Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 3.014
Filtrer
2.
Open Biol ; 14(6): 230427, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38862020

RÉSUMÉ

Hypertrophic cardiomyopathy (HCM) is a monogenic cardiac disorder commonly induced by sarcomere gene mutations. However, the mechanism for HCM is not well defined. Here, we generated transgenic MYH7 R453C and MYH6 R453C piglets and found both developed typical cardiac hypertrophy. Unexpectedly, we found serious fibrosis and cardiomyocyte loss in the ventricular of MYH7 R453C, not MYH6 R453C piglets, similar to HCM patients. Then, RNA-seq analysis and western blotting identified the activation of ERK1/2 and PI3K-Akt pathways in MYH7 R453C. Moreover, we observed an increased expression of fetal genes and an excess of reactive oxygen species (ROS) in MYH7 R453C piglet models, which was produced by Nox4 and subsequently induced inflammatory response. Additionally, the phosphorylation levels of Smad2/3, ERK1/2 and NF-kB p65 proteins were elevated in cardiomyocytes with the MYH7 R453C mutation. Furthermore, epigallocatechin gallate, a natural bioactive compound, could be used as a drug to reduce cell death by adjusting significant downregulation of the protein expression of Bax and upregulated Bcl-2 levels in the H9C2 models with MYH7 R453C mutation. In conclusion, our study illustrated that TGF-ß/Smad2/3, ERK1/2 and Nox4/ROS pathways have synergistic effects on cardiac remodelling and inflammation in MYH7 R453C mutation.


Sujet(s)
Chaînes lourdes de myosine , NADPH Oxidase 4 , Facteur de transcription NF-kappa B , Espèces réactives de l'oxygène , Transduction du signal , Facteur de croissance transformant bêta , Animaux , Chaînes lourdes de myosine/métabolisme , Chaînes lourdes de myosine/génétique , Facteur de croissance transformant bêta/métabolisme , NADPH Oxidase 4/métabolisme , NADPH Oxidase 4/génétique , Espèces réactives de l'oxygène/métabolisme , Facteur de transcription NF-kappa B/métabolisme , Suidae , Myocytes cardiaques/métabolisme , Humains , Myosines cardiaques/métabolisme , Myosines cardiaques/génétique , Modèles animaux de maladie humaine , Système de signalisation des MAP kinases , Animal génétiquement modifié , Protéine Smad2/métabolisme , Protéine Smad2/génétique , Mutation , Protéine Smad-3/métabolisme , Protéine Smad-3/génétique , Remodelage ventriculaire , Cardiomyopathie hypertrophique/métabolisme , Cardiomyopathie hypertrophique/génétique , Cardiomyopathie hypertrophique/anatomopathologie , Rats
3.
Circ Genom Precis Med ; 17(3): e004369, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38853772

RÉSUMÉ

BACKGROUND: Hypertrophic cardiomyopathy (HCM) is caused by sarcomere gene mutations (genotype-positive HCM) in ≈50% of patients and occurs in the absence of mutations (genotype-negative HCM) in the other half of patients. We explored how alterations in the metabolomic and lipidomic landscape are involved in cardiac remodeling in both patient groups. METHODS: We performed proteomics, metabolomics, and lipidomics on myectomy samples (genotype-positive N=19; genotype-negative N=22; and genotype unknown N=6) from clinically well-phenotyped patients with HCM and on cardiac tissue samples from sex- and age-matched and body mass index-matched nonfailing donors (N=20). These data sets were integrated to comprehensively map changes in lipid-handling and energy metabolism pathways. By linking metabolomic and lipidomic data to variability in clinical data, we explored patient group-specific associations between cardiac and metabolic remodeling. RESULTS: HCM myectomy samples exhibited (1) increased glucose and glycogen metabolism, (2) downregulation of fatty acid oxidation, and (3) reduced ceramide formation and lipid storage. In genotype-negative patients, septal hypertrophy and diastolic dysfunction correlated with lowering of acylcarnitines, redox metabolites, amino acids, pentose phosphate pathway intermediates, purines, and pyrimidines. In contrast, redox metabolites, amino acids, pentose phosphate pathway intermediates, purines, and pyrimidines were positively associated with septal hypertrophy and diastolic impairment in genotype-positive patients. CONCLUSIONS: We provide novel insights into both general and genotype-specific metabolic changes in HCM. Distinct metabolic alterations underlie cardiac disease progression in genotype-negative and genotype-positive patients with HCM.


Sujet(s)
Cardiomyopathie hypertrophique , Génotype , Phénotype , Humains , Cardiomyopathie hypertrophique/génétique , Cardiomyopathie hypertrophique/métabolisme , Cardiomyopathie hypertrophique/anatomopathologie , Mâle , Femelle , Adulte d'âge moyen , Adulte , Myocarde/métabolisme , Myocarde/anatomopathologie , Métabolomique , Protéomique , Lipidomique , Métabolisme lipidique/génétique , Sarcomères/métabolisme , Sarcomères/génétique , Métabolisme énergétique/génétique , Sujet âgé , Multi-omique
4.
Int J Mol Sci ; 25(11)2024 May 28.
Article de Anglais | MEDLINE | ID: mdl-38892064

RÉSUMÉ

Hypertrophic cardiomyopathy (HCM) is a heart condition characterized by cellular and metabolic dysfunction, with mitochondrial dysfunction playing a crucial role. Although the direct relationship between genetic mutations and mitochondrial dysfunction remains unclear, targeting mitochondrial dysfunction presents promising opportunities for treatment, as there are currently no effective treatments available for HCM. This review adhered to the Preferred Reporting Items for Systematic Reviews and Meta-Analysis Extension for Scoping Reviews guidelines. Searches were conducted in databases such as PubMed, Embase, and Scopus up to September 2023 using "MESH terms". Bibliographic references from pertinent articles were also included. Hypertrophic cardiomyopathy (HCM) is influenced by ionic homeostasis, cardiac tissue remodeling, metabolic balance, genetic mutations, reactive oxygen species regulation, and mitochondrial dysfunction. The latter is a common factor regardless of the cause and is linked to intracellular calcium handling, energetic and oxidative stress, and HCM-induced hypertrophy. Hypertrophic cardiomyopathy treatments focus on symptom management and complication prevention. Targeted therapeutic approaches, such as improving mitochondrial bioenergetics, are being explored. This includes coenzyme Q and elamipretide therapies and metabolic strategies like therapeutic ketosis. Understanding the biomolecular, genetic, and mitochondrial mechanisms underlying HCM is crucial for developing new therapeutic modalities.


Sujet(s)
Cardiomyopathie hypertrophique , Mutation , Oxydoréduction , Transduction du signal , Humains , Cardiomyopathie hypertrophique/génétique , Cardiomyopathie hypertrophique/métabolisme , Animaux , Mitochondries/métabolisme , Mitochondries/génétique , Stress oxydatif , Espèces réactives de l'oxygène/métabolisme
6.
Int J Cardiol ; 409: 132213, 2024 Aug 15.
Article de Anglais | MEDLINE | ID: mdl-38801835

RÉSUMÉ

BACKGROUND: Hypertrophic cardiomyopathy (HCM) is an inherited heart disease that can lead to sudden cardiac death. Impact of genetic testing for the prognosis and treatment of patients with HCM needs to be improved. We conducted a systematic review and meta-analysis to investigate the characteristics and outcomes associated with sarcomere genotypes in index patients with HCM. METHODS: A systematic search was conducted in Medline, Embase, and Cochrane Library up to Dec 31, 2023. Data on clinical characteristics, morphological and imaging features, outcomes and interventions were collected from published studies and pooled using a random-effects meta-analysis. RESULTS: A total of 30 studies with 10,825 HCM index patients were included in the pooled analyses. The frequency of sarcomere genes in HCM patients was 41%. Sarcomere mutations were more frequent in women (p < 0.00001), and were associated with lower body mass index (26.1 ± 4.7 versus 27.5 ± 4.3; p = 0.003) and left ventricular ejection fraction (65.7% ± 10.1% vs. 67.1% ± 8.6%; p = 0.03), less apical hypertrophy (6.5% vs. 20.1%; p < 0.0001) and left ventricular outflow tract obstruction (29.1% vs. 33.2%; p = 0.03), greater left atrial volume index (43.6 ± 21.1 ml/m2 vs. 37.3 ± 13.0 ml/m2; p = 0.02). Higher risks of ventricular tachycardia (23.4% vs. 14.1%; p < 0.0001), syncope (18.3% vs. 10.9%; p = 0.01) and heart failure (17.3% vs. 14.6%; p = 0.002) were also associated with sarcomere mutations. CONCLUSIONS: Sarcomere mutations are more frequent in women, and are associated with worse clinical characteristics and poor outcomes.


Sujet(s)
Cardiomyopathie hypertrophique , Mutation , Sarcomères , Humains , Sarcomères/génétique , Cardiomyopathie hypertrophique/génétique , Cardiomyopathie hypertrophique/diagnostic
7.
Hum Cell ; 37(4): 1205-1214, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38762696

RÉSUMÉ

Hypertrophic cardiomyopathy (HCM) is frequently caused by mutations in the MYPBC3 gene, which encodes the cardiac myosin-binding protein C (cMyBP-C). Most pathogenic variants in MYPBC3 are either nonsense mutations or result in frameshifts, suggesting that the primary disease mechanism involves reduced functional cMyBP-C protein levels within sarcomeres. However, a subset of MYPBC3 variants are missense mutations, and the molecular mechanisms underlying their pathogenicity remain elusive. Upon in vitro differentiation into cardiomyocytes, induced pluripotent stem cells (iPSCs) derived from HCM patients represent a valuable resource for disease modeling. In this study, we generated two iPSC lines from peripheral blood mononuclear cells (PBMCs) of a female with early onset and severe HCM linked to the MYBPC3: c.772G > A variant. Although this variant was initially classified as a missense mutation, recent studies indicate that it interferes with splicing and results in a frameshift. The generated iPSC lines exhibit a normal karyotype and display hallmark characteristics of pluripotency, including the ability to undergo trilineage differentiation. These novel iPSCs expand the existing repertoire of MYPBC3-mutated cell lines, broadening the spectrum of resources for exploring how diverse mutations induce HCM. They additionally offer a platform to study potential secondary genetic elements contributing to the pronounced disease severity observed in this individual.


Sujet(s)
Cardiomyopathie hypertrophique , Protéines de transport , Différenciation cellulaire , Cellules souches pluripotentes induites , Humains , Cellules souches pluripotentes induites/métabolisme , Cardiomyopathie hypertrophique/génétique , Cardiomyopathie hypertrophique/étiologie , Femelle , Protéines de transport/génétique , Protéines de transport/métabolisme , Différenciation cellulaire/génétique , Myocytes cardiaques/métabolisme , Myocytes cardiaques/anatomopathologie , Mutation faux-sens/génétique , Indice de gravité de la maladie , Mutation/génétique , Lignée cellulaire , Mutation avec décalage du cadre de lecture/génétique , Agranulocytes/métabolisme , Cellules cultivées
8.
Int J Cardiol ; 408: 132117, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-38710232

RÉSUMÉ

BACKGROUND: Hypertrophic cardiomyopathy (HCM) is believed to have low overall mortality rate, that could be influenced by gender, particularly among probands. We aimed to evaluate the survival rates and possible gender differences in a homogeneous cohort of HCM proband patients, referred for genetic testing, from the same geographical area, without differences in medical care access nor clinical referral pathways. METHODS: we compared the mortality rates of a cohort of consecutive HCM probands referred for genetic testing (2000-2022), from a Spanish region (xxx1) with a centralized genetic testing pathway, with its control reference population by Ederer II method. Gender differences were analyzed. RESULTS: Among the 649 HCM probands included in this study, there were significantly more men than women (61.3% vs 38.7, p < 0.05), with an earlier diagnosis (53.5 vs 61.1 years old, p < 0.05). Clinical evolution or arrhythmogenic HCM profile did no show no significant gender differences. Mean follow up was 9,8 years ±6,6 SD (9,9 ± 7 vs 9,6 ± 6,1, p = 0.59). No statistically significant differences in observed mortality, expected survival and excess mortality were found in the general HCM proband cohort. However, we found a significant excess mortality in female probands with HCM. No additional differences in analysis by genetic status were identified. CONCLUSION: Expected survival in our HCM probands did not differ from its reference population. However, despite no gender differences in phenotype severity were identified, proband HCM women did present a diagnosis delay and worse mortality outcomes.


Sujet(s)
Cardiomyopathie hypertrophique , Dépistage génétique , Humains , Mâle , Femelle , Adulte d'âge moyen , Cardiomyopathie hypertrophique/génétique , Cardiomyopathie hypertrophique/mortalité , Cardiomyopathie hypertrophique/diagnostic , Dépistage génétique/méthodes , Adulte , Sujet âgé , Analyse de survie , Études de cohortes , Études de suivi , Taux de survie/tendances , Orientation vers un spécialiste , Espagne/épidémiologie , Facteurs sexuels , Caractères sexuels
9.
Int J Cardiol ; 410: 132220, 2024 Sep 01.
Article de Anglais | MEDLINE | ID: mdl-38815672

RÉSUMÉ

BACKGROUND: Hypertrophic cardiomyopathy (HCM) is a widespread hereditary cardiac pathology characterized by thickened heart walls and rearrangement of cardiomyocytes. Despite extensive research, the mechanisms underlying HCM development remain poorly understood, impeding the development of effective therapeutic and diagnostic strategies. Recent studies have suggested a polygenic nature of HCM development alongside monogenic forms. Transcriptomic profiling is a valuable tool for investigating such diseases. In this study, we propose a novel approach to study regulatory microRNAs (miRNAs) in the context of HCM, utilizing state-of-the-art data analysis tools. METHODS AND RESULTS: Our method involves applying the Monte Carlo simulation and machine learning algorithm to transcriptomic data to generate high-capacity classifiers for HCM. From these classifiers, we extract key genes crucial for their performance, resulting in the identification of 16 key genes. Subsequently, we narrow down the pool of miRNAs by selecting those that may target the greatest number of key genes within the best models. We particularly focused on miR-124-3p, which we validated to have an association with HCM on an independent dataset. Subsequent investigation of its function revealed involvement of miR-124-3p in the RhoA signaling pathway. CONCLUSIONS: In this study we propose a new approach to analyze transcriptomic data to search for microRNAs associated with a disease. Using this approach for transcriptomic profiling data of patients with HCM, we identified miR-124-3p as a potential regulator of the RhoA signaling pathway in the pathogenesis of HCM.


