1
|
Huang S, Li J, Li Q, Wang Q, Zhou X, Chen J, Chen X, Bellou A, Zhuang J, Lei L. Cardiomyopathy: pathogenesis and therapeutic interventions. MedComm (Beijing) 2024; 5:e772. [PMID: 39465141 PMCID: PMC11502724 DOI: 10.1002/mco2.772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 09/12/2024] [Accepted: 09/16/2024] [Indexed: 10/29/2024] Open
Abstract
Cardiomyopathy is a group of disease characterized by structural and functional damage to the myocardium. The etiologies of cardiomyopathies are diverse, spanning from genetic mutations impacting fundamental myocardial functions to systemic disorders that result in widespread cardiac damage. Many specific gene mutations cause primary cardiomyopathy. Environmental factors and metabolic disorders may also lead to the occurrence of cardiomyopathy. This review provides an in-depth analysis of the current understanding of the pathogenesis of various cardiomyopathies, highlighting the molecular and cellular mechanisms that contribute to their development and progression. The current therapeutic interventions for cardiomyopathies range from pharmacological interventions to mechanical support and heart transplantation. Gene therapy and cell therapy, propelled by ongoing advancements in overarching strategies and methodologies, has also emerged as a pivotal clinical intervention for a variety of diseases. The increasing number of causal gene of cardiomyopathies have been identified in recent studies. Therefore, gene therapy targeting causal genes holds promise in offering therapeutic advantages to individuals diagnosed with cardiomyopathies. Acting as a more precise approach to gene therapy, they are gradually emerging as a substitute for traditional gene therapy. This article reviews pathogenesis and therapeutic interventions for different cardiomyopathies.
Collapse
Affiliation(s)
- Shitong Huang
- Department of Cardiac Surgical Intensive Care UnitGuangdong Cardiovascular InstituteGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhouChina
| | - Jiaxin Li
- Department of Cardiac Surgical Intensive Care UnitGuangdong Cardiovascular InstituteGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhouChina
| | - Qiuying Li
- Department of Cardiac Surgical Intensive Care UnitGuangdong Cardiovascular InstituteGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhouChina
| | - Qiuyu Wang
- Department of Cardiac Surgical Intensive Care UnitGuangdong Cardiovascular InstituteGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhouChina
| | - Xianwu Zhou
- Department of Cardiovascular SurgeryZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Jimei Chen
- Department of Cardiovascular SurgeryGuangdong Cardiovascular InstituteGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhouChina
- Department of Cardiovascular SurgeryGuangdong Provincial Key Laboratory of South China Structural Heart DiseaseGuangzhouChina
| | - Xuanhui Chen
- Department of Medical Big Data CenterGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhouChina
| | - Abdelouahab Bellou
- Department of Emergency Medicine, Institute of Sciences in Emergency MedicineGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhouChina
- Department of Emergency MedicineWayne State University School of MedicineDetroitMichiganUSA
| | - Jian Zhuang
- Department of Cardiovascular SurgeryGuangdong Cardiovascular InstituteGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhouChina
- Department of Cardiovascular SurgeryGuangdong Provincial Key Laboratory of South China Structural Heart DiseaseGuangzhouChina
| | - Liming Lei
- Department of Cardiac Surgical Intensive Care UnitGuangdong Cardiovascular InstituteGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhouChina
- Department of Cardiovascular SurgeryGuangdong Provincial Key Laboratory of South China Structural Heart DiseaseGuangzhouChina
| |
Collapse
|
2
|
Pavić J, Živanović M, Tanasković I, Pavić O, Stanković V, Virijević K, Mladenović T, Košarić J, Milićević B, Qamar SUR, Velicki L, Novaković I, Preveden A, Popović D, Tesić M, Seman S, Filipović N. A Machine Learning Approach to Gene Expression in Hypertrophic Cardiomyopathy. Pharmaceuticals (Basel) 2024; 17:1364. [PMID: 39459004 PMCID: PMC11510441 DOI: 10.3390/ph17101364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 10/08/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND/OBJECTIVES Hypertrophic cardiomyopathy (HCM) is a common heart disorder characterized by the thickening of the heart muscle, particularly in the left ventricle, which increases the risk of cardiac complications. This study aims to analyze the expression of apoptosis-regulating genes (CASP8, CASP9, CASP3, BAX, and BCL2) in blood samples from HCM patients, to better understand their potential as biomarkers for disease progression. METHODS Quantitative real-time PCR (qPCR) was used to evaluate gene expression in blood samples from 93 HCM patients. The correlation between apoptosis-regulating genes was conducted and clinical parameters were integrated for feature importance and clustering analysis. RESULTS Most patients exhibited significant downregulation of CASP8, CASP9, and CASP3. In contrast, BAX expression was elevated in 71 out of 93 patients, while BCL2 was increased in 55 out of 93 patients. Correlation analysis revealed weak negative correlations between the BAX/BCL2 ratio and CASP gene expression. CONCLUSIONS These findings suggest that reduced expression of apoptotic genes may indicate a protective cellular mechanism, which could serve as a biomarker for disease progression. Further studies are needed to investigate the potential for therapeutic modulation of these pathways to improve patient outcomes.
Collapse
Affiliation(s)
- Jelena Pavić
- Institute for Information Technologies Kragujevac, University of Kragujevac, 34000 Kragujevac, Serbia; (M.Ž.); (O.P.); (K.V.); (T.M.); (J.K.); (B.M.)
- Faculty of Engineering, University of Kragujevac, 34000 Kragujevac, Serbia (N.F.)
| | - Marko Živanović
- Institute for Information Technologies Kragujevac, University of Kragujevac, 34000 Kragujevac, Serbia; (M.Ž.); (O.P.); (K.V.); (T.M.); (J.K.); (B.M.)
| | - Irena Tanasković
- Department of Histology and Embryology, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia;
| | - Ognjen Pavić
- Institute for Information Technologies Kragujevac, University of Kragujevac, 34000 Kragujevac, Serbia; (M.Ž.); (O.P.); (K.V.); (T.M.); (J.K.); (B.M.)
| | - Vesna Stanković
- Department of Pathology, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia;
| | - Katarina Virijević
- Institute for Information Technologies Kragujevac, University of Kragujevac, 34000 Kragujevac, Serbia; (M.Ž.); (O.P.); (K.V.); (T.M.); (J.K.); (B.M.)
- Faculty of Engineering, University of Kragujevac, 34000 Kragujevac, Serbia (N.F.)
| | - Tamara Mladenović
- Institute for Information Technologies Kragujevac, University of Kragujevac, 34000 Kragujevac, Serbia; (M.Ž.); (O.P.); (K.V.); (T.M.); (J.K.); (B.M.)
- Faculty of Engineering, University of Kragujevac, 34000 Kragujevac, Serbia (N.F.)
| | - Jelena Košarić
- Institute for Information Technologies Kragujevac, University of Kragujevac, 34000 Kragujevac, Serbia; (M.Ž.); (O.P.); (K.V.); (T.M.); (J.K.); (B.M.)
| | - Bogdan Milićević
- Institute for Information Technologies Kragujevac, University of Kragujevac, 34000 Kragujevac, Serbia; (M.Ž.); (O.P.); (K.V.); (T.M.); (J.K.); (B.M.)
- Bioengineering Research and Development Center (BioIRC), 34000 Kragujevac, Serbia
| | - Safi Ur Rehman Qamar
- Faculty of Engineering, University of Kragujevac, 34000 Kragujevac, Serbia (N.F.)
- Bioengineering Research and Development Center (BioIRC), 34000 Kragujevac, Serbia
| | - Lazar Velicki
- Faculty of Medicine, University of Novi Sad, 21000 Novi Sad, Serbia; (L.V.); (A.P.)
- Institute of Cardiovascular Diseases Vojvodina, 21204 Sremska Kamenica, Serbia
| | - Ivana Novaković
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (I.N.); (M.T.)
| | - Andrej Preveden
- Faculty of Medicine, University of Novi Sad, 21000 Novi Sad, Serbia; (L.V.); (A.P.)
- Institute of Cardiovascular Diseases Vojvodina, 21204 Sremska Kamenica, Serbia
| | - Dejana Popović
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN 55905, USA;
| | - Milorad Tesić
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (I.N.); (M.T.)
- Clinic for Cardiology, University Clinical Center of Serbia, 11000 Belgrade, Serbia
| | - Stefan Seman
- Faculty of Sports and Physical Education, University of Belgrade, 11000 Belgrade, Serbia;
| | - Nenad Filipović
- Faculty of Engineering, University of Kragujevac, 34000 Kragujevac, Serbia (N.F.)
- Bioengineering Research and Development Center (BioIRC), 34000 Kragujevac, Serbia
| |
Collapse
|
3
|
Torbey AFM, Couto RGT, Grippa A, Maia EC, Miranda SA, Santos MACD, Peres ET, Costa OPS, Oliveira EMD, Mesquita ET. Cardiomyopathy in Children and Adolescents in the Era of Precision Medicine. Arq Bras Cardiol 2024; 121:e20230154. [PMID: 39442130 PMCID: PMC11634207 DOI: 10.36660/abc.20230154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 04/18/2024] [Accepted: 05/15/2024] [Indexed: 10/25/2024] Open
Abstract
In childhood and adolescence, cardiomyopathies have their own characteristics and are an important cause of heart failure, arrhythmias, sudden death, and indication for heart transplantation. Diagnosis is a challenge in daily practice due to its varied clinical presentation, heterogeneous etiologies, and limited knowledge of tools related to clinical and molecular genetics. However, it is essential to recognize the different phenotypes and prioritize the search for the etiology. Recent advances in precision medicine have made molecular diagnosis accessible, which makes it possible to individualize therapeutic approaches, stratify the prognosis, and identify individuals in the family who are at risk of developing the disease. The objective of this review is to emphasize the particularities of cardiomyopathies in pediatrics and how the individualized approach impacts the therapy and prognosis of the patient. Through a systematized approach, the five-stage protocol used in our service is presented. These stages bring together clinical evaluation for determining the morphofunctional phenotype, identification of etiology, classification, establishment of prognosis, and the search for personalized therapies.
Collapse
Affiliation(s)
- Ana Flávia Mallheiros Torbey
- Universidade Federal Fluminense, Niterói, RJ - Brasil
- Programa de Pós-Graduação em Ciências Cardiovasculares da Universidade Federal Fluminense, Niterói, RJ - Brasil
| | - Raquel Germer Toja Couto
- Universidade Federal Fluminense Hospital Universitário Antônio Pedro (EBSERH), Niterói, RJ - Brasil
| | - Aurea Grippa
- Universidade Federal Fluminense, Niterói, RJ - Brasil
| | | | | | | | | | | | | | - Evandro Tinoco Mesquita
- Universidade Federal Fluminense, Niterói, RJ - Brasil
- Programa de Pós-Graduação em Ciências Cardiovasculares da Universidade Federal Fluminense, Niterói, RJ - Brasil
- Complexo Hospitalar de Niteroi, Niterói, RJ - Brasil
| |
Collapse
|
4
|
Mikšiūnas R, Labeit S, Bironaite D. Class I and II Histone Deacetylase Inhibitors as Therapeutic Modulators of Dilated Cardiac Tissue-Derived Mesenchymal Stem/Stromal Cells. Int J Mol Sci 2024; 25:6758. [PMID: 38928463 PMCID: PMC11203858 DOI: 10.3390/ijms25126758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 06/14/2024] [Accepted: 06/15/2024] [Indexed: 06/28/2024] Open
Abstract
The prevalence of dilated cardiomyopathy (DCM) is increasing globally, highlighting the need for innovative therapeutic approaches to prevent its onset. In this study, we examined the energetic and epigenetic distinctions between dilated and non-dilated human myocardium-derived mesenchymal stem/stromal cells (hmMSCs) and assessed the effects of class I and II HDAC inhibitors (HDACi) on these cells and their cardiomyogenic differentiation. Cells were isolated from myocardium biopsies using explant outgrowth methods. Mitochondrial and histone deacetylase activities, ATP levels, cardiac transcription factors, and structural proteins were assessed using flow cytometry, PCR, chemiluminescence, Western blotting, and immunohistochemistry. The data suggest that the tested HDAC inhibitors improved acetylation and enhanced the energetic status of both types of cells, with significant effects observed in dilated myocardium-derived hmMSCs. Additionally, the HDAC inhibitors activated the cardiac transcription factors Nkx2-5, HOPX, GATA4, and Mef2C, and upregulated structural proteins such as cardiac troponin T and alpha cardiac actin at both the protein and gene levels. In conclusion, our findings suggest that HDACi may serve as potential modulators of the energetic status and cardiomyogenic differentiation of human heart hmMSCs. This avenue of exploration could broaden the search for novel therapeutic interventions for dilated cardiomyopathy, ultimately leading to improvements in heart function.
Collapse
Affiliation(s)
- Rokas Mikšiūnas
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Santariškių 5, LT-08406 Vilnius, Lithuania;
| | | | - Daiva Bironaite
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Santariškių 5, LT-08406 Vilnius, Lithuania;
| |
Collapse
|
5
|
Eden M, Leye M, Hahn J, Heilein E, Luzarowski M, Völschow B, Tannert C, Sossalla S, Lucena-Porcel C, Frank D, Frey N. Mst4, a novel cardiac STRIPAK complex-associated kinase, regulates cardiomyocyte growth and survival and is upregulated in human cardiomyopathy. J Biol Chem 2024; 300:107255. [PMID: 38579991 PMCID: PMC11087964 DOI: 10.1016/j.jbc.2024.107255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 03/15/2024] [Accepted: 03/19/2024] [Indexed: 04/07/2024] Open
Abstract
Myocardial failure is associated with adverse remodeling, including loss of cardiomyocytes, hypertrophy, and alterations in cell-cell contacts. Striatin-interacting phosphatase and kinase (STRIPAK) complexes and their mammalian STE20-like kinase 4 (Mst4) have been linked to development of different diseases. The role and targets of Mst4 in cardiomyocytes have not been investigated yet. Multitissue immunoblot experiments show highly enriched Mst4 expression in rodent hearts. Analyses of human biopsy samples from patients suffering from dilated cardiomyopathy revealed that Mst4 is upregulated (5- to 8-fold p < 0.001) compared with nonfailing controls. Increased abundance of Mst4 could also be detected in mouse models of cardiomyopathy. We confirmed that Mst4 interacts with STRIPAK components in neonatal rat ventricular cardiomyocytes, indicating that STRIPAK is present in the heart. Immunofluorescence stainings and molecular interaction studies revealed that Mst4 is localized to the intercalated disc and interacts with several intercalated disc proteins. Overexpression of Mst4 in cardiomyocytes results in hypertrophy compared with controls. In adult rat cardiomyocytes, Mst4 overexpression increases cellular and sarcomeric fractional shortening (p < 0.05), indicating enhanced contractility. Overexpression of Mst4 also inhibits apoptosis shown by reduction of cleaved caspase3 (-69%, p < 0.0001), caspase7 (-80%, p < 0.0001), and cleaved Parp1 (-27%, p < 0.001). To elucidate potential Mst4 targets, we performed phosphoproteomics analyses in neonatal rat cardiomyocytes after Mst4 overexpression and inhibition. The results revealed target candidates of Mst4 at the intercalated disc. We identified Mst4 as a novel cardiac kinase that is upregulated in cardiomyopathy-regulating cardiomyocyte growth and survival.
