Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros











Intervalo de ano de publicação
1.
Rev. chil. cardiol ; 43(1): 55-63, abr. 2024. ilus, graf, tab
Artigo em Espanhol | LILACS | ID: biblio-1559644

RESUMO

La miocardiopatía hipertrófica (MCH), definida clínicamente como una hipertrofia ventricular izquierda no explicada por sobrecarga hemodinámica ni por infiltración, es la enfermedad miocárdica hereditaria más común. El tratamiento actual se basa en terapias de reducción septal en casos de Miocardiopatía hipertrófica obstructiva (MCHo), prevención de muerte súbita cardíaca mediante desfibrilador automático implantable y en el uso de fármacos para disminuir la obstrucción dinámica del tracto de salida del ventrículo izquierdo. Recientemente se han desarrollado diversos agentes que actúan sobre los mecanismos que, a nivel celular, llevan a la aparición de hipertrofia ventricular. Este artículo se centra en formas de tratamiento farmacológico desarrolladas en los últimos años, en especial con inhibidores de miosina. Se detallan los mecanismos de acción como también los resultados de diversos estudios clínicos de evaluaciones recientes.


Current clinical management of Hypertrophic Cardiomyopathy is mainly based on modifying physiologic mechanisms to decrease left ventricular outflow tract obstruction. Also important are agents that prevent and treat ventricular arrythmias. This review article is focused on the cellular mechanisms that underlie the development of ventricular hypertrophy and the effect of new myosin inhibitors such as mavcamten and aficamten. Promising results of recent clinical trials evaluating the effect of these agents are analyzed. The potential benefits of genetic therapy are also discussed.


Assuntos
Humanos , Cardiomiopatia Hipertrófica/tratamento farmacológico , Fármacos Cardiovasculares/uso terapêutico , Miosinas Cardíacas/antagonistas & inibidores , Cardiomiopatia Hipertrófica/fisiopatologia , Terapia Genética
2.
J Am Coll Cardiol ; 78(25): 2518-2532, 2021 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-34915982

RESUMO

BACKGROUND: EXPLORER-HCM (Clinical Study to Evaluate Mavacamten [MYK-461] in Adults With Symptomatic Obstructive Hypertrophic Cardiomyopathy) demonstrated that mavacamten, a cardiac myosin inhibitor, improves symptoms, exercise capacity, and left ventricular outflow tract (LVOT) obstruction in patients with obstructive hypertrophic cardiomyopathy (oHCM). OBJECTIVES: The purpose of this study was to evaluate mavacamten's effect on measures of cardiac structure and function and its association with changes in other clinical measures. METHODS: Key echocardiographic parameters from serial echocardiograms over 30 weeks from 251 symptomatic oHCM patients (mavacamten [n = 123], placebo [n = 128]) were assessed in a core laboratory. RESULTS: More patients on mavacamten (80.9%; n = 76 of 94) vs placebo (34.0%; n = 33 of 97) showed complete resolution of mitral valve systolic anterior motion after 30 weeks (difference, 46.8%; P < 0.0001). Mavacamten also improved measures of diastolic function vs placebo, including left atrial volume index (LAVI) (mean ± SD baseline: 40 ± 12 mL/m2 vs 41 ± 14 mL/m2; mean change from baseline of -7.5 mL/m2 [95% CI: -9.0 to -6.1 mL/m2] vs -0.09 mL/m2 [95% CI: -1.6 to 1.5 mL/m2]; P < 0.0001) and lateral E/e' (baseline, 15 ± 6 vs 15 ± 8; change of -3.8 [95% CI: -4.7 to -2.8] vs 0.04 [95% CI: -0.9 to 1.0]; P < 0.0001). Among mavacamten-treated patients, improvement in resting, Valsalva, and post-exercise LVOT gradients, LAVI, and lateral E/e' was associated with reduction in N-terminal pro-B-type natriuretic peptide (P ≤ 0.03 for all). Reduction in LAVI was associated with improved peak exercise oxygen consumption (P = 0.04). CONCLUSIONS: Mavacamten significantly improved measures of left ventricular diastolic function and systolic anterior motion. Improvement in LVOT obstruction, LAVI, and E/e' was associated with reduction in a biomarker of myocardial wall stress (N-terminal pro-B-type natriuretic peptide). These findings demonstrate improvement in important markers of the pathophysiology of oHCM with mavacamten. (Clinical Study to Evaluate Mavacamten [MYK-461] in Adults With Symptomatic Obstructive Hypertrophic Cardiomyopathy; NCT03470545).


Assuntos
Benzilaminas/uso terapêutico , Cardiomiopatia Hipertrófica/tratamento farmacológico , Coração/efeitos dos fármacos , Uracila/análogos & derivados , Idoso , Benzilaminas/farmacologia , Biomarcadores/sangue , Miosinas Cardíacas/antagonistas & inibidores , Cardiomiopatia Hipertrófica/sangue , Cardiomiopatia Hipertrófica/diagnóstico por imagem , Método Duplo-Cego , Ecocardiografia , Tolerância ao Exercício/efeitos dos fármacos , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Uracila/farmacologia , Uracila/uso terapêutico
3.
Mol Med Rep ; 14(5): 4293-4300, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27748856

RESUMO

The role of myosin light chain II (MLC­II) in cellular differentiation of rat mandibular condylar chondrocytes (MCCs) induced by cyclical uniaxial compressive stress (CUCS) remains unclear. In the current study, a four­point bending system was used to apply CUCS to primary cultured MCCs from rats. It was identified that CUCS stimulated features of cellular differentiation including morphological alterations, cytoskeleton rearrangement and overproduction of proteoglycans. Furthermore, CUCS promoted runt­related transcription factor­2 (RUNX2) expression at mRNA (P<0.01) and protein levels (P<0.05) and elevated alkaline phosphatase (ALP) activity (P<0.01), which are both markers of osteogenic differentiation. Under conditions of stress, western blotting indicated that the ratio of phosphorylated MLC­II to total MLC­II was increased significantly (P<0.05). Silencing MLC­II by RNA interference reduced ALP activity (P<0.01), and eliminated RUNX2 mRNA expression (P<0.01). Addition of the MLC kinase inhibitor, ML­7, reduced the CUCS­associated upregulation of RUNX2 expression (P<0.01) and ALP activity (P<0.01). The data indicated that CUCS promoted cellular differentiation of rat primary MCCs, and this was suggested to be via the phosphorylation of MLC­II.


Assuntos
Miosinas Cardíacas/genética , Condrócitos/citologia , Subunidade alfa 1 de Fator de Ligação ao Core/biossíntese , Côndilo Mandibular/crescimento & desenvolvimento , Cadeias Leves de Miosina/genética , Osteogênese/genética , Fosfatase Alcalina/antagonistas & inibidores , Fosfatase Alcalina/genética , Animais , Azepinas/administração & dosagem , Miosinas Cardíacas/antagonistas & inibidores , Miosinas Cardíacas/biossíntese , Diferenciação Celular/genética , Condrócitos/metabolismo , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Citoesqueleto/genética , Citoesqueleto/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Côndilo Mandibular/citologia , Côndilo Mandibular/metabolismo , Cadeias Leves de Miosina/antagonistas & inibidores , Cadeias Leves de Miosina/biossíntese , Naftalenos/administração & dosagem , Fosforilação , Pressão , Interferência de RNA , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Ratos
4.
Peptides ; 81: 29-37, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27020249

RESUMO

Adropin is a peptide encoded by the energy homeostasis associated gene (Enho) and plays a critical role in the regulation of lipid metabolism, insulin sensitivity, and endothelial function. Little is known of the effects of adropin in the brain and whether this peptide modulates ischemia-induced blood-brain barrier (BBB) injury. Here, we used an in vitro BBB model of rat brain microvascular endothelial cells (RBE4) and hypothesized that adropin would reduce endothelial permeability during ischemic conditions. To mimic ischemic conditions in vitro, RBE4 cell monolayers were subjected to 16h hypoxia/low glucose (HLG). This resulted in a significant increase in paracellular permeability to FITC-labeled dextran (40kDa), a dramatic upregulation of vascular endothelial growth factor (VEGF), and the loss of junction proteins occludin and VE-cadherin. Notably, HLG also significantly decreased Enho expression and adropin levels. Treatment of RBE4 cells with synthetic adropin (1, 10 and 100ng/ml) concentration-dependently reduced endothelial permeability after HLG, but this was not mediated through protection to junction proteins or through reduced levels of VEGF. We found that HLG dramatically increased myosin light chain 2 (MLC2) phosphorylation in RBE4 cells, which was significantly reduced by adropin treatment. We also found that HLG significantly increased Rho-associated kinase (ROCK) activity, a critical upstream effector of MLC2 phosphorylation, and that adropin treatment attenuated that effect. These data indicate that treatment with adropin reduces endothelial cell permeability after HLG insult by inhibition of the ROCK-MLC2 signaling pathway. These promising findings suggest that adropin protects against endothelial barrier dysfunction during ischemic conditions.


Assuntos
Barreira Hematoencefálica/efeitos dos fármacos , Isquemia Encefálica/fisiopatologia , Permeabilidade Capilar/efeitos dos fármacos , Miosinas Cardíacas/antagonistas & inibidores , Cadeias Leves de Miosina/antagonistas & inibidores , Quinases Associadas a rho/antagonistas & inibidores , Junções Aderentes/efeitos dos fármacos , Junções Aderentes/metabolismo , Animais , Antígenos CD/metabolismo , Proteínas Sanguíneas/genética , Proteínas Sanguíneas/metabolismo , Barreira Hematoencefálica/metabolismo , Caderinas/metabolismo , Linhagem Celular , Relação Dose-Resposta a Droga , Células Endoteliais/efeitos dos fármacos , Glucose/metabolismo , Ocludina/metabolismo , Peptídeos/genética , Peptídeos/metabolismo , Fosforilação , Ratos , Junções Íntimas/efeitos dos fármacos , Junções Íntimas/metabolismo , Regulação para Cima/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/metabolismo
6.
Science ; 351(6273): 617-21, 2016 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-26912705

RESUMO

Hypertrophic cardiomyopathy (HCM) is an inherited disease of heart muscle that can be caused by mutations in sarcomere proteins. Clinical diagnosis depends on an abnormal thickening of the heart, but the earliest signs of disease are hyperdynamic contraction and impaired relaxation. Whereas some in vitro studies of power generation by mutant and wild-type sarcomere proteins are consistent with mutant sarcomeres exhibiting enhanced contractile power, others are not. We identified a small molecule, MYK-461, that reduces contractility by decreasing the adenosine triphosphatase activity of the cardiac myosin heavy chain. Here we demonstrate that early, chronic administration of MYK-461 suppresses the development of ventricular hypertrophy, cardiomyocyte disarray, and myocardial fibrosis and attenuates hypertrophic and profibrotic gene expression in mice harboring heterozygous human mutations in the myosin heavy chain. These data indicate that hyperdynamic contraction is essential for HCM pathobiology and that inhibitors of sarcomere contraction may be a valuable therapeutic approach for HCM.


Assuntos
Adenosina Trifosfatases/antagonistas & inibidores , Benzilaminas/administração & dosagem , Miosinas Cardíacas/antagonistas & inibidores , Cardiomiopatia Hipertrófica Familiar/tratamento farmacológico , Contração Miocárdica/efeitos dos fármacos , Cadeias Pesadas de Miosina/antagonistas & inibidores , Sarcômeros/efeitos dos fármacos , Uracila/análogos & derivados , Animais , Benzilaminas/química , Miosinas Cardíacas/genética , Cardiomiopatia Hipertrófica Familiar/patologia , Cardiomiopatia Hipertrófica Familiar/fisiopatologia , Células Cultivadas , Modelos Animais de Doenças , Fibrose , Ventrículos do Coração/efeitos dos fármacos , Ventrículos do Coração/patologia , Heterozigoto , Humanos , Masculino , Camundongos , Camundongos Endogâmicos , Mutação , Miocárdio/patologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/patologia , Cadeias Pesadas de Miosina/genética , Ratos , Uracila/administração & dosagem , Uracila/química
7.
Curr Biol ; 23(23): 2434-9, 2013 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-24268417

RESUMO

In development and differentiation, morphological changes often accompany mechanical changes [1], but it is unclear whether or when cells in embryos sense tissue elasticity. The earliest embryo is uniformly pliable, while adult tissues vary widely in mechanics from soft brain and stiff heart to rigid bone [2]. However, cell sensitivity to microenvironment elasticity is debated based in part on results from complex three-dimensional culture models [3]. Regenerative cardiology provides strong motivation to clarify any cell-level sensitivities to tissue elasticity because rigid postinfarct regions limit pumping by the adult heart [4]. Here, we focus on the spontaneously beating embryonic heart and sparsely cultured cardiomyocytes, including cells derived from pluripotent stem cells. Tissue elasticity, Et, increases daily for heart to 1-2 kPa by embryonic day 4 (E4), and although this is ~10-fold softer than adult heart, the beating contractions of E4 cardiomyocytes prove optimal at ~Et,E4 both in vivo and in vitro. Proteomics reveals daily increases in a small subset of proteins, namely collagen plus cardiac-specific excitation-contraction proteins. Rapid softening of the heart's matrix with collagenase or stiffening it with enzymatic crosslinking suppresses beating. Sparsely cultured E4 cardiomyocytes on collagen-coated gels likewise show maximal contraction on matrices with native E4 stiffness, highlighting cell-intrinsic mechanosensitivity. While an optimal elasticity for striation proves consistent with the mathematics of force-driven sarcomere registration, contraction wave speed is linear in Et as theorized for excitation-contraction coupled to matrix elasticity. Pluripotent stem cell-derived cardiomyocytes also prove to be mechanosensitive to matrix and thus generalize the main observation that myosin II organization and contractile function are optimally matched to the load contributed by matrix elasticity.


Assuntos
Proteínas da Matriz Extracelular/biossíntese , Frequência Cardíaca , Coração/embriologia , Contração Miocárdica/fisiologia , Miosinas/biossíntese , Miosinas Cardíacas/antagonistas & inibidores , Diferenciação Celular , Células Cultivadas , Colágeno/biossíntese , Colagenases/farmacologia , Elasticidade , Células-Tronco Embrionárias/metabolismo , Compostos Heterocíclicos de 4 ou mais Anéis/farmacologia , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Miócitos Cardíacos/citologia , Miofibrilas/fisiologia , Sarcômeros/fisiologia
8.
Mol Pharmacol ; 83(2): 454-69, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23188716

RESUMO

The iron-regulated metastasis suppressor, N-myc downstream-regulated gene 1 (NDRG1), is up-regulated by cellular iron depletion mediated by iron chelators and can inhibit cancer cell migration. However, the mechanism of how NDRG1 achieves this effect remains unclear. In this study, we implemented established and newly constructed NDRG1 overexpression and knockdown models using the DU145, HT29, and HCT116 cancer cell lines to investigate the molecular basis by which NDRG1 exerts its inhibitory effect on cell migration. Using these models, we demonstrated that NDRG1 overexpression inhibits cell migration by preventing actin-filament polymerization, stress fiber assembly and formation. In contrast, NDRG1 knockdown had the opposite effect. Moreover, we identified that NDRG1 inhibited an important regulatory pathway mediated by the Rho-associated, coiled-coil containing protein kinase 1 (ROCK1)/phosphorylated myosin light chain 2 (pMLC2) pathway that modulates stress fiber assembly. The phosphorylation of MLC2 is a key process in inducing stress fiber contraction, and this was shown to be markedly decreased or increased by NDRG1 overexpression or knockdown, respectively. The mechanism involved in the inhibition of MLC2 phosphorylation by NDRG1 was mediated by a significant (P < 0.001) decrease in ROCK1 expression that is a key kinase involved in MLC2 phosphorylation. Considering that NDRG1 is up-regulated after cellular iron depletion, novel thiosemicarbazone iron chelators (e.g., di-2-pyridylketone 4,4-dimethyl-3-thiosemicarbazone) were demonstrated to inhibit ROCK1/pMLC2-modulated actin-filament polymerization, stress fiber assembly, and formation via a mechanism involving NDRG1. These results highlight the role of the ROCK1/pMLC2 pathway in the NDRG1-mediated antimetastatic signaling network and the therapeutic potential of iron chelators at inhibiting metastasis.


Assuntos
Miosinas Cardíacas/metabolismo , Proteínas de Ciclo Celular/metabolismo , Movimento Celular/efeitos dos fármacos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Quelantes de Ferro/farmacologia , Cadeias Leves de Miosina/metabolismo , Fibras de Estresse/metabolismo , Quinases Associadas a rho/metabolismo , Actinas/genética , Actinas/metabolismo , Miosinas Cardíacas/antagonistas & inibidores , Miosinas Cardíacas/genética , Proteínas de Ciclo Celular/genética , Linhagem Celular Tumoral , Movimento Celular/genética , Técnicas de Silenciamento de Genes , Células HCT116 , Células HT29 , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Terapia de Alvo Molecular , Cadeias Leves de Miosina/antagonistas & inibidores , Cadeias Leves de Miosina/genética , Metástase Neoplásica , Fosforilação/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Tiossemicarbazonas/farmacologia , Quinases Associadas a rho/antagonistas & inibidores , Quinases Associadas a rho/genética
9.
Cancer Cell ; 19(1): 45-57, 2011 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-21215707

RESUMO

We show that in melanoma cells oncogenic BRAF, acting through MEK and the transcription factor BRN2, downregulates the cGMP-specific phosphodiesterase PDE5A. Although PDE5A downregulation causes a small decrease in proliferation, its major impact is to stimulate a dramatic increase in melanoma cell invasion. This is because PDE5A downregulation leads to an increase in cGMP, which induces an increase in cytosolic Ca(2+), stimulating increased contractility and inducing invasion. PDE5A downregulation also this leads to an increase in short-term and long-term colonization of the lungs by melanoma cells. We do not observe this pathway in NRAS mutant melanoma or BRAF mutant colorectal cells. Thus, we show that in melanoma cells oncogenic BRAF induces invasion through downregulation of PDE5A.


Assuntos
Nucleotídeo Cíclico Fosfodiesterase do Tipo 5/metabolismo , Regulação para Baixo/genética , Regulação Neoplásica da Expressão Gênica/fisiologia , Melanoma/patologia , Proteínas Proto-Oncogênicas B-raf/metabolismo , Animais , Calcimicina/farmacologia , Cálcio/antagonistas & inibidores , Cálcio/metabolismo , Miosinas Cardíacas/antagonistas & inibidores , Miosinas Cardíacas/metabolismo , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Movimento Celular/genética , Proliferação de Células , GMP Cíclico/análogos & derivados , GMP Cíclico/metabolismo , GMP Cíclico/farmacologia , Nucleotídeo Cíclico Fosfodiesterase do Tipo 5/genética , Expressão Gênica/efeitos dos fármacos , Expressão Gênica/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Compostos Heterocíclicos de 4 ou mais Anéis/farmacologia , Proteínas de Homeodomínio/metabolismo , Humanos , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/prevenção & controle , Neoplasias Pulmonares/secundário , Melanoma/metabolismo , Camundongos , Camundongos Nus , Cadeias Leves de Miosina/antagonistas & inibidores , Cadeias Leves de Miosina/metabolismo , Invasividade Neoplásica/patologia , Invasividade Neoplásica/prevenção & controle , Fatores do Domínio POU/metabolismo , Inibidores da Fosfodiesterase 5/farmacologia , Fosforilação/efeitos dos fármacos , Regiões Promotoras Genéticas/genética , Ligação Proteica/genética , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas B-raf/antagonistas & inibidores , Proteínas Proto-Oncogênicas B-raf/genética , RNA Interferente Pequeno/genética , Transplante Heterólogo/patologia
10.
Cell Motil Cytoskeleton ; 65(2): 100-15, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17968985

RESUMO

Phosphorylation of myosin regulatory light chain (RLC) at Ser19 (mono-phosphorylation) promotes filament assembly and enhances actin-activated ATPase activity of non-muscle myosin, while phosphorylation at both Ser19 and Thr18 (di-phosphorylation) further enhances the ATPase activity. However, it has not well been addressed which type of phosphorylation is important in regulating myosin during cytokinesis. Here, we investigated subcellular localization in sea urchin eggs of mono-phosphorylated and di-phosphorylated RLC by both quantitative biochemical and spatiotemporal cytological approaches. Mono-phosphorylated RLC was dominant in the equatorial cortex throughout the whole process of cytokinesis. Inhibition of myosin light chain kinase (MLCK) decreased mono-phosphorylated RLC both in the cortex and in the cleavage furrow, and blocked both formation and contraction of the contractile ring. Two different types of ROCK inhibitor gave inconsistent results: H1152 blocked both RLC mono-phosphorylation in the cleavage furrow and contraction of the contractile ring, while Y27632 affected neither the mono-phosphorylation nor cell division. These results suggest that there may be other targets of H1152 than ROCK, which is involved in the RLC phosphorylation in the cleavage furrow. Furthermore, it was revealed that localization of myosin heavy chain in the cleavage furrow, but not in the cortex, was perturbed by inhibition of RLC mono-phosphorylation. These results suggested that RLC mono-phosphorylation by more than two RLC kinases play a main role in regulation and localization of myosin in the dividing sea urchin eggs.


Assuntos
Miosinas Cardíacas/metabolismo , Fase de Clivagem do Zigoto/citologia , Fase de Clivagem do Zigoto/metabolismo , Cadeias Leves de Miosina/metabolismo , Óvulo/citologia , Óvulo/metabolismo , Adenosina Trifosfatases/metabolismo , Amidas/farmacologia , Compostos de Anilina , Animais , Azepinas/farmacologia , Miosinas Cardíacas/antagonistas & inibidores , Divisão Celular/fisiologia , Células Cultivadas , Clonagem Molecular , Citocinese/fisiologia , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Cadeias Leves de Miosina/antagonistas & inibidores , Quinase de Cadeia Leve de Miosina/metabolismo , Naftalenos/farmacologia , Fosforilação , Piridinas/farmacologia , Ouriços-do-Mar , Serina/metabolismo , Quinases Associadas a rho/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA