1
|
Johnson N, Qi B, Wen J, Du B, Banerjee S. KLHL24 associated cardiomyopathy: Gene function to clinical management. Gene 2025; 939:149185. [PMID: 39708934 DOI: 10.1016/j.gene.2024.149185] [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: 07/20/2024] [Revised: 10/26/2024] [Accepted: 12/17/2024] [Indexed: 12/23/2024]
Abstract
BACKGROUND KLHL24 (Kelch-like protein 24) is a significant component of the ubiquitin-proteasome system (UPS), involved in regulating protein turnover through targeted ubiquitination and degradation. Germline mutations in KLHL24 gene have been known to cause Epidermolysis Bullosa Simplex characterized by skin fragility but has recently been found to cause Cardiomyopathy. MAIN BODY Various cardiomyopathies, including hypertrophic cardiomyopathy and dilated cardiomyopathy, leading to abnormal protein degradation and affecting the stability and function of essential cardiac proteins which finally results into structural and functional abnormalities in cardiac muscle. In this review, in order to understand the disease association of germline mutations of KLHL24, we summarize all the studies performed with KLHL24 gene including studies from 2016 when KLHL24 was first identified to be associated with epidermolysis bullosa simplex till the recent studies in 2024 by using keywords such as KLHL24 gene, hypertrophic cardiomyopathy, dilated cardiomyopathy and epidermolysis bullosa simplex. Furthermore, we explored the proposed molecular mechanisms and pathophysiologies of KLHL24 associated diseases. Patients with KLHL24 mutations were usually presented with variable clinical symptoms. The main clinical presentations have been cutaneous lesions, cardiac symptoms associated with cardiomyopathies and there have been reports of skeletal muscle weakness and neurological symptoms as well. Current treatments focus on managing clinical symptoms and preventing complications through medications, lifestyle changes, and surgical interventions. In addition, researches have also been conducted cell culture based in vitro studies for reducing the clinical symptoms of KLHL24 associated diseases. However, currently there are no specific clinical trials going on regarding the therapeutic strategies among patients with KLHL24 mutations. Understanding the role of KLHL24 in cardiomyopathies is very important for developing targeted diagnostic approach with therapeutic strategies. CONCLUSION This review emphasizes the importance of KLHL24 mutations as a newly recognized cause of cardiomyopathy, paving the way for improved clinical diagnosis, targeted therapies, and ultimately, for better patient outcomes.
Collapse
Affiliation(s)
- Neil Johnson
- Department of Genetics, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, 130021, China; Department of Cardiology, China-Japan Union Hospital of Jilin University, Norman Bethune Health Science Center, Changchun, China
| | - Baiyu Qi
- Department of Genetics, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, 130021, China
| | - Jianping Wen
- Department of Genetics, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, 130021, China
| | - Beibei Du
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Norman Bethune Health Science Center, Changchun, China
| | - Santasree Banerjee
- Department of Genetics, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, 130021, China.
| |
Collapse
|
2
|
Hanna AD, Chang T, Ho KS, Yee RSZ, Walker WC, Agha N, Hsu CW, Jung SY, Dickinson ME, Samee MAH, Ward CS, Lee CS, Rodney GG, Hamilton SL. Mechanisms underlying dilated cardiomyopathy associated with FKBP12 deficiency. J Gen Physiol 2025; 157:e202413583. [PMID: 39661086 PMCID: PMC11633665 DOI: 10.1085/jgp.202413583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 08/14/2024] [Accepted: 10/22/2024] [Indexed: 12/12/2024] Open
Abstract
Dilated cardiomyopathy (DCM) is a highly prevalent and genetically heterogeneous condition that results in decreased contractility and impaired cardiac function. The FK506-binding protein FKBP12 has been implicated in regulating the ryanodine receptor in skeletal muscle, but its role in cardiac muscle remains unclear. To define the effect of FKBP12 in cardiac function, we generated conditional mouse models of FKBP12 deficiency. We used Cre recombinase driven by either the α-myosin heavy chain, (αMHC) or muscle creatine kinase (MCK) promoter, which are expressed at embryonic day 9 (E9) and E13, respectively. Both conditional models showed an almost total loss of FKBP12 in adult hearts compared with control animals. However, only the early embryonic deletion of FKBP12 (αMHC-Cre) resulted in an early-onset and progressive DCM, increased cardiac oxidative stress, altered expression of proteins associated with cardiac remodeling and disease, and sarcoplasmic reticulum Ca2+ leak. Our findings indicate that FKBP12 deficiency during early development results in cardiac remodeling and altered expression of DCM-associated proteins that lead to progressive DCM in adult hearts, thus suggesting a major role for FKBP12 in embryonic cardiac muscle.
Collapse
Affiliation(s)
- Amy D. Hanna
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
| | - Ting Chang
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
| | - Kevin S. Ho
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
| | - Rachel Sue Zhen Yee
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
| | | | - Nadia Agha
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
| | - Chih-Wei Hsu
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
| | - Sung Yun Jung
- Department of Biochemistry, Baylor College of Medicine, Houston, TX, USA
| | - Mary E. Dickinson
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
| | | | - Christopher S. Ward
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
| | - Chang Seok Lee
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
| | - George G. Rodney
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
| | - Susan L. Hamilton
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
3
|
She P, Gao B, Li D, Wu C, Zhu X, He Y, Mo F, Qi Y, Jin D, Chen Y, Zhao X, Lin J, Hu H, Li J, Zhang B, Xie P, Lin C, Christoffels VM, Wu Y, Zhu P, Zhong TP. The transcriptional repressor HEY2 regulates mitochondrial oxidative respiration to maintain cardiac homeostasis. Nat Commun 2025; 16:232. [PMID: 39747914 PMCID: PMC11696871 DOI: 10.1038/s41467-024-55557-4] [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: 11/06/2023] [Accepted: 12/17/2024] [Indexed: 01/04/2025] Open
Abstract
Energy deprivation and metabolic rewiring of cardiomyocytes are widely recognized hallmarks of heart failure. Here, we report that HEY2 (a Hairy/Enhancer-of-split-related transcriptional repressor) is upregulated in hearts of patients with dilated cardiomyopathy. Induced Hey2 expression in zebrafish hearts or mammalian cardiomyocytes impairs mitochondrial respiration, accompanied by elevated ROS, resulting in cardiomyocyte apoptosis and heart failure. Conversely, Hey2 depletion in adult mouse hearts and zebrafish enhances the expression of mitochondrial oxidation genes and cardiac function. Multifaceted genome-wide analyses reveal that HEY2 enriches at the promoters of genes known to regulate metabolism (including Ppargc1, Esrra and Cpt1) and colocalizes with HDAC1 to effectuate histone deacetylation and transcriptional repression. Consequently, restoration of PPARGC1A/ESRRA in Hey2- overexpressing zebrafish hearts or human cardiomyocyte-like cells rescues deficits in mitochondrial bioenergetics. Knockdown of Hey2 in adult mouse hearts protects against doxorubicin-induced cardiac dysfunction. These studies reveal an evolutionarily conserved HEY2/HDAC1-Ppargc1/Cpt transcriptional module that controls energy metabolism to preserve cardiac function.
Collapse
Affiliation(s)
- Peilu She
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, 510100, China
| | - Bangjun Gao
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Dongliang Li
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Chen Wu
- School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu, 210096, China
| | - Xuejiao Zhu
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Yuan He
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Fei Mo
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Yao Qi
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Daqing Jin
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Yewei Chen
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Xin Zhao
- School of Life Sciences, Fudan University, Shanghai, 200438, China
| | - Jinzhong Lin
- School of Life Sciences, Fudan University, Shanghai, 200438, China
| | - Hairong Hu
- School of Life Sciences, Fudan University, Shanghai, 200438, China
| | - Jia Li
- Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Bing Zhang
- Shanghai Center for Systems Biomedicine, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Peng Xie
- School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu, 210096, China
| | - Chengqi Lin
- School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu, 210096, China
| | - Vincent M Christoffels
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, 1105AZ, The Netherlands
| | - Yueheng Wu
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, 510100, China.
| | - Ping Zhu
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, 510100, China.
| | - Tao P Zhong
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China.
| |
Collapse
|
4
|
Corrado D, Thiene G, Bauce B, Calore C, Cipriani A, De Lazzari M, Migliore F, Perazzolo Marra M, Pilichou K, Rigato I, Rizzo S, Zorzi A, Basso C. The "Padua classification" of cardiomyopathies: Combining pathobiological basis and morpho-functional remodeling. Int J Cardiol 2025; 418:132571. [PMID: 39306295 DOI: 10.1016/j.ijcard.2024.132571] [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: 05/16/2024] [Revised: 09/12/2024] [Accepted: 09/16/2024] [Indexed: 10/19/2024]
Abstract
Over the last 20 years, the scientific progresses in molecular biology and genetics in combination with the increasing use in the clinical setting of contrast-enhanced cardiac magnetic resonance (CMR) for morpho-functional imaging and structural myocardial tissue characterization have provided important new insights into our understanding of the distinctive aspects of cardiomyopathy, regarding both the genetic and biologic background and the clinical phenotypic features. This has led to the need of an appropriate revision and upgrading of current nosographic framework and pathobiological categorization of heart muscle disorders. This article proposes a new definition and classification of cardiomyopathies that rely on the combination of the distinctive pathobiological basis (genetics, molecular biology and pathology) and the clinical phenotypic pattern (morpho-functional and structural features), leading to the proposal of three different disease categories, each of either genetic or non-genetic etiology and characterized by a combined designation based on both "anatomic" and "functional" features, i.e., hypertrophic/restrictive (H/RC), dilated/hypokinetic (D/HC) and scarring/arrhythmogenic cardiomyopathy (S/AC). The clinical application of the newly proposed classification approach in the real-world practice appears crucial to design a targeted clinical management and evaluation of outcomes of affected patients. Although current treatment of cardiomyopathies is largely palliative and based on drugs, catheter ablation, device or surgical interventions aimed to prevent and manage heart failure and malignant arrhythmias, better knowledge of basic mechanisms involved in the onset and progression of pathobiologically different heart muscle diseases may allow to the development of disease-specific curative therapy.
Collapse
Affiliation(s)
- Domenico Corrado
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, Italy.
| | - Gaetano Thiene
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, Italy
| | - Barbara Bauce
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, Italy
| | - Chiara Calore
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, Italy
| | - Alberto Cipriani
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, Italy
| | - Manuel De Lazzari
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, Italy
| | - Federico Migliore
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, Italy
| | - Martina Perazzolo Marra
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, Italy
| | - Kalliopi Pilichou
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, Italy
| | - Ilaria Rigato
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, Italy
| | - Stefania Rizzo
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, Italy
| | - Alessandro Zorzi
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, Italy
| | - Cristina Basso
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, Italy
| |
Collapse
|
5
|
Cleary SR, Teng ACT, Kongmeneck AD, Fang X, Phillips TA, Cho EE, Smith RA, Karkut P, Makarewich CA, Kekenes-Huskey PM, Gramolini AO, Robia SL. Dilated cardiomyopathy variant R14del increases phospholamban pentamer stability, blunting dynamic regulation of calcium. J Biol Chem 2024:108118. [PMID: 39710323 DOI: 10.1016/j.jbc.2024.108118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 10/28/2024] [Accepted: 12/17/2024] [Indexed: 12/24/2024] Open
Abstract
The sarco(endo)plasmic reticulum Ca2+ ATPase (SERCA) is a membrane transporter that creates and maintains intracellular Ca2+ stores. In the heart, SERCA is regulated by an inhibitory interaction with the monomeric form of the transmembrane micropeptide phospholamban (PLB). PLB also forms avid homo-pentamers, and dynamic exchange of PLB between pentamers and SERCA is an important determinant of cardiac responsiveness to exercise. Here, we investigated two naturally occurring pathogenic variants of PLB: a cysteine substitution of Arg9 (R9C) and an in-frame deletion of Arg14 (R14del). Both variants are associated with dilated cardiomyopathy. We previously showed that the R9C mutation causes disulfide crosslinking and hyperstabilization of pentamers. While the pathogenic mechanism of R14del is unclear, we hypothesized this mutation may also alter pentamer stability. Immunoblots revealed a significantly increased pentamer:monomer ratio for R14del-PLB compared to WT-PLB. We quantified homo-oligomerization and SERCA-binding in live cells using fluorescence resonance energy transfer (FRET) microscopy. R14del-PLB showed increased affinity for homo-oligomerization and decreased binding affinity for SERCA compared to WT. The data suggest that, like R9C, the R14del mutation stabilizes PLB in pentamers, decreasing its ability to regulate SERCA. The R14del mutation reduces the rate of PLB unbinding from pentamers after transient elevations of Ca2+, limiting the recovery of PLB-SERCA complexes. A computational model predicted that hyperstabilization of PLB pentamers by R14del impairs the ability of cardiac Ca2+ handling to respond to changing heart rates between rest and exercise. We postulate that impaired responsiveness to physiological stress contributes to arrhythmogenesis in human carriers of the R14del mutation.
Collapse
Affiliation(s)
- Sean R Cleary
- Department of Cell and Molecular Physiology, Loyola University Chicago, Maywood, IL, USA
| | - Allen C T Teng
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | | | - Xuan Fang
- Department of Cell and Molecular Physiology, Loyola University Chicago, Maywood, IL, USA
| | - Taylor A Phillips
- Department of Cell and Molecular Physiology, Loyola University Chicago, Maywood, IL, USA
| | - Ellen E Cho
- Department of Cell and Molecular Physiology, Loyola University Chicago, Maywood, IL, USA
| | - Rhys A Smith
- Department of Cell and Molecular Physiology, Loyola University Chicago, Maywood, IL, USA
| | - Patryk Karkut
- Department of Cell and Molecular Physiology, Loyola University Chicago, Maywood, IL, USA
| | - Catherine A Makarewich
- Division of Molecular Cardiovascular Biology of the Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Peter M Kekenes-Huskey
- Department of Cell and Molecular Physiology, Loyola University Chicago, Maywood, IL, USA
| | | | - Seth L Robia
- Department of Cell and Molecular Physiology, Loyola University Chicago, Maywood, IL, USA.
| |
Collapse
|
6
|
Agarwalla A, Austin MA, Reza N. Genetics in clinical cardiology: the current state and opportunities ahead. Future Cardiol 2024; 20:815-818. [PMID: 39540255 DOI: 10.1080/14796678.2024.2426883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024] Open
Affiliation(s)
- Anjali Agarwalla
- Division of Cardiovascular Medicine, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Melissa A Austin
- Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Nosheen Reza
- Division of Cardiovascular Medicine, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
7
|
Xu S, Zhang G, Tan X, Zeng Y, Fan H, Gao J, Qin Z, Yu F, Ma B, Zhang T, Jiang H, Li X, Wang X, Fan J, Bo X, Zhou Y, Tang J. Differential Expression Spectrum of circRNA in Plasma Exosomes in Dilated Cardiomyopathy With Heart Failure. J Cell Mol Med 2024; 28:e70258. [PMID: 39719688 DOI: 10.1111/jcmm.70258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 10/03/2024] [Accepted: 10/29/2024] [Indexed: 12/26/2024] Open
Abstract
Dilated cardiomyopathy (DCM), a form of non-ischaemic myocardial disease, is characterised by structural and functional cardiac abnormalities. As defined by the World Health Organisation, DCM constitutes a significant cardiac pathology, leading to increased morbidity and mortality due to complications such as heart failure and arrhythmias. The diagnostic process for DCM predominantly employs echocardiography and MRI, with biomarkers like NT-pro BNP and troponin providing supportive, yet non-specific, evidence. Exosomes, small extracellular vesicles, play a critical role in intercellular communications by transferring biomolecules including lipids, proteins, messenger RNA (mRNA) and non-coding RNA (ncRNA) to target cells, thereby influencing key cellular processes such as proliferation, differentiation, apoptosis, angiogenesis and immune modulation. Within the ncRNA category, circular RNAs (circRNAs) are notable for their cellular specificity and evolutionary conservation and are often implicated in the regulatory mechanisms underlying DCM and heart failure. This investigation employed next-generation sequencing technology to analyse plasma exosomal circRNA profiles in DCM patients with chronic heart failure (CHF), compared to healthy controls. The analysis revealed distinct circRNA expression patterns, identifying 49 uniquely expressed circRNAs in the DCM cohort with CHF. These circRNAs were associated with several critical biological pathways, including the sequestration of extracellular ligands from receptors, N-acetyltransferase activity, histone acetyltransferase activity and endocytic vesicle membrane composition. The findings of this study provide valuable insights into the pathophysiological mechanisms of DCM and offer evidence for improving clinical diagnostic methodologies.
Collapse
Affiliation(s)
- Shuai Xu
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Province Key Laboratory of Cardiac Injury and Repair, Zhengzhou, China
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, China
- Department of Cardiology, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Medical Center of Soochow University, Suzhou, China
- Institute for Hypertension, Soochow University, Suzhou, China
| | - Ge Zhang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Province Key Laboratory of Cardiac Injury and Repair, Zhengzhou, China
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, China
| | - Xin Tan
- Department of Cardiology, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Medical Center of Soochow University, Suzhou, China
- Institute for Hypertension, Soochow University, Suzhou, China
| | - Yiyao Zeng
- Department of Cardiology, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Medical Center of Soochow University, Suzhou, China
- Institute for Hypertension, Soochow University, Suzhou, China
| | - Huimin Fan
- Center of Translational Medicine and Clinical Laboratory, The Fourth Affiliated Hospital to Soochow University, Suzhou, China
| | - Jiamin Gao
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Province Key Laboratory of Cardiac Injury and Repair, Zhengzhou, China
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, China
| | - Zhen Qin
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Province Key Laboratory of Cardiac Injury and Repair, Zhengzhou, China
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, China
| | - Fengyi Yu
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Province Key Laboratory of Cardiac Injury and Repair, Zhengzhou, China
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, China
| | - Bin Ma
- Luoyang Central Hospital Affiliated of Zhengzhou University, Luoyang, China
| | - Ting Zhang
- Department of Cardiology, the Second People's Hospital of Hefei, Hefei Hospital Affiliated to Ahhui Medical University, Hefei, China
| | - Hezi Jiang
- Department of Cardiology, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Medical Center of Soochow University, Suzhou, China
- Institute for Hypertension, Soochow University, Suzhou, China
| | - Xian Li
- Department of Cardiology, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Medical Center of Soochow University, Suzhou, China
- Institute for Hypertension, Soochow University, Suzhou, China
| | - Xiangyu Wang
- Department of Cardiology, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Medical Center of Soochow University, Suzhou, China
- Institute for Hypertension, Soochow University, Suzhou, China
| | - Jili Fan
- Department of Cardiovascular Disease, Taihe County People's Hospital, Fuyang, China
| | - Xiaohong Bo
- Department of Cardiovascular Disease, Taihe County People's Hospital, Fuyang, China
| | - Yafeng Zhou
- Department of Cardiology, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Medical Center of Soochow University, Suzhou, China
- Institute for Hypertension, Soochow University, Suzhou, China
| | - Junnan Tang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Province Key Laboratory of Cardiac Injury and Repair, Zhengzhou, China
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, China
| |
Collapse
|
8
|
Ruan J, Wu M, Xiang J, Hui X, Yang L, Lin R, Xu W, Shu Q. Generation of a human induced pluripotent stem cell line from a female patient carrying LZTR1 gene mutation. Stem Cell Res 2024; 81:103616. [PMID: 39577308 DOI: 10.1016/j.scr.2024.103616] [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: 09/23/2024] [Revised: 10/31/2024] [Accepted: 11/18/2024] [Indexed: 11/24/2024] Open
Abstract
The leucine zipper-like transcriptional regulator 1 (LZTR1) gene has been reported to be associated with many kinds of human diseases, including cardiac disease, Noonan syndrome, and schwannomatosis. In this study, peripheral blood mononuclear cells (PBMCs) derived from patient diagnosed with dilated cardiomyopathy (DCM) was successfully reprogrammed into the human induced pluripotent stem cells (iPSCs) line, harboring a distinct heterozygous mutation in the LZTR1 gene. The established patient-derived iPSCs expressed endogenous pluripotent markers, demonstrated the potential to differentiate into three germ layers (endoderm, mesoderm, and ectoderm), and exhibited a normal karyotype.
Collapse
Affiliation(s)
- Jinghua Ruan
- Department of Cardiac Surgery, the Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China; Binjiang Institute of Zhejiang University, Hangzhou, Zhejiang, China.
| | - Mengqing Wu
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Jiakai Xiang
- Department of Cardiac Surgery, the Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China
| | - Xianrui Hui
- Binjiang Institute of Zhejiang University, Hangzhou, Zhejiang, China
| | - Lijun Yang
- Department of Cardiac Surgery, the Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China
| | - Ru Lin
- Department of Cardiac Surgery, the Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China; Binjiang Institute of Zhejiang University, Hangzhou, Zhejiang, China
| | - Weize Xu
- Department of Cardiac Surgery, the Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China; Binjiang Institute of Zhejiang University, Hangzhou, Zhejiang, China; School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Qiang Shu
- Department of Cardiac Surgery, the Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China; Binjiang Institute of Zhejiang University, Hangzhou, Zhejiang, China; Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
9
|
Qin Q, Zhou ZY, Liu Y, Zhou F, Cao C, Teng L. Unraveling the nexus of nesprin in dilated cardiomyopathy: From molecular insights to therapeutic prospects. Life Sci 2024; 358:123126. [PMID: 39396640 DOI: 10.1016/j.lfs.2024.123126] [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: 07/19/2024] [Revised: 09/25/2024] [Accepted: 10/08/2024] [Indexed: 10/15/2024]
Abstract
Dilated cardiomyopathy is a complex and debilitating heart disorder characterized by the enlargement and weakening of the cardiac chambers, leading to impaired contractility and heart failure. Nesprins, a family of nuclear envelope spectrin repeat proteins that include isoforms Nesprin-1/-2, are integral components of the LInker of Nucleoskeleton and Cytoskeleton complex. They facilitate the connection between the nuclear envelope and the cytoskeleton, crucial for maintaining nuclear architecture, migration and positioning, and mechanical transduction and signaling. Nesprin-1/-2 are abundantly expressed in cardiac and skeletal muscles.They have emerged as key players in the pathogenesis of dilated cardiomyopathy. Mutations in synaptic nuclear envelope-1/-2 genes encoding Nesprin-1/-2 are associated with dilated cardiomyopathy, underscoring their significance in cardiac health. This review highlights the all known cases of Nesprin-1/-2 related dilated cardiomyopathy, focusing on their interactions with the nuclear envelope, their role in mechanical transduction, and their influence on gene expression. Moreover, it delves into the underlying mechanisms through which Nesprin dysfunction disrupts nuclear-cytoskeletal coupling, leading to abnormal nuclear morphology, impaired mechanotransduction, and altered gene regulation. The exploration of Nesprin's impact on dilated cardiomyopathy offers a promising avenue for therapeutic interventions aimed at ameliorating the disease. This review provides a comprehensive overview of recent advancements in understanding the pivotal role of Nesprins in dilated cardiomyopathy research.
Collapse
Affiliation(s)
- Qin Qin
- Department of Cardiology, Yichang Central People's Hospital/The First Clinical Medical College, Three Gorges University, Yichang 443003, Hubei, People's Republic of China; School of Basic Medicine, China Three Gorges University, Yichang 443000, Hubei, People's Republic of China
| | - Zi-Yi Zhou
- Department of Cardiology, Yichang Central People's Hospital/The First Clinical Medical College, Three Gorges University, Yichang 443003, Hubei, People's Republic of China; School of Basic Medicine, China Three Gorges University, Yichang 443000, Hubei, People's Republic of China
| | - Yangyuanzhi Liu
- Department of Cardiology, Yichang Central People's Hospital/The First Clinical Medical College, Three Gorges University, Yichang 443003, Hubei, People's Republic of China; School of Basic Medicine, China Three Gorges University, Yichang 443000, Hubei, People's Republic of China
| | - Fei Zhou
- Department of Cardiology, Yichang Central People's Hospital/The First Clinical Medical College, Three Gorges University, Yichang 443003, Hubei, People's Republic of China
| | - Chunyu Cao
- School of Basic Medicine, China Three Gorges University, Yichang 443000, Hubei, People's Republic of China; College of Basic Medical Sciences, Hubei Key Laboratory of Tumor Microencironment and Immunotherapy, China Three Gorges University, Yichang 443000, Hubei, People's Republic of China
| | - Lin Teng
- Department of Cardiology, Yichang Central People's Hospital/The First Clinical Medical College, Three Gorges University, Yichang 443003, Hubei, People's Republic of China; King's College London British Heart Foundation Centre of Research Excellence, School of Cardiovascular and Metabolic Medicine & Sciences, London SE5 9NU, UK.
| |
Collapse
|
10
|
Kubanek M, Binova J, Piherova L, Krebsova A, Kotrc M, Hartmannova H, Hodanova K, Musalkova D, Stranecky V, Palecek T, Chaloupka A, Grochova I, Krejci J, Petrkova J, Melenovsky V, Kmoch S, Kautzner J. Genotype is associated with left ventricular reverse remodelling and early events in recent-onset dilated cardiomyopathy. ESC Heart Fail 2024; 11:4127-4138. [PMID: 39129193 PMCID: PMC11631235 DOI: 10.1002/ehf2.15009] [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: 10/23/2023] [Revised: 06/05/2024] [Accepted: 07/15/2024] [Indexed: 08/13/2024] Open
Abstract
AIMS Recent-onset dilated cardiomyopathy (RODCM) is characterized by heterogeneous aetiology and diverse clinical outcomes, with scarce data on genotype-phenotype correlates. Our aim was to correlate individual RODCM genotypes with left ventricular reverse remodelling (LVRR) and clinical outcomes. METHODS AND RESULTS In this prospective study, a total of 386 Czech RODCM patients with symptom duration ≤6 months underwent genetic counselling and whole-exome sequencing (WES). The presence of pathogenic (class 5) or likely pathogenic (class 4) variants in a set of 72 cardiomyopathy-related genes was correlated with the occurrence of all-cause death, heart transplantation, or implantation of a ventricular assist device (primary outcome) and/or ventricular arrhythmia event (secondary outcome). LVRR was defined as an improvement of left ventricular ejection fraction to >50% or ≥10% absolute increase, with a left ventricular end-diastolic diameter ≤33 mm/m2 or ≥10% relative decrease. Median follow-up was 41 months. RODCM was familial in 98 (25%) individuals. Class 4-5 variants of interest (VOIs) were identified in 125 (32%) cases, with 69 (18%) having a single titin-truncating variant (TTNtv) and 56 (14%) having non-titin (non-TTN) VOIs. The presence of class 4-5 non-TTN VOIs, but not of TTNtv, heralded a lower probability of 12-month LVRR and proved to be an independent baseline predictor both of the primary and the secondary outcome. The negative result of genetic testing was a strong protective baseline variable against occurrence of life-threatening ventricular arrhythmias. Detection of class 4-5 VOIs in genes coding nuclear envelope proteins was another independent predictor of both study outcomes at baseline and also of life-threatening ventricular arrhythmias after 12 months. Class 4-5 VOIs of genes coding cytoskeleton were associated with an increased risk of life-threatening ventricular arrhythmias after baseline assessment. A positive family history of dilated cardiomyopathy alone only related to a lower probability of LVRR at 12 months and at the final follow-up. CONCLUSIONS RODCM patients harbouring class 4-5 non-TTN VOIs are at higher risk of progressive heart failure and life-threatening ventricular arrhythmias. Genotyping may improve their early risk stratification at baseline assessment.
Collapse
Affiliation(s)
- Milos Kubanek
- Department of CardiologyInstitute for Clinical and Experimental MedicinePragueCzech Republic
- European Reference Network for Rare, Low Prevalence and Complex Diseases of the Heart, ERN GUARD‐Heart, IKEMPragueCzech Republic
| | - Jana Binova
- Department of CardiologyInstitute for Clinical and Experimental MedicinePragueCzech Republic
- Institute of Physiology, First Faculty of MedicineCharles UniversityPragueCzech Republic
| | - Lenka Piherova
- Department of Pediatrics and Adolescent Medicine, First Faculty of Medicine, Research Unit for Rare DiseasesCharles UniversityPragueCzech Republic
| | - Alice Krebsova
- Department of CardiologyInstitute for Clinical and Experimental MedicinePragueCzech Republic
- European Reference Network for Rare, Low Prevalence and Complex Diseases of the Heart, ERN GUARD‐Heart, IKEMPragueCzech Republic
| | - Martin Kotrc
- Department of CardiologyInstitute for Clinical and Experimental MedicinePragueCzech Republic
- Institute of Physiology, First Faculty of MedicineCharles UniversityPragueCzech Republic
| | - Hana Hartmannova
- Department of Pediatrics and Adolescent Medicine, First Faculty of Medicine, Research Unit for Rare DiseasesCharles UniversityPragueCzech Republic
| | - Katerina Hodanova
- Department of Pediatrics and Adolescent Medicine, First Faculty of Medicine, Research Unit for Rare DiseasesCharles UniversityPragueCzech Republic
| | - Dita Musalkova
- Department of Pediatrics and Adolescent Medicine, First Faculty of Medicine, Research Unit for Rare DiseasesCharles UniversityPragueCzech Republic
| | - Viktor Stranecky
- Department of Pediatrics and Adolescent Medicine, First Faculty of Medicine, Research Unit for Rare DiseasesCharles UniversityPragueCzech Republic
| | - Tomas Palecek
- Department of Cardiovascular Medicine, Second Department of Medicine, First Faculty of MedicineCharles University and General University HospitalPragueCzech Republic
| | - Anna Chaloupka
- First Internal Clinic of Cardio‐AngiologySt. Anne's University Hospital and Medical School of Masaryk UniversityBrnoCzech Republic
| | - Ilga Grochova
- First Internal Clinic of Cardio‐AngiologySt. Anne's University Hospital and Medical School of Masaryk UniversityBrnoCzech Republic
| | - Jan Krejci
- First Internal Clinic of Cardio‐AngiologySt. Anne's University Hospital and Medical School of Masaryk UniversityBrnoCzech Republic
| | - Jana Petrkova
- Department of Internal Medicine I – CardiologyUniversity Hospital OlomoucOlomoucCzech Republic
- Department of Pathological Physiology, Faculty of Medicine and DentistryPalacky University OlomoucOlomoucCzech Republic
| | - Vojtech Melenovsky
- Department of CardiologyInstitute for Clinical and Experimental MedicinePragueCzech Republic
| | - Stanislav Kmoch
- Department of Pediatrics and Adolescent Medicine, First Faculty of Medicine, Research Unit for Rare DiseasesCharles UniversityPragueCzech Republic
| | - Josef Kautzner
- Department of CardiologyInstitute for Clinical and Experimental MedicinePragueCzech Republic
| |
Collapse
|
11
|
Jurgens SJ, Rämö JT, Kramarenko DR, Wijdeveld LFJM, Haas J, Chaffin MD, Garnier S, Gaziano L, Weng LC, Lipov A, Zheng SL, Henry A, Huffman JE, Challa S, Rühle F, Verdugo CD, Krijger Juárez C, Kany S, van Orsouw CA, Biddinger K, Poel E, Elliott AL, Wang X, Francis C, Ruan R, Koyama S, Beekman L, Zimmerman DS, Deleuze JF, Villard E, Trégouët DA, Isnard R, Boomsma DI, de Geus EJC, Tadros R, Pinto YM, Wilde AAM, Hottenga JJ, Sinisalo J, Niiranen T, Walsh R, Schmidt AF, Choi SH, Chang KM, Tsao PS, Matthews PM, Ware JS, Lumbers RT, van der Crabben S, Laukkanen J, Palotie A, Amin AS, Charron P, Meder B, Ellinor PT, Daly M, Aragam KG, Bezzina CR. Genome-wide association study reveals mechanisms underlying dilated cardiomyopathy and myocardial resilience. Nat Genet 2024; 56:2636-2645. [PMID: 39572784 PMCID: PMC11631763 DOI: 10.1038/s41588-024-01975-5] [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: 12/08/2023] [Accepted: 10/08/2024] [Indexed: 12/06/2024]
Abstract
Dilated cardiomyopathy (DCM) is a heart muscle disease that represents an important cause of morbidity and mortality, yet causal mechanisms remain largely elusive. Here, we perform a large-scale genome-wide association study and multitrait analysis for DCM using 9,365 cases and 946,368 controls. We identify 70 genome-wide significant loci, which show broad replication in independent samples and map to 63 prioritized genes. Tissue, cell type and pathway enrichment analyses highlight the central role of the cardiomyocyte and contractile apparatus in DCM pathogenesis. Polygenic risk scores constructed from our genome-wide association study predict DCM across different ancestry groups, show differing contributions to DCM depending on rare pathogenic variant status and associate with systolic heart failure across various clinical settings. Mendelian randomization analyses reveal actionable potential causes of DCM, including higher bodyweight and higher systolic blood pressure. Our findings provide insights into the genetic architecture and mechanisms underlying DCM and myocardial function more broadly.
Collapse
Affiliation(s)
- Sean J Jurgens
- Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Amsterdam UMC location, University of Amsterdam, Amsterdam, the Netherlands
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Joel T Rämö
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Daria R Kramarenko
- Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Amsterdam UMC location, University of Amsterdam, Amsterdam, the Netherlands
- European Reference Network for rare low prevalence and complex diseases of the heart: ERN GUARD-Heart, Amsterdam, the Netherlands
| | - Leonoor F J M Wijdeveld
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Physiology, Amsterdam UMC location, Vrije Universiteit, Amsterdam, the Netherlands
| | - Jan Haas
- Department of Medicine III, Institute for Cardiomyopathies Heidelberg (ICH), University Hospital Heidelberg, Heidelberg, Germany
- Site Heidelberg/Mannheim, DZHK, Heidelberg, Germany
| | - Mark D Chaffin
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Sophie Garnier
- Research Unit on Cardiovascular Disorders, Metabolism and Nutrition, Team Genomics and Pathophysiology of Cardiovascular Disease, Sorbone Université, INSERM, Paris, France
- ICAN Institute for Cardiometabolism and Nutrition, Paris, France
| | - Liam Gaziano
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Lu-Chen Weng
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Alex Lipov
- Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Amsterdam UMC location, University of Amsterdam, Amsterdam, the Netherlands
| | - Sean L Zheng
- National Heart and Lung Institute, Imperial College London, London, UK
- MRC Laboratory of Medical Sciences, Imperial College London, London, UK
- Royal Brompton and Harefield Hospitals, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - Albert Henry
- Institute of Cardiovascular Science, University College London, London, UK
- Institute of Health Informatics, University College London, London, UK
| | - Jennifer E Huffman
- Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC), VA Boston Healthcare System, Boston, MA, USA
- Palo Alto Veterans Institute for Research (PAVIR), Palo Alto Health Care System, Palo Alto, CA, USA
- Harvard Medical School, Boston, MA, USA
| | - Saketh Challa
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Frank Rühle
- Bioinformatics Core Facility, Institute of Molecular Biology gGmbH (IMB), Mainz, Germany
- Department of Genetic Epidemiology, Institute of Human Genetics, University of Münster, Münster, Germany
| | - Carmen Diaz Verdugo
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Christian Krijger Juárez
- Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Amsterdam UMC location, University of Amsterdam, Amsterdam, the Netherlands
| | - Shinwan Kany
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Department of Cardiology, University Heart and Vascular Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Constance A van Orsouw
- Department of Clinical Genetics, Amsterdam UMC location, University of Amsterdam, Amsterdam, the Netherlands
| | - Kiran Biddinger
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Edwin Poel
- Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Amsterdam UMC location, University of Amsterdam, Amsterdam, the Netherlands
| | - Amanda L Elliott
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
- Harvard Medical School, Boston, MA, USA
- Department of Psychiatry and Center for Genomic Medicine, Psychiatric and Neurodevelopmental Genetics Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Xin Wang
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Catherine Francis
- National Heart and Lung Institute, Imperial College London, London, UK
- Royal Brompton and Harefield Hospitals, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - Richard Ruan
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Satoshi Koyama
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Leander Beekman
- Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Amsterdam UMC location, University of Amsterdam, Amsterdam, the Netherlands
| | - Dominic S Zimmerman
- Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Amsterdam UMC location, University of Amsterdam, Amsterdam, the Netherlands
| | - Jean-François Deleuze
- CEA, Centre National de Recherche en Génomique Humaine, Université Paris-Saclay, Evry, France
- Laboratory of Excellence in Medical Genomics, GENMED, Evry, France
- Fondation Jean Dausset, Centre d'Etude du Polymorphisme Humain, Paris, France
| | - Eric Villard
- Research Unit on Cardiovascular Disorders, Metabolism and Nutrition, Team Genomics and Pathophysiology of Cardiovascular Disease, Sorbone Université, INSERM, Paris, France
- ICAN Institute for Cardiometabolism and Nutrition, Paris, France
| | - David-Alexandre Trégouët
- Laboratory of Excellence in Medical Genomics, GENMED, Evry, France
- Bordeaux Population Health Research Center, UMR 1219, University of Bordeaux, INSERM, Bordeaux, France
| | - Richard Isnard
- Research Unit on Cardiovascular Disorders, Metabolism and Nutrition, Team Genomics and Pathophysiology of Cardiovascular Disease, Sorbone Université, INSERM, Paris, France
- ICAN Institute for Cardiometabolism and Nutrition, Paris, France
- APHP, Cardiology and Genetics Departments, Pitié-Salpêtrière Hospital, Paris, France
| | - Dorret I Boomsma
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Eco J C de Geus
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Amsterdam Public Health Research Institute, Amsterdam UMC location, Vrije Universiteit, Amsterdam, the Netherlands
| | - Rafik Tadros
- Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Amsterdam UMC location, University of Amsterdam, Amsterdam, the Netherlands
- Cardiovascular Genetics Centre, Montreal Heart Institute, Montreal, QC, Canada
- Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Yigal M Pinto
- Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Amsterdam UMC location, University of Amsterdam, Amsterdam, the Netherlands
- European Reference Network for rare low prevalence and complex diseases of the heart: ERN GUARD-Heart, Amsterdam, the Netherlands
- Department of Clinical Cardiology, Amsterdam Cardiovascular Sciences, Heart Failure and Arrhythmias, Amsterdam UMC location, University of Amsterdam, Amsterdam, the Netherlands
| | - Arthur A M Wilde
- Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Amsterdam UMC location, University of Amsterdam, Amsterdam, the Netherlands
- European Reference Network for rare low prevalence and complex diseases of the heart: ERN GUARD-Heart, Amsterdam, the Netherlands
- Department of Clinical Cardiology, Amsterdam Cardiovascular Sciences, Heart Failure and Arrhythmias, Amsterdam UMC location, University of Amsterdam, Amsterdam, the Netherlands
| | - Jouke-Jan Hottenga
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- The Netherlands Twin Register, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Juha Sinisalo
- Department of Cardiology, Helsinki University Hospital, Helsinki, Finland
- Heart and Lung Center, Helsinki University Hospital and Helsinki University, Helsinki, Finland
| | - Teemu Niiranen
- Department of Internal Medicine, University of Turku, Helsinki, Finland
- Division of Medicine, Turku University Hospital, Helsinki, Finland
- Finnish Institute for Health and Welfare (THL), Helsinki, Finland
| | - Roddy Walsh
- Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Amsterdam UMC location, University of Amsterdam, Amsterdam, the Netherlands
| | - Amand F Schmidt
- Department of Clinical Cardiology, Amsterdam Cardiovascular Sciences, Heart Failure and Arrhythmias, Amsterdam UMC location, University of Amsterdam, Amsterdam, the Netherlands
- Institute of Cardiovascular Science, Faculty of Population Health, University College London, London, UK
- University College London British Heart Foundation Research Accelerator, London, UK
- Department of Cardiology, Division Heart and Lungs, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Seung Hoan Choi
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Biostatistics, Boston University, Boston, MA, USA
| | - Kyong-Mi Chang
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Philip S Tsao
- Palo Alto Health Care System, Palo Alto, CA, USA
- Department of Medicine and Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Paul M Matthews
- National Heart and Lung Institute, Imperial College London, London, UK
- MRC Laboratory of Medical Sciences, Imperial College London, London, UK
| | - James S Ware
- National Heart and Lung Institute, Imperial College London, London, UK
- MRC Laboratory of Medical Sciences, Imperial College London, London, UK
- Royal Brompton and Harefield Hospitals, Guy's and St. Thomas' NHS Foundation Trust, London, UK
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - R Thomas Lumbers
- Institute of Health Informatics, University College London, London, UK
- The National Institute for Health Research, University College London Hospitals Biomedical Research Centre, University College London, London, UK
| | - Saskia van der Crabben
- Department of Clinical Genetics, Amsterdam UMC location, University of Amsterdam, Amsterdam, the Netherlands
| | - Jari Laukkanen
- Department of Medicine, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
- Central Finland Biobank, Central Finland Health Care District, Jyväskylä, Finland
| | - Aarno Palotie
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
- Program in Medical and Population Genetics and Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
| | - Ahmad S Amin
- Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Amsterdam UMC location, University of Amsterdam, Amsterdam, the Netherlands
- European Reference Network for rare low prevalence and complex diseases of the heart: ERN GUARD-Heart, Amsterdam, the Netherlands
- Department of Clinical Cardiology, Amsterdam Cardiovascular Sciences, Heart Failure and Arrhythmias, Amsterdam UMC location, University of Amsterdam, Amsterdam, the Netherlands
| | - Philippe Charron
- Research Unit on Cardiovascular Disorders, Metabolism and Nutrition, Team Genomics and Pathophysiology of Cardiovascular Disease, Sorbone Université, INSERM, Paris, France
- ICAN Institute for Cardiometabolism and Nutrition, Paris, France
- APHP, Cardiology and Genetics Departments, Pitié-Salpêtrière Hospital, Paris, France
| | - Benjamin Meder
- Department of Medicine III, Institute for Cardiomyopathies Heidelberg (ICH), University Hospital Heidelberg, Heidelberg, Germany
- Site Heidelberg/Mannheim, DZHK, Heidelberg, Germany
| | - Patrick T Ellinor
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA.
| | - Mark Daly
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland.
- Program in Medical and Population Genetics and Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA.
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA.
| | - Krishna G Aragam
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA.
| | - Connie R Bezzina
- Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Amsterdam UMC location, University of Amsterdam, Amsterdam, the Netherlands.
- European Reference Network for rare low prevalence and complex diseases of the heart: ERN GUARD-Heart, Amsterdam, the Netherlands.
| |
Collapse
|
12
|
Tran DD, Lien NTK, Tung NV, Huu NC, Nguyen PT, Tien DA, Thu DTH, Huy BQ, Oanh TTK, Lien NTP, Hien NT, Lan NN, Thanh LT, Duc NM, Hoang NH. Three Novel Pathogenic Variants in Unrelated Vietnamese Patients with Cardiomyopathy. Diagnostics (Basel) 2024; 14:2709. [PMID: 39682617 DOI: 10.3390/diagnostics14232709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 11/26/2024] [Accepted: 11/29/2024] [Indexed: 12/18/2024] Open
Abstract
Background: Cardiomyopathy, including dilated cardiomyopathy (DCM) and hypertrophic cardiomyopathy (HCM), is a major cause of heart failure (HF) and a leading indication for heart transplantation. Of these patients, 20-50% have a genetic cause, so understanding the genetic basis of cardiomyopathy will provide knowledge about the pathogenesis of the disease for diagnosis, treatment, prevention, and genetic counseling for families. Methods: This study collected nine patients from different Vietnamese families for genetic analysis at The Cardiovascular Center, E Hospital, Hanoi, Vietnam. The patients were diagnosed with cardiomyopathy based on clinical symptoms. Whole-exome sequencing (WES) was performed in the Vietnamese patients to identify variants associated with cardiomyopathy, and the Sanger sequencing method was used to validate the variants in the patients' families. The influence of the variants was predicted using in silico analysis tools. Results: Nine heterozygous variants were detected as a cause of disease in the patients, three of which were novel variants, including c.284C>G, p.Pro95Arg in the MYL2 gene, c.2356A>G, p.Thr786Ala in the MYH7 gene, and c.1223T>A, p.Leu408Gln in the DES gene. Two other variants were pathogenic variants (c.602T>C, p.Ile201Thr in the MYH7 gene and c.1391G>C, p.Gly464Ala in the PTPN11 gene), and four were variants of uncertain significance in the ACTA2, ANK2, MYOZ2, and PRKAG2 genes. The results of the in silico prediction software showed that the identified variants were pathogenic and responsible for the patients' DCM. Conclusions: Our results contribute to the understanding of cardiomyopathy pathogenesis and provide a basis for diagnosis, treatment, prevention, and genetic counseling.
Collapse
Affiliation(s)
- Dac Dai Tran
- E Hospital, Ministry of Health, 89 Tran Cung Str., Cau Giay, Hanoi 100000, Vietnam
| | - Nguyen Thi Kim Lien
- Institute of Genome Research, Vietnam Academy of Science and Technology, 18-Hoang Quoc Viet Str., Cau Giay, Hanoi 100000, Vietnam
| | - Nguyen Van Tung
- Institute of Genome Research, Vietnam Academy of Science and Technology, 18-Hoang Quoc Viet Str., Cau Giay, Hanoi 100000, Vietnam
- Faculty of Biotechnology, Graduate University of Science and Technology, Vietnam Academy of Science and Technology, Cau Giay, Hanoi 100000, Vietnam
| | - Nguyen Cong Huu
- E Hospital, Ministry of Health, 89 Tran Cung Str., Cau Giay, Hanoi 100000, Vietnam
| | - Phan Thao Nguyen
- E Hospital, Ministry of Health, 89 Tran Cung Str., Cau Giay, Hanoi 100000, Vietnam
| | - Do Anh Tien
- E Hospital, Ministry of Health, 89 Tran Cung Str., Cau Giay, Hanoi 100000, Vietnam
| | - Doan Thi Hoai Thu
- E Hospital, Ministry of Health, 89 Tran Cung Str., Cau Giay, Hanoi 100000, Vietnam
| | - Bui Quang Huy
- E Hospital, Ministry of Health, 89 Tran Cung Str., Cau Giay, Hanoi 100000, Vietnam
| | - Tran Thi Kim Oanh
- E Hospital, Ministry of Health, 89 Tran Cung Str., Cau Giay, Hanoi 100000, Vietnam
| | | | - Nguyen Thanh Hien
- Institute of Genome Research, Vietnam Academy of Science and Technology, 18-Hoang Quoc Viet Str., Cau Giay, Hanoi 100000, Vietnam
| | - Nguyen Ngoc Lan
- Institute of Genome Research, Vietnam Academy of Science and Technology, 18-Hoang Quoc Viet Str., Cau Giay, Hanoi 100000, Vietnam
- Center for Gene and Protein Research, Hanoi Medical University, 1st Ton That Tung Str., Dong Da, Hanoi 100000, Vietnam
| | - Le Tat Thanh
- Institute of Genome Research, Vietnam Academy of Science and Technology, 18-Hoang Quoc Viet Str., Cau Giay, Hanoi 100000, Vietnam
| | - Nguyen Minh Duc
- Institute of Genome Research, Vietnam Academy of Science and Technology, 18-Hoang Quoc Viet Str., Cau Giay, Hanoi 100000, Vietnam
- National Research Center for Medicinal Plant Germplasm & Breeding, National Institute of Medicinal Materials, Thanh Tri, Hanoi 100000, Vietnam
| | - Nguyen Huy Hoang
- Institute of Genome Research, Vietnam Academy of Science and Technology, 18-Hoang Quoc Viet Str., Cau Giay, Hanoi 100000, Vietnam
- Faculty of Biotechnology, Graduate University of Science and Technology, Vietnam Academy of Science and Technology, Cau Giay, Hanoi 100000, Vietnam
| |
Collapse
|
13
|
Sheikh M, Fallah SA, Moradi M, Jalali A, Vakili-Basir A, Sahebjam M, Ashraf H, Zoroufian A. Comparing HeartModel AI and cardiac magnetic resonance imaging for left ventricular volume and function evaluation in patients with dilated cardiomyopathy. BMC Cardiovasc Disord 2024; 24:670. [PMID: 39580388 PMCID: PMC11585202 DOI: 10.1186/s12872-024-04355-3] [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: 03/12/2024] [Accepted: 11/18/2024] [Indexed: 11/25/2024] Open
Abstract
BACKGROUND Integration of artificial intelligence enhances precision, yielding dependable evaluations of left ventricular volumes and ejection fraction despite image quality variations. Commercial software like HeartModelAI provides fully automated 3DE quantification, simplifying the measurement of left chamber volumes and ejection fraction. In this manuscript, we present a cross-sectional study to assess and compare the diagnostic accuracy of automated 3D echocardiography (HeartModelAI) to the standard Cardiac Magnetic Resonance Imaging in patients with dilated cardiomyopathy. METHODS In this cross-sectional study, 30 patients with dilated cardiomyopathy referring to the Tehran Heart Center with cardiac magnetic resonance imaging and comprehensive 3D transthoracic echocardiography within 24 h were included. All 3D volume analysis was performed with fully automated quantification software (HeartModelAI) using 3D images of 2,3, and 4-chamber views at the end of systole and diastole. RESULTS Excellent Inter- and Intra-observer correlation coefficient was reported for HeartModelAI software for all indexes. HeartModelAI displayed a remarkable correlation with cardiac magnetic resonance for left ventricular end-systolic volume index (r = 0.918 and r = 0.911); nevertheless, it underestimated left ventricular end-systolic volume index and left ventricular end-diastolic volume index. Conversely, ejection fraction, stroke volume, and left ventricular mass were overestimated. It was found that manual contour correction can enhance the accuracy of automated model estimations, particularly concerning EF in participants needing correction. CONCLUSION HeartModelAI software emerges as a rapid and viable imaging approach for evaluating the left ventricle's structure and function. In our study, LV volumes assessed by HeartModelAI demonstrated strong correlations with cardiac magnetic resonance imaging.
Collapse
Affiliation(s)
| | - Sahar Asl Fallah
- Cardiovascular Diseases Research Institute, Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Muhammadhosein Moradi
- Cardiac Primary Prevention Research Center, Cardiovascular Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Arash Jalali
- Cardiac Primary Prevention Research Center, Cardiovascular Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
- Department of Epidemiology and Biostatistics, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Ahmad Vakili-Basir
- Cardiovascular Diseases Research Institute, Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Epidemiology and Biostatistics, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Sahebjam
- Cardiovascular Diseases Research Institute, Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Haleh Ashraf
- Cardiac Primary Prevention Research Center, Cardiovascular Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Arezou Zoroufian
- Cardiovascular Diseases Research Institute, Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran.
- Echocardiography Department, Cardiovascular Diseases Research Institute, Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
14
|
Patel YB, David W, Terzic M, Mehla A, Swadia TV. BAG-3 Mutation Dilated Cardiomyopathy With Left Ventricular Noncompaction in Young Healthy Adult. JACC Case Rep 2024; 29:102648. [PMID: 39691879 PMCID: PMC11646925 DOI: 10.1016/j.jaccas.2024.102648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 08/17/2024] [Accepted: 08/23/2024] [Indexed: 12/19/2024]
Abstract
Cardiomyopathies are generally divided into ischemic and nonischemic types. Dilated cardiomyopathies, which are a type of nonischemic cardiomyopathy, may have a trait of left ventricular noncompaction. We present the case of a 34-year-old man with new-onset decompensated heart failure and left ventricular noncompaction from a BAG3 (Bcl-2 associated athanogene 3) truncating mutation.
Collapse
Affiliation(s)
- Yash B. Patel
- Department of Internal Medicine, Trinity Health Ann Arbor, Michigan, USA
| | - Wadie David
- Department of Cardiology, Ascension Southfield, Michigan, USA
| | - Milan Terzic
- Department of Internal Medicine, Trinity Health Ann Arbor, Michigan, USA
| | - Akanksha Mehla
- Department of Cardiology, University of Michigan Health Sparrow Hospital, Michigan, USA
| | | |
Collapse
|
15
|
Yan J, Wang X, Cao P, Li Q, Wu H. Downregulation of miR-214 promotes dilated Cardiomyopathy Progression through PDE5A-Mediated cGMP regulation. Sci Rep 2024; 14:28070. [PMID: 39543318 PMCID: PMC11564883 DOI: 10.1038/s41598-024-78983-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 11/05/2024] [Indexed: 11/17/2024] Open
Abstract
Dilated cardiomyopathy (DCM) is a myocardial disorder resulting in a substantial decline in cardiac function and potentially leading to heart failure. This research combines bioinformatics analysis with empirical validation to explore the roles and mechanisms of miR-214 in DCM. Using the DEseq2 R package, a total of 125 differentially expressed circulating miRNAs (DE c-miRNAs) and 784 DE genes (DEGs) were identified. Cross-analysis between target genes of DE c-miRNAs and DEGs identified 124 common genes, and protein-protein interaction analysis of common genes identified 11 hub genes. Twelve DE c-miRNAs were further verified by quantifying their levels in the serum of DCM patients and healthy individuals. miR-214 levels were significantly decreased in serum from DCM patients, positively correlated with left ventricular ejection fraction and left ventricular fractional shortening. Further analysis showed that miR-214 directly targets and negatively regulates phosphodiesterase 5 A (PDE5A). Elevated PDE5A expression reduced cGMP levels; however, using sildenafil, a PDE5A inhibitor, reversed this effect, substantiating the regulatory mechanism of miR-214 on cGMP via PDE5A. These results provide new potential targets for the diagnosis and treatment of DCM.
Collapse
Affiliation(s)
- Jingjing Yan
- Graduate School, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Xinzhou Wang
- Second Clinical Medical College, Henan University of Chinese Medicine, Zhengzhou, 450002, China
- Laboratory of Cell Imaging, Henan University of Chinese Medicine, 6 Dongfeng Rd, Zhengzhou, 450002, Henan, China
| | - Panxia Cao
- Graduate School, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Qiaozhi Li
- Heart Center, First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, 450002, China
| | - Hong Wu
- Second Clinical Medical College, Henan University of Chinese Medicine, Zhengzhou, 450002, China.
- Laboratory of Cell Imaging, Henan University of Chinese Medicine, 6 Dongfeng Rd, Zhengzhou, 450002, Henan, China.
- Institute of Cardiovascular Disease, Henan University of Chinese Medicine, Zhengzhou, 450002, China.
| |
Collapse
|
16
|
Garg A, Jansen S, Greenberg L, Zhang R, Lavine KJ, Greenberg MJ. Dilated cardiomyopathy-associated skeletal muscle actin (ACTA1) mutation R256H disrupts actin structure and function and causes cardiomyocyte hypocontractility. Proc Natl Acad Sci U S A 2024; 121:e2405020121. [PMID: 39503885 PMCID: PMC11572969 DOI: 10.1073/pnas.2405020121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Accepted: 09/17/2024] [Indexed: 11/13/2024] Open
Abstract
Skeletal muscle actin (ACTA1) mutations are a prevalent cause of skeletal myopathies consistent with ACTA1's high expression in skeletal muscle. Rare de novo mutations in ACTA1 associated with combined cardiac and skeletal myopathies have been reported, but ACTA1 represents only ~20% of the total actin pool in cardiomyocytes, making its role in cardiomyopathy controversial. Here we demonstrate how a mutation in an actin isoform expressed at low levels in cardiomyocytes can cause cardiomyopathy by focusing on a unique ACTA1 variant, R256H. We previously identified this variant in a family with dilated cardiomyopathy, who had reduced systolic function without clinical skeletal myopathy. Using a battery of multiscale biophysical tools, we show that R256H has potent effects on ACTA1 function at the molecular scale and in human cardiomyocytes. Importantly, we demonstrate that R256H acts in a dominant manner, where the incorporation of small amounts of mutant protein into thin filaments is sufficient to disrupt molecular contractility, and that this effect is dependent on the presence of troponin and tropomyosin. To understand the structural basis of this change in regulation, we resolved a structure of R256H filaments using cryoelectron microscopy, and we see alterations in actin's structure that have the potential to disrupt interactions with tropomyosin. Finally, we show that ACTA1R256H/+ human-induced pluripotent stem cell cardiomyocytes demonstrate reduced contractility and sarcomeric organization. Taken together, we demonstrate that R256H has multiple effects on ACTA1 function that are sufficient to cause reduced contractility and establish a likely causative relationship between ACTA1 R256H and clinical cardiomyopathy.
Collapse
Affiliation(s)
- Ankit Garg
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, MD21205
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO63110
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO63110
| | - Silvia Jansen
- Department of Cell Biology and Physiology, Washington University in St. Louis, St. Louis, MO63110
| | - Lina Greenberg
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO63110
| | - Rui Zhang
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO63110
| | - Kory J. Lavine
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO63110
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO63110
| | - Michael J. Greenberg
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO63110
| |
Collapse
|
17
|
de Noronha IM, Almeida LX, de Souza Silva Andrade NV, de França EET, de Morais Lima JH, Pedrosa R, Siqueira F, Onofre T. Respiratory Muscle Strength and Quality of Life in Patients With Heart Failure and Their Main Correlated Factors: A Cross-sectional Study. J Cardiovasc Nurs 2024; 39:535-542. [PMID: 37955376 DOI: 10.1097/jcn.0000000000001062] [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] [Indexed: 11/14/2023]
Abstract
BACKGROUND Heart failure may cause peripheral and respiratory muscle alterations, dyspnea, fatigue, and exercise intolerance, worsening the quality of life of patients. OBJECTIVES The aims of this study were to analyze respiratory muscle strength and quality of life of patients with heart failure and correlate them with clinical variables and functional classification. METHODS This cross-sectional study involved patients with heart failure. A manovacuometer assessed maximum inspiratory and expiratory pressures, and quality of life was assessed using the Minnesota Living with Heart Failure Questionnaire. Functional classification was categorized according to the New York Heart Association (NYHA) class in I, II, III, or IV. RESULTS We included 60 patients (66.7% male) with a mean age of 62.0 years and mean left ventricular ejection fraction of 42.0%. Maximum inspiratory pressure and maximum expiratory pressure were close to normal (>70% of predicted) in most patients; however, a subgroup composed mostly of patients with dilated heart failure and NYHA class III (n = 21) presented low maximum inspiratory pressure values (59.2%; 95% confidence interval, 55.7%-62.8%). The mean total score of the Minnesota Living with Heart Failure Questionnaire was 44.4 points, being negatively correlated with left ventricular ejection fraction ( r = -0.29, P = .02). Patients with NYHA class III and disease duration longer than 120 months presented higher total ( P < .01) and physical dimension scores. CONCLUSIONS Most patients had respiratory muscle strength close to normal; however, those with dilated heart failure and NYHA class III presented low maximum inspiratory pressure values. Quality of life was moderately compromised, mainly because of long disease duration, NYHA class III, and low left ventricular ejection fraction.
Collapse
|
18
|
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
|
19
|
Gigli M, Stolfo D, Merlo M, Sinagra G, Taylor MRG, Mestroni L. Pathophysiology of dilated cardiomyopathy: from mechanisms to precision medicine. Nat Rev Cardiol 2024:10.1038/s41569-024-01074-2. [PMID: 39394525 DOI: 10.1038/s41569-024-01074-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/20/2024] [Indexed: 10/13/2024]
Abstract
Dilated cardiomyopathy (DCM) is a complex disease with multiple causes and various pathogenic mechanisms. Despite improvements in the prognosis of patients with DCM in the past decade, this condition remains a leading cause of heart failure and premature death. Conventional treatment for DCM is based on the foundational therapies for heart failure with reduced ejection fraction. However, increasingly, attention is being directed towards individualized treatments and precision medicine. The ability to confirm genetic causality is gradually being complemented by an increased understanding of genotype-phenotype correlations. Non-genetic factors also influence the onset of DCM, and growing evidence links genetic background with concomitant non-genetic triggers or precipitating factors, increasing the extreme complexity of the pathophysiology of DCM. This Review covers the spectrum of pathophysiological mechanisms in DCM, from monogenic causes to the coexistence of genetic abnormalities and triggering environmental factors (the 'two-hit' hypothesis). The roles of common genetic variants in the general population and of gene modifiers in disease onset and progression are also discussed. Finally, areas for future research are highlighted, particularly novel therapies, such as small molecules, RNA and gene therapy, and measures for the prevention of arrhythmic death.
Collapse
Affiliation(s)
- Marta Gigli
- Cardiothoracovascular Department, Centre for Diagnosis and Treatment of Cardiomyopathies, European Reference Network for Rare, Low Prevalence and Complex Diseases of the Heart (ERN GUARD-Heart), Azienda Sanitaria Universitaria Giuliano-Isontina (ASUGI) and University of Trieste, Trieste, Italy
| | - Davide Stolfo
- Cardiothoracovascular Department, Centre for Diagnosis and Treatment of Cardiomyopathies, European Reference Network for Rare, Low Prevalence and Complex Diseases of the Heart (ERN GUARD-Heart), Azienda Sanitaria Universitaria Giuliano-Isontina (ASUGI) and University of Trieste, Trieste, Italy
- Division of Cardiology, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Marco Merlo
- Cardiothoracovascular Department, Centre for Diagnosis and Treatment of Cardiomyopathies, European Reference Network for Rare, Low Prevalence and Complex Diseases of the Heart (ERN GUARD-Heart), Azienda Sanitaria Universitaria Giuliano-Isontina (ASUGI) and University of Trieste, Trieste, Italy
| | - Gianfranco Sinagra
- Cardiothoracovascular Department, Centre for Diagnosis and Treatment of Cardiomyopathies, European Reference Network for Rare, Low Prevalence and Complex Diseases of the Heart (ERN GUARD-Heart), Azienda Sanitaria Universitaria Giuliano-Isontina (ASUGI) and University of Trieste, Trieste, Italy
| | - Matthew R G Taylor
- Adult Medical Genetics Program, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Luisa Mestroni
- Molecular Genetics Program, Cardiovascular Institute, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
20
|
Nasonova SN, Meshkov AN, Zhirov IV, Osmolovskaya YF, Shoshina AA, Gagloev AV, Dzhumaniiazova IH, Zelenova EA, Erema VV, Gusakova MS, Ivanov MV, Terekhov MV, Kashtanova DA, Nekrasova AI, Mitrofanov SI, Shingaliev AS, Yudin VS, Keskinov AA, Gomyranova NV, Chubykina UV, Ezhov MV, Tereshchenko SN, Yudin SM, Boytsov SA. [A clinical case of reverse left ventricular remodeling in patient with pathogenic TTN mutation. Case report]. TERAPEVT ARKH 2024; 96:901-908. [PMID: 39467245 DOI: 10.26442/00403660.2024.09.202852] [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: 07/22/2024] [Accepted: 07/22/2024] [Indexed: 10/30/2024]
Abstract
Dilated cardiomyopathy (DCM) is a leading cause of heart failure, sudden cardiac death, and heart transplantation in young patients. The causes of DCM are varied and include genetic factors and metabolic, infectious, toxic and others factors. Today it is known that germline mutations in more than 98 genes can be associated with the occurrence of DCM. However, the penetrance of these genes often depends on a combination of factors, including modifiable ones, i.e. those that change under the influence of the environment. About 20-25% of genetically determined forms of DCM are due to mutations in the titin gene (TTN). Titin is the largest protein in the body, which is an important component of the sarcomer. Although titin is the largest protein in the human body, its role in the physiology of heart and disease is not yet fully understood. However, a mutation in the TTN gene may later represent a potential therapeutic target for genetic and acquired cardiomyopathy. Thus, the analysis of clinical cases of cardiomyopathy in patients with identified mutations in the TTN gene is of great scientific interest. The article presents a clinical case of manifestation of DCM in patient with a revealed pathogenic variant of mutation in the gene TTN and reverse left ventricular remodeling of the against the background of optimal therapy of heart failure in a subsequent outpatient observation.
Collapse
Affiliation(s)
- S N Nasonova
- Chazov National Medical Research Center of Cardiology
| | - A N Meshkov
- Chazov National Medical Research Center of Cardiology
- National Research Center for Therapy and Preventive Medicine
| | - I V Zhirov
- Chazov National Medical Research Center of Cardiology
| | | | - A A Shoshina
- Chazov National Medical Research Center of Cardiology
| | - A V Gagloev
- Chazov National Medical Research Center of Cardiology
| | | | - E A Zelenova
- Centre for Strategic Planning and Management of Biomedical Health Risks
| | - V V Erema
- Centre for Strategic Planning and Management of Biomedical Health Risks
| | - M S Gusakova
- Centre for Strategic Planning and Management of Biomedical Health Risks
| | - M V Ivanov
- Centre for Strategic Planning and Management of Biomedical Health Risks
| | - M V Terekhov
- Centre for Strategic Planning and Management of Biomedical Health Risks
| | - D A Kashtanova
- Centre for Strategic Planning and Management of Biomedical Health Risks
| | - A I Nekrasova
- Centre for Strategic Planning and Management of Biomedical Health Risks
| | - S I Mitrofanov
- Centre for Strategic Planning and Management of Biomedical Health Risks
| | - A S Shingaliev
- Centre for Strategic Planning and Management of Biomedical Health Risks
| | - V S Yudin
- Centre for Strategic Planning and Management of Biomedical Health Risks
| | - A A Keskinov
- Centre for Strategic Planning and Management of Biomedical Health Risks
| | | | - U V Chubykina
- Chazov National Medical Research Center of Cardiology
| | - M V Ezhov
- Chazov National Medical Research Center of Cardiology
| | | | - S M Yudin
- Centre for Strategic Planning and Management of Biomedical Health Risks
| | - S A Boytsov
- Chazov National Medical Research Center of Cardiology
| |
Collapse
|
21
|
Qu L, Duan X, Chen H. The effects of sodium-glucose cotransporter 2 inhibitors on the 'forgotten' right ventricle. ESC Heart Fail 2024. [PMID: 39370371 DOI: 10.1002/ehf2.15103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 09/11/2024] [Accepted: 09/17/2024] [Indexed: 10/08/2024] Open
Abstract
With the progress in diagnosis, treatment and imaging techniques, there is a growing recognition that impaired right ventricular (RV) function profoundly affects the prognosis of patients with heart failure (HF), irrespective of their left ventricular ejection fraction (LVEF). In addition, right HF (RHF) is a common complication associated with various diseases, including congenital heart disease, myocardial infarction (MI), pulmonary arterial hypertension (PAH) and dilated cardiomyopathy (DCM), and it can manifest at any time after left ventricular assist devices (LVADs). The sodium-glucose cotransporter 2 (SGLT2) inhibition by gliflozins has emerged as a cornerstone medicine for managing type 2 diabetes mellitus (T2DM) and HF, with an increasing focus on its potential to enhance RV function. In this review, we aim to present an updated perspective on the pleiotropic effects of gliflozins on the right ventricle and offer insights into the underlying mechanisms. We can ascertain their advantageous impact on the right ventricle by discussing the evidence obtained in animal models and monumental clinical trials. In light of the pathophysiological changes in RHF, we attempt to elucidate crucial mechanisms regarding their beneficial effects, including alleviation of RV overload, reduction of hyperinsulinaemia and inflammatory responses, regulation of nutrient signalling pathways and cellular energy metabolism, inhibition of oxidative stress and myocardial fibrosis, and maintenance of ion balance. Finally, this drug class's potential application and benefits in various clinical settings are described, along with a prospective outlook on future clinical practice and research directions.
Collapse
Affiliation(s)
- Liangzhen Qu
- Department of Cardiovascular Medicine, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, China
- International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| | - Xueting Duan
- Department of Cardiovascular Medicine, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, China
- International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| | - Han Chen
- Department of Cardiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
22
|
Hu Y, Zou Y, Qiao L, Lin L. Integrative proteomic and metabolomic elucidation of cardiomyopathy with in vivo and in vitro models and clinical samples. Mol Ther 2024; 32:3288-3312. [PMID: 39233439 PMCID: PMC11489546 DOI: 10.1016/j.ymthe.2024.08.030] [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: 04/30/2024] [Revised: 07/16/2024] [Accepted: 08/30/2024] [Indexed: 09/06/2024] Open
Abstract
Cardiomyopathy is a prevalent cardiovascular disease that affects individuals of all ages and can lead to life-threatening heart failure. Despite its variety in types, each with distinct characteristics and causes, our understanding of cardiomyopathy at a systematic biology level remains incomplete. Mass spectrometry-based techniques have emerged as powerful tools, providing a comprehensive view of the molecular landscape and aiding in the discovery of biomarkers and elucidation of mechanisms. This review highlights the significant potential of integrating proteomic and metabolomic approaches with specialized databases to identify biomarkers and therapeutic targets across different types of cardiomyopathies. In vivo and in vitro models, such as genetically modified mice, patient-derived or induced pluripotent stem cells, and organ chips, are invaluable in exploring the pathophysiological complexities of this disease. By integrating omics approaches with these sophisticated modeling systems, our comprehension of the molecular underpinnings of cardiomyopathy can be greatly enhanced, facilitating the development of diagnostic markers and therapeutic strategies. Among the promising therapeutic targets are those involved in extracellular matrix remodeling, sarcomere damage, and metabolic remodeling. These targets hold the potential to advance precision therapy in cardiomyopathy, offering hope for more effective treatments tailored to the specific molecular profiles of patients.
Collapse
Affiliation(s)
- Yiwei Hu
- Department of Chemistry, Zhongshan Hospital, and Minhang Hospital, Fudan University, Shanghai 200000, China
| | - Yunzeng Zou
- Department of Chemistry, Zhongshan Hospital, and Minhang Hospital, Fudan University, Shanghai 200000, China.
| | - Liang Qiao
- Department of Chemistry, Zhongshan Hospital, and Minhang Hospital, Fudan University, Shanghai 200000, China.
| | - Ling Lin
- Department of Chemistry, Zhongshan Hospital, and Minhang Hospital, Fudan University, Shanghai 200000, China.
| |
Collapse
|
23
|
Du Z, Zhou Y, Li Q, Xie Y, Zhu T, Qiao J, Zhang R, Bao Y, Wang L, Xie Y, Quan J, Lin M, Zhang N, Jin Q, Liang W, Wu L, Yin T, Xie Y. SIRT1 Ameliorates Lamin A/C Deficiency-Induced Cardiac Dysfunction by Promoting Mitochondrial Bioenergetics. JACC Basic Transl Sci 2024; 9:1211-1230. [PMID: 39534638 PMCID: PMC11551877 DOI: 10.1016/j.jacbts.2024.05.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 05/10/2024] [Accepted: 05/29/2024] [Indexed: 11/16/2024]
Abstract
Dilated cardiomyopathy (DCM) is associated with high mortality despite advanced therapies. The LMNA gene encodes lamin A/C and is the second most frequently mutated gene associated with DCM, for which therapeutic options are limited. Here we generated Lmna -/- mice and found they exhibited cardiac dysfunction at the age of 1 month but not at 2 weeks. Proteomics showed down-regulation of mitochondrial function-related pathways in Lmna -/- hearts. Moreover, early injured mitochondria with decreased cristae density and sirtuin 1 (SIRT1) down-regulation were observed in 2-week-old Lmna -/- hearts. Adenoviral overexpression of SIRT1 in lamin A/C knockdown neonatal rat ventricular myocytes improved mitochondrial oxidative respiration capacity. Adeno-associated virus-mediated SIRT1 overexpression alleviated mitochondrial injury, cardiac systolic dysfunction, ventricular dilation, and fibrosis, and prolonged lifespan in Lmna -/- mice. Mechanistically, LMNA maintains mitochondrial bioenergetics through the SIRT1-PARKIN axis. Our results suggest that targeting the SIRT1 signaling pathway is expected to be a novel therapeutic strategy for LMNA mutation-associated DCM.
Collapse
Affiliation(s)
- Zunhui Du
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yanting Zhou
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qiheng Li
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuan Xie
- College of Osteopathic Medicine, Kansas City University, Kansas City, Missouri, USA
| | - Tingfang Zhu
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jing Qiao
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ruihong Zhang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yangyang Bao
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lingjie Wang
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yinyin Xie
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jinwei Quan
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Menglu Lin
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ning Zhang
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qi Jin
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenbin Liang
- University of Ottawa Heart Institute, Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Liqun Wu
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tong Yin
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yucai Xie
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
24
|
Karadas H, Tosun H, Ceylan H. Identification of dilated cardiomyopathy-linked key genes by bioinformatics methods and evaluating the impact of tannic acid and monosodium glutamate in rats. Biotechnol Appl Biochem 2024. [PMID: 39318238 DOI: 10.1002/bab.2670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 09/05/2024] [Indexed: 09/26/2024]
Abstract
Dilated cardiomyopathy (DCM) is the most common type of myocardial dysfunction, affecting mostly young adults, but its therapeutic diagnosis and biomarkers for prognosis are lacking. This study aimed to investigate the possible effect of the common food additive monosodium glutamate (MSG) and tannic acid (TA), a phenolic compound, on the key molecular actors responsible for DCM. DCM-related publicly available microarray datasets (GSE120895, GSE17800, and GSE19303) were downloaded from the comprehensive Gene Expression Omnibus (GEO) database, and analyzed to identify differentially expressed genes (DEGs). By integrating DEGs and gene-disease validity curation results, overlapping genes were screened and identified as hub genes. Protein-protein interaction (PPI) network and ontology analysis were performed to make sense of the identified biological data. Finally, mRNA expression changes of identified hub genes in the heart tissues of rats treated with MSG and TA were measured by the qPCR method. Six upregulated (IGF1, TTN, ACTB, LMNA, EDN1, and NPPB) DEGs were identified between the DCM and healthy control samples as the hub genes. qPCR results revealed that the mRNA levels of these genes involved in DCM development increased significantly in rat heart tissues exposed to MSG. In contrast, this increase was remarkably alleviated by TA treatment. Our results provide new insights into critical molecular mechanisms that should be focused on in future DCM studies. Moreover, MSG may play a critical role in DCM formation, and TA may be used as a promising therapeutic agent in DCM.
Collapse
Affiliation(s)
- Habibe Karadas
- Department of Molecular Biology and Genetics, Faculty of Science, Atatürk University, Erzurum, Turkey
| | - Hilal Tosun
- Department of Molecular Biology and Genetics, Faculty of Science, Atatürk University, Erzurum, Turkey
| | - Hamid Ceylan
- Department of Molecular Biology and Genetics, Faculty of Science, Atatürk University, Erzurum, Turkey
| |
Collapse
|
25
|
Du H, Zhao Y, Wen J, Dai B, Hu G, Zhou Y, Yin Z, Ding N, Li H, Fan J, Nie X, Wang F, Liu Q, Wen Z, Xu G, Wang DW, Chen C. LncRNA DCRT Protects Against Dilated Cardiomyopathy by Preventing NDUFS2 Alternative Splicing by Binding to PTBP1. Circulation 2024; 150:1030-1049. [PMID: 38841852 DOI: 10.1161/circulationaha.123.067861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 05/15/2024] [Indexed: 06/07/2024]
Abstract
BACKGROUND Dilated cardiomyopathy is characterized by left ventricular dilation and continuous systolic dysfunction. Mitochondrial impairment is critical in dilated cardiomyopathy; however, the underlying mechanisms remain unclear. Here, we explored the cardioprotective role of a heart-enriched long noncoding RNA, the dilated cardiomyopathy repressive transcript (DCRT), in maintaining mitochondrial function. METHODS The DCRT knockout (DCRT-/-) mice and DCRT knockout cells were developed using CRISPR-Cas9 technology. Cardiac-specific DCRT transgenic mice were generated using α-myosin heavy chain promoter. Chromatin coimmunoprecipitation, RNA immunoprecipitation, Western blot, and isoform sequencing were performed to investigate the underlying mechanisms. RESULTS We found that the long noncoding RNA DCRT was highly enriched in the normal heart tissues and that its expression was significantly downregulated in the myocardium of patients with dilated cardiomyopathy. DCRT-/- mice spontaneously developed cardiac dysfunction and enlargement with mitochondrial impairment. DCRT transgene or overexpression with the recombinant adeno-associated virus system in mice attenuated cardiac dysfunction induced by transverse aortic constriction treatment. Mechanistically, DCRT inhibited the third exon skipping of NDUFS2 (NADH dehydrogenase ubiquinone iron-sulfur protein 2) by directly binding to PTBP1 (polypyrimidine tract binding protein 1) in the nucleus of cardiomyocytes. Skipping of the third exon of NDUFS2 induced mitochondrial dysfunction by competitively inhibiting mitochondrial complex I activity and binding to PRDX5 (peroxiredoxin 5) and suppressing its antioxidant activity. Furthermore, coenzyme Q10 partially alleviated mitochondrial dysfunction in cardiomyocytes caused by DCRT reduction. CONCLUSIONS Our study revealed that the loss of DCRT contributed to PTBP1-mediated exon skipping of NDUFS2, thereby inducing cardiac mitochondrial dysfunction during dilated cardiomyopathy development, which could be partially treated with coenzyme Q10 supplementation.
Collapse
MESH Headings
- Animals
- Cardiomyopathy, Dilated/genetics
- Cardiomyopathy, Dilated/metabolism
- Cardiomyopathy, Dilated/pathology
- Polypyrimidine Tract-Binding Protein/genetics
- Polypyrimidine Tract-Binding Protein/metabolism
- RNA, Long Noncoding/genetics
- RNA, Long Noncoding/metabolism
- Mice
- Alternative Splicing
- Humans
- Mice, Knockout
- Heterogeneous-Nuclear Ribonucleoproteins/genetics
- Heterogeneous-Nuclear Ribonucleoproteins/metabolism
- Electron Transport Complex I/metabolism
- Electron Transport Complex I/genetics
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Male
- Mitochondria, Heart/metabolism
- Mitochondria, Heart/pathology
- Mitochondria, Heart/genetics
- Mice, Transgenic
Collapse
Affiliation(s)
- Hengzhi Du
- Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (H.D., Y.Z., J.W., B.D., G.H., Y.Z., Z.Y., N.D., H.L., J.F., X.N., F.W., Z.W., D.W.W., C.C.)
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China (H.D., Y.Z., J.W., B.D., G.H., Y.Z., Z.Y., N.D., H.L., J.F., X.N., F.W., Z.W., D.W.W., C.C.)
| | - Yanru Zhao
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China (H.D., Y.Z., J.W., B.D., G.H., Y.Z., Z.Y., N.D., H.L., J.F., X.N., F.W., Z.W., D.W.W., C.C.)
| | - Jianpei Wen
- Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (H.D., Y.Z., J.W., B.D., G.H., Y.Z., Z.Y., N.D., H.L., J.F., X.N., F.W., Z.W., D.W.W., C.C.)
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China (H.D., Y.Z., J.W., B.D., G.H., Y.Z., Z.Y., N.D., H.L., J.F., X.N., F.W., Z.W., D.W.W., C.C.)
| | - Beibei Dai
- Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (H.D., Y.Z., J.W., B.D., G.H., Y.Z., Z.Y., N.D., H.L., J.F., X.N., F.W., Z.W., D.W.W., C.C.)
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China (H.D., Y.Z., J.W., B.D., G.H., Y.Z., Z.Y., N.D., H.L., J.F., X.N., F.W., Z.W., D.W.W., C.C.)
| | - Guo Hu
- Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (H.D., Y.Z., J.W., B.D., G.H., Y.Z., Z.Y., N.D., H.L., J.F., X.N., F.W., Z.W., D.W.W., C.C.)
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China (H.D., Y.Z., J.W., B.D., G.H., Y.Z., Z.Y., N.D., H.L., J.F., X.N., F.W., Z.W., D.W.W., C.C.)
| | - Yufei Zhou
- Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (H.D., Y.Z., J.W., B.D., G.H., Y.Z., Z.Y., N.D., H.L., J.F., X.N., F.W., Z.W., D.W.W., C.C.)
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China (H.D., Y.Z., J.W., B.D., G.H., Y.Z., Z.Y., N.D., H.L., J.F., X.N., F.W., Z.W., D.W.W., C.C.)
| | - Zhongwei Yin
- Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (H.D., Y.Z., J.W., B.D., G.H., Y.Z., Z.Y., N.D., H.L., J.F., X.N., F.W., Z.W., D.W.W., C.C.)
