Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 3.791
Filtrer
1.
Sci Adv ; 10(31): eadp0443, 2024 Aug 02.
Article de Anglais | MEDLINE | ID: mdl-39093974

RÉSUMÉ

Mitochondrial fusion and fission accompany adaptive responses to stress and altered metabolic demands. Inner membrane fusion and cristae morphogenesis depends on optic atrophy 1 (Opa1), which is expressed in different isoforms and is cleaved from a membrane-bound, long to a soluble, short form. Here, we have analyzed the physiological role of Opa1 isoforms and Opa1 processing by generating mouse lines expressing only one cleavable Opa1 isoform or a non-cleavable variant thereof. Our results show that expression of a single cleavable or non-cleavable Opa1 isoform preserves embryonic development and the health of adult mice. Opa1 processing is dispensable under metabolic and thermal stress but prolongs life span and protects against mitochondrial cardiomyopathy in OXPHOS-deficient Cox10-/- mice. Mechanistically, loss of Opa1 processing disturbs the balance between mitochondrial biogenesis and mitophagy, suppressing cardiac hypertrophic growth in Cox10-/- hearts. Our results highlight the critical regulatory role of Opa1 processing, mitochondrial dynamics, and metabolism for cardiac hypertrophy.


Sujet(s)
Cardiomyopathies , dGTPases , Animaux , dGTPases/métabolisme , dGTPases/génétique , Souris , Cardiomyopathies/métabolisme , Cardiomyopathies/génétique , Cardiomyopathies/anatomopathologie , Dynamique mitochondriale , Mitophagie/génétique , Souris knockout , Isoformes de protéines/métabolisme , Isoformes de protéines/génétique , Mitochondries/métabolisme , Modèles animaux de maladie humaine , Développement embryonnaire/génétique
2.
Int J Mol Sci ; 25(15)2024 Jul 27.
Article de Anglais | MEDLINE | ID: mdl-39125771

RÉSUMÉ

Cardiomyopathy is the predominant defect in Barth syndrome (BTHS) and is caused by a mutation of the X-linked Tafazzin (TAZ) gene, which encodes an enzyme responsible for remodeling mitochondrial cardiolipin. Despite the known importance of mitochondrial dysfunction in BTHS, how specific TAZ mutations cause diverse BTHS heart phenotypes remains poorly understood. We generated a patient-tailored CRISPR/Cas9 knock-in mouse allele (TazPM) that phenocopies BTHS clinical traits. As TazPM males express a stable mutant protein, we assessed cardiac metabolic dysfunction and mitochondrial changes and identified temporally altered cardioprotective signaling effectors. Specifically, juvenile TazPM males exhibit mild left ventricular dilation in systole but have unaltered fatty acid/amino acid metabolism and normal adenosine triphosphate (ATP). This occurs in concert with a hyperactive p53 pathway, elevation of cardioprotective antioxidant pathways, and induced autophagy-mediated early senescence in juvenile TazPM hearts. However, adult TazPM males exhibit chronic heart failure with reduced growth and ejection fraction, cardiac fibrosis, reduced ATP, and suppressed fatty acid/amino acid metabolism. This biphasic changeover from a mild-to-severe heart phenotype coincides with p53 suppression, downregulation of cardioprotective antioxidant pathways, and the onset of terminal senescence in adult TazPM hearts. Herein, we report a BTHS genotype/phenotype correlation and reveal that absent Taz acyltransferase function is sufficient to drive progressive cardiomyopathy.


Sujet(s)
Acyltransferases , Syndrome de Barth , Cardiomyopathies , Syndrome de Barth/génétique , Syndrome de Barth/métabolisme , Syndrome de Barth/anatomopathologie , Animaux , Souris , Acyltransferases/génétique , Cardiomyopathies/génétique , Cardiomyopathies/métabolisme , Cardiomyopathies/anatomopathologie , Mâle , Humains , Mutation ponctuelle , Modèles animaux de maladie humaine , Facteurs de transcription/génétique , Facteurs de transcription/métabolisme , Phénotype
3.
J Cardiovasc Pharmacol ; 84(2): 239-249, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-39115722

RÉSUMÉ

ABSTRACT: The study aimed to investigate the pathogenesis of sepsis-induced cardiomyopathy, a leading cause of mortality in septic patients. Transcriptome data from cecal ligation and puncture-induced septic mice were analyzed at different time points (24, 48, and 72 hours) using GSE171546 data. Through weighted gene co-expression network analysis, time series, and differential expression analyses, key time-series differentially expressed genes were identified. In addition, single-cell sequencing data (GSE207363) were used for both differential and pseudotime analyses to pinpoint differentially expressed genes specific to endothelial cells. The study highlighted Spock2, S100a9, S100a8, and Xdh as differential genes specific to endothelial cells in a time-dependent manner. Immunofluorescence validation confirmed the increased expression of SPOCK2 in the endothelial cells of cecal ligation and puncture-induced septic mice. Furthermore, in vitrostudies showed that deletion of Spock2 significantly increased LPS-induced apoptosis and necrosis in human umbilical vein endothelial cells. In conclusion, SPOCK2 expression was increased in septic cardiac endothelial cells and LPS-induced human umbilical vein endothelial cells and may play a protective role.