Sujet(s)
Cardiomyopathie hypertrophique , Apprentissage machine , microARN , microARN/génétique , Cardiomyopathie hypertrophique/génétique , Cardiomyopathie hypertrophique/diagnostic , Humains , Marqueurs biologiques/métabolisme , Analyse de profil d'expression de gènes/méthodes , Mâle , Femelle
11.
J Am Heart Assoc ; 13(10): e033565, 2024 May 21.
Article de Anglais | MEDLINE | ID: mdl-38757491

RÉSUMÉ

BACKGROUND: The genetic basis of hypertrophic cardiomyopathy (HCM) is complex, and the relationship between genotype status and clinical outcome is incompletely resolved. METHODS AND RESULTS: We assessed a large international HCM cohort to define in contemporary terms natural history and clinical consequences of genotype. Consecutive patients (n=1468) with established HCM diagnosis underwent genetic testing. Patients with pathogenic (or likely pathogenic) variants were considered genotype positive (G+; n=312; 21%); those without definite disease-causing mutations (n=651; 44%) or variants of uncertain significance (n=505; 35%) were considered genotype negative (G-). Patients were followed up for a median of 7.8 years (interquartile range, 3.5-13.4 years); HCM end points were examined by cumulative event incidence. Over follow-up, 135 (9%) patients died, 33 from a variety of HCM-related causes. After adjusting for age, all-cause and HCM-related mortality did not differ between G- versus G+ patients (hazard ratio [HR], 0.78 [95% CI, 0.46-1.31]; P=0.37; HR, 0.93 [95% CI, 0.38-2.30]; P=0.87, respectively). Adverse event rates, including heart failure progression to class III/IV, heart transplant, or heart failure death, did not differ (G- versus G+) when adjusted for age (HR, 1.20 [95% CI, 0.63-2.26]; P=0.58), nor was genotype independently associated with sudden death event risk (HR, 1.39 [95% CI, 0.88-2.21]; P=0.16). In multivariable analysis, age was the only independent predictor of all-cause and HCM-related mortality, heart failure progression, and sudden death events. CONCLUSIONS: In this large consecutive cohort of patients with HCM, genotype (G+ or G-) was not a predictor of clinical course, including all-cause and HCM-related mortality and risk for heart failure progression or sudden death. G+ status should not be used to dictate clinical management or predict outcome in HCM.


Sujet(s)
Cardiomyopathie hypertrophique , Génotype , Humains , Cardiomyopathie hypertrophique/génétique , Cardiomyopathie hypertrophique/mortalité , Cardiomyopathie hypertrophique/diagnostic , Mâle , Femelle , Adulte d'âge moyen , Adulte , Mutation , Phénotype , Évolution de la maladie , Facteurs de risque , Prédisposition génétique à une maladie , Sujet âgé , Dépistage génétique/méthodes , Pronostic , Facteurs temps , Défaillance cardiaque/génétique , Défaillance cardiaque/mortalité , Mort subite cardiaque/étiologie , Mort subite cardiaque/épidémiologie , Transplantation cardiaque
12.
Arch Cardiovasc Dis ; 117(6-7): 427-432, 2024.
Article de Anglais | MEDLINE | ID: mdl-38762345

RÉSUMÉ

BACKGROUND: The efficacy of current pharmacological therapies in hypertrophic cardiomyopathy is limited. A cardiac myosin inhibitor, mavacamten, has recently been approved as a first-in-class treatment for symptomatic hypertrophic obstructive cardiomyopathy. AIMS: To assess the profile and burden of cardiac myosin inhibitor candidates in the hypertrophic cardiomyopathy prospective Register of hypertrophic cardiomyopathy (REMY) held by the French Society of Cardiology. METHODS: Data were collected at baseline and during follow-up from patients with hypertrophic cardiomyopathy enrolled in REMY by the three largest participating centres. RESULTS: Among 1059 adults with hypertrophic cardiomyopathy, 461 (43.5%) had obstruction; 325 (30.7%) of these were also symptomatic, forming the "cardiac myosin inhibitor candidates" group. Baseline features of this group were: age 58±15years; male sex (n=196; 60.3%); diagnosis-to-inclusion delay 5 (1-12)years; maximum wall thickness 20±6mm; left ventricular ejection fraction 69±6%; family history of hypertrophic cardiomyopathy or sudden cardiac death (n=133; 40.9%); presence of a pathogenic sarcomere gene mutation (n=101; 31.1%); beta-blocker or verapamil treatment (n=304; 93.8%), combined with disopyramide (n=28; 8.7%); and eligibility for septal reduction therapy (n=96; 29%). At the end of a median follow-up of 66 (34-106) months, 319 (98.2%) were treated for obstruction (n=43 [13.2%] received disopyramide), 46 (14.2%) underwent septal reduction therapy and the all-cause mortality rate was 1.9/100 person-years (95% confidence interval 1.4-2.6) (46 deaths). Moreover, 41 (8.9%) patients from the initial hypertrophic obstructive cardiomyopathy group became eligible for a cardiac myosin inhibitor. CONCLUSIONS: In this cohort of patients with hypertrophic cardiomyopathy selected from the REMY registry, one third were eligible for a cardiac myosin inhibitor.


Sujet(s)
Cardiomyopathie hypertrophique , Agents cardiovasculaires , Enregistrements , Fonction ventriculaire gauche , Humains , Mâle , Cardiomyopathie hypertrophique/traitement médicamenteux , Cardiomyopathie hypertrophique/physiopathologie , Cardiomyopathie hypertrophique/mortalité , Cardiomyopathie hypertrophique/génétique , Cardiomyopathie hypertrophique/diagnostic , Femelle , Adulte d'âge moyen , France/épidémiologie , Résultat thérapeutique , Sujet âgé , Facteurs temps , Fonction ventriculaire gauche/effets des médicaments et des substances chimiques , Agents cardiovasculaires/usage thérapeutique , Agents cardiovasculaires/effets indésirables , Sélection de patients , Études prospectives , Myosines cardiaques/génétique , Benzylamines/usage thérapeutique , Adulte , Facteurs de risque , Obstacle à l'éjection ventriculaire/physiopathologie , Obstacle à l'éjection ventriculaire/traitement médicamenteux , Obstacle à l'éjection ventriculaire/étiologie , Uracile/analogues et dérivés
13.
J Clin Invest ; 134(9)2024 May 01.
Article de Anglais | MEDLINE | ID: mdl-38690729

RÉSUMÉ

The myosin inhibitor mavacamten has transformed the management of obstructive hypertrophic cardiomyopathy (HCM) by targeting myosin ATPase activity to mitigate cardiac hypercontractility. This therapeutic mechanism has proven effective for patients with HCM independent of having a primary gene mutation in myosin. In this issue of the JCI, Buvoli et al. report that muscle hypercontractility is a mechanism of pathogenesis underlying muscle dysfunction in Laing distal myopathy, a disorder characterized by mutations altering the rod domain of ß myosin heavy chain. The authors performed detailed physiological, molecular, and biomechanical analyses and demonstrated that myosin ATPase inhibition can correct a large extent of muscle abnormalities. The findings offer a therapeutic avenue for Laing distal myopathy and potentially other myopathies. This Commentary underscores the importance of reevaluating myosin activity's role across myopathies in general for the potential development of targeted myosin inhibitors to treat skeletal muscle disorders.


Sujet(s)
Benzylamines , Muscles squelettiques , Uracile/analogues et dérivés , Humains , Muscles squelettiques/métabolisme , Muscles squelettiques/anatomopathologie , Cardiomyopathie hypertrophique/traitement médicamenteux , Cardiomyopathie hypertrophique/métabolisme , Cardiomyopathie hypertrophique/génétique , Chaînes lourdes de myosine/génétique , Chaînes lourdes de myosine/métabolisme , Myopathies distales/génétique , Myopathies distales/traitement médicamenteux , Myopathies distales/métabolisme , Myopathies distales/anatomopathologie , Animaux , Mutation , Myosines/métabolisme , Myosines/génétique
14.
BMC Cardiovasc Disord ; 24(1): 282, 2024 May 29.
Article de Anglais | MEDLINE | ID: mdl-38811883

RÉSUMÉ

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.


Sujet(s)
Autopsie , Mort subite cardiaque , Herpèsvirus humain de type 6 , Myocardite , Parvovirus humain B19 , Humains , Myocardite/virologie , Myocardite/anatomopathologie , Myocardite/génétique , Mort subite cardiaque/étiologie , Mort subite cardiaque/anatomopathologie , Mort subite cardiaque/prévention et contrôle , Mâle , Adulte , Femelle , Herpèsvirus humain de type 6/génétique , Herpèsvirus humain de type 6/isolement et purification , Parvovirus humain B19/génétique , Cardiomyopathie dilatée/génétique , Cardiomyopathie dilatée/virologie , Cardiomyopathie dilatée/anatomopathologie , Infections à roséolovirus/complications , Infections à roséolovirus/virologie , Infections à roséolovirus/diagnostic , Infections à roséolovirus/anatomopathologie , Cardiomyopathie hypertrophique/génétique , Cardiomyopathie hypertrophique/anatomopathologie , Infections à Parvoviridae/complications , Jeune adulte , Prédisposition génétique à une maladie , Issue fatale , Infections à virus Epstein-Barr/complications , Herpèsvirus humain de type 4/génétique , Co-infection , Cause de décès , Mutation , Adulte d'âge moyen
15.
J Phys Chem B ; 128(19): 4716-4727, 2024 May 16.
Article de Anglais | MEDLINE | ID: mdl-38708944

