Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 3 de 3
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
FASEB J ; 31(6): 2674-2685, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28280005

RESUMO

Type 2 diabetes mellitus (T2DM) represents a rapidly increasing threat to global public health. T2DM arises largely from obesity, poor diet, and lack of exercise, but it also involves genetic predisposition. Here we report that the KCNE2 potassium channel transmembrane regulatory subunit is expressed in human and mouse pancreatic ß cells. Kcne2 deletion in mice impaired glucose tolerance as early as 5 wk of age in pups fed a Western diet, ultimately causing diabetes. In adult mice fed normal chow, skeletal muscle expression of insulin receptor ß and insulin receptor substrate 1 were down-regulated 2-fold by Kcne2 deletion, characteristic of T2DM. Kcne2 deletion also caused extensive pancreatic transcriptome changes consistent with facets of T2DM, including endoplasmic reticulum stress, inflammation, and hyperproliferation. Kcne2 deletion impaired ß-cell insulin secretion in vitro up to 8-fold and diminished ß-cell peak outward K+ current at positive membrane potentials, but also left-shifted its voltage dependence and slowed inactivation. Interestingly, we also observed an aging-dependent reduction in ß-cell outward currents in both Kcne2+/+ and Kcne2-/- mice. Our results demonstrate that KCNE2 is required for normal ß-cell electrical activity and insulin secretion, and that Kcne2 deletion causes T2DM. KCNE2 may regulate multiple K+ channels in ß cells, including the T2DM-linked KCNQ1 potassium channel α subunit.-Lee, S. M., Baik, J., Nguyen, D., Nguyen, V., Liu, S., Hu, Z., Abbott, G. W. Kcne2 deletion impairs insulin secretion and causes type 2 diabetes mellitus.


Assuntos
Diabetes Mellitus Tipo 2/genética , Insulina/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Animais , Fenômenos Eletrofisiológicos , Feminino , Regulação da Expressão Gênica/fisiologia , Humanos , Ilhotas Pancreáticas/metabolismo , Potenciais da Membrana , Camundongos , Camundongos Knockout , Técnicas de Patch-Clamp , Potássio/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Transdução de Sinais , Técnicas de Cultura de Tecidos
2.
J Mol Cell Cardiol ; 87: 148-51, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26307149

RESUMO

Coronary artery disease (CAD) is the leading cause of death worldwide. An estimated half of cases involve genetic predisposition. Sequence variants in human KCNE2, which encodes a cardiac and epithelial K(+) channel ß subunit, cause inherited cardiac arrhythmias. Unexpectedly, human KCNE2 polymorphisms also associate with predisposition to atherosclerosis, with unestablished causality or mechanisms. Here, we report that germline Kcne2 deletion promotes atherosclerosis in mice, overcoming the relative resistance of this species to plaque deposition. In female western diet-fed mice, Kcne2 deletion increased plaque deposition >6-fold and also caused premature ventricular complexes and sudden death. The data establish causality for the first example of ion channel-linked atherosclerosis, and demonstrate that the severity of Kcne2-linked cardiac arrhythmias is strongly diet-dependent.


Assuntos
Arritmias Cardíacas/genética , Aterosclerose/genética , Predisposição Genética para Doença , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Animais , Arritmias Cardíacas/fisiopatologia , Aterosclerose/fisiopatologia , Morte Súbita Cardíaca/patologia , Dieta , Genótipo , Mutação em Linhagem Germinativa , Humanos , Camundongos , Deleção de Sequência
3.
Sci Rep ; 6: 23118, 2016 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-26984260

RESUMO

Nonalcoholic fatty liver disease (NAFLD) is an increasing health problem worldwide, with genetic, epigenetic, and environmental components. Here, we describe the first example of NAFLD caused by genetic disruption of a mammalian potassium channel subunit. Mice with germline deletion of the KCNE2 potassium channel ß subunit exhibited NAFLD as early as postnatal day 7. Using mouse genetics, histology, liver damage assays and transcriptomics we discovered that iron deficiency arising from KCNE2-dependent achlorhydria is a major factor in early-onset NAFLD in Kcne2(─/─) mice, while two other KCNE2-dependent defects did not initiate NAFLD. The findings uncover a novel genetic basis for NAFLD and an unexpected potential factor in human KCNE2-associated cardiovascular pathologies, including atherosclerosis.


Assuntos
Anemia Ferropriva/complicações , Hepatopatia Gordurosa não Alcoólica/etiologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Animais , Proteína C-Reativa/análise , Dieta Hiperlipídica , Feminino , Redes Reguladoras de Genes , Mutação em Linhagem Germinativa , Homocisteína/sangue , Fígado/metabolismo , Fígado/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Hepatopatia Gordurosa não Alcoólica/genética , Hepatopatia Gordurosa não Alcoólica/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana/deficiência , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Deleção de Sequência , Transcriptoma , Triglicerídeos/sangue
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA