Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 819
Filtrer
1.
Sci Rep ; 14(1): 15422, 2024 07 04.
Article de Anglais | MEDLINE | ID: mdl-38965264

RÉSUMÉ

Hypertrophic cardiomyopathy (HCM) is an inherited disorder characterized by left ventricular hypertrophy and diastolic dysfunction, and increases the risk of arrhythmias and heart failure. Some patients with HCM develop a dilated phase of hypertrophic cardiomyopathy (D-HCM) and have poor prognosis; however, its pathogenesis is unclear and few pathological models exist. This study established disease-specific human induced pluripotent stem cells (iPSCs) from a patient with D-HCM harboring a mutation in MYBPC3 (c.1377delC), a common causative gene of HCM, and investigated the associated pathophysiological mechanisms using disease-specific iPSC-derived cardiomyocytes (iPSC-CMs). We confirmed the expression of pluripotent markers and the ability to differentiate into three germ layers in D-HCM patient-derived iPSCs (D-HCM iPSCs). D-HCM iPSC-CMs exhibited disrupted myocardial sarcomere structures and an increased number of damaged mitochondria. Ca2+ imaging showed increased abnormal Ca2+ signaling and prolonged decay time in D-HCM iPSC-CMs. Cell metabolic analysis revealed increased basal respiration, maximal respiration, and spare-respiratory capacity in D-HCM iPSC-CMs. RNA sequencing also showed an increased expression of mitochondrial electron transport system-related genes. D-HCM iPSC-CMs showed abnormal Ca2+ handling and hypermetabolic state, similar to that previously reported for HCM patient-derived iPSC-CMs. Although further studies are required, this is expected to be a useful pathological model for D-HCM.


Sujet(s)
Calcium , Cardiomyopathie hypertrophique , Protéines de transport , Mutation avec décalage du cadre de lecture , Cellules souches pluripotentes induites , Myocytes cardiaques , Cellules souches pluripotentes induites/métabolisme , Humains , Myocytes cardiaques/métabolisme , Myocytes cardiaques/anatomopathologie , Cardiomyopathie hypertrophique/génétique , Cardiomyopathie hypertrophique/métabolisme , Cardiomyopathie hypertrophique/anatomopathologie , Calcium/métabolisme , Protéines de transport/génétique , Protéines de transport/métabolisme , Signalisation calcique , Différenciation cellulaire , Mâle
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.
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
4.
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
5.
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
6.
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
7.
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
8.
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
9.
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
10.
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
11.
JCI Insight ; 9(8)2024 Mar 14.
Article de Anglais | MEDLINE | ID: mdl-38483507

RÉSUMÉ

The polymerization of myosin molecules into thick filaments in muscle sarcomeres is essential for cardiac contractility, with the attenuation of interactions between the heads of myosin molecules within the filaments being proposed to result in hypercontractility, as observed in hypertrophic cardiomyopathy (HCM). However, experimental evidence demonstrates that the structure of these giant macromolecular complexes is highly dynamic, with molecules exchanging between the filaments and a pool of soluble molecules on the minute timescale. Therefore, we sought to test the hypothesis that the enhancement of interactions between the heads of myosin molecules within thick filaments limits the mobility of myosin by taking advantage of mavacamten, a small molecule approved for the treatment of HCM. Myosin molecules were labeled in vivo with a green fluorescent protein (GFP) and imaged in intact hearts using multiphoton microscopy. Treatment of the intact hearts with mavacamten resulted in an unexpected > 5-fold enhancement in GFP-myosin mobility within the sarcomere. In vitro biochemical assays suggested that mavacamten enhanced the mobility of GFP-myosin by increasing the solubility of myosin molecules, through the stabilization of a compact/folded conformation of the molecules, once disassociated from the thick filaments. These findings provide alternative insight into the mechanisms by which molecules exchange into and out of thick filaments and have implications for how mavacamten may affect cardiac contractility.


Sujet(s)
Benzylamines , Myocarde , Sarcomères , Solubilité , Uracile/analogues et dérivés , Animaux , Sarcomères/métabolisme , Myocarde/métabolisme , Souris , Myosines/métabolisme , Pliage des protéines , Protéines à fluorescence verte/métabolisme , Protéines à fluorescence verte/génétique , Cardiomyopathie hypertrophique/métabolisme , Contraction myocardique , Humains , Mâle
12.
Cell Mol Life Sci ; 81(1): 158, 2024 Mar 31.
Article de Anglais | MEDLINE | ID: mdl-38556571

RÉSUMÉ

Mutations in cysteine and glycine-rich protein 3 (CSRP3)/muscle LIM protein (MLP), a key regulator of striated muscle function, have been linked to hypertrophic cardiomyopathy (HCM) and dilated cardiomyopathy (DCM) in patients. However, the roles of CSRP3 in heart development and regeneration are not completely understood. In this study, we characterized a novel zebrafish gene-trap line, gSAIzGFFM218A, which harbors an insertion in the csrp3 genomic locus, heterozygous fish served as a csrp3 expression reporter line and homozygous fish served as a csrp3 mutant line. We discovered that csrp3 is specifically expressed in larval ventricular cardiomyocytes (CMs) and that csrp3 deficiency leads to excessive trabeculation, a common feature of CSRP3-related HCM and DCM. We further revealed that csrp3 expression increased in response to different cardiac injuries and was regulated by several signaling pathways vital for heart regeneration. Csrp3 deficiency impeded zebrafish heart regeneration by impairing CM dedifferentiation, hindering sarcomere reassembly, and reducing CM proliferation while aggravating apoptosis. Csrp3 overexpression promoted CM proliferation after injury and ameliorated the impairment of ventricle regeneration caused by pharmacological inhibition of multiple signaling pathways. Our study highlights the critical role of Csrp3 in both zebrafish heart development and regeneration, and provides a valuable animal model for further functional exploration that will shed light on the molecular pathogenesis of CSRP3-related human cardiac diseases.


Sujet(s)
Cardiomyopathie hypertrophique , Protéines à domaine LIM , Danio zébré , Animaux , Humains , Danio zébré/génétique , Danio zébré/métabolisme , Cystéine/génétique , Cystéine/métabolisme , Protéines du muscle/génétique , Protéines du muscle/métabolisme , Cardiomyopathie hypertrophique/génétique , Cardiomyopathie hypertrophique/métabolisme , Myocytes cardiaques/métabolisme
13.
Eur J Med Res ; 29(1): 109, 2024 Feb 09.
Article de Anglais | MEDLINE | ID: mdl-38336819

RÉSUMÉ

INTRODUCTION: Salusins, which are translated from the alternatively spliced mRNA of torsin family 2 member A (TOR2A), play a vital role in regulation of various cardiovascular diseases. However, it remains unclear precisely regarding their roles in hypertrophic cardiomyopathy (HCM). Therefore, this study was conducted to explore therapeutic effect and the underlying mechanisms of salusins on HCM. MATERIAL AND METHODS: In vivo experiments, Sprague-Dawley rats were used to induce HCM model by angiotensin (Ang) II infusion for 4 weeks. The rats were randomly divided into four groups, namely, Saline + Control shRNA (n = 7), Ang II + Control shRNA (n = 8), Saline + TOR2A shRNA (n = 7), and Ang II + TOR2A shRNA groups (n = 8). After HCM induction, doppler echocardiography is recommended to evaluate heart function. In vitro experiments, primary neonatal rat cardiomyocytes (NRCMs) and cardiac fibroblasts (NRCFs) were obtained from newborn rats, and were treated with Ang II (10-6 M) for 24 h. RESULTS: After treatment with Ang II, levels of salusin-α and salusin-ß were elevated in serum and cardiac tissues of rats and in the neonatal rat cardiomyocytes and cardiac fibroblasts. Downregulation of salusins alleviated the Ang II-induced cardiac hypertrophy by suppressing the increased atrial natriuretic peptide (ANP), brain natriuretic peptide (BNP) and beta-myosin heavy chain (ß-MHC) and cardiac fibrosis by blocking collagen I, collagen III and transforming growth factor-beta (TGF-ß), and it also attenuated oxidative stress by suppressing the increased reactive oxygen species (ROS) and malondialdehyde (MDA) levels and reversing the decreased superoxide dismutase (SOD) activity and autophagy by inhibiting the increased microtubule-associated protein light chain 3B (LC3B), Beclin1, autophagy related gene (Atg) 3 and Atg5 in the cardiac tissues of Ang II-infused rats and in the Ang II-treated NRCMs. CONCLUSIONS: All these findings suggest that the levels of salusins were elevated in the HCM, and targeting of salusins contributes to alleviation of cardiac hypertrophy and fibrosis probably via attenuating oxidative stress and autophagy. Accordingly, targeting of salusins may be a strategy for HCM therapy.


Sujet(s)
Cardiomyopathie hypertrophique , Rats , Animaux , Rat Sprague-Dawley , Régulation négative , Cardiomyopathie hypertrophique/génétique , Cardiomyopathie hypertrophique/métabolisme , Cardiomégalie/induit chimiquement , Cardiomégalie/génétique , Cardiomégalie/métabolisme , Myocytes cardiaques , Angiotensine-II/génétique , Angiotensine-II/métabolisme , Angiotensine-II/pharmacologie , Stress oxydatif , Petit ARN interférent/effets indésirables , Petit ARN interférent/métabolisme , Autophagie/génétique , Collagène/génétique
14.
Circ Genom Precis Med ; 17(2): e004377, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38362799

RÉSUMÉ

BACKGROUND: Pathogenic autosomal-dominant missense variants in MYH7 (myosin heavy chain 7), which encodes the sarcomeric protein (ß-MHC [beta myosin heavy chain]) expressed in cardiac and skeletal myocytes, are a leading cause of hypertrophic cardiomyopathy and are clinically actionable. However, ≈75% of MYH7 missense variants are of unknown significance. While human-induced pluripotent stem cells (hiPSCs) can be differentiated into cardiomyocytes to enable the interrogation of MYH7 variant effect in a disease-relevant context, deep mutational scanning has not been executed using diploid hiPSC derivates due to low hiPSC gene-editing efficiency. Moreover, multiplexable phenotypes enabling deep mutational scanning of MYH7 variant hiPSC-derived cardiomyocytes are unknown. METHODS: To overcome these obstacles, we used CRISPRa On-Target Editing Retrieval enrichment to generate an hiPSC library containing 113 MYH7 codon variants suitable for deep mutational scanning. We first established that ß-MHC protein loss occurs in a hypertrophic cardiomyopathy human heart with a pathogenic MYH7 variant. We then differentiated the MYH7 missense variant hiPSC library to cardiomyocytes for multiplexed assessment of ß-MHC variant abundance by massively parallel sequencing and hiPSC-derived cardiomyocyte survival. RESULTS: Both the multiplexed assessment of ß-MHC abundance and hiPSC-derived cardiomyocyte survival accurately segregated all known pathogenic variants from synonymous variants. Functional data were generated for 4 variants of unknown significance and 58 additional MYH7 missense variants not yet detected in patients. CONCLUSIONS: This study leveraged hiPSC differentiation into disease-relevant cardiomyocytes to enable multiplexed assessments of MYH7 missense variants for the first time. Phenotyping strategies used here enable the application of deep mutational scanning to clinically actionable genes, which should reduce the burden of variants of unknown significance on patients and clinicians.


Sujet(s)
Cardiomyopathie hypertrophique , Cellules souches pluripotentes induites , Humains , Myocytes cardiaques/métabolisme , Chaînes lourdes de myosine/génétique , Cellules souches pluripotentes induites/métabolisme , Cardiomyopathie hypertrophique/génétique , Cardiomyopathie hypertrophique/métabolisme , Différenciation cellulaire/génétique , Myosines cardiaques/génétique
15.
Circ Res ; 134(3): 290-306, 2024 02 02.
Article de Anglais | MEDLINE | ID: mdl-38197258

RÉSUMÉ

BACKGROUND: Hypertrophic cardiomyopathy (HCM) is the most prevalent monogenic heart disorder. However, the pathogenesis of HCM, especially its nongenetic mechanisms, remains largely unclear. Transcription factors are known to be involved in various biological processes including cell growth. We hypothesized that SP1 (specificity protein 1), the first purified TF in mammals, plays a role in the cardiomyocyte growth and cardiac hypertrophy of HCM. METHODS: Cardiac-specific conditional knockout of Sp1 mice were constructed to investigate the role of SP1 in the heart. The echocardiography, histochemical experiment, and transmission electron microscope were performed to analyze the cardiac phenotypes of cardiac-specific conditional knockout of Sp1 mice. RNA sequencing, chromatin immunoprecipitation sequencing, and adeno-associated virus experiments in vivo were performed to explore the downstream molecules of SP1. To examine the therapeutic effect of SP1 on HCM, an SP1 overexpression vector was constructed and injected into the mutant allele of Myh6 R404Q/+ (Myh6 c. 1211C>T) HCM mice. The human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) from a patient with HCM were used to detect the potential therapeutic effects of SP1 in human HCM. RESULTS: The cardiac-specific conditional knockout of Sp1 mice developed a typical HCM phenotype, displaying overt myocardial hypertrophy, interstitial fibrosis, and disordered myofilament. In addition, Sp1 knockdown dramatically increased the cell area of hiPSC-CMs and caused intracellular myofibrillar disorganization, which was similar to the hypertrophic cardiomyocytes of HCM. Mechanistically, Tuft1 was identified as the key target gene of SP1. The hypertrophic phenotypes induced by Sp1 knockdown in both hiPSC-CMs and mice could be rescued by TUFT1 (tuftelin 1) overexpression. Furthermore, SP1 overexpression suppressed the development of HCM in the mutant allele of Myh6 R404Q/+ mice and also reversed the hypertrophic phenotype of HCM hiPSC-CMs. CONCLUSIONS: Our study demonstrates that SP1 deficiency leads to HCM. SP1 overexpression exhibits significant therapeutic effects on both HCM mice and HCM hiPSC-CMs, suggesting that SP1 could be a potential intervention target for HCM.


Sujet(s)
Cardiomyopathie hypertrophique , Cellules souches pluripotentes induites , Humains , Souris , Animaux , Cellules souches pluripotentes induites/métabolisme , Cardiomyopathie hypertrophique/métabolisme , Myofibrilles/métabolisme , Myocytes cardiaques/métabolisme , Cardiomégalie/métabolisme , Facteurs de transcription/métabolisme , Mammifères
16.
Cell Prolif ; 57(4): e13573, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-37916452

RÉSUMÉ

Pompe disease (PD) is a rare autosomal recessive disorder that presents with progressive hypertrophic cardiomyopathy. However, the detailed mechanism remains clarified. Herein, PD patient-specific induced pluripotent stem cells were differentiated into cardiomyocytes (PD-iCMs) that exhibited cardiomyopathic features of PD, including decreased acid alpha-glucosidase activity, lysosomal glycogen accumulation and hypertrophy. The defective mitochondria were involved in the cardiac pathology as shown by the significantly decreased number of mitochondria and impaired respiratory function and ATP production in PD-iCMs, which was partially due to elevated levels of intracellular reactive oxygen species produced from depolarized mitochondria. Further analysis showed that impaired fusion and autophagy of mitochondria and declined expression of mitochondrial complexes underlies the mechanism of dysfunctional mitochondria. This was alleviated by supplementation with recombinant human acid alpha-glucosidase that improved the mitochondrial function and concomitantly mitigated the cardiac pathology. Therefore, this study suggests that defective mitochondria underlie the pathogenesis of cardiomyopathy in patients with PD.


Sujet(s)
Cardiomyopathie hypertrophique , Glycogénose de type II , Cellules souches pluripotentes induites , Maladies mitochondriales , Humains , Glycogénose de type II/métabolisme , Glycogénose de type II/anatomopathologie , Myocytes cardiaques/métabolisme , Cellules souches pluripotentes induites/métabolisme , Cardiomyopathie hypertrophique/métabolisme , Cardiomyopathie hypertrophique/anatomopathologie , Maladies mitochondriales/métabolisme , Maladies mitochondriales/anatomopathologie
17.
J Mol Cell Cardiol ; 186: 45-56, 2024 01.
Article de Anglais | MEDLINE | ID: mdl-37979444

RÉSUMÉ

Cardiac hypertrophy can develop to end-stage heart failure (HF), which inevitably leading to heart transplantation or death. Preserving cardiac function in cardiomyocytes (CMs) is essential for improving prognosis in hypertrophic cardiomyopathy (HCM) patients. Therefore, understanding transcriptomic heterogeneity of CMs in HCM would be indispensable to aid potential therapeutic targets investigation. We isolated primary CM from HCM patients who had extended septal myectomy, and obtained transcriptomes in 338 human primary CM with single-cell tagged reverse transcription (STRT-seq) approach. Our results revealed that CMs could be categorized into three subsets in nonfailing HCM heart: high energy synthesis cluster, high cellular metabolism cluster and intermediate cluster. The expression of electron transport chain (ETC) was up-regulated in larger-sized CMs from high energy synthesis cluster. Of note, we found the expression of Cytochrome c oxidase subunit 7B (COX7B), a subunit of Complex IV in ETC had trends of positively correlation with CMs size. Further, by assessing COX7B expression in HCM patients, we speculated that COX7B was compensatory up-regulated at early-stage but down-regulated in failing HCM heart. To test the hypothesis that COX7B might participate both in hypertrophy and HF progression, we used adeno associated virus 9 (AAV9) to mediate the expression of Cox7b in pressure overload-induced mice. Mice in vivo data supported that knockdown of Cox7b would accelerate HF and Cox7b overexpression could restore partial cardiac function in hypertrophy. Our result highlights targeting COX7B and preserving energy synthesis in hypertrophic CMs could be a promising translational direction for HF therapeutic strategy.


Sujet(s)
Cardiomyopathie hypertrophique , Défaillance cardiaque , Transplantation cardiaque , Humains , Animaux , Souris , Myocytes cardiaques/métabolisme , Défaillance cardiaque/génétique , Défaillance cardiaque/métabolisme , Cardiomyopathie hypertrophique/métabolisme , Cardiomégalie/génétique , Cardiomégalie/métabolisme
18.
Int J Mol Sci ; 24(19)2023 Oct 02.
Article de Anglais | MEDLINE | ID: mdl-37834293

RÉSUMÉ

Hypertrophic cardiomyopathy (HCM) is the most common inherited heart disease and the leading cause of sudden cardiac death in young people. Mutations in genes that encode structural proteins of the cardiac sarcomere are the more frequent genetic cause of HCM. The disease is characterized by cardiomyocyte hypertrophy and myocardial fibrosis, which is defined as the excessive deposition of extracellular matrix proteins, mainly collagen I and III, in the myocardium. The development of fibrotic tissue in the heart adversely affects cardiac function. In this review, we discuss the latest evidence on how cardiac fibrosis is promoted, the role of cardiac fibroblasts, their interaction with cardiomyocytes, and their activation via the TGF-ß pathway, the primary intracellular signalling pathway regulating extracellular matrix turnover. Finally, we summarize new findings on profibrotic genes as well as genetic and non-genetic factors involved in the pathophysiology of HCM.


Sujet(s)
Cardiomyopathie hypertrophique , Humains , Adolescent , Cardiomyopathie hypertrophique/métabolisme , Myocarde/métabolisme , Myocytes cardiaques/métabolisme , Fibroblastes/métabolisme , Fibrose
19.
J Mol Cell Cardiol ; 185: 26-37, 2023 12.
Article de Anglais | MEDLINE | ID: mdl-37797718

RÉSUMÉ

Hypertrophic cardiomyopathy (HCM) is the most prevalent inherited cardiac disease. Up to 40% of cases are associated with heterozygous mutations in myosin binding protein C (cMyBP-C, MYBPC3). Most of these mutations lead to premature termination codons (PTC) and patients show reduction of functional cMyBP-C. This so-called haploinsufficiency most likely contributes to disease development. We analyzed mechanisms underlying haploinsufficiency using cardiac tissue from HCM-patients with truncation mutations in MYBPC3 (MYBPC3trunc). We compared transcriptional activity, mRNA and protein expression to donor controls. To differentiate between HCM-specific and general hypertrophy-induced mechanisms we used patients with left ventricular hypertrophy due to aortic stenosis (AS) as an additional control. We show that cMyBP-C haploinsufficiency starts at the mRNA level, despite hypertrophy-induced increased transcriptional activity. Gene set enrichment analysis (GSEA) of RNA-sequencing data revealed an increased expression of NMD-components. Among them, Up-frameshift protein UPF3B, a regulator of NMD was upregulated in MYBPC3trunc patients and not in AS-patients. Strikingly, we show that in sarcomeres UPF3B but not UPF1 and UPF2 are localized to the Z-discs, the presumed location of sarcomeric protein translation. Our data suggest that cMyBP-C haploinsufficiency in HCM-patients is established by UPF3B-dependent NMD during the initial translation round at the Z-disc.


Sujet(s)
Cardiomyopathie hypertrophique , Myocytes cardiaques , Humains , Cardiomyopathie hypertrophique/métabolisme , Haploinsuffisance , Hypertrophie/métabolisme , Mutation , Myocytes cardiaques/métabolisme , ARN messager/génétique , ARN messager/métabolisme , Protéines de liaison à l'ARN/métabolisme
20.
Cardiovasc Res ; 119(15): 2550-2562, 2023 11 25.
Article de Anglais | MEDLINE | ID: mdl-37648651

RÉSUMÉ

AIMS: Cardiac fibrosis drives the progression of heart failure in ischaemic and hypertrophic cardiomyopathy. Therefore, the development of specific anti-fibrotic treatment regimens to counteract cardiac fibrosis is of high clinical relevance. Hence, this study examined the presence of persistent fibroblast activation during longstanding human heart disease at a single-cell resolution to identify putative therapeutic targets to counteract pathological cardiac fibrosis in patients. METHODS AND RESULTS: We used single-nuclei RNA sequencing with human tissues from two samples of one healthy donor, and five hypertrophic and two failing hearts. Unsupervised sub-clustering of 7110 nuclei led to the identification of 7 distinct fibroblast clusters. De-convolution of cardiac fibroblast heterogeneity revealed a distinct population of human cardiac fibroblasts with a molecular signature of persistent fibroblast activation and a transcriptional switch towards a pro-fibrotic extra-cellular matrix composition in patients with established cardiac hypertrophy and heart failure. This sub-cluster was characterized by high expression of POSTN, RUNX1, CILP, and a target gene adipocyte enhancer-binding protein 1 (AEBP1) (all P < 0.001). Strikingly, elevated circulating AEBP1 blood level were also detected in a validation cohort of patients with confirmed cardiac fibrosis and hypertrophic cardiomyopathy by cardiac magnetic resonance imaging (P < 0.01). Since endogenous AEBP1 expression was increased in patients with established cardiac hypertrophy and heart failure, we assessed the functional consequence of siRNA-mediated AEBP1 silencing in human cardiac fibroblasts. Indeed, AEBP1 silencing reduced proliferation, migration, and fibroblast contractile capacity and α-SMA gene expression, which is a hallmark of fibroblast activation (all P < 0.05). Mechanistically, the anti-fibrotic effects of AEBP1 silencing were linked to transforming growth factor-beta pathway modulation. CONCLUSION: Together, this study identifies persistent fibroblast activation in patients with longstanding heart disease, which might be detected by circulating AEBP1 and therapeutically modulated by its targeted silencing in human cardiac fibroblasts.


Sujet(s)
Cardiomyopathies , Cardiomyopathie hypertrophique , Cardiopathies , Défaillance cardiaque , Humains , Défaillance cardiaque/métabolisme , Cardiopathies/anatomopathologie , Cardiomégalie/métabolisme , Cardiomyopathie hypertrophique/métabolisme , Cardiomyopathies/métabolisme , Fibrose , Fibroblastes/métabolisme , Analyse de profil d'expression de gènes , Carboxypeptidases/métabolisme , Protéines de répression/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...