RÉSUMÉ

Hypertrophic cardiomyopathy is one of the most common forms of genetic cardiomyopathy. Mavacamten is a first-in-class myosin modulator that was identified via activity screening on the wild type, and it is FDA-approved for the treatment of obstructive hypertrophic cardiomyopathy (HCM). The drug selectively binds to the cardiac ß-myosin, inhibiting myosin function to decrease cardiac contractility. Though the drug is thought to affect multiple steps of the myosin cross-bridge cycle, its detailed mechanism of action is still under investigation. Individual steps in the overall cross-bridge cycle must be queried to elucidate the full mechanism of action. In this study, we utilize the rare-event method of transition path sampling to generate reactive trajectories to gain insights into the action of the drug on the dynamics and rate of the ATP hydrolysis step for human cardiac ß-myosin. We study three known HCM causative myosin mutations: R453C, P710R, and R712L to observe the effect of the drug on the alterations caused by these mutations in the chemical step. Since the crystal structure of the drug-bound myosin was not available at the time of this work, we created a model of the drug-bound system utilizing a molecular docking approach. We find a significant effect of the drug in one case, where the actual mechanism of the reaction is altered from the wild type by mutation. The drug restores both the rate of hydrolysis to the wildtype level and the mechanism of the reaction. This is a way to check the effect of the drug on untested mutations.


Sujet(s)
Adénosine triphosphate , Cardiomyopathie hypertrophique , Mutation , Humains , Hydrolyse , Adénosine triphosphate/métabolisme , Adénosine triphosphate/composition chimique , Cardiomyopathie hypertrophique/métabolisme , Cardiomyopathie hypertrophique/génétique , Cardiomyopathie hypertrophique/traitement médicamenteux , Biocatalyse , Simulation de dynamique moléculaire , Myosines/composition chimique , Myosines/métabolisme , Myosines/génétique , Benzylamines , Uracile/analogues et dérivés
16.
Comput Biol Med ; 175: 108499, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38677172

RÉSUMÉ

Familial hypertrophic cardiomyopathy (HCM) is a significant precursor of heart failure and sudden cardiac death, primarily caused by mutations in sarcomeric and structural proteins. Despite the extensive research on the HCM genotype, the complex and context-specific nature of many signaling and metabolic pathways linking the HCM genotype to phenotype has hindered therapeutic advancements for patients. Here, we have developed a computational model of HCM encompassing cardiomyocyte signaling and metabolic networks and their associated interactions. Utilizing a stochastic logic-based ODE approach, we linked cardiomyocyte signaling to the metabolic network through a gene regulatory network and post-translational modifications. We validated the model against published data on activities of signaling species in the HCM context and transcriptomes of two HCM mouse models (i.e., R403Q-αMyHC and R92W-TnT). Our model predicts that HCM mutation induces changes in metabolic functions such as ATP synthase deficiency and a transition from fatty acids to carbohydrate metabolism. The model indicated major shifts in glutamine-related metabolism and increased apoptosis after HCM-induced ATP synthase deficiency. We predicted that the transcription factors STAT, SRF, GATA4, TP53, and FoxO are the key regulators of cardiomyocyte hypertrophy and apoptosis in HCM in alignment with experiments. Moreover, we identified shared (e.g., activation of PGC1α by AMPK, and FHL1 by titin) and context-specific mechanisms (e.g., regulation of Ca2+ sensitivity by titin in HCM patients) that may control genotype-to-phenotype transition in HCM across different species or mutations. We also predicted potential combination drug targets for HCM (e.g., mavacamten plus ROS inhibitors) preventing or reversing HCM phenotype (i.e., hypertrophic growth, apoptosis, and metabolic remodeling) in cardiomyocytes. This study provides new insights into mechanisms linking genotype to phenotype in familial hypertrophic cardiomyopathy and offers a framework for assessing new treatments and exploring variations in HCM experimental models.


Sujet(s)
Cardiomyopathie hypertrophique , Myocytes cardiaques , Myocytes cardiaques/métabolisme , Animaux , Souris , Humains , Cardiomyopathie hypertrophique/génétique , Cardiomyopathie hypertrophique/métabolisme , Transduction du signal , Modèles cardiovasculaires , Phénotype , Génotype
17.
Proc Natl Acad Sci U S A ; 121(19): e2318413121, 2024 May 07.
Article de Anglais | MEDLINE | ID: mdl-38683993

RÉSUMÉ

Determining the pathogenicity of hypertrophic cardiomyopathy-associated mutations in the ß-myosin heavy chain (MYH7) can be challenging due to its variable penetrance and clinical severity. This study investigates the early pathogenic effects of the incomplete-penetrant MYH7 G256E mutation on myosin function that may trigger pathogenic adaptations and hypertrophy. We hypothesized that the G256E mutation would alter myosin biomechanical function, leading to changes in cellular functions. We developed a collaborative pipeline to characterize myosin function across protein, myofibril, cell, and tissue levels to determine the multiscale effects on structure-function of the contractile apparatus and its implications for gene regulation and metabolic state. The G256E mutation disrupts the transducer region of the S1 head and reduces the fraction of myosin in the folded-back state by 33%, resulting in more myosin heads available for contraction. Myofibrils from gene-edited MYH7WT/G256E human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) exhibited greater and faster tension development. This hypercontractile phenotype persisted in single-cell hiPSC-CMs and engineered heart tissues. We demonstrated consistent hypercontractile myosin function as a primary consequence of the MYH7 G256E mutation across scales, highlighting the pathogenicity of this gene variant. Single-cell transcriptomic and metabolic profiling demonstrated upregulated mitochondrial genes and increased mitochondrial respiration, indicating early bioenergetic alterations. This work highlights the benefit of our multiscale platform to systematically evaluate the pathogenicity of gene variants at the protein and contractile organelle level and their early consequences on cellular and tissue function. We believe this platform can help elucidate the genotype-phenotype relationships underlying other genetic cardiovascular diseases.


Sujet(s)
Myosines cardiaques , Cardiomyopathie hypertrophique , Cellules souches pluripotentes induites , Contraction myocardique , Myocytes cardiaques , Chaînes lourdes de myosine , Humains , Chaînes lourdes de myosine/génétique , Chaînes lourdes de myosine/métabolisme , Myosines cardiaques/génétique , Myosines cardiaques/métabolisme , Cardiomyopathie hypertrophique/génétique , Cardiomyopathie hypertrophique/métabolisme , Cellules souches pluripotentes induites/métabolisme , Myocytes cardiaques/métabolisme , Myocytes cardiaques/anatomopathologie , Contraction myocardique/génétique , Mutation , Mitochondries/métabolisme , Mitochondries/génétique , Myofibrilles/métabolisme , Respiration cellulaire/génétique
18.
Cell Rep Med ; 5(5): 101520, 2024 May 21.
Article de Anglais | MEDLINE | ID: mdl-38642550

RÉSUMÉ

Pathogenic variants in MYH7 and MYBPC3 account for the majority of hypertrophic cardiomyopathy (HCM). Targeted drugs like myosin ATPase inhibitors have not been evaluated in children. We generate patient and variant-corrected iPSC-cardiomyocytes (CMs) from pediatric HCM patients harboring single variants in MYH7 (V606M; R453C), MYBPC3 (G148R) or digenic variants (MYBPC3 P955fs, TNNI3 A157V). We also generate CMs harboring MYBPC3 mono- and biallelic variants using CRISPR editing of a healthy control. Compared with isogenic and healthy controls, variant-positive CMs show sarcomere disorganization, higher contractility, calcium transients, and ATPase activity. However, only MYH7 and biallelic MYBPC3 variant-positive CMs show stronger myosin-actin binding. Targeted myosin ATPase inhibitors show complete rescue of the phenotype in variant-positive CMs and in cardiac Biowires to mirror isogenic controls. The response is superior to verapamil or metoprolol. Myosin inhibitors can be effective in genotypically diverse HCM highlighting the need for myosin inhibitor drug trials in pediatric HCM.


Sujet(s)
Myosines cardiaques , Cardiomyopathie hypertrophique , Cellules souches pluripotentes induites , Myocytes cardiaques , Chaînes lourdes de myosine , Humains , Cellules souches pluripotentes induites/métabolisme , Cellules souches pluripotentes induites/effets des médicaments et des substances chimiques , Cardiomyopathie hypertrophique/génétique , Cardiomyopathie hypertrophique/traitement médicamenteux , Cardiomyopathie hypertrophique/anatomopathologie , Cardiomyopathie hypertrophique/métabolisme , Myosines cardiaques/génétique , Myosines cardiaques/métabolisme , Enfant , Myocytes cardiaques/métabolisme , Myocytes cardiaques/effets des médicaments et des substances chimiques , Myocytes cardiaques/anatomopathologie , Chaînes lourdes de myosine/génétique , Chaînes lourdes de myosine/métabolisme , Protéines de transport/génétique , Protéines de transport/métabolisme , Génotype , Myosines/métabolisme , Myosines/génétique , Mâle , Femelle , Sarcomères/métabolisme , Sarcomères/génétique
19.
J Mol Cell Cardiol ; 191: 27-39, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38648963

RÉSUMÉ

Approximately 40% of hypertrophic cardiomyopathy (HCM) mutations are linked to the sarcomere protein cardiac myosin binding protein-C (cMyBP-C). These mutations are either classified as missense mutations or truncation mutations. One mutation whose nature has been inconsistently reported in the literature is the MYBPC3-c.772G > A mutation. Using patient-derived human induced pluripotent stem cells differentiated to cardiomyocytes (hiPSC-CMs), we have performed a mechanistic study of the structure-function relationship for this MYBPC3-c.772G > A mutation versus a mutation corrected, isogenic cell line. Our results confirm that this mutation leads to exon skipping and mRNA truncation that ultimately suggests ∼20% less cMyBP-C protein (i.e., haploinsufficiency). This, in turn, results in increased myosin recruitment and accelerated myofibril cycling kinetics. Our mechanistic studies suggest that faster ADP release from myosin is a primary cause of accelerated myofibril cross-bridge cycling due to this mutation. Additionally, the reduction in force generating heads expected from faster ADP release during isometric contractions is outweighed by a cMyBP-C phosphorylation mediated increase in myosin recruitment that leads to a net increase of myofibril force, primarily at submaximal calcium activations. These results match well with our previous report on contractile properties from myectomy samples of the patients from whom the hiPSC-CMs were generated, demonstrating that these cell lines are a good model to study this pathological mutation and extends our understanding of the mechanisms of altered contractile properties of this HCM MYBPC3-c.772G > A mutation.


Sujet(s)
Cardiomyopathie hypertrophique , Protéines de transport , Haploinsuffisance , Cellules souches pluripotentes induites , Mutation , Myocytes cardiaques , Humains , Cardiomyopathie hypertrophique/génétique , Cardiomyopathie hypertrophique/métabolisme , Myocytes cardiaques/métabolisme , Protéines de transport/génétique , Protéines de transport/métabolisme , Cellules souches pluripotentes induites/métabolisme , Myosines/métabolisme , Myosines/génétique , Différenciation cellulaire/génétique , Cinétique
20.
Cell Mol Biol (Noisy-le-grand) ; 70(4): 61-67, 2024 Apr 28.
Article de Anglais | MEDLINE | ID: mdl-38678626

RÉSUMÉ

The purpose of this study was to explore the correlations of interleukin-1 (IL-1) and IL-6 gene polymorphisms with hypertrophic cardiomyopathy (HCM). A total of 200 patients with HCM were enrolled as disease group, and 200 healthy individuals were included as control group. Peripheral blood was collected from all subjects in both disease and control groups. Gene polymorphisms and serum expression levels of IL-1 and IL-6 were detected, and conjoint analysis was performed based on results of cardiac color Doppler ultrasound examination. The allele distribution of IL-1 rs1878320 showed a difference between disease and control groups (P=0.000). The frequency of the allele T was lower in disease group. The genotype distribution of IL-1 rs1878320 (P=0.001) and IL-6 rs1474347 (P=0.000) in disease group was different from that in control. The frequency of TC genotype of IL-1 rs1878320 was lower in disease group, and that of CA genotype of IL-6 rs1474347 was higher in disease group. There was a difference in the distribution of the dominant model of IL-6 rs1474347 between disease and control groups (P=0.021), and the frequency of CC + CA in the dominant model was 171 (0.855). The frequency of AC haplotype of IL-1 gene was overtly higher in disease group (P=0.000), while the frequency of AT haplotype was lower in disease group (P=0.000). The IL-1 rs1516792 polymorphism had an association with serum IL-1 level (P<0.05), the IL-1 level was notably increased in the patients with the genotype AA, and it was higher in disease group. The polymorphism of rs1878320 locus in IL-1 gene was correlated with interventricular septal (IVS) (P=0.047), and IVS was reduced in the patients with TC genotype. The polymorphism of rs1516792 locus in IL-1 gene was distinctly related to left ventricular outflow tract (LVOT) (P=0.041), and LVOT was lowered in the patients with GG genotype. The IL-6 rs2069831 polymorphism was associated with left ventricular ejection fraction (LVEF) (P=0.035), and LVEF declined in the patients with TT genotype. The IL-1 and IL-6 gene polymorphisms are correlated with the susceptibility and progression of HCM.


Sujet(s)
Cardiomyopathie hypertrophique , Interleukine-1 , Interleukine-6 , Polymorphisme de nucléotide simple , Adulte , Femelle , Humains , Mâle , Adulte d'âge moyen , Allèles , Cardiomyopathie hypertrophique/génétique , Études cas-témoins , Fréquence d'allèle/génétique , Prédisposition génétique à une maladie , Génotype , Interleukine-1/sang , Interleukine-1/génétique , Interleukine-6/sang , Interleukine-6/génétique , Polymorphisme de nucléotide simple/génétique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...