1
|
Bracamonte JH, Watkins L, Pat B, Dell’Italia LJ, Saucerman JJ, Holmes JW. Contributions of mechanical loading and hormonal changes to eccentric hypertrophy during volume overload: a Bayesian analysis using logic-based network models. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.12.612768. [PMID: 39345523 PMCID: PMC11429691 DOI: 10.1101/2024.09.12.612768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Primary mitral regurgitation (MR) is a pathology that alters mechanical loading on the left ventricle, triggers an array of compensatory neurohormonal responses, and induces a distinctive ventricular remodeling response known as eccentric hypertrophy. Drug therapies may alleviate symptoms, but only mitral valve repair or replacement can provide significant recovery of cardiac function and dimensions. Questions remain about the optimal timing of surgery, with 20% of patients developing systolic dysfunction post-operatively despite being treated according to the current guidelines. Thus, better understanding of the hypertrophic process in the setting of ventricular volume overload (VO) is needed to improve and better personalize the management of MR. To address this knowledge gap, we employ a Bayesian approach to combine data from 70 studies on experimental volume overload in dogs and rats and use it to calibrate a logic-based network model of hypertrophic signaling in myocytes. The calibrated model predicts that growth in experimental VO is mostly driven by the neurohormonal response, with an initial increase in myocardial tissue stretch being compensated by subsequent remodeling fairly early in the time course of VO. This observation contrasts with a common perception that volume-overload hypertrophy is driven primarily by increased myocyte strain. The model reproduces many aspects of 43 studies not used in its calibration, including infusion of individual hypertrophic agonists alone or in combination with various drugs commonly employed to treat heart failure, as well as administration of some of those drugs in the setting of experimental volume overload. We believe this represents a promising approach to using the known structure of an intracellular signaling network to integrate information from multiple studies into quantitative predictions of the range of expected responses to potential interventions in the complex setting of cardiac hypertrophy driven by a combination of hormonal and mechanical factors.
Collapse
Affiliation(s)
- Johane H. Bracamonte
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Lionel Watkins
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, United States of America
| | - Betty Pat
- Birmingham Veterans Affairs Health Care System, Birmingham, Alabama, United States of America
- Division of Cardiovascular Disease, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Louis J. Dell’Italia
- Birmingham Veterans Affairs Health Care System, Birmingham, Alabama, United States of America
- Division of Cardiovascular Disease, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Jeffrey J. Saucerman
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, United States of America
| | - Jeffrey W. Holmes
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- Division of Cardiovascular Disease, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- Division of Cardiothoracic Surgery, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| |
Collapse
|
2
|
Hassanein EHM, Althagafy HS, Baraka MA, Abd-Alhameed EK, Ibrahim IM, Abd El-Maksoud MS, Mohamed NM, Ross SA. The promising antioxidant effects of lignans: Nrf2 activation comes into view. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:6439-6458. [PMID: 38695909 PMCID: PMC11422461 DOI: 10.1007/s00210-024-03102-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 04/11/2024] [Indexed: 09/25/2024]
Abstract
Lignans are biologically active compounds widely distributed, recognized, and identified in seeds, fruits, and vegetables. Lignans have several intriguing bioactivities, including anti-inflammatory, antioxidant, and anticancer activities. Nrf2 controls the expression of many cytoprotective genes. Activation of Nrf2 is a promising therapeutic approach for treating and preventing diseases resulting from oxidative injury and inflammation. Lignans have been demonstrated to stimulate Nrf2 signaling in a variety of in vitro and experimental animal models. The review summarizes the findings of fourteen lignans (Schisandrin A, Schisandrin B, Schisandrian C, Magnolol, Honokiol, Sesamin, Sesamol, Sauchinone, Pinoresinol, Phyllanthin, Nectandrin B, Isoeucommin A, Arctigenin, Lariciresinol) as antioxidative and anti-inflammatory agents, affirming how Nrf2 activation affects their pharmacological effects. Therefore, lignans may offer therapeutic candidates for the treatment and prevention of various diseases and may contribute to the development of effective Nrf2 modulators.
Collapse
Affiliation(s)
- Emad H M Hassanein
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Assiut, 71524, Egypt
| | - Hanan S Althagafy
- Department of Biochemistry, Faculty of Science, University of Jeddah, Jeddah, Saudi Arabia
| | - Mohammad A Baraka
- Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Assiut, 71524, Egypt
| | - Esraa K Abd-Alhameed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| | - Islam M Ibrahim
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| | - Mostafa S Abd El-Maksoud
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| | - Nesma M Mohamed
- Department of Pharmacognosy, Faculty of Pharmacy, Assiut University, Assiut, 71526, Egypt.
- Department of Pharmacognosy, Faculty of Pharmacy, Badr University in Assiut, Assiut, 77771, Egypt.
| | - Samir A Ross
- National Center for Natural Products Research, Research Institute of Pharmaceutical Sciences, School of Pharmacy, The University of Mississippi, University, MS, 38677, USA
- Department of BioMolecular Sciences, Division of Pharmacognosy, School of Pharmacy, University of Mississippi, University, MS, 38677, USA
| |
Collapse
|
3
|
Kim TS, Hong CY, Oh SJ, Choe YH, Hwang TS, Kim J, Lee SL, Yoon H, Bok EY, Cho AR, Do YJ, Kim E. RNA sequencing provides novel insights into the pathogenesis of naturally occurring myxomatous mitral valve disease stage B1 in beagle dogs. PLoS One 2024; 19:e0300813. [PMID: 38753730 PMCID: PMC11098313 DOI: 10.1371/journal.pone.0300813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Accepted: 03/05/2024] [Indexed: 05/18/2024] Open
Abstract
Myxomatous mitral valve disease (MMVD) is the most common cardiovascular disorder in dogs with a high prevalence, accounting for approximately 75% of all canine heart disease cases. MMVD is a complex disease and shows variable progression from mild valve leakage to severe regurgitation, potentially leading to heart failure. However, the molecular mechanisms and age-related changes that govern disease progression, especially at the early stage (B1) before the development of discernable clinical signs, remain poorly understood. In this prospective study, we aimed to compare gene expression differences between blood samples of aged beagle dogs with stage B1 MMVD and those of healthy controls using RNA sequencing. Clinical evaluation was also conducted, which revealed minimal differences in radiographic and echocardiographic measurements despite distinct biomarker variations between the two groups. Comparative transcriptomics revealed differentially expressed genes associated with extracellular matrix remodeling, prostaglandin metabolism, immune modulation, and interferon-related pathways, which bear functional relevance for MMVD. In particular, the top 10 over- and under-expressed genes represent promising candidates for influencing pathogenic changes in MMVD stage B1. Our research findings, which include identified variations in clinical markers and gene expression, enhance our understanding of MMVD. Furthermore, they underscore the need for further research into early diagnosis and treatment strategies, as, to the best of our knowledge, no prior studies have explored the precise molecular mechanisms of stage B1 in MMVD through total RNA sequencing.
Collapse
Affiliation(s)
- Tae-Seok Kim
- College of Veterinary Medicine, Gyeongsang National University, Jinju, Gyeongsangnam-do, Republic of Korea
| | - Chae-Yeon Hong
- College of Veterinary Medicine, Gyeongsang National University, Jinju, Gyeongsangnam-do, Republic of Korea
| | - Seong-Ju Oh
- College of Veterinary Medicine, Gyeongsang National University, Jinju, Gyeongsangnam-do, Republic of Korea
| | - Yong-Ho Choe
- College of Veterinary Medicine, Gyeongsang National University, Jinju, Gyeongsangnam-do, Republic of Korea
| | - Tae-Sung Hwang
- College of Veterinary Medicine, Gyeongsang National University, Jinju, Gyeongsangnam-do, Republic of Korea
| | - Jaemin Kim
- Division of Applied Life Science, Gyeongsang National University, Jinju, Gyeongsangnam-do, Republic of Korea
- Institute of Agriculture and Life Sciences, Gyeongsang National University, Jinju, Gyeongsangnam-do, Republic of Korea
| | - Sung-Lim Lee
- College of Veterinary Medicine, Gyeongsang National University, Jinju, Gyeongsangnam-do, Republic of Korea
- Research Institute of Life Sciences, Gyeongsang National University, Jinju, Gyeongsangnam-do, Republic of Korea
| | - Hakyoung Yoon
- College of Veterinary Medicine, Jeonbuk National University, Iksan, Jeollabuk-do, Republic of Korea
| | - Eun-Yeong Bok
- Division of Animal Diseases & Health, National Institute of Animal Science, Rural Development Administration, Wanju, Jeollabuk-do, Republic of Korea
| | - A-ra Cho
- Division of Animal Diseases & Health, National Institute of Animal Science, Rural Development Administration, Wanju, Jeollabuk-do, Republic of Korea
| | - Yoon Jung Do
- Division of Animal Diseases & Health, National Institute of Animal Science, Rural Development Administration, Wanju, Jeollabuk-do, Republic of Korea
| | - Eunju Kim
- Division of Animal Diseases & Health, National Institute of Animal Science, Rural Development Administration, Wanju, Jeollabuk-do, Republic of Korea
| |
Collapse
|
4
|
Bonet F, Hernandez-Torres F, Ramos-Sánchez M, Quezada-Feijoo M, Bermúdez-García A, Daroca T, Alonso-Villa E, García-Padilla C, Mangas A, Toro R. Unraveling the Etiology of Dilated Cardiomyopathy through Differential miRNA-mRNA Interactome. Biomolecules 2024; 14:524. [PMID: 38785931 PMCID: PMC11117812 DOI: 10.3390/biom14050524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 04/23/2024] [Accepted: 04/25/2024] [Indexed: 05/25/2024] Open
Abstract
Dilated cardiomyopathy (DCM) encompasses various acquired or genetic diseases sharing a common phenotype. The understanding of pathogenetic mechanisms and the determination of the functional effects of each etiology may allow for tailoring different therapeutic strategies. MicroRNAs (miRNAs) have emerged as key regulators in cardiovascular diseases, including DCM. However, their specific roles in different DCM etiologies remain elusive. Here, we applied mRNA-seq and miRNA-seq to identify the gene and miRNA signature from myocardial biopsies from four patients with DCM caused by volume overload (VCM) and four with ischemic DCM (ICM). Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis were used for differentially expressed genes (DEGs). The miRNA-mRNA interactions were identified by Pearson correlation analysis and miRNA target-prediction programs. mRNA-seq and miRNA-seq were validated by qRT-PCR and miRNA-mRNA interactions were validated by luciferase assays. We found 112 mRNAs and five miRNAs dysregulated in VCM vs. ICM. DEGs were positively enriched for pathways related to the extracellular matrix (ECM), mitochondrial respiration, cardiac muscle contraction, and fatty acid metabolism in VCM vs. ICM and negatively enriched for immune-response-related pathways, JAK-STAT, and NF-kappa B signaling. We identified four pairs of negatively correlated miRNA-mRNA: miR-218-5p-DDX6, miR-218-5p-TTC39C, miR-218-5p-SEMA4A, and miR-494-3p-SGMS2. Our study revealed novel miRNA-mRNA interaction networks and signaling pathways for VCM and ICM, providing novel insights into the development of these DCM etiologies.
Collapse
Affiliation(s)
- Fernando Bonet
- Medicine Department, School of Medicine, University of Cádiz (UCA), 11003 Cádiz, Spain; (F.B.); (E.A.-V.); (A.M.)
- Research Unit, Biomedical Research and Innovation Institute of Cádiz (INiBICA), Puerta del Mar University Hospital, 11009 Cádiz, Spain
| | - Francisco Hernandez-Torres
- Department of Biochemistry and Molecular Biology III and Immunology, Faculty of Medicine, University of Granada, 18016 Granada, Spain
| | - Mónica Ramos-Sánchez
- Cardiology Department, Central de la Cruz Roja Hospital, 28003 Madrid, Spain; (M.R.-S.); (M.Q.-F.)
- Medicine Department, School of Medicine, Alfonso X EL Sabio University, 28691 Madrid, Spain
| | - Maribel Quezada-Feijoo
- Cardiology Department, Central de la Cruz Roja Hospital, 28003 Madrid, Spain; (M.R.-S.); (M.Q.-F.)
- Medicine Department, School of Medicine, Alfonso X EL Sabio University, 28691 Madrid, Spain
| | - Aníbal Bermúdez-García
- Cardiovascular Surgery Department, Puerta del Mar University Hospital, 11009 Cádiz, Spain (T.D.)
| | - Tomás Daroca
- Cardiovascular Surgery Department, Puerta del Mar University Hospital, 11009 Cádiz, Spain (T.D.)
| | - Elena Alonso-Villa
- Medicine Department, School of Medicine, University of Cádiz (UCA), 11003 Cádiz, Spain; (F.B.); (E.A.-V.); (A.M.)
- Research Unit, Biomedical Research and Innovation Institute of Cádiz (INiBICA), Puerta del Mar University Hospital, 11009 Cádiz, Spain
| | | | - Alipio Mangas
- Medicine Department, School of Medicine, University of Cádiz (UCA), 11003 Cádiz, Spain; (F.B.); (E.A.-V.); (A.M.)
- Research Unit, Biomedical Research and Innovation Institute of Cádiz (INiBICA), Puerta del Mar University Hospital, 11009 Cádiz, Spain
- Internal Medicine Department, Puerta del Mar University Hospital, 11009 Cádiz, Spain
| | - Rocio Toro
- Medicine Department, School of Medicine, University of Cádiz (UCA), 11003 Cádiz, Spain; (F.B.); (E.A.-V.); (A.M.)
- Research Unit, Biomedical Research and Innovation Institute of Cádiz (INiBICA), Puerta del Mar University Hospital, 11009 Cádiz, Spain
| |
Collapse
|
5
|
Reimann MJ, Cremer S, Christiansen L, Ibragimov E, Gao F, Cirera S, Fredholm M, Olsen LH, Karlskov-Mortensen P. Mitral valve transcriptome analysis in thirty-four age-matched Cavalier King Charles Spaniels with or without congestive heart failure caused by myxomatous mitral valve disease. Mamm Genome 2024; 35:77-89. [PMID: 37938355 PMCID: PMC10884180 DOI: 10.1007/s00335-023-10024-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 10/08/2023] [Indexed: 11/09/2023]
Abstract
We here report the results of a mitral valve transcriptome study designed to identify genes and molecular pathways involved in development of congestive heart failure (CHF) following myxomatous mitral valve disease (MMVD) in dogs. The study is focused on a cohort of elderly age-matched dogs (n = 34, age ~ 10 years) from a single breed-Cavalier King Charles Spaniels (CKCS)-with a high incidence of MMVD. The cohort comprises 19 dogs (10♀, 9♂) without MMVD-associated CHF, and 15 dogs (6♀, 9♂) with CHF caused by MMVD; i.e., we compare gene expression in breed and age-matched groups of dogs, which only differ with respect to CHF status. We identify 56 genes, which are differentially expressed between the two groups. In this list of genes, we confirm an enrichment of genes related to the TNFβ-signaling pathway, extracellular matrix organization, vascular development, and endothelium damage, which also have been identified in previous studies. However, the genes with the greatest difference in expression between the two groups are CNTN3 and MYH1. Both genes encode proteins, which are predicted to have an effect on the contractile activity of myocardial cells, which in turn may have an effect on valvular performance and hemodynamics across the mitral valve. This may result in shear forces with impact on MMVD progression.
Collapse
Affiliation(s)
- Maria J Reimann
- Preclinical Disease Biology, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Signe Cremer
- Preclinical Disease Biology, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Liselotte Christiansen
- Preclinical Disease Biology, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Emil Ibragimov
- Animal Genetics and Breeding, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Fei Gao
- Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
- Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Susanna Cirera
- Animal Genetics and Breeding, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Merete Fredholm
- Animal Genetics and Breeding, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Lisbeth H Olsen
- Preclinical Disease Biology, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Peter Karlskov-Mortensen
- Animal Genetics and Breeding, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark.
| |
Collapse
|
6
|
Manzoor S, Kane MS, Grenett M, Oh JY, Pat B, Lewis C, Davies JE, Steele C, Patel RP, Dell'Italia LJ. Elevated cardiac hemoglobin expression is associated with a pro-oxidative and inflammatory environment in primary mitral regurgitation. Free Radic Biol Med 2023; 208:126-133. [PMID: 37543167 DOI: 10.1016/j.freeradbiomed.2023.08.007] [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: 05/24/2023] [Revised: 08/01/2023] [Accepted: 08/02/2023] [Indexed: 08/07/2023]
Abstract
BACKGROUND Primary mitral regurgitation (PMR) is associated with oxidative and inflammatory myocardial damage. We reported greater exosome hemoglobin (Hb) in pericardial fluid (PCF) versus plasma, suggesting a cardiac source of Hb. OBJECTIVE Test the hypothesis that Hb is produced in the PMR heart and is associated with increased inflammation. METHODS AND RESULTS Hb gene expression for subunits alpha (HBA) and beta (HBB) was assessed in right atria (RA), left atria (LA) and left ventricular (LV) tissue from donor hearts (n = 10) and PMR patient biopsies at surgery (n = 11). PMR patients (n = 22) had PCF and blood collected for macrophage markers, pro-inflammatory cytokines, and matrix metalloproteinases (MMPs). In-situ hybridization for HBA mRNA and immunohistochemistry for Hb-alpha (Hbα) and Hb-beta (Hbβ) protein was performed on PMR tissue. RESULTS HBA and HBB genes are significantly increased (>4-fold) in RA, LA, and LV in PMR vs. normal hearts. In PMR tissue, HBA mRNA is expressed in both LV cardiomyocytes and interstitial cells by in-situ hybridization; however, Hbα and Hbβ protein is only expressed in interstitial cells by immunohistochemistry. PCF oxyHb is significantly increased over plasma along with low ratios (<1.0) of haptoglobin:oxyHb and hemopexin:heme supporting a highly oxidative environment. Macrophage chemotactic protein-1, tumor necrosis factor-α, interleukin-6, and MMPs are significantly higher in PCF vs. plasma. CONCLUSION There is increased Hb production in the PMR heart coupled with the inflammatory state of the heart, suggests a myocardial vulnerability of further Hb delivery and/or production during cardiac surgery that could adversely affect LV functional recovery.
Collapse
Affiliation(s)
- Shajer Manzoor
- Department of Medicine, Division of Cardiology, University of Alabama at Birmingham (UAB), Birmingham, AL, USA
| | - Mariame Selma Kane
- Department of Medicine, Division of Cardiology, University of Alabama at Birmingham (UAB), Birmingham, AL, USA; Birmingham Veterans Affairs Health Care System, Birmingham, AL, USA
| | - Maximiliano Grenett
- Department of Medicine, Division of Cardiology, University of Alabama at Birmingham (UAB), Birmingham, AL, USA; Birmingham Veterans Affairs Health Care System, Birmingham, AL, USA
| | - Joo-Yeun Oh
- Department of Medicine, Division of Cardiology, University of Alabama at Birmingham (UAB), Birmingham, AL, USA; Birmingham Veterans Affairs Health Care System, Birmingham, AL, USA
| | - Betty Pat
- Department of Medicine, Division of Cardiology, University of Alabama at Birmingham (UAB), Birmingham, AL, USA; Birmingham Veterans Affairs Health Care System, Birmingham, AL, USA
| | - Clifton Lewis
- Department of Surgery, Division of Thoracic and Cardiovascular Surgery, UAB, USA
| | - James E Davies
- Department of Surgery, Division of Thoracic and Cardiovascular Surgery, UAB, USA
| | - Chad Steele
- Department of Microbiology and Immunology, Tulane University, New Orleans, LA, USA
| | - Rakesh P Patel
- Department of Pathology and Center for Free Radical Biology, UAB, USA
| | - Louis J Dell'Italia
- Department of Medicine, Division of Cardiology, University of Alabama at Birmingham (UAB), Birmingham, AL, USA; Birmingham Veterans Affairs Health Care System, Birmingham, AL, USA.
| |
Collapse
|
7
|
Ahmed MI, Andrikopoulou E, Zheng J, Ulasova E, Pat B, Kelley EE, Powell PC, Denney TS, Lewis C, Davies JE, Darley-Usmar V, Dell’Italia LJ. Interstitial Collagen Loss, Myocardial Remodeling, and Function in Primary Mitral Regurgitation. JACC Basic Transl Sci 2022; 7:973-981. [PMID: 36337921 PMCID: PMC9626893 DOI: 10.1016/j.jacbts.2022.04.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 04/27/2022] [Accepted: 04/27/2022] [Indexed: 11/12/2022]
Abstract
Interstitial collagen loss and cardiomyocyte ultrastructural damage accounts for left ventricular (LV) sphericity and decrease in LV twist and circumferential strain. Normal LV diastolic function belies significantly abnormal left atrial (LA) function and early LV diastolic untwist rate. This underscores the complex interplay of LV and LA myocardial remodeling and function in the pathophysiology of primary mitral regurgitation. In this study, we connect LA function with LV systolic and diastolic myocardial remodeling and function using cardiac magnetic resonance tissue tagging in primary mitral regurgitation.
Collapse
Key Words
- BNP, brain natriuretic peptide
- CMR, cardiac magnetic resonance
- ED, end diastole
- ES, end systole
- ICTP, carboxy-terminal telopeptide of collagen type I
- LA, left atrial
- LV, left ventricle
- LVEF, LV ejection fraction
- PICP, carboxy-terminal propeptide of procollagen type I
- PMR, primary mitral regurgitation
- RV, right ventricle
- SV, stroke volume
- XO, xanthine oxidase
- cardiac magnetic resonance
- collagen loss
- left ventricular remodeling
- primary mitral regurgitation
Collapse
Affiliation(s)
- Mustafa I. Ahmed
- Division of Cardiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | | | - Jingyi Zheng
- Department of Mathematics and Statistics, Auburn University, Auburn, Alabama, USA
| | - Elena Ulasova
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Betty Pat
- Division of Cardiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
- Birmingham Veterans Affairs Health Care System, Birmingham, Alabama, USA
| | - Eric E. Kelley
- Department of Physiology and Pharmacology, West Virginia University, Morgantown, West Virginia, USA
| | - Pamela Cox Powell
- Division of Cardiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
- Birmingham Veterans Affairs Health Care System, Birmingham, Alabama, USA
| | - Thomas S. Denney
- Samuel Ginn College of Engineering, Auburn University, Auburn, Alabama, USA
| | - Clifton Lewis
- Division of Thoracic and Cardiovascular Surgery, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - James E. Davies
- Division of Thoracic and Cardiovascular Surgery, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Victor Darley-Usmar
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Louis J. Dell’Italia
- Division of Cardiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
- Birmingham Veterans Affairs Health Care System, Birmingham, Alabama, USA
| |
Collapse
|
8
|
Fragasso G, Sanvito F, Monaca G, Ardizzone V, De Bonis M, Pappalardo F, Smart C, Montanaro C, Lapenna E, Calabrese MC, Castiglioni A, Benussi S, Maisano F, Zangrillo A, Ambrosi A, Doglioni C, Alfieri O, Margonato A. Myocardial fibrosis in asymptomatic patients undergoing surgery for mitral and aortic valve regurgitation. J Cardiovasc Med (Hagerstown) 2022; 23:505-512. [PMID: 35904996 DOI: 10.2459/jcm.0000000000001347] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Chronic heart valve regurgitation induces left ventricular (LV) volume overload, leading to the development of hypertrophy and progressive dilatation of the ventricle to maintain physiological cardiac output. In order to prevent potential irreversible LV structural changes, the identification of the best timing for treatment is pivotal. OBJECTIVE To assess the presence and extent of fibrosis in myocardial tissue in asymptomatic patients with valvular heart disease (VHD) and preserved LV dimensions and function undergoing cardiac surgery. METHODS Thirty-nine patients were enrolled. Sixteen patients were affected by aortic or mitral regurgitation: they were all asymptomatic, undergoing valve surgery according to VHD European Society of Cardiology guidelines. Twenty-three patients with end-stage nonischemic dilated cardiomyopathy (DCM) and severe LV dysfunction undergoing cardiac surgery for implantation of a durable left ventricular assist device (LVAD) served as controls. During surgery, VHD patients underwent three myocardial biopsies at the level of the septum, the lateral wall and LV apex, while in LVAD patients the coring of the apex of the LV was used. For both groups, the tissue samples were analyzed on one section corresponding to the apical area. All slides were stained with hematoxylin and eosin and Masson's trichrome staining and further digitalized. The degree of fibrosis was then calculated as a percentage of the total area. RESULTS Of 39 patients, 23 met the inclusion criteria: 12 had mitral or aortic insufficiency with a preserved ejection fraction and 11 had idiopathic dilated cardiomyopathy. Quantitative analysis of apical sections revealed a myocardial fibrosis amount of 10 ± 6% in VHD patients, while in LVAD patients the mean apical myocardial fibrosis rate was 38 ± 9%. In VHD patients, fibrosis was also present in the lateral wall (9 ± 4%) and in the septum (9 ± 6%). CONCLUSION Our case series study highlights the presence of tissue remodeling with fibrosis in asymptomatic patients with VHD and preserved LV function. According to our results, myocardial fibrosis is present at an early stage of the disease, well before developing detectable LV dysfunction and symptoms. Since the relationship between the progressive magnitude of myocardial fibrosis and potential prognostic implications are not yet defined, further studies on this topic are warranted.
Collapse
Affiliation(s)
| | | | | | | | | | - Federico Pappalardo
- Department of Anesthesia and Intensive Care, IRCCS San Raffaele Scientific Institute, Milano
- Department of Cardiothoracic and Vascular Anesthesia and Intensive Care, AO SS Antonio e Biagio e Cesare Arrigo, Alessandria
| | - Chanel Smart
- Pathology Unit, Division of Experimental Oncology
| | - Claudia Montanaro
- Department of Clinical Cardiology
- Department of Cardiology, Royal Brompton Hospital, London, United Kingdom
| | | | | | | | - Stefano Benussi
- Department of Cardiac Surgery
- Department of Cardiac Surgery, ASST degli Spedali Civili di Brescia
| | | | - Alberto Zangrillo
- Department of Anesthesia and Intensive Care, IRCCS San Raffaele Scientific Institute, Milano
| | - Alessandro Ambrosi
- Department of Biostatistics, University Vita/Salute San Raffaele, Milano, Italy
| | | | | | | |
Collapse
|
9
|
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
|
10
|
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
|
11
|
O'Brien MJ, Beijerink NJ, Wade CM. Genetics of canine myxomatous mitral valve disease. Anim Genet 2021; 52:409-421. [PMID: 34028063 DOI: 10.1111/age.13082] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/03/2021] [Indexed: 12/26/2022]
Abstract
Myxomatous mitral valve disease (MMVD) is the most common heart disease and cause of cardiac death in domestic dogs. MMVD is characterised by slow progressive myxomatous degeneration from the tips of the mitral valves onwards with subsequent mitral valve regurgitation, and left atrial and ventricular dilatation. Although the disease usually has a long asymptomatic period, in dogs with severe disease, mortality is typically secondary to left-sided congestive heart failure. Although it is not uncommon for dogs to survive long enough in the asymptomatic period to die from unrelated causes; a proportion of dogs rapidly advance into congestive heart failure. Heightened prevalence in certain breeds, such as the Cavalier King Charles Spaniel, has indicated that MMVD is under a genetic influence. The genetic characterisation of the factors that underlie the difference in progression of disease is of strong interest to those concerned with dog longevity and welfare. Advanced genomic technologies have the potential to provide information that may impact treatment, prevalence, or severity of MMVD through the elucidation of pathogenic mechanisms and the detection of predisposing genetic loci of major effect. Here we describe briefly the clinical nature of the disorder and consider the physiological mechanisms that might impact its occurrence in the domestic dog. Using results from comparative genomics we suggest possible genetic approaches for identifying genetic risk factors within breeds. The Cavalier King Charles Spaniel breed represents a robust resource for uncovering the genetic basis of MMVD.
Collapse
Affiliation(s)
- M J O'Brien
- School of Life and Environmental Sciences, University of Sydney, Sydney, NSW, 2006, Australia
| | - N J Beijerink
- Sydney School of Veterinary Science, Faculty of Science, University of Sydney, Sydney, NSW, 2006, Australia.,Veterinaire Specialisten Vught, Reutsedijk 8a, Vught, 5264 PC, The Netherlands
| | - C M Wade
- School of Life and Environmental Sciences, University of Sydney, Sydney, NSW, 2006, Australia
| |
Collapse
|
12
|
Lin Y, Chen C, Shih J, Cheng B, Chang C, Lin M, Ho C, Chen Z, Fisch S, Chang W. Dapagliflozin Improves Cardiac Hemodynamics and Mitigates Arrhythmogenesis in Mitral Regurgitation-Induced Myocardial Dysfunction. J Am Heart Assoc 2021; 10:e019274. [PMID: 33749310 PMCID: PMC8174384 DOI: 10.1161/jaha.120.019274] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Background Mitral regurgitation (MR) is a major contributor for heart failure (HF) and atrial fibrillation. Despite the advancement of MR surgeries, an effective medical therapy to mitigate MR progression is lacking. Sodium glucose cotransporter 2 inhibitors, a new class of antidiabetic drugs, has shown measurable benefits in reduction of HF hospitalization and cardiovascular mortality but the mechanism is unclear. We hypothesized that dapagliflozin (DAPA), a sodium glucose cotransporter 2 inhibitor, can improve cardiac hemodynamics in MR‐induced HF. Methods and Results Using a novel, mini‐invasive technique, we established a MR model in rats, in which MR induced left heart dilatation and functional decline. Half of the rats were randomized to be administered with DAPA at 10 mg/kg per day for 6 weeks. After evaluation of electrocardiography and echocardiography, hemodynamic studies were performed, followed by postmortem tissue analyses. Results showed that DAPA partially rescued MR‐induced impairment including partial restoration of left ventricular ejection fraction and end‐systolic pressure volume relationship. Despite no significant changes in electrocardiography at rest, rats treated with DAPA exhibited lower inducibility and decreased duration of pacing‐induced atrial fibrillation. DAPA also significantly attenuated cardiac fibrosis, cardiac expression of apoptosis, and endoplasmic reticulum stress‐associated proteins. Conclusions DAPA was able to suppress cardiac fibrosis and endoplasmic reticulum stress and improve hemodynamics in an MR‐induced HF rat model. The demonstrated DAPA effect on the heart and its association with key molecular contributors in eliciting its cardio‐protective function, provides a plausible point of DAPA as a potential strategy for MR‐induced HF.
Collapse
Affiliation(s)
- Yu‐Wen Lin
- Division of CardiologyDepartment of Internal MedicineChi‐Mei Medical CenterTainanTaiwan
| | - Chin‐Yu Chen
- Department of RadiologyChi‐Mei Medical CenterTainanTaiwan
| | - Jhih‐Yuan Shih
- Division of CardiologyDepartment of Internal MedicineChi‐Mei Medical CenterTainanTaiwan
| | - Bor‐Chih Cheng
- Division of Cardiovascular SurgeryChi‐Mei Medical CenterTainanTaiwan
- Department of BiotechnologySouthern Taiwan University of Science and TechnologyTainanTaiwan
| | - Ching‐Ping Chang
- Department of Medical ResearchChi Mei Medical CenterTainanTaiwan
| | - Mao‐Tsun Lin
- Department of Medical ResearchChi Mei Medical CenterTainanTaiwan
| | - Chung‐Han Ho
- Department of Hospital and Health Care AdministrationChi‐Mei Medical CenterTainanTaiwan
| | - Zhih‐Cherng Chen
- Division of CardiologyDepartment of Internal MedicineChi‐Mei Medical CenterTainanTaiwan
| | - Sudeshna Fisch
- Department of MedicineBrigham and Women's HospitalHarvard Medical SchoolBostonMA
| | - Wei‐Ting Chang
- Division of CardiologyDepartment of Internal MedicineChi‐Mei Medical CenterTainanTaiwan
- Department of BiotechnologySouthern Taiwan University of Science and TechnologyTainanTaiwan
- Institute of Clinical MedicineCollege of MedicineNational Cheng Kung UniversityTainanTaiwan
| |
Collapse
|
13
|
Corporan D, Onohara D, Amedi A, Saadeh M, Guyton RA, Kumar S, Padala M. Hemodynamic and transcriptomic studies suggest early left ventricular dysfunction in a preclinical model of severe mitral regurgitation. J Thorac Cardiovasc Surg 2021; 161:961-976.e22. [PMID: 33277035 PMCID: PMC7889661 DOI: 10.1016/j.jtcvs.2020.08.119] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 08/06/2020] [Accepted: 08/24/2020] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Primary mitral regurgitation is a valvular lesion in which the left ventricular ejection fraction remains preserved for long periods, delaying a clinical trigger for mitral valve intervention. In this study, we sought to investigate whether adverse left ventricular remodeling occurs before a significant fall in ejection fraction and characterize these changes. METHODS Sixty-five rats were induced with severe mitral regurgitation by puncturing the mitral valve leaflet with a 23-G needle using ultrasound guidance. Rats underwent longitudinal cardiac echocardiography at biweekly intervals and hearts explanted at 2 weeks (n = 15), 10 weeks (n = 15), 20 weeks (n = 15), and 40 weeks (n = 15). Sixty age- and weight-matched healthy rats were used as controls. Unbiased RNA-sequencing was performed at each terminal point. RESULTS Regurgitant fraction was 40.99 ± 9.40%, with pulmonary flow reversal in the experimental group, and none in the control group. Significant fall in ejection fraction occurred at 14 weeks after mitral regurgitation induction. However, before 14 weeks, end-diastolic volume increased by 93.69 ± 52.38% (P < .0001 compared with baseline), end-systolic volume increased by 118.33 ± 47.54% (P < .0001 compared with baseline), and several load-independent pump function indices were reduced. Transcriptomic data at 2 and 10 weeks before fall in ejection fraction indicated up-regulation of myocyte remodeling and oxidative stress pathways, whereas those at 20 and 40 weeks indicated extracellular matrix remodeling. CONCLUSIONS In this rodent model of mitral regurgitation, left ventricular ejection fraction was preserved for a long duration, yet rapid and severe left ventricular dilatation, and biological remodeling occurred before a clinically significant fall in ejection fraction.
Collapse
Affiliation(s)
- Daniella Corporan
- Structural Heart Research and Innovation Laboratory, Carlyle Fraser Heart Center at Emory University Hospital Midtown, Atlanta, Ga
| | - Daisuke Onohara
- Structural Heart Research and Innovation Laboratory, Carlyle Fraser Heart Center at Emory University Hospital Midtown, Atlanta, Ga
| | - Alan Amedi
- Structural Heart Research and Innovation Laboratory, Carlyle Fraser Heart Center at Emory University Hospital Midtown, Atlanta, Ga
| | - Maher Saadeh
- Structural Heart Research and Innovation Laboratory, Carlyle Fraser Heart Center at Emory University Hospital Midtown, Atlanta, Ga
| | - Robert A Guyton
- Structural Heart Research and Innovation Laboratory, Carlyle Fraser Heart Center at Emory University Hospital Midtown, Atlanta, Ga; Division of Cardiothoracic Surgery, Department of Surgery, Emory University School of Medicine, Atlanta, Ga
| | - Sandeep Kumar
- Department of Biomedical Engineering, Emory University School of Medicine, Atlanta, Ga
| | - Muralidhar Padala
- Structural Heart Research and Innovation Laboratory, Carlyle Fraser Heart Center at Emory University Hospital Midtown, Atlanta, Ga; Division of Cardiothoracic Surgery, Department of Surgery, Emory University School of Medicine, Atlanta, Ga.
| |
Collapse
|
14
|
Abstract
Myocardial fibrosis, the expansion of the cardiac interstitium through deposition of extracellular matrix proteins, is a common pathophysiologic companion of many different myocardial conditions. Fibrosis may reflect activation of reparative or maladaptive processes. Activated fibroblasts and myofibroblasts are the central cellular effectors in cardiac fibrosis, serving as the main source of matrix proteins. Immune cells, vascular cells and cardiomyocytes may also acquire a fibrogenic phenotype under conditions of stress, activating fibroblast populations. Fibrogenic growth factors (such as transforming growth factor-β and platelet-derived growth factors), cytokines [including tumour necrosis factor-α, interleukin (IL)-1, IL-6, IL-10, and IL-4], and neurohumoral pathways trigger fibrogenic signalling cascades through binding to surface receptors, and activation of downstream signalling cascades. In addition, matricellular macromolecules are deposited in the remodelling myocardium and regulate matrix assembly, while modulating signal transduction cascades and protease or growth factor activity. Cardiac fibroblasts can also sense mechanical stress through mechanosensitive receptors, ion channels and integrins, activating intracellular fibrogenic cascades that contribute to fibrosis in response to pressure overload. Although subpopulations of fibroblast-like cells may exert important protective actions in both reparative and interstitial/perivascular fibrosis, ultimately fibrotic changes perturb systolic and diastolic function, and may play an important role in the pathogenesis of arrhythmias. This review article discusses the molecular mechanisms involved in the pathogenesis of cardiac fibrosis in various myocardial diseases, including myocardial infarction, heart failure with reduced or preserved ejection fraction, genetic cardiomyopathies, and diabetic heart disease. Development of fibrosis-targeting therapies for patients with myocardial diseases will require not only understanding of the functional pluralism of cardiac fibroblasts and dissection of the molecular basis for fibrotic remodelling, but also appreciation of the pathophysiologic heterogeneity of fibrosis-associated myocardial disease.
Collapse
Affiliation(s)
- Nikolaos G Frangogiannis
- Department of Medicine (Cardiology), The Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, 1300 Morris Park Avenue Forchheimer G46B, Bronx, NY 10461, USA
| |
Collapse
|
15
|
Tsai FC, Chang GJ, Lai YJ, Chang SH, Chen WJ, Yeh YH. Ubiquitin Pathway Is Associated with Worsening Left Ventricle Function after Mitral Valve Repair: A Global Gene Expression Study. Int J Mol Sci 2020; 21:ijms21145073. [PMID: 32708358 PMCID: PMC7404186 DOI: 10.3390/ijms21145073] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 07/13/2020] [Accepted: 07/15/2020] [Indexed: 12/25/2022] Open
Abstract
The molecular mechanism for worsening left ventricular (LV) function after mitral valve (MV) repair for chronic mitral regurgitation remains unknown. We wished to assess the LV transcriptome and identify determinants associated with worsening LV function post-MV repair. A total of 13 patients who underwent MV repair for chronic primary mitral regurgitation were divided into two groups, preserved LV function (N = 8) and worsening LV function (N = 5), for the study. Specimens of LV from the patients taken during surgery were used for the gene microarray study. Cardiomyocyte cell line HL-1 cells were transfected with gene-containing plasmids and further evaluated for mRNA and protein expression, apoptosis, and contractile protein degradation. Of 67,258 expressed sequence tags, microarrays identified 718 genes to be differentially expressed between preserved-LVF and worsening-LVF, including genes related to the protein ubiquitination pathway, bone morphogenetic protein (BMP) receptors, and regulation of eIF4 and p70S6K signaling. In addition, worsening-LVF was associated with altered expressions of genes pathologically relevant to heart failure, such asdownregulated apelin receptors and upregulated peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PPARGC1A). HL-1 cardiomyocyte cells transfected with ubiquitination-related genes demonstrated activation of the protein ubiquitination pathwaywith an increase in the ubiquitin activating enzyme E1 (UAE-E1). It also led to increased apoptosis, downregulated and ubiquitinated X-linked inhibitor of apoptosis protein (XIAP), and reduced cell viability. Overexpression of ubiquitination-related genes also resulted in degradation and increased ubiquitination of α-smooth muscle actin (SMA). In conclusion, worsening-LVF presented differential gene expression profiles from preserved-LVF after MV repair. Upregulation of protein ubiquitination-related genes associated with worsening-LVF after MV repair may exert adverse effects on LV through increased apoptosis and contractile protein degradation.
Collapse
Affiliation(s)
- Feng-Chun Tsai
- Division of Cardiovascular and Thoracic Surgery, Chang-Gung Memorial Hospital, Taoyuan 333, Taiwan;
- College of Medicine, Chang-Gung University, Taoyuan 333, Taiwan; (S.-H.C.); (W.-J.C.)
| | - Gwo-Jyh Chang
- Graduate Institute of Clinical Medical Sciences, Chang-Gung University, Taoyuan 333, Taiwan;
| | - Ying-Ju Lai
- Department of Respiratory Therapy, Chang-Gung University College of Medicine, Taoyuan 333, Taiwan;
| | - Shang-Hung Chang
- College of Medicine, Chang-Gung University, Taoyuan 333, Taiwan; (S.-H.C.); (W.-J.C.)
- Cardiovascular Department, Chang-Gung Memorial Hospital, Taoyuan 333, Taiwan
| | - Wei-Jan Chen
- College of Medicine, Chang-Gung University, Taoyuan 333, Taiwan; (S.-H.C.); (W.-J.C.)
- Cardiovascular Department, Chang-Gung Memorial Hospital, Taoyuan 333, Taiwan
| | - Yung-Hsin Yeh
- College of Medicine, Chang-Gung University, Taoyuan 333, Taiwan; (S.-H.C.); (W.-J.C.)
- Cardiovascular Department, Chang-Gung Memorial Hospital, Taoyuan 333, Taiwan
- Correspondence: ; Tel./Fax: +886-3-3271192
| |
Collapse
|
16
|
Liu W, Wang Z. Current Understanding of the Biomechanics of Ventricular Tissues in Heart Failure. Bioengineering (Basel) 2019; 7:E2. [PMID: 31861916 PMCID: PMC7175293 DOI: 10.3390/bioengineering7010002] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 12/17/2019] [Accepted: 12/18/2019] [Indexed: 12/17/2022] Open
Abstract
Heart failure is the leading cause of death worldwide, and the most common cause of heart failure is ventricular dysfunction. It is well known that the ventricles are anisotropic and viscoelastic tissues and their mechanical properties change in diseased states. The tissue mechanical behavior is an important determinant of the function of ventricles. The aim of this paper is to review the current understanding of the biomechanics of ventricular tissues as well as the clinical significance. We present the common methods of the mechanical measurement of ventricles, the known ventricular mechanical properties including the viscoelasticity of the tissue, the existing computational models, and the clinical relevance of the ventricular mechanical properties. Lastly, we suggest some future research directions to elucidate the roles of the ventricular biomechanics in the ventricular dysfunction to inspire new therapies for heart failure patients.
Collapse
Affiliation(s)
- Wenqiang Liu
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO 80523, USA;
| | - Zhijie Wang
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO 80523, USA;
- Department of Mechanical Engineering, Colorado State University, Fort Collins, CO 80523, USA
| |
Collapse
|
17
|
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
|
18
|
Humeres C, Frangogiannis NG. Fibroblasts in the Infarcted, Remodeling, and Failing Heart. JACC Basic Transl Sci 2019; 4:449-467. [PMID: 31312768 PMCID: PMC6610002 DOI: 10.1016/j.jacbts.2019.02.006] [Citation(s) in RCA: 222] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 02/15/2019] [Accepted: 02/19/2019] [Indexed: 02/07/2023]
Abstract
Expansion and activation of fibroblasts following cardiac injury is important for repair but may also contribute to fibrosis, remodeling, and dysfunction. The authors discuss the dynamic alterations of fibroblasts in failing and remodeling myocardium. Emerging concepts suggest that fibroblasts are not unidimensional cells that act exclusively by secreting extracellular matrix proteins, thus promoting fibrosis and diastolic dysfunction. In addition to their involvement in extracellular matrix expansion, activated fibroblasts may also exert protective actions, preserving the cardiac extracellular matrix, transducing survival signals to cardiomyocytes, and regulating inflammation and angiogenesis. The functional diversity of cardiac fibroblasts may reflect their phenotypic heterogeneity.
Collapse
Key Words
- AT1, angiotensin type 1
- ECM, extracellular matrix
- FAK, focal adhesion kinase
- FGF, fibroblast growth factor
- IL, interleukin
- MAPK, mitogen-activated protein kinase
- MRTF, myocardin-related transcription factor
- PDGF, platelet-derived growth factor
- RNA, ribonucleic acid
- ROCK, Rho-associated coiled-coil containing kinase
- ROS, reactive oxygen species
- SMA, smooth muscle actin
- TGF, transforming growth factor
- TRP, transient receptor potential
- cytokines
- extracellular matrix
- fibroblast
- infarction
- lncRNA, long noncoding ribonucleic acid
- miRNA, micro–ribonucleic acid
- remodeling
Collapse
Affiliation(s)
- Claudio Humeres
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx, New York
| | - Nikolaos G Frangogiannis
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx, New York
| |
Collapse
|
19
|
Abstract
The ECM (extracellular matrix) network plays a crucial role in cardiac homeostasis, not only by providing structural support, but also by facilitating force transmission, and by transducing key signals to cardiomyocytes, vascular cells, and interstitial cells. Changes in the profile and biochemistry of the ECM may be critically implicated in the pathogenesis of both heart failure with reduced ejection fraction and heart failure with preserved ejection fraction. The patterns of molecular and biochemical ECM alterations in failing hearts are dependent on the type of underlying injury. Pressure overload triggers early activation of a matrix-synthetic program in cardiac fibroblasts, inducing myofibroblast conversion, and stimulating synthesis of both structural and matricellular ECM proteins. Expansion of the cardiac ECM may increase myocardial stiffness promoting diastolic dysfunction. Cardiomyocytes, vascular cells and immune cells, activated through mechanosensitive pathways or neurohumoral mediators may play a critical role in fibroblast activation through secretion of cytokines and growth factors. Sustained pressure overload leads to dilative remodeling and systolic dysfunction that may be mediated by changes in the interstitial protease/antiprotease balance. On the other hand, ischemic injury causes dynamic changes in the cardiac ECM that contribute to regulation of inflammation and repair and may mediate adverse cardiac remodeling. In other pathophysiologic conditions, such as volume overload, diabetes mellitus, and obesity, the cell biological effectors mediating ECM remodeling are poorly understood and the molecular links between the primary insult and the changes in the matrix environment are unknown. This review article discusses the role of ECM macromolecules in heart failure, focusing on both structural ECM proteins (such as fibrillar and nonfibrillar collagens), and specialized injury-associated matrix macromolecules (such as fibronectin and matricellular proteins). Understanding the role of the ECM in heart failure may identify therapeutic targets to reduce geometric remodeling, to attenuate cardiomyocyte dysfunction, and even to promote myocardial regeneration.
Collapse
Affiliation(s)
- Nikolaos G Frangogiannis
- From the Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx, NY
| |
Collapse
|
20
|
Dell'Italia LJ, Collawn JF, Ferrario CM. Multifunctional Role of Chymase in Acute and Chronic Tissue Injury and Remodeling. Circ Res 2019; 122:319-336. [PMID: 29348253 DOI: 10.1161/circresaha.117.310978] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Chymase is the most efficient Ang II (angiotensin II)-forming enzyme in the human body and has been implicated in a wide variety of human diseases that also implicate its many other protease actions. Largely thought to be the product of mast cells, the identification of other cellular sources including cardiac fibroblasts and vascular endothelial cells demonstrates a more widely dispersed production and distribution system in various tissues. Furthermore, newly emerging evidence for its intracellular presence in cardiomyocytes and smooth muscle cells opens an entirely new compartment of chymase-mediated actions that were previously thought to be limited to the extracellular space. This review illustrates how these multiple chymase-mediated mechanisms of action can explain the residual risk in clinical trials of cardiovascular disease using conventional renin-angiotensin system blockade.
Collapse
Affiliation(s)
- Louis J Dell'Italia
- From the Department of Medicine, Division of Cardiology, Birmingham Veteran Affairs Medical Center (L.J.D.), Division of Cardiovascular Disease, Department of Medicine (L.J.D.), and Department of Cell, Developmental and Integrative Biology (J.F.C.), University of Alabama at Birmingham; and Division of Surgical Sciences, Wake Forest University School of Medicine, Winston-Salem, NC (C.M.F.).
| | - James F Collawn
- From the Department of Medicine, Division of Cardiology, Birmingham Veteran Affairs Medical Center (L.J.D.), Division of Cardiovascular Disease, Department of Medicine (L.J.D.), and Department of Cell, Developmental and Integrative Biology (J.F.C.), University of Alabama at Birmingham; and Division of Surgical Sciences, Wake Forest University School of Medicine, Winston-Salem, NC (C.M.F.)
| | - Carlos M Ferrario
- From the Department of Medicine, Division of Cardiology, Birmingham Veteran Affairs Medical Center (L.J.D.), Division of Cardiovascular Disease, Department of Medicine (L.J.D.), and Department of Cell, Developmental and Integrative Biology (J.F.C.), University of Alabama at Birmingham; and Division of Surgical Sciences, Wake Forest University School of Medicine, Winston-Salem, NC (C.M.F.)
| |
Collapse
|
21
|
Saucerman JJ, Tan PM, Buchholz KS, McCulloch AD, Omens JH. Mechanical regulation of gene expression in cardiac myocytes and fibroblasts. Nat Rev Cardiol 2019; 16:361-378. [PMID: 30683889 PMCID: PMC6525041 DOI: 10.1038/s41569-019-0155-8] [Citation(s) in RCA: 128] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The intact heart undergoes complex and multiscale remodelling processes in response to altered mechanical cues. Remodelling of the myocardium is regulated by a combination of myocyte and non-myocyte responses to mechanosensitive pathways, which can alter gene expression and therefore function in these cells. Cellular mechanotransduction and its downstream effects on gene expression are initially compensatory mechanisms during adaptations to the altered mechanical environment, but under prolonged and abnormal loading conditions, they can become maladaptive, leading to impaired function and cardiac pathologies. In this Review, we summarize mechanoregulated pathways in cardiac myocytes and fibroblasts that lead to altered gene expression and cell remodelling under physiological and pathophysiological conditions. Developments in systems modelling of the networks that regulate gene expression in response to mechanical stimuli should improve integrative understanding of their roles in vivo and help to discover new combinations of drugs and device therapies targeting mechanosignalling in heart disease.
Collapse
Affiliation(s)
- Jeffrey J Saucerman
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Philip M Tan
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Kyle S Buchholz
- Departments of Bioengineering and Medicine, University of California San Diego, La Jolla, CA, USA
| | - Andrew D McCulloch
- Departments of Bioengineering and Medicine, University of California San Diego, La Jolla, CA, USA.
| | - Jeffrey H Omens
- Departments of Bioengineering and Medicine, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
22
|
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
|
23
|
Xiao X, Song D, Cheng Y, Hu Y, Wang F, Lu Z, Wang Y. Biogenic nanoselenium particles activate Nrf2‐ARE pathway by phosphorylating p38, ERK1/2, and AKT on IPEC‐J2 cells. J Cell Physiol 2018; 234:11227-11234. [DOI: 10.1002/jcp.27773] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 10/30/2018] [Indexed: 01/28/2023]
Affiliation(s)
- Xiao Xiao
- National Engineering Laboratory of Biological Feed Safety and Pollution Prevention and Control, Key Laboratory of Animal Nutrition and Feed Science in Eastern China, Zhejiang University Hangzhou China
- Key Laboratory of Feed and Animal Nutrition of Zhejiang Province, Ministry of Agriculture, Institute of Feed Science, Zhejiang University Hangzhou China
| | - Deguang Song
- National Engineering Laboratory of Biological Feed Safety and Pollution Prevention and Control, Key Laboratory of Animal Nutrition and Feed Science in Eastern China, Zhejiang University Hangzhou China
- Key Laboratory of Feed and Animal Nutrition of Zhejiang Province, Ministry of Agriculture, Institute of Feed Science, Zhejiang University Hangzhou China
| | - Yuanzhi Cheng
- National Engineering Laboratory of Biological Feed Safety and Pollution Prevention and Control, Key Laboratory of Animal Nutrition and Feed Science in Eastern China, Zhejiang University Hangzhou China
- Key Laboratory of Feed and Animal Nutrition of Zhejiang Province, Ministry of Agriculture, Institute of Feed Science, Zhejiang University Hangzhou China
| | - Yuhan Hu
- National Engineering Laboratory of Biological Feed Safety and Pollution Prevention and Control, Key Laboratory of Animal Nutrition and Feed Science in Eastern China, Zhejiang University Hangzhou China
- Key Laboratory of Feed and Animal Nutrition of Zhejiang Province, Ministry of Agriculture, Institute of Feed Science, Zhejiang University Hangzhou China
| | - Fengqin Wang
- National Engineering Laboratory of Biological Feed Safety and Pollution Prevention and Control, Key Laboratory of Animal Nutrition and Feed Science in Eastern China, Zhejiang University Hangzhou China
- Key Laboratory of Feed and Animal Nutrition of Zhejiang Province, Ministry of Agriculture, Institute of Feed Science, Zhejiang University Hangzhou China
| | - Zeqing Lu
- National Engineering Laboratory of Biological Feed Safety and Pollution Prevention and Control, Key Laboratory of Animal Nutrition and Feed Science in Eastern China, Zhejiang University Hangzhou China
- Key Laboratory of Feed and Animal Nutrition of Zhejiang Province, Ministry of Agriculture, Institute of Feed Science, Zhejiang University Hangzhou China
| | - Yizhen Wang
- National Engineering Laboratory of Biological Feed Safety and Pollution Prevention and Control, Key Laboratory of Animal Nutrition and Feed Science in Eastern China, Zhejiang University Hangzhou China
- Key Laboratory of Feed and Animal Nutrition of Zhejiang Province, Ministry of Agriculture, Institute of Feed Science, Zhejiang University Hangzhou China
| |
Collapse
|
24
|
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
|
25
|
Roussel E, Drolet MC, Lavigne AM, Arsenault M, Couet J. Multiple short-chain dehydrogenases/reductases are regulated in pathological cardiac hypertrophy. FEBS Open Bio 2018; 8:1624-1635. [PMID: 30338214 PMCID: PMC6168690 DOI: 10.1002/2211-5463.12506] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 07/04/2018] [Accepted: 07/23/2018] [Indexed: 12/18/2022] Open
Abstract
Cardiac hypertrophy (CH) is an important and independent predictor of morbidity and mortality. Through expression profiling, we recently identified a subset of genes (Dhrs7c, Decr, Dhrs11, Dhrs4, Hsd11b1, Hsd17b10, Hsd17b8, Blvrb, Pecr), all of which are members of the short‐chain dehydrogenase/reductase (SDR) superfamily and are highly expressed in the heart, that were significantly dysregulated in a rat model of CH caused by severe aortic valve regurgitation (AR). Here, we studied their expression in various models of CH, as well as factors influencing their regulation. Among the nine SDR genes studied, all but Hsd11b1 were down‐regulated in CH models (AR rats or mice infused with either isoproterenol or angiotensin II). This regulation showed a clear sex dimorphism, being more evident in males than in females irrespective of CH levels. In neonatal rat cardiomyocytes, we observed that treatment with the α1‐adrenergic receptor agonist phenylephrine mostly reproduced the observations made in CH animals models. Retinoic acid, on the other hand, stimulated the expression of most of the SDR genes studied, suggesting that their expression may be related to cardiomyocyte differentiation. Indeed, levels of expression were found to be higher in the hearts of adult animals than in neonatal cardiomyocytes. In conclusion, we identified a group of genes modulated in animal models of CH and mostly in males. This could be related to the activation of the fetal gene expression program in pathological CH situations, in which these highly expressed genes are down‐regulated in the adult heart.