Collapse
Affiliation(s)
- Matthias Eden
- Department of Internal Medicine III, University of Heidelberg, Heidelberg, Germany; German Centre for Cardiovascular Research, Mannheim/Heidelberg, Germany
| | - Marius Leye
- Department of Internal Medicine III, University of Heidelberg, Heidelberg, Germany; German Centre for Cardiovascular Research, Mannheim/Heidelberg, Germany
| | - Justus Hahn
- Department of Internal Medicine III, University of Heidelberg, Heidelberg, Germany; German Centre for Cardiovascular Research, Mannheim/Heidelberg, Germany
| | - Emanuel Heilein
- Department of Internal Medicine III, University of Heidelberg, Heidelberg, Germany; German Centre for Cardiovascular Research, Mannheim/Heidelberg, Germany
| | - Marcin Luzarowski
- Core Facility for Mass Spectrometry and Proteomics, Center for Molecular Biology at Heidelberg University (ZMBH), Heidelberg, Germany
| | - Bill Völschow
- German Centre for Cardiovascular Research, Kiel, Lübeck, Hamburg, Germany; Department of Cardiology, University Heart and Vascular Center Hamburg, Hamburg, Germany
| | - Christin Tannert
- German Centre for Cardiovascular Research, Kiel, Lübeck, Hamburg, Germany; Department of Internal Medicine III (Cardiology and Angiology), University Hospital Schleswig-Holstein, Kiel, Germany
| | - Samuel Sossalla
- Department of Cardiology, University of Giessen, Giessen and Kerckhoff Heart and Lung Centre, Giessen, Germany
| | - Carlota Lucena-Porcel
- Tissue Bank of the National Center of Tumor Diseases (NCT) Heidelberg, Heidelberg University Hospital, Heidelberg, Germany
| | - Derk Frank
- German Centre for Cardiovascular Research, Kiel, Lübeck, Hamburg, Germany; Department of Internal Medicine III (Cardiology and Angiology), University Hospital Schleswig-Holstein, Kiel, Germany
| | - Norbert Frey
- Department of Internal Medicine III, University of Heidelberg, Heidelberg, Germany; German Centre for Cardiovascular Research, Mannheim/Heidelberg, Germany.
| |
Collapse
|
6
|
de Oliveira RM, Paiva MUB, Picossi CRC, Paiva DVN, Ricart CAO, Ruperez FJ, Barbas C, Atik FA, Martins AMA. Metabolomic insights in advanced cardiomyopathy of chronic chagasic and idiopathic patients that underwent heart transplant. Sci Rep 2024; 14:9810. [PMID: 38684702 PMCID: PMC11059181 DOI: 10.1038/s41598-024-53875-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 02/06/2024] [Indexed: 05/02/2024] Open
Abstract
Heart failure (HF) studies typically focus on ischemic and idiopathic heart diseases. Chronic chagasic cardiomyopathy (CCC) is a progressive degenerative inflammatory condition highly prevalent in Latin America that leads to a disturbance of cardiac conduction system. Despite its clinical and epidemiological importance, CCC molecular pathogenesis is poorly understood. Here we characterize and discriminate the plasma metabolomic profile of 15 patients with advanced HF referred for heart transplantation - 8 patients with CCC and 7 with idiopathic dilated cardiomyopathy (IDC) - using gas chromatography/quadrupole time-of-flight mass spectrometry. Compared to the 12 heart donor individuals, also included to represent the control (CTRL) scenario, patients with advanced HF exhibited a metabolic imbalance with 21 discriminating metabolites, mostly indicative of accumulation of fatty acids, amino acids and important components of the tricarboxylic acid (TCA) cycle. CCC vs. IDC analyses revealed a metabolic disparity between conditions, with 12 CCC distinctive metabolites vs. 11 IDC representative metabolites. Disturbances were mainly related to amino acid metabolism profile. Although mitochondrial dysfunction and loss of metabolic flexibility may be a central mechanistic event in advanced HF, metabolic imbalance differs between CCC and IDC populations, possibly explaining the dissimilar clinical course of Chagas' patients.
Collapse
Affiliation(s)
- Raphaela M de Oliveira
- School of Medicine, University of Brasilia, Brasilia, Brazil
- Laboratory of Protein Chemistry and Biochemistry, University of Brasilia, Brasilia, Brazil
| | | | - Carolina R C Picossi
- Center of Excellence in Metabolomics and Bioanalysis, University of San Pablo CEU, Madrid, Spain
| | - Diego V N Paiva
- School of Medicine, University of Brasilia, Brasilia, Brazil
| | - Carlos A O Ricart
- Laboratory of Protein Chemistry and Biochemistry, University of Brasilia, Brasilia, Brazil
| | - Francisco J Ruperez
- Center of Excellence in Metabolomics and Bioanalysis, University of San Pablo CEU, Madrid, Spain
| | - Coral Barbas
- Center of Excellence in Metabolomics and Bioanalysis, University of San Pablo CEU, Madrid, Spain
| | - Fernando A Atik
- School of Medicine, University of Brasilia, Brasilia, Brazil
- Institute of Cardiology and Transplantation of the Federal District, Brasilia, Brazil
| | - Aline M A Martins
- School of Medicine, University of Brasilia, Brasilia, Brazil.
- Center of Excellence in Metabolomics and Bioanalysis, University of San Pablo CEU, Madrid, Spain.
| |
Collapse
|
7
|
Zhao J, Wang B, Ta S, Lu X, Zhao X, Liu J, Yuan J, Wang J, Liu L. Association between Hypertrophic Cardiomyopathy and Variations in Sarcomere Gene and Calcium Channel Gene in Adults. Cardiology 2024; 149:440-450. [PMID: 38615672 DOI: 10.1159/000538747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 04/02/2024] [Indexed: 04/16/2024]
Abstract
INTRODUCTION Calcium channel gene variations have been reported to be associated with hypertrophic cardiomyopathy (HCM) in family, but the relationship between calcium channel gene variations and HCM remains undefined in the population. METHODS A total of 719 HCM unrelated patients were initially enrolled. Finally, 371 patients were identified based on inclusion and exclusion criteria, including 145 patients with gene negative, 28 patients with a single rare calcium channel gene variation (calcium gene variation), 162 patients with a single pathogenic/likely pathogenic sarcomere gene variation (sarcomere gene variation) and 36 patients with a single pathogenic/likely pathogenic sarcomere gene variation and a single rare calcium channel gene variation (double gene variations). Then the demographic, electrocardiographic, echocardiographic, and follow-up data were collected. RESULTS Patients with double gene variations were at an earlier age and had more percent of family history of HCM, and had thicker walls, higher left ventricular outflow tract pressure gradient, more pathological Q waves, and more bundle branch blocks as compared with those with single sarcomere gene variation. During the follow-up period, patients with double gene variations had more primary endpoints than the other three groups (p = 0.0013). Multivariate analysis showed that double gene variations were the independent predictor of primary endpoint events in patients (HR: 4.82, 95% CI: 1.77-13.2; p = 0.002). CONCLUSION We found that patients with double gene variations had more severe HCM phenotype and prognosis. The pathogenesis effects of sarcomere gene variation and calcium channel gene variation may be cumulative in HCM populations.
Collapse
Affiliation(s)
- Jia Zhao
- Department of Ultrasound, Xijing Hospital, Xijing Hypertrophic Cardiomyopathy Center, Air Force Military Medical University, Xi'an, China,
| | - Bo Wang
- Department of Ultrasound, Xijing Hospital, Xijing Hypertrophic Cardiomyopathy Center, Air Force Military Medical University, Xi'an, China
| | - Shengjun Ta
- Department of Ultrasound, Xijing Hospital, Xijing Hypertrophic Cardiomyopathy Center, Air Force Military Medical University, Xi'an, China
| | - Xiaonan Lu
- Department of Ultrasound, Xijing Hospital, Xijing Hypertrophic Cardiomyopathy Center, Air Force Military Medical University, Xi'an, China
| | - Xueli Zhao
- Department of Ultrasound, Xijing Hospital, Xijing Hypertrophic Cardiomyopathy Center, Air Force Military Medical University, Xi'an, China
| | - Jiao Liu
- Department of Ultrasound, Xijing Hospital, Xijing Hypertrophic Cardiomyopathy Center, Air Force Military Medical University, Xi'an, China
| | - Jiarui Yuan
- Department of Ultrasound, Xijing Hospital, Xijing Hypertrophic Cardiomyopathy Center, Air Force Military Medical University, Xi'an, China
| | - Jing Wang
- Department of Ultrasound, Xijing Hospital, Xijing Hypertrophic Cardiomyopathy Center, Air Force Military Medical University, Xi'an, China
| | - Liwen Liu
- Department of Ultrasound, Xijing Hospital, Xijing Hypertrophic Cardiomyopathy Center, Air Force Military Medical University, Xi'an, China
| |
Collapse
|
8
|
Yang W, Zhu Y, Tang F, Jian Z, Xiao Y. Cardiac proteomic profiling suggests that hypertrophic and dilated cardiomyopathy share a common pathogenetic pathway of the calcium signalling pathway. Eur J Clin Invest 2023; 53:e14051. [PMID: 37381592 DOI: 10.1111/eci.14051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 06/04/2023] [Accepted: 06/21/2023] [Indexed: 06/30/2023]
Abstract
OBJECTIVE Hypertrophic cardiomyopathy (HCM) and dilated cardiomyopathy (DCM) are classified as different diseases but have many similar pathogenic genes and clinical symptoms. Previous research has focused on mutated genes. This study was conducted to identify key molecular mechanisms and explore effective therapeutic targets. METHODS Myocardial tissue was harvested from patients with HCM (n = 3) or DCM (n = 4) during surgery. Hearts donated by healthy traffic accident victims were treated as controls (n = 4). Total proteins were extracted for liquid chromatography-tandem mass spectrometry. Differentially expressed proteins (DEPs) were annotated via GO and KEGG analyses. Selected distinguishing protein abundance was confirmed by western blotting. RESULTS Compared with the control group, there were 121 and 76 DEPs in the HCM and DCM groups, respectively. GO terms for these two comparisons are associated with contraction-related components and actin binding. Additionally, the most significantly upregulated and downregulated proteins were periostin and tropomyosin alpha-3 chain in both comparisons. Moreover, when comparing the HCM and DCM groups, we found 60 significant DEPs, and the GO and KEGG terms are related to the calcium signalling pathway. Expression of the calcium regulation-related protein peptidyl-prolyl cis-trans isomerase (FKBP1A) was significantly upregulated in multiple samples. CONCLUSION HCM and DCM have many mutual pathogenetic pathways. Calcium ion-related processes are among the most significant factors affecting disease development. For HCM and DCM, research on regulating linchpin protein expression or interfering with key calcium-related pathways may be more beneficial than genetic research.
Collapse
Affiliation(s)
- Wenjuan Yang
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Yu Zhu
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Army Medical University, Chongqing, China
- Department of Cardiovascular Surgery, Hainan Hospital of Chinese PLA General Hospital, Sanya, China
| | - Fuqin Tang
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Zhao Jian
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Yingbin Xiao
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Army Medical University, Chongqing, China
| |
Collapse
|
9
|
Harutyunyan KR, Abrahamyan HT, Adamyan SH, Mkrtchian S, Ter-Markosyan AS. Calcium-Regulating Hormonal System and HMGB1 in Cardiomyopathies. Endocr Metab Immune Disord Drug Targets 2023; 23:115-121. [PMID: 35980074 DOI: 10.2174/1871530322666220817110538] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 05/31/2022] [Accepted: 06/03/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND Calcium ions play a key role in the heart's functional activity. The steadystate levels of calcium are contingent on the calcium regulating hormonal system, impairment of which might result in the development of cardiac pathology. An important role in these processes is also attributed to the specific inflammatory mediator, HMGB1, one of the damage-associated molecular patterns (DAMPs) released by immune cells or cell damage. OBJECTIVE This study investigated the cardioprotective potential of the calcium-regulating hormonal system in cardiomyopathies with an emphasis on the possible role of HMGB1. METHODS Ca2+ and inorganic phosphate levels were determined in the serum using an electrolyte analyzer and spectrophotometric analyzer correspondingly. The 1-34 fragment of parathyroid hormone (PTH), calcitonin, vitamin D, and HMGB1 were detected using ELISA kits. RESULTS The levels of PTH, calcitonin, phosphate, and HMGB1 were found elevated in females suffering from cardiomyopathy. The same tendency was observed in men; however, statistically significant changes were registered only for PTH and phosphate. CONCLUSION It can be suggested that among other reasons, the decrease of the left ventricular function in cardiomyopathy patients can be linked to the high HMGB1, whereas the activation of the calciumregulating system as manifested by the elevated PTH aims at restoration of calcium homeostasis and thus have positive, i.e. cardioprotective consequences.
Collapse
|
10
|
Ji L, Chen J, Qin Y, Yang S. Case report: Rare restrictive cardiomyopathy with ventricular fibrillation as initial symptom rescued by automatic external defibrillator in a pediatric patient. Front Cardiovasc Med 2022; 9:1058341. [DOI: 10.3389/fcvm.2022.1058341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 10/31/2022] [Indexed: 11/13/2022] Open
Abstract
Restrictive cardiomyopathy (RCM) is a rare form of heart muscle disease with poor prognosis. Its primary manifestations were caused by systemic or pulmonary circulation congestion. Here, we reported a case of RCM with ventricular fibrillation as initial symptom in a 7-year-old boy. The child suffered cardiac and respiratory arrest suddenly while exercising at school and immediately was given external chest compression and defibrillation by the school’s equipped automatic external defibrillator (AED). The rescue was successful. At the time of the AED discharge, his electrocardiogram (ECG) indicated ventricular fibrillation. Upon further examination, the echocardiogram revealed enlarged bilateral atria, decreased diastolic function and normal ventricular thickness. Genetic analysis identified a heterozygous missense mutation [c.611(exon8)G>A,p.R204H] of TNNI3 in the proband boy. This case contributes to our understanding of RCM in children and emphasizes the importance of having AEDs available in public places.
Collapse
|
11
|
Chen H, Chen C, Spanos M, Li G, Lu R, Bei Y, Xiao J. Exercise training maintains cardiovascular health: signaling pathways involved and potential therapeutics. Signal Transduct Target Ther 2022; 7:306. [PMID: 36050310 PMCID: PMC9437103 DOI: 10.1038/s41392-022-01153-1] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/22/2022] [Accepted: 08/12/2022] [Indexed: 11/09/2022] Open
Abstract
Exercise training has been widely recognized as a healthy lifestyle as well as an effective non-drug therapeutic strategy for cardiovascular diseases (CVD). Functional and mechanistic studies that employ animal exercise models as well as observational and interventional cohort studies with human participants, have contributed considerably in delineating the essential signaling pathways by which exercise promotes cardiovascular fitness and health. First, this review summarizes the beneficial impact of exercise on multiple aspects of cardiovascular health. We then discuss in detail the signaling pathways mediating exercise's benefits for cardiovascular health. The exercise-regulated signaling cascades have been shown to confer myocardial protection and drive systemic adaptations. The signaling molecules that are necessary for exercise-induced physiological cardiac hypertrophy have the potential to attenuate myocardial injury and reverse cardiac remodeling. Exercise-regulated noncoding RNAs and their associated signaling pathways are also discussed in detail for their roles and mechanisms in exercise-induced cardioprotective effects. Moreover, we address the exercise-mediated signaling pathways and molecules that can serve as potential therapeutic targets ranging from pharmacological approaches to gene therapies in CVD. We also discuss multiple factors that influence exercise's effect and highlight the importance and need for further investigations regarding the exercise-regulated molecules as therapeutic targets and biomarkers for CVD as well as the cross talk between the heart and other tissues or organs during exercise. We conclude that a deep understanding of the signaling pathways involved in exercise's benefits for cardiovascular health will undoubtedly contribute to the identification and development of novel therapeutic targets and strategies for CVD.
Collapse
Affiliation(s)
- Huihua Chen
- School of Basic Medical Science, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Chen Chen
- Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai, 200444, China.,Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science, Shanghai University, Shanghai, 200444, China
| | - Michail Spanos
- Cardiovascular Division of the Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Guoping Li
- Cardiovascular Division of the Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Rong Lu
- School of Basic Medical Science, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Yihua Bei
- Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai, 200444, China. .,Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science, Shanghai University, Shanghai, 200444, China.
| | - Junjie Xiao
- Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai, 200444, China. .,Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science, Shanghai University, Shanghai, 200444, China.
| |
Collapse
|
12
|
Malakootian M, Bagheri Moghaddam M, Kalayinia S, Farrashi M, Maleki M, Sadeghipour P, Amin A. Dilated cardiomyopathy caused by a pathogenic nucleotide variant in RBM20 in an Iranian family. BMC Med Genomics 2022; 15:106. [PMID: 35527250 PMCID: PMC9079971 DOI: 10.1186/s12920-022-01262-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 05/05/2022] [Indexed: 12/22/2022] Open
Abstract
Abstract
Introduction
Dilated cardiomyopathy (DCM) is characterized by the dilation and impaired contraction of 1 or both ventricles and can be caused by a variety of disorders. Up to 50% of idiopathic DCM cases have heritable familial diseases, and the clinical screening of family members is recommended. Identifying a genetic cause that can explain the DCM risk in the family can help with better screening planning and clinical decision-making. Whole-exome sequencing (WES) has aided significantly in the detection of causative genes in many genetically heterogeneous diseases. In the present study, we applied WES to identify the causative genetic variant in a family with heritable DCM.
Methods
WES was applied to identify genetic variants on a 26-year-old man as the proband of a family with DCM. Subsequently, Sanger sequencing was performed to confirm the variant in the patient and all the available affected and unaffected family members. The pathogenicity of the variant was evaluated through co-segregation analysis in the family and employment of in silico predictive software.
Results
WES demonstrated the missense pathogenic heterozygous nucleotide variant, c.1907G > A, (p.Arg636His, rs267607004, NM_0011343), in exon 9 of the RBM20 gene in the proband. The variant was co-segregated in all the affected family members in a heterozygous form and the unaffected family members. The in silico analysis confirmed the variant as pathogenic.
Conclusion
Pathogenic RBM20 nucleotide variants are associated with arrhythmogenic DCM. We believe that our report is the first to show an RBM20 variant in Iranian descent associated with DCM.
Collapse
|
13
|
Tomczyk MM, Cheung KG, Xiang B, Tamanna N, Fonseca Teixeira AL, Agarwal P, Kereliuk SM, Spicer V, Lin L, Treberg J, Tong Q, Dolinsky VW. Mitochondrial Sirtuin-3 (SIRT3) Prevents Doxorubicin-Induced Dilated Cardiomyopathy by Modulating Protein Acetylation and Oxidative Stress. Circ Heart Fail 2022; 15:e008547. [PMID: 35418250 PMCID: PMC9117478 DOI: 10.1161/circheartfailure.121.008547] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND High doses of doxorubicin put cancer patients at risk for developing dilated cardiomyopathy. Previously, we showed that doxorubicin treatment decreases SIRT3 (sirtuin 3), the main mitochondrial deacetylase and increases protein acetylation in rat cardiomyocytes. Here, we hypothesize that SIRT3 expression can attenuate doxorubicin induced dilated cardiomyopathy in vivo by preventing the acetylation of mitochondrial proteins. METHODS Nontransgenic, M3-SIRT3 (truncated SIRT3; short isoform), and M1-SIRT3 (full-length SIRT3; mitochondrial localized) transgenic mice were treated with doxorubicin for 4 weeks (8 mg/kg body weight per week). Echocardiography was performed to assess cardiac structure and function and validated by immunohistochemistry and immunofluorescence (n=4-10). Mass spectrometry was performed on cardiac mitochondrial peptides in saline (n=6) and doxorubicin (n=5) treated hearts. Validation was performed in doxorubicin treated primary rat and human induced stem cell derived cardiomyocytes transduced with adenoviruses for M3-SIRT3 and M1-SIRT3 and deacetylase deficient mutants (n=4-10). RESULTS Echocardiography revealed that M3-SIRT3 transgenic mice were partially resistant to doxorubicin induced changes to cardiac structure and function whereas M1-SIRT3 expression prevented cardiac remodeling and dysfunction. In doxorubicin hearts, 37 unique acetylation sites on mitochondrial proteins were altered. Pathway analysis revealed these proteins are involved in energy production, fatty acid metabolism, and oxidative stress resistance. Increased M1-SIRT3 expression in primary rat and human cardiomyocytes attenuated doxorubicin-induced superoxide formation, whereas deacetylase deficient mutants were unable to prevent oxidative stress. CONCLUSIONS Doxorubicin reduced SIRT3 expression and markedly affected the cardiac mitochondrial acetylome. Increased M1-SIRT3 expression in vivo prevented doxorubicin-induced cardiac dysfunction, suggesting that SIRT3 could be a potential therapeutic target for mitigating doxorubicin-induced dilated cardiomyopathy.
Collapse
Affiliation(s)
- Mateusz M Tomczyk
- Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Theme of the Children's Hospital Research Institute of Manitoba' Winnipeg' Canada (M.M.T., K.G.C., B.X., P.A., S.M.K., V.W.D.).,Department of Pharmacology and Therapeutics (M.M.T., K.G.C., B.X., P.A., S.M.K., V.W.D.), University of Manitoba, Winnipeg, Canada.,Rady Faculty of Health Science, College of Medicine (M.M.T., K.G.C., B.X., P.A., S.M.K., V.S., V.W.D.), University of Manitoba, Winnipeg, Canada
| | - Kyle G Cheung
- Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Theme of the Children's Hospital Research Institute of Manitoba' Winnipeg' Canada (M.M.T., K.G.C., B.X., P.A., S.M.K., V.W.D.).,Department of Pharmacology and Therapeutics (M.M.T., K.G.C., B.X., P.A., S.M.K., V.W.D.), University of Manitoba, Winnipeg, Canada.,Rady Faculty of Health Science, College of Medicine (M.M.T., K.G.C., B.X., P.A., S.M.K., V.S., V.W.D.), University of Manitoba, Winnipeg, Canada
| | - Bo Xiang
- Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Theme of the Children's Hospital Research Institute of Manitoba' Winnipeg' Canada (M.M.T., K.G.C., B.X., P.A., S.M.K., V.W.D.).,Department of Pharmacology and Therapeutics (M.M.T., K.G.C., B.X., P.A., S.M.K., V.W.D.), University of Manitoba, Winnipeg, Canada.,Rady Faculty of Health Science, College of Medicine (M.M.T., K.G.C., B.X., P.A., S.M.K., V.S., V.W.D.), University of Manitoba, Winnipeg, Canada
| | - Nahid Tamanna
- Department of Biological Sciences (N.T., A.L.F.T., J.T.), University of Manitoba, Winnipeg, Canada
| | - Ana L Fonseca Teixeira
- Department of Biological Sciences (N.T., A.L.F.T., J.T.), University of Manitoba, Winnipeg, Canada
| | - Prasoon Agarwal
- Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Theme of the Children's Hospital Research Institute of Manitoba' Winnipeg' Canada (M.M.T., K.G.C., B.X., P.A., S.M.K., V.W.D.).,Department of Pharmacology and Therapeutics (M.M.T., K.G.C., B.X., P.A., S.M.K., V.W.D.), University of Manitoba, Winnipeg, Canada.,Rady Faculty of Health Science, College of Medicine (M.M.T., K.G.C., B.X., P.A., S.M.K., V.S., V.W.D.), University of Manitoba, Winnipeg, Canada.,KTH Royal Institute of Technology, School of Electrical Engineering and Computer Science, Stockholm, Sweden (P.A.).,Science for Life Laboratory, Solna, Sweden (P.A.)
| | - Stephanie M Kereliuk
- Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Theme of the Children's Hospital Research Institute of Manitoba' Winnipeg' Canada (M.M.T., K.G.C., B.X., P.A., S.M.K., V.W.D.).,Department of Pharmacology and Therapeutics (M.M.T., K.G.C., B.X., P.A., S.M.K., V.W.D.), University of Manitoba, Winnipeg, Canada.,Rady Faculty of Health Science, College of Medicine (M.M.T., K.G.C., B.X., P.A., S.M.K., V.S., V.W.D.), University of Manitoba, Winnipeg, Canada
| | - Victor Spicer
- Department of Internal Medicine (V.S.), University of Manitoba, Winnipeg, Canada.,Rady Faculty of Health Science, College of Medicine (M.M.T., K.G.C., B.X., P.A., S.M.K., V.S., V.W.D.), University of Manitoba, Winnipeg, Canada.,Manitoba Center for Proteomics and Systems Biology, Winnipeg, Canada (V.S.)
| | - Ligen Lin
- Children's Nutrition Research Center, Baylor College of Medicine, Houston, TX (L.L., Q.T.).,Institute of Chinese Medical Sciences, University of Macau, China (L.L.)
| | - Jason Treberg
- Department of Biological Sciences (N.T., A.L.F.T., J.T.), University of Manitoba, Winnipeg, Canada
| | - Qiang Tong
- Children's Nutrition Research Center, Baylor College of Medicine, Houston, TX (L.L., Q.T.)
| | - Vernon W Dolinsky
- Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Theme of the Children's Hospital Research Institute of Manitoba' Winnipeg' Canada (M.M.T., K.G.C., B.X., P.A., S.M.K., V.W.D.).,Department of Pharmacology and Therapeutics (M.M.T., K.G.C., B.X., P.A., S.M.K., V.W.D.), University of Manitoba, Winnipeg, Canada.,Rady Faculty of Health Science, College of Medicine (M.M.T., K.G.C., B.X., P.A., S.M.K., V.S., V.W.D.), University of Manitoba, Winnipeg, Canada
| |
Collapse
|
14
|
Szabó D, Sárszegi Z, Polgár B, Sághy É, Reglődi D, Tóth T, Onódi Z, Leszek P, Varga ZV, Helyes Z, Kemény Á, Ferdinandy P, Tamás A. PACAP-38 and PAC1 Receptor Alterations in Plasma and Cardiac Tissue Samples of Heart Failure Patients. Int J Mol Sci 2022; 23:ijms23073715. [PMID: 35409075 PMCID: PMC8998504 DOI: 10.3390/ijms23073715] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/23/2022] [Accepted: 03/24/2022] [Indexed: 02/07/2023] Open
Abstract
Pituitary adenylate cyclase activating polypeptide-38 (PACAP-38) is a multifunctional neuropeptide, which may play a role in cardioprotection. However, little is known about the presence of PACAP-38 in heart failure (HF) patients. The aim of our study was to measure the alterations of PACAP-38 like immunoreactivity (LI) in acute (n = 13) and chronic HF (n = 33) and to examine potential correlations between PACAP-38 and HF predictors (cytokines, NT-proBNP). Tissue PACAP-38 LI and PAC1 receptor levels were also investigated in heart tissue samples of patients with HF. Significantly higher plasma PACAP-38 LI was detected in patients with acute HF, while in chronic HF patients, a lower level of immunoreactivity was observed compared to healthy controls (n = 13). Strong negative correlation was identified between plasma PACAP-38 and NT-proBNP levels in chronic HF, as opposed to the positive connection seen in the acute HF group. Plasma IL-1 β, IL-2 and IL-4 levels were significantly lower in chronic HF, and IL-10 was significantly higher in patients with acute HF. PACAP-38 levels of myocardial tissues were lower in all end-stage HF patients and lower PAC1 receptor levels were detected in the primary dilated cardiomyopathy group compared to the controls. We conclude that PACAP-38 and PAC1 expression correlates with some biomarkers of acute and chronic HF; therefore, further studies are necessary to explore whether PACAP could be a suitable prognostic biomarker in HF patients.
Collapse
Affiliation(s)
- Dóra Szabó
- Heart Institute, Clinical Centre, Medical School, University of Pecs, 7624 Pecs, Hungary; (D.S.); (Z.S.)
- Department of Anatomy, MTA-PTE PACAP Research Team, Centre for Neuroscience, Medical School, University of Pecs, 7624 Pecs, Hungary; (D.R.); (T.T.)
- Szentagothai Research Centre, University of Pecs, 7624 Pecs, Hungary; (Z.H.); (Á.K.)
| | - Zsolt Sárszegi
- Heart Institute, Clinical Centre, Medical School, University of Pecs, 7624 Pecs, Hungary; (D.S.); (Z.S.)
| | - Beáta Polgár
- Department of Medical Microbiology and Immunology, Clinical Centre, Medical School, University of Pecs, 7624 Pecs, Hungary;
| | - Éva Sághy
- Cardiometabolic Research Group, MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1089 Budapest, Hungary; (É.S.); (Z.O.); (Z.V.V.); (P.F.)
| | - Dóra Reglődi
- Department of Anatomy, MTA-PTE PACAP Research Team, Centre for Neuroscience, Medical School, University of Pecs, 7624 Pecs, Hungary; (D.R.); (T.T.)
- Szentagothai Research Centre, University of Pecs, 7624 Pecs, Hungary; (Z.H.); (Á.K.)
| | - Tünde Tóth
- Department of Anatomy, MTA-PTE PACAP Research Team, Centre for Neuroscience, Medical School, University of Pecs, 7624 Pecs, Hungary; (D.R.); (T.T.)
- Szentagothai Research Centre, University of Pecs, 7624 Pecs, Hungary; (Z.H.); (Á.K.)
| | - Zsófia Onódi
- Cardiometabolic Research Group, MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1089 Budapest, Hungary; (É.S.); (Z.O.); (Z.V.V.); (P.F.)
- HCEMM-SU Cardiometabolic Immunology Research Group, Semmelweis University, 1089 Budapest, Hungary
| | - Przemyslaw Leszek
- Department of Heart Failure and Transplantology, Cardinal Stefan Wyszyński National Institute of Cardiology, 04-628 Warszawa, Poland;
| | - Zoltán V. Varga
- Cardiometabolic Research Group, MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1089 Budapest, Hungary; (É.S.); (Z.O.); (Z.V.V.); (P.F.)
- HCEMM-SU Cardiometabolic Immunology Research Group, Semmelweis University, 1089 Budapest, Hungary
| | - Zsuzsanna Helyes
- Szentagothai Research Centre, University of Pecs, 7624 Pecs, Hungary; (Z.H.); (Á.K.)
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pecs, 7624 Pecs, Hungary
| | - Ágnes Kemény
- Szentagothai Research Centre, University of Pecs, 7624 Pecs, Hungary; (Z.H.); (Á.K.)
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pecs, 7624 Pecs, Hungary
- Department of Medical Biology, Medical School, University of Pecs, 7624 Pecs, Hungary
| | - Péter Ferdinandy
- Cardiometabolic Research Group, MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1089 Budapest, Hungary; (É.S.); (Z.O.); (Z.V.V.); (P.F.)
- Pharmahungary Group, 6720 Szeged, Hungary
| | - Andrea Tamás
- Department of Anatomy, MTA-PTE PACAP Research Team, Centre for Neuroscience, Medical School, University of Pecs, 7624 Pecs, Hungary; (D.R.); (T.T.)
- Szentagothai Research Centre, University of Pecs, 7624 Pecs, Hungary; (Z.H.); (Á.K.)
- Correspondence: or ; Tel.: +36-72-536-001 (ext. 36421)
| |
Collapse
|
15
|
Chahine M, Fontaine JM, Boutjdir M. Racial Disparities in Ion Channelopathies and Inherited Cardiovascular Diseases Associated With Sudden Cardiac Death. J Am Heart Assoc 2022; 11:e023446. [PMID: 35243873 PMCID: PMC9075281 DOI: 10.1161/jaha.121.023446] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 11/11/2021] [Indexed: 12/19/2022]
Abstract
Cardiovascular disease (CVD) continues to be the most common cause of death worldwide, and cardiac arrhythmias account for approximately one half of these deaths. The morbidity and mortality from CVD have been reduced significantly over the past few decades; however, disparities in racial or ethnic populations still exist. This review is based on available literature to date and focuses on known cardiac channelopathies and other inherited disorders associated with sudden cardiac death in African American/Black subjects and the role of epigenetics in phenotypic manifestations of CVD, and illustrates existing disparities in treatment and outcomes. The review also highlights the knowledge gaps that limit understanding of the manifestation of phenotypic abnormalities across racial or ethnic groups and discusses disparities associated with device underuse in the management of patients at risk for sudden cardiac death. We discuss factors related to reports in the United States, that the overall mortality attributed to CVD and the number of out-of-hospital cardiac arrests are higher among African American/Black subjects when compared with other racial or ethnic groups. African American/Black subjects are disproportionally affected by CVD, including cardiac arrhythmias and sudden cardiac death, thus highlighting a major concern in this population that remains underrepresented in clinical trials with limited genetic testing and device underuse. The proposed solutions include (1) early identification of genetic variants, which is crucial in tailoring a preventive management strategy; (2) inclusion of diverse racial or ethnic groups in clinical trials; (3) compliance with guideline-directed medical treatment and referral to cardiovascular subspecialists; and (4) training and mentoring of underrepresented junior faculty in cardiovascular health disparities research.
Collapse
Affiliation(s)
- Mohamed Chahine
- Department of MedicineFaculty of MedicineUniversité LavalQuebec CityQCCanada
- CERVO Brain Research CenterQuebec CityQCCanada
| | - John M. Fontaine
- University of Pittsburgh Medical CenterWilliamsportPA
- University of Central Florida School of Medicine Affiliate–West Florida HospitalPensacolaFL
| | - Mohamed Boutjdir
- Cardiovascular Research ProgramVeterans Administration New York Harbor Healthcare SystemNew YorkNY
- Department of Medicine, Cell Biology and PharmacologyState University of New York Downstate Medical CenterNew YorkNY
- Department of MedicineNew York University School of MedicineNew YorkNY
| |
Collapse
|
16
|
Hua CC, Liu XM, Liang LR, Wang LF, Zhong JC. Targeting the microRNA-34a as a Novel Therapeutic Strategy for Cardiovascular Diseases. Front Cardiovasc Med 2022; 8:784044. [PMID: 35155600 PMCID: PMC8828972 DOI: 10.3389/fcvm.2021.784044] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 12/28/2021] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular diseases (CVDs) are still the main cause of morbidity and mortality worldwide and include a group of disorders varying from vasculature, myocardium, arrhythmias and cardiac development. MicroRNAs (miRs) are endogenous non-coding RNAs with 18–23 nucleotides that regulate gene expression. The miR-34 family, including miR-34a/b/c, plays a vital role in the regulation of myocardial physiology and pathophysiological processes. Recently, miR-34a has been implicated in cardiovascular fibrosis, dysfunction and related cardiovascular disorders as an essential regulator. Interestingly, there is a pivotal link among miR-34a, cardiovascular fibrosis, and Smad4/TGF-β1 signaling. Notably, both loss-of-function and gain-of-function approaches identified the critical roles of miR-34a in cardiovascular apoptosis, autophagy, inflammation, senescence and remodeling by modulating multifunctional signaling pathways. In this article, we focus on the current understanding of miR-34a in biogenesis, its biological effects and its implications for cardiac pathologies including myocardial infarction, heart failure, ischaemia reperfusion injury, cardiomyopathy, atherosclerosis, hypertension and atrial fibrillation. Thus, further understanding of the effects of miR-34a on cardiovascular diseases will aid the development of effective interventions. Targeting for miR-34a has emerged as a potential therapeutic target for cardiovascular dysfunction and related diseases.
Collapse
Affiliation(s)
- Cun-Cun Hua
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Xin-Ming Liu
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Li-Rong Liang
- Department of Clinical Epidemiology and Tobacco Dependence Treatment Research, Beijing Institute of Respiratory Medicine, Beijing, China
| | - Le-Feng Wang
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
- *Correspondence: Jiu-Chang Zhong
| | - Jiu-Chang Zhong
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
- Department of Clinical Epidemiology and Tobacco Dependence Treatment Research, Beijing Institute of Respiratory Medicine, Beijing, China
- Beijing Institute of Respiratory Medicine, Capital Medical University, Beijing, China
- Le-Feng Wang
| |
Collapse
|
17
|
Li TT, Li HY, Cheng J. Changes of serum uric acid and its clinical correlation in children with dilated cardiomyopathy. Transl Pediatr 2021; 10:3211-3217. [PMID: 35070835 PMCID: PMC8753472 DOI: 10.21037/tp-21-537] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Accepted: 12/14/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Dilated cardiomyopathy (DCM) is the most common type of childhood cardiomyopathy and uric acid (UA) is considered closely associated with cardiovascular disease. There are few reports about the relationship between serum UA level and DCM in children, and the present study aimed to analyze the changes and clinical correlation of the two. METHODS The clinical data of 49 children under 16 years old and who were hospitalized with DCM, and 44 healthy children who underwent physical examination in the same period at Tianjin Children's Hospital from June 2015 to November 2019 were analyzed retrospectively. RESULTS The 49 children in the case group included 17 males and 32 females, aged from 2 to 172 months. The case group were divided into New York Heart Association (NYHA) functional class I (n=2), class II (n=17), class III (n=11), and class IV (n=19). The 44 healthy children selected as the control group included 20 males and 24 females aged from 2 to 161 months. The serum UA level was detected, and an ultrasonic cardiogram was conducted in each child. The serum UA level, left ventricular end-diastolic diameter (LVEDD), left ventricular end-systolic diameter (LVESD), and left atrial diameter (LAD) of the case group were higher than that of the control group, while the left ventricular ejection fraction (LVEF) and left ventricular fractional shortening (LVFS) were lower than that of the control group, and significant statistical differences were seen between the two groups (P<0.01). The serum UA level, LVEDD, LVESD, and LAD of NYHAIII-IV class patients were higher than that of the NYHAI-II class, but LVEF and LVFS were lower than that of the NYHA I-II class, and there were significant statistical differences between the two groups (P<0.01). Statistical correlations were seen between the serum UA level and NYHA functional class, LVEDD, LVESD, LAD, LVEF, and LVFS (rs=0.599, 0.567, 0.579, 0.475, -0.333, -0.341, respectively, P<0.05). CONCLUSIONS Elevated serum UA levels exist in children with DCM and correlate with NYHA functional class and ultrasonic values. Change in serum UA levels may be used as a biomarker reflecting the severity of DCM in children.
Collapse
Affiliation(s)
- Ting-Ting Li
- Department of Cardiology, Tianjin Children's Hospital/Tianjin University Children's Hospital, Tianjin, China
| | - Hao-Ying Li
- Department of Cardiology, Tianjin Children's Hospital/Tianjin University Children's Hospital, Tianjin, China
| | - Ji Cheng
- Department of Cardiology, Tianjin Children's Hospital/Tianjin University Children's Hospital, Tianjin, China
| |
Collapse
|
18
|
Circ-SIRT1 inhibits cardiac hypertrophy via activating SIRT1 to promote autophagy. Cell Death Dis 2021; 12:1069. [PMID: 34759275 PMCID: PMC8580993 DOI: 10.1038/s41419-021-04059-y] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Revised: 07/20/2021] [Accepted: 07/26/2021] [Indexed: 12/18/2022]
Abstract
Mounting studies have substantiated that abrogating autophagy contributes to cardiac hypertrophy (CH). Sirtuin 1 (SIRT1) has been reported to support autophagy and inhibit CH. However, the upstream regulation mechanism behind the regulation of SIRT1 level in CH remains unclear. Circular RNAs (circRNAs) are vital modulators in diverse human diseases including CH. This study intended to investigate the regulatory mechanism of circRNA on SIRT1 expression in CH. CH model was established by angiotensin II (Ang II) fusion or transverse aortic constriction (TAC) surgery and Ang II treatment on hiPSC-CMs and H9c2 cells in vitro. Our results showed that circ-SIRT1 (hsa_circ_0093884) expression was downregulated in Ang II-treated hiPSC-CMs, and confirmed that its conserved mouse homolog circ-Sirt1 (mmu_circ_0002354) was expressed at low levels in Ang II-treated H9c2 cells and TAC-induced mice model. Functionally, circ-SIRT1/circ-Sirt1 attenuated Ang II-induced CH and induced autophagy in hiPSC-CMs and H9c2 cardiomyocytes. Mechanistically, circ-SIRT1 could upregulate its host gene SIRT1 at the post-transcriptional level by sponging miR-3681-3p/miR-5195-3p and stabilized SIRT1 protein at the post-translational level by recruiting USP22 to induce deubiquitination on SIRT1 protein. Further, SIRT1 knockdown could rescue the effect of circ-SIRT1 upregulation on Ang II-induced CH and autophagy in vitro and in vivo. In conclusion, we first uncovered that circ-SIRT1 restrains CH via activating SIRT1 to promote autophagy, indicating circ-SIRT1 as a promising target to alleviate CH.
Collapse
|
19
|
Abstract
Practical relevance: Hypertrophic cardiomyopathy (HCM) is the most common form of feline
cardiomyopathy observed clinically and may affect up to approximately 15% of
the domestic cat population, primarily as a subclinical disease.
Fortunately, severe HCM, leading to heart failure or arterial
thromboembolism (ATE), only occurs in a small proportion of these cats. Patient group: Domestic cats of any age from 3 months upward, of either sex and of any
breed, can be affected. A higher prevalence in male and domestic shorthair
cats has been reported. Diagnostics: Subclinical feline HCM may or may not produce a heart murmur or gallop sound.
Substantial left atrial enlargement can often be identified radiographically
in cats with severe HCM. Biomarkers should not be relied on solely to
diagnose the disease. While severe feline HCM can usually be diagnosed via
echocardiography alone, feline HCM with mild to moderate left ventricular
(LV) wall thickening is a diagnosis of exclusion, which means there is no
definitive test for HCM in these cats and so other disorders that can cause
mild to moderate LV wall thickening (eg, hyperthyroidism, systemic
hypertension, acromegaly, dehydration) need to be ruled out. Key findings: While a genetic cause of HCM has been identified in two breeds and is
suspected in another, for most cats the cause is unknown. Systolic anterior
motion of the mitral valve (SAM) is the most common cause of dynamic left
ventricular outflow tract obstruction (DLVOTO) and, in turn, the most common
cause of a heart murmur with feline HCM. While severe DLVOTO is probably
clinically significant and so should be treated, lesser degrees probably are
not. Furthermore, since SAM can likely be induced in most cats with HCM, the
distinction between HCM without obstruction and HCM with obstruction (HOCM)
is of limited importance in cats. Diastolic dysfunction, and its
consequences of abnormally increased atrial pressure leading to signs of
heart failure, and sluggish atrial blood flow leading to ATE, is the primary
abnormality that causes clinical signs and death in affected cats. Treatment
(eg, loop diuretics) is aimed at controlling heart failure. Preventive
treatment (eg, antithrombotic drugs) is aimed at reducing the risk of
complications (eg, ATE). Conclusions: Most cats with HCM show no overt clinical signs and live a normal or
near-normal life despite this disease. However, a substantial minority of
cats develop overt clinical signs referable to heart failure or ATE that
require treatment. For most cats with clinical signs caused by HCM, the
long-term prognosis is poor to grave despite therapy. Areas of uncertainty: Genetic mutations (variants) that cause HCM have been identified in a few
breeds, but, despite valiant efforts, the cause of HCM in the vast majority
of cats remains unknown. No treatment currently exists that reverses or even
slows the cardiomyopathic process in HCM, again despite valiant efforts. The
search goes on.
Collapse
Affiliation(s)
- Mark D Kittleson
- School of Veterinary Medicine, Department of Medicine and Epidemiology, University of California, Davis, and Veterinary Information Network, 777 West Covell Boulevard, Davis, CA 95616, USA
| | - Etienne Côté
- Department of Companion Animals, Atlantic Veterinary College, University of Prince Edward Island, Charlottetown, Prince Edward Island, Canada
| |
Collapse
|
20
|
Yeh JN, Yue Y, Chu YC, Huang CR, Yang CC, Chiang JY, Yip HK, Guo J. Entresto protected the cardiomyocytes and preserved heart function in cardiorenal syndrome rat fed with high-protein diet through regulating the oxidative stress and Mfn2-mediated mitochondrial functional integrity. Biomed Pharmacother 2021; 144:112244. [PMID: 34601193 DOI: 10.1016/j.biopha.2021.112244] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 09/15/2021] [Accepted: 09/22/2021] [Indexed: 12/11/2022] Open
Abstract
This study tested the hypothesis that Entresto (En) therapy protected the cardiomyocytes and heart function in cardiorenal syndrome (CRS) rats fed with high-protein diet (HPD) through regulating the oxidative-stress and Mfn2-mediated mitochondrial functional integrity. En (12.5 μM for the in-vitro study) protected the H9C2-cells against H2O2-induced cell apoptosis, whereas stepwise-increased H2O2 concentrations induced a significant increase in protein expressions of Mfn2/phosphorylated (p)-DRP1/mitochondrial-Bax in H9C2-cells. En downregulated H2O2-induced mitochondrial fission/upregulated mitochondrial fusion and deletion of Mfn2 gene (i.e., shMfn2) to significantly reduce H2O2-induced ROS production. En significantly suppressed and shMfn2 further significantly suppressed both H2O2-reduced mitochondrial-membrane potential and H2O2-induced ROS production/cell apoptosis/mitochondrial damage/mitochondrial-Bax released from mitochondria in H9C2 cells. En significantly reduced protein expressions of Mfn2 and p-DRP1. Additionally, En significantly suppressed and shMfn2 further significantly suppressed the protein expressions of mitochondrial-damaged (DRP1)/oxidative-stress (NOX-1/NOX-2)/apoptosis (mitochondrial-Bax/caspase-3/PARP)/autophagic (LC3B-II/LC3B-I) biomarkers (all p < 0.01). Rats were categorized into group 1 [sham-control + high-protein-diet (HPD)], group 2 (CRS + HPD) and group 3 (CRS+ HPD + En/100 mg/kg/day). By day 63 after CRS induction, the LVEF was significantly lower in group 3 and more significantly lower in group 2 than in group 1, whereas the protein expressions of oxidative-stress (NOX-1/NOX-2/p22phox/oxidized protein)/apoptotic (mitochondrial-Bax/caspase-3/PARP), fibrotic (Smad-3/TGF-ß), autophagic (Beclin-1/Atg5/ratio of LC3B-II/LC3B-I) and mitochondrial-damaged (DRP1/cyclophilin-D/cytosolic-cytochrome-C) biomarkers exhibited an opposite pattern of LVEF among the groups. Downregulation of Mfn2 by En or shMfn2 in cardiomyocytes avoided H2O2 damage and En improved the cardiac function in HPD-feeding CRS rat via adjusting Mfn2-mediated mitochondrial functional integrity.
Collapse
Affiliation(s)
- Jui-Ning Yeh
- Department of Cardiology, The First Affiliated Hospital, Jinan University, 613W. Huangpu Avenue, Guangzhou 510630, China
| | - Ya Yue
- Department of Nephrology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Yi-Ching Chu
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, 123, Dapi Road, Niaosung Dist., Kaohsiung City 83301, Taiwan; Center for Shockwave Medicine and Tissue Engineering, Kaohsiung Chang Gung Memorial Hospital Kaohsiung, Taiwan
| | - Chi-Ruei Huang
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, 123, Dapi Road, Niaosung Dist., Kaohsiung City 83301, Taiwan; Center for Shockwave Medicine and Tissue Engineering, Kaohsiung Chang Gung Memorial Hospital Kaohsiung, Taiwan
| | - Chih-Chao Yang
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - John Y Chiang
- Department of Computer Science and Engineering, National Sun Yat-Sen University, Kaohsiung, Taiwan; Department of Healthcare Administration and Medical Informatics, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Hon-Kan Yip
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, 123, Dapi Road, Niaosung Dist., Kaohsiung City 83301, Taiwan; Center for Shockwave Medicine and Tissue Engineering, Kaohsiung Chang Gung Memorial Hospital Kaohsiung, Taiwan; Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital Kaohsiung, Taiwan; Department of Nursing, Asia University Taichung, Taiwan; Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan; Division of Cardiology, Department of Internal Medicine, Xiamen Chang Gung Hospital, Xiamen, Fujian, China.
| | - Jun Guo
- Department of Cardiology, The First Affiliated Hospital, Jinan University, 613W. Huangpu Avenue, Guangzhou 510630, China.
| |
Collapse
|
21
|
Zhang H, Hua X, Song J. Phenotypes of Cardiovascular Diseases: Current Status and Future Perspectives. PHENOMICS (CHAM, SWITZERLAND) 2021; 1:229-241. [PMID: 36939805 PMCID: PMC9590492 DOI: 10.1007/s43657-021-00022-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 08/11/2021] [Accepted: 08/16/2021] [Indexed: 10/20/2022]
Abstract
Cardiovascular diseases (CVDs) are a large group of diseases and have become the leading cause of morbidity and mortality worldwide. Although considerable progresses have been made in the diagnosis, treatment and prognosis of CVD, communication barriers between clinicians and researchers still exist because the phenotypes of CVD are complex and diverse in clinical practice and lack of unity. Therefore, it is particularly important to establish a standardized and unified terminology to describe CVD. In recent years, there have been several studies, such as the Human Phenotype Ontology, attempting to provide a standardized description of the disease phenotypes. In the present article, we outline recent advances in the classification of the major types of CVD to retrospectively review the current progresses of phenotypic studies in the cardiovascular field and provide a reference for future cardiovascular research.
Collapse
Affiliation(s)
- Hang Zhang
- grid.506261.60000 0001 0706 7839The Cardiomyopathy Research Group, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167A Beilishi Road, Xi Cheng District, Beijing, 100037 China
| | - Xiumeng Hua
- grid.506261.60000 0001 0706 7839The Cardiomyopathy Research Group, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167A Beilishi Road, Xi Cheng District, Beijing, 100037 China
| | - Jiangping Song
- grid.506261.60000 0001 0706 7839The Cardiomyopathy Research Group, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167A Beilishi Road, Xi Cheng District, Beijing, 100037 China
| |
Collapse
|
22
|
Pagiatakis C, Di Mauro V. The Emerging Role of Epigenetics in Therapeutic Targeting of Cardiomyopathies. Int J Mol Sci 2021; 22:ijms22168721. [PMID: 34445422 PMCID: PMC8395924 DOI: 10.3390/ijms22168721] [Citation(s) in RCA: 10] [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: 07/05/2021] [Revised: 08/06/2021] [Accepted: 08/10/2021] [Indexed: 02/06/2023] Open
Abstract
Cardiomyopathies (CMPs) are a heterogeneous group of myocardial diseases accountable for the majority of cases of heart failure (HF) and/or sudden cardiac death (SCD) worldwide. With the recent advances in genomics, the original classification of CMPs on the basis of morphological and functional criteria (dilated (DCM), hypertrophic (HCM), restrictive (RCM), and arrhythmogenic ventricular cardiomyopathy (AVC)) was further refined into genetic (inherited or familial) and acquired (non-inherited or secondary) forms. Despite substantial progress in the identification of novel CMP-associated genetic variations, as well as improved clinical recognition diagnoses, the functional consequences of these mutations and the exact details of the signaling pathways leading to hypertrophy, dilation, and/or contractile impairment remain elusive. To date, global research has mainly focused on the genetic factors underlying CMP pathogenesis. However, growing evidence shows that alterations in molecular mediators associated with the diagnosis of CMPs are not always correlated with genetic mutations, suggesting that additional mechanisms, such as epigenetics, may play a role in the onset or progression of CMPs. This review summarizes published findings of inherited CMPs with a specific focus on the potential role of epigenetic mechanisms in regulating these cardiac disorders.
Collapse
Affiliation(s)
- Christina Pagiatakis
- IRCCS-Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Italy
- Correspondence: (C.P.); (V.D.M.)
| | - Vittoria Di Mauro
- IRCCS-Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Italy
- Institute of Genetic and Biomedical Research (IRGB), Milan Unit, National Research Council, Via Fantoli 16/15, 20138 Milan, Italy
- Correspondence: (C.P.); (V.D.M.)
| |
Collapse
|
23
|
Maiuolo J, Carresi C, Gliozzi M, Musolino V, Scarano F, Coppoletta AR, Guarnieri L, Nucera S, Scicchitano M, Bosco F, Ruga S, Zito MC, Macri R, Cardamone A, Serra M, Mollace R, Tavernese A, Mollace V. Effects of Bergamot Polyphenols on Mitochondrial Dysfunction and Sarcoplasmic Reticulum Stress in Diabetic Cardiomyopathy. Nutrients 2021; 13:nu13072476. [PMID: 34371986 PMCID: PMC8308586 DOI: 10.3390/nu13072476] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/18/2021] [Accepted: 07/18/2021] [Indexed: 12/14/2022] Open
Abstract
Cardiovascular disease is the leading cause of death and disability in the Western world. In order to safeguard the structure and the functionality of the myocardium, it is extremely important to adequately support the cardiomyocytes. Two cellular organelles of cardiomyocytes are essential for cell survival and to ensure proper functioning of the myocardium: mitochondria and the sarcoplasmic reticulum. Mitochondria are responsible for the energy metabolism of the myocardium, and regulate the processes that can lead to cell death. The sarcoplasmic reticulum preserves the physiological concentration of the calcium ion, and triggers processes to protect the structural and functional integrity of the proteins. The alterations of these organelles can damage myocardial functioning. A proper nutritional balance regarding the intake of macronutrients and micronutrients leads to a significant improvement in the symptoms and consequences of heart disease. In particular, the Mediterranean diet, characterized by a high consumption of plant-based foods, small quantities of red meat, and high quantities of olive oil, reduces and improves the pathological condition of patients with heart failure. In addition, nutritional support and nutraceutical supplementation in patients who develop heart failure can contribute to the protection of the failing myocardium. Since polyphenols have numerous beneficial properties, including anti-inflammatory and antioxidant properties, this review gathers what is known about the beneficial effects of polyphenol-rich bergamot fruit on the cardiovascular system. In particular, the role of bergamot polyphenols in mitochondrial and sarcoplasmic dysfunctions in diabetic cardiomyopathy is reported.
Collapse
Affiliation(s)
- Jessica Maiuolo
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, Campus Universitario di Germaneto, 88100 Catanzaro, Italy; (J.M.); (C.C.); (M.G.); (V.M.); (F.S.); (A.R.C.); (L.G.); (S.N.); (M.S.); (F.B.); (S.R.); (M.C.Z.); (R.M.); (A.C.); (M.S.); (R.M.); (A.T.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Cristina Carresi
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, Campus Universitario di Germaneto, 88100 Catanzaro, Italy; (J.M.); (C.C.); (M.G.); (V.M.); (F.S.); (A.R.C.); (L.G.); (S.N.); (M.S.); (F.B.); (S.R.); (M.C.Z.); (R.M.); (A.C.); (M.S.); (R.M.); (A.T.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Micaela Gliozzi
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, Campus Universitario di Germaneto, 88100 Catanzaro, Italy; (J.M.); (C.C.); (M.G.); (V.M.); (F.S.); (A.R.C.); (L.G.); (S.N.); (M.S.); (F.B.); (S.R.); (M.C.Z.); (R.M.); (A.C.); (M.S.); (R.M.); (A.T.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Vincenzo Musolino
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, Campus Universitario di Germaneto, 88100 Catanzaro, Italy; (J.M.); (C.C.); (M.G.); (V.M.); (F.S.); (A.R.C.); (L.G.); (S.N.); (M.S.); (F.B.); (S.R.); (M.C.Z.); (R.M.); (A.C.); (M.S.); (R.M.); (A.T.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Federica Scarano
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, Campus Universitario di Germaneto, 88100 Catanzaro, Italy; (J.M.); (C.C.); (M.G.); (V.M.); (F.S.); (A.R.C.); (L.G.); (S.N.); (M.S.); (F.B.); (S.R.); (M.C.Z.); (R.M.); (A.C.); (M.S.); (R.M.); (A.T.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Anna Rita Coppoletta
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, Campus Universitario di Germaneto, 88100 Catanzaro, Italy; (J.M.); (C.C.); (M.G.); (V.M.); (F.S.); (A.R.C.); (L.G.); (S.N.); (M.S.); (F.B.); (S.R.); (M.C.Z.); (R.M.); (A.C.); (M.S.); (R.M.); (A.T.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Lorenza Guarnieri
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, Campus Universitario di Germaneto, 88100 Catanzaro, Italy; (J.M.); (C.C.); (M.G.); (V.M.); (F.S.); (A.R.C.); (L.G.); (S.N.); (M.S.); (F.B.); (S.R.); (M.C.Z.); (R.M.); (A.C.); (M.S.); (R.M.); (A.T.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Saverio Nucera
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, Campus Universitario di Germaneto, 88100 Catanzaro, Italy; (J.M.); (C.C.); (M.G.); (V.M.); (F.S.); (A.R.C.); (L.G.); (S.N.); (M.S.); (F.B.); (S.R.); (M.C.Z.); (R.M.); (A.C.); (M.S.); (R.M.); (A.T.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Miriam Scicchitano
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, Campus Universitario di Germaneto, 88100 Catanzaro, Italy; (J.M.); (C.C.); (M.G.); (V.M.); (F.S.); (A.R.C.); (L.G.); (S.N.); (M.S.); (F.B.); (S.R.); (M.C.Z.); (R.M.); (A.C.); (M.S.); (R.M.); (A.T.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Francesca Bosco
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, Campus Universitario di Germaneto, 88100 Catanzaro, Italy; (J.M.); (C.C.); (M.G.); (V.M.); (F.S.); (A.R.C.); (L.G.); (S.N.); (M.S.); (F.B.); (S.R.); (M.C.Z.); (R.M.); (A.C.); (M.S.); (R.M.); (A.T.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Stefano Ruga
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, Campus Universitario di Germaneto, 88100 Catanzaro, Italy; (J.M.); (C.C.); (M.G.); (V.M.); (F.S.); (A.R.C.); (L.G.); (S.N.); (M.S.); (F.B.); (S.R.); (M.C.Z.); (R.M.); (A.C.); (M.S.); (R.M.); (A.T.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Maria Caterina Zito
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, Campus Universitario di Germaneto, 88100 Catanzaro, Italy; (J.M.); (C.C.); (M.G.); (V.M.); (F.S.); (A.R.C.); (L.G.); (S.N.); (M.S.); (F.B.); (S.R.); (M.C.Z.); (R.M.); (A.C.); (M.S.); (R.M.); (A.T.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Roberta Macri
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, Campus Universitario di Germaneto, 88100 Catanzaro, Italy; (J.M.); (C.C.); (M.G.); (V.M.); (F.S.); (A.R.C.); (L.G.); (S.N.); (M.S.); (F.B.); (S.R.); (M.C.Z.); (R.M.); (A.C.); (M.S.); (R.M.); (A.T.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Antonio Cardamone
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, Campus Universitario di Germaneto, 88100 Catanzaro, Italy; (J.M.); (C.C.); (M.G.); (V.M.); (F.S.); (A.R.C.); (L.G.); (S.N.); (M.S.); (F.B.); (S.R.); (M.C.Z.); (R.M.); (A.C.); (M.S.); (R.M.); (A.T.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Maria Serra
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, Campus Universitario di Germaneto, 88100 Catanzaro, Italy; (J.M.); (C.C.); (M.G.); (V.M.); (F.S.); (A.R.C.); (L.G.); (S.N.); (M.S.); (F.B.); (S.R.); (M.C.Z.); (R.M.); (A.C.); (M.S.); (R.M.); (A.T.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Rocco Mollace
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, Campus Universitario di Germaneto, 88100 Catanzaro, Italy; (J.M.); (C.C.); (M.G.); (V.M.); (F.S.); (A.R.C.); (L.G.); (S.N.); (M.S.); (F.B.); (S.R.); (M.C.Z.); (R.M.); (A.C.); (M.S.); (R.M.); (A.T.)
- IRCCS San Raffaele, Via di Valcannuta 247, 00133 Rome, Italy
| | - Annamaria Tavernese
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, Campus Universitario di Germaneto, 88100 Catanzaro, Italy; (J.M.); (C.C.); (M.G.); (V.M.); (F.S.); (A.R.C.); (L.G.); (S.N.); (M.S.); (F.B.); (S.R.); (M.C.Z.); (R.M.); (A.C.); (M.S.); (R.M.); (A.T.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Vincenzo Mollace
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, Campus Universitario di Germaneto, 88100 Catanzaro, Italy; (J.M.); (C.C.); (M.G.); (V.M.); (F.S.); (A.R.C.); (L.G.); (S.N.); (M.S.); (F.B.); (S.R.); (M.C.Z.); (R.M.); (A.C.); (M.S.); (R.M.); (A.T.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
- IRCCS San Raffaele, Via di Valcannuta 247, 00133 Rome, Italy
- Correspondence: ; Tel.: +39-327-475-8006
| |
Collapse
|
24
|
Qin J, Guo N, Tong J, Wang Z. Function of histone methylation and acetylation modifiers in cardiac hypertrophy. J Mol Cell Cardiol 2021; 159:120-129. [PMID: 34175302 DOI: 10.1016/j.yjmcc.2021.06.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 06/14/2021] [Accepted: 06/19/2021] [Indexed: 12/15/2022]
Abstract
Cardiac hypertrophy is an adaptive response of the heart to increased workload induced by various physiological or pathological stimuli. It is a common pathological process in multiple cardiovascular diseases, and it ultimately leads to heart failure. The development of cardiac hypertrophy is accompanied by gene expression reprogramming, a process that is largely dependent on epigenetic regulation. Histone modifications such as methylation and acetylation are dynamically regulated under cardiac stress. These consequently contribute to the pathogenesis of cardiac hypertrophy via compensatory or maladaptive transcriptome reprogramming. Histone methylation and acetylation modifiers play crucial roles in epigenetic remodeling during the pathogenesis of cardiac hypertrophy. Regulation of histone methylation and acetylation modifiers serves as a bridge between signal transduction and downstream gene reprogramming. Exploring the role of histone modifiers in cardiac hypertrophy provides novel therapeutic strategies to treat cardiac hypertrophy and heart failure. In this review, we summarize the recent advancements in functional histone methylation and acetylation modifiers in cardiac hypertrophy, with an emphasis on the underlying mechanisms and the therapeutic potential.
Collapse
Affiliation(s)
- Jian Qin
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ningning Guo
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jingjing Tong
- School of Life Sciences, Central China Normal University, Wuhan, China
| | - Zhihua Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen, Shenzhen, China; State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
25
|
Luo J, Zhao W, Gan Y, Pan B, Liu L, Liu Z, Tian J. Cardiac Troponin I R193H Mutation Is Associated with Mitochondrial Damage in Cardiomyocytes. DNA Cell Biol 2021; 40:184-191. [PMID: 33465007 DOI: 10.1089/dna.2020.5828] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Malfunction of myocardial mitochondria plays a crucial role in the development of cardiovascular disorders, especially hypertrophic and dilated cardiomyopathies. Cardiac troponin I (cTnI) is an important structural protein and essential to contraction and relaxation of cardiomyocytes. Recent studies suggest that mutated cTnIR193H could function as a regulatory molecule for other cell functions. This study was to determine whether mutated cTnI could contribute to mitochondrial dysfunction of cardiomyocytes. Primary cardiomyocytes were transfected with cTnIR193H adenovirus with empty vector as control. Mitochondrial structure and function were evaluated in the cells 72 h after transfection. Transmission electron microscopy examination showed mitochondria in the cardiomyocytes with R193H mutation displayed broken cristae, vacuolation, and mitophagy. Mitochondrial function studies revealed a significant decrease in complex I activity, ATP and reactive oxygen species levels, and oxygen consumption rate compared with controls. Western blot analysis demonstrated that expressions of mitochondria-related genes, including ND5 (ubiquinone oxidoreductase chain 5), LRPPRC (a leucine-rich protein of pentatricopeptide repeat family), and PGC-1α (PPARG co-activator 1 alpha), were significantly downregulated in R193H mutation cardiomyocytes compared with the control. Swelling and broken cristae were observed in the mitochondria of cardiomyocytes from cTnIR193H mutation transgenic mice with decreased mitochondrial function, not from the littermate control mice. The data from the present study demonstrated that mitochondrial structure and function were significantly impaired in cardiomyocytes with cTnIR193H mutation, suggesting that cTnI might be critically involved in maintaining the structural and functional integrity of myocardial mitochondria.
Collapse
Affiliation(s)
- Jing Luo
- Department of Cardiovascular Medicine, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, P.R. China
| | - Weian Zhao
- Department of Cardiovascular Medicine, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, P.R. China
| | - Yi Gan
- Department of Cardiovascular Medicine, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, P.R. China
| | - Bo Pan
- Department of Cardiovascular Medicine, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, P.R. China
| | - Lingjuan Liu
- Department of Cardiovascular Medicine, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, P.R. China
| | - Zhenguo Liu
- Department of Medicine, Center for Precision Medicine, University of Missouri School of Medicine, Columbia, Missouri, USA
| | - Jie Tian
- Department of Cardiovascular Medicine, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, P.R. China
| |
Collapse
|
26
|
Stafford F, Thomson K, Butters A, Ingles J. Hypertrophic Cardiomyopathy: Genetic Testing and Risk Stratification. Curr Cardiol Rep 2021; 23:9. [PMID: 33433738 DOI: 10.1007/s11886-020-01437-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/18/2020] [Indexed: 12/13/2022]
Abstract
PURPOSE OF REVIEW Our knowledge of the genetic basis and molecular pathogenesis of hypertrophic cardiomyopathy (HCM) continues to evolve. We describe the genetic basis of HCM, recent advances in genetic testing and the role of genetics in guiding risk stratification and management, both now and in the future. RECENT FINDINGS While initially thought to be an exclusively Mendelian disease, we now know there are important HCM sub-groups. A proportion will have sarcomere variants as the cause of their disease, while others will have genetic variants in genes that can give rise to conditions that can mimic HCM. The role of genetics is primarily for cascade genetic testing, though there is emerging evidence of a role for prognosis and patient management. Genetic testing is a useful addition to management. Genotype may play a greater role in risk stratification, management, treatment and prognosis in future, offering improved outcomes for patients and their families with HCM.
Collapse
Affiliation(s)
- Fergus Stafford
- Cardio Genomics Program at Centenary Institute, The University of Sydney, Locked Bag 6, Newtown, NSW, 2042, Australia
| | - Kate Thomson
- Oxford Medical Genetics Laboratories, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
- Radcliffe Department of Medicine, Division of Cardiovascular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Alexandra Butters
- Cardio Genomics Program at Centenary Institute, The University of Sydney, Locked Bag 6, Newtown, NSW, 2042, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Jodie Ingles
- Cardio Genomics Program at Centenary Institute, The University of Sydney, Locked Bag 6, Newtown, NSW, 2042, Australia.
- Faculty of Medicine and Health, The University of Sydney, Sydney, Australia.
- Department of Cardiology, Royal Prince Alfred Hospital, Sydney, Australia.
| |
Collapse
|
27
|
Zhou Z, Zheng L, Tang C, Chen Z, Zhu R, Peng X, Wu X, Zhu P. Identification of Potentially Relevant Genes for Excessive Exercise-Induced Pathological Cardiac Hypertrophy in Zebrafish. Front Physiol 2020; 11:565307. [PMID: 33329019 PMCID: PMC7734032 DOI: 10.3389/fphys.2020.565307] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 10/05/2020] [Indexed: 12/24/2022] Open
Abstract
Exercise-induced cardiac remodeling has aroused public concern for some time, as sudden cardiac death is known to occur in athletes; however, little is known about the underlying mechanism of exercise-induced cardiac injury. In the present study, we established an excessive exercise-induced pathologic cardiac hypertrophy model in zebrafish with increased myocardial fibrosis, myofibril disassembly, mitochondrial degradation, upregulated expression of the pathological hypertrophy marker genes in the heart, contractile impairment, and cardiopulmonary function impairment. High-throughput RNA-seq analysis revealed that the differentially expressed genes were enriched in the regulation of autophagy, protein folding, and degradation, myofibril development, angiogenesis, metabolic reprogramming, and insulin and FoxO signaling pathways. FOXO proteins may be the core mediator of the regulatory network needed to promote the pathological response. Further, PPI network analysis showed that pik3c3, gapdh, fbox32, fzr1, ubox5, lmo7a, kctd7, fbxo9, lonrf1l, fbxl4, nhpb2l1b, nhp2, fbl, hsp90aa1.1, snrpd3l, dhx15, mrto4, ruvbl1, hspa8b, and faub are the hub genes that correlate with the pathogenesis of pathological cardiac hypertrophy. The underlying regulatory pathways and cardiac pressure-responsive molecules identified in the present study will provide valuable insights for the supervision and clinical treatment of pathological cardiac hypertrophy induced by excessive exercise.
Collapse
Affiliation(s)
- Zuoqiong Zhou
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Lan Zheng
- Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, College of Physical Education, Hunan Normal University, Changsha, China
| | - Changfa Tang
- Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, College of Physical Education, Hunan Normal University, Changsha, China
| | - Zhanglin Chen
- Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, College of Physical Education, Hunan Normal University, Changsha, China
| | - Runkang Zhu
- Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, College of Physical Education, Hunan Normal University, Changsha, China
| | - Xiyang Peng
- Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, College of Physical Education, Hunan Normal University, Changsha, China
| | - Xiushan Wu
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Ping Zhu
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| |
Collapse
|
28
|
Chen Y, Wu X, Hu D, Wang W. Importance of Mitochondrial-Related Genes in Dilated Cardiomyopathy Based on Bioinformatics Analysis. CARDIOVASCULAR INNOVATIONS AND APPLICATIONS 2020; 5. [DOI: 10.15212/cvia.2019.0588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
We designed this study to identify potential key protein interaction networks, genes, and correlated pathways in dilated cardiomyopathy (DCM) via bioinformatics methods. We selected the GSE3586 microarray dataset, consisting of 15 dilated cardiomyopathic heart biopsy samples and 13 nonfailing heart biopsy samples. Initially, the GSE3586 dataset was downloaded and was analyzed with the limma package to identify differentially expressed genes (DEGs). A total of 172 DEGs consisting of 162 upregulated genes and ten downregulated genes in DCM were selected by the criterion of adjusted Pvalues less than 0.01 and the log2-fold change of 0.6 or greater. Gene Ontology functional enrichment analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis were performed to view the biological processes, cellular components, molecular function, and KEGG pathways of the DEGs. Next, protein-protein interactions were constructed, and the hub protein modules were identified. Then we selected the key genes DLD, UQCRC2, DLAT, SUCLA2, ATP5A1, PRDX3, FH, SDHD, and NDUFV1, which are involved in a wide range of biological activities, such as the citrate cycle, oxidation-reduction processes and cellular respiration, and energy derivation by oxidation of organic compounds in mitochondria. Finally, we found that currently there are no related gene-targeting drugs after exploring the predicted interactions between key genes and drugs, and transcription factors. In conclusion, our study provides greater understanding of the pathogenesis and underlying molecular mechanisms in DCM. This contributes to the exploration of potential gene therapy targets.
Collapse
Affiliation(s)
- Yukuan Chen
- Shantou University Medical College, Shantou, 515041 Guangdong, People’s Republic of China
- Department of Cardiology, Second Affiliated Hospital of Shantou University Medical College, Shantou, 515041 Guangdong, People’s Republic of China
| | - Xiaohui Wu
- Shantou University Medical College, Shantou, 515041 Guangdong, People’s Republic of China
- Department of Cardiology, Second Affiliated Hospital of Shantou University Medical College, Shantou, 515041 Guangdong, People’s Republic of China
| | - Danchun Hu
- Shantou University Medical College, Shantou, 515041 Guangdong, People’s Republic of China
- Department of Cardiology, Second Affiliated Hospital of Shantou University Medical College, Shantou, 515041 Guangdong, People’s Republic of China
| | - Wei Wang
- Department of Cardiology, Second Affiliated Hospital of Shantou University Medical College, Shantou, 515041 Guangdong, People’s Republic of China
| |
Collapse
|
29
|
Increased O-GlcNAcylation induces myocardial hypertrophy. In Vitro Cell Dev Biol Anim 2020; 56:735-743. [PMID: 32996013 DOI: 10.1007/s11626-020-00503-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 08/31/2020] [Indexed: 01/15/2023]
Abstract
Myocardial hypertrophy is a common precursor of many diseases, and it can lead to myocardial ischemia and weaken cardiac contractility. High-sugar diets and diabetes are high risk factors for cardiac hypertrophy. O-GlcNAcylation, a dynamic and ubiquitous post-translational glycosylation of proteins on serine/threonine residues, has been usually considered as a nutrient sensor. Hyperglycemia, hyperlipidemia, and hyperinsulinemia lead to an enhancement of protein O-GlcNAcylation; however, whether excessive O-linked β-N-acetylglucosamine (O-GlcNAc) glycosylation of proteins in cardiomyocytes causes cardiac hypertrophy remains unclear. In this study, we treated cultured primary cardiomyocytes or mice with streptozotocin (STZ) or PUGNAc, two inhibitors of O-GlcNAcase (OGA) to elevate cellular O-GlcNAcylation. We found that increased O-GlcNAcylation induced hypertrophy-like changes by detecting cardiomyocyte morphology or measuring the thickness of mice left ventricular wall with HE staining. The mRNA levels of cardiac hypertrophy-related genes, atrial natriuretic peptide (ANP) and β-myosin heavy chain (β-MHC), are increased in drug treatment groups. We further found that the increase of O-GlcNAcylation upregulated the activity of cAMP response element-binding protein (CREB) in cultured primary cells and in vivo by detecting the phosphorylation level of CREB by Western blot and the mRNA levels of CREB downstream targets C-fos and C-jun by RT-qPCR. These results suggest that the increased O-GlcNAcylation in cardiomyocytes is associated with cardiac hypertrophy both in cultured cells and in vivo, which provides possible intervention targets and approaches for the clinical treatment of myocardial hypertrophy triggered by high carbohydrate diets.
Collapse
|
30
|
Abstract
Myosins constitute a superfamily of actin-based molecular motor proteins that mediates a variety of cellular activities including muscle contraction, cell migration, intracellular transport, the formation of membrane projections, cell adhesion, and cell signaling. The 12 myosin classes that are expressed in humans share sequence similarities especially in the N-terminal motor domain; however, their enzymatic activities, regulation, ability to dimerize, binding partners, and cellular functions differ. It is becoming increasingly apparent that defects in myosins are associated with diseases including cardiomyopathies, colitis, glomerulosclerosis, neurological defects, cancer, blindness, and deafness. Here, we review the current state of knowledge regarding myosins and disease.
Collapse
|
31
|
Histone Deacetylase Inhibitor Suberoylanilide Hydroxamic Acid Improves Energetic Status and Cardiomyogenic Differentiation of Human Dilated Myocardium-Derived Primary Mesenchymal Cells. Int J Mol Sci 2020; 21:ijms21144845. [PMID: 32650632 PMCID: PMC7402340 DOI: 10.3390/ijms21144845] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 07/03/2020] [Accepted: 07/05/2020] [Indexed: 02/08/2023] Open
Abstract
Background. In this study the effect of histone deacetylase (HDAC) inhibitor suberoylanilide hydroxamic acid (SAHA) on the energetic status and cardiomyogenic differentiation of human healthy and dilated myocardium-derived mesenchymal stromal cells (hmMSC) have been investigated. Methods. The hmMSC were isolated from the healthy and dilated post-operation heart biopsies by explant outgrowth method. Cell proliferation, HDAC activity, mitochondrial membrane potential, and level of adenosine triphosphate (ATP) were evaluated. The effect of SAHA on mitochondrial parameters has been investigated also by Seahorse XF analyzer and cardiomyogenic differentiation was confirmed by the expression of transcription factor NK2 Homeobox 5 (Nkx2.5), cardiac troponin T and alpha cardiac actin at gene and protein levels. Results. Dilated myocardium-derived hmMSC had almost 1.5 folds higher HDAC activity compared to the healthy cells and significantly lower mitochondrial membrane potential and ATP level. HDAC class I and II inhibitor SAHA improved energetic status of mitochondria in dilated myocardium-isolated hmMSC and increased expression of cardiac specific proteins during 14 days of exposure of cells to SAHA. Conclusions. HDAC inhibitor SAHA can be a promising therapeutic for dilated cardiomyopathy (DCM). Dilated hmMSC exposed to SAHA improved energetic status and, subsequently, cardiomyogenic differentiation. Data suggest that human dilated myocardium-derived MSC still have cardio tissue regenerative potential, which might be stimulated by HDAC inhibitors.
Collapse
|
32
|
Tomczyk MM, Dolinsky VW. The Cardiac Lipidome in Models of Cardiovascular Disease. Metabolites 2020; 10:E254. [PMID: 32560541 PMCID: PMC7344916 DOI: 10.3390/metabo10060254] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 06/11/2020] [Accepted: 06/11/2020] [Indexed: 12/19/2022] Open
Abstract
Cardiovascular disease (CVD) is the leading cause of death worldwide. There are numerous factors involved in the development of CVD. Among these, lipids have an important role in maintaining the myocardial cell structure as well as cardiac function. Fatty acids (FA) are utilized for energy, but also contribute to the pathogenesis of CVD and heart failure. Advances in mass spectrometry methods have enabled the comprehensive analysis of a plethora of lipid species from a single sample comprised of a heterogeneous population of lipid molecules. Determining cardiac lipid alterations in different models of CVD identifies novel biomarkers as well as reveals molecular mechanisms that underlie disease development and progression. This information could inform the development of novel therapeutics in the treatment of CVD. Herein, we provide a review of recent studies of cardiac lipid profiles in myocardial infarction, obesity, and diabetic and dilated cardiomyopathy models of CVD by methods of mass spectrometry analysis.
Collapse
Affiliation(s)
- Mateusz M. Tomczyk
- Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Theme of the Children’s Hospital Research Institute of Manitoba, 715 McDermot Avenue, Winnipeg, MB R3E 3P4, Canada;
- Department of Pharmacology and Therapeutics, University of Manitoba, Winnipeg, MB R3E 0T6, Canada
- Rady Faculty of Health Science, College of Medicine, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Vernon W. Dolinsky
- Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Theme of the Children’s Hospital Research Institute of Manitoba, 715 McDermot Avenue, Winnipeg, MB R3E 3P4, Canada;
- Department of Pharmacology and Therapeutics, University of Manitoba, Winnipeg, MB R3E 0T6, Canada
- Rady Faculty of Health Science, College of Medicine, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| |
Collapse
|
33
|
Petroniene J, Morkvenaite‐Vilkonciene I, Miksiunas R, Bironaite D, Ramanaviciene A, Mikoliunaite L, Kisieliute A, Rucinskas K, Janusauskas V, Plikusiene I, Labeit S, Ramanavicius A. Evaluation of Redox Activity of Human Myocardium‐derived Mesenchymal Stem Cells by Scanning Electrochemical Microscopy. ELECTROANAL 2020. [DOI: 10.1002/elan.201900723] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Jurate Petroniene
- Department of Physical ChemistryFaculty of Chemistry and GeosciencesVilnius University Naugadruko str. 24 LT-03225 Vilnius Lithuania
| | - Inga Morkvenaite‐Vilkonciene
- Laboratory of Electrochemical Energy ConversionState Research Institute Centre for Physical Sciences and Technology Universiteto str. 3 LT-01513 Vilnius Lithuania
- Department of Mechatronics and RoboticsFaculty of MechanicsVilnius Gediminas Technical University Universiteto str. 3 LT-01513 Vilnius Lithuania
| | - Rokas Miksiunas
- Department of Regenerative medicineState Research Institute Centre for Innovative Medicine Universiteto str. 3 LT-01513 Vilnius Lithuania
| | - Daiva Bironaite
- Department of Regenerative medicineState Research Institute Centre for Innovative Medicine Universiteto str. 3 LT-01513 Vilnius Lithuania
| | - Almira Ramanaviciene
- Nanotechnas-Centre of Nanotechnology and Materials ScienceFaculty of Chemistry and GeosciencesVilnius University Naugadruko str. 24 LT-03225 Vilnius Lithuania
| | - Lina Mikoliunaite
- Department of Physical ChemistryFaculty of Chemistry and GeosciencesVilnius University Naugadruko str. 24 LT-03225 Vilnius Lithuania
| | - Aura Kisieliute
- Department of Physical ChemistryFaculty of Chemistry and GeosciencesVilnius University Naugadruko str. 24 LT-03225 Vilnius Lithuania
| | - Kestutis Rucinskas
- Centre of Cardiothoracic Surgery of Vilnius University Hospital Santariskiu Klinikos Universiteto str. 3 LT-01513 Vilnius Lithuania
| | - Vilius Janusauskas
- Centre of Cardiothoracic Surgery of Vilnius University Hospital Santariskiu Klinikos Universiteto str. 3 LT-01513 Vilnius Lithuania
| | - Ieva Plikusiene
- Department of Physical ChemistryFaculty of Chemistry and GeosciencesVilnius University Naugadruko str. 24 LT-03225 Vilnius Lithuania
| | - Siegfried Labeit
- Department of Integrative PathophysiologyUniversitätsmedizin Mannheim Theodor-Kutzer-Uferstr. 1–3 DE-68167 Mannheim Germany
| | - Arunas Ramanavicius
- Department of Physical ChemistryFaculty of Chemistry and GeosciencesVilnius University Naugadruko str. 24 LT-03225 Vilnius Lithuania
- Laboratory of NanotechnologyState Research Institute Centre for Physical Sciences and Technology Sauletekio str. LT-10257 Vilnius Lithuania
| |
Collapse
|
34
|
Cardiac troponin I R193H mutant interacts with HDAC1 to repress phosphodiesterase 4D expression in cardiomyocytes. Genes Dis 2020; 8:569-579. [PMID: 34179318 PMCID: PMC8209310 DOI: 10.1016/j.gendis.2020.01.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 12/13/2019] [Accepted: 01/06/2020] [Indexed: 01/12/2023] Open
Abstract
Cardiac Troponin I (cTnI) is a subunit of the thin filament involved in regulation of heart contraction. Mutated cTnI accounts for most genetic mutations associated with restrictive cardiomyopathy (RCM). We previously found phosphodiesterase 4D (PDE4D) decreased in RCM mice with cTnIR193H mutation and the mutant cTnI might be involved in PDE4D reduction. This study aims to elucidate a novel role of cTnIR193H mutant as a gene regulator. Overexpression of cTnIR193H mutant in cardiomyocytes showed decrease in PDED4D protein expression, while the enrichment of histone deacetylase 1 (HDAC1) was increased along with decreases in acetylated lysine 4 (acH3K4) and 9 (acH3K9) levels in the PDE4D promoter. HDAC1 overexpression could also downregulate PDE4D via reducing acH3K4 and acH3K9 levels. Co-IP assays showed that cTnIR193H mutant owed increased binding ability to HDAC1 compared with wild type cTnI. EGCG as a HDAC1 inhibitor could diminish the strength of cTnIR193H-HDAC1 interactions and alleviate the reduction in PDE4D expression. Together, our data indicated that cTnIR193H mutant could repress PDE4D expression in cardiomyocytes through HDAC1 associated histone deacetylation modification. Unlike the typical function of cTnI in cytoplasm, our study suggested a novel role of cTnI mutants in nuclei in regulating gene expression.
Collapse
|
35
|
Lamounier Júnior A, Ferrari F, Max R, Ritt LEF, Stein R. Importance of Genetic Testing in Dilated Cardiomyopathy: Applications and Challenges in Clinical Practice. Arq Bras Cardiol 2019; 113:274-281. [PMID: 31483024 PMCID: PMC6777894 DOI: 10.5935/abc.20190144] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2019] [Revised: 03/13/2019] [Accepted: 04/10/2019] [Indexed: 12/29/2022] Open
Abstract
Dilated cardiomyopathy (DCM) is a clinical syndrome characterized by left ventricular dilatation and contractile dysfunction. It is the most common cause of heart failure in young adults. The advent of next-generation sequencing has contributed to the discovery of a large amount of genomic data related to DCM. Mutations involving genes that encode cytoskeletal proteins, the sarcomere, and ion channels account for approximately 40% of cases previously classified as idiopathic DCM. In this scenario, geneticists and cardiovascular genetics specialists have begun to work together, building knowledge and establishing more accurate diagnoses. However, proper interpretation of genetic results is essential and multidisciplinary teams dedicated to the management and analysis of the obtained information should be considered. In this review, we approach genetic factors associated with DCM and their prognostic relevance and discuss how the use of genetic testing, when well recommended, can help cardiologists in the decision-making process.
Collapse
Affiliation(s)
- Arsonval Lamounier Júnior
- Health in Code S.L., Scientific Department, A Coruña -
Spain
- Universidade da Coruña, GRINCAR (Cardiovascular Research
Group), A Coruña - Spain
| | - Filipe Ferrari
- Graduate Program in Cardiology and Cardiovascular Sciences, Hospital
de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto
Alegre, RS - Brazil
- Exercise Cardiology Research Group (CardioEx), Hospital de
Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto
Alegre, RS - Brazil
| | - Renato Max
- Hospital Universitário Onofre Lopes, Natal, RN - Brazil
| | - Luiz Eduardo Fonteles Ritt
- Escola Bahiana de Medicina e Saúde Pública, Salvador,
BA - Brazil
- Hospital Cárdio Pulmonar, Salvador, BA - Brazil
| | - Ricardo Stein
- Graduate Program in Cardiology and Cardiovascular Sciences, Hospital
de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto
Alegre, RS - Brazil
- Exercise Cardiology Research Group (CardioEx), Hospital de
Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto
Alegre, RS - Brazil
| |
Collapse
|
36
|
Pourrazi H, Jafari A. Effects of a Combination of Dietary Restriction and Exercise Training on Myocardial Apoptosis in Male Rats. NUTRITION AND FOOD SCIENCES RESEARCH 2019. [DOI: 10.29252/nfsr.6.2.13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
|
37
|
García-Otero L, López M, Guitart-Mampel M, Morén C, Goncé A, Esteve C, Salazar L, Gómez O, Martínez JM, Torres B, César S, Garrabou G, Crispi F, Gratacós E. Cardiac and mitochondrial function in HIV-uninfected fetuses exposed to antiretroviral treatment. PLoS One 2019; 14:e0213279. [PMID: 30830946 PMCID: PMC6398922 DOI: 10.1371/journal.pone.0213279] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 02/18/2019] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Mitochondrial toxicity related to maternal combined antiretroviral treatment (cART) may have an impact on the heart of HIV-exposed uninfected (HEU) fetuses. Our objective was to evaluate fetal cardiovascular and mitochondrial biomarkers in HIV pregnancies. METHODS Prospective cohort including 47 HIV-infected and 47 non HIV-infected pregnancies. Fetal echocardiography was performed at 26-32 weeks of pregnancy. Umbilical cord blood and placental tissue were collected to study mitochondrial DNA content (mtDNA) (ratio 12SrRNA/RNAseP) and mitochondrial function (cytochrome c oxidase, COX, enzymatic activity) normalized by mitochondrial content (citrate synthase, CS). RESULTS HEU fetuses showed hypertrophic hearts (left myocardial wall thickness: HIV mean 3.21 mm (SD 0.81) vs. non-HIV 2.72 (0.42), p = 0.012), with signs of systolic and diastolic dysfunction (isovolumic relaxation time: HIV 52.2 ms (8.85) vs. non-HIV 42.5 ms (7.30); p<0.001). Cord blood mitochondrial content was significantly increased in HIV-exposed fetuses (CS activity: HIV 82.9 nmol/min.mg of protein (SD 40.5) vs. non-HIV 56.7 nmol/min.mg of protein (28.4); p = 0.007), with no differences in mtDNA content and COX activity. Both myocardial and mitochondrial mass parameters were significantly associated with zidovudine exposure. CONCLUSIONS HEU fetuses showed signs of increased myocardial and mitochondrial mass associated with maternal zidovudine treatment, suggesting a fetal adaptive response to cART toxicity.
Collapse
Affiliation(s)
- Laura García-Otero
- Fetal i+D Fetal Medicine Research Center, BCNatal—Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu), Institut Clinic de Ginecologia, Obstetricia i Neonatologia (ICGON), Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona and Centre for Biomedical Research on Rare Diseases (CIBER-ER), Barcelona, Spain
| | - Marta López
- Fetal i+D Fetal Medicine Research Center, BCNatal—Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu), Institut Clinic de Ginecologia, Obstetricia i Neonatologia (ICGON), Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona and Centre for Biomedical Research on Rare Diseases (CIBER-ER), Barcelona, Spain
| | - Mariona Guitart-Mampel
- Muscle Research and Mitochondrial Function Laboratory, Cellex IDIBAPS, Faculty of Medicine and Health Sciences-University of Barcelona, Internal Medicine Service-Hospital Clínic of Barcelona (Barcelona, Spain) and CIBER-ER
| | - Constanza Morén
- Muscle Research and Mitochondrial Function Laboratory, Cellex IDIBAPS, Faculty of Medicine and Health Sciences-University of Barcelona, Internal Medicine Service-Hospital Clínic of Barcelona (Barcelona, Spain) and CIBER-ER
| | - Anna Goncé
- Fetal i+D Fetal Medicine Research Center, BCNatal—Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu), Institut Clinic de Ginecologia, Obstetricia i Neonatologia (ICGON), Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona and Centre for Biomedical Research on Rare Diseases (CIBER-ER), Barcelona, Spain
| | - Carol Esteve
- Fetal i+D Fetal Medicine Research Center, BCNatal—Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu), Institut Clinic de Ginecologia, Obstetricia i Neonatologia (ICGON), Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona and Centre for Biomedical Research on Rare Diseases (CIBER-ER), Barcelona, Spain
| | - Laura Salazar
- Fetal i+D Fetal Medicine Research Center, BCNatal—Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu), Institut Clinic de Ginecologia, Obstetricia i Neonatologia (ICGON), Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona and Centre for Biomedical Research on Rare Diseases (CIBER-ER), Barcelona, Spain
| | - Olga Gómez
- Fetal i+D Fetal Medicine Research Center, BCNatal—Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu), Institut Clinic de Ginecologia, Obstetricia i Neonatologia (ICGON), Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona and Centre for Biomedical Research on Rare Diseases (CIBER-ER), Barcelona, Spain
| | - Josep María Martínez
- Fetal i+D Fetal Medicine Research Center, BCNatal—Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu), Institut Clinic de Ginecologia, Obstetricia i Neonatologia (ICGON), Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona and Centre for Biomedical Research on Rare Diseases (CIBER-ER), Barcelona, Spain
| | - Berta Torres
- Infectious Diseases Department, Hospital Clínic, Fundació Clínic per a la Recerca Biomèdica (FCRB), Barcelona, Spain
| | - Sergi César
- Department of Pediatric Cardiology, Hospital Sant Joan de Déu Barcelona, University of Barcelona, Barcelona, Spain
| | - Glòria Garrabou
- Muscle Research and Mitochondrial Function Laboratory, Cellex IDIBAPS, Faculty of Medicine and Health Sciences-University of Barcelona, Internal Medicine Service-Hospital Clínic of Barcelona (Barcelona, Spain) and CIBER-ER
| | - Fàtima Crispi
- Fetal i+D Fetal Medicine Research Center, BCNatal—Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu), Institut Clinic de Ginecologia, Obstetricia i Neonatologia (ICGON), Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona and Centre for Biomedical Research on Rare Diseases (CIBER-ER), Barcelona, Spain
| | - Eduard Gratacós
- Fetal i+D Fetal Medicine Research Center, BCNatal—Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu), Institut Clinic de Ginecologia, Obstetricia i Neonatologia (ICGON), Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona and Centre for Biomedical Research on Rare Diseases (CIBER-ER), Barcelona, Spain
| |
Collapse
|
38
|
Watanabe T, Kimura A, Kuroyanagi H. Alternative Splicing Regulator RBM20 and Cardiomyopathy. Front Mol Biosci 2018; 5:105. [PMID: 30547036 PMCID: PMC6279932 DOI: 10.3389/fmolb.2018.00105] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Accepted: 11/09/2018] [Indexed: 12/17/2022] Open
Abstract
RBM20 is a vertebrate-specific RNA-binding protein with two zinc finger (ZnF) domains, one RNA-recognition motif (RRM)-type RNA-binding domain and an arginine/serine (RS)-rich region. RBM20 has initially been identified as one of dilated cardiomyopathy (DCM)-linked genes. RBM20 is a regulator of heart-specific alternative splicing and Rbm20ΔRRM mice lacking the RRM domain are defective in the splicing regulation. The Rbm20ΔRRM mice, however, do not exhibit a characteristic DCM-like phenotype such as dilatation of left ventricles or systolic dysfunction. Considering that most of the RBM20 mutations identified in familial DCM cases were heterozygous missense mutations in an arginine-serine-arginine-serine-proline (RSRSP) stretch whose phosphorylation is crucial for nuclear localization of RBM20, characterization of a knock-in animal model is awaited. One of the major targets for RBM20 is the TTN gene, which is comprised of the largest number of exons in mammals. Alternative splicing of the TTN gene is exceptionally complicated and RBM20 represses >160 of its consecutive exons, yet detailed mechanisms for such extraordinary regulation are to be elucidated. The TTN gene encodes the largest known protein titin, a multi-functional sarcomeric structural protein specific to striated muscles. As titin is the most important factor for passive tension of cardiomyocytes, extensive heart-specific and developmentally regulated alternative splicing of the TTN pre-mRNA by RBM20 plays a critical role in passive stiffness and diastolic function of the heart. In disease models with diastolic dysfunctions, the phenotypes were rescued by increasing titin compliance through manipulation of the Ttn pre-mRNA splicing, raising RBM20 as a potential therapeutic target.
Collapse
Affiliation(s)
- Takeshi Watanabe
- Laboratory of Gene Expression, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan.,Department of Psychosomatic Dentistry, Graduate School of Medical and Dental Science, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Akinori Kimura
- Division of Pathology, Department of Molecular Pathogenesis, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan.,Laboratory for Integrated Research Projects on Intractable Diseases Advanced Technology Laboratories, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Hidehito Kuroyanagi
- Laboratory of Gene Expression, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan.,Laboratory for Integrated Research Projects on Intractable Diseases Advanced Technology Laboratories, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan.,Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
39
|
Dong Y, Xu S, Liu J, Ponnusamy M, Zhao Y, Zhang Y, Wang Q, Li P, Wang K. Non-coding RNA-linked epigenetic regulation in cardiac hypertrophy. Int J Biol Sci 2018; 14:1133-1141. [PMID: 29989099 PMCID: PMC6036733 DOI: 10.7150/ijbs.26215] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Accepted: 06/04/2018] [Indexed: 12/11/2022] Open
Abstract
Cardiac hypertrophy is an adaptive enlargement of myocardium in response to pressure overload caused various pathological insults, which is accompanied by alteration of a complex cascade of signaling pathways. During the hypertrophy process, many changes occur at cellular level including gene reprogramming by turning off chromatin regulators. Studies from the past decade have demonstrated that the abnormal epigenetic modifications, such as DNA methylation, histone modification, and oxidative modification of nucleic acid, could lead to changes in chromosome structure and cardiac dysfunction. Increasing evidence indicates that non-coding RNAs (ncRNAs) have functional significance in modulating the gene expression during those pathological events in the heart. Emerging evidences have highlighted that ncRNAs might serve as a signal for changing the state of chromatin, however, the knowledge about the ncRNA-linked epigenetic regulatory mechanisms in cardiac pathologies is still largely unexplored. In this review, we summarize the current information on association between ncRNAs and epigenetic modifications in cardiac hypertrophy, and we have discussed their crosstalk. In addition, this review provides insights into their therapeutic and diagnostic potential for treating hypertrophic heart disease.
Collapse
Affiliation(s)
- Yanhan Dong
- Institute for Translational Medicine, Qingdao University, Deng Zhou Road 38, Qingdao 266021, China
| | - Sheng Xu
- Institute for Translational Medicine, Qingdao University, Deng Zhou Road 38, Qingdao 266021, China
| | - Jing Liu
- Institute for Translational Medicine, Qingdao University, Deng Zhou Road 38, Qingdao 266021, China
| | - Murugavel Ponnusamy
- Institute for Translational Medicine, Qingdao University, Deng Zhou Road 38, Qingdao 266021, China
| | - Yanfang Zhao
- Institute for Translational Medicine, Qingdao University, Deng Zhou Road 38, Qingdao 266021, China
| | - Yanhui Zhang
- Institute for Translational Medicine, Qingdao University, Deng Zhou Road 38, Qingdao 266021, China
| | - Qi Wang
- Institute for Translational Medicine, Qingdao University, Deng Zhou Road 38, Qingdao 266021, China
| | - Peifeng Li
- Institute for Translational Medicine, Qingdao University, Deng Zhou Road 38, Qingdao 266021, China
| | - Kun Wang
- Institute for Translational Medicine, Qingdao University, Deng Zhou Road 38, Qingdao 266021, China
| |
Collapse
|
40
|
Li Y, Wang J, Sun L, Zhu S. RETRACTED: LncRNA myocardial infarction-associated transcript (MIAT) contributed to cardiac hypertrophy by regulating TLR4 via miR-93. Eur J Pharmacol 2018; 818:508-517. [PMID: 29157986 DOI: 10.1016/j.ejphar.2017.11.031] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 11/14/2017] [Accepted: 11/16/2017] [Indexed: 11/17/2022]
Abstract
This article has been retracted: please see Elsevier Policy on Article Withdrawal (http://www.elsevier.com/locate/withdrawalpolicy). This article has been retracted at the request of the Editor-in-Chief after concerns were raised about the article on PubPeer (https://pubpeer.com/publications/88E67BAC524E92069B2AA3474D2BB8?utm_source=Chrome&utm_medium=BrowserExtension&utm_campaign=Chrome). In addition the editor found more issues in some Western Blot figures. 1. Bands of figure 1C ANF 30min , 60min, 12h, 24h are identical to those in Fig 4C ANF control, AngII, AngII+si-NC and AngII+si-MAT 2. Beta-actin bands of Fig 1C are identical to those in Fig 4C, but blots represent different experimental conditions/samples. 3. Bands in Fig 6 D AngII and AngII+mir-NC show similarities with these of AngII+mir-93 and AngII+mir93+vector (band shapes and irregularities appear identical) After several attempts to reach out to the corresponding author, no response was provided. The manipulation of images casts doubts on all the data, and accordingly also the conclusions based on that data, in this publication. As such this article represents a severe abuse of the scientific publishing system. The scientific community takes a very strong view on this matter and apologies are offered to readers of the journal that this was not detected during the submission process.
Collapse
Affiliation(s)
- Yunwei Li
- Department of Cardiology, Huaihe Hospital of Henan University, Kaifeng 475000, China.
| | - Juan Wang
- Department of Cardiology, Huaihe Hospital of Henan University, Kaifeng 475000, China
| | - Lili Sun
- Department of Cardiology, Huaihe Hospital of Henan University, Kaifeng 475000, China
| | - Shengnan Zhu
- Department of Cardiology, Huaihe Hospital of Henan University, Kaifeng 475000, China
| |
Collapse
|
41
|
Ji F, Liu Q, Feng Z, Han X, Li Z. Genetic association between 1425G/A SNP in PRKCH and hypertrophic cardiomyopathy in a Chinese population. Oncotarget 2017; 8:114839-114844. [PMID: 29383124 PMCID: PMC5777736 DOI: 10.18632/oncotarget.22214] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2017] [Accepted: 09/23/2017] [Indexed: 12/21/2022] Open
Abstract
Hypertrophic cardiomyopathy is a heterogeneous myocardial disorder with a broad spectrum of clinical presentation and morphologic features. Previous reports indicated that protein kinase C pathway as a major determinant of cardiac hypertrophy and heart failure. Population-based analyses of the association between PRKCH gene (encoded PKCη) and HCM has not been performed yet. The purpose of this study is to investigate the association of the nonsynonymous SNP (1425G/A) in PRKCH gene and hypertrophic cardiomyopathy in a Chinese population. 323 patients with HCM and 326 controls were examined using a case-control methodology. The 1425G/A SNP in PRKCH was genotyped by allele-specific real-time PCR assay. The 1425G/A SNP in PRKCH increased the risk of HOCM (hypertrophic obstructive cardiomyopathy) (OR=1.427, 95% confidence interval, 1.013 to 2.012, P=0.046) under a dominant model. After age- and sex-adjustment, the significant associations remained in HOCM (for GG +AG versus AA, OR= 2.497, 95% confidence interval, 1.01 to 6.17; P=0.047). The 1425G/A SNP in PRKCH increases the risk of hypertrophic obstructive cardiomyopathy in the Chinese population.
Collapse
Affiliation(s)
- Feng Ji
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Qun Liu
- Johns Hopkins University School of Medicine, Baltimore, MD 21224, USA
| | - Zeyu Feng
- Medical School of Nantong University, Nantong 226001, China
| | - Xinwei Han
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Zhitong Li
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| |
Collapse
|