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China (H.D., Y.Z., J.W., B.D., G.H., Y.Z., Z.Y., N.D., H.L., J.F., X.N., F.W., Z.W., D.W.W., C.C.)
| | - Nan Ding
- Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (H.D., Y.Z., J.W., B.D., G.H., Y.Z., Z.Y., N.D., H.L., J.F., X.N., F.W., Z.W., D.W.W., C.C.)
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China (H.D., Y.Z., J.W., B.D., G.H., Y.Z., Z.Y., N.D., H.L., J.F., X.N., F.W., Z.W., D.W.W., C.C.)
| | - Huaping Li
- Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (H.D., Y.Z., J.W., B.D., G.H., Y.Z., Z.Y., N.D., H.L., J.F., X.N., F.W., Z.W., D.W.W., C.C.)
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China (H.D., Y.Z., J.W., B.D., G.H., Y.Z., Z.Y., N.D., H.L., J.F., X.N., F.W., Z.W., D.W.W., C.C.)
| | - Jiahui Fan
- Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (H.D., Y.Z., J.W., B.D., G.H., Y.Z., Z.Y., N.D., H.L., J.F., X.N., F.W., Z.W., D.W.W., C.C.)
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China (H.D., Y.Z., J.W., B.D., G.H., Y.Z., Z.Y., N.D., H.L., J.F., X.N., F.W., Z.W., D.W.W., C.C.)
| | - Xiang Nie
- Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (H.D., Y.Z., J.W., B.D., G.H., Y.Z., Z.Y., N.D., H.L., J.F., X.N., F.W., Z.W., D.W.W., C.C.)
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China (H.D., Y.Z., J.W., B.D., G.H., Y.Z., Z.Y., N.D., H.L., J.F., X.N., F.W., Z.W., D.W.W., C.C.)
| | - Feng Wang
- Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (H.D., Y.Z., J.W., B.D., G.H., Y.Z., Z.Y., N.D., H.L., J.F., X.N., F.W., Z.W., D.W.W., C.C.)
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China (H.D., Y.Z., J.W., B.D., G.H., Y.Z., Z.Y., N.D., H.L., J.F., X.N., F.W., Z.W., D.W.W., C.C.)
| | - Qian Liu
- Tongji Hospital, and Department of Forensic Medicine (Q.L.), Huazhong University of Science and Technology, Wuhan, China
| | - Zheng Wen
- Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (H.D., Y.Z., J.W., B.D., G.H., Y.Z., Z.Y., N.D., H.L., J.F., X.N., F.W., Z.W., D.W.W., C.C.)
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China (H.D., Y.Z., J.W., B.D., G.H., Y.Z., Z.Y., N.D., H.L., J.F., X.N., F.W., Z.W., D.W.W., C.C.)
| | - Gang Xu
- Divisions of Cardiology and Nephrology (G.X.), Huazhong University of Science and Technology, Wuhan, China
| | - Dao Wen Wang
- Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (H.D., Y.Z., J.W., B.D., G.H., Y.Z., Z.Y., N.D., H.L., J.F., X.N., F.W., Z.W., D.W.W., C.C.)
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China (H.D., Y.Z., J.W., B.D., G.H., Y.Z., Z.Y., N.D., H.L., J.F., X.N., F.W., Z.W., D.W.W., C.C.)
| | - Chen Chen
- Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (H.D., Y.Z., J.W., B.D., G.H., Y.Z., Z.Y., N.D., H.L., J.F., X.N., F.W., Z.W., D.W.W., C.C.)
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China (H.D., Y.Z., J.W., B.D., G.H., Y.Z., Z.Y., N.D., H.L., J.F., X.N., F.W., Z.W., D.W.W., C.C.)
| |
Collapse
|
26
|
Iwanski JB, Pappas CT, Mayfield RM, Farman GP, Ahrens-Nicklas R, Churko JM, Gregorio CC. Leiomodin 2 neonatal dilated cardiomyopathy mutation results in altered actin gene signatures and cardiomyocyte dysfunction. NPJ Regen Med 2024; 9:21. [PMID: 39285234 PMCID: PMC11405699 DOI: 10.1038/s41536-024-00366-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 08/23/2024] [Indexed: 09/19/2024] Open
Abstract
Neonatal dilated cardiomyopathy (DCM) is a poorly understood muscular disease of the heart. Several homozygous biallelic variants in LMOD2, the gene encoding the actin-binding protein Leiomodin 2, have been identified to result in severe DCM. Collectively, LMOD2-related cardiomyopathies present with cardiac dilation and decreased heart contractility, often resulting in neonatal death. Thus, it is evident that Lmod2 is essential to normal human cardiac muscle function. This study aimed to understand the underlying pathophysiology and signaling pathways related to the first reported LMOD2 variant (c.1193 G > A, p.Trp398*). Using patient-specific human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) and a mouse model harboring the homologous mutation to the patient, we discovered dysregulated actin-thin filament lengths, altered contractility and calcium handling properties, as well as alterations in the serum response factor (SRF)-dependent signaling pathway. These findings reveal that LMOD2 may be regulating SRF activity in an actin-dependent manner and provide a potential new strategy for the development of biologically active molecules to target LMOD2-related cardiomyopathies.
Collapse
Grants
- R01HL123078 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R00 HL128906 NHLBI NIH HHS
- R01 HL164644 NHLBI NIH HHS
- R01 GM120137 NIGMS NIH HHS
- F30HL151139 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- T32HL007249 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- T32 HL007249 NHLBI NIH HHS
- R01 HL123078 NHLBI NIH HHS
- R01HL164644 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- F30 HL151139 NHLBI NIH HHS
- R01GM120137 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
Collapse
Affiliation(s)
- Jessika B Iwanski
- Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson, AZ, 85724, USA
| | - Christopher T Pappas
- Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson, AZ, 85724, USA
| | - Rachel M Mayfield
- Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson, AZ, 85724, USA
| | - Gerrie P Farman
- Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson, AZ, 85724, USA
| | - Rebecca Ahrens-Nicklas
- Department of Pediatrics and Division of Human Genetics and Metabolism, The Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Jared M Churko
- Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson, AZ, 85724, USA.
| | - Carol C Gregorio
- Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson, AZ, 85724, USA.
- Department of Medicine and Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| |
Collapse
|
27
|
Zeng X, Zeng Q, Wang X, Li K, Wu J, Luo J. Causal association between 1400 metabolites and dilated cardiomyopathy: a bidirectional two-sample Mendelian randomization analysis. Front Endocrinol (Lausanne) 2024; 15:1423142. [PMID: 39329103 PMCID: PMC11424463 DOI: 10.3389/fendo.2024.1423142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 08/26/2024] [Indexed: 09/28/2024] Open
Abstract
Background Dilated cardiomyopathy (DCM) is a cardiac disease with a poor prognosis of unclear etiology. Previous studies have shown that metabolism is associated with DCM. This study investigates the causal relationship between 1400 metabolites and DCM using a two-sample Mendelian randomization (MR) approach. Methods The study utilized data from the OpenGWAS database, comprising 355,381 Europeans, including 1,444 DCM cases. A total of 1,400 metabolites were evaluated for their causal association with DCM. Instrumental variables (IVs) were selected based on genetic variation and used in the MR analysis. The primary analysis method was inverse variance weighting (IVW), supplemented by weighted median-based estimation and sensitivity analyses. Results Of the 1,400 metabolites analyzed, 52 were identified as causally associated with DCM. The analysis revealed both positively and negatively correlated metabolites with DCM risk. Notable findings include the positive correlation of Tryptophan betaine and 5-methyluridine (ribothymidine) levels, and an inverse association of Myristoleate and Erythronate levels with DCM. Conclusions The study provides significant insights into the metabolites potentially involved in the pathogenesis of DCM. These findings could pave the way for new therapeutic strategies and biomarker identification in DCM management.
Collapse
Affiliation(s)
- Xianghui Zeng
- Department of Cardiology, Ganzhou Hospital of Traditional Chinese Medicine, Ganzhou, Jiangxi, China
| | - Qingfeng Zeng
- Department of Cardiology, Ganzhou Hospital of Traditional Chinese Medicine, Ganzhou, Jiangxi, China
| | - Xianggui Wang
- Department of Cardiology, Ganzhou People's Hospital, Ganzhou, Jiangxi, China
| | - Kening Li
- Department of Cardiology, Ganzhou People's Hospital, Ganzhou, Jiangxi, China
| | - Jincheng Wu
- Department of Cardiology, Ganzhou People's Hospital, Ganzhou, Jiangxi, China
| | - Jianping Luo
- Department of Cardiology, Ganzhou People's Hospital, Ganzhou, Jiangxi, China
| |
Collapse
|
28
|
Micolonghi C, Perrone F, Fabiani M, Caroselli S, Savio C, Pizzuti A, Germani A, Visco V, Petrucci S, Rubattu S, Piane M. Unveiling the Spectrum of Minor Genes in Cardiomyopathies: A Narrative Review. Int J Mol Sci 2024; 25:9787. [PMID: 39337275 PMCID: PMC11431948 DOI: 10.3390/ijms25189787] [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: 07/22/2024] [Revised: 09/04/2024] [Accepted: 09/06/2024] [Indexed: 09/30/2024] Open
Abstract
Hereditary cardiomyopathies (CMPs), including arrhythmogenic cardiomyopathy (ACM), dilated cardiomyopathy (DCM), and hypertrophic cardiomyopathy (HCM), represent a group of heart disorders that significantly contribute to cardiovascular morbidity and mortality and are often driven by genetic factors. Recent advances in next-generation sequencing (NGS) technology have enabled the identification of rare variants in both well-established and minor genes associated with CMPs. Nowadays, a set of core genes is included in diagnostic panels for ACM, DCM, and HCM. On the other hand, despite their lesser-known status, variants in the minor genes may contribute to disease mechanisms and influence prognosis. This review evaluates the current evidence supporting the involvement of the minor genes in CMPs, considering their potential pathogenicity and clinical significance. A comprehensive analysis of databases, such as ClinGen, ClinVar, and GeneReviews, along with recent literature and diagnostic guidelines provides a thorough overview of the genetic landscape of minor genes in CMPs and offers guidance in clinical practice, evaluating each case individually based on the clinical referral, and insights for future research. Given the increasing knowledge on these less understood genetic factors, future studies are essential to clearly assess their roles, ultimately leading to improved diagnostic precision and therapeutic strategies in hereditary CMPs.
Collapse
Affiliation(s)
- Caterina Micolonghi
- Department of Experimental Medicine, Faculty of Medicine and Dentistry, Sapienza University of Rome, 00161 Rome, Italy
| | - Federica Perrone
- Department of Experimental Medicine, Faculty of Medicine and Dentistry, Sapienza University of Rome, 00161 Rome, Italy
- Department of Neuroscience, Istituto Superiore di Sanità, 00161 Rome, Italy
| | - Marco Fabiani
- Department of Experimental Medicine, Faculty of Medicine and Dentistry, Sapienza University of Rome, 00161 Rome, Italy
- ALTAMEDICA, Human Genetics, 00198 Rome, Italy
| | - Silvia Caroselli
- Department of Experimental Medicine, Faculty of Medicine and Dentistry, Sapienza University of Rome, 00161 Rome, Italy
- Juno Genetics, Reproductive Genetics, 00188 Rome, Italy
| | | | - Antonio Pizzuti
- Department of Experimental Medicine, Faculty of Medicine and Dentistry, Sapienza University of Rome, 00161 Rome, Italy
- Medical Genetics Unit, IRCCS Mendel Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy
| | - Aldo Germani
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Psychology, Sapienza University of Rome, 00189 Rome, Italy
| | - Vincenzo Visco
- S. Andrea University Hospital, 00189 Rome, Italy
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Psychology, Sapienza University of Rome, 00189 Rome, Italy
| | - Simona Petrucci
- S. Andrea University Hospital, 00189 Rome, Italy
- Medical Genetics Unit, IRCCS Mendel Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Psychology, Sapienza University of Rome, 00189 Rome, Italy
| | - Speranza Rubattu
- S. Andrea University Hospital, 00189 Rome, Italy
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Psychology, Sapienza University of Rome, 00189 Rome, Italy
- IRCCS Neuromed, 86077 Pozzilli, Italy
| | - Maria Piane
- S. Andrea University Hospital, 00189 Rome, Italy
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Psychology, Sapienza University of Rome, 00189 Rome, Italy
| |
Collapse
|
29
|
Kim OH, Kim J, Kim Y, Lee S, Lee BH, Kim BJ, Park HY, Park MH. Exploring novel MYH7 gene variants using in silico analyses in Korean patients with cardiomyopathy. BMC Med Genomics 2024; 17:225. [PMID: 39237976 PMCID: PMC11378590 DOI: 10.1186/s12920-024-02000-8] [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: 02/23/2024] [Accepted: 08/29/2024] [Indexed: 09/07/2024] Open
Abstract
BACKGROUND Pathogenic variants of MYH7, which encodes the beta-myosin heavy chain protein, are major causes of dilated and hypertrophic cardiomyopathy. METHODS In this study, we used whole-genome sequencing data to identify MYH7 variants in 397 patients with various cardiomyopathy subtypes who were participating in the National Project of Bio Big Data pilot study in Korea. We also performed in silico analyses to predict the pathogenicity of the novel variants, comparing them to known pathogenic missense variants. RESULTS We identified 27 MYH7 variants in 41 unrelated patients with cardiomyopathy, consisting of 20 previously known pathogenic/likely pathogenic variants, 2 variants of uncertain significance, and 5 novel variants. Notably, the pathogenic variants predominantly clustered within the myosin motor domain of MYH7. We confirmed that the novel identified variants could be pathogenic, as indicated by high prediction scores in the in silico analyses, including SIFT, Mutation Assessor, PROVEAN, PolyPhen-2, CADD, REVEL, MetaLR, MetaRNN, and MetaSVM. Furthermore, we assessed their damaging effects on protein dynamics and stability using DynaMut2 and Missense3D tools. CONCLUSIONS Overall, our study identified the distribution of MYH7 variants among patients with cardiomyopathy in Korea, offering new insights for improved diagnosis by enriching the data on the pathogenicity of novel variants using in silico tools and evaluating the function and structural stability of the MYH7 protein.
Collapse
Affiliation(s)
- Oc-Hee Kim
- Division of Genome Science, Department of Precision Medicine, National Institute of Health, Cheongju, 28159, Republic of Korea
| | - Jihyun Kim
- Division of Genome Science, Department of Precision Medicine, National Institute of Health, Cheongju, 28159, Republic of Korea
| | - Youngjun Kim
- Division of Genome Science, Department of Precision Medicine, National Institute of Health, Cheongju, 28159, Republic of Korea
| | - Soyoung Lee
- Department of Pediatrics, Hallym University Sacred Heart Hospital, Anyang, 14068, Republic of Korea
| | - Beom Hee Lee
- Medical Genetics Center, Asan Medical Center Children's Hospital, University of Ulsan College of Medicines, Seoul, 05505, Republic of Korea
| | - Bong-Jo Kim
- Division of Genome Science, Department of Precision Medicine, National Institute of Health, Cheongju, 28159, Republic of Korea
| | - Hyun-Young Park
- National Institute of Health, Cheongju, 28159, Republic of Korea
| | - Mi-Hyun Park
- Division of Genome Science, Department of Precision Medicine, National Institute of Health, Cheongju, 28159, Republic of Korea.
| |
Collapse
|
30
|
Zhang X, Burattini M, Duru J, Chala N, Wyssen N, Cofiño-Fabres C, Rivera-Arbeláez JM, Passier R, Poulikakos D, Ferrari A, Tringides C, Vörös J, Luciani GB, Miragoli M, Zambelli T. Multimodal Mapping of Electrical and Mechanical Latency of Human-Induced Pluripotent Stem Cell-Derived Cardiomyocyte Layers. ACS NANO 2024; 18:24060-24075. [PMID: 39172696 DOI: 10.1021/acsnano.4c03896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2024]
Abstract
The synchronization of the electrical and mechanical coupling assures the physiological pump function of the heart, but life-threatening pathologies may jeopardize this equilibrium. Recently, human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) have emerged as a model for personalized investigation because they can recapitulate human diseased traits, such as compromised electrical capacity or mechanical circuit disruption. This research avails the model of hiPSC-CMs and showcases innovative techniques to study the electrical and mechanical properties as well as their modulation due to inherited cardiomyopathies. In this work, hiPSC-CMs carrying either Brugada syndrome (BRU) or dilated cardiomyopathy (DCM), were organized in a bilayer configuration to first validate the experimental methods and second mimic the physiological environment. High-density CMOS-based microelectrode arrays (HD-MEA) have been employed to study the electrical activity. Furthermore, mechanical function was investigated via quantitative video-based evaluation, upon stimulation with a β-adrenergic agonist. This study introduces two experimental methods. First, high-throughput mechanical measurements in the hiPSC-CM layers (xy-inspection) are obtained using both a recently developed optical tracker (OPT) and confocal reference-free traction force microscopy (cTFM) aimed to quantify cardiac kinematics. Second, atomic force microscopy (AFM) with FluidFM probes, combined with the xy-inspection methods, supplemented a three-dimensional understanding of cell-cell mechanical coupling (xyz-inspection). This particular combination represents a multi-technique approach to detecting electrical and mechanical latency among the cell layers, examining differences and possible implications following inherited cardiomyopathies. It can not only detect disease characteristics in the proposed in vitro model but also quantitatively assess its response to drugs, thereby demonstrating its feasibility as a scalable tool for clinical and pharmacological studies.
Collapse
Affiliation(s)
- Xinyu Zhang
- Laboratory of Biosensors and Bioelectronics, Institute for Biomedical Engineering, ETH Zürich, 8092 Zürich,Switzerland
| | - Margherita Burattini
- Laboratory of Experimental and Applied Medical Technologies, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
- Department of Maternity, Surgery and Dentistry, University of Verona, 37134 Verona, Italy
| | - Jens Duru
- Laboratory of Biosensors and Bioelectronics, Institute for Biomedical Engineering, ETH Zürich, 8092 Zürich,Switzerland
| | - Nafsika Chala
- Laboratory of Thermodynamics in Emerging Technologies, Department of Mechanical and Process Engineering, ETH Zürich, 8092 Zurich,Switzerland
| | - Nino Wyssen
- Laboratory of Biosensors and Bioelectronics, Institute for Biomedical Engineering, ETH Zürich, 8092 Zürich,Switzerland
| | - Carla Cofiño-Fabres
- Department of Applied Stem Cell Technologies, TechMed Centre, University of Twente, 7500 AE Enschede, The Netherland
| | - José Manuel Rivera-Arbeláez
- Department of Applied Stem Cell Technologies, TechMed Centre, University of Twente, 7500 AE Enschede, The Netherland
| | - Robert Passier
- Department of Applied Stem Cell Technologies, TechMed Centre, University of Twente, 7500 AE Enschede, The Netherland
| | - Dimos Poulikakos
- Laboratory of Thermodynamics in Emerging Technologies, Department of Mechanical and Process Engineering, ETH Zürich, 8092 Zurich,Switzerland
| | - Aldo Ferrari
- Laboratory of Thermodynamics in Emerging Technologies, Department of Mechanical and Process Engineering, ETH Zürich, 8092 Zurich,Switzerland
- Experimental Continuum Mechanics, EMPA, Swiss Federal Laboratories for Material Science and Technologies, 8600 Dübendorf, Switzerland
| | - Christina Tringides
- Laboratory of Biosensors and Bioelectronics, Institute for Biomedical Engineering, ETH Zürich, 8092 Zürich,Switzerland
| | - János Vörös
- Laboratory of Biosensors and Bioelectronics, Institute for Biomedical Engineering, ETH Zürich, 8092 Zürich,Switzerland
| | | | - Michele Miragoli
- Laboratory of Experimental and Applied Medical Technologies, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
- Humanitas Research Hospital ─ IRCCS, 20089 Rozzano, Italy
| | - Tomaso Zambelli
- Laboratory of Biosensors and Bioelectronics, Institute for Biomedical Engineering, ETH Zürich, 8092 Zürich,Switzerland
| |
Collapse
|
31
|
Belcher AM, Annie FH, Rinehart S, Elashery A, Amer M. Severe familial dilated cardiomyopathy in a young adult due to a rare LMNA mutation: a case report. Eur Heart J Case Rep 2024; 8:ytae423. [PMID: 39239135 PMCID: PMC11375576 DOI: 10.1093/ehjcr/ytae423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 05/01/2024] [Accepted: 08/07/2024] [Indexed: 09/07/2024]
Abstract
Background Familial dilated cardiomyopathy prognosis and disease progression vary greatly depending upon the type of genetic mutation. Family history and genetic testing are paramount in developing the best treatment plan for a patient. However, with rare or novel mutations, the significance may be unknown. Regarding this, the following case report highlights the importance of vigilance and suspicion when treating a patient with a variant of unknown significance. Additionally, it shows the importance of thoroughly investigating the family history of cardiovascular disease. Case summary A 25-year-old Caucasian male was found to have a right bundle branch block and dilated cardiomyopathy upon presentation to the emergency department. Later testing showed that the dilated cardiomyopathy was due to an incredibly rare lamin A/C (LMNA) gene mutation, R349L. Despite treatment with a maximum-tolerable medication regimen and an automatic implantable cardioverter-defibrillator, the patient continued to decline and required a heart transplant. Discussion This case provides more information on the severity of this specific LMNA mutation that has only been documented once before. Of note, the time from the initial emergency department visit to the heart transplant was approximately 2 years. Given the patient's young age and rapid disease progression, in addition to a strong family history of sudden cardiac death, the significance of this mutation should not be understated. The additional knowledge gained from this case report can be used to aid in timely interventions and prognosis evaluation.