Collapse
Affiliation(s)
- Elise Roussel
- Groupe de recherche sur les valvulopathies Centre de Recherche Institut universitaire de cardiologie et de pneumologie de Québec Université Laval Quebec City Canada
| | - Marie-Claude Drolet
- Groupe de recherche sur les valvulopathies Centre de Recherche Institut universitaire de cardiologie et de pneumologie de Québec Université Laval Quebec City Canada
| | - Anne-Marie Lavigne
- Groupe de recherche sur les valvulopathies Centre de Recherche Institut universitaire de cardiologie et de pneumologie de Québec Université Laval Quebec City Canada
| | - Marie Arsenault
- Groupe de recherche sur les valvulopathies Centre de Recherche Institut universitaire de cardiologie et de pneumologie de Québec Université Laval Quebec City Canada
| | - Jacques Couet
- Groupe de recherche sur les valvulopathies Centre de Recherche Institut universitaire de cardiologie et de pneumologie de Québec Université Laval Quebec City Canada
| |
Collapse
|
26
|
Frangogiannis NG. Cardiac fibrosis: Cell biological mechanisms, molecular pathways and therapeutic opportunities. Mol Aspects Med 2018; 65:70-99. [PMID: 30056242 DOI: 10.1016/j.mam.2018.07.001] [Citation(s) in RCA: 533] [Impact Index Per Article: 76.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Accepted: 07/23/2018] [Indexed: 12/13/2022]
Abstract
Cardiac fibrosis is a common pathophysiologic companion of most myocardial diseases, and is associated with systolic and diastolic dysfunction, arrhythmogenesis, and adverse outcome. Because the adult mammalian heart has negligible regenerative capacity, death of a large number of cardiomyocytes results in reparative fibrosis, a process that is critical for preservation of the structural integrity of the infarcted ventricle. On the other hand, pathophysiologic stimuli, such as pressure overload, volume overload, metabolic dysfunction, and aging may cause interstitial and perivascular fibrosis in the absence of infarction. Activated myofibroblasts are the main effector cells in cardiac fibrosis; their expansion following myocardial injury is primarily driven through activation of resident interstitial cell populations. Several other cell types, including cardiomyocytes, endothelial cells, pericytes, macrophages, lymphocytes and mast cells may contribute to the fibrotic process, by producing proteases that participate in matrix metabolism, by secreting fibrogenic mediators and matricellular proteins, or by exerting contact-dependent actions on fibroblast phenotype. The mechanisms of induction of fibrogenic signals are dependent on the type of primary myocardial injury. Activation of neurohumoral pathways stimulates fibroblasts both directly, and through effects on immune cell populations. Cytokines and growth factors, such as Tumor Necrosis Factor-α, Interleukin (IL)-1, IL-10, chemokines, members of the Transforming Growth Factor-β family, IL-11, and Platelet-Derived Growth Factors are secreted in the cardiac interstitium and play distinct roles in activating specific aspects of the fibrotic response. Secreted fibrogenic mediators and matricellular proteins bind to cell surface receptors in fibroblasts, such as cytokine receptors, integrins, syndecans and CD44, and transduce intracellular signaling cascades that regulate genes involved in synthesis, processing and metabolism of the extracellular matrix. Endogenous pathways involved in negative regulation of fibrosis are critical for cardiac repair and may protect the myocardium from excessive fibrogenic responses. Due to the reparative nature of many forms of cardiac fibrosis, targeting fibrotic remodeling following myocardial injury poses major challenges. Development of effective therapies will require careful dissection of the cell biological mechanisms, study of the functional consequences of fibrotic changes on the myocardium, and identification of heart failure patient subsets with overactive fibrotic responses.
Collapse
Affiliation(s)
- Nikolaos G Frangogiannis
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, 1300 Morris Park Avenue, Forchheimer G46B, Bronx, NY, 10461, USA.
| |
Collapse
|
27
|
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
|
28
|
Nguyen-Truong M, Wang Z. Biomechanical Properties and Mechanobiology of Cardiac ECM. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1098:1-19. [PMID: 30238363 DOI: 10.1007/978-3-319-97421-7_1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
The heart is comprised of cardiac cells and extracellular matrix (ECM) which function together to pump blood throughout the body, provide organs with nutrients and oxygen, and remove metabolic wastes. Cardiac ECM provides a scaffold to cardiac cells and contributes to the mechanical properties and function of the cardiac tissue. Recently, more evidence suggests that cardiac ECM plays an active role in cardiac remodeling in response to mechanical loads. To that end, we provide an overview of the structure and function of the heart and the currently available in vivo and ex vivo mechanical measurements of cardiac tissues. We also review the biomechanical properties of cardiac tissues including the myocardium and heart valves, with a discussion on the differences between the right ventricle and left ventricle. Lastly, we go into the mechanical factors involved in cardiac remodeling and review the mechanobiology of cardiac tissues, i.e., the biomechanical responses at the cellular and tissue level, with an emphasis on the impact on the cardiac ECM. The regulation of cardiac ECM on cell function, which is a new and open area of research, is also briefly discussed. Future investigation into the ECM deposition and the interaction of cardiac cells and ECM components for mechanotransduction can assist to understand cardiac remodeling and inspire new therapies for cardiac diseases.
Collapse
Affiliation(s)
| | - Zhijie Wang
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO, USA. .,Department of Mechanical Engineering, Colorado State University, Fort Collins, CO, USA.
| |
Collapse
|
29
|
Guichard JL, Rogowski M, Agnetti G, Fu L, Powell P, Wei CC, Collawn J, Dell'Italia LJ. Desmin loss and mitochondrial damage precede left ventricular systolic failure in volume overload heart failure. Am J Physiol Heart Circ Physiol 2017; 313:H32-H45. [PMID: 28455287 PMCID: PMC5538858 DOI: 10.1152/ajpheart.00027.2017] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 03/27/2017] [Accepted: 04/10/2017] [Indexed: 01/10/2023]
Abstract
Heart failure due to chronic volume overload (VO) in rats and humans is characterized by disorganization of the cardiomyocyte desmin/mitochondrial network. Here, we tested the hypothesis that desmin breakdown is an early and continuous process throughout VO. Male Sprague-Dawley rats had aortocaval fistula (ACF) or sham surgery and were examined 24 h and 4 and 12 wk later. Desmin/mitochondrial ultrastructure was examined by transmission electron microscopy (TEM) and immunohistochemistry (IHC). Protein and kinome analysis were performed in isolated cardiomyocytes, and desmin cleavage was assessed by mass spectrometry in left ventricular (LV) tissue. Echocardiography demonstrated a 40% decrease in the LV mass-to-volume ratio with spherical remodeling at 4 wk with ACF and LV systolic dysfunction at 12 wk. Starting at 24 h and continuing to 4 and 12 wk, with ACF there is TEM evidence of extensive mitochondrial clustering, IHC evidence of disorganization associated with desmin breakdown, and desmin protein cleavage verified by Western blot analysis and mass spectrometry. IHC results revealed that ACF cardiomyocytes at 4 and 12 wk had perinuclear translocation of αB-crystallin from the Z disk with increased α, β-unsaturated aldehyde 4-hydroxynonelal. Use of protein markers with verification by TUNEL staining and kinome analysis revealed an absence of cardiomyocyte apoptosis at 4 and 12 wk of ACF. Significant increases in protein indicators of mitophagy were countered by a sixfold increase in p62/sequestosome-1, which is indicative of an inability to complete autophagy. An early and continuous disruption of the desmin/mitochondrial architecture, accompanied by oxidative stress and inhibition of apoptosis and mitophagy, suggests its causal role in LV dilatation and systolic dysfunction in VO.NEW & NOTEWORTHY This study provides new evidence of early onset (24 h) and continuous (4-12 wk) desmin misarrangement and disruption of the normal sarcomeric and mitochondrial architecture throughout the progression of volume overload heart failure, suggesting a causal link between desmin cleavage and mitochondrial disorganization and damage.
Collapse
Affiliation(s)
- Jason L Guichard
- Department of Medicine, Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, Alabama.,Center for Heart Failure Research, University of Alabama at Birmingham, Birmingham, Alabama
| | - Michael Rogowski
- Department of Medicine, Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, Alabama.,Center for Exercise Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Giulio Agnetti
- Division of Cardiology, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland.,Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy; and
| | - Lianwu Fu
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Pamela Powell
- Department of Veterans Affairs Medical Center, Birmingham, Alabama
| | - Chih-Chang Wei
- Center for Heart Failure Research, University of Alabama at Birmingham, Birmingham, Alabama.,Department of Veterans Affairs Medical Center, Birmingham, Alabama
| | - James Collawn
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Louis J Dell'Italia
- Department of Medicine, Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, Alabama; .,Center for Heart Failure Research, University of Alabama at Birmingham, Birmingham, Alabama.,Department of Veterans Affairs Medical Center, Birmingham, Alabama
| |
Collapse
|
30
|
Mohamed BA, Asif AR, Schnelle M, Qasim M, Khadjeh S, Lbik D, Schott P, Hasenfuss G, Toischer K. Proteomic analysis of short-term preload-induced eccentric cardiac hypertrophy. J Transl Med 2016; 14:149. [PMID: 27234427 PMCID: PMC4884361 DOI: 10.1186/s12967-016-0898-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Accepted: 05/07/2016] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Hemodynamic load leads to cardiac hypertrophy and heart failure. While afterload (pressure overload) induces concentric hypertrophy, elevation of preload (volume overload) yields eccentric hypertrophy and is associated with a better outcome. Here we analysed the proteomic pattern of mice subjected to short-term preload. METHODS AND RESULTS Female FVB/N mice were subjected to aortocaval shunt-induced volume overload that leads to an eccentric hypertrophy (left ventricular weight/tibia length +31 %) with sustained systolic heart function at 1 week after operation. Two-dimensional gel electrophoresis (2-DE) followed by mass spectrometric analysis showed alteration in the expression of 25 protein spots representing 21 different proteins. 64 % of these protein spots were up-regulated and 36 % of the protein spots were consistently down-regulated. Interestingly, α-1-antitrypsin was down-regulated, indicating higher elastin degradation and possibly contributing to the early dilatation. In addition to contractile and mitochondrial proteins, polymerase I and transcript release factor protein (PTRF) was also up-regulated, possibly contributing to the preload-induced signal transduction. CONCLUSIONS Our findings reveal the proteomic changes of early-stage eccentric myocardial remodeling after volume overload. Induced expression of some of the respiratory chain enzymes suggests a metabolic shift towards an oxidative phosphorylation that might contribute to the favorable remodeling seen in early VO. Down-regulation of α-1-antitrypsin might contribute to extracellular matrix remodeling and left ventricular dilatation. We also identified PTRF as a potential signaling regulator of volume overload-induced cardiac hypertrophy.
Collapse
Affiliation(s)
- Belal A Mohamed
- Department of Cardiology and Pneumology, University Medical Center, Goettingen, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Goettingen, Germany.,Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Abdul R Asif
- Institute of Clinical Chemistry/UMG-Laboratories, University Medical Center, Goettingen, Germany
| | - Moritz Schnelle
- Department of Cardiology and Pneumology, University Medical Center, Goettingen, Germany
| | - Mohamed Qasim
- Institute of Clinical Chemistry/UMG-Laboratories, University Medical Center, Goettingen, Germany.,Department of Microbiology, Kohat University of Science and Technology, Kohat, Pakistan
| | - Sara Khadjeh
- Department of Cardiology and Pneumology, University Medical Center, Goettingen, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Goettingen, Germany
| | - Dawid Lbik
- Department of Cardiology and Pneumology, University Medical Center, Goettingen, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Goettingen, Germany
| | - Peter Schott
- Department of Cardiology and Pneumology, University Medical Center, Goettingen, Germany
| | - Gerd Hasenfuss
- Department of Cardiology and Pneumology, University Medical Center, Goettingen, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Goettingen, Germany
| | - Karl Toischer
- Department of Cardiology and Pneumology, University Medical Center, Goettingen, Germany. .,DZHK (German Centre for Cardiovascular Research), Partner Site Goettingen, Germany. .,Abteilung Kardiologie und Pneumologie, Universitätsmedizin Göttingen, Robert-Koch-Str. 40, 37075, Göttingen, Germany.
| |
Collapse
|
31
|
Dal-Bianco JP, Aikawa E, Bischoff J, Guerrero JL, Hjortnaes J, Beaudoin J, Szymanski C, Bartko PE, Seybolt MM, Handschumacher MD, Sullivan S, Garcia ML, Mauskapf A, Titus JS, Wylie-Sears J, Irvin WS, Chaput M, Messas E, Hagège AA, Carpentier A, Levine RA. Myocardial Infarction Alters Adaptation of the Tethered Mitral Valve. J Am Coll Cardiol 2016; 67:275-87. [PMID: 26796392 DOI: 10.1016/j.jacc.2015.10.092] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Revised: 10/16/2015] [Accepted: 10/20/2015] [Indexed: 02/06/2023]
Abstract
BACKGROUND In patients with myocardial infarction (MI), leaflet tethering by displaced papillary muscles induces mitral regurgitation (MR), which doubles mortality. Mitral valves (MVs) are larger in such patients but fibrosis sets in counterproductively. The investigators previously reported that experimental tethering alone increases mitral valve area in association with endothelial-to-mesenchymal transition. OBJECTIVES The aim of this study was to explore the clinically relevant situation of tethering and MI, testing the hypothesis that ischemic milieu modifies mitral valve adaptation. METHODS Twenty-three adult sheep were examined. Under cardiopulmonary bypass, the papillary muscle tips in 6 sheep were retracted apically to replicate tethering, short of producing MR (tethered alone). Papillary muscle retraction was combined with apical MI created by coronary ligation in another 6 sheep (tethered plus MI), and left ventricular remodeling was limited by external constraint in 5 additional sheep (left ventricular constraint). Six sham-operated sheep were control subjects. Diastolic mitral valve surface area was quantified by 3-dimensional echocardiography at baseline and after 58 ± 5 days, followed by histopathology and flow cytometry of excised leaflets. RESULTS Tethered plus MI leaflets were markedly thicker than tethered-alone valves and sham control subjects. Leaflet area also increased significantly. Endothelial-to-mesenchymal transition, detected as α-smooth muscle actin-positive endothelial cells, significantly exceeded that in tethered-alone and control valves. Transforming growth factor-β, matrix metalloproteinase expression, and cellular proliferation were markedly increased. Uniquely, tethering plus MI showed endothelial activation with vascular adhesion molecule expression, neovascularization, and cells positive for CD45, considered a hematopoietic cell marker. Tethered plus MI findings were comparable with external ventricular constraint. CONCLUSIONS MI altered leaflet adaptation, including a profibrotic increase in valvular cell activation, CD45-positive cells, and matrix turnover. Understanding cellular and molecular mechanisms underlying leaflet adaptation and fibrosis could yield new therapeutic opportunities for reducing ischemic MR.
