Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 18.546
Filter
1.
Circ Heart Fail ; 17(6): e011057, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38847093

ABSTRACT

BACKGROUND: The immune systems and chronic inflammation are implicated in the pathogenesis of dilated cardiomyopathy (DCM) and heart failure. However, the significance of neutrophil extracellular traps (NETs) in heart failure remains to be elucidated. METHODS: We enrolled consecutive 62 patients with heart failure with idiopathic DCM who underwent endomyocardial biopsy. Biopsy specimens were subjected to fluorescent immunostaining to detect NETs, and clinical and outcome data were collected. Ex vivo and in vivo experiments were conducted. RESULTS: The numbers of NETs per myocardial tissue area and the proportion of NETs per neutrophil were significantly higher in patients with DCM compared with non-DCM control subjects without heart failure, and the numbers of NETs were negatively correlated with left ventricular ejection fraction. Patients with DCM with NETs (n=32) showed lower left ventricular ejection fraction and higher BNP (B-type natriuretic peptide) than those without NETs (n=30). In a multivariable Cox proportional hazard model, the presence of NETs was independently associated with an increased risk of adverse cardiac events in patients with DCM. To understand specific underlying mechanisms, extracellular flux analysis in ex vivo revealed that NETs-containing conditioned medium from wild-type neutrophils or purified NET components led to impaired mitochondrial oxygen consumption of cardiomyocytes, while these effects were abolished when PAD4 (peptidyl arginine deiminase 4) in neutrophils was genetically ablated. In a murine model of pressure overload, NETs in myocardial tissue were predominantly detected in the acute phase and persisted throughout the ongoing stress. Four weeks after transverse aortic constriction, left ventricular ejection fraction was reduced in wild-type mice, whereas PAD4-deficient mice displayed preserved left ventricular ejection fraction without inducing NET formation. CONCLUSIONS: NETs in myocardial tissue contribute to cardiac dysfunction and adverse outcomes in patients with heart failure with DCM, potentially through mitochondrial dysfunction of cardiomyocytes.


Subject(s)
Cardiomyopathy, Dilated , Extracellular Traps , Heart Failure , Myocardium , Neutrophils , Cardiomyopathy, Dilated/physiopathology , Cardiomyopathy, Dilated/metabolism , Humans , Extracellular Traps/metabolism , Heart Failure/physiopathology , Male , Female , Middle Aged , Animals , Myocardium/pathology , Myocardium/metabolism , Neutrophils/metabolism , Stroke Volume/physiology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Ventricular Function, Left/physiology , Mice , Aged , Mitochondria, Heart/metabolism , Mitochondria, Heart/pathology , Mice, Inbred C57BL , Biopsy
2.
Stem Cell Res ; 78: 103467, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38861774

ABSTRACT

Dilated cardiomyopathy (DCM) is one of the main causes of sudden cardiac death and heart failure and is the leading indication for cardiac transplantation worldwide. Mutations in dozens of cardiac genes have been connected to the development of DCM including the Troponin T2 gene (TNNT2). Here, we generated a human induced pluripotent stem cells (hiPSCs) from a DCM patient with a familial history that carries a missense mutation in TNNT2. The hiPSCs show typical morphology of pluripotent stem cells, expression of pluripotency markers, normal karyotype, and in vitro capacity to differentiate into all three germ layers.


Subject(s)
Cardiomyopathy, Dilated , Induced Pluripotent Stem Cells , Troponin T , Humans , Cardiomyopathy, Dilated/pathology , Induced Pluripotent Stem Cells/metabolism , Troponin T/metabolism , Troponin T/genetics , Cell Differentiation , Cell Line , Male , Karyotype
3.
Commun Biol ; 7(1): 702, 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38849449

ABSTRACT

The Drosophila model is pivotal in deciphering the pathophysiological underpinnings of various human ailments, notably aging and cardiovascular diseases. Cutting-edge imaging techniques and physiology yield vast high-resolution videos, demanding advanced analysis methods. Our platform leverages deep learning to segment optical microscopy images of Drosophila hearts, enabling the quantification of cardiac parameters in aging and dilated cardiomyopathy (DCM). Validation using experimental datasets confirms the efficacy of our aging model. We employ two innovative approaches deep-learning video classification and machine-learning based on cardiac parameters to predict fly aging, achieving accuracies of 83.3% (AUC 0.90) and 79.1%, (AUC 0.87) respectively. Moreover, we extend our deep-learning methodology to assess cardiac dysfunction associated with the knock-down of oxoglutarate dehydrogenase (OGDH), revealing its potential in studying DCM. This versatile approach promises accelerated cardiac assays for modeling various human diseases in Drosophila and holds promise for application in animal and human cardiac physiology under diverse conditions.