Collapse
Affiliation(s)
- Adam M Belcher
- CAMC Institute for Academic Medicine, Charleston Area Medical Center, 3044 Chesterfield Ave, Charleston, WV 25304, USA
| | - Frank H Annie
- CAMC Institute for Academic Medicine, Charleston Area Medical Center, 3044 Chesterfield Ave, Charleston, WV 25304, USA
| | - Sarah Rinehart
- CAMC Department of Cardiology, Charleston Area Medical Center, 3200 MacCorkle Ave SE, Charleston, WV 25304, USA
| | - Ahmad Elashery
- CAMC Department of Cardiology, Charleston Area Medical Center, 3200 MacCorkle Ave SE, Charleston, WV 25304, USA
| | - Muhammad Amer
- CAMC Department of Cardiology, Charleston Area Medical Center, 3200 MacCorkle Ave SE, Charleston, WV 25304, USA
| |
Collapse
|
32
|
Serpa F, Finn CM, Tahir UA. Navigating the penetrance and phenotypic spectrum of inherited cardiomyopathies. Heart Fail Rev 2024; 29:873-881. [PMID: 38898187 DOI: 10.1007/s10741-024-10405-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/08/2024] [Indexed: 06/21/2024]
Abstract
Inherited cardiomyopathies are genetic diseases that can lead to heart failure and sudden cardiac death. These conditions tend to run in families, following an autosomal dominant pattern where first-degree relatives have a 50% chance of carrying the pathogenic variant. Despite significant advancements and increased accessibility of genetic testing, accurately predicting the phenotypic expression of these conditions remains challenging due to the inherent variability in their clinical manifestations and the incomplete penetrance observed. This poses challenges in providing patient care and effectively communicating the potential risk of future disease to patients and their families. To address these challenges, this review aims to synthesize the available evidence on penetrance, expressivity, and factors influencing disease expression to improve communication and risk assessment for patients with inherited cardiomyopathies and their family members.
Collapse
Affiliation(s)
- Frans Serpa
- Division of Cardiology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Caitlin M Finn
- Division of Cardiology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Usman A Tahir
- Division of Cardiology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
33
|
Creso JG, Gokhan I, Rynkiewicz MJ, Lehman W, Moore JR, Campbell SG. In silico and in vitro models reveal the molecular mechanisms of hypocontractility caused by TPM1 M8R. Front Physiol 2024; 15:1452509. [PMID: 39282088 PMCID: PMC11392859 DOI: 10.3389/fphys.2024.1452509] [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: 06/21/2024] [Accepted: 08/19/2024] [Indexed: 09/18/2024] Open
Abstract
Dilated cardiomyopathy (DCM) is an inherited disorder often leading to severe heart failure. Linkage studies in affected families have revealed hundreds of different mutations that can cause DCM, with most occurring in genes associated with the cardiac sarcomere. We have developed an investigational pipeline for discovering mechanistic genotype-phenotype relationships in DCM and here apply it to the DCM-linked tropomyosin mutation TPM1 M8R. Atomistic simulations predict that M8R increases flexibility of the tropomyosin chain and enhances affinity for the blocked or inactive state of tropomyosin on actin. Applying these molecular effects to a Markov model of the cardiac thin filament reproduced the shifts in Ca2+sensitivity, maximum force, and a qualitative drop in cooperativity that were observed in an in vitro system containing TPM1 M8R. The model was then used to simulate the impact of M8R expression on twitch contractions of intact cardiac muscle, predicting that M8R would reduce peak force and duration of contraction in a dose-dependent manner. To evaluate this prediction, TPM1 M8R was expressed via adenovirus in human engineered heart tissues and isometric twitch force was observed. The mutant tissues manifested depressed contractility and twitch duration that agreed in detail with model predictions. Additional exploratory simulations suggest that M8R-mediated alterations in tropomyosin-actin interactions contribute more potently than tropomyosin chain stiffness to cardiac twitch dysfunction, and presumably to the ultimate manifestation of DCM. This study is an example of the growing potential for successful in silico prediction of mutation pathogenicity for inherited cardiac muscle disorders.
Collapse
Affiliation(s)
- Jenette G. Creso
- Department of Biomedical Engineering, Yale University, New Haven, CT, United States
| | - Ilhan Gokhan
- Department of Biomedical Engineering, Yale University, New Haven, CT, United States
| | - Michael J. Rynkiewicz
- Department of Pharmacology, Physiology and Biophysics, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - William Lehman
- Department of Pharmacology, Physiology and Biophysics, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Jeffrey R. Moore
- Department of Biological Sciences, University of Massachusetts–Lowell, Lowell, MA, United States
| | - Stuart G. Campbell
- Department of Biomedical Engineering, Yale University, New Haven, CT, United States
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT, United States
| |
Collapse
|
34
|
Ma Q, Zhang YH, Guo W, Feng K, Huang T, Cai YD. Machine Learning in Identifying Marker Genes for Congenital Heart Diseases of Different Cardiac Cell Types. Life (Basel) 2024; 14:1032. [PMID: 39202774 PMCID: PMC11355424 DOI: 10.3390/life14081032] [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: 05/13/2024] [Revised: 07/31/2024] [Accepted: 08/14/2024] [Indexed: 09/03/2024] Open
Abstract
Congenital heart disease (CHD) represents a spectrum of inborn heart defects influenced by genetic and environmental factors. This study advances the field by analyzing gene expression profiles in 21,034 cardiac fibroblasts, 73,296 cardiomyocytes, and 35,673 endothelial cells, utilizing single-cell level analysis and machine learning techniques. Six CHD conditions: dilated cardiomyopathy (DCM), donor hearts (used as healthy controls), hypertrophic cardiomyopathy (HCM), heart failure with hypoplastic left heart syndrome (HF_HLHS), Neonatal Hypoplastic Left Heart Syndrome (Neo_HLHS), and Tetralogy of Fallot (TOF), were investigated for each cardiac cell type. Each cell sample was represented by 29,266 gene features. These features were first analyzed by six feature-ranking algorithms, resulting in several feature lists. Then, these lists were fed into incremental feature selection, containing two classification algorithms, to extract essential gene features and classification rules and build efficient classifiers. The identified essential genes can be potential CHD markers in different cardiac cell types. For instance, the LASSO identified key genes specific to various heart cell types in CHD subtypes. FOXO3 was found to be up-regulated in cardiac fibroblasts for both Dilated and hypertrophic cardiomyopathy. In cardiomyocytes, distinct genes such as TMTC1, ART3, ARHGAP24, SHROOM3, and XIST were linked to dilated cardiomyopathy, Neo-Hypoplastic Left Heart Syndrome, hypertrophic cardiomyopathy, HF-Hypoplastic Left Heart Syndrome, and Tetralogy of Fallot, respectively. Endothelial cell analysis further revealed COL25A1, NFIB, and KLF7 as significant genes for dilated cardiomyopathy, hypertrophic cardiomyopathy, and Tetralogy of Fallot. LightGBM, Catboost, MCFS, RF, and XGBoost further delineated key genes for specific CHD subtypes, demonstrating the efficacy of machine learning in identifying CHD-specific genes. Additionally, this study developed quantitative rules for representing the gene expression patterns related to CHDs. This research underscores the potential of machine learning in unraveling the molecular complexities of CHD and establishes a foundation for future mechanism-based studies.
Collapse
Affiliation(s)
- Qinglan Ma
- School of Life Sciences, Shanghai University, Shanghai 200444, China;
| | - Yu-Hang Zhang
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA;
| | - Wei Guo
- Key Laboratory of Stem Cell Biology, Shanghai Jiao Tong University School of Medicine (SJTUSM) & Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai 200030, China;
| | - Kaiyan Feng
- Department of Computer Science, Guangdong AIB Polytechnic College, Guangzhou 510507, China;
| | - Tao Huang
- Bio-Med Big Data Center, CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yu-Dong Cai
- School of Life Sciences, Shanghai University, Shanghai 200444, China;
| |
Collapse
|
35
|
Rath P, Jena B, Behera SS, Sathapathy S, Rath PK, Sarangi S. Comparative study of echocardiographic parameters in healthy and dilated cardiomyopathy-affected dogs. VETERINARY RESEARCH FORUM : AN INTERNATIONAL QUARTERLY JOURNAL 2024; 15:403-409. [PMID: 39280861 PMCID: PMC11401138 DOI: 10.30466/vrf.2023.2018338.4108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 08/15/2024] [Indexed: 09/18/2024]
Abstract
Echocardiography is a non-invasive and gold standard imaging tool for diagnosing dilated cardiomyopathy (DCM) in dogs. This study aimed to compare the echocardiographic parameters between healthy and DCM-affected dogs. A total of 52 client-owned dogs, comprising 38 males and 14 females, were included. Among these, 24 dogs (46.15%) were classified as healthy controls and 28 dogs (53.85%) were part of DCM group. On breed-wise prevalence, it was reported that Labrador Retriever breeds showed a higher incidence of DCM than the others. The comparative studies of echocardiographic parameters showed that DCM-affected dogs had significantly higher values in left ventricular long axis length at -end diastole (LVLdA4C) and -end systole (LVLsA4C), end diastolic volume (EDV), end systolic volume (ESV), left atrium (LA)/aorta diameter (Ao) ratio, left ventricular internal dimension at systole (LVIDs), and end point septal separation (EPSS), as well as significantly lower values in left ventricular contractibility indices such as fractional shortening (FS) and ejection fraction (EF) compared to healthy dogs. Also, receiver operating characteristic curves were made to determine the optimal cut-off points for each echocardiographic parameter with specificity and sensitivity for diagnosing DCM. Significant areas under the curve were observed for parameters such as LVIDs, EF, FS, LA/Ao, EPSS, LVLdA4C, LVLsA4C, left ventricular EDV, left ventricular ESV, and ESV for DCM-affected dogs. This cut-off value can be used as an early diagnosis of DCM through echocardiography, facilitating timely clinical interventions and management strategies for improved quality of life in dogs.
Collapse
Affiliation(s)
- Pritish Rath
- Department of Veterinary Surgery and Radiology, Faculty of Veterinary and Animal Sciences, West Bengal University of Animal and Fishery Sciences, Kolkata, India
| | - Biswadeep Jena
- Department of Veterinary Surgery and Radiology, College of Veterinary Science and Animal Husbandry, Odisha University of Agriculture and Technology, Bhubaneswar, India
| | - Sidhartha Sankar Behera
- Department of Veterinary Surgery and Radiology, College of Veterinary Science and Animal Husbandry, Odisha University of Agriculture and Technology, Bhubaneswar, India
| | - Srinivas Sathapathy
- Department of Veterinary Anatomy and Histology, College of Veterinary Science and Animal Husbandry, Odisha University of Agriculture and Technology, Bhubaneswar, India
| | - Prasana Kumar Rath
- Department of Veterinary Pathology, College of Veterinary Science and Animal Husbandry, Odisha University of Agriculture and Technology, Bhubaneswar, India
| | - Swetapadma Sarangi
- Department of Veterinary Preventive Medicine, Madras Veterinary College, Tamil Nadu Veterinary and Animal Sciences University, Chennai, India
| |
Collapse
|
36
|
Wu XY, Lee YK, Lau YM, Au KW, Tse YL, Ng KM, Wong CK, Tse HF. The Pathogenic Mechanisms of and Novel Therapies for Lamin A/C-Related Dilated Cardiomyopathy Based on Patient-Specific Pluripotent Stem Cell Platforms and Animal Models. Pharmaceuticals (Basel) 2024; 17:1030. [PMID: 39204134 PMCID: PMC11357512 DOI: 10.3390/ph17081030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 07/12/2024] [Accepted: 07/16/2024] [Indexed: 09/03/2024] Open
Abstract
Variants (pathogenic) of the LMNA gene are a common cause of familial dilated cardiomyopathy (DCM), which is characterised by early-onset atrioventricular (AV) block, atrial fibrillation and ventricular tachyarrhythmias (VTs), and progressive heart failure. The unstable internal nuclear lamina observed in LMNA-related DCM is a consequence of the disassembly of lamins A and C. This suggests that LMNA variants produce truncated or alternative forms of protein that alter the nuclear structure and the signalling pathway related to cardiac muscle diseases. To date, the pathogenic mechanisms and phenotypes of LMNA-related DCM have been studied using different platforms, such as patient-specific induced pluripotent stem-cell-derived cardiomyocytes (iPSC-CMs) and transgenic mice. In this review, point variants in the LMNA gene that cause autosomal dominantly inherited forms of LMNA-related DCM are summarised. In addition, potential therapeutic targets based on preclinical studies of LMNA variants using transgenic mice and human iPSC-CMs are discussed. They include mitochondria deficiency, variants in nuclear deformation, chromatin remodelling, altered platelet-derived growth factor and ERK1/2-related pathways, and abnormal calcium handling.
Collapse
Affiliation(s)
- Xin-Yi Wu
- Cardiology Division, Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China; (X.-Y.W.); (Y.-K.L.); (Y.-M.L.); (K.-W.A.); (Y.-L.T.); (K.-M.N.); (C.-K.W.)
| | - Yee-Ki Lee
- Cardiology Division, Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China; (X.-Y.W.); (Y.-K.L.); (Y.-M.L.); (K.-W.A.); (Y.-L.T.); (K.-M.N.); (C.-K.W.)
| | - Yee-Man Lau
- Cardiology Division, Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China; (X.-Y.W.); (Y.-K.L.); (Y.-M.L.); (K.-W.A.); (Y.-L.T.); (K.-M.N.); (C.-K.W.)
| | - Ka-Wing Au
- Cardiology Division, Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China; (X.-Y.W.); (Y.-K.L.); (Y.-M.L.); (K.-W.A.); (Y.-L.T.); (K.-M.N.); (C.-K.W.)
| | - Yiu-Lam Tse
- Cardiology Division, Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China; (X.-Y.W.); (Y.-K.L.); (Y.-M.L.); (K.-W.A.); (Y.-L.T.); (K.-M.N.); (C.-K.W.)
| | - Kwong-Man Ng
- Cardiology Division, Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China; (X.-Y.W.); (Y.-K.L.); (Y.-M.L.); (K.-W.A.); (Y.-L.T.); (K.-M.N.); (C.-K.W.)
- Centre for Stem Cell Translational Biology, Hong Kong SAR, China
| | - Chun-Ka Wong
- Cardiology Division, Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China; (X.-Y.W.); (Y.-K.L.); (Y.-M.L.); (K.-W.A.); (Y.-L.T.); (K.-M.N.); (C.-K.W.)
| | - Hung-Fat Tse
- Cardiology Division, Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China; (X.-Y.W.); (Y.-K.L.); (Y.-M.L.); (K.-W.A.); (Y.-L.T.); (K.-M.N.); (C.-K.W.)
- Centre for Stem Cell Translational Biology, Hong Kong SAR, China
- Cardiac and Vascular Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518053, China
- Hong Kong-Guangdong Stem Cell and Regenerative Medicine Research Centre, The University of Hong Kong and Guangzhou Institutes of Biomedicine and Health, Hong Kong SAR, China
- Advanced Biomedical Instrumentation Centre, Hong Kong SAR, China
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong SAR, China
| |
Collapse
|
37
|
Myers MC, Wang S, Zhong Y, Maruyama S, Bueno C, Bastien A, Fazeli MS, Golchin N. Prevalence of Genetically Associated Dilated Cardiomyopathy: A Systematic Literature Review and Meta-Analysis. Cardiol Res 2024; 15:233-245. [PMID: 39205965 PMCID: PMC11349141 DOI: 10.14740/cr1680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 07/30/2024] [Indexed: 09/04/2024] Open
Abstract
Background Dilated cardiomyopathy (DCM) is a leading cause of heart failure and cardiac transplantation globally. Disease-associated genetic variants play a significant role in the development of DCM. Accurately determining the prevalence of genetically associated DCM (genetic DCM) is important for developing targeted prevention strategies. This review synthesized published literature on the global prevalence of genetic DCM across various populations, focusing on two of the most common variants: titin (TTN) and myosin heavy chain 7 (MYH7). Methods MEDLINE® and Embase were searched from database inception to September 19, 2022 for English-language studies reporting the prevalence of genetic DCM within any population. Studies using family history as a proxy for genetic DCM were excluded. Results Of 2,736 abstracts, 57 studies were included. Among the global adult or mixed (mostly adults with few pediatric patients) DCM population, median prevalence was 20.2% (interquartile range (IQR): 16.3-36.0%) for overall genetic DCM, 11.4% (IQR: 8.2-17.8%) for TTN-associated DCM, and 3.2% (IQR: 1.8-5.2%) for MYH7-associated DCM. Global prevalence of overall pediatric genetic DCM within the DCM population was similar (weighted mean: 21.3%). Few studies reported data on the prevalence of genetic DCM within the general population. Conclusions Our study identified variable prevalence estimates of genetic DCM across different populations and geographic locations. The current evidence may underestimate the genetic contributions due to limited screening and detection of potential DCM patients. Epidemiological studies using long-read whole genome sequencing to identify structural variants or non-coding variants are needed, as well as large cohort datasets with genotype-phenotype correlation analyses.
Collapse
Affiliation(s)
| | - Su Wang
- Evidinno Outcomes Research Inc., Vancouver, BC, Canada
| | - Yue Zhong
- Bristol Myers Squibb, Princeton, NJ, USA
| | | | | | | | | | | |
Collapse
|
38
|
Chao T, Ge Y, Sun J, Wang C. Research landscape of genetics in dilated cardiomyopathy: insight from a bibliometric analysis. Front Cardiovasc Med 2024; 11:1362551. [PMID: 39070560 PMCID: PMC11272475 DOI: 10.3389/fcvm.2024.1362551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 06/27/2024] [Indexed: 07/30/2024] Open
Abstract
Background Dilated cardiomyopathy (DCM) is a heterogeneous myocardial disorder with diverse genetic or acquired origins. Notable advances have been achieved in discovering and understanding the genetics of DCM. This study aimed to depict the distribution of the main research forces, hotspots, and frontiers in the genetics of DCM, thus shaping future research directions. Methods Based on the documents published in the Web of Science Core Collection database from 2013 to 2022, co-authorship of authors, institutions, and countries/regions, co-citation of references, and co-occurrence of keywords were conducted respectively to present the distribution of the leading research forces, research hotspots, and emerging trends in the genetics of DCM. Results 4,141 documents were included, and the annual publications have steadily increased. Seidman, Christine E, Meder, Benjamin, Sinagra, Gianfranco were the most productive authors, German Centre for Cardiovascular Research was the most productive institution, and the USA, China, and Germany were the most prolific countries. The co-occurrence of keywords has generated 8 clusters, including DCM, lamin a/c, heart failure, sudden cardiac death, hypertrophic cardiomyopathy, cardiac hypertrophy, arrhythmogenic cardiomyopathy, and next-generation sequencing. Frequent keywords with average publication time after 2019 mainly included arrhythmogenic cardiomyopathy, whole-exome sequencing, RBM 20, phenotype, risk stratification, precision medicine, genotype, and machine learning. Conclusion The research landscape of genetics in DCM is continuously evolving. Deciphering the genetic profiles by next-generation sequencing and illustrating pathogenic mechanisms of gene variants, establishing innovative treatments for heart failure and improved risk stratification for SCD, uncovering the genetic overlaps between DCM and other inherited cardiomyopathies, as well as identifying genotype-phenotype correlations are the main research hotspots and frontiers in this field.
Collapse
Affiliation(s)
- Tiantian Chao
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yaru Ge
- Community Medical Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Jinghui Sun
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Chenglong Wang
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
39
|
Martin TG, Leinwand LA. Hearts apart: sex differences in cardiac remodeling in health and disease. J Clin Invest 2024; 134:e180074. [PMID: 38949027 PMCID: PMC11213513 DOI: 10.1172/jci180074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/02/2024] Open
Abstract
Biological sex is an important modifier of physiology and influences pathobiology in many diseases. While heart disease is the number one cause of death worldwide in both men and women, sex differences exist at the organ and cellular scales, affecting clinical presentation, diagnosis, and treatment. In this Review, we highlight baseline sex differences in cardiac structure, function, and cellular signaling and discuss the contribution of sex hormones and chromosomes to these characteristics. The heart is a remarkably plastic organ and rapidly responds to physiological and pathological cues by modifying form and function. The nature and extent of cardiac remodeling in response to these stimuli are often dependent on biological sex. We discuss organ- and molecular-level sex differences in adaptive physiological remodeling and pathological cardiac remodeling from pressure and volume overload, ischemia, and genetic heart disease. Finally, we offer a perspective on key future directions for research into cardiac sex differences.
Collapse
Affiliation(s)
- Thomas G. Martin
- Department of Molecular, Cellular, and Developmental Biology and
- BioFrontiers Institute, University of Colorado Boulder, Boulder, Colorado, USA
| | - Leslie A. Leinwand
- Department of Molecular, Cellular, and Developmental Biology and
- BioFrontiers Institute, University of Colorado Boulder, Boulder, Colorado, USA
| |
Collapse
|
40
|
Goanta EV, Vacarescu C, Tartea G, Ungureanu A, Militaru S, Muraretu A, Faur-Grigori AA, Petrescu L, Vătăsescu R, Cozma D. Unexpected Genetic Twists in Patients with Cardiac Devices. J Clin Med 2024; 13:3801. [PMID: 38999368 PMCID: PMC11242405 DOI: 10.3390/jcm13133801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 06/12/2024] [Accepted: 06/26/2024] [Indexed: 07/14/2024] Open
Abstract
Objective: To assess the frequency and types of genetic mutations in patients with arrhythmias who underwent cardiac device implantation. Methods: Retrospective observational study, including 38 patients with different arrhythmias and cardiac arrest as a first cardiac event. Treatment modalities encompass pacemakers, transvenous defibrillators, loop recorders, subcutaneous defibrillators, and cardiac resynchronization therapy. All patients underwent genetic testing, using commercially available panels (106-174 genes). Outcome measures include mortality, arrhythmia recurrence, and device-related complications. Results: Clinical parameters revealed a family history of sudden cardiac death in 19 patients (50%), who were predominantly male (58%) and had a mean age of 44.5 years and a mean left ventricle ejection fraction of 40.3%. Genetic testing identified mutations in various genes, predominantly TMEM43 (11%). In two patients (3%) with arrhythmogenic cardiomyopathy, complete subcutaneous defibrillator extraction with de novo transvenous implantable cardioverter-defibrillator implantation was needed. The absence of multiple associations among severe gene mutations was crucial for cardiac resynchronization therapy response. Mortality in this group was around 3% in titin dilated cardiomyopathy patients. Conclusions: Integration of genetic testing into the decision-making process for patients with electronic devices represents a paradigm shift in personalized medicine. By identifying genetic markers associated with arrhythmia susceptibility, heart failure etiology, and cardiac resynchronization therapy response, clinicians can tailor device choices to optimize patient outcomes.
Collapse
Affiliation(s)
- Emilia-Violeta Goanta
- Doctoral School, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania;
- Cardiology Department, Emergency County Hospital of Craiova, Tabaci Street, Nr. 1, 200642 Craiova, Romania; (G.T.); (A.U.); (A.M.)
| | - Cristina Vacarescu
- Department of Cardiology, “Victor Babes” University of Medicine and Pharmacy, 2 Eftimie Murgu Square, 300041 Timisoara, Romania; (L.P.); (D.C.)
- Institute of Cardiovascular Diseases Timisoara, 13A Gheorghe Adam Street, 300310 Timisoara, Romania;
- Research Center of the Institute of Cardiovascular Diseases Timisoara, 13A Gheorghe Adam Street, 300310 Timisoara, Romania
| | - Georgica Tartea
- Cardiology Department, Emergency County Hospital of Craiova, Tabaci Street, Nr. 1, 200642 Craiova, Romania; (G.T.); (A.U.); (A.M.)
- Department of Physiology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Adrian Ungureanu
- Cardiology Department, Emergency County Hospital of Craiova, Tabaci Street, Nr. 1, 200642 Craiova, Romania; (G.T.); (A.U.); (A.M.)
| | - Sebastian Militaru
- Department of Cardiology, Craiova University of Medicine and Pharmacy, 200349 Craiova, Romania;
| | - Alexandra Muraretu
- Cardiology Department, Emergency County Hospital of Craiova, Tabaci Street, Nr. 1, 200642 Craiova, Romania; (G.T.); (A.U.); (A.M.)
| | | | - Lucian Petrescu
- Department of Cardiology, “Victor Babes” University of Medicine and Pharmacy, 2 Eftimie Murgu Square, 300041 Timisoara, Romania; (L.P.); (D.C.)
| | - Radu Vătăsescu
- Cardiology Department, Clinical Emergency Hospital, 014461 Bucharest, Romania;
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Dragos Cozma
- Department of Cardiology, “Victor Babes” University of Medicine and Pharmacy, 2 Eftimie Murgu Square, 300041 Timisoara, Romania; (L.P.); (D.C.)
- Institute of Cardiovascular Diseases Timisoara, 13A Gheorghe Adam Street, 300310 Timisoara, Romania;
- Research Center of the Institute of Cardiovascular Diseases Timisoara, 13A Gheorghe Adam Street, 300310 Timisoara, Romania
| |
Collapse
|
41
|
Wali R, Xu H, Cheruiyot C, Saleem HN, Janshoff A, Habeck M, Ebert A. Integrated machine learning and multimodal data fusion for patho-phenotypic feature recognition in iPSC models of dilated cardiomyopathy. Biol Chem 2024; 405:427-439. [PMID: 38651266 DOI: 10.1515/hsz-2024-0023] [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: 02/07/2024] [Accepted: 03/27/2024] [Indexed: 04/25/2024]
Abstract
Integration of multiple data sources presents a challenge for accurate prediction of molecular patho-phenotypic features in automated analysis of data from human model systems. Here, we applied a machine learning-based data integration to distinguish patho-phenotypic features at the subcellular level for dilated cardiomyopathy (DCM). We employed a human induced pluripotent stem cell-derived cardiomyocyte (iPSC-CM) model of a DCM mutation in the sarcomere protein troponin T (TnT), TnT-R141W, compared to isogenic healthy (WT) control iPSC-CMs. We established a multimodal data fusion (MDF)-based analysis to integrate source datasets for Ca2+ transients, force measurements, and contractility recordings. Data were acquired for three additional layer types, single cells, cell monolayers, and 3D spheroid iPSC-CM models. For data analysis, numerical conversion as well as fusion of data from Ca2+ transients, force measurements, and contractility recordings, a non-negative blind deconvolution (NNBD)-based method was applied. Using an XGBoost algorithm, we found a high prediction accuracy for fused single cell, monolayer, and 3D spheroid iPSC-CM models (≥92 ± 0.08 %), as well as for fused Ca2+ transient, beating force, and contractility models (>96 ± 0.04 %). Integrating MDF and XGBoost provides a highly effective analysis tool for prediction of patho-phenotypic features in complex human disease models such as DCM iPSC-CMs.
Collapse
Affiliation(s)
- Ruheen Wali
- Department of Cardiology and Pneumology, Heart Research Center, University Medical Center, 27177 Göttingen University , Robert-Koch-Strasse 40, D-37075 Göttingen, Germany
- Partner Site Göttingen, DZHK (German Center for Cardiovascular Research), Robert-Koch-Strasse 40, D-37075 Göttingen, Germany
| | - Hang Xu
- Department of Cardiology and Pneumology, Heart Research Center, University Medical Center, 27177 Göttingen University , Robert-Koch-Strasse 40, D-37075 Göttingen, Germany
- Partner Site Göttingen, DZHK (German Center for Cardiovascular Research), Robert-Koch-Strasse 40, D-37075 Göttingen, Germany
| | - Cleophas Cheruiyot
- Department of Cardiology and Pneumology, Heart Research Center, University Medical Center, 27177 Göttingen University , Robert-Koch-Strasse 40, D-37075 Göttingen, Germany
- Partner Site Göttingen, DZHK (German Center for Cardiovascular Research), Robert-Koch-Strasse 40, D-37075 Göttingen, Germany
| | - Hafiza Nosheen Saleem
- Department of Cardiology and Pneumology, Heart Research Center, University Medical Center, 27177 Göttingen University , Robert-Koch-Strasse 40, D-37075 Göttingen, Germany
- Partner Site Göttingen, DZHK (German Center for Cardiovascular Research), Robert-Koch-Strasse 40, D-37075 Göttingen, Germany
| | - Andreas Janshoff
- Institute for Physical Chemistry, Göttingen University, Tammannstraße 6, D-37077 Göttingen, Germany
| | - Michael Habeck
- Microscopic Image Analysis, 39065 Jena University Hospital , Kollegiengasse 10, D-07743 Jena, Germany
| | - Antje Ebert
- Department of Cardiology and Pneumology, Heart Research Center, University Medical Center, 27177 Göttingen University , Robert-Koch-Strasse 40, D-37075 Göttingen, Germany
- Partner Site Göttingen, DZHK (German Center for Cardiovascular Research), Robert-Koch-Strasse 40, D-37075 Göttingen, Germany
| |
Collapse
|
42
|
Wiethoff I, Sikking M, Evers S, Gabrio A, Henkens M, Michels M, Verdonschot J, Heymans S, Hiligsmann M. Quality of life and societal costs in patients with dilated cardiomyopathy. EUROPEAN HEART JOURNAL. QUALITY OF CARE & CLINICAL OUTCOMES 2024; 10:334-344. [PMID: 37709575 PMCID: PMC11187720 DOI: 10.1093/ehjqcco/qcad056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 09/06/2023] [Accepted: 09/13/2023] [Indexed: 09/16/2023]
Abstract
AIMS Dilated cardiomyopathy (DCM) is a major cause of heart failure impairing patient wellbeing and imposing a substantial economic burden on society, but respective data are missing. This study aims to measure the quality of life (QoL) and societal costs of DCM patients. METHODS AND RESULTS A cross-sectional evaluation of QoL and societal costs of DCM patients was performed through the 5-level EuroQol and the Medical Consumption Questionnaire and Productivity Cost Questionnaire, respectively. QoL was translated into numerical values (i.e. utilities). Costs were measured from a Dutch societal perspective. Final costs were extrapolated to 1 year, reported in 2022 Euros, and compared between DCM severity according to NYHA classes. A total of 550 DCM patients from the Maastricht cardiomyopathy registry were included. Mean age was 61 years, and 34% were women. Overall utility was slightly lower for DCM patients than the population mean (0.840 vs. 0.869, P = 0.225). Among EQ-5D dimensions, DCM patients scored lowest in 'usual activities'. Total societal DCM costs were €14 843 per patient per year. Cost drivers were productivity losses (€7037) and medical costs (€4621). Patients with more symptomatic DCM (i.e. NYHA class III or IV) had significantly higher average DCM costs per year compared to less symptomatic DCM (€31 099 vs. €11 446, P < 0.001) and significantly lower utilities (0.631 vs. 0.883, P < 0.001). CONCLUSION DCM is associated with high societal costs and reduced QoL, in particular with high DCM severity.
Collapse
Affiliation(s)
- Isabell Wiethoff
- Department of Health Services Research, Care and Public Health Research Institute (CAPHRI), Maastricht University, 6200 MD Maastricht, The Netherlands
| | - Maurits Sikking
- Department of Cardiology, CARIM, Maastricht University Medical Centre, 6229 ER Maastricht, The Netherlands
| | - Silvia Evers
- Department of Health Services Research, Care and Public Health Research Institute (CAPHRI), Maastricht University, 6200 MD Maastricht, The Netherlands
- Trimbos Institute, Netherlands Institute of Mental Health and Addiction, Centre for Economic Evaluation and Machine Learning, 3500 AS Utrecht, The Netherlands
| | - Andrea Gabrio
- Department of Methodology and Statistics, Faculty of Health Medicine and Life Science, Maastricht University, 6229 HA Maastricht, The Netherlands
| | - Michiel Henkens
- Department of Cardiology, CARIM, Maastricht University Medical Centre, 6229 ER Maastricht, The Netherlands
- Department of Pathology, Maastricht University Medical Centre, 6229 HX Maastricht, The Netherlands
- Netherlands Heart Institute (NLHI), 3511 EP Utrecht, The Netherlands
| | - Michelle Michels
- Department of Cardiology, Thoraxcenter, Erasmus MC, 3000 CA Rotterdam, The Netherlands
| | - Job Verdonschot
- Department of Cardiology, CARIM, Maastricht University Medical Centre, 6229 ER Maastricht, The Netherlands
- European Reference Network for Rare, Low Prevalence and Complex Diseases of the Heart (ERN GUARD-Heart), Maastricht University Medical Centre, 6202 AZ Maastricht, The Netherlands
- Department of Clinical Genetics, Maastricht University Medical Centre, 6202 AZ Maastricht, The Netherlands
| | - Stephane Heymans
- Department of Cardiology, CARIM, Maastricht University Medical Centre, 6229 ER Maastricht, The Netherlands
- European Reference Network for Rare, Low Prevalence and Complex Diseases of the Heart (ERN GUARD-Heart), Maastricht University Medical Centre, 6202 AZ Maastricht, The Netherlands
- Department of Cardiovascular Research, University of Leuven, 3000 Leuven, Belgium
| | - Mickaël Hiligsmann
- Department of Health Services Research, Care and Public Health Research Institute (CAPHRI), Maastricht University, 6200 MD Maastricht, The Netherlands
| |
Collapse
|
43
|
Malinow I, Fong DC, Miyamoto M, Badran S, Hong CC. Pediatric dilated cardiomyopathy: a review of current clinical approaches and pathogenesis. Front Pediatr 2024; 12:1404942. [PMID: 38966492 PMCID: PMC11223501 DOI: 10.3389/fped.2024.1404942] [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: 03/21/2024] [Accepted: 06/05/2024] [Indexed: 07/06/2024] Open
Abstract
Pediatric dilated cardiomyopathy (DCM) is a rare, yet life-threatening cardiovascular condition characterized by systolic dysfunction with biventricular dilatation and reduced myocardial contractility. Therapeutic options are limited with nearly 40% of children undergoing heart transplant or death within 2 years of diagnosis. Pediatric patients are currently diagnosed based on correlating the clinical picture with echocardiographic findings. Patient age, etiology of disease, and parameters of cardiac function significantly impact prognosis. Treatments for pediatric DCM aim to ameliorate symptoms, reduce progression of disease, and prevent life-threatening arrhythmias. Many therapeutic agents with known efficacy in adults lack the same evidence in children. Unlike adult DCM, the pathogenesis of pediatric DCM is not well understood as approximately two thirds of cases are classified as idiopathic disease. Children experience unique gene expression changes and molecular pathway activation in response to DCM. Studies have pointed to a significant genetic component in pediatric DCM, with variants in genes related to sarcomere and cytoskeleton structure implicated. In this regard, pediatric DCM can be considered pediatric manifestations of inherited cardiomyopathy syndromes. Yet exciting recent studies in infantile DCM suggest that this subset has a distinct etiology involving defective postnatal cardiac maturation, such as the failure of programmed centrosome breakdown in cardiomyocytes. Improved knowledge of pathogenesis is central to developing child-specific treatment approaches. This review aims to discuss the established biological pathogenesis of pediatric DCM, current clinical guidelines, and promising therapeutic avenues, highlighting differences from adult disease. The overarching goal is to unravel the complexities surrounding this condition to facilitate the advancement of novel therapeutic interventions and improve prognosis and overall quality of life for pediatric patients affected by DCM.
Collapse
Affiliation(s)
- Ian Malinow
- Division of Cardiovascular Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Daniel C. Fong
- Division of Cardiovascular Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Matthew Miyamoto
- Division of Cardiovascular Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Sarah Badran
- Department of Pediatric Cardiology, Michigan State University College of Human Medicine Helen Devos Children’s Hospital, Grand Rapids, MI, United States
| | - Charles C. Hong
- Department of Medicine, Division of Cardiology, Michigan State University College of Human Medicine, East Lansing, MI, United States
| |
Collapse
|
44
|
Li X, Li J, Samuelsson AM, Thakur H, Kapiloff MS. Protein phosphatase 2A anchoring disruptor gene therapy for familial dilated cardiomyopathy. Mol Ther Methods Clin Dev 2024; 32:101233. [PMID: 38572067 PMCID: PMC10988123 DOI: 10.1016/j.omtm.2024.101233] [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: 11/06/2023] [Accepted: 03/08/2024] [Indexed: 04/05/2024]
Abstract
Familial dilated cardiomyopathy is a prevalent cause of heart failure that results from the mutation of genes encoding proteins of diverse function. Despite modern therapy, dilated cardiomyopathy typically has a poor outcome and is the leading cause of cardiac transplantation. The phosphatase PP2A at cardiomyocyte perinuclear mAKAPβ signalosomes promotes pathological eccentric cardiac remodeling, as is characteristic of dilated cardiomyopathy. Displacement of PP2A from mAKAPβ, inhibiting PP2A function in that intracellular compartment, can be achieved by expression of a mAKAPβ-derived PP2A binding domain-derived peptide. To test whether PP2A anchoring disruption would be effective at preventing dilated cardiomyopathy-associated cardiac dysfunction, the adeno-associated virus gene therapy vector AAV9sc.PBD was devised to express the disrupting peptide in cardiomyocytes in vivo. Proof-of-concept is now provided that AAV9sc.PBD improves the cardiac structure and function of a cardiomyopathy mouse model involving transgenic expression of a mutant α-tropomyosin E54K Tpm1 allele, while AAV9sc.PBD has no effect on normal non-transgenic mice. At the cellular level, AAV9sc.PBD restores cardiomyocyte morphology and gene expression in the mutant Tpm1 mouse. As the mechanism of AAV9sc.PBD action suggests potential efficacy in dilated cardiomyopathy regardless of the underlying etiology, these data support the further testing of AAV9sc.PBD as a broad-based treatment for dilated cardiomyopathy.
Collapse
Affiliation(s)
- Xueyi Li
- Stanford Cardiovascular Institute, Departments of Ophthalmology and Medicine, Stanford University, Palo Alto, CA 94304, USA
| | - Jinliang Li
- Stanford Cardiovascular Institute, Departments of Ophthalmology and Medicine, Stanford University, Palo Alto, CA 94304, USA
| | - Anne-Maj Samuelsson
- Stanford Cardiovascular Institute, Departments of Ophthalmology and Medicine, Stanford University, Palo Alto, CA 94304, USA
| | - Hrishikesh Thakur
- Stanford Cardiovascular Institute, Departments of Ophthalmology and Medicine, Stanford University, Palo Alto, CA 94304, USA
| | - Michael S. Kapiloff
- Stanford Cardiovascular Institute, Departments of Ophthalmology and Medicine, Stanford University, Palo Alto, CA 94304, USA
| |
Collapse
|
45
|
León P, Franco P, Hinojosa N, Torres K, Moreano A, Romero VI. TTN novel splice variant in familial dilated cardiomyopathy and splice variants review: a case report. Front Cardiovasc Med 2024; 11:1387063. [PMID: 38938651 PMCID: PMC11210389 DOI: 10.3389/fcvm.2024.1387063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 05/03/2024] [Indexed: 06/29/2024] Open
Abstract
This case report details the identification of a novel likely pathogenic splicing variant in the TTN gene, associated with dilated cardiomyopathy (DCM), in a 42-year-old male patient presenting with early-onset heart failure and reduced ejection fraction. DCM is a nonischemic heart condition characterized by left biventricular dilation and systolic dysfunction, with approximately one-third of cases being familial and often linked to genetic mutations. The TTN gene, encoding the largest human protein essential for muscle contraction and sarcomere structure, is implicated in about 25% of DCM cases through mutations, especially truncating variants. Our investigation revealed a previously unreported G > C mutation at the splice acceptor site in intron 356 of TTN, confirmed by Sanger sequencing and not found in population databases, suggesting a novel contribution to the understanding of DCM etiology. The case emphasizes the critical role of the TTN gene in cardiac function and the genetic complexity underlying DCM. A comprehensive literature review highlighted the prevalence and significance of splice variants in the TTN gene, particularly those affecting the titin A-band, which is known for its role in muscle contraction and stability. This variant's identification underscores the importance of genetic screening in patients with DCM, offering insights into the disease's familial transmission and potential therapeutic targets. Our findings contribute to the expanding knowledge of genetic factors in DCM, demonstrating the necessity of integrating genetic diagnostics in cardiovascular medicine. This case supports the growing evidence linking splicing mutations in specific regions of the TTN gene to DCM development and underscores the importance of genetic counseling and testing in managing heart disease.
Collapse
Affiliation(s)
- Paul León
- College of Biological and Environmental Sciences, Universidad San Francisco de Quito, Quito, Ecuador
| | - Paula Franco
- School of Medicine, Universidad San Francisco de Quito, Quito, Ecuador
| | - Nicole Hinojosa
- School of Medicine, Universidad San Francisco de Quito, Quito, Ecuador
| | - Kevin Torres
- School of Medicine, Universidad San Francisco de Quito, Quito, Ecuador
| | - Andrés Moreano
- Department of Cardiology, Universidad de Sao Paulo, Sao Paulo, Brazil
| | - Vanessa I. Romero
- College of Biological and Environmental Sciences, Universidad San Francisco de Quito, Quito, Ecuador
- School of Medicine, Universidad San Francisco de Quito, Quito, Ecuador
| |
Collapse
|
46
|
Auguin D, Robert-Paganin J, Réty S, Kikuti C, David A, Theumer G, Schmidt AW, Knölker HJ, Houdusse A. Omecamtiv mecarbil and Mavacamten target the same myosin pocket despite opposite effects in heart contraction. Nat Commun 2024; 15:4885. [PMID: 38849353 PMCID: PMC11161628 DOI: 10.1038/s41467-024-47587-9] [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: 10/30/2023] [Accepted: 04/03/2024] [Indexed: 06/09/2024] Open
Abstract
Inherited cardiomyopathies are common cardiac diseases worldwide, leading in the late stage to heart failure and death. The most promising treatments against these diseases are small molecules directly modulating the force produced by β-cardiac myosin, the molecular motor driving heart contraction. Omecamtiv mecarbil and Mavacamten are two such molecules that completed phase 3 clinical trials, and the inhibitor Mavacamten is now approved by the FDA. In contrast to Mavacamten, Omecamtiv mecarbil acts as an activator of cardiac contractility. Here, we reveal by X-ray crystallography that both drugs target the same pocket and stabilize a pre-stroke structural state, with only few local differences. All-atom molecular dynamics simulations reveal how these molecules produce distinct effects in motor allostery thus impacting force production in opposite way. Altogether, our results provide the framework for rational drug development for the purpose of personalized medicine.
Collapse
Affiliation(s)
- Daniel Auguin
- Structural Motility, Institut Curie, Université Paris Sciences et Lettres, Sorbonne Université, CNRS UMR144, Paris, 75248, France
- Laboratoire de Physiologie, Ecologie et Environnement (P2E), UPRES EA 1207/USC INRAE-1328, UFR Sciences et Techniques, Université d'Orléans, Orléans, France
| | - Julien Robert-Paganin
- Structural Motility, Institut Curie, Université Paris Sciences et Lettres, Sorbonne Université, CNRS UMR144, Paris, 75248, France
| | - Stéphane Réty
- Laboratoire de Biologie et Modélisation de la Cellule, ENS de Lyon, CNRS, UMR 5239, Inserm, U1293, Université Claude Bernard Lyon 1, Lyon, France
| | - Carlos Kikuti
- Structural Motility, Institut Curie, Université Paris Sciences et Lettres, Sorbonne Université, CNRS UMR144, Paris, 75248, France
| | - Amandine David
- Structural Motility, Institut Curie, Université Paris Sciences et Lettres, Sorbonne Université, CNRS UMR144, Paris, 75248, France
| | | | | | | | - Anne Houdusse
- Structural Motility, Institut Curie, Université Paris Sciences et Lettres, Sorbonne Université, CNRS UMR144, Paris, 75248, France.
| |
Collapse
|
47
|
Duno-Miranda S, Nelson SR, Rasicci DV, Bodt SM, Cirilo JA, Vang D, Sivaramakrishnan S, Yengo CM, Warshaw DM. Tail length and E525K dilated cardiomyopathy mutant alter human β-cardiac myosin super-relaxed state. J Gen Physiol 2024; 156:e202313522. [PMID: 38709176 PMCID: PMC11074782 DOI: 10.1085/jgp.202313522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 03/18/2024] [Accepted: 04/17/2024] [Indexed: 05/07/2024] Open
Abstract
Dilated cardiomyopathy (DCM) is a condition characterized by impaired cardiac function, due to myocardial hypo-contractility, and is associated with point mutations in β-cardiac myosin, the molecular motor that powers cardiac contraction. Myocardial function can be modulated through sequestration of myosin motors into an auto-inhibited "super-relaxed" state (SRX), which may be further stabilized by a structural state known as the "interacting heads motif" (IHM). Here, we sought to determine whether hypo-contractility of DCM myocardium results from reduced function of individual myosin molecules or from decreased myosin availability to interact with actin due to increased IHM/SRX stabilization. We used an established DCM myosin mutation, E525K, and characterized the biochemical and mechanical activity of wild-type and mutant human β-cardiac myosin constructs that differed in the length of their coiled-coil tail, which dictates their ability to form the IHM/SRX state. We found that short-tailed myosin constructs exhibited low IHM/SRX content, elevated actin-activated ATPase activity, and fast velocities in unloaded motility assays. Conversely, longer-tailed constructs exhibited higher IHM/SRX content and reduced actomyosin ATPase and velocity. Our modeling suggests that reduced velocities may be attributed to IHM/SRX-dependent sequestration of myosin heads. Interestingly, longer-tailed E525K mutants showed no apparent impact on velocity or actomyosin ATPase at low ionic strength but stabilized IHM/SRX state at higher ionic strength. Therefore, the hypo-contractility observed in DCM may be attributable to reduced myosin head availability caused by enhanced IHM/SRX stability in E525K mutants.
Collapse
Affiliation(s)
- Sebastian Duno-Miranda
- Department of Molecular Physiology and Biophysics, Cardiovascular Research Institute, University of Vermont, Burlington, VT, USA
| | - Shane R. Nelson
- Department of Molecular Physiology and Biophysics, Cardiovascular Research Institute, University of Vermont, Burlington, VT, USA
| | - David V. Rasicci
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, PA, USA
| | - Skylar M.L. Bodt
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, PA, USA
| | - Joseph A. Cirilo
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, PA, USA
| | - Duha Vang
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN, USA
| | - Sivaraj Sivaramakrishnan
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN, USA
| | - Christopher M. Yengo
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, PA, USA
| | - David M. Warshaw
- Department of Molecular Physiology and Biophysics, Cardiovascular Research Institute, University of Vermont, Burlington, VT, USA
| |
Collapse
|
48
|
Marchal GA, Rivaud MR, Wolswinkel R, Basso C, van Veen TAB, Bezzina CR, Remme CA. Genetic background determines the severity of age-dependent cardiac structural abnormalities and arrhythmia susceptibility in Scn5a-1798insD mice. Europace 2024; 26:euae153. [PMID: 38875491 PMCID: PMC11203918 DOI: 10.1093/europace/euae153] [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: 02/09/2024] [Accepted: 05/12/2024] [Indexed: 06/16/2024] Open
Abstract
AIMS Patients with mutations in SCN5A encoding NaV1.5 often display variable severity of electrical and structural alterations, but the underlying mechanisms are not fully elucidated. We here investigate the combined modulatory effect of genetic background and age on disease severity in the Scn5a1798insD/+ mouse model. METHODS AND RESULTS In vivo electrocardiogram and echocardiograms, ex vivo electrical and optical mapping, and histological analyses were performed in adult (2-7 months) and aged (8-28 months) wild-type (WT) and Scn5a1798insD/+ (mutant, MUT) mice from the FVB/N and 129P2 inbred strains. Atrio-ventricular (AV) conduction, ventricular conduction, and ventricular repolarization are modulated by strain, genotype, and age. An aging effect was present in MUT mice, with aged MUT mice of both strains showing prolonged QRS interval and right ventricular (RV) conduction slowing. 129P2-MUT mice were severely affected, with adult and aged 129P2-MUT mice displaying AV and ventricular conduction slowing, prolonged repolarization, and spontaneous arrhythmias. In addition, the 129P2 strain appeared particularly susceptible to age-dependent electrical, functional, and structural alterations including RV conduction slowing, reduced left ventricular (LV) ejection fraction, RV dilatation, and myocardial fibrosis as compared to FVB/N mice. Overall, aged 129P2-MUT mice displayed the most severe conduction defects, RV dilatation, and myocardial fibrosis, in addition to the highest frequency of spontaneous arrhythmia and inducible arrhythmias. CONCLUSION Genetic background and age both modulate disease severity in Scn5a1798insD/+ mice and hence may explain, at least in part, the variable disease expressivity observed in patients with SCN5A mutations. Age- and genetic background-dependent development of cardiac structural alterations furthermore impacts arrhythmia risk. Our findings therefore emphasize the importance of continued assessment of cardiac structure and function in patients carrying SCN5A mutations.
Collapse
Affiliation(s)
- Gerard A Marchal
- Department of Experimental Cardiology, Heart Centre, Amsterdam UMC location University of Amsterdam, Meibergdreef 15, PO Box 22660, 1100 DD Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Meibergdreef 15, PO Box 22660, 1100 DD Amsterdam, The Netherlands
- OptoCARD Lab, Institute of Clinical Physiology (IFC-CNR), Florence, Italy
| | - Mathilde R Rivaud
- Department of Experimental Cardiology, Heart Centre, Amsterdam UMC location University of Amsterdam, Meibergdreef 15, PO Box 22660, 1100 DD Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Meibergdreef 15, PO Box 22660, 1100 DD Amsterdam, The Netherlands
| | - Rianne Wolswinkel
- Department of Experimental Cardiology, Heart Centre, Amsterdam UMC location University of Amsterdam, Meibergdreef 15, PO Box 22660, 1100 DD Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Meibergdreef 15, PO Box 22660, 1100 DD Amsterdam, The Netherlands
| | - Cristina Basso
- Department of Cardiac, Thoracic and Vascular Sciences and Public Health, University of Padova, Padua, Italy
| | - Toon A B van Veen
- Department of Medical Physiology, Division Heart & Lungs, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Connie R Bezzina
- Department of Experimental Cardiology, Heart Centre, Amsterdam UMC location University of Amsterdam, Meibergdreef 15, PO Box 22660, 1100 DD Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Meibergdreef 15, PO Box 22660, 1100 DD Amsterdam, The Netherlands
| | - Carol Ann Remme
- Department of Experimental Cardiology, Heart Centre, Amsterdam UMC location University of Amsterdam, Meibergdreef 15, PO Box 22660, 1100 DD Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Meibergdreef 15, PO Box 22660, 1100 DD Amsterdam, The Netherlands
| |
Collapse
|
49
|
Petmezas G, Papageorgiou VE, Vassilikos V, Pagourelias E, Tsaklidis G, Katsaggelos AK, Maglaveras N. Recent advancements and applications of deep learning in heart failure: Α systematic review. Comput Biol Med 2024; 176:108557. [PMID: 38728995 DOI: 10.1016/j.compbiomed.2024.108557] [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: 03/15/2024] [Revised: 04/12/2024] [Accepted: 05/05/2024] [Indexed: 05/12/2024]
Abstract
BACKGROUND Heart failure (HF), a global health challenge, requires innovative diagnostic and management approaches. The rapid evolution of deep learning (DL) in healthcare necessitates a comprehensive review to evaluate these developments and their potential to enhance HF evaluation, aligning clinical practices with technological advancements. OBJECTIVE This review aims to systematically explore the contributions of DL technologies in the assessment of HF, focusing on their potential to improve diagnostic accuracy, personalize treatment strategies, and address the impact of comorbidities. METHODS A thorough literature search was conducted across four major electronic databases: PubMed, Scopus, Web of Science and IEEE Xplore, yielding 137 articles that were subsequently categorized into five primary application areas: cardiovascular disease (CVD) classification, HF detection, image analysis, risk assessment, and other clinical analyses. The selection criteria focused on studies utilizing DL algorithms for HF assessment, not limited to HF detection but extending to any attempt in analyzing and interpreting HF-related data. RESULTS The analysis revealed a notable emphasis on CVD classification and HF detection, with DL algorithms showing significant promise in distinguishing between affected individuals and healthy subjects. Furthermore, the review highlights DL's capacity to identify underlying cardiomyopathies and other comorbidities, underscoring its utility in refining diagnostic processes and tailoring treatment plans to individual patient needs. CONCLUSIONS This review establishes DL as a key innovation in HF management, highlighting its role in advancing diagnostic accuracy and personalized care. The insights provided advocate for the integration of DL in clinical settings and suggest directions for future research to enhance patient outcomes in HF care.
Collapse
Affiliation(s)
- Georgios Petmezas
- 2nd Department of Obstetrics and Gynecology, Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece; Centre for Research and Technology Hellas, Thessaloniki, Greece.
| | | | - Vasileios Vassilikos
- 3rd Department of Cardiology, Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Efstathios Pagourelias
- 3rd Department of Cardiology, Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - George Tsaklidis
- Department of Mathematics, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Aggelos K Katsaggelos
- Department of Electrical and Computer Engineering, Northwestern University, Evanston, IL, USA
| | - Nicos Maglaveras
- 2nd Department of Obstetrics and Gynecology, Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
50
|
Ichimura S, Misaka T, Ogawara R, Tomita Y, Anzai F, Sato Y, Miura S, Yokokawa T, Sato T, Oikawa M, Kobayashi A, Yoshihisa A, Takeishi Y. Neutrophil Extracellular Traps in Myocardial Tissue Drive Cardiac Dysfunction and Adverse Outcomes in Patients With Heart Failure With Dilated Cardiomyopathy. Circ Heart Fail 2024; 17:e011057. [PMID: 38847093 DOI: 10.1161/circheartfailure.123.011057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 04/26/2024] [Indexed: 06/20/2024]
Abstract
BACKGROUND The immune systems and chronic inflammation are implicated in the pathogenesis of dilated cardiomyopathy (DCM) and heart failure. However, the significance of neutrophil extracellular traps (NETs) in heart failure remains to be elucidated. METHODS We enrolled consecutive 62 patients with heart failure with idiopathic DCM who underwent endomyocardial biopsy. Biopsy specimens were subjected to fluorescent immunostaining to detect NETs, and clinical and outcome data were collected. Ex vivo and in vivo experiments were conducted. RESULTS The numbers of NETs per myocardial tissue area and the proportion of NETs per neutrophil were significantly higher in patients with DCM compared with non-DCM control subjects without heart failure, and the numbers of NETs were negatively correlated with left ventricular ejection fraction. Patients with DCM with NETs (n=32) showed lower left ventricular ejection fraction and higher BNP (B-type natriuretic peptide) than those without NETs (n=30). In a multivariable Cox proportional hazard model, the presence of NETs was independently associated with an increased risk of adverse cardiac events in patients with DCM. To understand specific underlying mechanisms, extracellular flux analysis in ex vivo revealed that NETs-containing conditioned medium from wild-type neutrophils or purified NET components led to impaired mitochondrial oxygen consumption of cardiomyocytes, while these effects were abolished when PAD4 (peptidyl arginine deiminase 4) in neutrophils was genetically ablated. In a murine model of pressure overload, NETs in myocardial tissue were predominantly detected in the acute phase and persisted throughout the ongoing stress. Four weeks after transverse aortic constriction, left ventricular ejection fraction was reduced in wild-type mice, whereas PAD4-deficient mice displayed preserved left ventricular ejection fraction without inducing NET formation. CONCLUSIONS NETs in myocardial tissue contribute to cardiac dysfunction and adverse outcomes in patients with heart failure with DCM, potentially through mitochondrial dysfunction of cardiomyocytes.
Collapse
Affiliation(s)
- Shohei Ichimura
- Department of Cardiovascular Medicine (S.I., T.M., R.O., Y.T., F.A., Y.S., S.M., T.Y., T.S., M.O., A.K., A.Y., Y.T.), Fukushima Medical University, Japan
| | - Tomofumi Misaka
- Department of Cardiovascular Medicine (S.I., T.M., R.O., Y.T., F.A., Y.S., S.M., T.Y., T.S., M.O., A.K., A.Y., Y.T.), Fukushima Medical University, Japan
- Department of Community Cardiovascular Medicine (T.M., A.K.), Fukushima Medical University, Japan
| | - Ryo Ogawara
- Department of Cardiovascular Medicine (S.I., T.M., R.O., Y.T., F.A., Y.S., S.M., T.Y., T.S., M.O., A.K., A.Y., Y.T.), Fukushima Medical University, Japan
| | - Yusuke Tomita
- Department of Cardiovascular Medicine (S.I., T.M., R.O., Y.T., F.A., Y.S., S.M., T.Y., T.S., M.O., A.K., A.Y., Y.T.), Fukushima Medical University, Japan
| | - Fumiya Anzai
- Department of Cardiovascular Medicine (S.I., T.M., R.O., Y.T., F.A., Y.S., S.M., T.Y., T.S., M.O., A.K., A.Y., Y.T.), Fukushima Medical University, Japan
| | - Yu Sato
- Department of Cardiovascular Medicine (S.I., T.M., R.O., Y.T., F.A., Y.S., S.M., T.Y., T.S., M.O., A.K., A.Y., Y.T.), Fukushima Medical University, Japan
| | - Shunsuke Miura
- Department of Cardiovascular Medicine (S.I., T.M., R.O., Y.T., F.A., Y.S., S.M., T.Y., T.S., M.O., A.K., A.Y., Y.T.), Fukushima Medical University, Japan
| | - Tetsuro Yokokawa
- Department of Cardiovascular Medicine (S.I., T.M., R.O., Y.T., F.A., Y.S., S.M., T.Y., T.S., M.O., A.K., A.Y., Y.T.), Fukushima Medical University, Japan
| | - Takamasa Sato
- Department of Cardiovascular Medicine (S.I., T.M., R.O., Y.T., F.A., Y.S., S.M., T.Y., T.S., M.O., A.K., A.Y., Y.T.), Fukushima Medical University, Japan
| | - Masayoshi Oikawa
- Department of Cardiovascular Medicine (S.I., T.M., R.O., Y.T., F.A., Y.S., S.M., T.Y., T.S., M.O., A.K., A.Y., Y.T.), Fukushima Medical University, Japan
| | - Atsushi Kobayashi
- Department of Community Cardiovascular Medicine (T.M., A.K.), Fukushima Medical University, Japan
| | - Akiomi Yoshihisa
- Department of Clinical Laboratory Sciences (A.Y.), Fukushima Medical University, Japan
| | - Yasuchika Takeishi
- Department of Cardiovascular Medicine (S.I., T.M., R.O., Y.T., F.A., Y.S., S.M., T.Y., T.S., M.O., A.K., A.Y., Y.T.), Fukushima Medical University, Japan
| |
Collapse
|