Collapse
Affiliation(s)
- Jacob P Dal-Bianco
- Cardiac Ultrasound Laboratory, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts; Leducq Transatlantic Mitral Network, Fondation Leducq, Paris, France
| | - Elena Aikawa
- Leducq Transatlantic Mitral Network, Fondation Leducq, Paris, France; Center for Excellence in Vascular Biology, Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Joyce Bischoff
- Leducq Transatlantic Mitral Network, Fondation Leducq, Paris, France; Vascular Biology Program, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts; Department of Surgery, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts
| | - J Luis Guerrero
- Surgical Cardiovascular Laboratory, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Jesper Hjortnaes
- Center for Excellence in Vascular Biology, Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Jonathan Beaudoin
- Cardiac Ultrasound Laboratory, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts; Leducq Transatlantic Mitral Network, Fondation Leducq, Paris, France
| | - Catherine Szymanski
- Cardiac Ultrasound Laboratory, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts; Leducq Transatlantic Mitral Network, Fondation Leducq, Paris, France
| | - Philipp E Bartko
- Cardiac Ultrasound Laboratory, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Margo M Seybolt
- Surgical Cardiovascular Laboratory, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Mark D Handschumacher
- Cardiac Ultrasound Laboratory, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Suzanne Sullivan
- Surgical Cardiovascular Laboratory, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Michael L Garcia
- Surgical Cardiovascular Laboratory, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Adam Mauskapf
- Surgical Cardiovascular Laboratory, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - James S Titus
- Surgical Cardiovascular Laboratory, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Jill Wylie-Sears
- Vascular Biology Program, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Whitney S Irvin
- Center for Excellence in Vascular Biology, Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Miguel Chaput
- Cardiac Ultrasound Laboratory, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts; Leducq Transatlantic Mitral Network, Fondation Leducq, Paris, France
| | - Emmanuel Messas
- Leducq Transatlantic Mitral Network, Fondation Leducq, Paris, France; Departments of Cardiology and Cardiovascular Surgery, Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, University Paris Descartes, INSERM Unit 633, Paris, France
| | - Albert A Hagège
- Leducq Transatlantic Mitral Network, Fondation Leducq, Paris, France; Departments of Cardiology and Cardiovascular Surgery, Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, University Paris Descartes, INSERM Unit 633, Paris, France
| | - Alain Carpentier
- Leducq Transatlantic Mitral Network, Fondation Leducq, Paris, France; Departments of Cardiology and Cardiovascular Surgery, Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, University Paris Descartes, INSERM Unit 633, Paris, France
| | - Robert A Levine
- Cardiac Ultrasound Laboratory, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts; Leducq Transatlantic Mitral Network, Fondation Leducq, Paris, France; Departments of Cardiology and Cardiovascular Surgery, Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, University Paris Descartes, INSERM Unit 633, Paris, France.
| | | |
Collapse
|
32
|
Yu A, Zhang J, Liu H, Liu B, Meng L. Identification of nondiabetic heart failure-associated genes by bioinformatics approaches in patients with dilated ischemic cardiomyopathy. Exp Ther Med 2016; 11:2602-2608. [PMID: 27284354 DOI: 10.3892/etm.2016.3252] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 03/03/2016] [Indexed: 12/24/2022] Open
Abstract
Heart failure (HF) is a common pathological condition affecting 4% of the worldwide population. However, approaches for predicting or treating nondiabetic HF (ND-HF) progression are insufficient. In the current study, the gene expression profile GSE26887 was analyzed, which contained samples from 5 healthy controls, 7 diabetes mellitus-HF patients and 12 ND-HF patients with dilated ischemic cardiomyopathy. The dataset of 5 healthy controls and 12 ND-HF patients was normalized with robust multichip average analysis and the differentially expressed genes (DEGs) were screened by unequal variance t-test and multiple-testing correction. In addition, the protein-protein interaction (PPI) network of the upregulated and downregulated genes was constructed using the Search Tool for the Retrieval of Interacting Genes/Proteins database and the Cytoscape software platform. Subsequently, gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were performed. A total of 122 upregulated and 133 downregulated genes were detected. The most significantly up- and downregulated genes were EIF1AY and SERPINE1, respectively. In addition, 38 and 77 nodes were obtained in the up- and downregulated PPI network. DEGs that owned the highest connectivity degree were USP9Y and UTY in the upregulated network, and CD44 in the downregulated networks, respectively. NPPA and SERPINE1 were also found to be hub genes in the PPI network. Several GO terms and pathways that were enriched by DEGs were identified, and the most significantly enriched KEGG pathways were drug metabolism and extracellular matrix-receptor interaction. In conclusion, the two DEGs, NPPA and SERPINE1, may be important in the pathogenesis of HF and may be used for the diagnosis and treatment of HF.
Collapse
Affiliation(s)
- Anzhong Yu
- Department of Cardiology, Jinan No. 4 People's Hospital, Jinan, Shandong 250031, P.R. China
| | - Jingyao Zhang
- Department of Blood Purification, Jinan Infectious Disease Hospital, Jinan, Shandong 250021, P.R. China
| | - Haiyan Liu
- Department of Internal Medicine, Jinan Minzu Hospital, Jinan, Shandong 250014, P.R. China
| | - Bing Liu
- Department of Cardiology, Jinan No. 4 People's Hospital, Jinan, Shandong 250031, P.R. China
| | - Lingdong Meng
- Department of Cardiology, Jinan No. 4 People's Hospital, Jinan, Shandong 250031, P.R. China
| |
Collapse
|
33
|
Takai S, Jin D. Improvement of cardiovascular remodelling by chymase inhibitor. Clin Exp Pharmacol Physiol 2016; 43:387-93. [DOI: 10.1111/1440-1681.12549] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Revised: 01/14/2016] [Accepted: 01/16/2016] [Indexed: 11/27/2022]
Affiliation(s)
- Shinji Takai
- Department of Innovative Medicine; Graduate School of Medicine; Osaka Medical College; Takatsuki Japan
| | - Denan Jin
- Department of Pharmacology; Osaka Medical College; Takatsuki Japan
| |
Collapse
|
34
|
Correlation between growth differentiation factor-15 and collagen metabolism indicators in patients with myocardial infarction and heart failure. JOURNAL OF GERIATRIC CARDIOLOGY : JGC 2016; 13:88-93. [PMID: 26918019 PMCID: PMC4753018 DOI: 10.11909/j.issn.1671-5411.2016.01.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Background Growth differentiation factor (GDF)-15, a divergent member of the transforming growth factor beta super-family does appear to be up-regulated in response to experimental pressure overload and progression of heart failure (HF). HF frequently develops after myocardial infarction (MI), contributing to worse outcome. The aim of this study is to assess the correlation between GDF-15 levels and markers related to collagen turnover in different stages of HF. Methods The study consists of a cohort of 179 patients, including stable angina pectoris patients (AP group, n = 50), old MI patients without HF (OMI group, n = 56), old MI patients with HF (OMI-HF group, n = 38) and normal Control group (n = 35). Both indicators reflecting the synthesis and degradation rates of collagen including precollagen I N-terminal peptide (PINP), type I collagen carboxy-terminal peptide (ICTP), precollagen III N-terminal peptide (PIIINP) and GDF-15 were measured using an enzyme-linked inmunosorbent assay. Results The plasma GDF-15 level was higher in OMI-HF group (1373.4 ± 275.4 ng/L) than OMI group (1036.1 ± 248.6 ng/L), AP group (784.6 ± 222.4 ng/L) and Control group (483.8 ± 186.4 ng/L) (P < 0.001). The indicators of collagen turnover (ICTP, PINP, PIIINP) all increased in the OMI-HF group compared with Control group (3.03 ± 1.02 µg/L vs. 2.08 ± 0.95 µg/L, 22.2 ± 6.6 µg/L vs. 16.7 ± 5.1 µg/L and 13.2 ± 7.9 µg/L vs. 6.4 ± 2.1 µg/L, respectively; P < 0.01). GDF-15 positively correlated with ICTP and PIIINP (r = 0.302, P < 0.001 and r = 0.206, P = 0.006, respectively). GDF-15 positively correlated to the echocardiographic diastolic indicators E/Em and left atrial pressure (r = 0.349 and r = 0.358, respectively; P < 0.01), and inversely correlated to the systolic indicators left ventricular ejection fraction and the average of peak systolic myocardial velocities (Sm) (r = −0.623 and r = −0.365, respectively; P < 0.01). Conclusion Plasma GDF-15 is associated with the indicators of type I and III collagen turnover.
Collapse
|
35
|
Mohamed BA, Schnelle M, Khadjeh S, Lbik D, Herwig M, Linke WA, Hasenfuss G, Toischer K. Molecular and structural transition mechanisms in long-term volume overload. Eur J Heart Fail 2015; 18:362-71. [PMID: 26694078 PMCID: PMC5064674 DOI: 10.1002/ejhf.465] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Revised: 11/08/2015] [Accepted: 11/14/2015] [Indexed: 01/09/2023] Open
Abstract
Aim We have previously reported that early phase (1 week) of experimental volume overload (VO) has an adaptive phenotype while wall stress‐matched pressure overload (PO) is maladaptive. Here we investigate the transition from adaptation to heart failure (HF) in long‐term VO. Methods and results FVB/N wild‐type mice were subjected to VO induced by aortocaval shunt, and were followed by serial echocardiography until in vivo left ventricular ejection fraction was below <50% (135 ± 35 days). Heart failure was evident from increased lung and liver weight and increased mortality compared with sham. Maladaptive remodelling resulted in significantly reduced sarcomeric titin phosphorylation (causing increased sarcomeric stiffness), whereas interstitial fibrosis was not increased. This was paralleled by re‐expression of the fetal gene program, activation of calcium/calmodulin‐dependent protein kinase II (CaMKII), decreased protein kinase B (Akt) phosphorylation, high oxidative stress, and increased apoptosis. Consistently, development of HF and mortality were significantly aggravated in Akt‐deficient mice. Conclusion Transition to HF in VO is associated with decreased Akt and increased CaMKII signalling pathways together with increased oxidative stress and apoptosis. Lack of interstitial fibrosis together with sarcomeric titin hypophosphorylation indicates an increased stiffness at the sarcomeric but not matrix level in VO‐induced HF (in contrast to PO). Transition to HF may result from myocyte loss and myocyte dysfunction owing to increased stiffness.
Collapse
Affiliation(s)
- Belal A Mohamed
- Department of Cardiology and Pneumology, Georg-August-University, Goettingen, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Goettingen, Germany.,Faculty of Medicine, Mansoura University, Egypt
| | - Moritz Schnelle
- Department of Cardiology and Pneumology, Georg-August-University, Goettingen, Germany
| | - Sara Khadjeh
- Department of Cardiology and Pneumology, Georg-August-University, Goettingen, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Goettingen, Germany
| | - Dawid Lbik
- Department of Cardiology and Pneumology, Georg-August-University, Goettingen, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Goettingen, Germany
| | - Melissa Herwig
- Department of Cardiovascular Physiology, Ruhr University Bochum, Germany
| | - Wolfgang A Linke
- Department of Cardiology and Pneumology, Georg-August-University, Goettingen, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Goettingen, Germany.,Department of Cardiovascular Physiology, Ruhr University Bochum, Germany
| | - Gerd Hasenfuss
- Department of Cardiology and Pneumology, Georg-August-University, Goettingen, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Goettingen, Germany
| | - Karl Toischer
- Department of Cardiology and Pneumology, Georg-August-University, Goettingen, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Goettingen, Germany
| |
Collapse
|
36
|
Transcriptional Changes Associated with Long-Term Left Ventricle Volume Overload in Rats: Impact on Enzymes Related to Myocardial Energy Metabolism. BIOMED RESEARCH INTERNATIONAL 2015; 2015:949624. [PMID: 26583150 PMCID: PMC4637065 DOI: 10.1155/2015/949624] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 09/13/2015] [Indexed: 01/12/2023]
Abstract
Patients with left ventricle (LV) volume overload (VO) remain in a compensated state for many years although severe dilation is present. The myocardial capacity to fulfill its energetic demand may delay decompensation. We performed a gene expression profile, a model of chronic VO in rat LV with severe aortic valve regurgitation (AR) for 9 months, and focused on the study of genes associated with myocardial energetics. Methods. LV gene expression profile was performed in rats after 9 months of AR and compared to sham-operated controls. LV glucose and fatty acid (FA) uptake was also evaluated in vivo by positron emission tomography in 8-week AR rats treated or not with fenofibrate, an activator of FA oxidation (FAO). Results. Many LV genes associated with mitochondrial function and metabolism were downregulated in AR rats. FA β-oxidation capacity was significantly impaired as early as two weeks after AR. Treatment with fenofibrate, a PPARα agonist, normalized both FA and glucose uptake while reducing LV dilation caused by AR. Conclusion. Myocardial energy substrate preference is affected early in the evolution of LV-VO cardiomyopathy. Maintaining a relatively normal FA utilization in the myocardium could translate into less glucose uptake and possibly lesser LV remodeling.
Collapse
|
37
|
Tan HT, Lim TK, Richards AM, Kofidis T, Teoh KLK, Ling LH, Chung MCM. Unravelling the proteome of degenerative human mitral valves. Proteomics 2015; 15:2934-44. [DOI: 10.1002/pmic.201500040] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Revised: 03/05/2015] [Accepted: 04/21/2015] [Indexed: 12/26/2022]
Affiliation(s)
- Hwee Tong Tan
- Department of Biochemistry; Yong Loo Lin School of Medicine, National University of Singapore; Singapore
| | - Teck Kwang Lim
- Department of Biological Sciences; Faculty of Science, National University of Singapore; Singapore
| | - Arthur Mark Richards
- Department of Medicine; Yong Loo Lin School of Medicine, National University of Singapore; Singapore
| | - Theodoros Kofidis
- Department of Cardiac; Thoracic and Vascular Surgery; National University Heart Centre; Singapore
| | - Kristine Leok-Kheng Teoh
- Department of Cardiac; Thoracic and Vascular Surgery; National University Heart Centre; Singapore
| | - Lieng H. Ling
- Department of Medicine; Yong Loo Lin School of Medicine, National University of Singapore; Singapore
- Cardiac Department; National University Heart Centre; Singapore
| | - Maxey C. M. Chung
- Department of Biochemistry; Yong Loo Lin School of Medicine, National University of Singapore; Singapore
- Department of Biological Sciences; Faculty of Science, National University of Singapore; Singapore
| |
Collapse
|
38
|
Ketoconazole induces apoptosis in rat cardiomyocytes through reactive oxygen species-mediated parkin overexpression. Arch Toxicol 2015; 89:1871-80. [PMID: 25787151 DOI: 10.1007/s00204-015-1502-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2015] [Accepted: 03/05/2015] [Indexed: 12/25/2022]
Abstract
Azole antifungals such as ketoconazole are generally known to induce a variety of heart function side effects, e.g., long-QT syndrome and ventricular arrhythmias. However, a clear mechanism for the action of ketoconazole in heart cells has not been reported. In the present study, we assessed the correlation between ketoconazole-induced apoptosis and the alteration of genes in response to ketoconazole in rat cardiomyocytes. Cardiomyocyte viability was significantly inhibited by treatment with ketoconazole. Ketoconazole also stimulated H2O2 generation and TUNEL-positive apoptosis in a dose-dependent manner. DNA microarray technology revealed that 10,571 genes were differentially expressed by more than threefold in ketoconazole-exposed cardiomyocytes compared with untreated controls. Among these genes, parkin, which encodes a component of the multiprotein E3 ubiquitin ligase complex, was predominantly overexpressed among those classified as apoptosis- and reactive oxygen species (ROS)-related genes. The expression of parkin was also elevated in cardiomyocytes treated with exogenous H2O2. Moreover, cell viability and apoptosis in response to ketoconazole were inhibited in cardiomyocytes treated with ROS inhibitors and transfected with parkin siRNA. From the present findings, we concluded that ketoconazole may increase the expression of parkin via the ROS-mediated pathway, which consequently results in the apoptosis and decreased viability of cardiomyocytes.
Collapse
|
39
|
Abstract
: Mitral regurgitation and other conditions marked by a pure isolated volume overload (VO) of the heart result in a progressive form of eccentric left ventricular remodeling and dysfunction. As opposed to the more extensively studied pressure overload, there are no approved medical therapies because an understanding of the underlying pathological mechanisms at work in VO is lacking. Over the past 20 years, our laboratory has identified multiple key biological functions involved in the pathological remodeling in VO. Specifically, we have noted perturbed matrix homeostasis, detrimental adrenergic signaling, increased intracellular reactive oxygen species and an intense inflammatory response that implicates mast cells and their product chymase, which seems to cause extensive remodeling both inside and outside the cardiomyocyte. How these multiple pathways intersect over the course of VO and their response to various single and combined interventions are now the subject of intense investigation.
Collapse
|
40
|
Yancey DM, Guichard JL, Ahmed MI, Zhou L, Murphy MP, Johnson MS, Benavides GA, Collawn J, Darley-Usmar V, Dell'Italia LJ. Cardiomyocyte mitochondrial oxidative stress and cytoskeletal breakdown in the heart with a primary volume overload. Am J Physiol Heart Circ Physiol 2015; 308:H651-63. [PMID: 25599572 DOI: 10.1152/ajpheart.00638.2014] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Left ventricular (LV) volume overload (VO) results in cardiomyocyte oxidative stress and mitochondrial dysfunction. Because mitochondria are both a source and target of ROS, we hypothesized that the mitochondrially targeted antioxidant mitoubiquinone (MitoQ) will improve cardiomyocyte damage and LV dysfunction in VO. Isolated cardiomyocytes from Sprague-Dawley rats were exposed to stretch in vitro and VO of aortocaval fistula (ACF) in vivo. ACF rats were treated with and without MitoQ. Isolated cardiomyocytes were analyzed after 3 h of cyclical stretch or 8 wk of ACF with MitoSox red or 5-(and-6)-chloromethyl-2',7'-dichlorodihydrofluorescein diacetate to measure ROS and with tetramethylrhodamine to measure mitochondrial membrane potential. Transmission electron microscopy and immunohistochemistry were used for cardiomyocyte structural assessment. In vitro cyclical stretch and 8-wk ACF resulted in increased cardiomyocyte mitochondrial ROS production and decreased mitochondrial membrane potential, which were significantly improved by MitoQ. ACF had extensive loss of desmin and β₂-tubulin that was paralleled by mitochondrial disorganization, loss of cristae, swelling, and clustering identified by mitochondria complex IV staining and transmission electron microscopy. MitoQ improved mitochondrial structural damage and attenuated desmin loss/degradation evidenced by immunohistochemistry and protein expression. However, LV dilatation and fractional shortening were unaffected by MitoQ treatment in 8-wk ACF. In conclusion, although MitoQ did not affect LV dilatation or function in ACF, these experiments suggest a connection of cardiomyocyte mitochondria-derived ROS production with cytoskeletal disruption and mitochondrial damage in the VO of ACF.
Collapse
Affiliation(s)
- Danielle M Yancey
- UAB Comprehensive Cardiovascular Center, University of Alabama at Birmingham, Birmingham, Alabama; Department of Medicine, Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, Alabama
| | - Jason L Guichard
- UAB Comprehensive Cardiovascular Center, University of Alabama at Birmingham, Birmingham, Alabama; Department of Medicine, Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, Alabama
| | - Mustafa I Ahmed
- UAB Comprehensive Cardiovascular Center, University of Alabama at Birmingham, Birmingham, Alabama; Department of Medicine, Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, Alabama
| | - Lufang Zhou
- UAB Comprehensive Cardiovascular Center, University of Alabama at Birmingham, Birmingham, Alabama; Department of Medicine, Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, Alabama
| | | | - Michelle S Johnson
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama; UAB Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Gloria A Benavides
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama; UAB Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, Alabama
| | - James Collawn
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Victor Darley-Usmar
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama; UAB Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Louis J Dell'Italia
- Department of Veterans Affairs Medical Center, Birmingham, Alabama; UAB Comprehensive Cardiovascular Center, University of Alabama at Birmingham, Birmingham, Alabama; Department of Medicine, Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, Alabama;
| |
Collapse
|
41
|
Cangemi C, Hansen ML, Argraves WS, Rasmussen LM. Fibulins and their role in cardiovascular biology and disease. Adv Clin Chem 2014; 67:245-65. [PMID: 25735864 DOI: 10.1016/bs.acc.2014.09.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Fibulins are a group of extracellular matrix proteins of which many are present in high amounts in the cardiovascular system. They share common biochemical properties and are often found in relation to basement membranes or elastic fibers. Observations in humans with specific mutations in fibulin genes, together with results from genetically engineered mice and data from human cardiovascular tissue suggest that the fibulin family of proteins play important functional roles in the cardiovascular system. Moreover, fibulin-1 circulates in high concentrations in plasma and may function as a cardiovascular disease marker.
Collapse
Affiliation(s)
- Claudia Cangemi
- Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Odense, Denmark
| | - Maria Lyck Hansen
- Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Odense, Denmark
| | - William Scott Argraves
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Lars Melholt Rasmussen
- Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Odense, Denmark.
| |
Collapse
|
42
|
Hulanicka M, Garncarz M, Parzeniecka-Jaworska M, Jank M. The transcriptomic profile of peripheral blood nuclear cells in dogs with heart failure. BMC Genomics 2014; 15:509. [PMID: 24952741 PMCID: PMC4092214 DOI: 10.1186/1471-2164-15-509] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Accepted: 06/13/2014] [Indexed: 01/17/2023] Open
Abstract
Background In recent years advances have been made in the investigative methods of molecular background of canine heart disease. Studies have been conducted to identify specific genes which, when pathologically expressed, could lead to the dysfunction of the canine heart or are correlated with heart failure. For this purpose genome wide microarray experiments on tissues from failing hearts have been performed. In the presented study a whole genome microarray analysis was used for the first time to describe the transcription profile of peripheral blood nuclear cells in dogs with heart failure. Dogs with recognized heart disease were classified according the ISACHC (International Small Animal Cardiac Health Council) classification scheme as class 1 (asymptomatic) - 13 dogs, class 2 (mild to moderate heart failure) - 13 dogs and class 3 (severe heart failure) - 12 dogs. The control group consisted of 14 healthy dogs. The clinical picture of the animals included: animal history, clinical examination, echocardiographic examination and where applicable electrocardiographic and radiographic examinations. Results In the present study we identified four sets of differentially expressed genes, namely heart-failure-specific genes and ISACHC1-specific genes, ISACHC2-sepcific genes and ISACHC-3 specific genes. The most important set consisted of genes differentially expressed in all dogs with heart failure, despite the ISACHC stage. We identified 71 heart-failure-specific genes which were involved in two statistically significant receptor signalling pathways, namely angiotensinR - > CREB/ELK-SRF/TP53 signalling and ephrinR - > actin signalling. The number of ISACHC1-specific genes was 83; ISACHC2-specific genes - 1247 and ISACHC3-specific - 200. Conclusions The transcriptomic profile of peripheral blood nuclear cells in dogs with heart failure seems to reflect the presence of clinical signs of the disease in patients based on the observation that the largest number of differentially expressed genes was identified in ISACHC 2 group of patients. This group consists of dogs just starting to show clinical signs of heart failure. A set of genes was also found to have changed expression in all dogs with heart failure, despite the stage of the disease. Electronic supplementary material The online version of this article (doi:10.1186/1471-2164-15-509) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Magdalena Hulanicka
- Department of Physiological Sciences, Faculty of Veterinary Medicine, Warsaw University of Life Sciences, Nowoursynowska str, 159c, 02-776 Warsaw, Poland.
| | | | | | | |
Collapse
|
43
|
Li AH, Liu PP, Villarreal FJ, Garcia RA. Dynamic changes in myocardial matrix and relevance to disease: translational perspectives. Circ Res 2014; 114:916-27. [PMID: 24577970 DOI: 10.1161/circresaha.114.302819] [Citation(s) in RCA: 95] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The cardiac extracellular matrix (ECM) provides the architectural scaffold to support efficient contraction and relaxation of cardiomyocytes. The elegant design of the ECM facilitates optimal force transduction, electric transmission, intercellular communication, and metabolic exchange within the myocardial microenvironment. In the setting of increased wall stress, injury, or disease, the ECM can undergo a series of dynamic changes that lead to favorable chamber remodeling and functional adaptation. Over time, sustained matrix remodeling can impair diastolic and systolic function caused by excess deposition of interstitial fibrous tissue. These pathological alterations in ECM structure/function are considered central to the evolution of adverse cardiac remodeling and the development of heart failure. This review discusses the complex dynamics of the cardiac ECM in the setting of myocardial infarction, pressure overload, and volume overload. We also summarize the current status of ECM biomarkers that may have clinical value in prognosticating cardiac disease progression in patients. Finally, we discuss the most current status of drugs under evaluation for use in cardiac fibrosis.
Collapse
Affiliation(s)
- Ai-Hsien Li
- From the University of Ottawa Heart Institute, Ottawa, Ontario, Canada (A.-H.L., P.P.L.); University of Toronto, Heart & Stroke/Lewar Centre of Excellence, Toronto, Ontario, Canada (P.P.L.); University of California, San Diego, School of Medicine (F.J.V., R.A.G.); and Bristol-Myers Squibb Company, Pennington, NJ (R.A.G.)
| | | | | | | |
Collapse
|
44
|
Rienks M, Papageorgiou AP, Frangogiannis NG, Heymans S. Myocardial extracellular matrix: an ever-changing and diverse entity. Circ Res 2014; 114:872-88. [PMID: 24577967 DOI: 10.1161/circresaha.114.302533] [Citation(s) in RCA: 250] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The cardiac extracellular matrix (ECM) is a complex architectural network consisting of structural and nonstructural proteins, creating strength and plasticity. The nonstructural compartment of the ECM houses a variety of proteins, which are vital for ECM plasticity, and can be divided into 3 major groups: glycoproteins, proteoglycans, and glycosaminoglycans. The common denominator for these groups is glycosylation, which refers to the decoration of proteins or lipids with sugars. This review will discuss the fundamental role of the matrix in cardiac development, homeostasis, and remodeling, from a glycobiology point of view. Glycoproteins (eg, thrombospondins, secreted protein acidic and rich in cysteine, tenascins), proteoglycans (eg, versican, syndecans, biglycan), and glycosaminoglycans (eg, hyaluronan, heparan sulfate) are upregulated on cardiac injury and regulate key processes in the remodeling myocardium such as inflammation, fibrosis, and angiogenesis. Albeit some parallels can be made regarding the processes these proteins are involved in, their specific functions are extremely diverse. In fact, under varying conditions, individual proteins can even have opposing functions, making spatiotemporal contribution of these proteins in the rearrangement of multifaceted ECM very hard to grasp. Alterations of protein characteristics by the addition of sugars may explain the immense, yet tightly regulated, variability of the remodeling cardiac matrix. Understanding the role of glycosylation in altering the ultimate function of glycoproteins, proteoglycans, and glycosaminoglycans in the myocardium may lead to the development of new biochemical structures or compounds with great therapeutic potential for patients with heart disease.
Collapse
Affiliation(s)
- Marieke Rienks
- From Maastricht University Medical Centre, Maastricht, The Netherlands
| | | | | | | |
Collapse
|
45
|
Zheng J, Wei CC, Hase N, Shi K, Killingsworth CR, Litovsky SH, Powell PC, Kobayashi T, Ferrario CM, Rab A, Aban I, Collawn JF, Dell'Italia LJ. Chymase mediates injury and mitochondrial damage in cardiomyocytes during acute ischemia/reperfusion in the dog. PLoS One 2014; 9:e94732. [PMID: 24733352 PMCID: PMC3986229 DOI: 10.1371/journal.pone.0094732] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Accepted: 03/18/2014] [Indexed: 12/22/2022] Open
Abstract
Cardiac ischemia and reperfusion (I/R) injury occurs because the acute increase in oxidative/inflammatory stress during reperfusion culminates in the death of cardiomyocytes. Currently, there is no drug utilized clinically that attenuates I/R injury in patients. Previous studies have demonstrated degranulation of mast cell contents into the interstitium after I/R. Using a dog model of I/R, we tested the role of chymase, a mast cell protease, in cardiomyocyte injury using a specific oral chymase inhibitor (CI). 15 adult mongrel dogs had left anterior descending artery occlusion for 60 min and reperfusion for 100 minutes. 9 dogs received vehicle and 6 were pretreated with a specific CI. In vivo cardiac microdialysis demonstrated a 3-fold increase in interstitial fluid chymase activity in I/R region that was significantly decreased by CI. CI pretreatment significantly attenuated loss of laminin, focal adhesion complex disruption, and release of troponin I into the circulation. Microarray analysis identified an I/R induced 17-fold increase in nuclear receptor subfamily 4A1 (NR4A1) and significantly decreased by CI. NR4A1 normally resides in the nucleus but can induce cell death on migration to the cytoplasm. I/R caused significant increase in NR4A1 protein expression and cytoplasmic translocation, and mitochondrial degradation, which were decreased by CI. Immunohistochemistry also revealed a high concentration of chymase within cardiomyocytes after I/R. In vitro, chymase added to culture HL-1 cardiomyocytes entered the cytoplasm and nucleus in a dynamin-dependent fashion, and promoted cytoplasmic translocation of NR4A1 protein. shRNA knockdown of NR4A1 on pre-treatment of HL-1 cells with CI significantly decreased chymase-induced cell death and mitochondrial damage. These results suggest that the beneficial effects of an orally active CI during I/R are mediated in the cardiac interstitium as well as within the cardiomyocyte due to a heretofore-unrecognized chymase entry into cardiomyocytes.
Collapse
Affiliation(s)
- Junying Zheng
- Birmingham Veteran Affairs Medical Center and Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Chih-Chang Wei
- Birmingham Veteran Affairs Medical Center and Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Naoki Hase
- Teijin Institute for Bio-Medical Research, Teijin Pharma Ltd, Tokyo, Japan
| | - Ke Shi
- Birmingham Veteran Affairs Medical Center and Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Cheryl R. Killingsworth
- Birmingham Veteran Affairs Medical Center and Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Silvio H. Litovsky
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Pamela C. Powell
- Birmingham Veteran Affairs Medical Center and Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | | | - Carlos M. Ferrario
- Wake Forest University, Winston Salem, North Carolina, United States of America
| | - Andras Rab
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Inmaculada Aban
- Department of Biostatistics University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - James F. Collawn
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Louis J. Dell'Italia
- Birmingham Veteran Affairs Medical Center and Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- * E-mail:
| |
Collapse
|
46
|
Kong P, Christia P, Frangogiannis NG. The pathogenesis of cardiac fibrosis. Cell Mol Life Sci 2014; 71:549-74. [PMID: 23649149 PMCID: PMC3769482 DOI: 10.1007/s00018-013-1349-6] [Citation(s) in RCA: 1150] [Impact Index Per Article: 104.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Revised: 04/19/2013] [Accepted: 04/22/2013] [Indexed: 12/16/2022]
Abstract
Cardiac fibrosis is characterized by net accumulation of extracellular matrix proteins in the cardiac interstitium, and contributes to both systolic and diastolic dysfunction in many cardiac pathophysiologic conditions. This review discusses the cellular effectors and molecular pathways implicated in the pathogenesis of cardiac fibrosis. Although activated myofibroblasts are the main effector cells in the fibrotic heart, monocytes/macrophages, lymphocytes, mast cells, vascular cells and cardiomyocytes may also contribute to the fibrotic response by secreting key fibrogenic mediators. Inflammatory cytokines and chemokines, reactive oxygen species, mast cell-derived proteases, endothelin-1, the renin/angiotensin/aldosterone system, matricellular proteins, and growth factors (such as TGF-β and PDGF) are some of the best-studied mediators implicated in cardiac fibrosis. Both experimental and clinical evidence suggests that cardiac fibrotic alterations may be reversible. Understanding the mechanisms responsible for initiation, progression, and resolution of cardiac fibrosis is crucial to design anti-fibrotic treatment strategies for patients with heart disease.
Collapse
Affiliation(s)
- Ping Kong
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, 1300 Morris Park Avenue Forchheimer G46B, Bronx, NY 10461 USA
| | - Panagiota Christia
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, 1300 Morris Park Avenue Forchheimer G46B, Bronx, NY 10461 USA
| | - Nikolaos G. Frangogiannis
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, 1300 Morris Park Avenue Forchheimer G46B, Bronx, NY 10461 USA
| |
Collapse
|
47
|
Fibroblasts in myocardial infarction: a role in inflammation and repair. J Mol Cell Cardiol 2013; 70:74-82. [PMID: 24321195 DOI: 10.1016/j.yjmcc.2013.11.015] [Citation(s) in RCA: 355] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2013] [Revised: 11/06/2013] [Accepted: 11/27/2013] [Indexed: 12/14/2022]
Abstract
Fibroblasts do not only serve as matrix-producing reparative cells, but exhibit a wide range of functions in inflammatory and immune responses, angiogenesis and neoplasia. The adult mammalian myocardium contains abundant fibroblasts enmeshed within the interstitial and perivascular extracellular matrix. The current review manuscript discusses the dynamic phenotypic and functional alterations of cardiac fibroblasts following myocardial infarction. Extensive necrosis of cardiomyocytes in the infarcted heart triggers an intense inflammatory reaction. In the early stages of infarct healing, fibroblasts become pro-inflammatory cells, activating the inflammasome and producing cytokines, chemokines and proteases. Pro-inflammatory cytokines (such as Interleukin-1) delay myofibroblast transformation, until the wound is cleared from dead cells and matrix debris. Resolution of the inflammatory infiltrate is associated with fibroblast migration, proliferation, matrix protein synthesis and myofibroblast conversion. Growth factors and matricellular proteins play an important role in myofibroblast activation during the proliferative phase of healing. Formation of a mature cross-linked scar is associated with clearance of fibroblasts, as poorly-understood inhibitory signals restrain the fibrotic response. However, in the non-infarcted remodeling myocardium, local fibroblasts may remain activated in response to volume and pressure overload and may promote interstitial fibrosis. Considering their abundance, their crucial role in cardiac inflammation and repair, and their involvement in myocardial dysfunction and arrhythmogenesis, cardiac fibroblasts may be key therapeutic targets in cardiac remodeling. This article is part of a Special Issue entitled Myocyte-Fibroblast Signalling in Myocardium.
Collapse
|
48
|
Zheng J, Yancey DM, Ahmed MI, Wei CC, Powell PC, Shanmugam M, Gupta H, Lloyd SG, McGiffin DC, Schiros CG, Denney TS, Babu GJ, Dell'Italia LJ. Increased sarcolipin expression and adrenergic drive in humans with preserved left ventricular ejection fraction and chronic isolated mitral regurgitation. Circ Heart Fail 2013; 7:194-202. [PMID: 24297688 DOI: 10.1161/circheartfailure.113.000519] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND There is currently no therapy proven to attenuate left ventricular (LV) dilatation and dysfunction in volume overload induced by isolated mitral regurgitation (MR). To better understand molecular signatures underlying isolated MR, we performed LV gene expression analyses and overlaid regulated genes into ingenuity pathway analysis in patients with isolated MR. METHODS AND RESULTS Gene arrays from LV tissue of 35 patients, taken at the time of surgical repair for isolated MR, were compared with 13 normal controls. Cine-MRI was performed in 31 patients before surgery to measure LV function and volume from serial short-axis summation. LV end-diastolic volume was 2-fold (P=0.005) higher in MR patients than in normal controls, and LV ejection fraction was 64±7% (50%-79%) in MR patients. Ingenuity pathway analysis identified significant activation of pathways involved in β-adrenergic, cAMP, and G-protein-coupled signaling, whereas there was downregulation of pathways associated with complement activation and acute phase response. SERCA2a and phospholamban protein were unchanged in MR versus control left ventricles. However, mRNA and protein levels of the sarcoplasmic reticulum Ca2+ ATPase (SERCA) regulatory protein sarcolipin, which is predominantly expressed in normal atria, were increased 12- and 6-fold, respectively. Immunofluorescence analysis confirmed the absence of sarcolipin in normal left ventricles and its marked upregulation in MR left ventricles. CONCLUSIONS These results demonstrate alterations in multiple pathways associated with β-adrenergic signaling and sarcolipin in the left ventricles of patients with isolated MR and LV ejection fraction>50%, suggesting a beneficial role for β-adrenergic blockade in isolated MR.
Collapse
|
49
|
Bigg PW, Baldo G, Sleeper MM, O'Donnell PA, Bai H, Rokkam VR, Liu Y, Wu S, Giugliani R, Casal ML, Haskins ME, Ponder KP. Pathogenesis of mitral valve disease in mucopolysaccharidosis VII dogs. Mol Genet Metab 2013; 110:319-28. [PMID: 23856419 PMCID: PMC3800211 DOI: 10.1016/j.ymgme.2013.06.013] [Citation(s) in RCA: 17] [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: 06/18/2013] [Accepted: 06/18/2013] [Indexed: 01/02/2023]
Abstract
Mucopolysaccharidosis VII (MPS VII) is due to the deficient activity of β-glucuronidase (GUSB) and results in the accumulation of glycosaminoglycans (GAGs) in lysosomes and multisystemic disease with cardiovascular manifestations. The goal here was to determine the pathogenesis of mitral valve (MV) disease in MPS VII dogs. Untreated MPS VII dogs had a marked reduction in the histochemical signal for structurally-intact collagen in the MV at 6 months of age, when mitral regurgitation had developed. Electron microscopy demonstrated that collagen fibrils were of normal diameter, but failed to align into large parallel arrays. mRNA analysis demonstrated a modest reduction in the expression of genes that encode collagen or collagen-associated proteins such as the proteoglycan decorin which helps collagen fibrils assemble, and a marked increase for genes that encode proteases such as cathepsins. Indeed, enzyme activity for cathepsin B (CtsB) was 19-fold normal. MPS VII dogs that received neonatal intravenous injection of a gamma retroviral vector had an improved signal for structurally-intact collagen, and reduced CtsB activity relative to that seen in untreated MPS VII dogs. We conclude that MR in untreated MPS VII dogs was likely due to abnormalities in MV collagen structure. This could be due to upregulation of enzymes that degrade collagen or collagen-associated proteins, to the accumulation of GAGs that compete with proteoglycans such as decorin for binding to collagen, or to other causes. Further delineation of the etiology of abnormal collagen structure may lead to treatments that improve biomechanical properties of the MV and other tissues.
Collapse
Affiliation(s)
- Paul W. Bigg
- Department of Internal Medicine, Washington University School of Medicine, St. Louis MO
| | - Guilherme Baldo
- Programa de Pos-Graduacao em Genetica e Biologia Molecular, Universidade Federal do Rio Grande do Sul, RS, Brazil
| | - Meg M. Sleeper
- Department of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Patricia A. O'Donnell
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Hanqing Bai
- Department of Internal Medicine, Washington University School of Medicine, St. Louis MO
| | - Venkata R.P. Rokkam
- Department of Internal Medicine, Washington University School of Medicine, St. Louis MO
| | - Yuli Liu
- Department of Internal Medicine, Washington University School of Medicine, St. Louis MO
| | - Susan Wu
- Department of Internal Medicine, Washington University School of Medicine, St. Louis MO
| | - Roberto Giugliani
- Programa de Pos-Graduacao em Genetica e Biologia Molecular, Universidade Federal do Rio Grande do Sul, RS, Brazil
| | - Margret L. Casal
- Department of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Mark E. Haskins
- Department of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Katherine P. Ponder
- Department of Internal Medicine, Washington University School of Medicine, St. Louis MO
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis MO
| |
Collapse
|
50
|
Chemaly ER, Kang S, Zhang S, McCollum L, Chen J, Bénard L, Purushothaman KR, Hajjar RJ, Lebeche D. Differential patterns of replacement and reactive fibrosis in pressure and volume overload are related to the propensity for ischaemia and involve resistin. J Physiol 2013; 591:5337-55. [PMID: 24018949 DOI: 10.1113/jphysiol.2013.258731] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Pathological left ventricle (LV) hypertrophy (LVH) results in reactive and replacement fibrosis. Volume overload LVH (VOH) is less profibrotic than pressure overload LVH (POH). Studies attribute subendocardial fibrosis in POH to ischaemia, and reduced fibrosis in VOH to collagen degradation favouring dilatation. However, the mechanical origin of the relative lack of fibrosis in VOH is incompletely understood. We hypothesized that reduced ischaemia propensity in VOH compared to POH accounted for the reduced replacement fibrosis, along with reduced reactive fibrosis. Rats with POH (ascending aortic banding) evolved into either compensated-concentric POH (POH-CLVH) or dilated cardiomyopathy (POH-DCM); they were compared to VOH (aorta-caval fistula). We quantified LV fibrosis, structural and haemodynamic factors of ischaemia propensity, and the activation of profibrotic pathways. Fibrosis in POH-DCM was severe, subendocardial and subepicardial, in contrast with subendocardial fibrosis in POH-CLVH and nearly no fibrosis in VOH. The propensity for ischaemia was more important in POH versus VOH, explaining different patterns of replacement fibrosis. LV collagen synthesis and maturation, and matrix metalloproteinase-2 expression, were more important in POH. The angiotensin II-transforming growth-factor β axis was enhanced in POH, and connective tissue growth factor (CTGF) was overexpressed in all types of LVH. LV resistin expression was markedly elevated in POH, mildly elevated in VOH and independently reflected chronic ischaemic injury after myocardial infarction. In vitro, resistin is induced by angiotensin II and induces CTGF in cardiomyocytes. Based on these findings, we conclude that a reduced ischaemia propensity and attenuated upstream reactive fibrotic pathways account for the attenuated fibrosis in VOH versus POH.
Collapse
Affiliation(s)
- Elie R Chemaly
- D. Lebeche: Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY 10029, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|