Subject(s)
Aging , Cardiomyopathy, Dilated , Disease Models, Animal , Machine Learning , Animals , Cardiomyopathy, Dilated/physiopathology , Cardiomyopathy, Dilated/genetics , Aging/physiology , Drosophila melanogaster/physiology , Deep Learning , Heart/physiopathology , Heart/physiology , Humans , Drosophila/physiology
4.
Sci Rep ; 14(1): 13942, 2024 06 17.
Article in English | MEDLINE | ID: mdl-38886541

ABSTRACT

Dilated cardiomyopathy (DCM) is a common cause of heart failure, thromboembolism, arrhythmias, and sudden cardiac death. The quality of life and long-term survival rates of patients with dilated DCM have greatly improved in recent decades. Nevertheless, the clinical prognosis for DCM patients remains unfavorable. The primary driving factors underlying the pathogenesis of DCM remain incompletely understood. The present study aimed to identify driving factors underlying the pathogenesis of DCM from the perspective of gene regulatory networks. Single-cell RNA sequencing data and bulk RNA data were obtained from the Gene Expression Omnibus (GEO) database. Differential gene analysis, single-cell genomics analysis, and functional enrichment analysis were conducted using R software. The construction of Gene Regulatory Networks was performed using Python. We used the pySCENIC method to analyze the single-cell data and identified 401 regulons. Through variance decomposition, we selected 19 regulons that showed significant responsiveness to DCM. Next, we employed the ssGSEA method to assess regulons in two bulk RNA datasets. Significant statistical differences were observed in 9 and 13 regulons in each dataset. By intersecting these differentiated regulons and identifying shared targets that appeared at least twice, we successfully pinpointed three differentially expressed targets across both datasets. In this study, we assessed and identified 19 gene regulatory networks that were responsive to the disease. Furthermore, we validated these networks using two bulk RNA datasets of DCM. The elucidation of dysregulated regulons and targets (CDKN1A, SAT1, ZFP36) enhances the molecular understanding of DCM, aiding in the development of tailored therapies for patients.


Subject(s)
Cardiomyopathy, Dilated , Gene Regulatory Networks , Sequence Analysis, RNA , Single-Cell Analysis , Cardiomyopathy, Dilated/genetics , Single-Cell Analysis/methods , Humans , Sequence Analysis, RNA/methods , Gene Expression Profiling , RNA/genetics , RNA/metabolism , Computational Biology/methods , Gene Expression Regulation
5.
BMC Cardiovasc Disord ; 24(1): 307, 2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38886700

ABSTRACT

BACKGROUND: Carney syndrome is an uncommon autosomal disorder closely linked to mutations in the PRKAR1A gene. Skin lesions are the most pronounced feature of Carney syndrome, affecting over 80% of individuals with this condition. This syndrome is characterized by a triad of myxomas, skin pigmentation, and endocrine hyperfunction, featuring multiple endocrine neoplasms with skin and cardiac involvement. Dilated cardiomyopathy, a primary cardiomyopathy, is defined as the dilation and impaired systolic function of the left or both ventricles. Its clinical presentation varies from being asymptomatic to heart failure or sudden cardiac death, making it a leading global cause of heart failure. Currently, Dilated cardiomyopathy has an estimated prevalence of 1/2500-1/250 individuals, predominantly affecting those aged 30-40 years, with a male-to-female ratio of 3:1. This case report describes a heart failure patient with cardiac myxoma caused by Carney syndrome combined with dilated cardiomyopathy. The patient was successfully treated for heart failure by heart transplantation. CASE PRESENTATION: Herein, we report a case of heart failure due to Carney syndrome that resulted in cardiac myxoma combined with dilated cardiomyopathy. A 35-year-old male was admitted to the hospital three years ago because of sudden chest tightness and shortness of breath. Echocardiography indicated myxoma, and a combination of genetic screening and physical examination confirmed Carney syndrome with cardiac myxoma. Following symptomatic management, he was discharged. Surgical interventions were not considered at the time. However, the patient's chest tightness and shortness of breath symptoms worsened, and he returned to the hospital. A New York Heart Association grade IV heart function was confirmed, and echocardiography indicated the presence of dilated cardiomyopathy accompanied by cardiac myxoma. Ultimately, the patient's heart failure was successfully treated with heart transplantation. CONCLUSIONS: Cardiac myxoma caused by Carney syndrome combined with heart failure caused by dilated cardiomyopathy can be resolved by heart transplantation.


Subject(s)
Cardiomyopathy, Dilated , Carney Complex , Heart Failure , Heart Neoplasms , Heart Transplantation , Myxoma , Humans , Cardiomyopathy, Dilated/surgery , Cardiomyopathy, Dilated/etiology , Cardiomyopathy, Dilated/diagnosis , Cardiomyopathy, Dilated/diagnostic imaging , Male , Carney Complex/genetics , Carney Complex/diagnosis , Carney Complex/surgery , Carney Complex/complications , Adult , Myxoma/complications , Myxoma/surgery , Myxoma/diagnostic imaging , Myxoma/diagnosis , Myxoma/genetics , Heart Failure/etiology , Heart Failure/diagnosis , Heart Failure/surgery , Heart Neoplasms/surgery , Heart Neoplasms/complications , Heart Neoplasms/diagnostic imaging , Heart Neoplasms/diagnosis , Heart Neoplasms/genetics , Treatment Outcome , Cyclic AMP-Dependent Protein Kinase RIalpha Subunit/genetics
6.
Cells ; 13(11)2024 May 27.
Article in English | MEDLINE | ID: mdl-38891055

ABSTRACT

Intracellular cargo delivery via distinct transport routes relies on vesicle carriers. A key trafficking route distributes cargo taken up by clathrin-mediated endocytosis (CME) via early endosomes. The highly dynamic nature of the endosome network presents a challenge for its quantitative analysis, and theoretical modelling approaches can assist in elucidating the organization of the endosome trafficking system. Here, we introduce a new computational modelling approach for assessment of endosome distributions. We employed a model of induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) with inherited mutations causing dilated cardiomyopathy (DCM). In this model, vesicle distribution is defective due to impaired CME-dependent signaling, resulting in plasma membrane-localized early endosomes. We recapitulated this in iPSC-CMs carrying two different mutations, TPM1-L185F and TnT-R141W (MUT), using 3D confocal imaging as well as super-resolution STED microscopy. We computed scaled distance distributions of EEA1-positive vesicles based on a spherical approximation of the cell. Employing this approach, 3D spherical modelling identified a bi-modal segregation of early endosome populations in MUT iPSC-CMs, compared to WT controls. Moreover, spherical modelling confirmed reversion of the bi-modal vesicle localization in RhoA II-treated MUT iPSC-CMs. This reflects restored, homogeneous distribution of early endosomes within MUT iPSC-CMs following rescue of CME-dependent signaling via RhoA II-dependent RhoA activation. Overall, our approach enables assessment of early endosome distribution in cell-based disease models. This new method may provide further insight into the dynamics of endosome networks in different physiological scenarios.


Subject(s)
Endosomes , Induced Pluripotent Stem Cells , Myocytes, Cardiac , Humans , Induced Pluripotent Stem Cells/metabolism , Endosomes/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Endocytosis , Mutation/genetics , Computer Simulation , rhoA GTP-Binding Protein/metabolism , Cardiomyopathies/metabolism , Cardiomyopathies/pathology , Imaging, Three-Dimensional , Cardiomyopathy, Dilated/metabolism , Cardiomyopathy, Dilated/pathology , Models, Biological , Tropomyosin/metabolism , Tropomyosin/genetics
7.
BMJ Case Rep ; 17(6)2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38901852

ABSTRACT

Mid-aortic syndrome (MAS) is a rare vascular disease that usually leads to renovascular hypertension. With the predominant manifestations being intractable arterial hypertension and lower extremity arterial insufficiency, it has rarely been associated with dilated cardiomyopathy. We report a young girl with congestive heart failure, where the cause was initially attributed to dilated cardiomyopathy. A repeated echocardiogram 6 months later brought the physician's suspicion of MAS because of the abnormal colour of Doppler from the subcostal view. Further assessment using CT angiography revealed discrete thoracic coarctation at the level of T10, with the narrowest diameter of 2.1 mm, thus confirming the diagnosis. Her inflammatory markers and connective tissue screening were negative. She underwent successful stenting of coarctation of the aorta, which later caused improvement in her cardiac function. We highlighted the importance of looking for treatable causes of dilated cardiomyopathy and vigilant clinical and echocardiogram assessment with high suspicion to diagnose MAS.


Subject(s)
Aortic Coarctation , Cardiomyopathy, Dilated , Humans , Cardiomyopathy, Dilated/diagnosis , Cardiomyopathy, Dilated/diagnostic imaging , Cardiomyopathy, Dilated/complications , Female , Aortic Coarctation/complications , Aortic Coarctation/diagnosis , Aortic Coarctation/diagnostic imaging , Computed Tomography Angiography , Echocardiography , Stents , Diagnosis, Differential , Syndrome , Heart Failure/etiology
8.
PLoS One ; 19(6): e0293105, 2024.
Article in English | MEDLINE | ID: mdl-38889130

ABSTRACT

Obg-like ATPase 1 (OLA1) protein has GTP and ATP hydrolyzing activities and is important for cellular growth and survival. The human OLA1 gene maps to chromosome 2 (locus 2q31.1), near Titin (TTN), which is associated with familial dilated cardiomyopathy (DCM). In this study, we found that expression of OLA1 was significantly downregulated in failing human heart tissue (HF) compared to non-failing hearts (NF). Using the Sanger sequencing method, we characterized the human OLA1 gene and screened for mutations in the OLA1 gene in patients with failing and non-failing hearts. Among failing and non-failing heart patients, we found 15 different mutations in the OLA1 gene, including two transversions, one substitution, one deletion, and eleven transitions. All mutations were intronic except for a non-synonymous 5144A>G, resulting in 254Tyr>Cys in exon 8 of the OLA1 gene. Furthermore, haplotype analysis of these mutations revealed that these single nucleotide polymorphisms (SNPs) are linked to each other, resulting in disease-specific haplotypes. Additionally, to screen the 254Tyr>Cys point mutation, we developed a cost-effective, rapid genetic screening PCR test that can differentiate between homozygous (AA and GG) and heterozygous (A/G) genotypes. Our results demonstrate that this PCR test can effectively screen for OLA1 mutation-associated cardiomyopathy in human patients using easily accessible cells or tissues, such as blood cells. These findings have important implications for the diagnosis and treatment of cardiomyopathy.


Subject(s)
Heart Failure , Polymorphism, Single Nucleotide , Humans , Heart Failure/genetics , Male , Female , Haplotypes , Polymerase Chain Reaction/methods , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/diagnosis , Middle Aged , Adult , Genetic Testing/methods , Mutation , Adenosine Triphosphatases/genetics
9.
J Am Coll Cardiol ; 83(22): 2214-2232, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38811098

ABSTRACT

Arrhythmias frequently accompany heart failure and left ventricular dysfunction. Tachycardias, atrial fibrillation, and premature ventricular contractions can induce a reversible form of dilated cardiomyopathy (CM) known as arrhythmia-induced CM (AiCM). The intriguing question is why certain individuals are more susceptible to AiCM, despite similar arrhythmia burdens. The primary challenge is determining the extent of arrhythmias' contribution to left ventricular systolic dysfunction. AiCM should be considered in patients with a mean heart rate of >100 beats/min, atrial fibrillation, or a PVC burden of >10%. Confirmation of AiCM occurs when CM reverses upon eliminating the responsible arrhythmia. Therapy choice depends on the specific arrhythmia, patient comorbidities, and preferences. After left ventricular function is restored, ongoing follow-up is essential if an abnormal myocardial substrate persists. Accurate diagnosis and treatment of AiCM have the potential to enhance patients' quality of life, improve clinical outcomes, and reduce hospital admissions and overall health care costs.


Subject(s)
Arrhythmias, Cardiac , Humans , Arrhythmias, Cardiac/etiology , Arrhythmias, Cardiac/therapy , Arrhythmias, Cardiac/physiopathology , Cardiomyopathies/etiology , Cardiomyopathies/therapy , Cardiomyopathies/physiopathology , Cardiomyopathies/diagnosis , Cardiomyopathy, Dilated/therapy , Cardiomyopathy, Dilated/physiopathology , Cardiomyopathy, Dilated/etiology
10.
Sci Rep ; 14(1): 11980, 2024 05 25.
Article in English | MEDLINE | ID: mdl-38796549

ABSTRACT

Pathogenic BAG5 variants recently linked to dilated cardiomyopathy (DCM) prompt further investigation into phenotypic, mutational, and pathomechanistic aspects. We explored the clinical and molecular characteristics of DCM associated with BAG5 variants, uncovering the consistently severe manifestations of the disease and its impact on the endoplasmic reticulum (ER) stress response. The analysis involved three siblings affected by DCM and arrhythmia, along with their four unaffected siblings, their unaffected father, and their mother who exhibited arrhythmia. The parents were consanguineous. Exome and Sanger sequencing identified a novel BAG5 variant, c.444_445delGA (p.Lys149AsnfsTer6), homozygous in affected siblings and heterozygous in parents and unaffected siblings. We generated heterozygous and homozygous Bag5 point mutant knock-in (KI) mice and evaluated cardiac pathophysiology under stress conditions, including tunicamycin (TN) administration. Bag5-/- mice displayed no abnormalities up to 12 months old and showed no anomalies during an exercise stress test. However, following TN injection, Bag5-/- mice exhibited significantly reduced left ventricular fractional shortening (LVFS) and ejection fraction (LVEF). Their cardiac tissues exhibited a notable increase in apoptotic cells, despite non-distinctive changes in CHOP and GRP78 levels. Interestingly, only Bag5 KI male mice demonstrated arrhythmia, which was more pronounced in Bag5-/- than in Bag5+/-males. Here, our study reveals a novel BAG5 mutation causing DCM by impairing the ER stress response, with observed sex-specific arrhythmia differences.


Subject(s)
Arrhythmias, Cardiac , Cardiomyopathy, Dilated , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress , Animals , Cardiomyopathy, Dilated/genetics , Endoplasmic Reticulum Stress/genetics , Humans , Arrhythmias, Cardiac/genetics , Male , Female , Mice , Pedigree , Mice, Knockout , Adult , Apoptosis/genetics , Mutation
11.
Sci China Life Sci ; 67(6): 1155-1169, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38811441

ABSTRACT

CFIRL is a long noncoding RNA (lncRNA), we previously identified as the most significantly upregulated lncRNA in the failing hearts of patients with dilated cardiomyopathy (DCM). In this study, we determined the function of CFIRL and its role in DCM. Real-time polymerase chain reaction and in situ hybridization assays revealed that CFIRL was primarily localized in the nucleus of cardiac fibroblasts and robustly increased in failing hearts. Global knockdown or fibroblast-specific knockout of CFIRL attenuated transverse aortic constriction (TAC)-induced cardiac dysfunction and fibrosis in vivo. Overexpression of CFIRL in vitro promoted fibroblast proliferation and aggravated angiotensin II-induced differentiation to myofibroblasts. CFIRL knockdown attenuated these effects. Mechanistically, RNA pull-down assay and gene expression profiling revealed that CFIRL recruited ENO1, a newly identified noncanonical transcriptional factor, to activate IL-6 transcription. IL-6 exerted a paracrine effect on cardiomyocytes to promote cardiac hypertrophy, which can be prevented by CFIRL knockdown. These findings uncover the critical role of CFIRL, a fibroblast-associated lncRNA, in heart failure by facilitating crosstalk between fibroblasts and cardiomyocytes. CFIRL knockdown might be a potent strategy to prevent cardiac remodeling in heart failure, particularly in DCM.


Subject(s)
Cardiomyopathy, Dilated , Fibroblasts , Fibrosis , Myocytes, Cardiac , RNA, Long Noncoding , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/metabolism , Animals , Fibroblasts/metabolism , Male , Humans , Myocytes, Cardiac/metabolism , Mice , Cell Proliferation , Interleukin-6/metabolism , Interleukin-6/genetics , Mice, Inbred C57BL , Myocardium/metabolism , Myocardium/pathology , Myofibroblasts/metabolism , Heart Failure/genetics , Heart Failure/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Cell Differentiation , Gene Knockdown Techniques
12.
Front Immunol ; 15: 1327372, 2024.
Article in English | MEDLINE | ID: mdl-38736889

ABSTRACT

Introduction: Growing evidence from animal models indicates that the myocardium hosts a population of B cells that play a role in the development of cardiomyopathy. However, there is minimal data on human myocardial B cells in the context of cardiomyopathy. Methods: We integrated single-cell and single-nuclei datasets from 45 healthy human hearts, 70 hearts with dilated cardiomyopathy (DCM), and 8 hearts with arrhythmogenic right ventricular cardiomyopathy (ARVC). Interactions between B cells and other cell types were investigated using the CellChat Package. Differential gene expression analysis comparing B cells across conditions was performed using DESeq2. Pathway analysis was performed using Ingenuity, KEGG, and GO pathways analysis. Results: We identified 1,100 B cells, including naive B cells and plasma cells. Cells showed an extensive network of interactions within the healthy myocardium that included outgoing signaling to macrophages, T cells, endothelial cells, and pericytes, and incoming signaling from endothelial cells, pericytes, and fibroblasts. This niche relied on ECM-receptor, contact, and paracrine interactions; and changed significantly in the context of cardiomyopathy, displaying disease-specific features. Differential gene expression analysis showed that in the context of DCM both naive and plasma B cells upregulated several pathways related to immune activation, including upregulation of oxidative phosphorylation, upregulation of leukocyte extravasation, and, in naive B cells, antigen presentation. Discussion: The human myocardium contains naive B cells and plasma cells, integrated into a diverse and dynamic niche that has distinctive features in healthy, DCM, and ARVC. Naive myocardial-associated B cells likely contribute to the pathogenesis of human DCM.


Subject(s)
Arrhythmogenic Right Ventricular Dysplasia , B-Lymphocytes , Cardiomyopathy, Dilated , Myocardium , Humans , Cardiomyopathy, Dilated/immunology , Cardiomyopathy, Dilated/genetics , Arrhythmogenic Right Ventricular Dysplasia/genetics , Arrhythmogenic Right Ventricular Dysplasia/metabolism , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Myocardium/metabolism , Myocardium/immunology , Myocardium/pathology , Male , Female , Cell Communication/immunology , Gene Expression Profiling , Middle Aged , Adult , Transcriptome , Gene Expression Regulation
13.
Genome Biol ; 25(1): 135, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38783323

ABSTRACT

BACKGROUND: Dilated cardiomyopathy (DCM) is one of the most common causes of heart failure. Multiple identified mutations in nexilin (NEXN) have been suggested to be linked with severe DCM. However, the exact association between multiple mutations of Nexn and DCM remains unclear. Moreover, it is critical for the development of precise and effective therapeutics in treatments of DCM. RESULTS: In our study, Nexn global knockout mice and mice carrying human equivalent G645del mutation are studied using functional gene rescue assays. AAV-mediated gene delivery is conducted through systemic intravenous injections at the neonatal stage. Heart tissues are analyzed by immunoblots, and functions are assessed by echocardiography. Here, we identify functional components of Nexilin and demonstrate that exogenous introduction could rescue the cardiac function and extend the lifespan of Nexn knockout mouse models. Similar therapeutic effects are also obtained in G645del mice, providing a promising intervention for future clinical therapeutics. CONCLUSIONS: In summary, we demonstrated that a single injection of AAV-Nexn was capable to restore the functions of cardiomyocytes and extended the lifespan of Nexn knockout and G645del mice. Our study represented a long-term gene replacement therapy for DCM that potentially covers all forms of loss-of-function mutations in NEXN.


Subject(s)
Cardiomyopathy, Dilated , Genetic Therapy , Mice, Knockout , Animals , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/therapy , Mice , Humans , Dependovirus/genetics , Myocytes, Cardiac/metabolism , Disease Models, Animal , Mutation , Genetic Vectors/administration & dosage , Gene Transfer Techniques
14.
Biomolecules ; 14(5)2024 Apr 27.
Article in English | MEDLINE | ID: mdl-38785931

ABSTRACT

Dilated cardiomyopathy (DCM) encompasses various acquired or genetic diseases sharing a common phenotype. The understanding of pathogenetic mechanisms and the determination of the functional effects of each etiology may allow for tailoring different therapeutic strategies. MicroRNAs (miRNAs) have emerged as key regulators in cardiovascular diseases, including DCM. However, their specific roles in different DCM etiologies remain elusive. Here, we applied mRNA-seq and miRNA-seq to identify the gene and miRNA signature from myocardial biopsies from four patients with DCM caused by volume overload (VCM) and four with ischemic DCM (ICM). Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis were used for differentially expressed genes (DEGs). The miRNA-mRNA interactions were identified by Pearson correlation analysis and miRNA target-prediction programs. mRNA-seq and miRNA-seq were validated by qRT-PCR and miRNA-mRNA interactions were validated by luciferase assays. We found 112 mRNAs and five miRNAs dysregulated in VCM vs. ICM. DEGs were positively enriched for pathways related to the extracellular matrix (ECM), mitochondrial respiration, cardiac muscle contraction, and fatty acid metabolism in VCM vs. ICM and negatively enriched for immune-response-related pathways, JAK-STAT, and NF-kappa B signaling. We identified four pairs of negatively correlated miRNA-mRNA: miR-218-5p-DDX6, miR-218-5p-TTC39C, miR-218-5p-SEMA4A, and miR-494-3p-SGMS2. Our study revealed novel miRNA-mRNA interaction networks and signaling pathways for VCM and ICM, providing novel insights into the development of these DCM etiologies.


Subject(s)
Cardiomyopathy, Dilated , MicroRNAs , RNA, Messenger , Humans , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/metabolism , Cardiomyopathy, Dilated/pathology , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Gene Regulatory Networks , Male , Gene Expression Profiling , Gene Expression Regulation , Middle Aged , Female
15.
J Gen Physiol ; 156(6)2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38727632

ABSTRACT

JGP study (Duno-Miranda et al. https://doi.org/10.1085/jgp.202313522) shows that a mutation linked to dilated cardiomyopathy stabilizes ß-cardiac myosin in its autoinhibited, super-relaxed state.


Subject(s)
Mutation , Humans , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/metabolism , Animals
16.
Korean J Radiol ; 25(6): 540-549, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38807335

ABSTRACT

OBJECTIVE: This study investigated the feasibility and prognostic relevance of threshold-based quantification of myocardial delayed enhancement (MDE) on CT in patients with nonischemic dilated cardiomyopathy (NIDCM). MATERIALS AND METHODS: Forty-three patients with NIDCM (59.3 ± 17.1 years; 21 male) were included in the study and underwent cardiac CT and MRI. MDE was quantified manually and with a threshold-based quantification method using cutoffs of 2, 3, and 4 standard deviations (SDs) on three sets of CT images (100 kVp, 120 kVp, and 70 keV). Interobserver agreement in MDE quantification was assessed using the intraclass correlation coefficient (ICC). Agreement between CT and MRI was evaluated using the Bland-Altman method and the concordance correlation coefficient (CCC). Patients were followed up for the subsequent occurrence of the primary composite outcome, including cardiac death, heart transplantation, heart failure hospitalization, or appropriate use of an implantable cardioverter-defibrillator. The Kaplan-Meier method was used to estimate event-free survival according to MDE levels. RESULTS: Late gadolinium enhancement (LGE) was observed in 29 patients (67%, 29/43), and the mean LGE found with the 5-SD threshold was 4.1% ± 3.6%. The 4-SD threshold on 70-keV CT showed excellent interobserver agreement (ICC = 0.810) and the highest concordance with MRI (CCC = 0.803). This method also yielded the smallest bias with the narrowest range of 95% limits of agreement compared to MRI (bias, -0.119%; 95% limits of agreement, -4.216% to 3.978%). During a median follow-up of 1625 days (interquartile range, 712-1430 days), 10 patients (23%, 10/43) experienced the primary composite outcome. Event-free survival significantly differed between risk subgroups divided by the optimal MDE cutoff of 4.3% (log-rank P = 0.005). CONCLUSION: The 4-SD threshold on 70-keV monochromatic CT yielded results comparable to those of MRI for quantifying MDE as a marker of myocardial fibrosis, which showed prognostic value in patients with NIDCM.


Subject(s)
Cardiomyopathy, Dilated , Contrast Media , Feasibility Studies , Fibrosis , Magnetic Resonance Imaging , Tomography, X-Ray Computed , Humans , Male , Cardiomyopathy, Dilated/diagnostic imaging , Female , Middle Aged , Prognosis , Tomography, X-Ray Computed/methods , Fibrosis/diagnostic imaging , Magnetic Resonance Imaging/methods , Myocardium/pathology , Adult , Aged
17.
J Gen Physiol ; 156(6)2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38709176

ABSTRACT

Dilated cardiomyopathy (DCM) is a condition characterized by impaired cardiac function, due to myocardial hypo-contractility, and is associated with point mutations in ß-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 ß-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.


Subject(s)
Cardiac Myosins , Cardiomyopathy, Dilated , Ventricular Myosins , Animals , Humans , Actins/metabolism , Actins/genetics , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/metabolism , Cardiomyopathy, Dilated/physiopathology , Mutation , Myocardial Contraction/physiology , Ventricular Myosins/genetics , Ventricular Myosins/metabolism , Cardiac Myosins/genetics , Cardiac Myosins/metabolism
18.
BMC Cardiovasc Disord ; 24(1): 282, 2024 May 29.
Article in English | MEDLINE | ID: mdl-38811883

ABSTRACT

Sudden cardiac death (SCD) is a major public health issue worldwide. In the young (< 40 years of age), genetic cardiomyopathies and viral myocarditis, sometimes in combination, are the most frequent, but underestimated, causes of SCD. Molecular autopsy is essential for prevention. Several studies have shown an association between genetic cardiomyopathies and viral myocarditis, which is probably underestimated due to insufficient post-mortem investigations. We report on four autopsy cases illustrating the pathogenesis of these combined pathologies. In two cases, a genetic hypertrophic cardiomyopathy was diagnosed in combination with Herpes Virus Type 6 (HHV6) and/or Parvovirus-B19 (PVB19) in the heart. In the third case, autopsy revealed a dilated cardiomyopathy and virological analyses revealed acute myocarditis caused by three viruses: PVB19, HHV6 and Epstein-Barr virus. Genetic analyses revealed a mutation in the gene coding for desmin. The fourth case illustrated a channelopathy and a PVB19/HHV6 coinfection. Our four cases illustrate the highly probable deleterious role of cardiotropic viruses in the occurrence of SCD in subjects with genetic cardiomyopathies. We discuss the pathogenetic link between viral myocarditis and genetic cardiomyopathy. Molecular autopsy is essential in prevention of these SCD, and a close collaboration between cardiologists, pathologists, microbiologists and geneticians is mandatory.


Subject(s)
Autopsy , Death, Sudden, Cardiac , Herpesvirus 6, Human , Myocarditis , Parvovirus B19, Human , Humans , Myocarditis/virology , Myocarditis/pathology , Myocarditis/genetics , Death, Sudden, Cardiac/etiology , Death, Sudden, Cardiac/pathology , Death, Sudden, Cardiac/prevention & control , Male , Adult , Female , Herpesvirus 6, Human/genetics , Herpesvirus 6, Human/isolation & purification , Parvovirus B19, Human/genetics , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/virology , Cardiomyopathy, Dilated/pathology , Roseolovirus Infections/complications , Roseolovirus Infections/virology , Roseolovirus Infections/diagnosis , Roseolovirus Infections/pathology , Cardiomyopathy, Hypertrophic/genetics , Cardiomyopathy, Hypertrophic/pathology , Parvoviridae Infections/complications , Young Adult , Genetic Predisposition to Disease , Fatal Outcome , Epstein-Barr Virus Infections/complications , Herpesvirus 4, Human/genetics , Coinfection , Cause of Death , Mutation , Middle Aged
19.
PLoS Genet ; 20(5): e1011279, 2024 May.
Article in English | MEDLINE | ID: mdl-38748723

ABSTRACT

The leiomodin (Lmod) family of actin-binding proteins play a critical role in muscle function, highlighted by the fact that mutations in all three family members (LMOD1-3) result in human myopathies. Mutations in the cardiac predominant isoform, LMOD2 lead to severe neonatal dilated cardiomyopathy. Most of the disease-causing mutations in the LMOD gene family are nonsense, or frameshift, mutations predicted to result in expression of truncated proteins. However, in nearly all cases of disease, little to no LMOD protein is expressed. We show here that nonsense-mediated mRNA decay, a cellular mechanism which eliminates mRNAs with premature termination codons, underlies loss of mutant protein from two independent LMOD2 disease-causing mutations. Furthermore, we generated steric-blocking oligonucleotides that obstruct deposition of the exon junction complex, preventing nonsense-mediated mRNA decay of mutant LMOD2 transcripts, thereby restoring mutant protein expression. Our investigation lays the initial groundwork for potential therapeutic intervention in LMOD-linked myopathies.


Subject(s)
Codon, Nonsense , Nonsense Mediated mRNA Decay , Humans , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/metabolism , Codon, Nonsense/genetics , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Muscle Proteins/genetics , Muscle Proteins/metabolism , Mutation , Nonsense Mediated mRNA Decay/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism
20.
Int J Mol Sci ; 25(10)2024 May 13.
Article in English | MEDLINE | ID: mdl-38791328

ABSTRACT

Inherited muscular diseases (MDs) are genetic degenerative disorders typically caused by mutations in a single gene that affect striated muscle and result in progressive weakness and wasting in affected individuals. Cardiac muscle can also be involved with some variability that depends on the genetic basis of the MD (Muscular Dystrophy) phenotype. Heart involvement can manifest with two main clinical pictures: left ventricular systolic dysfunction with evolution towards dilated cardiomyopathy and refractory heart failure, or the presence of conduction system defects and serious life-threatening ventricular arrhythmias. The two pictures can coexist. In these cases, heart transplantation (HTx) is considered the most appropriate option in patients who are not responders to the optimized standard therapeutic protocols. However, cardiac transplant is still considered a relative contraindication in patients with inherited muscle disorders and end-stage cardiomyopathies. High operative risk related to muscle impairment and potential graft involvement secondary to the underlying myopathy have been the two main reasons implicated in the generalized reluctance to consider cardiac transplant as a viable option. We report an overview of cardiac involvement in MDs and its possible association with the underlying molecular defect, as well as a systematic review of HTx outcomes in patients with MD-related end-stage dilated cardiomyopathy, published so far in the literature.


Subject(s)
Cardiomyopathy, Dilated , Heart Transplantation , Muscular Dystrophies , Humans , Cardiomyopathy, Dilated/surgery , Heart Transplantation/methods , Muscular Dystrophies/complications
SELECTION OF CITATIONS
SEARCH DETAIL
...