Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.977
Filter
1.
Theranostics ; 14(11): 4462-4480, 2024.
Article in English | MEDLINE | ID: mdl-39113806

ABSTRACT

Rationale: Cardiomyocytes (CMs) undergo dramatic structural and functional changes in postnatal maturation; however, the regulatory mechanisms remain greatly unclear. Cypher/Z-band alternatively spliced PDZ-motif protein (ZASP) is an essential sarcomere component maintaining Z-disc stability. Deletion of mouse Cypher and mutation in human ZASP result in dilated cardiomyopathy (DCM). Whether Cypher/ZASP participates in CM maturation and thereby affects cardiac function has not been answered. Methods: Immunofluorescence, transmission electron microscopy, real-time quantitative PCR, and Western blot were utilized to identify the role of Cypher in CM maturation. Subsequently, RNA sequencing and bioinformatics analysis predicted serum response factor (SRF) as the key regulator. Rescue experiments were conducted using adenovirus or adeno-associated viruses encoding SRF, both in vitro and in vivo. The molecular mechanisms were elucidated through G-actin/F-actin fractionation, nuclear-cytoplasmic extraction, actin disassembly assays, and co-sedimentation assays. Results: Cypher deletion led to impaired sarcomere isoform switch and morphological abnormalities in mitochondria, transverse-tubules, and intercalated discs. RNA-sequencing analysis revealed significant dysregulation of crucial genes related to sarcomere assembly, mitochondrial metabolism, and electrophysiology in the absence of Cypher. Furthermore, SRF was predicted as key transcription factor mediating the transcriptional differences. Subsequent rescue experiments showed that SRF re-expression during the critical postnatal period effectively rectified CM maturation defects and notably improved cardiac function in Cypher-depleted mice. Mechanistically, Cypher deficiency resulted in the destabilization of F-actin and a notable increase in G-actin levels, thereby impeding the nuclear localisation of myocardin-related transcription factor A (MRTFA) and subsequently initiating SRF transcription. Conclusion: Cypher/ZASP plays a crucial role in CM maturation through actin-mediated MRTFA-SRF signalling. The linkage between CM maturation abnormalities and the late-onset of DCM is suggested, providing further insights into the pathogenesis of DCM and potential treatment strategies.


Subject(s)
Actins , Cardiomyopathy, Dilated , Myocytes, Cardiac , Serum Response Factor , Signal Transduction , Trans-Activators , Animals , Myocytes, Cardiac/metabolism , Serum Response Factor/metabolism , Serum Response Factor/genetics , Mice , Actins/metabolism , Trans-Activators/metabolism , Trans-Activators/genetics , Cardiomyopathy, Dilated/metabolism , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/pathology , Sarcomeres/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Humans , Mice, Knockout
2.
Nat Commun ; 15(1): 7000, 2024 Aug 14.
Article in English | MEDLINE | ID: mdl-39143095

ABSTRACT

Mutations in the nuclear envelope (NE) protein lamin A/C (encoded by LMNA), cause a severe form of dilated cardiomyopathy (DCM) with early-onset life-threatening arrhythmias. However, molecular mechanisms underlying increased arrhythmogenesis in LMNA-related DCM (LMNA-DCM) remain largely unknown. Here we show that a frameshift mutation in LMNA causes abnormal Ca2+ handling, arrhythmias and disformed NE in LMNA-DCM patient-specific iPSC-derived cardiomyocytes (iPSC-CMs). Mechanistically, lamin A interacts with sirtuin 1 (SIRT1) where mutant lamin A/C accelerates degradation of SIRT1, leading to mitochondrial dysfunction and oxidative stress. Elevated reactive oxygen species (ROS) then activates the Ca2+/calmodulin-dependent protein kinase II (CaMKII)-ryanodine receptor 2 (RYR2) pathway and aggravates the accumulation of SUN1 in mutant iPSC-CMs, contributing to arrhythmias and NE deformation, respectively. Taken together, the lamin A/C deficiency-mediated ROS disorder is revealed as central to LMNA-DCM development. Manipulation of impaired SIRT1 activity and excessive oxidative stress is a potential future therapeutic strategy for LMNA-DCM.


Subject(s)
Cardiomyopathy, Dilated , Induced Pluripotent Stem Cells , Lamin Type A , Myocytes, Cardiac , Oxidative Stress , Reactive Oxygen Species , Sirtuin 1 , Cardiomyopathy, Dilated/metabolism , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/pathology , Lamin Type A/metabolism , Lamin Type A/genetics , Induced Pluripotent Stem Cells/metabolism , Reactive Oxygen Species/metabolism , Humans , Sirtuin 1/metabolism , Sirtuin 1/genetics , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Phenotype , Arrhythmias, Cardiac/metabolism , Arrhythmias, Cardiac/genetics , Arrhythmias, Cardiac/pathology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Frameshift Mutation , Calcium/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Ryanodine Receptor Calcium Release Channel/genetics , Nuclear Envelope/metabolism , Mitochondria/metabolism , Male , Microtubule-Associated Proteins/metabolism , Microtubule-Associated Proteins/genetics
3.
Nat Cardiovasc Res ; 3(3): 343-355, 2024 Mar.
Article in English | MEDLINE | ID: mdl-39183958

ABSTRACT

Heart failure affects millions of people worldwide, with men exhibiting a higher incidence than women. Our previous work has shown that mosaic loss of the Y chromosome (LOY) in leukocytes is causally associated with an increased risk for heart failure. Here, we show that LOY macrophages from the failing hearts of humans with dilated cardiomyopathy exhibit widespread changes in gene expression that correlate with cardiac fibroblast activation. Moreover, we identify the ubiquitously transcribed t et ratricopeptide Y-linked (Uty) gene in leukocytes as a causal locus for an accelerated progression of heart failure in male mice with LOY. We demonstrate that Uty disruption leads to epigenetic alterations in both monocytes and macrophages, increasing the propensity of differentiation into profibrotic macrophages. Treatment with a transforming growth factor-ß-neutralizing antibody prevented the cardiac pathology associated with Uty deficiency in leukocytes. These findings shed light on the mechanisms that contribute to the higher incidence of heart failure in men.


Subject(s)
Chromosomes, Human, Y , Epigenesis, Genetic , Heart Failure , Animals , Male , Heart Failure/genetics , Heart Failure/pathology , Humans , Chromosomes, Human, Y/genetics , Fibrosis/genetics , Fibrosis/pathology , Macrophages/metabolism , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/pathology , Disease Models, Animal , Mice , Female , Phenotype , Mice, Inbred C57BL , Cells, Cultured , Mice, Knockout
4.
Nat Cardiovasc Res ; 3(2): 140-144, 2024 Feb.
Article in English | MEDLINE | ID: mdl-39196186

ABSTRACT

High-proportion spliced-in titin truncating variants (hiPSI TTNtvs) have been associated with an increased risk of atrial fibrillation, dilated cardiomyopathy (DCM) and heart failure in individuals of European ancestry1. However, similar data in individuals of African ancestry are lacking. Here we examined the association of hiPSI TTNtvs with atrial fibrillation, DCM and heart failure in individuals of African ancestry using data from the All of Us Research Program. Among 38,154 individuals of African ancestry, 169 (0.4%) individuals carried a hiPSI TTNtv. hiPSI TTNtv carriers were at a higher risk of developing atrial fibrillation (adjusted hazard ratio (HRadj) 2.42, 95% confidence interval (CI) 1.52-3.85), DCM (HRadj 2.82, 95% CI 1.81-4.39) and heart failure (HRadj 2.07, 95% CI 1.43-3.00) compared with noncarriers. The association of hiPSI TTNtvs with atrial fibrillation, DCM and heart failure was similar in individuals of African ancestry and those of European ancestry. Therefore, genetic testing for hiPSI TTNtvs may permit early identification of carriers and support preventive measures to reduce the likelihood of heart failure development both in individuals of European ancestry and in individuals of African ancestry.


Subject(s)
Atrial Fibrillation , Cardiomyopathy, Dilated , Connectin , Genetic Predisposition to Disease , Heart Failure , Adult , Aged , Female , Humans , Male , Middle Aged , Atrial Fibrillation/genetics , Black or African American/genetics , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/ethnology , Connectin/genetics , Heart Failure/genetics , Heart Failure/ethnology , Phenotype , Risk Assessment , Risk Factors , RNA Splicing , United States/epidemiology , White/genetics
5.
Ital J Pediatr ; 50(1): 156, 2024 Aug 26.
Article in English | MEDLINE | ID: mdl-39183344

ABSTRACT

BACKGROUND: Dilated cardiomyopathy (DCM) is an etiologically heterogeneous group of diseases of the myocardium. With the rapid evolution in laboratory investigations, genetic background is increasingly determined including many genes with variable penetrance and expressivity. Biallelic NEXN variants are rare in humans and associated with poor prognosis: fetal and perinatal death or severe DCMs in infants. CASE PRESENTATION: We describe two male infants with prenatal diagnosis of dilated cardiomyopathy with impaired ventricular contractility. One of the patients showed hydrops and polyhydramnios. Postnatally, a DCM with severely reduced systolic function was confirmed and required medical treatment. In patient 1, Whole Exome Sequencing (WES) revealed a homozygous NEXN variant: c.1156dup (p.Met386fs) while in patient 2 a custom Next Generation Sequencing (NGS) panel revealed the homozygous NEXN variant c.1579_1584delp. (Glu527_Glu528del). These NEXN variants have not been previously described. Unlike the unfavorable prognosis described for biallelic NEXN variants, we observed in both our patients a favorable clinical course over time. CONCLUSION: This report might help to broaden the present knowledge regarding NEXN biallelic variants and their clinical expression. It might be worthy to consider the inclusion of the NEXN gene sequencing in the investigation of pediatric patients with DCM.


Subject(s)
Cardiomyopathy, Dilated , Humans , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/diagnosis , Male , Infant, Newborn , Female , Exome Sequencing , Pregnancy
6.
Nat Commun ; 15(1): 5812, 2024 Jul 10.
Article in English | MEDLINE | ID: mdl-38987251

ABSTRACT

RagGTPases (Rags) play an essential role in the regulation of cell metabolism by controlling the activities of both mechanistic target of rapamycin complex 1 (mTORC1) and Transcription factor EB (TFEB). Several diseases, herein named ragopathies, are associated to Rags dysfunction. These diseases may be caused by mutations either in genes encoding the Rags, or in their upstream regulators. The resulting phenotypes may encompass a variety of clinical features such as cataract, kidney tubulopathy, dilated cardiomyopathy and several types of cancer. In this review, we focus on the key clinical, molecular and physio-pathological features of ragopathies, aiming to shed light on their underlying mechanisms.


Subject(s)
Mutation , Humans , Neoplasms/genetics , Neoplasms/pathology , Neoplasms/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Mechanistic Target of Rapamycin Complex 1/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Animals , Cataract/genetics , Cardiomyopathy, Dilated/genetics
7.
Scand Cardiovasc J ; 58(1): 2379356, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39046218

ABSTRACT

AIMS: This study aimed to assess the practicality of using a stepwise pedigree-based approach to differentiate between familial and sporadic Dilated Cardiomyopathy (DCM), while also considering timing of the genetic analysis. The analysis includes an examination of the extent to which complete family investigations were conducted in real-world scenarios as well as the length of the investigation. METHODS: The stepwise pedigree approach involved conducting a comprehensive family history spanning 3 to 4 generations, reviewing medical records of relatives, and conducting clinical screening using echocardiography and electrocardiogram on first-degree relatives. Familial DCM was diagnosed when at least 2 family members were found to have DCM, and genetic analysis was considered as an option. This study involved a manual review of all DCM investigations conducted at the Centre of Cardiovascular Genetics at Umeå University Hospital, where the stepwise pedigree approach has been employed since 2007. RESULTS: The investigation process had a mean duration of 643 days (95% CI 560.5-724.9). Of the investigations preformed, 94 (68%) were complete, 12 (9%) were ongoing, and 33 (24%) were prematurely terminated and thus incomplete. At the conclusion of the investigations, 55 cases (43%) were classified as familial DCM, 50 (39%) as sporadic DCM, and 22 (18%) remained unassessed due to incomplete pedigrees. Among the familial cases, genetic verification was achieved in 40%. CONCLUSION: The stepwise pedigree approach is time consuming, and the investigations are often incomplete which may suggest that a more direct approach to genetic analysis, may be warranted.


Subject(s)
Cardiomyopathy, Dilated , Genetic Predisposition to Disease , Genetic Testing , Heredity , Pedigree , Phenotype , Predictive Value of Tests , Humans , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/diagnosis , Retrospective Studies , Female , Male , Middle Aged , Adult , Echocardiography , Medical History Taking , Electrocardiography , Time Factors , Aged , Risk Factors
8.
Int J Med Sci ; 21(9): 1769-1782, 2024.
Article in English | MEDLINE | ID: mdl-39006834

ABSTRACT

Dilated cardiomyopathy (DCM) causes heart failure and sudden death. Epigenetics is crucial in cardiomyopathy susceptibility and progression; however, the relationship between epigenetics, particularly DNA methylation, and DCM remains unknown. Therefore, this study identified aberrantly methylated differentially expressed genes (DEGs) associated with DCM using bioinformatics analysis and characterized their clinical utility in DCM. DNA methylation expression profiles and transcriptome data from public datasets of human DCM and healthy control cardiac tissues were obtained from the Gene Expression Omnibus public datasets. Then an epigenome-wide association study was performed. DEGs were identified in both DCM and healthy control cardiac tissues. In total, 3,353 cytosine-guanine dinucleotide sites annotated to 2,818 mRNAs were identified, and 479 DCM-related genes were identified. Subsequently, core genes were screened using logistic, least absolute shrinkage and selection operator, random forest, and support vector machine analyses. The overlapping of these genes resulted in DEGs with abnormal methylation patterns. Cross-tabulation analysis identified 8 DEGs with abnormal methylation. Real-time quantitative polymerase chain reaction confirmed the expression of aberrantly methylated DEGs in mice. In DCM murine cardiac tissues, the expressions of SLC16A9, SNCA, PDE5A, FNDC1, and HTRA1 were higher compared to normal murine cardiac tissues. Moreover, logistic regression model associated with aberrantly methylated DEGs was developed to evaluate the diagnostic value, and the area under the receiver operating characteristic curve was 0.949, indicating that the diagnostic model could reliably distinguish DCM from non-DCM samples. In summary, our study identified 5 DEGs through integrated bioinformatic analysis and in vivo experiments, which could serve as potential targets for further comprehensive investigation.


Subject(s)
Cardiomyopathy, Dilated , Computational Biology , DNA Methylation , Gene Expression Profiling , Cardiomyopathy, Dilated/genetics , DNA Methylation/genetics , Humans , Animals , Mice , Epigenesis, Genetic , Transcriptome/genetics , Male , Gene Expression Regulation/genetics
9.
J Clin Invest ; 134(12)2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38950288

ABSTRACT

Research advances over the past 30 years have confirmed a critical role for genetics in the etiology of dilated cardiomyopathies (DCMs). However, full knowledge of the genetic architecture of DCM remains incomplete. We identified candidate DCM causal gene, C10orf71, in a large family with 8 patients with DCM by whole-exome sequencing. Four loss-of-function variants of C10orf71 were subsequently identified in an additional group of492 patients with sporadic DCM from 2 independent cohorts. C10orf71 was found to be an intrinsically disordered protein specifically expressed in cardiomyocytes. C10orf71-KO mice had abnormal heart morphogenesis during embryonic development and cardiac dysfunction as adults with altered expression and splicing of contractile cardiac genes. C10orf71-null cardiomyocytes exhibited impaired contractile function with unaffected sarcomere structure. Cardiomyocytes and heart organoids derived from human induced pluripotent stem cells with C10orf71 frameshift variants also had contractile defects with normal electrophysiological activity. A rescue study using a cardiac myosin activator, omecamtiv mecarbil, restored contractile function in C10orf71-KO mice. These data support C10orf71 as a causal gene for DCM by contributing to the contractile function of cardiomyocytes. Mutation-specific pathophysiology may suggest therapeutic targets and more individualized therapy.


Subject(s)
Cardiomyopathy, Dilated , Frameshift Mutation , Mice, Knockout , Myocytes, Cardiac , Organoids , Adult , Animals , Female , Humans , Male , Mice , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/pathology , Cardiomyopathy, Dilated/metabolism , Disease Models, Animal , Myocardial Contraction/genetics , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Organoids/metabolism , Organoids/pathology
10.
Scand Cardiovasc J ; 58(1): 2373083, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39024033

ABSTRACT

OBJECTIVE: This paper was performed to decipher the serum microRNA (miR)-125b-5p expression in patients with dilated cardiomyopathy (DCM) combined with heart failure (HF) and its effect on myocardial fibrosis. METHODS: Serum miR-125b-5p expression, LVEDD, LVESD, LVEF, LVFS, and NT-proBNP levels were evaluated in clinical samples. A rat DCM model was established by continuous intraperitoneal injection of adriamycin and treated with miR-125b-5p agomir and its negative control. Cardiac function, serum TNF-α, hs-CRP, and NT-proBNP levels, pathological changes in myocardial tissues, cardiomyocyte apoptosis, and the expression levels of miR-125b-5p and fibrosis-related factors were detected in rats. RESULTS: In comparison to the control group, the case group had higher levels of LVEDD, LVESD, and NT-pro-BNP, and lower levels of LVEF, LVFS, and miR-125b-5p expression levels. Overexpression of miR-125b-5p effectively led to the improvement of cardiomyocyte hypertrophy and collagen arrangement disorder in DCM rats, the reduction of blue-stained collagen fibers in the interstitial myocardium, the reduction of the levels of TNF-α, hs-CRP, and NT-proBNP and the expression levels of TGF-1ß, Collagen I, and α-SMA, and the reduction of the number of apoptosis in cardiomyocytes. CONCLUSION: Overexpression of miR-125b-5p is effective in ameliorating myocardial fibrosis.


Subject(s)
Apoptosis , Cardiomyopathy, Dilated , Heart Failure , MicroRNAs , Myocardium , Ventricular Function, Left , Adult , Aged , Animals , Female , Humans , Male , Middle Aged , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/blood , Cardiomyopathy, Dilated/pathology , Case-Control Studies , Circulating MicroRNA/blood , Circulating MicroRNA/genetics , Disease Models, Animal , Fibrosis , Heart Failure/blood , Heart Failure/genetics , Heart Failure/metabolism , Heart Failure/pathology , MicroRNAs/blood , MicroRNAs/genetics , MicroRNAs/metabolism , Myocardium/pathology , Myocardium/metabolism , Myocytes, Cardiac/pathology , Myocytes, Cardiac/metabolism , Natriuretic Peptide, Brain/blood , Natriuretic Peptide, Brain/genetics , Peptide Fragments/blood , Rats, Sprague-Dawley , Stroke Volume , Ventricular Remodeling
11.
Stem Cell Res ; 80: 103497, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39053319