Sujet(s)
Apoptose , Cardiomyopathies , Modèles animaux de maladie humaine , Cellules endothéliales de la veine ombilicale humaine , Souris de lignée C57BL , Sepsie , Animaux , Sepsie/métabolisme , Sepsie/génétique , Sepsie/complications , Humains , Cellules endothéliales de la veine ombilicale humaine/métabolisme , Cellules endothéliales de la veine ombilicale humaine/anatomopathologie , Cardiomyopathies/métabolisme , Cardiomyopathies/génétique , Cardiomyopathies/anatomopathologie , Mâle , Facteurs temps , Transcriptome , Cellules cultivées , Souris knockout , Cellules endothéliales/métabolisme , Cellules endothéliales/anatomopathologie , Réseaux de régulation génique , Nécrose , Bases de données génétiques , Transduction du signal , Analyse de profil d'expression de gènes , Régulation de l'expression des gènes , Lipopolysaccharides/pharmacologie , Régulation positive , Analyse sur cellule unique , Souris , Calgranuline B
4.
Cells ; 13(15)2024 Jul 27.
Article de Anglais | MEDLINE | ID: mdl-39120296

RÉSUMÉ

Arrhythmogenic cardiomyopathy (AC) is a hereditary cardiac disorder characterized by the gradual replacement of cardiomyocytes with fibrous and adipose tissue, leading to ventricular wall thinning, chamber dilation, arrhythmias, and sudden cardiac death. Despite advances in treatment, disease management remains challenging. Animal models, particularly mice and zebrafish, have become invaluable tools for understanding AC's pathophysiology and testing potential therapies. Mice models, although useful for scientific research, cannot fully replicate the complexity of the human AC. However, they have provided valuable insights into gene involvement, signalling pathways, and disease progression. Zebrafish offer a promising alternative to mammalian models, despite the phylogenetic distance, due to their economic and genetic advantages. By combining animal models with in vitro studies, researchers can comprehensively understand AC, paving the way for more effective treatments and interventions for patients and improving their quality of life and prognosis.


Sujet(s)
Modèles animaux de maladie humaine , Animaux , Humains , Danio zébré , Troubles du rythme cardiaque/anatomopathologie , Troubles du rythme cardiaque/physiopathologie , Troubles du rythme cardiaque/génétique , Dysplasie ventriculaire droite arythmogène/génétique , Dysplasie ventriculaire droite arythmogène/anatomopathologie , Souris , Cardiomyopathies/anatomopathologie , Cardiomyopathies/génétique
5.
BMC Genomics ; 25(1): 707, 2024 Jul 20.
Article de Anglais | MEDLINE | ID: mdl-39033293

RÉSUMÉ

Most studied, investigating transcriptional changes in myocardial biopsies focus on human genes. However, the presence and potential consequence of persistent expression of viral genes within the myocardium is unclear. The aim of the study was to analyze viral gene expression in RNAseq data from endomyocardial biopsies. The NCBI Bioproject library was screened for published projects that included bulk RNA sequencing data from endomyocardial biopsies from both healthy and diseased patients with a sample size greater than 20. Diseased patients with hypertrophic, dilated, and ischemic cardiomyopathies were included. A total of 507 patients with 507 samples from 6 bioprojects were included and mapped to the human genome (hg38). Unmappable sequences were extracted and mapped to an artificial 'super-virus' genome comprising 12,182 curated viral reference genomes. Subsequently, the sequences were reiteratively permutated and mapped again to account for randomness. In total, sequences from 68 distinct viruses were found, all of which were potentially human pathogenic. No increase in cardiotropic viruses was found in patients with dilated cardiomyopathy. However, the expression levels of the particle forming human endogenous retrovirus K were significantly increased (q < 0.0003, ANOVA). Higher expression levels were associated with increased expression in mitochondrial pathways such as oxidative phosphorylation (p < 0.0001). In Conclusion, expression of human endogenous retrovirus K is significantly increased in patients with dilated cardiomyopathy, which in turn was associated with transcriptional alterations in major cellular pathways.


Sujet(s)
Cardiomyopathies , Myocarde , Humains , Cardiomyopathies/virologie , Cardiomyopathies/génétique , Cardiomyopathies/anatomopathologie , Cardiomyopathies/métabolisme , Biopsie , Myocarde/métabolisme , Myocarde/anatomopathologie , Rétrovirus endogènes/génétique , Analyse de profil d'expression de gènes , Transcriptome
6.
Sci Rep ; 14(1): 15141, 2024 07 02.
Article de Anglais | MEDLINE | ID: mdl-38956129

RÉSUMÉ

Pediatric cardiomyopathies are mostly attributed to variants in sarcomere-related genes. Unfortunately, the genetic architecture of pediatric cardiomyopathies has never been previously studied in Jordan. We sought to uncover the genetic landscape of 14 patients from nine families with several subtypes of pediatric cardiomyopathies in Jordan using Exome sequencing (ES). Our investigation identified pathogenic and likely pathogenic variants in seven out of nine families (77.8%), clustering in sarcomere-related genes. Surprisingly, phenocopies of sarcomere-related hypertrophic cardiomyopathies were evident in probands with glycogen storage disorder and mitochondrial-related disease. Our study underscored the significance of streamlining ES or expanding cardiomyopathy-related gene panels to identify plausible phenocopies of sarcomere-related cardiomyopathies. Our findings also pointed out the need for genetic testing in patients with cardiomyopathy and their at-risk family members. This can potentially lead to better management strategies, enabling early interventions, and ultimately enhancing their prognosis. Finally, our findings provide an initial contribution to the currently absent knowledge about the molecular underpinnings of cardiomyopathies in Jordan.


Sujet(s)
Cardiomyopathies , Pedigree , Sarcomères , Humains , Jordanie , Mâle , Femelle , Sarcomères/génétique , Enfant , Cardiomyopathies/génétique , Cardiomyopathies/diagnostic , Enfant d'âge préscolaire , , Nourrisson , Phénotype , Adolescent , Mutation , Dépistage génétique/méthodes
7.
Orphanet J Rare Dis ; 19(1): 248, 2024 Jul 03.
Article de Anglais | MEDLINE | ID: mdl-38961493

RÉSUMÉ

BACKGROUND: Primary carnitine deficiency (PCD) is a rare autosomal recessive fatty acid oxidation disorder caused by variants in SLC22A5, with its prevalence and SLC22A5 gene mutation spectrum varying across races and regions. This study aimed to systematically analyze the incidence of PCD in China and delineate regional differences in the prevalence of PCD and SLC22A5 gene variants. METHODS: PubMed, Embase, Web of Science, and Chinese databases were searched up to November 2023. Following quality assessment and data extraction, a meta-analysis was performed on screening results for PCD among Chinese newborns. RESULTS: After reviewing 1,889 articles, 22 studies involving 9,958,380 newborns and 476 PCD cases were included. Of the 476 patients with PCD, 469 underwent genetic diagnosis, revealing 890 variants of 934 alleles of SLC22A5, among which 107 different variants were detected. The meta-analysis showed that the prevalence of PCD in China was 0.05‰ [95%CI, (0.04‰, 0.06‰)] or 1/20 000 [95%CI, (1/16 667, 1/25 000)]. Subgroup analyses revealed a higher incidence in southern China [0.07‰, 95%CI, (0.05‰, 0.08‰)] than in northern China [0.02‰, 95%CI, (0.02‰, 0.03‰)] (P < 0.001). Furthermore, the result of the meta-analysis showed that the frequency of the variant with c.1400C > G, c.51C > G, c.760C > T, c.338G > A, and c.428C > T were 45% [95%CI, (34%, 59%)], 26% [95%CI, (22%, 31%)], 14% [95%CI, (10%, 20%)], 6% [95%CI, (4%, 8%)], and 5% [95%CI, (4%, 8%)], respectively. Among the subgroup analyses, the variant frequency of c.1400C > G in southern China [39%, 95%CI, (29%, 53%)] was significantly lower than that in northern China [79‰, 95%CI, (47‰, 135‰)] (P < 0.05). CONCLUSIONS: This study systematically analyzed PCD prevalence and identified common SLC22A5 gene variants in the Chinese population. The findings provide valuable epidemiological insights and guidance for future PCD screening effects in newborns.


Sujet(s)
Carnitine , Hyperammoniémie , Membre-5 de la famille-22 de transporteurs de solutés , Humains , Chine/épidémiologie , Carnitine/déficit , Nouveau-né , Membre-5 de la famille-22 de transporteurs de solutés/génétique , Hyperammoniémie/génétique , Hyperammoniémie/épidémiologie , Hyperammoniémie/diagnostic , Cardiomyopathies/génétique , Cardiomyopathies/épidémiologie , Maladies musculaires/génétique , Maladies musculaires/épidémiologie , Mutation/génétique , Dépistage néonatal/méthodes , Peuples d'Asie de l'Est
8.
J Pregnancy ; 2024: 7713590, 2024.
Article de Anglais | MEDLINE | ID: mdl-38957710

RÉSUMÉ

Preeclampsia and peripartum cardiomyopathy (PPCM) are significant obstetric problems that can arise during or after pregnancy. Both are known to be causes of maternal mortality and morbidity. Several recent studies have suggested a link between preeclampsia and the pathophysiology of PPCM. However, the common thread that connects the two has yet to be thoroughly and fully articulated. Here, we investigate the complex dynamics of preeclampsia and PPCM in this review. Our analysis focuses mainly on inflammatory and immunological responses, endothelial dysfunction as a shared pathway, and potential genetic predisposition to both diseases. To begin, we will look at how excessive inflammatory and immunological responses can lead to clinical symptoms of both illnesses, emphasizing the role of proinflammatory cytokines and immune cells in modifying vascular and tissue responses. Second, we consider endothelial dysfunction to be a crucial point at which endothelial damage and activation contribute to pathogenesis through increased vascular permeability, vascular dysfunction, and thrombus formation. Finally, we examine recent information suggesting genetic predispositions to preeclampsia and PPCM, such as genetic variants in genes involved in the management of blood pressure, the inflammatory response, and heart structural integrity. With this synergistic study, we seek to encourage more research and creative therapy solutions by emphasizing the need for an interdisciplinary approach to understanding and managing the connection between preeclampsia and PPCM.


Sujet(s)
Cardiomyopathies , Période de péripartum , Pré-éclampsie , Humains , Femelle , Pré-éclampsie/physiopathologie , Pré-éclampsie/génétique , Grossesse , Cardiomyopathies/étiologie , Cardiomyopathies/génétique , Cardiomyopathies/physiopathologie , Prédisposition génétique à une maladie , Endothélium vasculaire/physiopathologie , Complications cardiovasculaires de la grossesse/physiopathologie , Complications cardiovasculaires de la grossesse/génétique
9.
Int J Med Sci ; 21(9): 1629-1639, 2024.
Article de Anglais | MEDLINE | ID: mdl-39006843

RÉSUMÉ

The complete molecular mechanism underlying doxorubicin-induced cardiomyopathy remains incompletely elucidated. In this investigation, we engineered mice with cardiomyocyte-specific sorting nexin 3 knockout (SNX3Cko ) to probe the potential protective effects of SNX3 ablation on doxorubicin-triggered myocardial injury, focusing on GPX4-dependent ferroptosis. Our findings indicate that SNX3 deletion normalized heart contractile/relaxation function and thwarted the escalation of cardiac injury biomarkers following doxorubicin exposure. Additionally, SNX3 deletion in the heart mitigated the inflammatory response and oxidative stress in the presence of doxorubicin. At the molecular level, the detrimental effects of doxorubicin-induced cell death, endoplasmic reticulum (ER) stress, and mitochondrial dysfunction were alleviated by SNX3 deficiency. Molecular analysis revealed the activation of GPX4-mediated ferroptosis by doxorubicin, whereas loss of SNX3 prevented the initiation of GPX4-dependent ferroptosis. Furthermore, treatment with erastin, a ferroptosis inducer, markedly reduced cell viability, exacerbated ER stress, and induced mitochondrial dysfunction in SNX3-depleted cardiomyocytes upon doxorubicin exposure. In summary, our results demonstrate that SNX3 deficiency shielded the heart from doxorubicin-induced myocardial dysfunction by modulating GPX4-associated ferroptosis.


Sujet(s)
Cardiomyopathies , Doxorubicine , Ferroptose , Souris knockout , Myocytes cardiaques , Phospholipid hydroperoxide glutathione peroxidase , Nexines de tri , Ferroptose/effets des médicaments et des substances chimiques , Ferroptose/génétique , Animaux , Doxorubicine/effets indésirables , Doxorubicine/toxicité , Cardiomyopathies/induit chimiquement , Cardiomyopathies/anatomopathologie , Cardiomyopathies/génétique , Nexines de tri/génétique , Nexines de tri/métabolisme , Souris , Phospholipid hydroperoxide glutathione peroxidase/métabolisme , Phospholipid hydroperoxide glutathione peroxidase/génétique , Myocytes cardiaques/effets des médicaments et des substances chimiques , Myocytes cardiaques/métabolisme , Myocytes cardiaques/anatomopathologie , Humains , Stress oxydatif/effets des médicaments et des substances chimiques , Stress du réticulum endoplasmique/effets des médicaments et des substances chimiques
10.
Int J Mol Sci ; 25(14)2024 Jul 10.
Article de Anglais | MEDLINE | ID: mdl-39062799

RÉSUMÉ

A genetic diagnosis of primary cardiomyopathies can be a long-unmet need in patients with complex phenotypes. We investigated a three-generation family with cardiomyopathy and various extracardiac abnormalities that had long sought a precise diagnosis. The 41-year-old proband had hypertrophic cardiomyopathy (HCM), left ventricular noncompaction, myocardial fibrosis, arrhythmias, and a short stature. His sister showed HCM, myocardial hypertrabeculation and fibrosis, sensorineural deafness, and congenital genitourinary malformations. Their father had left ventricular hypertrophy (LVH). The proband's eldest daughter demonstrated developmental delay and seizures. We performed a clinical examination and whole-exome sequencing for all available family members. All patients with HCM/LVH shared a c.4411-2A>C variant in ALPK3, a recently known HCM-causative gene. Functional studies confirmed that this variant alters ALPK3 canonical splicing. Due to extracardiac symptoms in the female patients, we continued the search and found two additional single-gene disorders. The proband's sister had a p.Trp329Gly missense in GATA3, linked to hypoparathyroidism, sensorineural deafness, and renal dysplasia; his daughter had a p.Ser251del in WDR45, associated with beta-propeller protein-associated neurodegeneration. This unique case of three monogenic disorders in one family shows how a comprehensive approach with thorough phenotyping and extensive genetic testing of all symptomatic individuals provides precise diagnoses and appropriate follow-up, embodying the concept of personalized medicine. We also present the first example of a splicing functional study for ALPK3 and describe the genotype-phenotype correlations in cardiomyopathy.


Sujet(s)
Pedigree , Humains , Femelle , Mâle , Adulte , Cardiomyopathies/génétique , Cardiomyopathies/diagnostic , , Malformations multiples/génétique , Surdité neurosensorielle/génétique , Phénotype , Cardiomyopathie hypertrophique/génétique , Cardiomyopathie hypertrophique/diagnostic
11.
Int J Mol Sci ; 25(14)2024 Jul 16.
Article de Anglais | MEDLINE | ID: mdl-39063023

RÉSUMÉ

Mitochondrial fission and fusion are vital dynamic processes for mitochondrial quality control and for the maintenance of cellular respiration; they also play an important role in the formation and maintenance of cells with high energy demand including cardiomyocytes and neurons. The DNM1L (dynamin-1 like) gene encodes for the DRP1 protein, an evolutionary conserved member of the dynamin family that is responsible for the fission of mitochondria; it is ubiquitous but highly expressed in the developing neonatal heart. De novo heterozygous pathogenic variants in the DNM1L gene have been previously reported to be associated with neonatal or infantile-onset encephalopathy characterized by hypotonia, developmental delay and refractory epilepsy. However, cardiac involvement has been previously reported only in one case. Next-Generation Sequencing (NGS) was used to genetically assess a baby girl characterized by developmental delay with spastic-dystonic, tetraparesis and hypertrophic cardiomyopathy of the left ventricle. Histochemical analysis and spectrophotometric determination of electron transport chain were performed to characterize the muscle biopsy; moreover, the morphology of mitochondria and peroxisomes was evaluated in cultured fibroblasts as well. Herein, we expand the phenotype of DNM1L-related disorder, describing the case of a girl with a heterozygous mutation in DNM1L and affected by progressive infantile encephalopathy, with cardiomyopathy and fatal paroxysmal vomiting correlated with bulbar transitory abnormal T2 hyperintensities and diffusion-weighted imaging (DWI) restriction areas, but without epilepsy. In patients with DNM1L mutations, careful evaluation for cardiac involvement is recommended.


Sujet(s)
Cardiomyopathies , Dynamines , Mutation , Humains , Femelle , Dynamines/génétique , Cardiomyopathies/génétique , Mutation/génétique , Nourrisson , Issue fatale , Encéphalopathies/génétique , Encéphalopathies/anatomopathologie , dGTPases/génétique
12.
Int J Mol Sci ; 25(14)2024 Jul 17.
Article de Anglais | MEDLINE | ID: mdl-39063061

RÉSUMÉ

(1) Heart transplantation (HTX) improves the overall survival and functional status of end-stage heart failure patients with cardiomyopathies (CMPs). The majority of CMPs have genetic causes, and the overlap between CMPs and inherited myopathies is well documented. However, the long-term outcome in skeletal muscle function and possibility of an undiagnosed underlying genetic cause of both a cardiac and skeletal pathology remain unknown. (2) Thirty-nine patients were assessed using open and standardized interviews on muscle function, a quality-of-life (EuroQol EQ-5D-3L) questionnaire, and a physical examination (Medical Research Council Muscle scale). Whole-exome sequencing was completed in three stages for those with skeletal muscle weakness. (3) Seven patients (17.9%) reported new-onset muscle weakness and motor limitations. Objective muscle weakness in the upper and lower extremities was seen in four patients. In three of them, exome sequencing revealed pathogenic/likely pathogenic variants in the genes encoding nexilin, myosin heavy chain, titin, and SPG7. (4) Our findings support a positive long-term outcome of skeletal muscle function in HTX patients. However, 10% of patients showed clinical signs of myopathy due to a possible genetic cause. The integration of genetic testing and standardized neurological assessment of motor function during the peri-HTX period should be considered.


Sujet(s)
Transplantation cardiaque , Maladies neuromusculaires , Humains , Transplantation cardiaque/effets indésirables , Mâle , Femelle , Adulte d'âge moyen , Maladies neuromusculaires/génétique , Adulte , Qualité de vie , , Muscles squelettiques/anatomopathologie , Muscles squelettiques/physiopathologie , Sujet âgé , Défaillance cardiaque/génétique , Défaillance cardiaque/chirurgie , Défaillance cardiaque/étiologie , Cardiomyopathies/génétique , Cardiomyopathies/étiologie , Faiblesse musculaire/étiologie , Faiblesse musculaire/génétique , Connectine/génétique
13.
Biochem Biophys Res Commun ; 727: 150336, 2024 Oct 01.
Article de Anglais | MEDLINE | ID: mdl-38959731

RÉSUMÉ

Myocardial fibrosis involves the loss of cardiomyocytes, myocardial fibroblast proliferation, and a reduction in angiogenesis, ultimately leading to heart failure, Given its significant implications, it is crucial to explore novel therapies for myocardial fibrosis. Recently one emerging avenue has been the use of small extracellular vesicles (sEV)-carried miRNA. In this review, we summarize the regulatory role of sEV-carried miRNA in myocardial fibrosis. We explored not only the potential diagnostic value of circulating miRNA as biomarkers for heart disease but also the therapeutic implications of sEV-carried miRNA derived from various cellular sources and applications of modified sEV. This exploration is paramount for researchers striving to develop innovative, cell-free therapies as potential drug candidates for the management of myocardial fibrosis.


Sujet(s)
Vésicules extracellulaires , Fibrose , microARN , Humains , Vésicules extracellulaires/métabolisme , Vésicules extracellulaires/génétique , microARN/génétique , microARN/métabolisme , Fibrose/génétique , Animaux , Myocarde/anatomopathologie , Myocarde/métabolisme , Cardiomyopathies/génétique , Cardiomyopathies/anatomopathologie , Cardiomyopathies/métabolisme , Marqueurs biologiques/métabolisme , Myocytes cardiaques/métabolisme , Myocytes cardiaques/anatomopathologie
14.
Int Immunopharmacol ; 138: 112574, 2024 Sep 10.
Article de Anglais | MEDLINE | ID: mdl-38971104

RÉSUMÉ

BACKGROUND: Ischemic cardiomyopathy (IC) is primarily due to long-term ischemia/hypoxia of the coronary arteries, leading to impaired cardiac contractile or diastolic function. A new form of cell death induced by copper, called "cuproptosis" is related to the development and progression of multiple diseases. The cuproptosis-related gene (CuGs) plays an important role in acute myocardial infarction, while the specific mechanisms of CuGs in ischemic cardiomyopathy remain unclear. METHODS: The expressions of CuGs and their immune characteristics were analyzed with the IC datasets obtained from the Gene Expression Omnibus, namely GSE5406 and GSE57338, identifying core genes associated with IC development. By comparing RF, SVM, GLM and XGB models, the optimal machine learning model was selected. The expression of marker genes was validated based on the GSE57345, GSE48166 and GSE42955 datasets. Construct a CeRNA network based on core genes. Therapeutic chemiacals targeting core genes were acquired using the CTD database, and molecular docking was performed using Autodock vina software. By ligating the left anterior descending (LAD) coronary artery, an IC mouse model is established, and core genes were experimentally validated using Western blot (WB) and immunohistochemistry (IHC) methods. RESULTS: We identified 14 CuGs closely associated with the onset of IC. The SVM model exhibited superior discriminative power (AUC = 0.914), with core genes being DLST, ATP7B, FDX1, SLC31A1 and DLAT. Core genes were validated on the GSE42955, GSE48166 and GSE57345 datasets, showing excellent performance (AUC = 0.943, AUC = 0.800, and AUC = 0.932). The CeRNA network consists of 218 nodes and 264 lines, including 5 core diagnostic genes, 52 miRNAs, and 161 lncRNAs. Chemicals predictions indicated 8 chemicals have therapeutic effects on the core diagnostic genes, with benzo(a)pyrene molecular docking showing the highest affinity (-11.3 kcal/mol). Compared to the normal group, the IC group,which was established by LAD ligation, showed a significant decrease in LVEF as indicated by cardiac ultrasound, and increased fibrosis as shown by MASSON staining, WB results suggest increased expression of DLST and ATP7B, and decreased expression of FDX1, SLC31A1 and DLAT in the myocardial ischemic area (p < 0.05), which was also confirmed by IHC in tissue sections. CONCLUSION: In summary, this study comprehensively revealed that DLST, ATP7B, FDX1, SLC31A1 and DLAT could be identified as potential immunological biomarkers in IC, and validated through an IC mouse model, providing valuable insights for future research into the mechanisms of CuGs and its diagnostic value to IC.


Sujet(s)
Apoptose , Biologie informatique , Ischémie myocardique , Animaux , Humains , Mâle , Souris , Cardiomyopathies/génétique , Cardiomyopathies/immunologie , Bases de données génétiques , Modèles animaux de maladie humaine , Réseaux de régulation génique , Souris de lignée C57BL , Simulation de docking moléculaire , Ischémie myocardique/génétique , Ischémie myocardique/immunologie , Cuivre
15.
Heart Fail Rev ; 29(5): 1025-1037, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38985384

RÉSUMÉ

Cardiac disorders exhibit considerable heterogeneity, and understanding their genetic foundations is crucial for their diagnosis and treatment. Recent genetic analyses involving a growing number of participants have uncovered novel mutations within both coding and non-coding regions of DNA, contributing to the onset of cardiac conditions. The NEXN gene, encoding the Nexilin protein, an actin filament-binding protein, is integral to normal cardiac function. Mutations in this gene have been linked to cardiomyopathies, cardiovascular disorders, and sudden deaths. Heterozygous or homozygous variants of the NEXN gene are associated with the development of endocardial fibroelastosis (EFE), a rare cardiac condition characterized by excessive collagen and elastin deposition in the left ventricular endocardium predominantly affecting infants and young children. EFE occurs both primary and secondary to other conditions and often leads to unfavorable prognoses and outcomes. This review explores the role of NEXN genetic variants in cardiovascular disorders, particularly EFE, revealing that functional mutations are not clustered in a specific domain of Nexilin based on the cardiac disorder phenotype. Our review underscores the importance of understanding genetic mutations for the diagnosis and treatment of cardiac conditions.


Sujet(s)
Cardiomyopathies , Fibroélastose endocardique , Mutation , Humains , Cardiomyopathies/génétique , Cardiomyopathies/diagnostic , Cardiomyopathies/physiopathologie , Cardiomyopathies/métabolisme , Fibroélastose endocardique/génétique , Fibroélastose endocardique/métabolisme , Fibroélastose endocardique/diagnostic , Protéines des microfilaments/génétique , Protéines des microfilaments/métabolisme , Phénotype
16.
Commun Biol ; 7(1): 905, 2024 Jul 27.
Article de Anglais | MEDLINE | ID: mdl-39068302

RÉSUMÉ

ATTR amyloidosis results from the conversion of transthyretin into amyloid fibrils that deposit in tissues causing organ failure and death. This conversion is facilitated by mutations in ATTRv amyloidosis, or aging in ATTRwt amyloidosis. ATTRv amyloidosis exhibits extreme phenotypic variability, whereas ATTRwt amyloidosis presentation is consistent and predictable. Previously, we found unique structural variabilities in cardiac amyloid fibrils from polyneuropathic ATTRv-I84S patients. In contrast, cardiac fibrils from five genotypically different patients with cardiomyopathy or mixed phenotypes are structurally homogeneous. To understand fibril structure's impact on phenotype, it is necessary to study the fibrils from multiple patients sharing genotype and phenotype. Here we show the cryo-electron microscopy structures of fibrils extracted from four cardiomyopathic ATTRwt amyloidosis patients. Our study confirms that they share identical conformations with minimal structural variability, consistent with their homogenous clinical presentation. Our study contributes to the understanding of ATTR amyloidosis biopathology and calls for further studies.


Sujet(s)
Amyloïde , Cryomicroscopie électronique , Myocarde , Humains , Amyloïde/métabolisme , Amyloïde/composition chimique , Amyloïde/ultrastructure , Myocarde/anatomopathologie , Myocarde/ultrastructure , Préalbumine/génétique , Préalbumine/métabolisme , Préalbumine/composition chimique , Neuropathies amyloïdes familiales/génétique , Neuropathies amyloïdes familiales/anatomopathologie , Mâle , Femelle , Adulte d'âge moyen , Sujet âgé , Amyloïdose/métabolisme , Amyloïdose/anatomopathologie , Amyloïdose/génétique , Mutation , Cardiomyopathies/génétique , Cardiomyopathies/anatomopathologie , Cardiomyopathies/métabolisme
17.
Adv Exp Med Biol ; 1441: 977-990, 2024.
Article de Anglais | MEDLINE | ID: mdl-38884765

RÉSUMÉ

The identification of a disease-causing variant in a patient diagnosed with cardiomyopathy allows for presymptomatic testing in at risk relatives. Carriers of a pathogenic variant can subsequently be screened at intervals by a cardiologist to assess the risk for potentially life-threatening arrhythmias which can be life-saving. In addition, gene-specific recommendations for risk stratification and disease specific pharmacological options for therapy are beginning to emerge. The large variability in disease penetrance, symptoms, and prognosis, and in some families even in cardiomyopathy subtype, makes genetic counseling both of great importance and complicated.


Sujet(s)
Cardiomyopathies , Humains , Cardiomyopathies/génétique , Conseil génétique , Prédisposition génétique à une maladie/génétique , Dépistage génétique/méthodes , Mutation
18.
Adv Exp Med Biol ; 1441: 991-1019, 2024.
Article de Anglais | MEDLINE | ID: mdl-38884766

RÉSUMÉ

Cardiomyopathies are a heterogeneous group of disorders of the heart muscle that ultimately result in congestive heart failure. Rapid progress in genetics, molecular and cellular biology with breakthrough innovative genetic-engineering techniques, such as next-generation sequencing and multiomics platforms, stem cell reprogramming, as well as novel groundbreaking gene-editing systems over the past 25 years has greatly improved the understanding of pathogenic signaling pathways in inherited cardiomyopathies. This chapter will focus on intracellular and intercellular molecular signaling pathways that are activated by a genetic insult in cardiomyocytes to maintain tissue and organ level regulation and resultant cardiac remodeling in certain forms of cardiomyopathies. In addition, animal models of different clinical forms of human cardiomyopathies with their summaries of triggered key molecules and signaling pathways will be described.


Sujet(s)
Cardiomyopathies , Modèles animaux de maladie humaine , Myocytes cardiaques , Transduction du signal , Animaux , Humains , Cardiomyopathies/génétique , Cardiomyopathies/anatomopathologie , Cardiomyopathies/métabolisme , Myocytes cardiaques/métabolisme , Myocytes cardiaques/anatomopathologie , Transduction du signal/génétique
19.
Clin Epigenetics ; 16(1): 76, 2024 Jun 06.
Article de Anglais | MEDLINE | ID: mdl-38845031

RÉSUMÉ

Tatton-Brown-Rahman syndrome (TBRS) is a rare congenital genetic disorder caused by autosomal dominant pathogenic variants in the DNA methyltransferase DNMT3A gene. Typical TBRS clinical features are overgrowth, intellectual disability, and minor facial anomalies. However, since the syndrome was first described in 2014, a widening spectrum of abnormalities is being described. Cardiovascular abnormalities are less commonly reported but can be a major complication of the syndrome. This article describes a family of three individuals diagnosed with TBRS in adulthood and highlights the variable expression of cardiovascular features. A 34-year-old proband presented with progressive aortic dilatation, mitral valve (MV) regurgitation, left ventricular (LV) dilatation, and ventricular arrhythmias. The affected family members (mother and brother) were diagnosed with MV regurgitation, LV dilatation, and arrhythmias. Exome sequencing and computational protein analysis suggested that the novel familial DNMT3A mutation Ser775Tyr is located in the methyltransferase domain, however, distant from the active site or DNA-binding loops. Nevertheless, this bulky substitution may have a significant effect on DNMT3A protein structure, dynamics, and function. Analysis of peripheral blood cfDNA and transcriptome showed shortened mononucleosome fragments and altered gene expression in a number of genes related to cardiovascular health and of yet undescribed function, including several lncRNAs. This highlights the importance of epigenetic regulation by DNMT3A on cardiovascular system development and function. From the clinical perspective, we suggest that new patients diagnosed with congenital DNMT3A variants and TBRS require close examination and follow-up for aortic dilatation and valvular disease because these conditions can progress rapidly. Moreover, personalized treatments, based on the specific DNMT3A variants and the different pathways of their function loss, can be envisioned in the future.


Sujet(s)
DNA (cytosine-5-)-methyltransferase , DNA methyltransferase 3A , Pedigree , Humains , DNA methyltransferase 3A/génétique , Adulte , Mâle , DNA (cytosine-5-)-methyltransferase/génétique , Femelle , Cardiomyopathies/génétique , Maladies de l'aorte/génétique , /méthodes , Déficience intellectuelle/génétique , Mutation
20.
BMJ Paediatr Open ; 8(1)2024 May 31.
Article de Anglais | MEDLINE | ID: mdl-38823802

RÉSUMÉ

OBJECTIVES: This study aimed to describe the genetic and clinical characteristics of paediatric cardiomyopathy in a cohort of Chinese patients. METHODS: We retrospectively reviewed the clinical history and mutation spectrum of 75 unrelated Chinese paediatric patients who were diagnosed with cardiomyopathy and referred to our hospital between January 2016 and December 2022. RESULTS: Seventy-five children with cardiomyopathy were enrolled, including 32 (42.7%) boys and 43 (57.3%) girls. Dilated cardiomyopathy was the most prevalent cardiomyopathy (61.3%) in the patients, followed by hypertrophic cardiomyopathy (17.3%), ventricular non-compaction (14.7%), restrictive cardiomyopathy (5.3%) and arrhythmogenic right ventricular cardiomyopathy (1.3%). Whole-exome sequencing and targeted next-generation sequencing identified 34 pathogenic/likely pathogenic variants and 1 copy number variant in 14 genes related to cardiomyopathy in 30 children, accounting for 40% of all patients. TNNC1 p.Asp65Asn and MYH7 p.Glu500Lys have not been reported previously. The follow-up time ranged from 2 months to 6 years. Twenty-two children died (mortality rate 29%). CONCLUSIONS: Comprehensive genetic testing was associated with a 40% yield of causal genetic mutations in Chinese cardiomyopathy cases. We found diversity in the mutation profile in different patients, which suggests that the mutational background of cardiomyopathy in China is heterogeneous, and the findings may be helpful to those counselling patients and families.


Sujet(s)
Cardiomyopathies , Dépistage génétique , Mutation , Humains , Mâle , Femelle , Études rétrospectives , Enfant , Nourrisson , Cardiomyopathies/génétique , Enfant d'âge préscolaire , Chine/épidémiologie , , Adolescent
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE