1
|
Zhou C, Zhao H, Peng L, Dong Y, Wu Q, Wang X, Xu Y, Wang Y. Astragalus polysaccharide protects against cardiac injury in a tnnt2a mutant zebrafish model of dilated cardiomyopathy. BMC Complement Med Ther 2025; 25:197. [PMID: 40450272 DOI: 10.1186/s12906-025-04925-8] [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: 08/07/2023] [Accepted: 05/16/2025] [Indexed: 06/03/2025] Open
Abstract
BACKGROUND Dilated cardiomyopathy (DCM) is a severe and irreversible heart disease characterized by dilated ventricles and decreased myocardial function. DCM has a poor prognosis and a very low survival rate, with a 5-year mortality rate ranging from 15 to 50%, and is an important cause of sudden cardiac death and heart failure. Genetic factors play important roles in the pathogenesis of DCM. Mutations in the cardiac troponin T (tnnt2) gene represent an important subset of known pathogenic variants that bind to DCM. However, few specific drugs are currently available to treat DCM caused by these gene mutations. Astragalus polysaccharide (APS), the main active ingredient of Astragalus mongholicus Bunge (Huangqi), is widely used in China to treat cardiovascular diseases, including DCM. This study explored drugs for the treatment of DCM caused by tnnt2a mutation and revealed the protective effect of APS on tnnt2a-mutant dilated cardiomyopathy. METHODS The tnnt2a-/- mutant zebrafish were used as a DCM model for comparison with the APS-treated group. The survival rate and the sinus venosus‒bulbus arteriosus (SV‒BA) distance were used to observe changes in cardiac output. Histopathological changes were observed via hematoxylin and eosin (HE) staining and TUNEL staining. The transcriptomes of the zebrafish in the DCM group and APS-treated group were investigated via RNA-seq. qRT‒PCR detection of apoptosis-related gene expression. RESULTS We found that APS markedly increased the heart rate and ATP content, and significantly inhibited the level of cardiac tissue edema, which are essential for improving the survival rate of tnnt2a-/-. Furthermore, APS modulates key muscle fiber-related genes (including ttnb and myom3) and significantly impacts multiple signaling pathways, including Rap1, PI3K-Akt, Jak-STAT, and Wnt signaling. The qRT‒PCR results revealed that APS decreased the expression of bax, caspase-3, and caspase-9 but increased the expression of bcl-2 in DCM zebrafish. CONCLUSIONS Our findings suggest that APS can improve the survival rate in dilated cardiomyopathy and has a positive protective effect on the myocardium in the tnnt2a mutant zebrafish model of DCM.
Collapse
Affiliation(s)
- Chang Zhou
- Department of Cardiology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, 200240, China
| | - Hui Zhao
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Longping Peng
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Yidan Dong
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Qiong Wu
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Xu Wang
- Cancer Metabolism Laboratory, Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Yingjia Xu
- Department of Cardiology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, 200240, China.
| | - Youhua Wang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
| |
Collapse
|
2
|
Jadidi M, Babaali V, InanlooRahatloo K, Salehi N, Mollazadeh R. Identification of a rare variant in TNNT3 responsible for familial dilated cardiomyopathy through whole-exome sequencing and in silico analysis. Eur J Med Res 2025; 30:424. [PMID: 40437600 PMCID: PMC12117963 DOI: 10.1186/s40001-025-02692-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Accepted: 05/15/2025] [Indexed: 06/01/2025] Open
Abstract
BACKGROUND Dilated cardiomyopathy (DCM) is a prevalent etiology of heart failure, distinguished by the gradual and frequently irreversible myocardial muscle impairment. Roughly 50% of DCM occurrences stem from hereditary rare variants. In this study, our aim was to identify the genetic cause of DCM in a pedigree with several affected individuals across four generations. METHODS Whole exome sequencing was performed on the proband, with variants filtered and analyzed using in silico tools. Co-segregation analysis was conducted using Sanger sequencing. Protein structure modeling and protein-protein interaction evaluations were performed using AlphaFold3 and HADDOCK2.4, respectively. RESULTS We identified a missense rare variant in the TNNT3 gene, leading to the p.Glu125Gly alteration in the Troponin T3 (TNNT3). This rare variant is strongly implicated as the causative factor for DCM in the pedigree. Several key factors underscore its significance: the rare variant co-segregates with the disease in the pedigree, is absent in 850 control samples, alters a conserved amino acid, is predicted to detrimentally affect protein function, and results in structural changes. CONCLUSIONS Our findings suggest that TNNT3 rare variants can induce DCM by weakening the binding energy between TNNT3 and Tropomyosin (TPM), leading to functional deficiencies in muscle contraction, as demonstrated by our structural modeling and docking studies. Troponin T is essential for the proper contraction of striated muscles and is related to cardiac development. Bioinformatics investigations have elucidated the involvement of TNNT3-related pathways, notably the Striated Muscle Contraction pathway and Cardiac Conduction. TNNT3 resides within loci previously implicated in cardiomyopathy. Given its crucial role in muscle contractile function, rare variants in TNNT3 hold the potential to be a significant contributing factor in the pathogenesis of DCM. A wealth of literature substantiates the correlation between troponin T and cardiac disorders. Our findings further corroborate this association.
Collapse
Affiliation(s)
- Motahareh Jadidi
- Department of Cell and Molecular Biology, School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Vida Babaali
- Department of Cell and Molecular Biology, School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Kolsoum InanlooRahatloo
- Department of Cell and Molecular Biology, School of Biology, College of Science, University of Tehran, Tehran, Iran.
| | - Najmeh Salehi
- Department of Applied Biological Sciences, School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Reza Mollazadeh
- Department of Cardiology, School of Medicine, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
3
|
Vo QD, Nakamura K, Saito Y, Akagi S, Miyoshi T, Yuasa S. Induced Pluripotent Stem Cells in Cardiomyopathy: Advancing Disease Modeling, Therapeutic Development, and Regenerative Therapy. Int J Mol Sci 2025; 26:4984. [PMID: 40507801 PMCID: PMC12155407 DOI: 10.3390/ijms26114984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2025] [Revised: 05/19/2025] [Accepted: 05/19/2025] [Indexed: 06/16/2025] Open
Abstract
Cardiomyopathies are a heterogeneous group of heart muscle diseases that can lead to heart failure, arrhythmias, and sudden cardiac death. Traditional animal models and in vitro systems have limitations in replicating the complex pathology of human cardiomyopathies. Induced pluripotent stem cells (iPSCs) offer a transformative platform by enabling the generation of patient-specific cardiomyocytes, thus opening new avenues for disease modeling, drug discovery, and regenerative therapy. This process involves reprogramming somatic cells into iPSCs and subsequently differentiating them into functional cardiomyocytes, which can be characterized using techniques such as electrophysiology, contractility assays, and gene expression profiling. iPSC-derived cardiomyocyte (iPSC-CM) platforms are also being explored for drug screening and personalized medicine, including high-throughput testing for cardiotoxicity and the identification of patient-tailored therapies. While iPSC-CMs already serve as valuable models for understanding disease mechanisms and screening drugs, ongoing advances in maturation and bioengineering are bringing iPSC-based therapies closer to clinical application. Furthermore, the integration of multi-omics approaches and artificial intelligence (AI) is enhancing the predictive power of iPSC models. iPSC-based technologies are paving the way for a new era of personalized cardiology, with the potential to revolutionize the management of cardiomyopathies through patient-specific insights and regenerative strategies.
Collapse
Affiliation(s)
- Quan Duy Vo
- Department of Cardiovascular Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (Q.D.V.); (S.A.); (T.M.); (S.Y.)
| | - Kazufumi Nakamura
- Department of Cardiovascular Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (Q.D.V.); (S.A.); (T.M.); (S.Y.)
- Center for Advanced Heart Failure, Okayama University Hospital, Okayama 700-8558, Japan
| | - Yukihiro Saito
- Department of Cardiovascular Medicine, Okayama University Hospital, Okayama 700-8558, Japan;
| | - Satoshi Akagi
- Department of Cardiovascular Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (Q.D.V.); (S.A.); (T.M.); (S.Y.)
| | - Toru Miyoshi
- Department of Cardiovascular Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (Q.D.V.); (S.A.); (T.M.); (S.Y.)
| | - Shinsuke Yuasa
- Department of Cardiovascular Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (Q.D.V.); (S.A.); (T.M.); (S.Y.)
| |
Collapse
|
4
|
Khattab E, Myrianthefs MM, Sakellaropoulos S, Alexandrou K, Mitsis A. Precision medicine applications in dilated cardiomyopathy: Advancing personalized care. Curr Probl Cardiol 2025; 50:103076. [PMID: 40381754 DOI: 10.1016/j.cpcardiol.2025.103076] [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: 05/08/2025] [Accepted: 05/14/2025] [Indexed: 05/20/2025]
Abstract
Dilated cardiomyopathy (DCM) is a prevalent cardiac disorder affecting 1 in 250-500 individuals, characterized by ventricular dilation and impaired systolic function, leading to heart failure and increased mortality, including sudden cardiac death. DCM arises from genetic and environmental factors, such as drug-induced, inflammatory, and viral causes, resulting in diverse yet overlapping phenotypes. Advances in precision medicine are revolutionizing DCM management by leveraging genetic and molecular profiling for tailored diagnostic and therapeutic approaches. This review highlights comprehensive diagnostic evaluations, genetic discoveries, and multi-omics approaches integrating genomic, transcriptomic, proteomic, and metabolomic data to enhance understanding of DCM pathophysiology. Innovative risk stratification methods, including machine learning, are improving predictions of disease progression. Despite these advancements, the current one-size-fits-all management strategy contributes to persistently high morbidity and mortality. Emerging targeted therapies, such as CRISPR/Cas9 genome editing, aetiology-specific interventions, and pharmacogenomics, are reshaping treatment paradigms. Precision medicine holds promise for optimizing DCM diagnosis, treatment, and outcomes, aiming to reduce the burden of this debilitating condition.
Collapse
Affiliation(s)
- Elina Khattab
- Cardiology Department, Consultant Interventional Cardiologist, Nicosia General Hospital, State Health Services Organization, 215, Old Road Nicosia-Limassol, Nicosia 2029, Cyprus
| | - Michael M Myrianthefs
- Cardiology Department, Consultant Interventional Cardiologist, Nicosia General Hospital, State Health Services Organization, 215, Old Road Nicosia-Limassol, Nicosia 2029, Cyprus
| | - Stefanos Sakellaropoulos
- Department of Internal Medicine, Cardiology Clinic, Kantonsspital Baden, Baden 5404, Switzerland
| | - Kyriakos Alexandrou
- Department of Nursing, School of Health Sciences, Cyprus University of Technology; Archiepiskopou Kyprianou 30, Limassol 3036, Cyprus
| | - Andreas Mitsis
- Cardiology Department, Consultant Interventional Cardiologist, Nicosia General Hospital, State Health Services Organization, 215, Old Road Nicosia-Limassol, Nicosia 2029, Cyprus.
| |
Collapse
|
5
|
Gerritse M, van Ham WB, Denning C, van Veen TAB, Maas RGC. Characteristics and pharmacological responsiveness in hiPSC models of inherited cardiomyopathy. Pharmacol Ther 2025; 272:108845. [PMID: 40250811 DOI: 10.1016/j.pharmthera.2025.108845] [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: 11/11/2024] [Revised: 02/17/2025] [Accepted: 03/24/2025] [Indexed: 04/20/2025]
Abstract
Inherited cardiomyopathies are a major cause of heart failure in all age groups, often with an onset in adolescence or early adult life. More than a thousand variants in approximately one hundred genes are associated with cardiomyopathies. Interestingly, many genetic cardiomyopathies display overlapping phenotypical defects in patients, despite the diversity of the initial pathogenic variants. Understanding how the underlying pathophysiology of genetic cardiomyopathies leads to these phenotypes will improve insights into a patient's disease course, and creates the opportunity for conceiving treatment strategies. Moreover, therapeutic strategies can be used to treat multiple cardiomyopathies based on shared phenotypes. Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) offer reliable, high-throughput models for studying molecular and cellular characteristics of hereditary cardiomyopathies. hiPSC-CMs are produced relatively easily, either by directly originating them from patients, or by introducing patient-specific genetic variants in healthy lines. This review evaluates 90 studies on 24 cardiomyopathy-associated genes and systematically summarises the morphological and functional phenotypes observed in hiPSC-CMs. Additionally, treatment strategies applied in cardiomyopathic hiPSC-CMs are compiled and scored for effectiveness. Multiple overlapping phenotypic defects were identified in cardiomyocytes with different variants, whereas certain characteristics were only associated with specific genetic variants. Based on these findings, common mechanisms, therapeutic prospects, and considerations for future research are discussed with the aim to improve clinical translation from hiPSC-CMs to patients.
Collapse
Affiliation(s)
- Merel Gerritse
- Utrecht Regenerative Medicine Center, Circulatory Health Research Center, University Utrecht, 3584 CS Utrecht, the Netherlands; Department of Medical Physiology, Division Heart & Lungs, University Medical Center Utrecht, 3584 CM Utrecht, the Netherlands.
| | - Willem B van Ham
- Department of Medical Physiology, Division Heart & Lungs, University Medical Center Utrecht, 3584 CM Utrecht, the Netherlands.
| | - Chris Denning
- Department of Stem Cell Biology, Biodiscovery Institute, University of Nottingham, University Park, Nottingham, NG7 2RD, UK.
| | - Toon A B van Veen
- Department of Medical Physiology, Division Heart & Lungs, University Medical Center Utrecht, 3584 CM Utrecht, the Netherlands.
| | - Renee G C Maas
- Utrecht Regenerative Medicine Center, Circulatory Health Research Center, University Utrecht, 3584 CS Utrecht, the Netherlands; Department of Cardiology, Experimental Cardiology Laboratory, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, the Netherlands.
| |
Collapse
|
6
|
Massier M, de Groote P, Donal E, Magnin-Poull I, Coubes C, Le Guillou Horn X, Rooryck C, Réant P, Troadec Y, Bréhin AC, Proukhnitzky J, Gandjbakhch E, Charron P, Richard P, Ader F. Exploring the Familial Phenotypic Variability Associated With TTN Truncating Variants in Cardiomyopathies: Variant Spectrum, Genotype-Phenotype Correlation and Consequences in Genetic Counseling. Clin Genet 2025; 107:425-433. [PMID: 39844436 DOI: 10.1111/cge.14679] [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: 10/08/2024] [Revised: 12/07/2024] [Accepted: 12/11/2024] [Indexed: 01/24/2025]
Abstract
Titin truncating variants (TTNtv) are the main genetic cause of dilated cardiomyopathies (DCMs). The phenotype and prognosis of probands have been evaluated in several large cohorts. However, few data are available on intrafamilial expressivity. To evaluate the phenotypical variability, we selected probands and family members carrying a unique TTN variant and recorded cardiac and genetic information. The cohort included 332 probands (314 TTNtv probands and 18 probands with in silico predicted in-frame exon skipping probands) and 191 relatives of TTNtv probands including 98 affected family members. Within TTNtv families, 96% of the affected relatives presented the same cardiomyopathy subtype as the proband, and 60% shared severity criteria (heart transplantation, implantable cardioverter-defibrillator, personal sudden death). Furthermore, we reported 18 probands that carry predicted in-frame exon skipping variants; they presented DCM (84%) as TTNtv patients but lower rate of rhythm disorders (0% vs. 29% respectively). In this work, we extend the genetic spectrum of TTNtv associated with DCM and show that within a family, and the cardiomyopathy phenotype is homogenous but the expressivity could vary. Such results are helpful for appropriate genetic counseling to better predict and manage the phenotype of mutation carriers.
Collapse
Affiliation(s)
- Marie Massier
- Sorbonne Université- DMU BioGem-Unité Fonctionnelle de Cardiogénétique et Myogénétique Moléculaire et Cellulaire, Service de Biochimie Métabolique, APHP-Hôpital Universitaire Pitié Salpêtrière, Paris, France
- Service de génétique Clinique, CHU Reims, Reims, France
| | | | - Erwan Donal
- Service de Cardiologie, CHU Rennes, Rennes, France
| | | | | | | | | | | | | | | | - Julie Proukhnitzky
- Centre de référence Des Maladies Cardiaques héréditaires, Hôpital Pitié Salpêtrière, Paris, France
| | - Estelle Gandjbakhch
- Centre de référence Des Maladies Cardiaques héréditaires, Hôpital Pitié Salpêtrière, Paris, France
- INSERM UMRS1166 Équipe 1, ICAN Institute (Institut de cardiométabolisme et Nutrition), Paris, France
| | - Philippe Charron
- Centre de référence Des Maladies Cardiaques héréditaires, Hôpital Pitié Salpêtrière, Paris, France
- INSERM UMRS1166 Équipe 1, ICAN Institute (Institut de cardiométabolisme et Nutrition), Paris, France
| | - Pascale Richard
- Sorbonne Université- DMU BioGem-Unité Fonctionnelle de Cardiogénétique et Myogénétique Moléculaire et Cellulaire, Service de Biochimie Métabolique, APHP-Hôpital Universitaire Pitié Salpêtrière, Paris, France
- INSERM UMRS1166 Équipe 1, ICAN Institute (Institut de cardiométabolisme et Nutrition), Paris, France
| | - Flavie Ader
- Sorbonne Université- DMU BioGem-Unité Fonctionnelle de Cardiogénétique et Myogénétique Moléculaire et Cellulaire, Service de Biochimie Métabolique, APHP-Hôpital Universitaire Pitié Salpêtrière, Paris, France
- INSERM UMRS1166 Équipe 1, ICAN Institute (Institut de cardiométabolisme et Nutrition), Paris, France
- Université Paris Cité, UFR de Pharmacie, Paris, France
| |
Collapse
|
7
|
Pastori P, Balla C, Rasia M, Lo Jacono E, Guerra C, Schininà R, Gualandi F, Bertini M, Tortorella G. Dilated Cardiomyopathy: A Novel BAG3 Mutation Associated with Aggressive Disease Progression and Ventricular Arrhythmias. J Cardiovasc Dev Dis 2025; 12:121. [PMID: 40278180 PMCID: PMC12027914 DOI: 10.3390/jcdd12040121] [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: 02/20/2025] [Revised: 03/16/2025] [Accepted: 03/24/2025] [Indexed: 04/26/2025] Open
Abstract
We present the case of a 46-year-old man with a history of complex ventricular arrhythmias preceding the development of asymptomatic mild left ventricular dysfunction, who presented with acute-onset heart failure and was ultimately diagnosed with dilated cardiomyopathy. Genetic testing identified a novel, likely pathogenic mutation in exon 4 of the BAG3 gene (NM_004281, c.1128del, (p.(Ser377AlafsTer47)), not previously reported in the literature. Given the presence of multiple clinical features indicative of a poor prognosis, he underwent prophylactic placement of a subcutaneous implantable cardioverter-defibrillator. The clinical presentation of this novel BAG3 mutation suggests that it may be associated with a significant arrhythmic phenotype. This case underscores the importance of close follow-up and genetic testing in patients presenting with mild left ventricular dysfunction and ventricular arrhythmias.
Collapse
Affiliation(s)
- Paolo Pastori
- Cardiology Unit, Medical and Diagnostics Department, Fidenza Hospital, Azienda USL of Parma, 43036 Fidenza, Italy; (P.P.); (M.R.); (E.L.J.); (C.G.); (G.T.)
| | - Cristina Balla
- Cardiology Unit, Department of Translational Medicine, Sant’Anna University Hospital, University of Ferrara, Via Aldo Moro 8, 44124 Ferrara, Italy;
| | - Marta Rasia
- Cardiology Unit, Medical and Diagnostics Department, Fidenza Hospital, Azienda USL of Parma, 43036 Fidenza, Italy; (P.P.); (M.R.); (E.L.J.); (C.G.); (G.T.)
| | - Emilia Lo Jacono
- Cardiology Unit, Medical and Diagnostics Department, Fidenza Hospital, Azienda USL of Parma, 43036 Fidenza, Italy; (P.P.); (M.R.); (E.L.J.); (C.G.); (G.T.)
| | - Clelia Guerra
- Cardiology Unit, Medical and Diagnostics Department, Fidenza Hospital, Azienda USL of Parma, 43036 Fidenza, Italy; (P.P.); (M.R.); (E.L.J.); (C.G.); (G.T.)
| | - Roberta Schininà
- Medical Genetics Service, Department of Mother and Child, Sant’Anna University Hospital, Via Aldo Moro 8, 44124 Ferrara, Italy; (R.S.); (F.G.)
| | - Francesca Gualandi
- Medical Genetics Service, Department of Mother and Child, Sant’Anna University Hospital, Via Aldo Moro 8, 44124 Ferrara, Italy; (R.S.); (F.G.)
| | - Matteo Bertini
- Cardiology Unit, Department of Translational Medicine, Sant’Anna University Hospital, University of Ferrara, Via Aldo Moro 8, 44124 Ferrara, Italy;
| | - Giovanni Tortorella
- Cardiology Unit, Medical and Diagnostics Department, Fidenza Hospital, Azienda USL of Parma, 43036 Fidenza, Italy; (P.P.); (M.R.); (E.L.J.); (C.G.); (G.T.)
| |
Collapse
|
8
|
Garmany R, Castrichini M, Neves R, Pereira NL, MacIntyre C, Schneider JW, Ackerman MJ, Giudicessi JR. Age at onset and clinical course of RBM20-mediated cardiomyopathy. Sci Rep 2025; 15:10716. [PMID: 40155426 PMCID: PMC11953230 DOI: 10.1038/s41598-025-95409-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 03/19/2025] [Indexed: 04/01/2025] Open
Abstract
Disease-causative variants in RBM20-encoded RNA-binding motif protein 20 cause a severe arrhythmogenic dilated cardiomyopathy (DCM). We aimed to characterize the clinical course of RBM20-mediated DCM in comparison to other familial and non-familial forms of DCM. The Mayo Clinic Genetic Arrhythmogenic Cardiomyopathy (ACM) Registry was used to identify RBM20- and TTNtv-positive individuals. Additionally, patients with idiopathic non-ischemic DCM were included in this study. Ventricular arrhythmic (VA) events were defined as sustained ventricular arrhythmia, sudden cardiac arrest (SCA), or ventricular tachycardia (VT)/ventricular fibrillation (VF)-terminating implantable cardioverter defibrillator (ICD) therapy and advanced heart failure (AHF) events defined as ventricular assist device implantation or heart transplantation. Overall, we identified 43 RBM20- (49% female) and 108 TTNtv- positive individuals (44% female). RBM20 variant-positive individuals were younger at time of diagnosis at 31 years vs. 45 years old for TTNtv-positive individuals (p < 0.0001). Additionally, RBM20 variant-positive individuals were more likely to have a family history of SCA (67% vs. 30%; p < 0.0001) and cardiomyopathy (88% vs. 61%; p = 0.0008). Interestingly, the prevalence of AHF (14% vs. 5%) events was higher in RBM20 variant-positive patients. RBM20 variant-positive individuals had earlier times to both VA (p = 0.007) and AHF (p = 0.0008) events. Findings were similar in comparison to idiopathic DCM. In this single center study, RBM20 patients had an earlier progressing disease with a higher prevalence of advanced heart failure compared with TTNtv patients. RBM20 patients progressed to VA and AHF endpoints earlier.
Collapse
Affiliation(s)
- Ramin Garmany
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic Alix School of Medicine and the Mayo Clinic Medical Scientist Training Program, Rochester, MN, USA
- Department of Molecular Pharmacology & Experimental Therapeutics, Windland Smith Rice Sudden Death Genomics Laboratory, Mayo Clinic, Rochester, MN, USA
| | - Matteo Castrichini
- Department of Molecular Pharmacology & Experimental Therapeutics, Windland Smith Rice Sudden Death Genomics Laboratory, Mayo Clinic, Rochester, MN, USA
- Department of Cardiovascular Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Raquel Neves
- Department of Molecular Pharmacology & Experimental Therapeutics, Windland Smith Rice Sudden Death Genomics Laboratory, Mayo Clinic, Rochester, MN, USA
| | - Naveen L Pereira
- Department of Cardiovascular Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Ciorsti MacIntyre
- Department of Molecular Pharmacology & Experimental Therapeutics, Windland Smith Rice Sudden Death Genomics Laboratory, Mayo Clinic, Rochester, MN, USA
- Department of Cardiovascular Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Jay W Schneider
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, USA
| | - Michael J Ackerman
- Department of Molecular Pharmacology & Experimental Therapeutics, Windland Smith Rice Sudden Death Genomics Laboratory, Mayo Clinic, Rochester, MN, USA
- Department of Cardiovascular Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
- Department of Pediatric and Adolescent Medicine/Division of Pediatric Cardiology, Windland Smith Rice Genetic Heart Rhythm Clinic, Mayo Clinic, Rochester, MN, USA
| | - John R Giudicessi
- Department of Molecular Pharmacology & Experimental Therapeutics, Windland Smith Rice Sudden Death Genomics Laboratory, Mayo Clinic, Rochester, MN, USA.
- Department of Cardiovascular Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
| |
Collapse
|
9
|
Shameem M, Olson SL, Marron Fernandez de Velasco E, Kumar A, Singh BN. Cardiac Fibroblasts: Helping or Hurting. Genes (Basel) 2025; 16:381. [PMID: 40282342 PMCID: PMC12026832 DOI: 10.3390/genes16040381] [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: 02/28/2025] [Revised: 03/22/2025] [Accepted: 03/25/2025] [Indexed: 04/29/2025] Open
Abstract
Cardiac fibroblasts (CFs) are the essential cell type for heart morphogenesis and homeostasis. In addition to maintaining the structural integrity of the heart tissue, muscle fibroblasts are involved in complex signaling cascades that regulate cardiomyocyte proliferation, migration, and maturation. While CFs serve as the primary source of extracellular matrix proteins (ECM), tissue repair, and paracrine signaling, they are also responsible for adverse pathological changes associated with cardiovascular disease. Following activation, fibroblasts produce excessive ECM components that ultimately lead to fibrosis and cardiac dysfunction. Decades of research have led to a much deeper understanding of the role of CFs in cardiogenesis. Recent studies using the single-cell genomic approach have focused on advancing the role of CFs in cellular interactions, and the mechanistic implications involved during cardiovascular development and disease. Arguably, the unique role of fibroblasts in development, tissue repair, and disease progression categorizes them into the friend or foe category. This brief review summarizes the current understanding of cardiac fibroblast biology and discusses the key findings in the context of development and pathophysiological conditions.
Collapse
Affiliation(s)
- Mohammad Shameem
- Department of Rehabilitation Medicine, University of Minnesota, Minneapolis, MN 55455, USA;
| | - Shelby L. Olson
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN 55455, USA;
| | | | - Akhilesh Kumar
- Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA;
- Stem Cell Institute, University of Minnesota, Minneapolis, MN 55455, USA
| | - Bhairab N. Singh
- Department of Rehabilitation Medicine, University of Minnesota, Minneapolis, MN 55455, USA;
- Stem Cell Institute, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
10
|
Shepherd J. Biomimetic Approaches in the Development of Optimised 3D Culture Environments for Drug Discovery in Cardiac Disease. Biomimetics (Basel) 2025; 10:204. [PMID: 40277603 PMCID: PMC12024959 DOI: 10.3390/biomimetics10040204] [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: 12/31/2024] [Revised: 03/09/2025] [Accepted: 03/21/2025] [Indexed: 04/26/2025] Open
Abstract
Cardiovascular disease remains the leading cause of death worldwide, yet despite massive investment in drug discovery, the progress of cardiovascular drugs from lab to clinic remains slow. It is a complex, costly pathway from drug discovery to the clinic and failure becomes more expensive as a drug progresses along this pathway. The focus has begun to shift to optimisation of in vitro culture methodologies, not only because these must be undertaken are earlier on in the drug discovery pathway, but also because the principles of the 3Rs have become embedded in national and international legislation and regulation. Numerous studies have shown myocyte cell behaviour to be much more physiologically relevant in 3D culture compared to 2D culture, highlighting the advantages of using 3D-based models, whether microfluidic or otherwise, for preclinical drug screening. This review aims to provide an overview of the challenges in cardiovascular drug discovery, the limitations of traditional routes, and the successes in the field of preclinical models for cardiovascular drug discovery. It focuses on the particular role biomimicry can play, but also the challenges around implementation within commercial drug discovery.
Collapse
Affiliation(s)
- Jenny Shepherd
- School of Engineering, University of Leicester, Leicester LE1 7RH, UK
| |
Collapse
|
11
|
Li D, Ma Q. Ubiquitin-specific protease: an emerging key player in cardiomyopathy. Cell Commun Signal 2025; 23:143. [PMID: 40102846 PMCID: PMC11921692 DOI: 10.1186/s12964-025-02123-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 02/23/2025] [Indexed: 03/20/2025] Open
Abstract
Protein quality control (PQC) plays a vital role in maintaining normal heart function, as cardiomyocytes are relatively sensitive to misfolded or damaged proteins, which tend to accumulate under pathological conditions. Ubiquitin-specific protease (USP) is the largest deubiquitinating enzyme family and a key component of the ubiquitin proteasome system (UPS), which is a non-lysosomal protein degradation machinery to mediate PQC in cells. USPs regulate the stability or activity of the target proteins that involve intracellular signaling, transcriptional control of inflammation, antioxidation, and cell growth. Recent studies demonstrate that the USPs can regulate fibrosis, lipid metabolism, glucose homeostasis, hypertrophic response, post-ischemic recovery and cell death such as apoptosis and ferroptosis in cardiomyocytes. Since myocardial cell loss is an important component of cardiomyopathy, therefore, these findings suggest that the UPSs play emerging roles in cardiomyopathy. This review briefly summarizes recent literature on the regulatory roles of USPs in the occurrence and development of cardiomyopathy, giving us new insights into the molecular mechanisms of USPs in different cardiomyopathy and potential preventive strategies for cardiomyopathy.
Collapse
Affiliation(s)
- Danlei Li
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan Province, China
| | - Qilin Ma
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, Hunan Province, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan Province, China.
| |
Collapse
|
12
|
Tsampras T, Antonopoulos A, Kasiakogias A, Mika A, Kolovou A, Papadimitriou E, Lazaros G, Tsioufis K, Vlachopoulos C. Cardiac Magnetic Resonance to Reclassify Diagnosis and Detect Cardiomyopathies in Hospitalized Patients with Acute Presentation. Life (Basel) 2025; 15:470. [PMID: 40141814 PMCID: PMC11944084 DOI: 10.3390/life15030470] [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: 02/27/2025] [Revised: 03/12/2025] [Accepted: 03/13/2025] [Indexed: 03/28/2025] Open
Abstract
BACKGROUND Cardiomyopathies are a significant cause of heart failure, arrhythmia, and cardiac morbidity in the general population. Cardiovascular magnetic resonance (CMR) is a valuable tool for the diagnostic work-up of patients with acute cardiac events. OBJECTIVES This study evaluated the diagnostic value of CMR and the yield of cardiomyopathies in hospitalized cardiac patients with acute presentation. METHODS A retrospective analysis was conducted with 535 consecutive hospitalized patients who underwent CMR at Hippokration Hospital, Athens, Greece, to identify a subset of scans performed on an urgent basis of hospitalized patients. Demographic data, causes of admission, CMR findings, and plasma cardiac biomarkers (hs-Troponin I, NT-proBNP, and CRP) were systematically recorded. RESULTS Out of the initial 535 CMR scans evaluated, a further analysis was conducted with 104 patients who were in hospital and underwent CMR on an urgent basis. From the total population of hospitalized patients, 33% had CMR findings indicative of underlying cardiomyopathy, with dilated cardiomyopathy being the most common subtype (36%), followed by arrhythmogenic cardiomyopathy (27%), hypertrophic cardiomyopathy (15%), or other subtypes (e.g., cardiac amyloidosis, sarcoidosis, endomyocardial fibrosis, EGPA, or unclassified). CMR led to the reclassification of the initial diagnosis into that of underlying cardiomyopathy in 32% of cases. The highest reclassification rate was observed within the subgroup with heart failure (71%), followed by that of acute myocardial infarction/ischemic heart disease (24%) and myocarditis (22%). CONCLUSIONS CMR imaging effectively contributed to the differential diagnosis of hospitalized patients with acute cardiac events that remained without a definitive diagnosis after their initial work-up and uncovered underlying cardiomyopathy in almost one-third of this cohort.
Collapse
Affiliation(s)
- Theodoros Tsampras
- 1st Cardiology Department, Hippokration Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (T.T.); (A.K.); (G.L.); (K.T.); (C.V.)
| | - Alexios Antonopoulos
- 1st Cardiology Department, Hippokration Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (T.T.); (A.K.); (G.L.); (K.T.); (C.V.)
| | - Alexandros Kasiakogias
- 1st Cardiology Department, Hippokration Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (T.T.); (A.K.); (G.L.); (K.T.); (C.V.)
| | - Alexia Mika
- Radiology Department, Hippokration Hospital, 11527 Athens, Greece
| | - Antonia Kolovou
- Radiology Department, Hippokration Hospital, 11527 Athens, Greece
| | | | - George Lazaros
- 1st Cardiology Department, Hippokration Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (T.T.); (A.K.); (G.L.); (K.T.); (C.V.)
| | - Konstantinos Tsioufis
- 1st Cardiology Department, Hippokration Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (T.T.); (A.K.); (G.L.); (K.T.); (C.V.)
| | - Charalambos Vlachopoulos
- 1st Cardiology Department, Hippokration Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (T.T.); (A.K.); (G.L.); (K.T.); (C.V.)
| |
Collapse
|
13
|
Tan K, Tay D, Tan W, Ng HK, Wong E, Morley MP, Singhera GK, Lee CJM, Jain PR, Tai FL, Hanson PJ, Cappola TP, Margulies KB, Foo R, Loh M. Epigenome-wide association study for dilated cardiomyopathy in left ventricular heart tissue identifies putative gene sets associated with cardiac pathology and early indicators of cardiac risk. Clin Epigenetics 2025; 17:45. [PMID: 40057770 PMCID: PMC11890527 DOI: 10.1186/s13148-025-01854-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 02/28/2025] [Indexed: 05/13/2025] Open
Abstract
BACKGROUND Methylation changes linked to dilated cardiomyopathy (DCM) affect cardiac gene expression. We investigate DCM mechanisms regulated by CpG methylation using multi-omics and causal analyses in the largest cohort of left ventricular tissues available. METHODS We mapped DNA methylation at ~ 850,000 CpG sites, performed array-based genotyping and conducted RNA sequencing on left ventricular tissue samples from failing and non-failing hearts across two independent DCM cohorts (discovery n = 329, replication n = 85). Summary-data-based Mendelian Randomisation (SMR) was applied to explore the causal contribution of sentinel CpGs to DCM. Fine-mapping of regions surrounding sentinel CpGs revealed additional signals for cardiovascular disease risk factors. Coordinated changes across multiple CpG sites were examined using weighted gene co-expression network analysis (WGCNA). RESULTS We identified 194 epigenome-wide significant CpGs associated with DCM (discovery P < 5.96E-08), enriched in active chromatin states in heart tissue. Amongst these, 32 sentinel CpGs significantly influenced the expression of 30 unique proximal genes (± 1 Mb). SMR suggested the causal contribution of two sentinel CpGs to DCM and two other sentinel CpGs to the expression of two unique proximal genes (P < 0.05). For one sentinel CpG, colocalisation analyses provided suggestive evidence for a single causal variant underlying the methylation-gene expression relationship. Fine-mapping revealed additional signals linked to cardiovascular disease-relevant traits, including creatinine levels and the Framingham Risk Score. Co-methylation modules were enriched in gene sets and transcriptional regulators related to cardiac physiological and pathological processes, as well as in transcriptional regulators whose cardiac relevance has yet to be determined. CONCLUSIONS Using the largest series of left ventricular tissue to date, this study investigates the causal role of cardiac methylation changes in DCM and suggests targets for experimental studies to probe DCM pathogenesis.
Collapse
Affiliation(s)
- Konstanze Tan
- Lee Kong Chian School of Medicine, Nanyang Technological University, Clinical Sciences Building, 11 Mandalay Road, Singapore, 308232, Singapore
| | - Darwin Tay
- Lee Kong Chian School of Medicine, Nanyang Technological University, Clinical Sciences Building, 11 Mandalay Road, Singapore, 308232, Singapore
| | - Wilson Tan
- Cardiovascular Research Institute, National University Health System, Singapore, Singapore
| | - Hong Kiat Ng
- Lee Kong Chian School of Medicine, Nanyang Technological University, Clinical Sciences Building, 11 Mandalay Road, Singapore, 308232, Singapore
| | - Eleanor Wong
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Michael P Morley
- Penn Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Gurpreet K Singhera
- Bruce McManus Cardiovascular Biobank, UBC- Centre for Heart Lung Innovation, Vancouver, BC, Canada
| | - Chang Jie Mick Lee
- Cardiovascular-Metabolic Disease Translational Research Programme, National University of Singapore, Singapore, Singapore
| | - Pritesh R Jain
- Lee Kong Chian School of Medicine, Nanyang Technological University, Clinical Sciences Building, 11 Mandalay Road, Singapore, 308232, Singapore
| | - Fei Li Tai
- Lee Kong Chian School of Medicine, Nanyang Technological University, Clinical Sciences Building, 11 Mandalay Road, Singapore, 308232, Singapore
| | - Paul J Hanson
- Bruce McManus Cardiovascular Biobank, UBC- Centre for Heart Lung Innovation, Vancouver, BC, Canada
| | - Thomas P Cappola
- Penn Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kenneth B Margulies
- Penn Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Roger Foo
- Cardiovascular Research Institute, National University Health System, Singapore, Singapore
| | - Marie Loh
- Lee Kong Chian School of Medicine, Nanyang Technological University, Clinical Sciences Building, 11 Mandalay Road, Singapore, 308232, Singapore.
| |
Collapse
|
14
|
Bergan N, Prachee I, Curran L, McGurk KA, Lu C, de Marvao A, Bai W, Halliday BP, Gregson J, O’Regan DP, Ware JS, Tayal U. Systematic Review, Meta-Analysis, and Population Study to Determine the Biologic Sex Ratio in Dilated Cardiomyopathy. Circulation 2025; 151:442-459. [PMID: 39895490 PMCID: PMC11827689 DOI: 10.1161/circulationaha.124.070872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 01/08/2025] [Indexed: 02/04/2025]
Abstract
BACKGROUND Dilated cardiomyopathy (DCM) appears to be diagnosed twice as often in male than in female patients. This could be attributed to underdiagnosis in female patients or sex differences in susceptibility. Up to 30% of cases have an autosomal dominant monogenic cause, where equal sex prevalence would be expected. The aim of this systematic review, meta-analysis, and population study was to assess the sex ratio in patients with DCM, stratified by genetic status, and evaluate whether this is influenced by diagnostic bias. METHODS A literature search identified DCM patient cohorts with discernible sex ratios. Exclusion criteria were studies with a small (n<100), pediatric, or peripartum population. Meta-analysis and metaregression compared the proportion of female participants for an overall DCM cohort and the following subtypes: all genetic DCM, individual selected DCM genes (TTN and LMNA), and gene-elusive DCM. Population DCM sex ratios generated from diagnostic codes were also compared with those from sex-specific means using the UK Biobank imaging cohort; this established ICD coded, novel imaging-first, and genotype first determined sex ratios. RESULTS A total of 99 studies, with 37 525 participants, were included. The overall DCM cohort had a 0.30 female proportion (95% CI, 0.28-0.32), corresponding to a male:female ratio (M:F) of 2.38:1. This was similar to patients with an identified DCM variant (0.31 [95% CI, 0.26-0.36]; M:F 2.22:1; P=0.56). There was also no significant difference when compared with patients with gene-elusive DCM (0.30 [95% CI, 0.24-0.37]; M:F 2.29:1; P=0.81). Furthermore, the ratio within autosomal dominant gene variants was not significantly different for TTN (0.28 [95% CI, 0.22-0.36]; M:F 2.51:1; P=0.82) or LMNA (0.35 [95% CI, 0.27-0.44]; M:F 1.84:1; P=0.41). Overall, the sex ratio for DCM in people with disease attributed to autosomal dominant gene variants was similar to the all-cause group (0.34 [95% CI, 0.28-0.40]; M:F 1.98:1; P=0.19). In the UK Biobank (n=47 549), DCM defined by International Classification of Diseases, 10th revision, coding had 4.5:1 M:F. However, implementing sex-specific imaging-first and genotype-first diagnostic approaches changed this to 1.7:1 and 2.3:1, respectively. CONCLUSIONS This study demonstrates that DCM is twice as prevalent in male patients. This was partially mitigated by implementing sex-specific DCM diagnostic criteria. The persistent male excess in genotype-positive patients with an equally prevalent genetic risk suggests additional genetic or environmental drivers for sex-biased penetrance. REGISTRATION URL: https://www.crd.york.ac.uk/prospero; Unique identifier: CRD42023451944.
Collapse
Affiliation(s)
- Natalie Bergan
- National Heart Lung Institute, Imperial College London, UK (N.B., L.C., K.A.M., B.P.H., J.S.W., U.T.)
| | - Ishika Prachee
- Royal Brompton & Harefield Hospitals, Guy’s and St Thomas’ NHS Foundation Trust, London, UK (I.P., B.P.H., J.S.W., U.T.)
| | - Lara Curran
- National Heart Lung Institute, Imperial College London, UK (N.B., L.C., K.A.M., B.P.H., J.S.W., U.T.)
| | - Kathryn A. McGurk
- National Heart Lung Institute, Imperial College London, UK (N.B., L.C., K.A.M., B.P.H., J.S.W., U.T.)
- MRC Laboratory of Medical Sciences, London, UK (K.A.M., C.L., A.d.M., D.P.O., J.S.W.)
| | - Chang Lu
- MRC Laboratory of Medical Sciences, London, UK (K.A.M., C.L., A.d.M., D.P.O., J.S.W.)
| | - Antonio de Marvao
- MRC Laboratory of Medical Sciences, London, UK (K.A.M., C.L., A.d.M., D.P.O., J.S.W.)
- Department of Women and Children’s Health and British Heart Foundation Centre of Research Excellence, School of Cardiovascular and Metabolic Medicine and Sciences, King’s College London, UK (A.d.M.)
| | - Wenjia Bai
- Biomedical Image Analysis Group, Department of Computing, London, UK (W.B.)
- Department of Brain Sciences, London, UK (W.B.)
| | - Brian P. Halliday
- National Heart Lung Institute, Imperial College London, UK (N.B., L.C., K.A.M., B.P.H., J.S.W., U.T.)
- Royal Brompton & Harefield Hospitals, Guy’s and St Thomas’ NHS Foundation Trust, London, UK (I.P., B.P.H., J.S.W., U.T.)
| | - John Gregson
- London School of Hygiene and Tropical Medicine, UK (J.G.)
| | - Declan P. O’Regan
- Institute of Clinical Sciences, London, UK (D.P.O.)
- MRC Laboratory of Medical Sciences, London, UK (K.A.M., C.L., A.d.M., D.P.O., J.S.W.)
| | - James S. Ware
- National Heart Lung Institute, Imperial College London, UK (N.B., L.C., K.A.M., B.P.H., J.S.W., U.T.)
- Imperial College Healthcare NHS Trust, London, UK (J.S.W.)
- MRC Laboratory of Medical Sciences, London, UK (K.A.M., C.L., A.d.M., D.P.O., J.S.W.)
- Royal Brompton & Harefield Hospitals, Guy’s and St Thomas’ NHS Foundation Trust, London, UK (I.P., B.P.H., J.S.W., U.T.)
| | - Upasana Tayal
- National Heart Lung Institute, Imperial College London, UK (N.B., L.C., K.A.M., B.P.H., J.S.W., U.T.)
- Royal Brompton & Harefield Hospitals, Guy’s and St Thomas’ NHS Foundation Trust, London, UK (I.P., B.P.H., J.S.W., U.T.)
- The George Institute for Global Health, UK (U.T.)
| |
Collapse
|
15
|
Garcia YE, Sjögren B, Osei-Owusu P. G protein regulation by RGS proteins in the pathophysiology of dilated cardiomyopathy. Am J Physiol Heart Circ Physiol 2025; 328:H348-H360. [PMID: 39772618 PMCID: PMC12103878 DOI: 10.1152/ajpheart.00653.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 12/23/2024] [Accepted: 12/24/2024] [Indexed: 01/11/2025]
Abstract
Regulators of G protein signaling (RGS) proteins fine-tune signaling via heterotrimeric G proteins to maintain physiologic homeostasis in various organ systems of the human body including the brain, kidney, heart, and vasculature. Impaired regulation of G protein signaling by RGS proteins is implicated in the pathogenesis of several human diseases including various forms of cardiomyopathy such as hypertrophic cardiomyopathy and dilated cardiomyopathy (DCM). Both genetic and nongenetic changes that impinge on G protein signaling in cardiomyocytes are implicated in the etiology of DCM, and there is accumulating evidence that such genetic and nongenetic changes affecting G protein signaling in cell types other than cardiomyocytes could serve as a DCM trigger in humans. This review discusses and highlights mammalian RGS proteins and their roles in cardiac physiology and disease, with a specific focus on the current understanding of the etiology of DCM and the pathogenic roles of RGS proteins that are prominently expressed in the cardiovascular system. Growing evidence suggests that defects in G protein regulation by RGS proteins in the cardiovascular system likely contribute to cardiomyocyte structural damage and decreased contractile function that hallmark DCM. Further studies that enhance the understanding of the dynamics of G protein regulation by RGS proteins in several cell types in the myocardium and the vasculature are critical to gaining more insight into the etiology of DCM and heart failure, and to the identification of novel therapeutic targets.
Collapse
Affiliation(s)
- Yadhira E Garcia
- Department of Pharmaceutical Sciences, University of California, Irvine, California, United States
| | - Benita Sjögren
- Department of Pharmaceutical Sciences, University of California, Irvine, California, United States
- Department of Biological Chemistry, University of California, Irvine, California, United States
| | - Patrick Osei-Owusu
- Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States
| |
Collapse
|
16
|
Lorca R, Alén A, Salgado M, Misiego-Margareto R, Dolado-Cuello J, Gómez J, Alonso V, Coto E, Avanzas P, Martínez-Hernández A, Suárez Mier MP. RBM20 p.Arg636Cys: A Pathogenic Variant Identified in a Family with Several Cases of Unexpected Sudden Deaths. J Clin Med 2025; 14:743. [PMID: 39941414 PMCID: PMC11818836 DOI: 10.3390/jcm14030743] [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: 01/02/2025] [Revised: 01/14/2025] [Accepted: 01/16/2025] [Indexed: 02/16/2025] Open
Abstract
Background: Dilated cardiomyopathy (DCM) can be an inherited condition related to premature sudden cardiac death (SCD). Pathogenic variants in some genes, like LMNA, SCN5A, FLNC or RBM20, have been linked to an increased risk of SCD. Although genetic study can help to stratify the arrhythmic risk, there are no specific guidelines for RBM20 carriers' management. We aimed to evaluate the genetic profile and clinical features of all DCM patients with pathogenic variants in RBM20.Methods: We identified all carriers of pathogenic variants in RBM20 in a single national center that specializes in inherited cardiac conditions. Forensic and molecular autopsies provided crucial information. Results: We identified a large family with inherited DCM due to RBM20 p.Arg636Cy and several SCDs. The proband was a 37-year-old male who suffered an unexpected SCD despite presenting a mild DCM phenotype with normal left ventricular ejection fraction. Family screening identified four other carriers, who were asymptomatic, but presented concealed mild DCM phenotypes. Family history revealed that six other relatives (two of them obligate carriers) had also suffered sudden deaths at young ages. Conclusions: We present an informative family with DCM, due to RBM20 p.Arg636Cys, and high rates of SCD, even in members with mild DCM phenotypes. ICD implantation to prevent SCD should be carefully evaluated in all RBM20 p.Arg636Cys carriers. Moreover, the frequent development of AF and HF progression requires specific awareness.
Collapse
Affiliation(s)
- Rebeca Lorca
- Área del Corazón, Hospital Universitario Central Asturias, 33011 Oviedo, Spain
- Unidad de Cardiopatías Familiares, Área del Corazón y Departamento de Genética Molecular, Hospital Universitario Central Asturias, 33011 Oviedo, Spain
- Departamento de Biología Funcional, Universidad de Oviedo, 33003 Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias, ISPA, 33011 Oviedo, Spain
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORs), 28029 Madrid, Spain
| | - Alberto Alén
- Área del Corazón, Hospital Universitario Central Asturias, 33011 Oviedo, Spain
| | - María Salgado
- Área del Corazón, Hospital Universitario Central Asturias, 33011 Oviedo, Spain
| | | | - Javier Dolado-Cuello
- Instituto de Medicina Legal y Ciencias Forenses de Asturias, 33011 Oviedo, Spain
| | - Juan Gómez
- Instituto de Investigación Sanitaria del Principado de Asturias, ISPA, 33011 Oviedo, Spain
| | - Vanesa Alonso
- Área del Corazón, Hospital Universitario Central Asturias, 33011 Oviedo, Spain
| | - Eliecer Coto
- Unidad de Cardiopatías Familiares, Área del Corazón y Departamento de Genética Molecular, Hospital Universitario Central Asturias, 33011 Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias, ISPA, 33011 Oviedo, Spain
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORs), 28029 Madrid, Spain
- Departamento de Medicina, Universidad de Oviedo, 33003 Oviedo, Spain
| | - Pablo Avanzas
- Área del Corazón, Hospital Universitario Central Asturias, 33011 Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias, ISPA, 33011 Oviedo, Spain
- Departamento de Medicina, Universidad de Oviedo, 33003 Oviedo, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | | | - María Paz Suárez Mier
- Histopathology Service, National Institute of Toxicology and Forensic Sciences, 28232 Madrid, Spain;
| |
Collapse
|
17
|
Wang Z, Chen Y, Li W, Gao C, Zhang J, Zang X, Zhao Z, Fan H, Zhao Y. Identification and validation of diagnostic biomarkers and immune infiltration in dilated cardiomyopathies with heart failure and construction of diagnostic model. Gene 2025; 934:149007. [PMID: 39427832 DOI: 10.1016/j.gene.2024.149007] [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: 07/11/2024] [Revised: 09/14/2024] [Accepted: 10/15/2024] [Indexed: 10/22/2024]
Abstract
Dilated cardiomyopathy (DCM) is characterized by immune cell infiltration and can readily progress to heart failure (HF). In the study, differential expression analysis, enrichment analysis, and protein-protein interaction (PPI) network analysis were performed on DCM with HF-related datasets. The CytoHubba was used to identify hub genes. Diagnostic biomarkers were obtained by validating their expression and diagnostic value in another external dataset, and a diagnostic model was constructed. Finally, single-sample gene set enrichment analysis (ssGSEA) was used to predict immune cell infiltration in cardiac samples. The associations between diagnostic biomarkers and immune cells were investigated. The NetworkAnalyst and miRDB databases were used to predict transcription factors and microRNAs, followed by establishing regulatory networks. The DSigDB database was used to predict drug candidates. Subsequently, a mouse model of DCM with HF was used to validate the expression levels of these genes. The present study revealed that differentially expressed genes were enriched in the extracellular matrix organization, cardiac muscle hypertrophy, and other immune-related biological processes. OMD and THBS4 were finally identified, and the nomogram has satisfactory prediction and strong calibration ability. In addition, the two diagnostic biomarkers exhibited significant associations with multiple immune infiltrating cells. Finally, two TFs, 65 microRNAs, and 10 drug candidates were obtained. In animal experiments, two diagnostic biomarkers showed expression trends consistent with the results of bioinformatic analysis. OMD and THBS4 have been identified as hub immune-related diagnostic biomarkers for DCM with HF. Our research provides novel insights into the diagnosis and treatment of the disease.
Collapse
Affiliation(s)
- Zhaodi Wang
- Department of Cardiology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, 450000, China; Heart Center of Henan Provincial People's Hospital, Central China Fuwai Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, 450000, China
| | - Yihan Chen
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China
| | - Weidong Li
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China
| | - Chuanyu Gao
- Department of Cardiology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, 450000, China; Henan Provincial Key Lab for Control of Coronary Heart Disease, Zhengzhou University Central China Fuwai Hospital, Zhengzhou 450000, China
| | - Jing Zhang
- Department of Cardiology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, 450000, China; Heart Center of Henan Provincial People's Hospital, Central China Fuwai Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, 450000, China
| | - Xiaobiao Zang
- Heart Center of Henan Provincial People's Hospital, Central China Fuwai Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, 450000, China
| | - Zhihan Zhao
- Heart Center of Henan Provincial People's Hospital, Central China Fuwai Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, 450000, China
| | - Hongkun Fan
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China.
| | - Yonghui Zhao
- Department of Cardiology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, 450000, China; Heart Center of Henan Provincial People's Hospital, Central China Fuwai Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, 450000, China.
| |
Collapse
|
18
|
Jiang F, Tang J, Wei X, Pan H, Fan X, Zhang P, Guo S. BMP6, a potential biomarker of inflammatory fibrosis and promising protective factor for dilated cardiomyopathy. Chin Med 2025; 20:12. [PMID: 39825396 PMCID: PMC11740616 DOI: 10.1186/s13020-025-01062-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 01/03/2025] [Indexed: 01/20/2025] Open
Abstract
BACKGROUND Dilated cardiomyopathy (DCM) stands as one of the most prevalent and severe causes of heart failure. Inflammation plays a pivotal role throughout the progression of DCM to heart failure, while age acts as a natural predisposing factor for all cardiovascular diseases. These two factors often interact, contributing to cardiac fibrosis, which is both a common manifestation and a pathogenic driver of adverse remodeling in DCM-induced heart failure. METHOD Bulk RNA-seq, single-cell RNA-seq, Mendelian randomization analysis, animal model construction, and BMP6 knockdown were utilized to identify and validate potential specific markers and targets for intervention in DCM heart failure. RESULTS We found that DCM hearts exhibit pronounced inflammatory cell infiltration and fibrosis. Both bulk RNA-seq and single-cell RNA-seq analyses revealed aberrant BMP6 expression specifically in fibroblasts. The ROC curve underscores the high specificity of BMP6 in relation to DCM, while Mendelian randomization analysis further confirms BMP6 as a protective factor against DCM. Notably, BMP6 knockdown led to a decrease in SMAD6 expression and a marked elevation in COL1A1 expression levels, indicating its antifibrotic role. CONCLUSION BMP6 emerges as a promising biomarker for DCM, and its functional role in exerting an antifibrotic effect underscores its potential as a therapeutic target.
Collapse
Affiliation(s)
- Feng Jiang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Jiayang Tang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Xiaoqi Wei
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Hai Pan
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Xinyi Fan
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Peng Zhang
- Wuhan Hospital of Traditional Chinese Medicine, Wuhan, 430014, China.
| | - Shuzhen Guo
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 102488, China.
| |
Collapse
|
19
|
Li L, Xie S, Deng W. RNA binding proteins: Mechanistic considerations and perspectives in controlling cardiovascular diseases. Eur J Pharmacol 2025; 987:177101. [PMID: 39613174 DOI: 10.1016/j.ejphar.2024.177101] [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: 07/14/2024] [Revised: 11/01/2024] [Accepted: 11/05/2024] [Indexed: 12/01/2024]
Abstract
Cardiovascular diseases (CVDs) are becoming serious disease that endangering human health due to the increasing morbidity and mortality, and many molecular targets are involved in this complex pathologic process. Recently, RNA-binding proteins (RBPs) have received potential attention as a promising targets for preventing CVDs, including myocardial hypertrophy, dilated cardiomyopathy (DCM), myocardial fibrosis, and pulmonary hypertension (PH). As important regulators of RNA metabolism, RBPs play important roles in all steps of the gene expression cascade,and affect the occurrence and development of various diseases. In this review, we discuss the regulatory role of RBPs on various CVDs at the post transcriptional modification level based on current research. We also highlight the existing and potential RNA-based therapeutics that could impact future CVD treatments.
Collapse
Affiliation(s)
- Lanlan Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, China; Department of Endocrinology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute of Wuhan University, Wuhan, 430060, China
| | - Saiyang Xie
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, China; Department of Endocrinology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute of Wuhan University, Wuhan, 430060, China
| | - Wei Deng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, China; Department of Endocrinology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
20
|
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
|
21
|
Yang H, Wang Z, Xu Y, Du Y, Yang H, Lu Y. Prognostic signature and therapeutic drug identification for dilated cardiomyopathy based on necroptosis via bioinformatics and experimental validation. Sci Rep 2025; 15:319. [PMID: 39747333 PMCID: PMC11696111 DOI: 10.1038/s41598-024-83455-8] [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: 07/31/2024] [Accepted: 12/16/2024] [Indexed: 01/04/2025] Open
Abstract
Necroptosis, a type of programmed cell death, has been increasingly linked to cardiovascular disease development, yet its role in dilated cardiomyopathy (DCM) remains unclear. In this study, we analyzed the GSE5406 dataset from the GEO database to explore necroptosis-related prognostic signatures in DCM using LASSO regression. We identified five necroptosis-related genes (BID, CAMK2B, GLUL, HSP90AB1, CHMP5) that define a necroptosis-related signature with strong predictive value, evidenced by ROC curve areas of 0.852 and 0.957 in training and test sets, respectively. Our analyses, including GO and GSEA enrichment, focused on pathways associated with high necroptosis-related scores (NRS) and revealed significant immune cell infiltration. Notably, nTreg and iTreg cells were enriched in the high NRS group, while CD8 naive T cells and CD8 T cells positively correlated with NRS. Small molecule drugs fenofibrate, procyclidine, and tienilic acid emerged as potential therapeutic agents for high-risk patients, with fenofibrate showing efficacy in inhibiting DCM progression in an inflammatory animal model. These findings underscore the clinical relevance of necroptosis-related genes in assessing DCM progression and prognosis and highlight their potential for targeted therapeutic development.
Collapse
Affiliation(s)
- Han Yang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhenwei Wang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yawei Xu
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yimei Du
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Haibo Yang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Yang Lu
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
22
|
Qin S, Zhu C, Chen C, Sheng Z, Cao Y. An emerging double‑edged sword role of ferroptosis in cardiovascular disease (Review). Int J Mol Med 2025; 55:16. [PMID: 39540363 PMCID: PMC11573318 DOI: 10.3892/ijmm.2024.5457] [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: 09/02/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
The pathophysiology of cardiovascular disease (CVD) is complex and presents a serious threat to human health. Cardiomyocyte loss serves a pivotal role in both the onset and progression of CVD. Among various forms of programmed cell death, ferroptosis, along with apoptosis, autophagy and pyroptosis, is closely linked to the advancement of CVD. Ferroptosis, a mechanism of cell death, is driven by the buildup of oxidized lipids and excess iron. This pathway is modulated by lipid, amino acid and iron metabolism. Key characteristics of ferroptosis include disrupted iron homeostasis, increased peroxidation of polyunsaturated fatty acids due to reactive oxygen species, decreased glutathione levels and inactivation of glutathione peroxidase 4. Treatments targeting ferroptosis could potentially prevent or alleviate CVD by inhibiting the ferroptosis pathway. Ferroptosis is integral to the pathogenesis of several types of CVD and inhibiting its occurrence in cardiomyocytes could be a promising therapeutic strategy for the future treatment of CVD. The present review provided an in‑depth analysis of advancements in understanding the mechanisms underlying ferroptosis. The present manuscript summarized the interplay between ferroptosis and CVDs, highlighting its dual roles in these conditions. Additionally, potential therapeutic targets within the ferroptosis pathway were discussed, alongside the current limitations and future directions of these novel treatment strategies. The present review may offer novel insights into preventive and therapeutic approaches for CVDs.
Collapse
Affiliation(s)
- Sirun Qin
- Department of Cardiovascular Medicine, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Can Zhu
- Department of Cardiovascular Medicine, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Chenyang Chen
- Department of Cardiovascular Medicine, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Zhe Sheng
- Department of Cardiovascular Medicine, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Yu Cao
- Department of Cardiovascular Medicine, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| |
Collapse
|
23
|
Hao J, Li W, Jiao W, Li F, Xie Y. IL-17 affects the immune regulation of CD4+ T cells in dilated cardiomyopathy through JAK/STAT pathway. Cardiol Young 2025; 35:16-23. [PMID: 39757542 DOI: 10.1017/s104795112402688x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2025]
Abstract
PURPOSE Studying the effect of interleukin-17 (IL-17) on the mechanism of CD4+ T-cell immune regulation and the Janus kinase/signal transducer and activator of transcription (JAK/STAT) pathway may offer new ideas and methods for the therapy of dilated cardiomyopathy. METHODS Naive CD4+ T cells were isolated from mice using a magnetic bead sorting reagent and manipulated by overexpression or knockdown of IL-17. Protein levels of Janus kinase 2 (JAK2), phosphorylated JAK2 (p-JAK2), signal transducer and activator of transcription 3 (STAT3), phosphorylated STAT3 (p-STAT3), matrix metalloproteinase-2 (MMP-2), and matrix metalloproteinase-9 (MMP-9) were determined by Western blotting. Quantitative polymerase chain reaction was used to assess the levels of JAK2, STAT3, MMP-2, and MMP-9. Expression of tumour necrosis factor-alpha (TNF-α), interleukin-1 beta (IL-1β), interleukin-4 (IL-4), and interferon-gamma (IFNγ) was determined by enzyme-linked immunosorbent assay test kits. TNF-α, IL-1β, IL-4, and IFNγ secretion was measured by flow cytometry. RESULTS In CD4+ T cells, IL-17 overexpression increased TNF-α, IL-1β, IL-4, IFNγ, p-JAK2, p-STAT3, MMP-2, MMP-9 levels, and apoptosis. Knockdown of IL-17 reduced the levels of TNF-α, IL-1β, IL-4, IFNγ, p-JAK2, p-STAT3, MMP-2, and MMP-9, as well as the level of apoptosis. CONCLUSION Through regulation of IL-17 expression in CD4+ T cells, this study reveals its crucial role in regulating the secretion of inflammatory factors, activation of the JAK/STAT signaling pathway, expression of matrix metalloproteinases, and apoptosis of CD4+ T cells.
Collapse
Affiliation(s)
- Jie Hao
- Department of Cardiovascular, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Weiwei Li
- Department of Cardiovascular, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Wei Jiao
- Department of Cardiovascular, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Fang Li
- Department of Cardiovascular, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yanan Xie
- Department of Cardiovascular, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
24
|
Tang Q, Meng X, Tu X, Zhang J. Mendelian Randomization Study on the Associations Between Genetically Predicted Cardiovascular Disease Subtypes and the Risk of Developing Cardiomyopathies. Clin Appl Thromb Hemost 2025; 31:10760296251328011. [PMID: 40152048 PMCID: PMC11951890 DOI: 10.1177/10760296251328011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Revised: 02/15/2025] [Accepted: 02/28/2025] [Indexed: 03/29/2025] Open
Abstract
Cardiomyopathies are commonly believed to have genetic origins; however, the connection between cardiomyopathies and cardiovascular diseases remains uncertain. Thus, we employed a Mendelian randomization (MR) approach to investigate the potential causal effects of specific cardiovascular disease subtypes on dilated and hypertrophic cardiomyopathies, focusing primarily on a European population. Summary-level data for cardiomyopathies and other cardiovascular diseases were obtained from public genome-wide association studies. Random-effects inverse-variance weighting was used as the primary analysis, whereas sensitivity analyses, including weighted median, MR-Egger, and multivariable MR methods, were also conducted. A genetic predisposition to atrial fibrillation [odds ratio (OR): 1.33; 95% confidence interval (CI): 1.18-1.50; P < 0.001], heart failure (OR: 3.22; 95% CI: 1.92-5.41; P < 0.001), and hypertension (OR: 1.50; 95% CI: 1.25-1.81; P < 0.001) were causally linked to an increased risk of developing dilated cardiomyopathy. However, there was no direct causal connection between genetically predicted coronary heart disease, pulmonary embolism, or ischemic stroke and the risk of developing dilated cardiomyopathy. In contrast, no significant associations were found between genetically predicted CVD subtypes and the risk of developing hypertrophic cardiomyopathy. Genetically predicted heart failure is significantly associated with the risk of developing dilated cardiomyopathy, underscoring the importance of effective heart failure management for risk prevention. Moreover, individuals with hypertension and atrial fibrillation might have an increased predisposition to dilated cardiomyopathy, highlighting crucial implications for management.
Collapse
Affiliation(s)
- Qiaolin Tang
- Department of Cardiology, Jiangxi Province Hospital of Integrated Chinese and Western Medicine, Nanchang, China
| | - Xiangzhu Meng
- Department of Cardiology, Jiangxi Province Hospital of Integrated Chinese and Western Medicine, Nanchang, China
| | - Xiaowen Tu
- Department of Cardiology, Jiangxi Province Hospital of Integrated Chinese and Western Medicine, Nanchang, China
| | - Jian Zhang
- Department of Cardiology, Jiangxi Province Hospital of Integrated Chinese and Western Medicine, Nanchang, China
| |
Collapse
|
25
|
Esteban-Fernández A, Anguita-Sánchez M, Rosillo N, Bernal Sobrino JL, Del Prado N, Fernández-Pérez C, Rodríguez-Padial L, Elola Somoza FJ. Comprehensive analysis of clinical characteristics, management, and prognosis in patients with dilated cardiomyopathy discharged from Spanish hospitals. Hellenic J Cardiol 2024:S1109-9666(24)00268-9. [PMID: 39710048 DOI: 10.1016/j.hjc.2024.12.005] [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: 09/20/2024] [Revised: 12/15/2024] [Accepted: 12/16/2024] [Indexed: 12/24/2024] Open
Abstract
OBJECTIVE Dilated cardiomyopathy (DCM) is a leading cause of heart failure (HF) characterized by left ventricular dilatation and systolic dysfunction not explained by abnormal loading conditions. Despite its prevalence, DCM's epidemiology and prognosis remain poorly studied in our country. METHODS A retrospective observational study encompassed patients discharged from all Spanish public hospitals between 2016 and 2021 diagnosed with DCM. Data were extracted from the Minimum Basic Data Set. The study focused on hospital admissions, comorbidities, in-hospital mortality, and readmission rates for circulatory system diseases at 30 and 365 days. RESULTS Among 27,402 index episodes, DCM was the primary diagnosis in 12.4%, predominantly affecting men (72.5%). In-hospital mortality was 8.7%, with significant predictors including cardiogenic shock (OR: 12.4, 95% CI: 9.6-15.9), advanced or metastatic cancer (OR: 4.3, 95% CI: 3.8-5.0), renal failure (OR: 2.4, 95% CI: 2.2-2.7), and chronic liver disease (OR: 2.4, 95% CI: 2.1-2.8). Readmission rates were 7.9% at 30 days and 25.5% at 365 days, predominantly due to HF. Multivariate analysis identified age (IRR: 1.02, 95% CI: 1.01-1.02), female sex (IRR: 0.87, 95% CI: 0.79-0.96), severe hematological diseases (IRR: 2.12, 95% CI: 1.45-3.10), and metastatic cancer (IRR: 1.65, 95% CI: 1.31-2.07) as predictors of 30-day readmissions. At 365 days, predictors included age (IRR: 1.02, 95% CI: 1.01-1.02), female sex (IRR: 0.80, 95% CI: 0.74-0.86), severe hematological diseases (IRR: 2.43, 95% CI: 1.66-3.56), and renal failure (IRR: 1.42, 95% CI: 1.31-1.55). CONCLUSION This study highlights the substantial hospitalization burden and mortality risk among DCM patients, emphasizing the necessity for advanced management strategies and specialized cardiac care.
Collapse
Affiliation(s)
- Alberto Esteban-Fernández
- Cardiology Department, Hospital Universitario Severo Ochoa, Leganés, Madrid, Spain; Faculty of Health Sciences, Universidad Alfonso X el Sabio (UAX), Villanueva de la Cañada, Madrid, Spain.
| | - Manuel Anguita-Sánchez
- Cardiology Department, Hospital Universitario Reina Sofía, Córdoba, Spain; The Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Cordoba University, Córdoba, Spain; Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Nicolás Rosillo
- Institute for the Improvement of Health Care (IMAS Foundation), Madrid, Spain; Preventive Medicine Department, Hospital Universitario 12 de Octubre, Madrid, Spain
| | | | - Náyade Del Prado
- Institute for the Improvement of Health Care (IMAS Foundation), Madrid, Spain
| | - Cristina Fernández-Pérez
- Institute for the Improvement of Health Care (IMAS Foundation), Madrid, Spain; Preventive Medicine Department, Área Sanitaria de Santiago y Barbanza, Instituto de Investigaciones Sanitarias de Santiago, Santiago de Compostela (A Coruña), Spain
| | | | | |
Collapse
|
26
|
Zhou G, Yu R, Luo C, Li P, Huang Z, Zhang B, Liu G, Zhong Y, Liang J. Association between comorbid COPD or chronic bronchitis and the prognosis of patients with Dilated cardiomyopathy. Ann Med 2024; 56:2428857. [PMID: 39550350 PMCID: PMC11571775 DOI: 10.1080/07853890.2024.2428857] [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: 08/24/2023] [Revised: 03/29/2024] [Accepted: 10/21/2024] [Indexed: 11/18/2024] Open
Abstract
AIMS Dilated cardiomyopathy (DCM) is characterized by unilateral or bilateral ventricular enlargement and reduced ventricular systolic function, with or without heart failure. In previous studies, we found that a history of chronic obstructive pulmonary disease (COPD) or chronic bronchitis is a high risk factor for DCM combined with pulmonary hypertension (PH). Therefore, we propose that the comorbidity of COPD or chronic bronchitis will increase the cardiogenic mortality of patients with DCM. METHODS Data were collected from patients with DCM who were admitted to The First Affiliated Hospital of Guangzhou University of Chinese Medicine from October 2008 to April 2020. The primary endpoint was cardiac death. Multivariable Cox regression analyses were employed to assess the associations between the comorbidities COPD or chronic bronchitis with the study endpoints. Different adjusting models were used to adjust for potential confounders. RESULTS A total of 305 DCM patients were ultimately enrolled, among whom 46 patients had COPD or chronic bronchitis. The median follow-up was 50 months. The rate of cardiac death in the COPD or chronic bronchitis group was significantly greater than that in the non-COPD or nonchronic bronchitis group (p < 0.001). The associations between comorbid COPD or chronic bronchitis and cardiac death remained robust after eliminating the possible effects of confounders. After grouping by PH, the risk difference was mainly derived from the intermediate- or high-probability PH group. CONCLUSIONS Comorbid COPD or chronic bronchitis increased the risk of cardiac death among DCM patients with an intermediate or high PH probability.
Collapse
MESH Headings
- Humans
- Cardiomyopathy, Dilated/epidemiology
- Cardiomyopathy, Dilated/mortality
- Cardiomyopathy, Dilated/complications
- Male
- Female
- Middle Aged
- Pulmonary Disease, Chronic Obstructive/epidemiology
- Pulmonary Disease, Chronic Obstructive/mortality
- Pulmonary Disease, Chronic Obstructive/complications
- Pulmonary Disease, Chronic Obstructive/physiopathology
- Bronchitis, Chronic/epidemiology
- Bronchitis, Chronic/complications
- Bronchitis, Chronic/physiopathology
- Prognosis
- Comorbidity
- Risk Factors
- Adult
- Aged
- China/epidemiology
- Hypertension, Pulmonary/epidemiology
- Hypertension, Pulmonary/mortality
- Proportional Hazards Models
- Retrospective Studies
Collapse
Affiliation(s)
- Guiting Zhou
- Cardiovascular Disease, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Runjia Yu
- Cardiovascular Disease, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Chuanjin Luo
- Cardiovascular Disease, Guangzhou University of Traditional Chinese Medicine First Affiliated Hospital, Guangzhou, China
| | - Ping Li
- The Department of Cardiovascular Disease, Meizhou Hospital of Traditional Chinese Medicine, Meizhou, China
| | - Zhihua Huang
- The Department of Cardiovascular Disease, Meizhou Hospital of Traditional Chinese Medicine, Meizhou, China
| | - Bo Zhang
- The Department of Cardiovascular Disease, Meizhou Hospital of Traditional Chinese Medicine, Meizhou, China
| | - Guangjiao Liu
- The Department of Cardiovascular Disease, Meizhou Hospital of Traditional Chinese Medicine, Meizhou, China
| | - Yueqiao Zhong
- The Department of Cardiovascular Disease, Meizhou Hospital of Traditional Chinese Medicine, Meizhou, China
| | - Jiahua Liang
- The Department of Cardiovascular Disease, Meizhou Hospital of Traditional Chinese Medicine, Meizhou, China
| |
Collapse
|
27
|
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
|
28
|
Montvilaitė-Laurinavičienė A, Dirsienė R, Neverauskaitė-Piliponienė G, Banišauskaitė A, Šukys M, Šakalytė G, Ereminienė E. Heart failure of very rare aetiology-haemochromatosis Type 3: a case report. Eur Heart J Case Rep 2024; 8:ytae637. [PMID: 39687529 PMCID: PMC11647590 DOI: 10.1093/ehjcr/ytae637] [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: 09/19/2024] [Revised: 10/23/2024] [Accepted: 11/22/2024] [Indexed: 12/18/2024]
Abstract
Background Haemochromatosis is a pathological condition characterized by the accumulation of iron in parenchymal organs, leading to toxic damage and dysfunction. Cardiac haemochromatosis represents one of the rare causes of severe heart failure (HF) that can be potentially prevented with targeted treatment. Case Summary We present the case of a 41-year-old female who was hospitalized for decompensated HF. Echocardiography revealed severe systolic dysfunction with a phenotype of dilated cardiomyopathy, accompanied by secondary moderate mitral regurgitation and severe tricuspid regurgitation (TR). To differentiate potential causes of HF, coronary angiography, cardiac magnetic resonance imaging (MRI), and endomyocardial biopsy were performed. Based on clinical findings, laboratory results, cardiac MRI, and endomyocardial biopsy data, a diagnosis of haemochromatosis was confirmed, and mutations in the TFR2 gene, responsible for haemochromatosis Type 3, were identified. The patient was treated in accordance with the latest European Society of Cardiology HF guidelines, and specific treatment for haemochromatosis, including therapeutic phlebotomy and iron chelation therapy, was initiated, resulting in a significant positive outcome. Discussion Investigating the aetiology of HF is essential, as even rare causes can be identified, and specific treatments are available that significantly improve prognosis and survival.
Collapse
Affiliation(s)
| | - Rūta Dirsienė
- Faculty of Medicine, Lithuanian University of Health Sciences, A. Mickevičiaus Str. 9, 44307 Kaunas, Lithuania
- Department of Cardiology, Lithuanian University of Health Sciences, Eivenių Str. 2, 50161 Kaunas, Lithuania
| | - Gintarė Neverauskaitė-Piliponienė
- Faculty of Medicine, Lithuanian University of Health Sciences, A. Mickevičiaus Str. 9, 44307 Kaunas, Lithuania
- Department of Cardiology, Lithuanian University of Health Sciences, Eivenių Str. 2, 50161 Kaunas, Lithuania
| | - Audra Banišauskaitė
- Faculty of Medicine, Lithuanian University of Health Sciences, A. Mickevičiaus Str. 9, 44307 Kaunas, Lithuania
- Department of Radiology, Lithuanian University of Health Sciences, Eivenių Str. 2, 50161 Kaunas, Lithuania
| | - Marius Šukys
- Faculty of Medicine, Lithuanian University of Health Sciences, A. Mickevičiaus Str. 9, 44307 Kaunas, Lithuania
- Department of Genetics and Laboratory Medicine, Lithuanian University of Health Sciences, Eivenių Str. 2, 50161 Kaunas, Lithuania
| | - Gintarė Šakalytė
- Faculty of Medicine, Lithuanian University of Health Sciences, A. Mickevičiaus Str. 9, 44307 Kaunas, Lithuania
- Department of Cardiology, Lithuanian University of Health Sciences, Eivenių Str. 2, 50161 Kaunas, Lithuania
- Institute of Cardiology, Lithuanian University of Health Sciences, Sukilėlių Av. 15, 50161 Kaunas, Lithuania
| | - Eglė Ereminienė
- Faculty of Medicine, Lithuanian University of Health Sciences, A. Mickevičiaus Str. 9, 44307 Kaunas, Lithuania
- Department of Cardiology, Lithuanian University of Health Sciences, Eivenių Str. 2, 50161 Kaunas, Lithuania
- Institute of Cardiology, Lithuanian University of Health Sciences, Sukilėlių Av. 15, 50161 Kaunas, Lithuania
| |
Collapse
|
29
|
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
|
30
|
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
|
31
|
Li J, Hu L, Huang X. Causal relationship between leukocyte telomere length and two cardiomyopathies based on a bidirectional Mendelian randomization approach. Medicine (Baltimore) 2024; 103:e40308. [PMID: 39533571 PMCID: PMC11556983 DOI: 10.1097/md.0000000000040308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 10/11/2024] [Indexed: 11/16/2024] Open
Abstract
This study aims to employ the Mendelian randomization (MR) approach to investigate the relationship between leukocyte telomere length (TL) and 2 prevalent forms of cardiomyopathies. Using R software (4.3.1) for MR study, independent genetic variants associated with leukocyte TL were extracted from the Integrative Epidemiology Unit database, while cardiomyopathies data were pooled from FinnGen and European Bioinformatics Institute databases. Analytical methodologies included inverse-variance weighting, MR-Egger regression, and weighted median methods. Further analyses involved MR-Egger intercept and MR-PRESSO for handling horizontal pleiotropy and Cochran Q test for study heterogeneity. Our forward Mendelian randomization study indicates a positive correlation between longer leukocyte TL and the risk of 2 forms of cardiomyopathies: the longer the leukocyte telomere, the higher is the risk of cardiomyopathies. Specifically, for hypertrophic obstructive cardiomyopathy the OR is 2.23 (95% CI: 1.19-4.14, P = .01), for hypertrophic cardiomyopathy the OR is 1.80 (95% CI: 1.14-2.85, P = .01), and for dilated cardiomyopathy the OR is 1.32 (95% CI: 1.01-1.71, P = .04). In contrast, our reverse Mendelian randomization showed that cardiomyopathies were not directly associated with TL, and the inverse-variance-weighted test was not statistically significant for any of the 3 (P > .05). The reliability tests for the forward Mendelian randomization, including both MR-Egger intercept and MR-PRESSO tests, show no evidence of horizontal pleiotropy, and Cochran Q test indicates no heterogeneity. The "leave-one-out" sensitivity analysis revealed no outlier genes. The reliability tests for the reverse Mendelian randomization, including both MR-Egger intercept and MR-PRESSO tests, also indicate no genetic pleiotropy. Despite the heterogeneity shown in our study between hypertrophic cardiomyopathy and leukocyte TL, the sensitivity analysis did not identify any anomalies. Our Mendelian randomization study suggests that longer leukocyte TL is associated with an increased risk of hypertrophic obstructive cardiomyopathy, hypertrophic cardiomyopathy, and dilated cardiomyopathy. However, the onset of these 2 kinds of disease does not directly lead to changes in leukocyte TL.
Collapse
Affiliation(s)
- Jun Li
- Guang’anmen Hospital, China Academy of Traditional Chinese Medicine, Beijing, China
| | - Lanshuo Hu
- Xiyuan Hospital, China Academy of Traditional Chinese Medicine, Beijing, China
| | - Xuanchun Huang
- Guang’anmen Hospital, China Academy of Traditional Chinese Medicine, Beijing, China
| |
Collapse
|
32
|
Kang M, Lee CS, Son H, Lee J, Lee J, Seo HJ, Kim MK, Choi M, Cho HJ, Kim HS. Latrophilin-2 Deletion in Cardiomyocyte Disrupts Cell Junction, Leading to D-CMP. Circ Res 2024; 135:1098-1115. [PMID: 39421931 DOI: 10.1161/circresaha.124.324670] [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: 04/08/2024] [Revised: 09/30/2024] [Accepted: 10/01/2024] [Indexed: 10/19/2024]
Abstract
BACKGROUND Latrophilin-2 (Lphn2), an adhesive GPCR (G protein-coupled receptor), was found to be a specific marker of cardiac progenitors during the differentiation of pluripotent stem cells into cardiomyocytes or during embryonic heart development in our previous studies. Its role in adult heart physiology, however, remains unclear. METHODS The embryonic lethality resulting from Lphn2 deletion necessitates the establishment of cardiomyocyte-specific, tamoxifen-inducible Lphn2 knockout mice, which was achieved by crossing Lphn2 flox/flox mice with mice having MerCreMer (tamoxifen-inducible Cre [Cyclization recombinase] recombinase) under the α-myosin heavy chain promoter. RESULTS Tamoxifen treatment for several days completely suppressed Lphn2 expression, specifically in the myocardium, and induced the dilated cardiomyopathy (D-CMP) phenotype with serious arrhythmia and sudden death in a short period of time. Transmission electron microscopy showed mitochondrial abnormalities, blurred Z-discs, and dehiscent myofibrils. The D-CMP phenotype, or heart failure, worsened during myocardial infarction. In a mechanistic study of D-CMP, Lphn2 knockout suppressed PGC-1α (Peroxisome proliferator-activated receptor gamma coactivator 1-alpha) and mitochondrial dysfunction, leading to the accumulation of reactive oxygen species and the global suppression of junctional molecules, such as N-cadherin (adherens junction), DSC-2 (desmocollin-2; desmosome), and connexin-43 (gap junction), leading to the dehiscence of cardiac myofibers and serious arrhythmia. In an experimental therapeutic trial, activators of p38-MAPK (p38 mitogen-activated protein kinases), which is a downstream signaling molecule of Lphn2, remarkably rescued the D-CMP phenotype of Lphn2 knockout in the heart by restoring PGC-1α and mitochondrial function and recovering global junctional proteins. CONCLUSIONS Lphn2 is a critical regulator of heart integrity by controlling mitochondrial functions and cell-to-cell junctions in cardiomyocytes. Its deficiency leads to D-CMP, which can be rescued by activators of the p38-MAPK pathway.
Collapse
MESH Headings
- Animals
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/pathology
- Mice, Knockout
- Mice
- Cardiomyopathy, Dilated/genetics
- Cardiomyopathy, Dilated/metabolism
- Cardiomyopathy, Dilated/pathology
- Intercellular Junctions/metabolism
- Intercellular Junctions/drug effects
- Receptors, Peptide/genetics
- Receptors, Peptide/metabolism
- Mice, Inbred C57BL
- Receptors, G-Protein-Coupled/metabolism
- Receptors, G-Protein-Coupled/genetics
- Receptors, G-Protein-Coupled/deficiency
- Tamoxifen/pharmacology
- p38 Mitogen-Activated Protein Kinases/metabolism
- Gene Deletion
- Male
- Cells, Cultured
- Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism
- Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics
Collapse
Affiliation(s)
- Minjun Kang
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, College of Medicine or College of Pharmacy, Seoul National University, South Korea (M.K., C.-S.L., H.S., H.J.S., H.-S.K.)
- Biomedical Research Institute (M.K., C.-S.L., H.S., Jaewon Lee, H.J.S., M.-K.K., H.-S.K.), Seoul National University Hospital, South Korea
| | - Choon-Soo Lee
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, College of Medicine or College of Pharmacy, Seoul National University, South Korea (M.K., C.-S.L., H.S., H.J.S., H.-S.K.)
- Biomedical Research Institute (M.K., C.-S.L., H.S., Jaewon Lee, H.J.S., M.-K.K., H.-S.K.), Seoul National University Hospital, South Korea
| | - HyunJu Son
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, College of Medicine or College of Pharmacy, Seoul National University, South Korea (M.K., C.-S.L., H.S., H.J.S., H.-S.K.)
- Biomedical Research Institute (M.K., C.-S.L., H.S., Jaewon Lee, H.J.S., M.-K.K., H.-S.K.), Seoul National University Hospital, South Korea
| | - Jeongha Lee
- Department of Biomedical Sciences, Seoul National University College of Medicine, South Korea (Jeongha Lee, M.C.)
| | - Jaewon Lee
- Biomedical Research Institute (M.K., C.-S.L., H.S., Jaewon Lee, H.J.S., M.-K.K., H.-S.K.), Seoul National University Hospital, South Korea
| | - Hyun Ju Seo
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, College of Medicine or College of Pharmacy, Seoul National University, South Korea (M.K., C.-S.L., H.S., H.J.S., H.-S.K.)
- Biomedical Research Institute (M.K., C.-S.L., H.S., Jaewon Lee, H.J.S., M.-K.K., H.-S.K.), Seoul National University Hospital, South Korea
| | - Moo-Kang Kim
- Department of Internal Medicine (M.-K.K., H.-J.C., H.-S.K.), Seoul National University Hospital, South Korea
- Biomedical Research Institute (M.K., C.-S.L., H.S., Jaewon Lee, H.J.S., M.-K.K., H.-S.K.), Seoul National University Hospital, South Korea
| | - Murim Choi
- Department of Biomedical Sciences, Seoul National University College of Medicine, South Korea (Jeongha Lee, M.C.)
| | - Hyun-Jai Cho
- Department of Internal Medicine (M.-K.K., H.-J.C., H.-S.K.), Seoul National University Hospital, South Korea
| | - Hyo-Soo Kim
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, College of Medicine or College of Pharmacy, Seoul National University, South Korea (M.K., C.-S.L., H.S., H.J.S., H.-S.K.)
- Department of Internal Medicine (M.-K.K., H.-J.C., H.-S.K.), Seoul National University Hospital, South Korea
- Biomedical Research Institute (M.K., C.-S.L., H.S., Jaewon Lee, H.J.S., M.-K.K., H.-S.K.), Seoul National University Hospital, South Korea
| |
Collapse
|
33
|
Patel R, Patel R, Patel E, Patel M. Dilated cardiomyopathy due to novel LMNA mutation: a case report. Front Cardiovasc Med 2024; 11:1422151. [PMID: 39411178 PMCID: PMC11473315 DOI: 10.3389/fcvm.2024.1422151] [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: 04/23/2024] [Accepted: 09/17/2024] [Indexed: 10/19/2024] Open
Abstract
A case of a 44-year-old man presenting with a family history of LMNA mutation and cardiac symptoms (dizziness, weakness, palpitations, and shortness of breath) congruent with dilated cardiomyopathy. Genetic testing revealed a novel likely pathogenic mutation of the LMNA gene (c.513G>A, exon 2) not previously associated with dilated cardiomyopathy, and the patient underwent guideline direct treatment for dilated cardiomyopathy. In patients with LMNA mutations, VTA risk should be calculated to determine the need for prophylactic ICD placement.
Collapse
Affiliation(s)
- Riddhi Patel
- Lake Erie College of Osteopathic Medicine, Erie, PA, United States
| | - Raj Patel
- Lake Erie College of Osteopathic Medicine, Greensburg, PA, United States
| | - Ekta Patel
- St. Bonaventure University, St. Bonaventure, NY, United States
| | - Mehul Patel
- Premier Medical Group—Cardiology Division, Newburgh, NY, United States
| |
Collapse
|
34
|
Kan JY, Wang DC, Jiang ZH, Wu LD, Xu K, Gu Y. Progression from cardiomyopathy to heart failure with reduced ejection fraction: A CORIN deficient course. Heliyon 2024; 10:e37838. [PMID: 39315128 PMCID: PMC11417248 DOI: 10.1016/j.heliyon.2024.e37838] [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: 01/08/2024] [Revised: 09/09/2024] [Accepted: 09/11/2024] [Indexed: 09/25/2024] Open
Abstract
Cardiomyopathies, encompassing hypertrophic cardiomyopathy (HCM) and dilated cardiomyopathy (DCM), constitute a diverse spectrum of heart muscle diseases that often culminating in heart failure (HF). The inherent molecular heterogeneity of these conditions has implications for prognosis and therapeutic strategies. Publicly available microarray and RNA sequencing (RNA-seq) data sets of HCM (n = 106 from GSE36961) and DCM (n = 18 from GSE135055 and 166 from GSE141910) patients were employed for our analysis. The Non-negative Matrix Factorization (NMF) algorithm was applied to explore the molecular stratification within HCM and DCM, and enrichment analysis was performed to delineate their biological characteristics. By integrating bulk and single-nucleus RNA-seq (snRNA-seq) data, we identified a potential biomarker for HCM progression and cardiac fibrosis, which was subsequently validated using mendelian randomization and in vitro. Our application of NMF identified two distinct molecular clusters. Particularly, a profibrotic, heart failure with reduced ejection fraction (HFrEF)-resembling Cluster 1 emerged, characterized by diminished expression of CORIN and a high degree of fibroblast activation. This cluster also exhibited lower left ventricular ejection fraction (LVEF) and worse prognostic outcomes, establishing the significance of this molecular subclassification. We further found that overexpression of CORIN could mitigate TGFβ1-induced expression of col1a1 and α-SMA in neonatal rat cardiac fibroblasts. Our results indicated the heterogeneity of HCM population, and further evidenced the participation of corin in the progression of HCM, DCM and HFrEF. Nevertheless, our study is constrained by the lack of corresponding clinical data and experimental validation of the identified subtypes. Therefore, further studies are warranted to elucidate the downstream pathways of corin and to validate these findings in independent patient cohorts.
Collapse
Affiliation(s)
| | | | | | - Li-da Wu
- Division of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Ke Xu
- Division of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Yue Gu
- Division of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| |
Collapse
|
35
|
Xu S, Zhang G, Tan X, Zeng Y, Jiang H, Jiang Y, Wang X, Song Y, Fan H, Zhou Y. Plasma Olink Proteomics Reveals Novel Biomarkers for Prediction and Diagnosis in Dilated Cardiomyopathy with Heart Failure. J Proteome Res 2024; 23:4139-4150. [PMID: 39129220 PMCID: PMC11385702 DOI: 10.1021/acs.jproteome.4c00522] [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] [Indexed: 08/13/2024]
Abstract
In this study, we utilized the Olink Cardiovascular III panel to compare the expression levels of 92 cardiovascular-related proteins between patients with dilated cardiomyopathy combined with heart failure (DCM-HF) (n = 20) and healthy normal people (Normal) (n = 18). The top five most significant proteins, including SPP1, IGFBP7, F11R, CHI3L1, and Plaur, were selected by Olink proteomics. These proteins were further validated using ELISA in plasma samples collected from an additional cohort. ELISA validation confirmed significant increases in SPP1, IGFBP7, F11R, CHI3L1, and Plaur in DCM-HF patients compared to healthy controls. GO and KEGG analysis indicated that NT-pro BNP, SPP1, IGFBP7, F11R, CHI3L1, Plaur, BLM hydrolase, CSTB, Gal-4, CCL15, CDH5, SR-PSOX, and CCL2 were associated with DCM-HF. Correlation analysis revealed that these 13 differentially expressed proteins have strong correlations with clinical indicators such as LVEF and NT-pro BNP, etc. Additionally, in the GEO-DCM data sets, the combined diagnostic value of these five core proteins AUC values of 0.959, 0.773, and 0.803, respectively indicating the predictive value of the five core proteins for DCM-HF. Our findings suggest that these proteins may be useful biomarkers for the diagnosis and prediction of DCM-HF, and further research is prompted to explore their potential as therapeutic targets.
Collapse
Affiliation(s)
- Shuai Xu
- Department of Cardiology, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Medical Center of Soochow University,Suzhou 215000, China
- Institute for Hypertension, Soochow University, Suzhou 215000, China
| | - Ge Zhang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
- Henan Province Key Laboratory of Cardiac Injury and Repair, Zhengzhou 450052, China
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou 450052, China
| | - Xin Tan
- Department of Cardiology, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Medical Center of Soochow University,Suzhou 215000, China
- Institute for Hypertension, Soochow University, Suzhou 215000, China
| | - Yiyao Zeng
- Department of Cardiology, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Medical Center of Soochow University,Suzhou 215000, China
- Institute for Hypertension, Soochow University, Suzhou 215000, China
| | - Hezi Jiang
- Department of Cardiology, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Medical Center of Soochow University,Suzhou 215000, China
- Institute for Hypertension, Soochow University, Suzhou 215000, China
| | - Yufeng Jiang
- Department of Cardiology, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Medical Center of Soochow University,Suzhou 215000, China
- Institute for Hypertension, Soochow University, Suzhou 215000, China
| | - Xiangyu Wang
- Department of Cardiology, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Medical Center of Soochow University,Suzhou 215000, China
- Institute for Hypertension, Soochow University, Suzhou 215000, China
| | - Yahui Song
- Center of Translational Medicine and Clinical Laboratory, The Fourth Affiliated Hospital to Soochow University, Suzhou 215028, China
| | - Huimin Fan
- Center of Translational Medicine and Clinical Laboratory, The Fourth Affiliated Hospital to Soochow University, Suzhou 215028, China
| | - Yafeng Zhou
- Department of Cardiology, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Medical Center of Soochow University,Suzhou 215000, China
- Institute for Hypertension, Soochow University, Suzhou 215000, China
| |
Collapse
|
36
|
Xue S, Jiang H. Exploring the etiology of dilated cardiomyopathy using Mendelian randomization. Front Cardiovasc Med 2024; 11:1364126. [PMID: 39253394 PMCID: PMC11381399 DOI: 10.3389/fcvm.2024.1364126] [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: 01/01/2024] [Accepted: 07/25/2024] [Indexed: 09/11/2024] Open
Abstract
Background Observational clinical studies suggest an association between dilated cardiomyopathy (DCM) and various factors including titin, cardiac troponin I (CTnI), desmocollin-2, the perinatal period, alcoholism, Behçet's disease, systemic lupus erythematosus, hyperthyroidism and thyrotoxicosis, hypothyroidism, carnitine metabolic disorder, and renal insufficiency. The causal nature of these associations remains uncertain. This study aims to explore these correlations using the Mendelian randomization (MR) approach. Objective To investigate the etiology of DCM through Mendelian randomization analysis. Methods Data mining was conducted in genome-wide association study databases, focusing on variant target proteins (titin, CTnI, desmocollin-2), the perinatal period, alcoholism, Behçet's disease, systemic lupus erythematosus, hyperthyroidism and thyrotoxicosis, hypothyroidism, carnitine metabolic disorder, and renal insufficiency, with DCM as the outcome. The analysis employed various regression models, namely, the inverse-variance weighted (IVW), MR-Egger, simple mode, weighted median, and weighted mode methods. Results The IVW results showed a correlation between titin protein and DCM, identifying titin as a protective factor [OR = 0.856, 95% CI (0.744-0.985), P = 0.030]. CTnI protein correlated with DCM, marking it as a risk factor [OR = 1.204, 95% CI (1.010-1.436), P = 0.040]. Desmocollin-2 also correlated with DCM and was recognized as a risk factor [OR = 1.309, 95% CI (1.085-1.579), P = 0.005]. However, no causal relationship was found between the perinatal period, alcoholism, Behçet's disease, systemic lupus erythematosus, hyperthyroidism and thyrotoxicosis, hypothyroidism, carnitine metabolic disorder, renal insufficiency, and DCM (P > 0.05). The MR-Egger intercept test indicated no pleiotropy (P > 0.05), affirming the effectiveness of Mendelian randomization in causal inference. Conclusion Titin, CTnI, and desmocollin-2 proteins were identified as independent risk factors for DCM. Contrasting with previous observational studies, no causal relationship was observed between DCM and the perinatal period, alcoholism, Behçet's disease, systemic lupus erythematosus, hyperthyroidism and thyrotoxicosis, hypothyroidism, carnitine metabolic disorder, or renal insufficiency.
Collapse
Affiliation(s)
- SiYang Xue
- Department of Cardiology, The Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - HongJu Jiang
- Department of Cardiology, The Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| |
Collapse
|
37
|
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
|
38
|
McBenedict B, Hauwanga WN, Amadi ES, Yau MCY, Amuzie CR, Abdelraof MO, Alphonse B, Ibrahim AMA, Okere MO, Chikere O, Johnny C, Lima Pessôa B. The Role of Ventricular Assist Devices in Patients With Heart Failure Due to Dilated Cardiomyopathy: A Systematic Review. Cureus 2024; 16:e66259. [PMID: 39238676 PMCID: PMC11377123 DOI: 10.7759/cureus.66259] [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: 07/08/2024] [Accepted: 08/01/2024] [Indexed: 09/07/2024] Open
Abstract
Dilated cardiomyopathy (DCM) is a prevalent heart muscle disease characterized by ventricular dilation and systolic dysfunction, leading to severe heart failure (HF) and often requiring heart transplantation (HTx). This systematic review aimed to synthesize information regarding the role of ventricular assist devices (VADs) in managing HF patients due to DCM. A comprehensive search was conducted across PubMed, Embase, Scopus, Web of Science, and Cochrane databases for studies published between 2014 and 2024. Inclusion criteria were studies involving adult patients with HF due to DCM treated with VADs. Exclusion criteria included non-human studies, pediatric populations, and non-peer-reviewed articles. Thirty-one studies met the inclusion criteria. The included studies demonstrated that the use of VADs in patients with DCM resulted in significant improvements in left ventricular ejection fraction (LVEF), myocardial fibrosis reduction, and reverse ventricular remodeling. Studies reported enhanced survival rates, reduced symptoms, and better quality of life. VADs served as a critical bridge to HTx and, in some cases, as long-term destination therapy. However, complications such as thrombus formation, anemia, and kidney failure were noted, emphasizing the need for vigilant monitoring and management. Continuous advancements in VAD technology and patient management protocols were found to be essential for optimizing outcomes. We conclude that VADs play a crucial role in managing advanced HF due to DCM by providing mechanical circulatory support, improving cardiac function, and enhancing patient survival and quality of life. Despite associated complications, VADs are invaluable for patients with severe HF, offering both immediate and long-term therapeutic benefits. Future research should focus on minimizing complications and further improving VAD technology to enhance patient outcomes.
Collapse
Affiliation(s)
| | - Wilhelmina N Hauwanga
- Family Medicine, Faculty of Medicine, Federal University of the State of Rio de Janeiro, Rio de Janeiro, BRA
| | | | | | - Chibuike R Amuzie
- Public Health, Louisiana State University Shreveport, Shreveport, USA
| | | | - Berley Alphonse
- Internal Medicine, University Notre Dame of Haiti, Port-au-Prince, HTI
| | | | - Madeleine O Okere
- Internal Medicine, University of Port Harcourt Teaching Hospital, Port Harcourt, NGA
| | - Ogbonna Chikere
- Public Health, Louisiana State University Shreveport, Shreveport, USA
| | | | | |
Collapse
|
39
|
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: 1] [Impact Index Per Article: 1.0] [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
|
40
|
Wang J, Diao Y, Xu Y, Guo J, Li W, Li Y, Wan K, Sun J, Han Y, Chen Y. Liver T1 Mapping Derived From Cardiac Magnetic Resonance Imaging: A Potential Prognostic Marker in Idiopathic Dilated Cardiomyopathy. J Magn Reson Imaging 2024; 60:675-685. [PMID: 38174826 DOI: 10.1002/jmri.29223] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 11/27/2023] [Accepted: 11/30/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND Hepatic alterations are common aftereffects of heart failure (HF) and ventricular dysfunction. The prognostic value of liver injury markers derived from cardiac MRI studies in nonischemic dilated cardiomyopathy (DCM) patients is unclear. PURPOSE Evaluate the prognostic performance of liver injury markers derived from cardiac MRI studies in DCM patients. STUDY TYPE Prospective. POPULATION Three hundred fifty-six consecutive DCM patients diagnosed according to ESC guidelines (age 48.7 ± 14.2 years, males 72.6%). FIELD STRENGTH/SEQUENCE Steady-state free precession, modified Look-Locker inversion recovery T1 mapping and phase sensitive inversion recovery late gadolinium enhancement (LGE) sequences at 3 T. ASSESSMENT Clinical characteristics, conventional MRI parameters (ventricular volumes, function, mass), native myocardial and liver T1, liver extracellular volume (ECV), and myocardial LGE presence were assessed. Patients were followed up for a median duration of 48.3 months (interquartile range 42.0-69.9 months). Primary endpoints included HF death, sudden cardiac death, heart transplantation, and HF readmission; secondary endpoints included HF death, sudden cardiac death, and heart transplantation. Models were developed to predict endpoints and the incremental value of including liver parameters assessed. STATISTICAL TESTS Optimal cut-off value was determined using receiver operating characteristic curve and Youden method. Survival analysis was performed using Kaplan-Meier and Cox proportional hazard. Discriminative power of models was compared using net reclassification improvement and integrated discriminatory index. P value <0.05 was considered statistically significant. RESULTS 47.2% patients reached primary endpoints; 25.8% patients reached secondary endpoints. Patients with elevated liver ECV (cut-off 34.4%) had significantly higher risk reaching primary and secondary endpoints. Cox regression showed liver ECV was an independent prognostic predictor, and showed independent prognostic value for primary endpoints and long-term HF readmission compared to conventional clinical and cardiac MRI parameters. DATA CONCLUSIONS Liver ECV is an independent prognostic predictor and may serve as an innovative approach for risk stratification for DCM. EVIDENCE LEVEL 1 TECHNICAL EFFICACY: Stage 2.
Collapse
Affiliation(s)
- Jiaqi Wang
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, China
| | - Yike Diao
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
| | - Yuanwei Xu
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, China
| | - Jiajun Guo
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, China
| | - Weihao Li
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, China
| | - Yangjie Li
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, China
| | - Ke Wan
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, China
| | - Jiayu Sun
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
| | - Yuchi Han
- Cardiovascular Medicine, Wexner Medical Center, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Yucheng Chen
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
41
|
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
|
42
|
Kopeva K. Can cuproptosis-related genes be involved in the pathogenesis of dilated cardiomyopathy? Int J Cardiol 2024; 403:131860. [PMID: 38367885 DOI: 10.1016/j.ijcard.2024.131860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 02/10/2024] [Indexed: 02/19/2024]
Affiliation(s)
- Kristina Kopeva
- Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| |
Collapse
|
43
|
Solaro RJ, Goldspink PH, Wolska BM. Emerging Concepts of Mechanisms Controlling Cardiac Tension: Focus on Familial Dilated Cardiomyopathy (DCM) and Sarcomere-Directed Therapies. Biomedicines 2024; 12:999. [PMID: 38790961 PMCID: PMC11117855 DOI: 10.3390/biomedicines12050999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 04/28/2024] [Accepted: 04/30/2024] [Indexed: 05/26/2024] Open
Abstract
Novel therapies for the treatment of familial dilated cardiomyopathy (DCM) are lacking. Shaping research directions to clinical needs is critical. Triggers for the progression of the disorder commonly occur due to specific gene variants that affect the production of sarcomeric/cytoskeletal proteins. Generally, these variants cause a decrease in tension by the myofilaments, resulting in signaling abnormalities within the micro-environment, which over time result in structural and functional maladaptations, leading to heart failure (HF). Current concepts support the hypothesis that the mutant sarcomere proteins induce a causal depression in the tension-time integral (TTI) of linear preparations of cardiac muscle. However, molecular mechanisms underlying tension generation particularly concerning mutant proteins and their impact on sarcomere molecular signaling are currently controversial. Thus, there is a need for clarification as to how mutant proteins affect sarcomere molecular signaling in the etiology and progression of DCM. A main topic in this controversy is the control of the number of tension-generating myosin heads reacting with the thin filament. One line of investigation proposes that this number is determined by changes in the ratio of myosin heads in a sequestered super-relaxed state (SRX) or in a disordered relaxed state (DRX) poised for force generation upon the Ca2+ activation of the thin filament. Contrasting evidence from nanometer-micrometer-scale X-ray diffraction in intact trabeculae indicates that the SRX/DRX states may have a lesser role. Instead, the proposal is that myosin heads are in a basal OFF state in relaxation then transfer to an ON state through a mechano-sensing mechanism induced during early thin filament activation and increasing thick filament strain. Recent evidence about the modulation of these mechanisms by protein phosphorylation has also introduced a need for reconsidering the control of tension. We discuss these mechanisms that lead to different ideas related to how tension is disturbed by levels of mutant sarcomere proteins linked to the expression of gene variants in the complex landscape of DCM. Resolving the various mechanisms and incorporating them into a unified concept is crucial for gaining a comprehensive understanding of DCM. This deeper understanding is not only important for diagnosis and treatment strategies with small molecules, but also for understanding the reciprocal signaling processes that occur between cardiac myocytes and their micro-environment. By unraveling these complexities, we can pave the way for improved therapeutic interventions for managing DCM.
Collapse
Affiliation(s)
- R. John Solaro
- Department of Physiology and Biophysics, Center for Cardiovascular Research, University of Illinois at Chicago, Chicago, IL 60612, USA; (P.H.G.); (B.M.W.)
| | - Paul H. Goldspink
- Department of Physiology and Biophysics, Center for Cardiovascular Research, University of Illinois at Chicago, Chicago, IL 60612, USA; (P.H.G.); (B.M.W.)
| | - Beata M. Wolska
- Department of Physiology and Biophysics, Center for Cardiovascular Research, University of Illinois at Chicago, Chicago, IL 60612, USA; (P.H.G.); (B.M.W.)
- Department of Medicine, Section of Cardiology, University of Illinois at Chicago, Chicago, IL 60612, USA
| |
Collapse
|
44
|
Aryafar M, Mahdavi M, Shahzadi H, Ranjbar YR, Sohouli MH, Afzal S, Tehrani AN, Fotros D, Daftari G. Association between dietary selenium and zinc intake and risk of dilated cardiomyopathy in children: a case-control study. BMC Pediatr 2024; 24:251. [PMID: 38605385 PMCID: PMC11010394 DOI: 10.1186/s12887-024-04706-1] [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: 10/16/2023] [Accepted: 03/12/2024] [Indexed: 04/13/2024] Open
Abstract
BACKGROUND Dilated cardiomyopathy (DCMP) is characterized by the enlargement and weakening of the heart and is a major cause of heart failure in children. Infection and nutritional deficiencies are culprits for DCMP. Zinc is an important nutrient for human health due to its anti-oxidant effect that protects cell against oxidative damage. This case-control study aimed to investigate the relationship between dietary intake of zinc and selenium and the risk of DCMP in pediatric patients. METHODS A total of 36 DCMP patients and 72 matched controls were recruited, and their dietary intakes were assessed via a validated food frequency questionnaire. We used chi-square and sample T-test for qualitative and quantitative variables, respectively. Logistic regression analysis was applied to assess the relationship between selenium and zinc intake with the risk of DCMP. RESULTS After fully adjusting for confounding factors, analyses showed that selenium (OR = 0.19, CI = 0.057-0.069, P trend < 0.011) and zinc (OR = 0.12, CI = 0.035-0.046, P trend < 0.002) intake were strongly associated with 81% and 88% lower risk of pediatric DCMP, respectively. CONCLUSIONS This study highlights the protective role of adequate dietary intake of selenium and zinc in decreasing the risk of DCMP in children. Malnutrition may exacerbate the condition and addressing these micronutrient deficiencies may improve the cardiac function. Further studies are recommended to detect the underlying mechanisms and dietary recommendations for DCMP prevention.
Collapse
Affiliation(s)
- Maryam Aryafar
- Rajaei Cardiovascular Medical and Research Center, Iran University of Medical Sciences Tehran, Tehran, Iran
| | - Mohammad Mahdavi
- Rajaei Cardiovascular Medical and Research Center, Iran University of Medical Sciences Tehran, Tehran, Iran
| | - Hossein Shahzadi
- Rajaei Cardiovascular Medical and Research Center, Iran University of Medical Sciences Tehran, Tehran, Iran
| | - Yeganeh Rajabpour Ranjbar
- Department of Clinical Nutrition and Dietetics, Faculty of Nutrition Sciences and Food Technology, Research Institute, National Nutrition and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Sina Afzal
- Department of Orthopedic and Trauma Surgery, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Asal Neshatbini Tehrani
- Student Research Committee, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Department of Nutrition, School of Allied Medical Sciences, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Danial Fotros
- Department of Clinical Nutrition and Dietetics, Faculty of Nutrition Sciences and Food Technology, Research Institute, National Nutrition and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ghazal Daftari
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
- Keshavarz Boulevard, Tehran, Iran.
| |
Collapse
|
45
|
He B, Quan L, Li C, Yan W, Zhang Z, Zhou L, Wei Q, Li Z, Mo J, Zhang Z, Pan X, Huang J, Liu L. Targeting ERBB2 and PIK3R1 as a therapeutic strategy for dilated cardiomyopathy: A single-cell sequencing and mendelian randomization analysis. Heliyon 2024; 10:e25572. [PMID: 38434379 PMCID: PMC10907741 DOI: 10.1016/j.heliyon.2024.e25572] [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: 07/19/2023] [Revised: 01/13/2024] [Accepted: 01/29/2024] [Indexed: 03/05/2024] Open
Abstract
Background Dilated cardiomyopathy (DCM) is widely recognized as a significant contributor to heart failure. Nevertheless, the absence of pharmaceutical interventions capable of reversing disease progression and improving prognosis underscores the imperative for additional research in this area. Methods First, we identified and evaluated three gene sets, namely "SC-DCM", "EP-DCM" and "Drug", using big data and multiple bioinformatics analysis methods. Accordingly, drug-treatable ("Hub") genes in DCM were identified. Following this, four microarray expression profile datasets were employed to authenticate the expression levels and discriminatory efficacy of "Hub" genes. Additionally, mendelian randomization analysis was conducted to ascertain the causal association between the "Hub genes" and heart failure. Finally, the "DGIdb" was applied to identify "Hub" genes-targeted drugs. The "ssGSEA" algorithm assessed the level of immune cell infiltration in DCM. Results Enrichment analysis showed that the "SC-DCM" and "EP-DCM" gene sets were closely associated with DCM. PIK3R1 and ERBB2 were identified as drug-treatable genes in DCM. Additional analysis using MR supported a causal relationship between ERBB2 and heart failure, but not PIK3R1. Moreover, PIK3R1 was positively correlated with immune activation, while ERBB2 was negatively correlated. We found that everolimus was a pharmacological inhibitor for both PIK3R1 and ERBB2. However, no pharmacological agonist was found for ERBB2. Conclusion PIK3R1 and ERBB2 are drug-treatable genes in DCM. ERBB2 has a causal effect on heart failure, and its normal expression may play a role in preventing the progression of DCM to heart failure. In addition, there is a cross-expression of PIK3R1 and ERBB2 genes in both DCM and tumors. The adaptive immune system and PIK3R1 may be involved in DCM disease progression, while ERBB2 exerts a protective effect against DCM.
Collapse
Affiliation(s)
- Bin He
- Graduate School of Youjiang Medical University for Nationalities, Baise, China
| | - Liping Quan
- Graduate School of Youjiang Medical University for Nationalities, Baise, China
| | - Chengban Li
- Graduate School of Youjiang Medical University for Nationalities, Baise, China
| | - Wei Yan
- Graduate School of Youjiang Medical University for Nationalities, Baise, China
- Department of Cardiology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - ZhuoHua Zhang
- Department of Cardiology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - LiuFan Zhou
- Department of Cardiology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Qinjiang Wei
- Department of Cardiology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Zhile Li
- Department of Cardiology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Jianjiao Mo
- Department of Cardiology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Zhen Zhang
- Department of Cardiology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Xingshou Pan
- Department of Cardiology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - JianJun Huang
- College of Clinical Medicine, Youjiang Medical University for Nationalities, Baise, China
- Department of Neurology, Affiliated Hospital of Youjiang Medical University for Nationalities, Youjiang Medical University for Nationalities, Baise, 533000, Guangxi, China
| | - Li Liu
- Department of Cardiology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
- The Key Laboratory for High Incidence Prevention and Treatment in Guangxi Guixi Area, Baise, 533000, Youjiang Medical University for Nationalities, Baise, 533000, Guangxi, China
| |
Collapse
|
46
|
Li S, Liu S, Sun X, Hao L, Gao Q. Identification of endocrine-disrupting chemicals targeting key DCM-associated genes via bioinformatics and machine learning. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 274:116168. [PMID: 38460409 DOI: 10.1016/j.ecoenv.2024.116168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 02/04/2024] [Accepted: 02/27/2024] [Indexed: 03/11/2024]
Abstract
Dilated cardiomyopathy (DCM) is a primary cause of heart failure (HF), with the incidence of HF increasing consistently in recent years. DCM pathogenesis involves a combination of inherited predisposition and environmental factors. Endocrine-disrupting chemicals (EDCs) are exogenous chemicals that interfere with endogenous hormone action and are capable of targeting various organs, including the heart. However, the impact of these disruptors on heart disease through their effects on genes remains underexplored. In this study, we aimed to explore key DCM-related genes using machine learning (ML) and the construction of a predictive model. Using the Gene Expression Omnibus (GEO) database, we screened differentially expressed genes (DEGs) and performed enrichment analyses of Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways related to DCM. Through ML techniques combining maximum relevance minimum redundancy (mRMR) and least absolute shrinkage and selection operator (LASSO) logistic regression, we identified key genes for predicting DCM (IL1RL1, SEZ6L, SFRP4, COL22A1, RNASE2, HB). Based on these key genes, 79 EDCs with the potential to affect DCM were identified, among which 4 (3,4-dichloroaniline, fenitrothion, pyrene, and isoproturon) have not been previously associated with DCM. These findings establish a novel relationship between the EDCs mediated by key genes and the development of DCM.
Collapse
Affiliation(s)
- Shu Li
- Department of Health and Intelligent Engineering, College of Health Management, China Medical University, Shenyang, Liaoning Province 110122, PR China..
| | - Shuice Liu
- Department of Pharmacology, Shenyang Medical College, Shenyang, Liaoning Province 110001, PR China..
| | - Xuefei Sun
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang 110122, PR China..
| | - Liying Hao
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang 110122, PR China..
| | - Qinghua Gao
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, PR China..
| |
Collapse
|
47
|
Li X, He W, Song Q, Ding Q, Zhang X, Zeng Z, Deng W, Deng G, Guan L, Hong W, Liu Y, Shu F, Xu L, Tan N, Ma J, Jiang L. The Prognostic Value of Serum Calcium Levels in Elderly Dilated Cardiomyopathy Patients. Glob Heart 2024; 19:25. [PMID: 38434155 PMCID: PMC10906334 DOI: 10.5334/gh.1304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 02/05/2024] [Indexed: 03/05/2024] Open
Abstract
Background It is unclear whether serum calcium on admission is associated with clinical outcomes in dilated cardiomyopathy (DCM). In this study, we conducted a retrospective study spanning a decade to investigate the prognostic value of baseline calcium in elderly patients with DCM. Methods A total of 1,089 consecutive elderly patients (age ≥60 years) diagnosed with DCM were retrospectively enrolled from January 2010 to December 2019. Univariate and multivariate analyses were performed to investigate the association of serum calcium with their clinical outcomes. Results In this study, the average age of the subjects was 68.36 ± 6.31 years. Receiver operating characteristic (ROC) curve analysis showed that serum calcium level had a great sensitivity and specificity for predicting in-hospital death, with an AUC of 0.732. Kaplan-Meier survival analysis showed that patients with a serum calcium >8.62 mg/dL had a better prognosis than those with a serum calcium ≤8.62 mg/dL (log-rank χ2 40.84, p < 0.001). After adjusting for several common risk factors, a serum calcium ≤8.62 mg/dL was related to a higher risk of long-term mortality (HR: 1.449; 95% CI: 1.115~1.882; p = 0.005). Conclusions Serum calcium level could be served as a simple and affordable tool to evaluate patients' prognosis in DCM.
Collapse
Affiliation(s)
- Xinyi Li
- School of Medicine, South China University of Technology, Guangzhou 510006, China
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Wenfei He
- Department of Cardiology, Guangdong Provincial People’s Hospital’s Nanhai Hospital, The Second People’s Hospital of Nanhai District, Foshan 528000, China
| | - Qiqi Song
- School of Medicine, South China University of Technology, Guangzhou 510006, China
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Qingshan Ding
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Xiaonan Zhang
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Zhigang Zeng
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Weiping Deng
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Gang Deng
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Lichang Guan
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Wanzi Hong
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Yaoxin Liu
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Fen Shu
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Lishu Xu
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Ning Tan
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Jinjin Ma
- School of Medicine, South China University of Technology, Guangzhou 510006, China
| | - Lei Jiang
- School of Medicine, South China University of Technology, Guangzhou 510006, China
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| |
Collapse
|
48
|
Keil L, Berisha F, Ritter S, Skibowski J, Subramanian H, Nikolaev VO, Kubisch C, Woitschach R, Fabritz L, Twerenbold R, Blankenberg S, Weidemann S, Zeller T, Kirchhof P, Reichart D, Magnussen C. Multimodal characterization of dilated cardiomyopathy: Geno- And Phenotyping of PrImary Cardiomyopathy (GrAPHIC). ESC Heart Fail 2024; 11:541-549. [PMID: 37964758 PMCID: PMC10804161 DOI: 10.1002/ehf2.14544] [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: 04/11/2023] [Revised: 08/23/2023] [Accepted: 09/15/2023] [Indexed: 11/16/2023] Open
Abstract
AIMS Cardiomyopathies (CMPs) are a heterogeneous group of diseases that are defined by structural and functional abnormalities of the cardiac muscle. Dilated cardiomyopathy (DCM), the most common CMP, is defined by left ventricular dilation and impaired contractility and represents a common cause of heart failure. Different phenotypes result from various underlying genetic and acquired causes with variable effects on disease development and progression, prognosis, and response to medical treatment. Current treatment algorithms do not consider these different aetiologies, due to lack of insights into treatable drivers of cardiac failure in patients with DCM. Our study aims to precisely phenotype and genotype the various subtypes of DCM and hereby lay the foundation for individualized therapy. METHODS AND RESULTS The Geno- And Phenotyping of PrImary Cardiomyopathy (GrAPHIC) is a currently ongoing prospective observational monocentric cohort study that recruits patients with DCM after exclusion of other causes such as coronary artery disease, valvular dysfunction, myocarditis, exposure to toxins, and peripartum CMP. Patients are enrolled at our heart failure outpatient clinic or during hospitalization at the University Hospital Hamburg. Clinical parameters, multimodal imaging and functional assessment, cardiac biopsies, and blood samples are obtained to enable an integrated genomic, functional, and biomarker analysis. CONCLUSIONS The GrAPHIC will contribute to a better understanding of the heterogeneous nature of primary CMPs focusing on DCM and provide improved prognostic approaches and more individualized therapies.
Collapse
Affiliation(s)
- Laura Keil
- Department of CardiologyUniversity Heart and Vascular Center Hamburg, University Medical Center Hamburg‐EppendorfHamburgGermany
| | - Filip Berisha
- Department of CardiologyUniversity Heart and Vascular Center Hamburg, University Medical Center Hamburg‐EppendorfHamburgGermany
- German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/LuebeckHamburgGermany
- Institute of Experimental Cardiovascular ResearchUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Stella Ritter
- Department of CardiologyUniversity Heart and Vascular Center Hamburg, University Medical Center Hamburg‐EppendorfHamburgGermany
| | - Johanna Skibowski
- Department of CardiologyUniversity Heart and Vascular Center Hamburg, University Medical Center Hamburg‐EppendorfHamburgGermany
| | - Hariharan Subramanian
- German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/LuebeckHamburgGermany
- Institute of Experimental Cardiovascular ResearchUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Viacheslav O. Nikolaev
- German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/LuebeckHamburgGermany
- Institute of Experimental Cardiovascular ResearchUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Christian Kubisch
- Institute of Human GeneticsUniversity Hospital Hamburg‐EppendorfHamburgGermany
| | - Rixa Woitschach
- Institute of Human GeneticsUniversity Hospital Hamburg‐EppendorfHamburgGermany
| | - Larissa Fabritz
- Department of CardiologyUniversity Heart and Vascular Center Hamburg, University Medical Center Hamburg‐EppendorfHamburgGermany
- German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/LuebeckHamburgGermany
- University Centre of Cardiovascular Science, UKE HamburgHamburgGermany
| | - Raphael Twerenbold
- Department of CardiologyUniversity Heart and Vascular Center Hamburg, University Medical Center Hamburg‐EppendorfHamburgGermany
- German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/LuebeckHamburgGermany
- University Centre of Cardiovascular Science, UKE HamburgHamburgGermany
| | - Stefan Blankenberg
- Department of CardiologyUniversity Heart and Vascular Center Hamburg, University Medical Center Hamburg‐EppendorfHamburgGermany
- German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/LuebeckHamburgGermany
| | - Sören Weidemann
- Department of PathologyUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Tanja Zeller
- Department of CardiologyUniversity Heart and Vascular Center Hamburg, University Medical Center Hamburg‐EppendorfHamburgGermany
- German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/LuebeckHamburgGermany
- University Centre of Cardiovascular Science, UKE HamburgHamburgGermany
| | - Paulus Kirchhof
- Department of CardiologyUniversity Heart and Vascular Center Hamburg, University Medical Center Hamburg‐EppendorfHamburgGermany
- German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/LuebeckHamburgGermany
| | - Daniel Reichart
- Department of Medicine IUniversity Hospital, LMU MunichMunichGermany
| | - Christina Magnussen
- Department of CardiologyUniversity Heart and Vascular Center Hamburg, University Medical Center Hamburg‐EppendorfHamburgGermany
- German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/LuebeckHamburgGermany
| |
Collapse
|
49
|
Huang Y, Li LC, Li YX, Gui C, Yang LH. Development and validation of a risk model for intracardiac thrombosis in patients with dilated cardiomyopathy: a retrospective study. Sci Rep 2024; 14:1431. [PMID: 38228722 PMCID: PMC10791606 DOI: 10.1038/s41598-024-51745-w] [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: 08/15/2023] [Accepted: 01/09/2024] [Indexed: 01/18/2024] Open
Abstract
Intracardiac thrombosis is a severe complication in patients with non-ischemic dilated cardiomyopathy. This study aims to develop and validate an individualized nomogram to evaluate the risk of intracardiac thrombosis in patients with non-ischemic dilated cardiomyopathy. This retrospective study included patients diagnosed with dilated cardiomyopathy at first admission. Clinical baseline characteristics were acquired from electronic medical record systems. Multiple methods were applied to screen the key variables and generate multiple different variable combinations. Multivariable logistic regression was used to build the models, and the optimal model was chosen by comparing the discrimination. Then we checked the performance of the model in different thrombus subgroups. Finally, the model was presented using a nomogram and evaluated from the perspectives of discrimination, calibration, and clinical usefulness. Internal validation was performed by extracting different proportions of data for Bootstrapping. Ultimately, 564 eligible patients were enrolled, 67 of whom developed an intracardiac thrombosis. Risk factors included d-dimer, white blood cell count, high-sensitivity C-reactive protein, pulse pressure, history of stroke, hematocrit, and NT-proBNP in the optimal model. The model had good discrimination and calibration, and the area under the curve (AUC) was 0.833 (0.782-0.884), and the model's performance in each subgroup was stable. Clinical decision curve analysis showed that the model had clinical application value when the high-risk threshold was between 2% and 78%. The AUC of interval validation (30% and 70% data resampling) was 0.844 (0.765-0.924) and 0.833 (0.775-0.891), respectively. This novel intracardiac thrombosis nomogram could be conveniently applied to facilitate the individual intracardiac thrombosis risk assessment in patients with non-ischemic dilated cardiomyopathy.
Collapse
Affiliation(s)
- Yuan Huang
- Department of Cardiology, Jiangbin Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
| | - Long-Chang Li
- Department of Cardiology, The First People's Hospital of Nanning, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Yu-Xin Li
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
- Guangxi Key Laboratory Base of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Nanning, 530021, Guangxi, China
- Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, Nanning, 530021, Guangxi, China
| | - Chun Gui
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China.
- Guangxi Key Laboratory Base of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Nanning, 530021, Guangxi, China.
- Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, Nanning, 530021, Guangxi, China.
| | - Li-Hua Yang
- Department of Cardiology, Jiangbin Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China.
| |
Collapse
|
50
|
Amdani S, Auerbach SR, Bansal N, Chen S, Conway J, Silva JPDA, Deshpande SR, Hoover J, Lin KY, Miyamoto SD, Puri K, Price J, Spinner J, White R, Rossano JW, Bearl DW, Cousino MK, Catlin P, Hidalgo NC, Godown J, Kantor P, Masarone D, Peng DM, Rea KE, Schumacher K, Shaddy R, Shea E, Tapia HV, Valikodath N, Zafar F, Hsu D. Research Gaps in Pediatric Heart Failure: Defining the Gaps and Then Closing Them Over the Next Decade. J Card Fail 2024; 30:64-77. [PMID: 38065308 DOI: 10.1016/j.cardfail.2023.08.026] [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: 02/07/2023] [Revised: 08/18/2023] [Accepted: 08/19/2023] [Indexed: 01/13/2024]
Abstract
Given the numerous opportunities and the wide knowledge gaps in pediatric heart failure, an international group of pediatric heart failure experts with diverse backgrounds were invited and tasked with identifying research gaps in each pediatric heart failure domain that scientists and funding agencies need to focus on over the next decade.
Collapse
Affiliation(s)
- Shahnawaz Amdani
- Department of Pediatric Cardiology, Cleveland Clinic Children's, Cleveland, Ohio.
| | - Scott R Auerbach
- Division of Pediatric Cardiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Neha Bansal
- Division of Pediatric Cardiology, Mount Sinai Kravis Children's Hospital, Icahn School of Medicine, New York, New York
| | - Sharon Chen
- Division of Pediatric Cardiology, Lucile Packard Children's Hospital, Stanford University School of Medicine, Palo Alto, California
| | - Jennifer Conway
- Division of Pediatric Cardiology, Stollery Children's Hospital, Edmonton, Alberta, Canada
| | - Julie Pires DA Silva
- Division of Pediatric Cardiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | | | - Jessica Hoover
- Department of Pediatric Cardiology, Cleveland Clinic Children's, Cleveland, Ohio
| | - Kimberly Y Lin
- Division of Cardiology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Shelley D Miyamoto
- Division of Pediatric Cardiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Kriti Puri
- Department of Pediatrics, Section of Pediatric Cardiology, Baylor College of Medicine/Texas Children's Hospital, Houston, Texas
| | - Jack Price
- Department of Pediatrics, Section of Pediatric Cardiology, Baylor College of Medicine/Texas Children's Hospital, Houston, Texas
| | - Joseph Spinner
- Department of Pediatrics, Section of Pediatric Cardiology, Baylor College of Medicine/Texas Children's Hospital, Houston, Texas
| | - Rachel White
- Division of Cardiology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Joseph W Rossano
- Division of Cardiology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - David W Bearl
- Department of Pediatric Cardiology, Monroe Carell Jr. Children's Hospital, Nashville, Tennessee
| | - Melissa K Cousino
- Department of Pediatrics, University of Michigan, C. S. Mott Children's Hospital, Ann Arbor, Michigan
| | - Perry Catlin
- Department of Psychology, Marquette University, Milwaukee, Wisconsin
| | - Nicolas Corral Hidalgo
- Division of Pediatric Cardiology, Children's Hospital at Montefiore, Albert Einstein College of Medicine, Bronx, New York
| | - Justin Godown
- Department of Pediatric Cardiology, Monroe Carell Jr. Children's Hospital, Nashville, Tennessee
| | - Paul Kantor
- Children's Hospital Los Angeles and the Keck School of Medicine of the University of Southern California, Los Angeles, California
| | - Daniele Masarone
- Heart Failure Unit, Department of Cardiology, AORN dei Colli-Monaldi Hospital Naples, Naples, Italy
| | - David M Peng
- Department of Pediatrics, University of Michigan, C. S. Mott Children's Hospital, Ann Arbor, Michigan
| | - Kelly E Rea
- Department of Pediatrics, University of Michigan, C. S. Mott Children's Hospital, Ann Arbor, Michigan
| | - Kurt Schumacher
- Department of Pediatrics, University of Michigan, C. S. Mott Children's Hospital, Ann Arbor, Michigan
| | - Robert Shaddy
- Children's Hospital Los Angeles and the Keck School of Medicine of the University of Southern California, Los Angeles, California
| | - Erin Shea
- Heart Failure Unit, Department of Cardiology, AORN dei Colli-Monaldi Hospital Naples, Naples, Italy
| | - Henry Valora Tapia
- Division of Pediatric Cardiology, University of Utah. Salt Lake City, Utah
| | - Nishma Valikodath
- Department of Pediatrics, Section of Pediatric Cardiology, Baylor College of Medicine/Texas Children's Hospital, Houston, Texas
| | - Farhan Zafar
- The Heart Institute, Cincinnati Children's Hospital Medical Center, College of Medicine, University of Cincinnati, Cincinnati, Ohio
| | - Daphne Hsu
- Division of Pediatric Cardiology, Children's Hospital at Montefiore, Albert Einstein College of Medicine, Bronx, New York
| |
Collapse
|