Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.256
Filtrar
1.
Proc Natl Acad Sci U S A ; 121(42): e2323052121, 2024 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-39378095

RESUMO

Cardiac myosin-specific (MyHC) T cells drive the disease pathogenesis of immune checkpoint inhibitor-associated myocarditis (ICI-myocarditis). To determine whether MyHC T cells are tissue-resident memory T (TRM) cells, we characterized cardiac TRM cells in naive mice and established that they have a distinct phenotypic and transcriptional profile that can be defined by their upregulation of CD69, PD-1, and CXCR6. We then investigated the effects of cardiac injury through a modified experimental autoimmune myocarditis mouse model and an ischemia-reperfusion injury mouse model and determined that cardiac inflammation induces the recruitment of autoreactive MyHC TRM cells, which coexpress PD-1 and CD69. To investigate whether the recruited MyHC TRM cells could increase susceptibility to ICI-myocarditis, we developed a two-hit ICI-myocarditis mouse model where cardiac injury was induced, mice were allowed to recover, and then were treated with anti-PD-1 antibodies. We determined that mice who recover from cardiac injury are more susceptible to ICI-myocarditis development. We found that murine and human TRM cells share a similar location in the heart and aggregate along the perimyocardium. We phenotyped cells obtained from pericardial fluid from patients diagnosed with dilated cardiomyopathy and ischemic cardiomyopathy and established that pericardial T cells are predominantly CD69+ TRM cells that up-regulate PD-1. Finally, we determined that human pericardial macrophages produce IL-15, which supports and maintains pericardial TRM cells.


Assuntos
Inibidores de Checkpoint Imunológico , Células T de Memória , Miocardite , Animais , Miocardite/imunologia , Miocardite/patologia , Miocardite/metabolismo , Inibidores de Checkpoint Imunológico/farmacologia , Camundongos , Humanos , Células T de Memória/imunologia , Células T de Memória/metabolismo , Modelos Animais de Doenças , Masculino , Receptor de Morte Celular Programada 1/metabolismo , Miosinas Cardíacas/imunologia , Miosinas Cardíacas/metabolismo , Antígenos de Diferenciação de Linfócitos T/metabolismo , Antígenos de Diferenciação de Linfócitos T/imunologia , Camundongos Endogâmicos C57BL , Lectinas Tipo C/metabolismo , Feminino , Miosinas/metabolismo , Miocárdio/imunologia , Miocárdio/patologia , Miocárdio/metabolismo , Antígenos CD
2.
BMC Med Genomics ; 17(1): 225, 2024 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-39237976

RESUMO

BACKGROUND: Pathogenic variants of MYH7, which encodes the beta-myosin heavy chain protein, are major causes of dilated and hypertrophic cardiomyopathy. METHODS: In this study, we used whole-genome sequencing data to identify MYH7 variants in 397 patients with various cardiomyopathy subtypes who were participating in the National Project of Bio Big Data pilot study in Korea. We also performed in silico analyses to predict the pathogenicity of the novel variants, comparing them to known pathogenic missense variants. RESULTS: We identified 27 MYH7 variants in 41 unrelated patients with cardiomyopathy, consisting of 20 previously known pathogenic/likely pathogenic variants, 2 variants of uncertain significance, and 5 novel variants. Notably, the pathogenic variants predominantly clustered within the myosin motor domain of MYH7. We confirmed that the novel identified variants could be pathogenic, as indicated by high prediction scores in the in silico analyses, including SIFT, Mutation Assessor, PROVEAN, PolyPhen-2, CADD, REVEL, MetaLR, MetaRNN, and MetaSVM. Furthermore, we assessed their damaging effects on protein dynamics and stability using DynaMut2 and Missense3D tools. CONCLUSIONS: Overall, our study identified the distribution of MYH7 variants among patients with cardiomyopathy in Korea, offering new insights for improved diagnosis by enriching the data on the pathogenicity of novel variants using in silico tools and evaluating the function and structural stability of the MYH7 protein.


Assuntos
Miosinas Cardíacas , Cadeias Pesadas de Miosina , Humanos , Cadeias Pesadas de Miosina/genética , Cadeias Pesadas de Miosina/química , Miosinas Cardíacas/genética , República da Coreia , Feminino , Masculino , Pessoa de Meia-Idade , Cardiomiopatias/genética , Simulação por Computador , Adulto , Mutação de Sentido Incorreto , Idoso
3.
Am J Hum Genet ; 111(10): 2219-2231, 2024 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-39226896

RESUMO

Bicuspid aortic valve (BAV) is the most common congenital heart lesion with an estimated population prevalence of 1%. We hypothesize that specific gene variants predispose to early-onset complications of BAV (EBAV). We analyzed whole-exome sequences (WESs) to identify rare coding variants that contribute to BAV disease in 215 EBAV-affected families. Predicted damaging variants in candidate genes with moderate or strong supportive evidence to cause developmental cardiac phenotypes were present in 107 EBAV-affected families (50% of total), including genes that cause BAV (9%) or heritable thoracic aortic disease (HTAD, 19%). After appropriate filtration, we also identified 129 variants in 54 candidate genes that are associated with autosomal-dominant congenital heart phenotypes, including recurrent deleterious variation of FBN2, MYH6, channelopathy genes, and type 1 and 5 collagen genes. These findings confirm our hypothesis that unique rare genetic variants drive early-onset presentations of BAV disease.


Assuntos
Valva Aórtica , Doença da Válvula Aórtica Bicúspide , Sequenciamento do Exoma , Doenças das Valvas Cardíacas , Linhagem , Humanos , Doença da Válvula Aórtica Bicúspide/genética , Doença da Válvula Aórtica Bicúspide/patologia , Valva Aórtica/anormalidades , Valva Aórtica/patologia , Doenças das Valvas Cardíacas/genética , Masculino , Feminino , Predisposição Genética para Doença , Idade de Início , Fenótipo , Exoma/genética , Adulto , Cadeias Pesadas de Miosina/genética , Fibrilina-2/genética , Miosinas Cardíacas/genética
4.
Int J Mol Sci ; 25(18)2024 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-39337273

RESUMO

Omecamtiv mecarbil (OM) is a small molecule that has been shown to improve the function of the slow human ventricular myosin (MyHC) motor through a complex perturbation of the thin/thick filament regulatory state of the sarcomere mediated by binding to myosin allosteric sites coupled to inorganic phosphate (Pi) release. Here, myofibrils from samples of human left ventricle (ß-slow MyHC-7) and left atrium (α-fast MyHC-6) from healthy donors were used to study the differential effects of µmolar [OM] on isometric force in relaxing conditions (pCa 9.0) and at maximal (pCa 4.5) or half-maximal (pCa 5.75) calcium activation, both under control conditions (15 °C; equimolar DMSO; contaminant inorganic phosphate [Pi] ~170 µM) and in the presence of 5 mM [Pi]. The activation state and OM concentration within the contractile lattice were rapidly altered by fast solution switching, demonstrating that the effect of OM was rapid and fully reversible with dose-dependent and myosin isoform-dependent features. In MyHC-7 ventricular myofibrils, OM increased submaximal and maximal Ca2+-activated isometric force with a complex dose-dependent effect peaking (40% increase) at 0.5 µM, whereas in MyHC-6 atrial myofibrils, it had no effect or-at concentrations above 5 µM-decreased the maximum Ca2+-activated force. In both ventricular and atrial myofibrils, OM strongly depressed the kinetics of force development and relaxation up to 90% at 10 µM [OM] and reduced the inhibition of force by inorganic phosphate. Interestingly, in the ventricle, but not in the atrium, OM induced a large dose-dependent Ca2+-independent force development and an increase in basal ATPase that were abolished by the presence of millimolar inorganic phosphate, consistent with the hypothesis that the widely reported Ca2+-sensitising effect of OM may be coupled to a change in the state of the thick filaments that resembles the on-off regulation of thin filaments by Ca2+. The complexity of this scenario may help to understand the disappointing results of clinical trials testing OM as inotropic support in systolic heart failure compared with currently available inotropic drugs that alter the calcium signalling cascade.


Assuntos
Contração Miocárdica , Miofibrilas , Ureia , Humanos , Ureia/análogos & derivados , Ureia/farmacologia , Miofibrilas/metabolismo , Miofibrilas/efeitos dos fármacos , Contração Miocárdica/efeitos dos fármacos , Cálcio/metabolismo , Miocárdio/metabolismo , Isoformas de Proteínas/metabolismo , Miosinas/metabolismo , Ventrículos do Coração/efeitos dos fármacos , Ventrículos do Coração/metabolismo , Masculino , Miosinas Cardíacas/metabolismo , Feminino , Adulto
5.
Pharmacol Res Perspect ; 12(5): e70006, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39257068

RESUMO

Aficamten, a cardiac myosin inhibitor, is being developed for the treatment of patients with symptomatic hypertrophic cardiomyopathy (HCM). The purpose of this study was to determine the absorption, metabolism, and excretion of aficamten. Eight healthy male participants received a single oral dose of 20 mg aficamten (containing approximately 100 µCi of radiocarbon). Blood, urine, and feces samples were collected up to a maximum of Day 26. The pharmacokinetics of aficamten were characterized by moderate absorption, with a median tmax of 2.0 h postdose. The median t1/2 of aficamten was 99.6 h with similar t1/2 observed for metabolites and total radioactivity in plasma and whole blood. The overall total recovery of administered total radioactivity was 89.7% with 57.7% of the dose recovered in feces and 32.0% in urine. The main circulating metabolites in plasma included monohydroxylated metabolites M1a (CK-3834282) and M1b (CK-3834283) accounting for 10.5% and 36.4% of the total radioactivity AUC both with a median tmax of 5 h. The other major plasma metabolite was M5 (an oxygen-linked glucuronide conjugate of M1a), which accounted for 10.3% of the total plasma radioactivity exposure, with a tmax of 24 h. In urine, M5 was the most abundant metabolite with 8.02% total radioactive dose (TRD), followed by M1a and M1b with 6.16% and 2.85% TRD, respectively; however, there were no metabolites in urine observed at >10% of dose. The major metabolite in feces was M18 representing 44.1% of the radioactive dose. These findings indicated that aficamten was eliminated by metabolism, and to a minor extent, by fecal excretion of unchanged aficamten with renal excretion playing a minor role. Feces were the principal route of excretion of the radioactive dose.


Assuntos
Biotransformação , Humanos , Masculino , Adulto , Fezes/química , Adulto Jovem , Miosinas Cardíacas/metabolismo , Pessoa de Meia-Idade , Administração Oral , Voluntários Saudáveis
6.
Biochemistry (Mosc) ; 89(7): 1273-1282, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-39218024

RESUMO

Lead and cadmium are heavy metals widely distributed in the environment and contribute significantly to cardiovascular morbidity and mortality. Using Leadmium Green dye, we have shown that lead and cadmium enter cardiomyocytes, distributing throughout the cell. Using an in vitro motility assay, we have shown that sliding velocity of actin and native thin filaments over myosin decreases with increasing concentrations of Pb2+ and Cd2+. Significantly lower concentrations of Pb2+ and Cd2+ (0.6 mM) were required to stop sliding of thin filaments over myosin compared to the stopping actin sliding over the same myosin (1.1-1.6 mM). Lower concentration of Cd2+ (1.1 mM) needed to stop actin sliding over myosin compared to the Pb2++Cd2+ combination (1.3 mM) and lead alone (1.6 mM). There were no differences found in the effects of lead and cadmium cations on relative force developed by myosin heads or number of actin filaments bound to myosin. Sliding velocity of actin over myosin in the left atrium, right and left ventricles changed equally when exposed to the same dose of the same metal. Thus, we have demonstrated for the first time that Pb2+ and Cd2+ can directly affect myosin and thin filament function, with Cd2+ exerting a more toxic influence on myosin function compared to Pb2+.


Assuntos
Citoesqueleto de Actina , Cádmio , Cátions Bivalentes , Chumbo , Cádmio/farmacologia , Animais , Citoesqueleto de Actina/metabolismo , Citoesqueleto de Actina/efeitos dos fármacos , Miosinas Cardíacas/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Actinas/metabolismo , Coelhos
7.
Sci Rep ; 14(1): 19685, 2024 08 24.
Artigo em Inglês | MEDLINE | ID: mdl-39181964

RESUMO

Prostate cancer (PRAD) is one of the leading malignancies in men all around the world. Here, we identified Myosin Heavy Chain 6 (MYH6) as a potential tumor suppressor gene in the development of prostate cancer. We found lower expression of MYH6 in prostate cancer tissues, and its lower gene expression was also associated with worse clinical outcomes. In vitro and in vivo assays indicated that overexpressed MYH6 could suppress the proliferation and migration progression of prostate cancer cells. RNA-seq was employed to investigate the mechanism, and KIT Proto-Oncogen (KIT) was determined as the downstream gene of MYH6, which was further confirmed using rescue assays. In all, we provide the evidence that MYH6 could serve as a tumor suppressor in prostate cancer. Our results highlight the potential role of MYH6 in the development of prostate cancer.


Assuntos
Movimento Celular , Proliferação de Células , Progressão da Doença , Regulação Neoplásica da Expressão Gênica , Cadeias Pesadas de Miosina , Neoplasias da Próstata , Proteínas Proto-Oncogênicas c-kit , Masculino , Humanos , Cadeias Pesadas de Miosina/genética , Cadeias Pesadas de Miosina/metabolismo , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Neoplasias da Próstata/metabolismo , Proliferação de Células/genética , Linhagem Celular Tumoral , Proteínas Proto-Oncogênicas c-kit/genética , Proteínas Proto-Oncogênicas c-kit/metabolismo , Movimento Celular/genética , Animais , Regulação para Baixo , Camundongos , Miosinas Cardíacas
8.
Nat Cardiovasc Res ; 3(8): 1003-1016, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39196032

RESUMO

Hypertrophic cardiomyopathy (HCM) is an inherited disease of the sarcomere resulting in excessive cardiac contractility. The first-in-class cardiac myosin inhibitor, mavacamten, improves symptoms in obstructive HCM. Here we present aficamten, a selective small-molecule inhibitor of cardiac myosin that diminishes ATPase activity by strongly slowing phosphate release, stabilizing a weak actin-binding state. Binding to an allosteric site on the myosin catalytic domain distinct from mavacamten, aficamten prevents the conformational changes necessary to enter the strongly actin-bound force-generating state. In doing so, aficamten reduces the number of functional myosin heads driving sarcomere shortening. The crystal structure of aficamten bound to cardiac myosin in the pre-powerstroke state provides a basis for understanding its selectivity over smooth and fast skeletal muscle. Furthermore, in cardiac myocytes and in mice bearing the hypertrophic R403Q cardiac myosin mutation, aficamten reduces cardiac contractility. Our findings suggest aficamten holds promise as a therapy for HCM.


Assuntos
Miosinas Cardíacas , Cardiomiopatia Hipertrófica , Contração Miocárdica , Animais , Cardiomiopatia Hipertrófica/tratamento farmacológico , Cardiomiopatia Hipertrófica/metabolismo , Humanos , Contração Miocárdica/efeitos dos fármacos , Miosinas Cardíacas/metabolismo , Miosinas Cardíacas/genética , Modelos Animais de Doenças , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Camundongos , Cristalografia por Raios X , Mutação , Sarcômeros/metabolismo , Sarcômeros/efeitos dos fármacos , Actinas/metabolismo , Modelos Moleculares , Camundongos Transgênicos , Conformação Proteica
9.
Int J Mol Sci ; 25(16)2024 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-39201382

RESUMO

Hypertrophic cardiomyopathy (HCM) is a cardiovascular pathology that is caused by variants in genes encoding sarcomere-associated proteins. However, the clinical significance of numerous variants in HCM-associated genes is still unknown. CRISPR/Cas9 is a tool of nucleotide sequence editing that allows for the unraveling of different biological tasks. In this study, introducing a mutation with CRISPR/Cas9 into induced pluripotent stem cells (iPSCs) of a healthy donor and the directed differentiation of the isogenic iPSC lines into cardiomyocytes were used to assess the pathogenicity of a variant of unknown significance, p.M659I (c.1977G > A) in MYH7, which was found previously in an HCM patient. Using two single-stranded donor oligonucleotides with and without the p.M659I (c.1977G > A) mutation, together with CRISPR/Cas9, an iPSC line heterozygous at the p.M659I (c.1977G > A) variant in MYH7 was generated. No CRISPR/Cas9 off-target activity was observed. The iPSC line with the introduced p.M659I (c.1977G > A) mutation in MYH7 retained its pluripotent state and normal karyotype. Compared to the isogenic control, cardiomyocytes derived from the iPSCs with the introduced p.M659I (c.1977G > A) mutation in MYH7 recapitulated known HCM features: enlarged size, elevated diastolic calcium level, changes in the expression of HCM-related genes, and disrupted energy metabolism. These findings indicate the pathogenicity of the variant.


Assuntos
Sistemas CRISPR-Cas , Miosinas Cardíacas , Cardiomiopatia Hipertrófica , Células-Tronco Pluripotentes Induzidas , Miócitos Cardíacos , Cadeias Pesadas de Miosina , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/citologia , Miosinas Cardíacas/genética , Miosinas Cardíacas/metabolismo , Cadeias Pesadas de Miosina/genética , Cadeias Pesadas de Miosina/metabolismo , Humanos , Cardiomiopatia Hipertrófica/genética , Cardiomiopatia Hipertrófica/patologia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Miócitos Cardíacos/citologia , Diferenciação Celular/genética , Edição de Genes/métodos , Mutação , Linhagem Celular
10.
Biochem Biophys Res Commun ; 733: 150582, 2024 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-39191188

RESUMO

Hypertrophic cardiomyopathy (HCM) is an inherited disease of the heart muscle that is dominated by variations in eight genes encoding sarcomere proteins. Although there are clinical or basic research reports that carrying double mutations can lead to more severe HCM phenotypes, there are also research reports that after reanalyzing the reported mutations, the severity of clinical symptoms in patients with double mutations did not significantly increase compared to patients with only one mutation. To determine whether double pathogenic mutations can aggravate the phenotype of hypertrophic cardiomyopathy in mice, we constructed mice carrying single pathogenic heterozygous mutation Myh6-R453C or Tnnt2-R92W and mice carrying both pathogenic heterozygous mutations. Our results showed that mice with double heterozygous mutations exhibited significant hypertrophic cardiomyopathy phenotypes at 4 weeks of age, and the degree of hypertrophy was significantly higher than that of single heterozygous mutant mice of the same age. Our study suggests that carrying the two pathogenic heterozygous mutations simultaneously can aggravate the phenotype of HCM in mice, which provides experimental evidence for the genotype-phenotype relationship of double pathogenic mutations and provides reference significance for clinical risk stratification of HCM patients.


Assuntos
Cardiomiopatia Hipertrófica , Heterozigoto , Mutação , Cadeias Pesadas de Miosina , Fenótipo , Troponina T , Animais , Cardiomiopatia Hipertrófica/genética , Cardiomiopatia Hipertrófica/patologia , Camundongos , Cadeias Pesadas de Miosina/genética , Troponina T/genética , Camundongos Endogâmicos C57BL , Miosinas Cardíacas/genética , Masculino , Humanos , Modelos Animais de Doenças
11.
Int J Mol Sci ; 25(15)2024 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-39125703

RESUMO

Hypertrophic cardiomyopathy (HCM) is a genetic disease characterized by unexplained left ventricular hypertrophy (LVH), diastolic dysfunction, and increased sudden-death risk. Early detection of the phenotypic expression of the disease in genetic carriers without LVH (Gen+/Phen-) is crucial for emerging therapies. This clinical study aims to identify echocardiographic predictors of phenotypic development in Gen+/Phen-. Sixteen Gen+/Phen- (one subject with troponin T, six with myosin heavy chain-7, and nine with myosin-binding protein C3 mutations), represented the study population. At first and last visit we performed comprehensive 2D speckle-tracking strain echocardiography. During a follow-up of 8 ± 5 years, five carriers developed LVH (LVH+). At baseline, these patients were older than those who did not develop LVH (LVH-) (30 ± 8 vs. 15 ± 8 years, p = 0.005). LVH+ had reduced peak global strain rate during the isovolumic relaxation period (SRIVR) (0.28 ± 0.05 vs. 0.40 ± 0.11 1/s, p = 0.048) and lower global longitudinal strain (GLS) (-19.8 ± 0.4 vs. -22.3 ± 1.1%; p < 0.0001) than LVH- at baseline. SRIVR and GLS were not correlated with age (overall, p > 0.08). This is the first HCM study investigating subjects before they manifest clinically significant or relevant disease burden or symptomatology, comparing at baseline HCM Gen+/Phen- subjects who will develop LVH with those who will not. Furthermore, we identified highly sensitive, easily obtainable, age- and load-independent echocardiographic predictors of phenotype development in HCM gene carriers who may undergo early preventive treatment.


Assuntos
Cardiomiopatia Hipertrófica , Ecocardiografia , Hipertrofia Ventricular Esquerda , Mutação , Humanos , Masculino , Feminino , Ecocardiografia/métodos , Hipertrofia Ventricular Esquerda/genética , Hipertrofia Ventricular Esquerda/diagnóstico por imagem , Hipertrofia Ventricular Esquerda/etiologia , Adulto , Cardiomiopatia Hipertrófica/genética , Cardiomiopatia Hipertrófica/diagnóstico por imagem , Pessoa de Meia-Idade , Adolescente , Cadeias Pesadas de Miosina/genética , Troponina T/genética , Heterozigoto , Proteínas de Transporte/genética , Adulto Jovem , Fenótipo , Miosinas Cardíacas/genética
12.
Int J Mol Sci ; 25(15)2024 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-39125885

RESUMO

Nonsyndromic sporadic thoracic aortic aneurysm (nssTAA) is characterized by diverse genetic variants that may vary in different populations. Our aim was to identify clinically relevant variants in genes implicated in hereditary aneurysms in Russian patients with nssTAA. Forty-one patients with nssTAA without dissection were analyzed. Using massive parallel sequencing, we searched for variants in exons of 53 known disease-causing genes. Patients were found to have no (likely) pathogenic variants in the genes of hereditary TAA. Six variants of uncertain significance (VUSs) were identified in four (9.8%) patients. Three VUSs [FBN1 c.7841C>T (p.Ala2614Val), COL3A1 c.2498A>T (p.Lys833Ile), and MYH11 c.4993C>T (p.Arg1665Cys)] are located in genes with "definitive" disease association (ClinGen). The remaining variants are in "potentially diagnostic" genes or genes with experimental evidence of disease association [NOTCH1 c.964G>A (p.Val322Met), COL4A5 c.953C>G (p.Pro318Arg), and PLOD3 c.833G>A (p.Gly278Asp)]. Russian patients with nssTAA without dissection examined in this study have ≥1 VUSs in six known genes of hereditary TAA (FBN1, COL3A1, MYH11, NOTCH1, COL4A5, or PLOD3). Experimental studies expanded genetic testing, and clinical examination of patients and first/second-degree relatives may shift VUSs to the pathogenic (benign) category or to a new class of rare "predisposing" low-penetrance variants causing the pathology if combined with other risk factors.


Assuntos
Aneurisma da Aorta Torácica , Predisposição Genética para Doença , Humanos , Feminino , Masculino , Federação Russa/epidemiologia , Aneurisma da Aorta Torácica/genética , Pessoa de Meia-Idade , Adulto , Cadeias Pesadas de Miosina/genética , Fibrilina-1/genética , Colágeno Tipo III/genética , Idoso , Miosinas Cardíacas/genética , Sequenciamento de Nucleotídeos em Larga Escala , Mutação , Variação Genética , Adipocinas
13.
Dis Model Mech ; 17(8)2024 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-39189070

RESUMO

Hypertrophic cardiomyopathy (HCM) is an inherited heart muscle disease that is characterised by left ventricular wall thickening, cardiomyocyte disarray and fibrosis, and is associated with arrhythmias, heart failure and sudden death. However, it is unclear to what extent the electrophysiological disturbances that lead to sudden death occur secondary to structural changes in the myocardium or as a result of HCM cardiomyocyte electrophysiology. In this study, we used an induced pluripotent stem cell model of the R403Q variant in myosin heavy chain 7 (MYH7) to study the electrophysiology of HCM cardiomyocytes in electrically coupled syncytia, revealing significant conduction slowing and increased spatial dispersion of repolarisation - both well-established substrates for arrhythmia. Analysis of rhythmonome protein expression in MYH7 R403Q cardiomyocytes showed reduced expression of connexin-43 (also known as GJA1), sodium channels and inward rectifier potassium channels - a three-way hit that reduces electrotonic coupling and slows cardiac conduction. Our data represent a previously unreported, biophysical basis for arrhythmia in HCM that is intrinsic to cardiomyocyte electrophysiology. Later in the progression of the disease, these proarrhythmic phenotypes may be accentuated by myocyte disarray and fibrosis to contribute to sudden death.


Assuntos
Cardiomiopatia Hipertrófica , Conexina 43 , Sistema de Condução Cardíaco , Miócitos Cardíacos , Cadeias Pesadas de Miosina , Conexina 43/metabolismo , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Humanos , Cardiomiopatia Hipertrófica/patologia , Cardiomiopatia Hipertrófica/metabolismo , Cardiomiopatia Hipertrófica/fisiopatologia , Cadeias Pesadas de Miosina/metabolismo , Cadeias Pesadas de Miosina/genética , Sistema de Condução Cardíaco/metabolismo , Sistema de Condução Cardíaco/fisiopatologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Miosinas Cardíacas/metabolismo , Miosinas Cardíacas/genética , Células Gigantes/metabolismo , Células Gigantes/patologia , Arritmias Cardíacas/patologia , Arritmias Cardíacas/metabolismo , Arritmias Cardíacas/fisiopatologia , Potenciais de Ação
15.
Med ; 5(7): 655-659, 2024 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-39002536

RESUMO

A key area of therapeutic progress in obstructive hypertrophic cardiomyopathy revolves around the emergence of cardiac myosin inhibitors, of which mavacamten and aficamten represent the first and second molecules. We summarize the key research evidence, including many similarities and potential differences between various clinical trials studying these molecules.


Assuntos
Miosinas Cardíacas , Cardiomiopatia Hipertrófica , Humanos , Cardiomiopatia Hipertrófica/tratamento farmacológico , Miosinas Cardíacas/metabolismo , Barbitúricos/uso terapêutico , Ureia/análogos & derivados , Ureia/uso terapêutico , Ureia/farmacologia , Uracila/análogos & derivados , Uracila/uso terapêutico , Uracila/farmacologia , Benzilaminas/uso terapêutico , Ensaios Clínicos como Assunto
16.
Stem Cell Res ; 80: 103491, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39047410

RESUMO

Variants in MYH7 cause cardiomyopathies as well as myosin storage myopathy and Laing early-onset distal myopathy (MPD1). MPD1 is characterized by muscle weakness and atrophy usually beginning in the lower legs. Here, we generated iPSC lines from lymphoblastoid cells of three unrelated individuals heterozygous for the most common MPD1-causing variant; p.Lys1617del. iPSC lines showed typical morphology, expressed pluripotency markers, demonstrated trilineage differentiation potential, and had a normal karyotype. These lines represent the first iPSCs derived from MPD1 patients and complement existing MPD1 animal models. They can provide in vitro platforms to better understand and model MPD1 pathomechanisms and test therapies.


Assuntos
Miosinas Cardíacas , Miopatias Distais , Células-Tronco Pluripotentes Induzidas , Cadeias Pesadas de Miosina , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/patologia , Cadeias Pesadas de Miosina/genética , Cadeias Pesadas de Miosina/metabolismo , Miopatias Distais/genética , Miopatias Distais/patologia , Miopatias Distais/metabolismo , Miosinas Cardíacas/genética , Miosinas Cardíacas/metabolismo , Masculino , Feminino , Linhagem Celular , Diferenciação Celular , Adulto
17.
Am J Cardiovasc Drugs ; 24(5): 591-602, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39030459

RESUMO

Hypertrophic cardiomyopathy (HCM) affects as many as 1 in 200 people in the adult population globally. Patients may present with exertional dyspnea, presyncope or syncope, atrial and ventricular arrhythmias, heart failure, and even sudden cardiac death. Current guideline-based therapy involves medical therapy for treatment of symptoms in milder forms of the disease and surgical or catheter-based septal reduction therapies in obstructive HCM. Until recently, there has existed a gap between these two approaches that is now being filled by a new class of drugs, cardiac myosin inhibitors, which directly target the underlying disease process in HCM. Current investigations examine the effects of two cardiac myosin inhibitors on reported symptoms, echocardiographic evidence of disease, and the associated need for septal reduction. This paper reviews the contemporary evidence for the use of cardiac myosin inhibitors in HCM in adults and highlights future directions for this exciting field of cardiovascular medicine.


Assuntos
Miosinas Cardíacas , Cardiomiopatia Hipertrófica , Humanos , Cardiomiopatia Hipertrófica/tratamento farmacológico , Miosinas Cardíacas/antagonistas & inibidores , Adulto , Benzilaminas , Uracila/análogos & derivados
18.
J Gen Physiol ; 156(10)2024 Oct 07.
Artigo em Inglês | MEDLINE | ID: mdl-39083045

RESUMO

Hypertrophic cardiomyopathy (HCM) is a genetic disease of the heart characterized by thickening of the left ventricle (LV), hypercontractility, and impaired relaxation. HCM is caused primarily by heritable mutations in sarcomeric proteins, such as ß myosin heavy chain. Until recently, medications in clinical use for HCM did not directly target the underlying contractile changes in the sarcomere. Here, we investigate a novel small molecule, RLC-1, identified in a bovine cardiac myofibril high-throughput screen. RLC-1 is highly dependent on the presence of a regulatory light chain to bind to cardiac myosin and modulate its ATPase activity. In demembranated rat LV trabeculae, RLC-1 decreased maximal Ca2+-activated force and Ca2+ sensitivity of force, while it increased the submaximal rate constant for tension redevelopment. In myofibrils isolated from rat LV, both maximal and submaximal Ca2+-activated force are reduced by nearly 50%. Additionally, the fast and slow phases of relaxation were approximately twice as fast as DMSO controls, and the duration of the slow phase was shorter. Structurally, x-ray diffraction studies showed that RLC-1 moved myosin heads away from the thick filament backbone and decreased the order of myosin heads, which is different from other myosin inhibitors. In intact trabeculae and isolated cardiomyocytes, RLC-1 treatment resulted in decreased peak twitch magnitude and faster activation and relaxation kinetics. In conclusion, RLC-1 accelerated kinetics and decreased force production in the demembranated tissue, intact tissue, and intact whole cells, resulting in a smaller cardiac twitch, which could improve the underlying contractile changes associated with HCM.


Assuntos
Contração Miocárdica , Animais , Ratos , Contração Miocárdica/efeitos dos fármacos , Contração Miocárdica/fisiologia , Cadeias Leves de Miosina/metabolismo , Bovinos , Miofibrilas/metabolismo , Miosinas Cardíacas/metabolismo , Ratos Sprague-Dawley , Masculino , Cálcio/metabolismo
19.
Forensic Sci Int ; 361: 112121, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38971138

RESUMO

Sudden unexplained death (SUD) is not uncommon in forensic pathology. Yet, diagnosis of SUD remains challenging due to lack of specific biomarkers. This study aimed to screen differentially expressed proteins (DEPs) and validate their usefulness as diagnostic biomarkers for SUD cases. We designed a three-phase investigation, where in the discovery phase, formalin-fixed paraffin-embedded (FFPE) heart specimens were screened through label-free proteomic analysis of cases dying from SUD, mechanical injury and carbon monoxide (CO) intoxication. A total of 26 proteins were identified to be DEPs for the SUD cases after rigorous criterion. Bioinformatics and Adaboost-recursive feature elimination (RFE) analysis further revealed that three of the 26 proteins (MYH6, COX5B and TNNT2) were potential discriminative biomarkers. In the training phase, MYH6 and COX5B were verified to be true DEPs in cardiac tissues from 29 independent SUD cases as compared with a serial of control cases (n = 42). Receiver operating characteristic (ROC) analysis illustrated that combination of MYH6 and COX5B achieved optimal diagnostic sensitivity (89.7 %) and specificity (84.4 %), with area under the curve (AUC) being 0.91. A diagnostic software based on the logistic regression formula derived from the training phase was then constructed. In the validation phase, the diagnostic software was applied to eight authentic SUD cases, seven (87.5 %) of which were accurately recognized. Our study provides a valid strategy towards practical diagnosis of SUD by integrating cardiac MYH6 and COX5B as dual diagnostic biomarkers.


Assuntos
Biomarcadores , Miocárdio , Cadeias Pesadas de Miosina , Proteômica , Adulto , Idoso , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Adulto Jovem , Biomarcadores/metabolismo , Miosinas Cardíacas/genética , Miosinas Cardíacas/metabolismo , Estudos de Casos e Controles , Morte Súbita/etiologia , Patologia Legal/métodos , Miocárdio/metabolismo , Miocárdio/química , Cadeias Pesadas de Miosina/metabolismo , Cadeias Pesadas de Miosina/genética , Curva ROC , Sensibilidade e Especificidade
20.
Am J Physiol Cell Physiol ; 327(3): C634-C645, 2024 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-39010841

RESUMO

Phenotypic changes to endometrial epithelial cells underpin receptivity to embryo implantation at the onset of pregnancy but the effect of hyperglycemia on these processes remains poorly understood. Here, we show that physiological levels of glucose (5 mM) abolished receptivity in the endometrial epithelial cell line, Ishikawa. However, embryo attachment was supported by 17 mM glucose as a result of glucose flux through the hexosamine biosynthetic pathway (HBP) and modulation of cell function via protein O-GlcNAcylation. Pharmacological inhibition of HBP or protein O-GlcNAcylation reduced embryo attachment in cocultures at 17 mM glucose. Mass spectrometry analysis of the O-GlcNAcylated proteome in Ishikawa cells revealed that myosin phosphatase target subunit 1 (MYPT1) is more highly O-GlcNAcylated in 17 mM glucose, correlating with loss of its target protein, phospho-myosin light chain 2, from apical cell junctions of polarized epithelium. Two-dimensional (2-D) and three-dimensional (3-D) morphologic analysis demonstrated that the higher glucose level attenuates epithelial polarity through O-GlcNAcylation. Inhibition of Rho (ras homologous)A-associated kinase (ROCK) or myosin II led to reduced polarity and enhanced receptivity in cells cultured in 5 mM glucose, consistent with data showing that MYPT1 acts downstream of ROCK signaling. These data implicate regulation of endometrial epithelial polarity through RhoA signaling upstream of actomyosin contractility in the acquisition of endometrial receptivity. Glucose levels impinge on this pathway through O-GlcNAcylation of MYPT1, which may impact endometrial receptivity to an implanting embryo in women with diabetes.NEW & NOTEWORTHY Understanding how glucose regulates endometrial function will support preconception guidance and/or the development of targeted interventions for individuals living with diabetes wishing to embark on pregnancy. We found that glucose can influence endometrial epithelial cell receptivity to embryo implantation by regulating posttranslational modification of proteins involved in the maintenance of cell polarity. Impaired or inappropriate endometrial receptivity could contribute to fertility and/or early pregnancy complications caused by poor glucose control.


Assuntos
Citoesqueleto , Implantação do Embrião , Endométrio , Glucose , Fosfatase de Miosina-de-Cadeia-Leve , Feminino , Implantação do Embrião/fisiologia , Humanos , Endométrio/metabolismo , Glucose/metabolismo , Fosfatase de Miosina-de-Cadeia-Leve/metabolismo , Citoesqueleto/metabolismo , Quinases Associadas a rho/metabolismo , Células Epiteliais/metabolismo , Cadeias Leves de Miosina/metabolismo , Animais , Gravidez , Acetilglucosamina/metabolismo , Glicosilação , Polaridade Celular/fisiologia , Hexosaminas/metabolismo , Hexosaminas/biossíntese , Miosinas Cardíacas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...