Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Physiol Rep ; 11(10): e15696, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-37226398

RESUMO

The voltage-gated Nav 1.5 channels mediate the fast Na+ current (INa ) in cardiomyocytes initiating action potentials and cardiac contraction. Downregulation of INa , as occurs in Brugada syndrome (BrS), causes ventricular arrhythmias. The present study investigated whether the Wnt/ß-catenin signaling regulates Nav 1.5 in human-induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs). In healthy male and female iPSC-CMs, activation of Wnt/ß-catenin signaling by CHIR-99021 reduced (p < 0.01) both Nav 1.5 protein and SCN5A mRNA. In iPSC-CMs from a BrS patient, both Nav 1.5 protein and peak INa were reduced compared to those in healthy iPSC-CMs. Treatment of BrS iPSC-CMs with Wnt-C59, a small-molecule Wnt inhibitor, led to a 2.1-fold increase in Nav 1.5 protein (p = 0.0005) but surprisingly did not affect SCN5A mRNA (p = 0.146). Similarly, inhibition of Wnt signaling using shRNA-mediated ß-catenin knockdown in BrS iPSC-CMs led to a 4.0-fold increase in Nav 1.5, which was associated with a 4.9-fold increase in peak INa but only a 2.1-fold increase in SCN5A mRNA. The upregulation of Nav 1.5 by ß-catenin knockdown was verified in iPSC-CMs from a second BrS patient. This study demonstrated that Wnt/ß-catenin signaling inhibits Nav 1.5 expression in both male and female human iPSC-CMs, and inhibition of Wnt/ß-catenin signaling upregulates Nav 1.5 in BrS iPSC-CMs through both transcriptional and posttranscriptional mechanisms.


Assuntos
Síndrome de Brugada , Células-Tronco Pluripotentes Induzidas , Canal de Sódio Disparado por Voltagem NAV1.5 , Via de Sinalização Wnt , Feminino , Humanos , Masculino , beta Catenina , Miócitos Cardíacos , Canal de Sódio Disparado por Voltagem NAV1.5/genética
2.
Sci Rep ; 10(1): 11274, 2020 07 09.
Artigo em Inglês | MEDLINE | ID: mdl-32647145

RESUMO

3D printing was used to develop an open access device capable of simultaneous electrical and mechanical stimulation of human induced pluripotent stem cells in 6-well plates. The device was designed using Computer-Aided Design (CAD) and 3D printed with autoclavable, FDA-approved materials. The compact design of the device and materials selection allows for its use inside cell incubators working at high humidity without the risk of overheating or corrosion. Mechanical stimulation of cells was carried out through the cyclic deflection of flexible, translucent silicone membranes by means of a vacuum-controlled, open-access device. A rhythmic stimulation cycle was programmed to create a more physiologically relevant in vitro model. This mechanical stimulation was coupled and synchronized with in situ electrical stimuli. We assessed the capabilities of our device to support cardiac myocytes derived from human induced pluripotent stem cells, confirming that cells cultured under electromechanical stimulation presented a defined/mature cardiomyocyte phenotype. This 3D printed device provides a unique high-throughput in vitro system that combines both mechanical and electrical stimulation, and as such, we foresee it finding applications in the study of any electrically responsive tissue such as muscles and nerves.


Assuntos
Desenho Assistido por Computador , Estimulação Elétrica/instrumentação , Células-Tronco Pluripotentes Induzidas/citologia , Impressão Tridimensional , Elasticidade , Humanos , Miócitos Cardíacos/citologia , Fenótipo , Silicones/química , Software , Estresse Mecânico , Pesquisa Translacional Biomédica
3.
Can J Cardiol ; 36(4): 564-576, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32046907

RESUMO

BACKGROUND: Myocardial infarction and heart failure are associated with reduced voltage-gated Na+ current (INa) that promotes arrhythmias and sudden deaths. We have previously shown that the Wnt/ß-catenin signalling (Wnt signalling), which is active in heart disease, reduces cardiac INa, suggesting that Wnt signalling may be a potential therapeutic target. However, because Wnt signalling is required for the homeostasis of many noncardiac tissues, administration of Wnt inhibitors to heart patients would cause significant side effects. The present study aims to elucidate the molecular mechanisms of cardiac INa inhibition by Wnt, which would identify cardiac-specific therapeutic targets. METHODS: Wnt signalling was activated in neonatal rat ventricular myocytes by Wnt3a protein. Adenovirus expressing Wnt3a was injected into the adult rat ventricle. CRISPR/Cas9 and chromatin immunoprecipitation were used for mechanistic studies. RESULTS: Wnt signalling activation in neonatal rat ventricular myocytes reduced Nav1.5 protein and Scn5a mRNA, but increased Tbx3, a known suppressor of Scn5a. Chromatin immunoprecipitation showed that Wnt signalling inhibits Scn5a expression through downstream mediator (TCF4) binding to both Tbx3 and Scn5a promoters. Overexpression or knockdown of Tbx3 directly modified Nav1.5 and INa, whereas CRISPR/Cas9-induced mutations at TCF4 binding sites within the Scn5a promoter attenuated Wnt inhibition of Scn5a and Nav1.5. In adult rat hearts, adenovirus expressing Wnt3a reduced Nav1.5, increased QRS duration in electrocardiogram, and increased the susceptibility to ventricular tachycardia. CONCLUSIONS: Wnt signalling inhibits the Na+ channel by direct and indirect (via Tbx3) suppression of Scn5a transcription. Strategies to block TCF4 binding to the Tbx3 and Scn5a promoters would represent novel strategies for cardiac-specific inhibition of the Wnt pathway to rescue INa and prevent sudden cardiac deaths.


Assuntos
Miócitos Cardíacos/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.5/genética , Canais de Sódio/fisiologia , Via de Sinalização Wnt , Animais , Regulação da Expressão Gênica , Ventrículos do Coração/citologia , Ratos
4.
Pflugers Arch ; 471(11-12): 1551-1564, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31713764

RESUMO

Hydrogen sulfide (H2S) is endogenously produced in pancreatic ß cells and its level is elevated in diabetes. Here, we report that H2S affects insulin secretion via two mechanisms that converge on cytosolic free Ca2+ ([Ca2+]i), a key mediator of insulin exocytosis. Cellular calcium imaging, using Fura-2 or Fluo-4, showed that exposure of INS-1E cells to H2S (30-100 µM) reduced both [Ca2+]i levels (by 21.7 ± 2.3%) and oscillation frequency (p < 0.01, n = 4). Consistent with a role of plasma membrane KATP channels (plasma-KATP), the effects of H2S on [Ca2+]i were blocked by gliclazide (a blocker of plasma-KATP channels), but were mimicked by diazoxide (an activator of plasma-KATP channels). Surprisingly, when Ca2+ entry via plasma membrane was inhibited using Ca2+-free external solutions, H2S increased [Ca2+]i by 39.7 ± 3.6% suggesting Ca2+ release from intracellular stores. H2S-induced [Ca2+]i increases were abolished by either FCCP (which depletes Ca2+ stored in mitochondria) or cyclosporine A (an inhibitor of mitochondrial permeability transition pore, mPTP) suggesting that H2S induces Ca2+ release from mitochondria. Measurement of mitochondrial membrane potential (MMP) suggested that H2S causes MMP depolarization, which was blocked by cyclosporine A. Finally, insulin measurements by ELISA indicated that H2S decreased insulin release from INS-1E cells, but after plasma membrane Ca2+ entry was blocked by nifedipine, H2S-induced mitochondrial Ca2+ release is able to increase insulin release. Together, our results indicate that H2S has dual effects on insulin release suggesting that, with different metabolic conditions, H2S may differentially modulate the insulin release from pancreatic ß cells and play a role in ß cell dysfunction.


Assuntos
Cálcio/metabolismo , Citosol/metabolismo , Homeostase/fisiologia , Sulfeto de Hidrogênio/metabolismo , Secreção de Insulina/fisiologia , Células Secretoras de Insulina/metabolismo , Canais KATP/metabolismo , Proteínas de Transporte da Membrana Mitocondrial/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Sinalização do Cálcio/fisiologia , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Exocitose/fisiologia , Insulina/metabolismo , Potencial da Membrana Mitocondrial/fisiologia , Mitocôndrias/metabolismo , Poro de Transição de Permeabilidade Mitocondrial , Ratos
5.
Chem Commun (Camb) ; 53(57): 8050-8053, 2017 Jul 13.
Artigo em Inglês | MEDLINE | ID: mdl-28671697

RESUMO

A metal-free iodine-catalyzed intramolecular amination has been developed for the practical synthesis of pyrrolo[2,3-b]indoles from readily available tryptophan esters. The transformation has been applied to a wide array of substrates and can be performed on gram scale under very mild conditions.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...