ABSTRACT

A newly identified pathogenic variant (A527G) in alpha B-crystallin (αB-crystallin) has been linked to congenital cataract and young-onset dilated cardiomyopathy (DCM) within a Dutch family, although the disease mechanism remains unclear. Four human induced pluripotent stem cell (hiPSC) clones were generated from three symptomatic patients carrying the A527G variant, and one healthy proband. Peripheral blood mononuclear cells (PBMCs) were reprogrammed using integration-free Sendai viral pluripotency vectors. The established hiPSCs clones exhibited regular ESC-like morphology, expression of pluripotency markers, and normal karyotyping. These hiPSC lines can facilitate future studies to understand the chaperone function and its role in DCM disease progression.


Subject(s)
Induced Pluripotent Stem Cells , alpha-Crystallin B Chain , Humans , Induced Pluripotent Stem Cells/metabolism , alpha-Crystallin B Chain/genetics , alpha-Crystallin B Chain/metabolism , Male , Female , Mutation , Cell Line , Adult , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/pathology
12.
Stem Cell Res ; 80: 103498, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39067410

ABSTRACT

Dilated cardiomyopathy (DCM) is a disorder of cardiac ventricular dilation and contractile dysfunction that often progresses to heart failure. Multiple genes have been associated with DCM, including SCN5A which has been linked to 2 % of all DCM cases. Peripheral mononuclear blood cells from DCM patients with SCN5A variants (c.2440C>T and c.665G>A) were utilized to generate two human induced pluripotent stem cell (iPSC) lines. Both lines exhibited typical iPSC morphology, expressed pluripotency markers, normal karyotypes, and trilineage differentiation capabilities. These lines offer valuable resources for investigating the mechanism of SCN5A-associated DCM, facilitating studies of ion channel protein involvement in the disease.


Subject(s)
Cardiomyopathy, Dilated , Induced Pluripotent Stem Cells , NAV1.5 Voltage-Gated Sodium Channel , Humans , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/pathology , Induced Pluripotent Stem Cells/metabolism , NAV1.5 Voltage-Gated Sodium Channel/genetics , NAV1.5 Voltage-Gated Sodium Channel/metabolism , Cell Differentiation , Male , Cell Line , Female
13.
Clin Sci (Lond) ; 138(15): 941-962, 2024 Aug 07.
Article in English | MEDLINE | ID: mdl-39018488

ABSTRACT

Atrial fibrillation (AF) remains challenging to prevent and treat. A key feature of AF is atrial enlargement. However, not all atrial enlargement progresses to AF. Atrial enlargement in response to physiological stimuli such as exercise is typically benign and reversible. Understanding the differences in atrial function and molecular profile underpinning pathological and physiological atrial remodelling will be critical for identifying new strategies for AF. The discovery of molecular mechanisms responsible for pathological and physiological ventricular hypertrophy has uncovered new drug targets for heart failure. Studies in the atria have been limited in comparison. Here, we characterised mouse atria from (1) a pathological model (cardiomyocyte-specific transgenic (Tg) that develops dilated cardiomyopathy [DCM] and AF due to reduced protective signalling [PI3K]; DCM-dnPI3K), and (2) a physiological model (cardiomyocyte-specific Tg with an enlarged heart due to increased insulin-like growth factor 1 receptor; IGF1R). Both models presented with an increase in atrial mass, but displayed distinct functional, cellular, histological and molecular phenotypes. Atrial enlargement in the DCM-dnPI3K Tg, but not IGF1R Tg, was associated with atrial dysfunction, fibrosis and a heart failure gene expression pattern. Atrial proteomics identified protein networks related to cardiac contractility, sarcomere assembly, metabolism, mitochondria, and extracellular matrix which were differentially regulated in the models; many co-identified in atrial proteomics data sets from human AF. In summary, physiological and pathological atrial enlargement are associated with distinct features, and the proteomic dataset provides a resource to study potential new regulators of atrial biology and function, drug targets and biomarkers for AF.


Subject(s)
Atrial Fibrillation , Atrial Remodeling , Heart Atria , Mice, Transgenic , Myocytes, Cardiac , Atrial Fibrillation/physiopathology , Atrial Fibrillation/metabolism , Atrial Fibrillation/genetics , Animals , Heart Atria/metabolism , Heart Atria/physiopathology , Heart Atria/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Receptor, IGF Type 1/metabolism , Receptor, IGF Type 1/genetics , Cardiomyopathy, Dilated/physiopathology , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/metabolism , Cardiomyopathy, Dilated/pathology , Disease Models, Animal , Fibrosis , Mice , Humans , Signal Transduction , Phosphatidylinositol 3-Kinases/metabolism , Heart Failure/physiopathology , Heart Failure/genetics , Heart Failure/metabolism , Heart Failure/pathology
14.
Biochim Biophys Acta Mol Cell Res ; 1871(7): 119804, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39084528

ABSTRACT

BACKGROUND: Myocardial fibrosis is an important pathological feature of dilated cardiomyopathy (DCM). The roles of SOCS2 in fibrosis of different organs are controversial. Herein, we investigated the function and potential mechanism of SOCS2 in myocardial fibrosis. METHODS: Bioinformatics, immunohistochemistry (IHC), immunofluorescence (IF), western blot (WB), real-time fluorescence quantitative PCR (qPCR), rat primary myocardial fibroblasts (rCFs) culture, doxorubicin (DOX) induced mouse dilated cardiomyopathy (DCM) model, and in vivo adeno-associated virus (AAV) infection were used to explore the role of SOCS2 in DCM. RESULTS: Bioinformatics analysis showed that SOCS2 was positively correlated with fibrosis related factors. SOCS2 was significantly upregulated in patients and mice with DCM. In vivo experiments showed that targeted inhibition of cardiac SOCS2 could improve mouse cardiac function and alleviate myocardial fibrosis. Further research demonstrated that SOCS2 promoted the transformation of myofibroblasts. Knockdown of SOCS2 reduced the nuclear localization of ß-catenin, which inhibited the fibrogenic effect of Wnt/ß-catenin pathway. In addition, bioinformatics analysis suggested that lymphoid enhancer binding factor 1 (LEF1) was significantly positively correlated with SOCS2. Finally, dual luciferase assays demonstrated that LEF1 could bind to the promoter region of SOCS2, thereby mediating its transcriptional activation. CONCLUSION: SOCS2 could activate the Wnt/ß-catenin by regulating the nuclear translocation of ß-catenin, which induces the transcriptional activation of SOCS2. Overall, these results indicated a positive feedback activation phenomenon between SOCS2, ß-catenin and LEF1 in DCM. These results suggested that inhibition of SOCS2 could effectively alleviate the progression of myocardial fibrosis and improve cardiac function.


Subject(s)
Fibrosis , Myocardium , Suppressor of Cytokine Signaling Proteins , beta Catenin , Animals , beta Catenin/metabolism , beta Catenin/genetics , Mice , Fibrosis/metabolism , Humans , Rats , Myocardium/metabolism , Myocardium/pathology , Male , Suppressor of Cytokine Signaling Proteins/metabolism , Suppressor of Cytokine Signaling Proteins/genetics , Cardiomyopathy, Dilated/metabolism , Cardiomyopathy, Dilated/pathology , Cardiomyopathy, Dilated/genetics , Wnt Signaling Pathway , Disease Models, Animal , Cell Nucleus/metabolism , Myofibroblasts/metabolism , Myofibroblasts/pathology , Mice, Inbred C57BL
15.
Comput Biol Med ; 179: 108901, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39029429

ABSTRACT

BACKGROUND: Dilated cardiomyopathy (DCM) is a common cause of heart failure. However, the role of cellular senescence in DCM has not been fully elucidated. Here, we aimed to investigate senescence in DCM, identify senescence related characteristic genes, and explore the potential small molecule compounds for DCM treatment. METHODS: DCM-associated datasets and senescence-related genes were respectively obtained from Gene Expression Omnibus (GEO) database and CellAge database. The characteristic genes were identified through methods including weighted gene co-expression network analysis (WGCNA), least absolute shrinkage and selection operator (LASSO), and random forest. The expression of characteristic genes was verified in the mouse DCM model. Moreover, the CIBERSORT algorithm was applied to analyze immune characteristics of DCM. Finally, several therapeutic compounds were predicted by CMap analysis, and the potential mechanism of chlorogenic acid (CGA) was investigated by molecular docking and molecular dynamics simulation. RESULTS: Three DCM- and senescence-related characteristic genes (MME, GNMT and PLA2G2A) were ultimately identified through comprehensive transcriptome analysis, and were experimentally verified in the doxorubicin induced mouse DCM. Meanwhile, the established diagnostic model, derived from dataset analysis, showed ideal diagnostic performance for DCM. Immune cell infiltration analysis suggested dysregulation of inflammation in DCM, and the characteristic genes were significantly associated with invasive immune cells. Finally, based on the specific gene expression profile of DCM, several potential therapeutic compounds were predicted through CMap analysis. In addition, molecular docking and molecular dynamics simulations suggested that CGA could bind to the active pocket of MME protein. CONCLUSION: Our study presents three characteristic genes (MME, PLA2G2A, and GNMT) and a novel senescence-based diagnostic nomogram, and discusses potential therapeutic compounds, providing new insights into the diagnosis and treatment of DCM.


Subject(s)
Cardiomyopathy, Dilated , Gene Expression Profiling , Transcriptome , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/drug therapy , Cardiomyopathy, Dilated/metabolism , Mice , Animals , Transcriptome/drug effects , Cellular Senescence/drug effects , Cellular Senescence/genetics , Humans , Chlorogenic Acid/pharmacology , Molecular Docking Simulation , Male
16.
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
18.
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
19.
Mol Genet Genomic Med ; 12(6): e2486, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38924380

ABSTRACT

BACKGROUND: Dilated cardiomyopathy (DCM) is characterized by dilatation of the left ventricle, systolic dysfunction, and normal or reduced thickness of the left ventricular wall. It is a leading cause of heart failure and cardiac death at a young age. Cases with neonatal onset DCM were correlated with severe clinical presentation and poor prognosis. A monogenic molecular etiology accounts for nearly half of cases. FAMILY DESCRIPTION: Here, we report a family with three deceased offspring at the age of 1 year old. The autopsy of the first deceased infant revealed a DCM. The second infant presented a DCM phenotype with a severely reduced Left Ventricular Ejection Fraction (LVEF) of 10%. Similarly, the third infant showed a severe DCM phenotype with LVEF of 30% as well, in addition to eccentric mitral insufficiency. RESULTS: Exome sequencing was performed for the trio (the second deceased infant and her parents). Data analysis following the autosomal dominant and recessive patterns of inheritance was carried out along with a mitochondrial pathways-based analysis. We identified a homozygous frameshift variant in the TNNI3 gene (c.204delG; p.(Arg69AlafsTer8)). This variant has been recently reported in the ClinVar database in association with cardiac phenotypes as pathogenic or likely pathogenic and classified as pathogenic according to ACMG. CONCLUSION: Genetic counseling was provided for the family and a prenatal diagnosis of choronic villus was proposed in the absence of pre-implantation genetic diagnosis possibilities. Our study expands the case series of early-onset DCM patients with a protein-truncating variant in the TNNI3 gene by reporting three affected infant siblings.


Subject(s)
Cardiomyopathy, Dilated , Consanguinity , Frameshift Mutation , Homozygote , Pedigree , Humans , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/pathology , Female , Male , Infant , Phenotype , Troponin I
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL