou University Medical CollegeShantouGuangdongChina
| | - Bayi Xu
- Department of CardiologyThe First Affiliated Hospital of Shantou University Medical CollegeShantouGuangdongChina
- Shantou University Medical CollegeShantouGuangdongChina
| | - Chengzhi Yu
- Department of CardiologyThe First Affiliated Hospital of Shantou University Medical CollegeShantouGuangdongChina
- Shantou University Medical CollegeShantouGuangdongChina
| | - Minghui Yue
- Department of CardiologyThe First Affiliated Hospital of Shantou University Medical CollegeShantouGuangdongChina
- Shantou University Medical CollegeShantouGuangdongChina
| | - Xuerui Tan
- Department of CardiologyThe First Affiliated Hospital of Shantou University Medical CollegeShantouGuangdongChina
- Shantou University Medical CollegeShantouGuangdongChina
| |
Collapse
|
34
|
Effect of Aqueous Extract from Descurainia sophia (L.) Webb ex Prantl on Ventricular Remodeling in Chronic Heart Failure Rats. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2018; 2018:1904081. [PMID: 30008784 PMCID: PMC6020489 DOI: 10.1155/2018/1904081] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Revised: 04/07/2018] [Accepted: 05/22/2018] [Indexed: 02/08/2023]
Abstract
Objective Descurainia sophia (L.) Webb ex Prantl (DS) is a traditional Chinese medicine. Our current study was to evaluate the effect of DS on ventricular remodeling in chronic heart failure (HF) rats and its underlying mechanism. Methods The rat chronic heart failure model induced by suprarenal abdominal aortic coarctation surgery. The survival rats were randomly divided into 3 groups: the sham group (n=6), the HF group (n=6), and the HF+DS group (n=6). After 3 months of drug intervention, we examined the effects of DS by Sirius Red staining, electron microscopy, echocardiography, hemodynamic measurement, and TUNEL and explored the underlying mechanism by Western blotting. Results We found that rats treated with DS showed improved cardiac function and less tissue damage compared to untreated group. Additionally, DS could reduce the cardiomyocytes apoptosis, decrease the ratio of Bax/bcl-2 and Caspase-3 expression, and enhance the phosphorylation of Akt protein expression. Conclusion Our study suggested that rats treated with DS after suprarenal abdominal aortic coarctation surgery showed attenuated cardiac fibrosis and apoptosis, and the protective effect may be correlated with the activation of PI3k/Akt/mTOR dependent manner.
Collapse
|
35
|
Ma Q, Liu Y, Chen L. JIP3 deficiency attenuates cardiac hypertrophy by suppression of JNK pathway. Biochem Biophys Res Commun 2018; 503:1-7. [PMID: 29604277 DOI: 10.1016/j.bbrc.2018.03.208] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2018] [Accepted: 03/27/2018] [Indexed: 01/22/2023]
Abstract
Pathological cardiac hypertrophy is a leading cause of morbidity and mortality worldwide; however, our understanding of the molecular mechanisms revealing the disease is still unclear. In the present study, we suggested that c-Jun N-terminal kinase (JNK)-interacting protein 3 (JIP3), involved in various cellular processes, played an essential role in regulating pathological cardiac hypertrophy through in vivo and in vitro studies. JIP3 was highly expressed in human hearts with hypertrophic cardiomyopathy (HCM), and in mouse hypertrophic hearts. Following, the wild type (WT) and JIP3-knockout (KO) mice subjected to aortic banding (AB) challenge were used as animal models with cardiac hypertrophy. The results showed that JIP3-KO mice after AB operation exhibited attenuated cardiac function, reduced fibrosis levels and decreased hypertrophic marker proteins, including atrial natriuretic peptides (Anp) and brain/B-type natriuretic peptides (Bnp) and β-myosin heavy chain (β-Mhc). Loss of JIP3 also ameliorated oxidative stress, inflammatory response, apoptosis and endoplasmic reticulum (ER) stress in hearts of mice after AB surgery. Consistently, the expressions of ER stress-related molecules, such as phosphorylated-α-subunit of the eukaryotic initiation factor-2 (eIF2α), glucose-regulated protein (GRP) 78 and C/-EBP homologous protein (CHOP), were markedly decreased by JIP3-deficiency in hearts of AB-operated mice. JNK and its down-streaming signal of p90rsk was highly activated by AB operation in WT mice, while being significantly reversed by JIP3-ablation. Intriguingly, the in vitro results showed that promoting JNK activation by using its activator of anisomycin enhanced AngII-stimulated ER stress, oxidative stress, apoptosis and inflammatory response in cardiomyocytes isolated from WT mice. However, JIP3-KO-attenuated these pathologies was rescued by anisomycin treatment in AngII-incubated cardiomyocytes. Together, the findings indicated that blockage of JIP3 could alleviate cardiac hypertrophy via inactivating JNK pathway, and thus might be a promising strategy to prevent pathological cardiac hypertrophy.
Collapse
Affiliation(s)
- Qinghua Ma
- Department of Cardiology, Linyi Central Hospital of Shandong Province, Linyi 276400, China
| | - Yuxiu Liu
- Department of Geriatric Medicine, Linyi Central Hospital of Shandong Province, Linyi 276400, China
| | - Lianghua Chen
- Department of Cardiology, Shandong Provincial Hospital, Jinan 250021, China.
| |
Collapse
|
36
|
Yasmin, Maskari RA, McEniery CM, Cleary SE, Li Y, Siew K, Figg NL, Khir AW, Cockcroft JR, Wilkinson IB, O'Shaughnessy KM. The matrix proteins aggrecan and fibulin-1 play a key role in determining aortic stiffness. Sci Rep 2018; 8:8550. [PMID: 29867203 PMCID: PMC5986773 DOI: 10.1038/s41598-018-25851-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 04/18/2018] [Indexed: 12/14/2022] Open
Abstract
Stiffening of the aorta is an important independent risk factor for myocardial infarction and stroke. Yet its genetics is complex and little is known about its molecular drivers. We have identified for the first time, tagSNPs in the genes for extracellular matrix proteins, aggrecan and fibulin-1, that modulate stiffness in young healthy adults. We confirmed SNP associations with ex vivo stiffness measurements and expression studies in human donor aortic tissues. Both aggrecan and fibulin-1 were found in the aortic wall, but with marked differences in the distribution and glycosylation of aggrecan reflecting loss of chondroitin-sulphate binding domains. These differences were age-dependent but the striking finding was the acceleration of this process in stiff versus elastic young aortas. These findings suggest that aggrecan and fibulin-1 have critical roles in determining the biomechanics of the aorta and their modification with age could underpin age-related aortic stiffening.
Collapse
Affiliation(s)
- Yasmin
- Division of Experimental Medicine & Immunotherapeutics, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK.
| | - Raya Al Maskari
- Division of Experimental Medicine & Immunotherapeutics, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Carmel M McEniery
- Division of Experimental Medicine & Immunotherapeutics, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Sarah E Cleary
- Division of Experimental Medicine & Immunotherapeutics, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Ye Li
- Brunel Institute of Bioengineering, Brunel University, Uxbridge, Middlesex, UK
| | - Keith Siew
- Division of Experimental Medicine & Immunotherapeutics, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Nichola L Figg
- Division of Cardiovascular Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Ashraf W Khir
- Brunel Institute of Bioengineering, Brunel University, Uxbridge, Middlesex, UK
| | - John R Cockcroft
- Division of Cardiology, New York-Presbyterian Hospital, Columbia University, New York, USA
| | - Ian B Wilkinson
- Division of Experimental Medicine & Immunotherapeutics, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Kevin M O'Shaughnessy
- Division of Experimental Medicine & Immunotherapeutics, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| |
Collapse
|
37
|
Ninh VK, El Hajj EC, Mouton AJ, El Hajj MC, Gilpin NW, Gardner JD. Chronic Ethanol Administration Prevents Compensatory Cardiac Hypertrophy in Pressure Overload. Alcohol Clin Exp Res 2018; 42:10.1111/acer.13799. [PMID: 29846943 PMCID: PMC6269226 DOI: 10.1111/acer.13799] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 05/21/2018] [Indexed: 12/16/2022]
Abstract
BACKGROUND Alcohol is among the most commonly abused drugs worldwide and affects many organ systems, including the heart. Alcoholic cardiomyopathy is characterized by a dilated cardiac phenotype with extensive hypertrophy and extracellular matrix (ECM) remodeling. We have previously shown that chronic ethanol (EtOH) administration accelerates the progression to heart failure in a rat model of volume overload. However, the mechanism by which this decompensation occurs is unknown. For this study, we hypothesized that chronic EtOH administration would prevent compensatory hypertrophy and cardiac remodeling in a rodent model of pressure overload (PO). METHODS Abdominal aortic constriction was used to create PO in 8-week-old male Wistar rats. Alcohol administration was performed via chronic intermittent EtOH vapor inhalation for 2 weeks prior to surgery and for the duration of the 8-week study. Echocardiography measurements were taken to assess ventricular functional and structural changes. RESULTS PO increased posterior wall thickness and the hypertrophic markers, atrial and B-type natriuretic peptides (ANP and BNP). With the added stressor of EtOH, wall thickness, ANP, and BNP decreased in PO animals. The combination of PO and EtOH resulted in increased wall stress compared to PO alone. PO also caused increased expression of collagen I and III, whereas EtOH alone only increased collagen III. The combined stresses of PO and EtOH led to an increase in collagen I expression, but collagen III did not change, resulting in an increased collagen I/III ratio in the PO rats treated with EtOH. Lastly, Notch1 expression was significantly increased only in the PO rats treated with EtOH. CONCLUSIONS Our data indicate that chronic EtOH may limit the cardiac hypertrophy induced by PO which may be associated with a Notch1 mechanism, resulting in increased wall stress and altered ECM profile.
Collapse
Affiliation(s)
- Van K Ninh
- Department of Physiology, LSU Health Sciences Center, New Orleans, Louisiana
| | - Elia C El Hajj
- Department of Physiology, LSU Health Sciences Center, New Orleans, Louisiana
| | - Alan J Mouton
- Department of Physiology, LSU Health Sciences Center, New Orleans, Louisiana
| | - Milad C El Hajj
- Department of Physiology, LSU Health Sciences Center, New Orleans, Louisiana
| | - Nicholas W Gilpin
- Department of Physiology, LSU Health Sciences Center, New Orleans, Louisiana
| | - Jason D Gardner
- Department of Physiology, LSU Health Sciences Center, New Orleans, Louisiana
| |
Collapse
|
38
|
El Hajj EC, El Hajj MC, Ninh VK, Gardner JD. Inhibitor of lysyl oxidase improves cardiac function and the collagen/MMP profile in response to volume overload. Am J Physiol Heart Circ Physiol 2018; 315:H463-H473. [PMID: 29775412 DOI: 10.1152/ajpheart.00086.2018] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The cardiac extracellular matrix is a complex architectural network that serves many functions, including providing structural and biochemical support to surrounding cells and regulating intercellular signaling pathways. Cardiac function is directly affected by extracellular matrix (ECM) composition, and alterations of the ECM contribute to the progression of heart failure. Initially, collagen deposition is an adaptive response that aims to preserve tissue integrity and maintain normal ventricular function. However, the synergistic effects of proinflammatory and profibrotic responses induce a vicious cycle, which causes excess activation of myofibroblasts, significantly increasing collagen deposition and accumulation in the matrix. Furthermore, excess synthesis and activation of the enzyme lysyl oxidase (LOX) during disease increases collagen cross-linking, which significantly increases collagen resistance to degradation by matrix metalloproteinases (MMPs). In the present study, the aortocaval fistula model of volume overload (VO) was used to determine whether LOX inhibition could prevent adverse changes in the ECM and subsequent cardiac dysfunction. The major findings from this study were that LOX inhibition 1) prevented VO-induced increases in left ventricular wall stress; 2) partially attenuated VO-induced ventricular hypertrophy; 3) completely blocked the increases in fibrotic proteins, including collagens, MMPs, and their tissue inhibitors; and 4) prevented the VO-induced decline in cardiac function. It remains unclear whether a direct interaction between LOX and MMPs exists; however, our experiments suggest a potential link between the two because LOX inhibition completely attenuated VO-induced increases in MMPs. Overall, our study demonstrated key cardioprotective effects of LOX inhibition against adverse cardiac remodeling due to chronic VO. NEW & NOTEWORTHY Although the primary role of lysyl oxidase (LOX) is to cross-link collagens, we found that elevated LOX during cardiac disease plays a key role in the progression of heart failure. Here, we show that inhibition of LOX in volume-overloaded rats prevented the development of cardiac dysfunction and improved ventricular collagen and matrix metalloproteinase/tissue inhibitor of metalloproteinase profiles.
Collapse
Affiliation(s)
- Elia C El Hajj
- Department of Physiology, LSU Health Sciences Center , New Orleans, Louisiana
| | - Milad C El Hajj
- Department of Physiology, LSU Health Sciences Center , New Orleans, Louisiana
| | - Van K Ninh
- Department of Physiology, LSU Health Sciences Center , New Orleans, Louisiana
| | - Jason D Gardner
- Department of Physiology, LSU Health Sciences Center , New Orleans, Louisiana
| |
Collapse
|
39
|
Heger J, Bornbaum J, Würfel A, Hill C, Brockmann N, Gáspár R, Pálóczi J, Varga ZV, Sárközy M, Bencsik P, Csont T, Török S, Kojonazarov B, Schermuly RT, Böngler K, Parahuleva M, Ferdinandy P, Schulz R, Euler G. JDP2 overexpression provokes cardiac dysfunction in mice. Sci Rep 2018; 8:7647. [PMID: 29769710 PMCID: PMC5955919 DOI: 10.1038/s41598-018-26052-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 05/03/2018] [Indexed: 01/19/2023] Open
Abstract
The transcriptional regulator JDP2 (Jun dimerization protein 2) has been identified as a prognostic marker for patients to develop heart failure after myocardial infarction. We now performed in vivo studies on JDP2-overexpressing mice, to clarify the impact of JDP2 on heart failure progression. Therefore, during birth up to the age of 4 weeks cardiac-specific JDP2 overexpression was prevented by doxycycline feeding in transgenic mice. Then, JDP2 overexpression was started. Already after 1 week, cardiac function, determined by echocardiography, decreased which was also resembled on the cardiomyocyte level. After 5 weeks blood pressure declined, ejection fraction and cardiac output was reduced and left ventricular dilatation developed. Heart weight/body weight, and mRNA expression of ANP, inflammatory marker genes, collagen and fibronectin increased. Collagen 1 protein expression increased, and fibrosis developed. As an additional sign of elevated extracellular matrix remodeling, matrix metalloproteinase 2 activity increased in JDP2 mice. Thus, JDP2 overexpression is deleterious to heart function in vivo. It can be concluded that JDP2 overexpression provokes cardiac dysfunction in adult mice that is accompanied by hypertrophy and fibrosis. Thus, induction of JDP2 is a maladaptive response contributing to heart failure development.
Collapse
Affiliation(s)
- Jacqueline Heger
- Institute of Physiology, Justus Liebig University, Giessen, Germany
| | - Julia Bornbaum
- Institute of Physiology, Justus Liebig University, Giessen, Germany
| | - Alona Würfel
- Institute of Physiology, Justus Liebig University, Giessen, Germany
| | - Christian Hill
- Institute of Physiology, Justus Liebig University, Giessen, Germany
| | - Nils Brockmann
- Institute of Physiology, Justus Liebig University, Giessen, Germany
| | - Renáta Gáspár
- Department of Biochemistry, University of Szeged, Szeged, Hungary
| | - János Pálóczi
- Department of Biochemistry, University of Szeged, Szeged, Hungary.,Pharmahungary Group, Szeged, Hungary
| | - Zoltán V Varga
- Department of Biochemistry, University of Szeged, Szeged, Hungary
| | - Márta Sárközy
- Department of Biochemistry, University of Szeged, Szeged, Hungary
| | - Péter Bencsik
- Department of Biochemistry, University of Szeged, Szeged, Hungary.,Pharmahungary Group, Szeged, Hungary
| | - Tamás Csont
- Department of Biochemistry, University of Szeged, Szeged, Hungary
| | - Szilvia Török
- Department of Biochemistry, University of Szeged, Szeged, Hungary
| | - Baktybek Kojonazarov
- Universities of Giessen and Marburg Lung Center (UGMLC), Excellence Cluster Cardio-Pulmonary System (ECCPS), Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Ralph Theo Schermuly
- Universities of Giessen and Marburg Lung Center (UGMLC), Excellence Cluster Cardio-Pulmonary System (ECCPS), Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Kerstin Böngler
- Institute of Physiology, Justus Liebig University, Giessen, Germany
| | - Mariana Parahuleva
- Internal Medicine/Cardiology and Angiology, University Hospital of Giessen and Marburg, Location Marburg, Marburg, Germany
| | - Peter Ferdinandy
- Department of Biochemistry, University of Szeged, Szeged, Hungary.,Pharmahungary Group, Szeged, Hungary.,Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Rainer Schulz
- Institute of Physiology, Justus Liebig University, Giessen, Germany
| | - Gerhild Euler
- Institute of Physiology, Justus Liebig University, Giessen, Germany.
| |
Collapse
|
40
|
Li L, Zhao Q, Kong W. Extracellular matrix remodeling and cardiac fibrosis. Matrix Biol 2018; 68-69:490-506. [PMID: 29371055 DOI: 10.1016/j.matbio.2018.01.013] [Citation(s) in RCA: 249] [Impact Index Per Article: 35.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 01/15/2018] [Accepted: 01/16/2018] [Indexed: 12/19/2022]
Abstract
Cardiac fibrosis, characterized by excessive deposition of extracellular matrix (ECM) proteins in the myocardium, distorts the architecture of the myocardium, facilitates the progression of arrhythmia and cardiac dysfunction, and influences the clinical course and outcome in patients with heart failure. This review describes the composition and homeostasis in normal cardiac interstitial matrix and introduces cellular and molecular mechanisms involved in cardiac fibrosis. We also characterize the ECM alteration in the fibrotic response under diverse cardiac pathological conditions and depict the role of matricellular proteins in the pathogenesis of cardiac fibrosis. Moreover, the diagnosis of cardiac fibrosis based on imaging and biomarker detection and the therapeutic strategies are addressed. Understanding the comprehensive molecules and pathways involved in ECM homeostasis and remodeling may provide important novel potential targets for preventing and treating cardiac fibrosis.
Collapse
Affiliation(s)
- Li Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, China; Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing 100191, China
| | - Qian Zhao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, China; Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing 100191, China
| | - Wei Kong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, China; Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing 100191, China.
| |
Collapse
|
41
|
Circulating Biomarkers in Heart Failure. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1067:89-108. [PMID: 29392578 DOI: 10.1007/5584_2017_140] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Biological markers have served for diagnosis, risk stratification and guided therapy of heart failure (HF). Our knowledge regarding abilities of biomarkers to relate to several pathways of HF pathogenesis and reflect clinical worsening or improvement in the disease is steadily expanding. Although there are numerous clinical guidelines, which clearly diagnosis, prevention and evidence-based treatment of HF, a strategy regarding exclusion of HF, as well as risk stratification of HF, nature evolution of disease is not well established and requires more development. The aim of the chapter is to discuss a role of biomarker-based approaches for more accurate diagnosis, in-depth risk stratification and individual targeting in treatment of patients with HF.
Collapse
|
42
|
Zhang F, Wang C, Lin J, Wang X. Oxidized low-density lipoprotein (ox-LDL) promotes cardiac differentiation of bone marrow mesenchymal stem cells via activating ERK1/2 signaling. Cardiovasc Ther 2017; 35. [PMID: 28880487 DOI: 10.1111/1755-5922.12305] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Revised: 08/16/2017] [Accepted: 09/02/2017] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND/AIMS The differentiation efficiency of bone marrow mesenchymal stem cells (BM-MSCs) is low in vivo after transplantation. Therefore, it is necessary to look for effective reagents for enhancing cardiac differentiation of BM-MSCs. It has been reported that cardiac differentiation of stem cells depends on the activation of extracellular signal-regulated protein 1/2 (ERK1/2) signaling. Oxidized low-density lipoprotein (ox-LDL) is a potent reagent for ERK1/2 activation. This indicates that ox-LDL may be a potential reagent to stimulate cardiac differentiation of stem cells. In this study, we investigated the effect of ox-LDL on cardiac differentiation of BM-MSCs and its relationship with ERK1/2 signaling. METHODS BM-MSCs were isolated from mouse bone marrow, cultured in DMEM supplemented with 15% FBS, and passaged up to the 3rd passage. Following culture with 5 μg/mL ox-LDL for 3 weeks, the cardiac differentiation of the 3rd passage BM-MSCs was identified by immunostaining, Western blotting, and RT-PCR assays for measuring the expression of cardiac-specific markers. To further explore the role of ERK1/2 signaling in cardiac differentiation of BM-MSCs, we simultaneously exposed BM-MSCs to ERK1/2 inhibitor (U0126) and ox-LDL, and identified the cardiac differentiation again. RESULTS The expressions of cardiac-specific markers including α-cardiac actin, α-MHC, β-MHC, ANP, and BNP were markedly increased in BM-MSCs following treatment with ox-LDL (P < .05), which indicates a directional differentiation of BM-MSCs to cardiac cells. Further, ox-LDL could also activate ERK1/2 in BM-MSCs, and application of U0126 markedly inhibited ox-LDL-induced cardiac transformation of BM-MSCs. CONCLUSIONS Ox-LDL induces cardiac differentiation of BM-MSCs via activation of ERK1/2 signaling.
Collapse
Affiliation(s)
- Fenxi Zhang
- Stem Cell and Biotheraphy Technology Research Center, Xinxiang Medical University, Xinxiang, China
| | - Congrui Wang
- Stem Cell and Biotheraphy Technology Research Center, Xinxiang Medical University, Xinxiang, China
| | - Juntang Lin
- Stem Cell and Biotheraphy Technology Research Center, Xinxiang Medical University, Xinxiang, China.,Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, China
| | - Xianwei Wang
- Stem Cell and Biotheraphy Technology Research Center, Xinxiang Medical University, Xinxiang, China.,Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
43
|
Zhang Y, Bauersachs J, Langer HF. Immune mechanisms in heart failure. Eur J Heart Fail 2017; 19:1379-1389. [DOI: 10.1002/ejhf.942] [Citation(s) in RCA: 115] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2017] [Revised: 05/26/2017] [Accepted: 06/19/2017] [Indexed: 12/15/2022] Open
Affiliation(s)
- Yingying Zhang
- University Hospital, Department of Cardiology and Cardiovascular Medicine; Eberhard Karls University Tuebingen; Tuebingen Germany
- Section for Cardioimmunology; Eberhard Karls University Tuebingen; Tübingen Germany
- Affiliated Hospital of Qingdao University, Department of Cardiology and Cardiovascular Medicine; Qingdao University; Qingdao China
| | - Johann Bauersachs
- Department of Cardiology and Angiology; Hannover Medical School; Hannover Germany
| | - Harald F. Langer
- University Hospital, Department of Cardiology and Cardiovascular Medicine; Eberhard Karls University Tuebingen; Tuebingen Germany
- Section for Cardioimmunology; Eberhard Karls University Tuebingen; Tübingen Germany
| |
Collapse
|
44
|
Ichihara S, Suzuki Y, Chang J, Kuzuya K, Inoue C, Kitamura Y, Oikawa S. Involvement of oxidative modification of proteins related to ATP synthesis in the left ventricles of hamsters with cardiomyopathy. Sci Rep 2017; 7:9243. [PMID: 28835655 PMCID: PMC5569096 DOI: 10.1038/s41598-017-08546-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 07/14/2017] [Indexed: 12/21/2022] Open
Abstract
Inflammation enhanced by accumulation of reactive oxygen species plays an essential role in the progression of cardiovascular diseases. Using the 2D-oxyblot analysis and 2D-difference image gel electrophoresis (2D-DIGE), we compared the levels of ROS-induced carbonyl modification of myocardial proteins in the whole left ventricles between 6-week-old hamsters with dilated (TO-2) and hypertrophic cardiomyopathy (Bio14.6) and control hamsters (F1B). Then, 2D electrophoresis combined with MALDI-TOF/TOF tandem mass spectrometry detected 18 proteins with increased carbonyl level in cardiomyopathy hamsters compared with control hamster. Carbonyl modification of proteins related to ATP synthesis, including citric acid cycle and electron transport system, was observed in the hearts of hamsters with both types of cardiomyopathy. Further analysis indicated that left ventricular carbonyl production correlated negatively with succinyl-CoA:3-ketoacid-coenzyme A transferase 1 activity (r 2 = 0.60, P = 0.0007) and ATP concentration (r 2 = 0.29, P = 0.037), suggesting that protein carbonylation has negative effects on the levels of these biomolecules. Furthermore, carbonyl production significantly correlated with plasma Troponin T level (r 2 = 0.33, P = 0.026). Reduction of energy metabolism by oxidative damage may contribute to the development of left ventricular impairment in cardiomyopathy.
Collapse
Affiliation(s)
- Sahoko Ichihara
- Graduate School of Regional Innovation Studies, Tsu, Japan.
- Department of Human Functional Genomics, Life Science Research Center, Mie University, Tsu, Japan.
- Department of Environmental and Preventive Medicine, Jichi Medical University School of Medicine, Shimotsuke, Japan.
| | - Yuka Suzuki
- Graduate School of Regional Innovation Studies, Tsu, Japan
- Community-University Research Cooperation Center, Mie University, Tsu, Japan
| | - Jie Chang
- Graduate School of Regional Innovation Studies, Tsu, Japan
- School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Kentaro Kuzuya
- Department of Human Functional Genomics, Life Science Research Center, Mie University, Tsu, Japan
| | - Chisa Inoue
- Department of Human Functional Genomics, Life Science Research Center, Mie University, Tsu, Japan
| | - Yuki Kitamura
- Department of Environmental and Molecular Medicine, Mie University Graduate School of Medicine, Tsu, Japan
| | - Shinji Oikawa
- Department of Environmental and Molecular Medicine, Mie University Graduate School of Medicine, Tsu, Japan
| |
Collapse
|
45
|
El Hajj EC, El Hajj MC, Ninh VK, Bradley JM, Claudino MA, Gardner JD. Detrimental role of lysyl oxidase in cardiac remodeling. J Mol Cell Cardiol 2017; 109:17-26. [DOI: 10.1016/j.yjmcc.2017.06.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 06/15/2017] [Accepted: 06/27/2017] [Indexed: 01/02/2023]
|
46
|
Frangogiannis NG. The extracellular matrix in myocardial injury, repair, and remodeling. J Clin Invest 2017; 127:1600-1612. [PMID: 28459429 DOI: 10.1172/jci87491] [Citation(s) in RCA: 332] [Impact Index Per Article: 41.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The cardiac extracellular matrix (ECM) not only provides mechanical support, but also transduces essential molecular signals in health and disease. Following myocardial infarction, dynamic ECM changes drive inflammation and repair. Early generation of bioactive matrix fragments activates proinflammatory signaling. The formation of a highly plastic provisional matrix facilitates leukocyte infiltration and activates infarct myofibroblasts. Deposition of matricellular proteins modulates growth factor signaling and contributes to the spatial and temporal regulation of the reparative response. Mechanical stress due to pressure and volume overload and metabolic dysfunction also induce profound changes in ECM composition that contribute to the pathogenesis of heart failure. This manuscript reviews the role of the ECM in cardiac repair and remodeling and discusses matrix-based therapies that may attenuate remodeling while promoting repair and regeneration.
Collapse
|
47
|
Deddens JC, Sadeghi AH, Hjortnaes J, van Laake LW, Buijsrogge M, Doevendans PA, Khademhosseini A, Sluijter JPG. Modeling the Human Scarred Heart In Vitro: Toward New Tissue Engineered Models. Adv Healthc Mater 2017; 6. [PMID: 27906521 DOI: 10.1002/adhm.201600571] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2016] [Revised: 07/07/2016] [Indexed: 12/11/2022]
Abstract
Cardiac remodeling is critical for effective tissue healing, however, excessive production and deposition of extracellular matrix components contribute to scarring and failing of the heart. Despite the fact that novel therapies have emerged, there are still no lifelong solutions for this problem. An urgent need exists to improve the understanding of adverse cardiac remodeling in order to develop new therapeutic interventions that will prevent, reverse, or regenerate the fibrotic changes in the failing heart. With recent advances in both disease biology and cardiac tissue engineering, the translation of fundamental laboratory research toward the treatment of chronic heart failure patients becomes a more realistic option. Here, the current understanding of cardiac fibrosis and the great potential of tissue engineering are presented. Approaches using hydrogel-based tissue engineered heart constructs are discussed to contemplate key challenges for modeling tissue engineered cardiac fibrosis and to provide a future outlook for preclinical and clinical applications.
Collapse
Affiliation(s)
- Janine C. Deddens
- Department of Cardiology; University Medical Center Utrecht; 3584CX Utrecht The Netherlands
- Netherlands Heart Institute (ICIN); 3584CX Utrecht The Netherlands
| | - Amir Hossein Sadeghi
- Department of Cardiology; University Medical Center Utrecht; 3584CX Utrecht The Netherlands
- Department of Cardiothoracic Surgery; Division Heart and Lungs; University Medical Center Utrecht; 3584CX Utrecht The Netherlands
- Biomaterials Innovation Research Center; Department of Medicine; Brigham and Women's Hospital; Harvard Medical School; Cambridge MA 02139 USA
- Harvard-MIT Division of Health Sciences & Technology; Massachusetts Institute of Technology; Cambridge MA 02139 USA
| | - Jesper Hjortnaes
- Department of Cardiothoracic Surgery; Division Heart and Lungs; University Medical Center Utrecht; 3584CX Utrecht The Netherlands
- UMC Utrecht Regenerative Medicine Center; University Medical Center Utrecht; 3584CT Utrecht The Netherlands
| | - Linda W. van Laake
- Department of Cardiology; University Medical Center Utrecht; 3584CX Utrecht The Netherlands
- UMC Utrecht Regenerative Medicine Center; University Medical Center Utrecht; 3584CT Utrecht The Netherlands
| | - Marc Buijsrogge
- Department of Cardiothoracic Surgery; Division Heart and Lungs; University Medical Center Utrecht; 3584CX Utrecht The Netherlands
| | - Pieter A. Doevendans
- Department of Cardiology; University Medical Center Utrecht; 3584CX Utrecht The Netherlands
- Netherlands Heart Institute (ICIN); 3584CX Utrecht The Netherlands
- UMC Utrecht Regenerative Medicine Center; University Medical Center Utrecht; 3584CT Utrecht The Netherlands
| | - Ali Khademhosseini
- Biomaterials Innovation Research Center; Department of Medicine; Brigham and Women's Hospital; Harvard Medical School; Cambridge MA 02139 USA
- Harvard-MIT Division of Health Sciences & Technology; Massachusetts Institute of Technology; Cambridge MA 02139 USA
- Wyss Institute for Biologically Inspired Engineering; Harvard University; Boston MA 02115 USA
- Department of Physics; King Abdulaziz University; Jeddah 21569 Saudi Arabia
| | - Joost P. G. Sluijter
- Department of Cardiology; University Medical Center Utrecht; 3584CX Utrecht The Netherlands
- Netherlands Heart Institute (ICIN); 3584CX Utrecht The Netherlands
- UMC Utrecht Regenerative Medicine Center; University Medical Center Utrecht; 3584CT Utrecht The Netherlands
| |
Collapse
|
48
|
Cardamonin Alleviates Pressure Overload-induced Cardiac Remodeling and Dysfunction Through Inhibition of Oxidative Stress. J Cardiovasc Pharmacol 2016; 68:441-451. [DOI: 10.1097/fjc.0000000000000430] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
49
|
El Hajj MC, Ninh VK, El Hajj EC, Bradley JM, Gardner JD. Estrogen receptor antagonism exacerbates cardiac structural and functional remodeling in female rats. Am J Physiol Heart Circ Physiol 2016; 312:H98-H105. [PMID: 27769996 DOI: 10.1152/ajpheart.00348.2016] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 10/04/2016] [Accepted: 10/17/2016] [Indexed: 12/24/2022]
Abstract
We have previously demonstrated the cardioprotective effects of ovarian hormones against adverse ventricular remodeling imposed by chronic volume overload. Here, we assess the estrogen receptor dependence of this cardioprotection. Four groups of female rats were studied: sham-operated (Sham), volume overloaded [aortocaval fistula (ACF)], Sham treated with estrogen receptor antagonist ICI 182,780 (Sham + ICI), and ACF treated with ICI. Cardiac function was assessed temporally using echocardiogram, and tissue samples were collected at 5 days and 6 wk postsurgery. All rats with volume overload had significantly increased cardiac output (96 ± 32 ml/min for ACF and 108 ± 11 ml/min for ACF + ICI vs. 31 ± 2 for Sham, P < 0.05). At 6 wk, volume overload induced significant left ventricular (LV) hypertrophy in both untreated and treated ACF groups. Both ACF groups developed significantly increased LV end-diastolic diameter (LVEDD), indicating LV dilatation, with the ACF + ICI group having the greatest increase (340%, relative to Sham). Ejection fraction was significantly reduced in the ACF + ICI group (23% reduction) at 6 wk postsurgery compared with untreated ACF (P < 0.05). Interstitial collagen staining was significantly reduced by volume overload, with estrogen receptor antagonism causing greater collagen loss at both 5 days and 6 wk postsurgery. Furthermore, volume overload induced a significant increase in LV wall stress only in rats treated with estrogen antagonist. These data indicate that estrogen receptor signaling is essential for sex hormone-dependent cardioprotection against adverse remodeling. The maintenance of myocardial extracellular matrix collagen appears to play a key role in this cardioprotection. NEW & NOTEWORTHY We assessed the estrogen receptor (ER) dependence of female-specific cardioprotection using a rat model of chronic volume-overload stress. ER antagonism worsened ventricular wall stress, ventricular dilation, and cardiac dysfunction induced by volume overload. Further, blocking ERs resulted in cardiac remodeling and functional changes similar to that previously found in ovariectomized rats.
Collapse
Affiliation(s)
- M C El Hajj
- Department of Physiology, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| | - V K Ninh
- Department of Physiology, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| | - E C El Hajj
- Department of Physiology, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| | - J M Bradley
- Department of Physiology, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| | - J D Gardner
- Department of Physiology, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| |
Collapse
|
50
|
Benoist D, Dubes V, Roubertie F, Gilbert SH, Charron S, Constantin M, Elbes D, Vieillot D, Quesson B, Cochet H, Haïssaguerre M, Rooryck C, Bordachar P, Thambo JB, Bernus O. Proarrhythmic remodelling of the right ventricle in a porcine model of repaired tetralogy of Fallot. Heart 2016; 103:347-354. [PMID: 28051771 PMCID: PMC5529985 DOI: 10.1136/heartjnl-2016-309730] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 06/27/2016] [Accepted: 07/21/2016] [Indexed: 11/17/2022] Open
Abstract
Objective The growing adult population with surgically corrected tetralogy of Fallot (TOF) is at risk of arrhythmias and sudden cardiac death. We sought to investigate the contribution of right ventricular (RV) structural and electrophysiological remodelling to arrhythmia generation in a preclinical animal model of repaired TOF (rTOF). Methods and results Pigs mimicking rTOF underwent cardiac MRI functional characterisation and presented with pulmonary regurgitation, RV hypertrophy, dilatation and dysfunction compared with Sham-operated animals (Sham). Optical mapping of rTOF RV-perfused wedges revealed a significant prolongation of RV activation time with slower conduction velocities and regions of conduction slowing well beyond the surgical scar. A reduced protein expression and lateralisation of Connexin-43 were identified in rTOF RVs. A remodelling of extracellular matrix-related gene expression and an increase in collagen content that correlated with prolonged RV activation time were also found in these animals. RV action potential duration (APD) was prolonged in the epicardial anterior region at early and late repolarisation level, thus contributing to a greater APD heterogeneity and to altered transmural and anteroposterior APD gradients in rTOF RVs. APD remodelling involved changes in Kv4.3 and MiRP1 expression. Spontaneous arrhythmias were more frequent in rTOF wedges and more complex in the anterior than in the posterior RV. Conclusion Significant remodelling of RV conduction and repolarisation properties was found in pigs with rTOF. This remodelling generates a proarrhythmic substrate likely to facilitate re-entries and to contribute to sudden cardiac death in patients with rTOF.
Collapse
Affiliation(s)
- David Benoist
- IHU LIRYC, L'Institut de Rythmologie et Modélisation Cardiaque, Fondation Bordeaux Université, Bordeaux, France.,Inserm U1045, Centre de Recherche Cardio-Thoracique de Bordeaux, Université de Bordeaux, Bordeaux, France
| | - Virginie Dubes
- IHU LIRYC, L'Institut de Rythmologie et Modélisation Cardiaque, Fondation Bordeaux Université, Bordeaux, France.,Inserm U1045, Centre de Recherche Cardio-Thoracique de Bordeaux, Université de Bordeaux, Bordeaux, France
| | - François Roubertie
- IHU LIRYC, L'Institut de Rythmologie et Modélisation Cardiaque, Fondation Bordeaux Université, Bordeaux, France.,Inserm U1045, Centre de Recherche Cardio-Thoracique de Bordeaux, Université de Bordeaux, Bordeaux, France.,CHU de Bordeaux, Hôpital Cardiologique du Haut-Lévêque, Pessac, France
| | - Stephen H Gilbert
- IHU LIRYC, L'Institut de Rythmologie et Modélisation Cardiaque, Fondation Bordeaux Université, Bordeaux, France.,Inserm U1045, Centre de Recherche Cardio-Thoracique de Bordeaux, Université de Bordeaux, Bordeaux, France.,Max Delbrück Center for Molecular Medicine, Mathematical Cell Physiology, Berlin, Germany
| | - Sabine Charron
- IHU LIRYC, L'Institut de Rythmologie et Modélisation Cardiaque, Fondation Bordeaux Université, Bordeaux, France.,Inserm U1045, Centre de Recherche Cardio-Thoracique de Bordeaux, Université de Bordeaux, Bordeaux, France
| | - Marion Constantin
- IHU LIRYC, L'Institut de Rythmologie et Modélisation Cardiaque, Fondation Bordeaux Université, Bordeaux, France.,Inserm U1045, Centre de Recherche Cardio-Thoracique de Bordeaux, Université de Bordeaux, Bordeaux, France
| | - Delphine Elbes
- IHU LIRYC, L'Institut de Rythmologie et Modélisation Cardiaque, Fondation Bordeaux Université, Bordeaux, France.,Inserm U1045, Centre de Recherche Cardio-Thoracique de Bordeaux, Université de Bordeaux, Bordeaux, France.,Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, UK
| | - Delphine Vieillot
- Plateforme Technologique d'Innovation Biomédicale, Université de Bordeaux, Pessac, France
| | - Bruno Quesson
- IHU LIRYC, L'Institut de Rythmologie et Modélisation Cardiaque, Fondation Bordeaux Université, Bordeaux, France.,Inserm U1045, Centre de Recherche Cardio-Thoracique de Bordeaux, Université de Bordeaux, Bordeaux, France
| | - Hubert Cochet
- IHU LIRYC, L'Institut de Rythmologie et Modélisation Cardiaque, Fondation Bordeaux Université, Bordeaux, France.,Inserm U1045, Centre de Recherche Cardio-Thoracique de Bordeaux, Université de Bordeaux, Bordeaux, France.,CHU de Bordeaux, Hôpital Cardiologique du Haut-Lévêque, Pessac, France
| | - Michel Haïssaguerre
- IHU LIRYC, L'Institut de Rythmologie et Modélisation Cardiaque, Fondation Bordeaux Université, Bordeaux, France.,Inserm U1045, Centre de Recherche Cardio-Thoracique de Bordeaux, Université de Bordeaux, Bordeaux, France.,CHU de Bordeaux, Hôpital Cardiologique du Haut-Lévêque, Pessac, France
| | - Caroline Rooryck
- Inserm U1211, Maladies Rares: Génétique et Métabolisme, Université de Bordeaux, Bordeaux, France
| | - Pierre Bordachar
- IHU LIRYC, L'Institut de Rythmologie et Modélisation Cardiaque, Fondation Bordeaux Université, Bordeaux, France.,Inserm U1045, Centre de Recherche Cardio-Thoracique de Bordeaux, Université de Bordeaux, Bordeaux, France.,CHU de Bordeaux, Hôpital Cardiologique du Haut-Lévêque, Pessac, France
| | - Jean-Benoit Thambo
- IHU LIRYC, L'Institut de Rythmologie et Modélisation Cardiaque, Fondation Bordeaux Université, Bordeaux, France.,Inserm U1045, Centre de Recherche Cardio-Thoracique de Bordeaux, Université de Bordeaux, Bordeaux, France.,CHU de Bordeaux, Hôpital Cardiologique du Haut-Lévêque, Pessac, France
| | - Olivier Bernus
- IHU LIRYC, L'Institut de Rythmologie et Modélisation Cardiaque, Fondation Bordeaux Université, Bordeaux, France.,Inserm U1045, Centre de Recherche Cardio-Thoracique de Bordeaux, Université de Bordeaux, Bordeaux, France
| |
Collapse
|