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











Base de dados
Intervalo de ano de publicação
1.
FASEB J ; 33(9): 9762-9774, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31162977

RESUMO

The KCNE2 single transmembrane-spanning voltage-gated potassium (Kv) channel ß subunit is ubiquitously expressed and essential for normal function of a variety of cell types, often via regulation of the KCNQ1 Kv channel. A polymorphism upstream of KCNE2 is associated with reduced lung function in human populations, but the pulmonary consequences of KCNE2 gene disruption are unknown. Here, germline deletion of mouse Kcne2 reduced pulmonary expression of potassium channel α subunits Kcnq1 and Kcnb1 but did not alter expression of other Kcne genes. Kcne2 colocalized and coimmunoprecipitated with Kcnq1 in mouse lungs, suggesting the formation of pulmonary Kcnq1-Kcne2 potassium channel complexes. Kcne2 deletion reduced blood O2, increased CO2, increased pulmonary apoptosis, and increased inflammatory mediators TNF-α, IL-6, and leukocytes in bronchoalveolar lavage (BAL) fluids. Consistent with increased pulmonary vascular leakage, Kcne2 deletion increased plasma, BAL albumin, and the BAL:plasma albumin concentration ratio. Kcne2-/- mouse lungs exhibited baseline induction of the reperfusion injury salvage kinase pathway but were less able to respond via this pathway to imposed pulmonary ischemia/reperfusion injury (IRI). We conclude that KCNE2 regulates KCNQ1 in the lungs and is required for normal lung function and resistance to pulmonary IRI. Our data support a causal relationship between KCNE2 gene disruption and lung dysfunction.-Zhou, L., Köhncke, C., Hu, Z., Roepke, T. K., Abbott, G. W. The KCNE2 potassium channel ß subunit is required for normal lung function and resilience to ischemia and reperfusion injury.


Assuntos
Regulação da Expressão Gênica/fisiologia , Lesão Pulmonar/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Traumatismo por Reperfusão/metabolismo , Animais , Citocinas/genética , Citocinas/metabolismo , Feminino , Mutação em Linhagem Germinativa , Inflamação/metabolismo , Canal de Potássio KCNQ1/genética , Canal de Potássio KCNQ1/metabolismo , Camundongos , Camundongos Knockout , Fosforilação , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Traumatismo por Reperfusão/genética , Canais de Potássio Shab/genética , Canais de Potássio Shab/metabolismo
3.
FASEB J ; 30(7): 2476-89, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-26985008

RESUMO

Hyperaldosteronism is associated with an increased prevalence of atrial fibrillation (AF). Mutations in KCNE3 have been associated with AF, and Kcne3(-/-) mice exhibit hyperaldosteronism. In this study, we used recently developed Kcne3(-/-) mice to study atrial electrophysiology with respect to development of aldosterone-dependent AF. In invasive electrophysiology studies, Kcne3(-/-) mice displayed a reduced atrial effective refractory period (AERP) and inducible episodes of paroxysmal AF. The cellular arrhythmogenic correlate for AF predisposition was a significant increase in atrial Kv currents generated by the micromolar 4-aminopyridine-sensitive Kv current encoded by Kv1.5. Electrophysiological alterations in Kcne3(-/-) mice were aldosterone dependent and were associated with increased Rab4, -5, and -9-dependent recycling of Kv1.5 channels to the Z-disc/T-tubulus region and lateral membrane via activation of the Akt/AS160 pathway. Treatment with spironolactone inhibited Akt/AS160 phosphorylation, reduced Rab-dependent Kv1.5 recycling, normalized AERP and atrial Kv currents to the wild-type level, and reduced arrhythmia induction in Kcne3(-/-) mice. Kcne3 deletion in mice predisposes to AF by a heretofore unrecognized mechanism-namely, increased aldosterone-dependent Kv1.5 recycling via Rab GTPases. The findings uncover detailed molecular mechanisms underpinning a channelopathy-linked form of AF and emphasize the inevitability of considering extracardiac mechanisms in genetic arrhythmia syndromes.-Lisewski, U., Koehncke, C., Wilck, N., Buschmeyer, B., Pieske, B., Roepke, T. K. Increased aldosterone-dependent Kv1.5 recycling predisposes to pacing-induced atrial fibrillation in Kcne3(-/-) mice.


Assuntos
Aldosterona/metabolismo , Fibrilação Atrial/etiologia , Canal de Potássio Kv1.5/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Glândulas Suprarrenais/patologia , Animais , Fenômenos Eletrofisiológicos , Proteínas Ativadoras de GTPase/metabolismo , Regulação da Expressão Gênica/fisiologia , Hiperaldosteronismo/genética , Hiperaldosteronismo/metabolismo , Canal de Potássio Kv1.5/genética , Proteínas de Membrana , Camundongos , Camundongos Knockout , Miócitos Cardíacos/metabolismo , Técnicas de Patch-Clamp , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Proteínas Proto-Oncogênicas c-akt , Espironolactona/farmacologia , Proteínas rab de Ligação ao GTP/genética , Proteínas rab de Ligação ao GTP/metabolismo
4.
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
5.
Artigo em Inglês | MEDLINE | ID: mdl-25520848

RESUMO

UNLABELLED: Neuroendocrine tumours (NETs) represent a broad spectrum of tumours, of which the serotonin-producing carcinoid is the most common and has been shown to cause right ventricular heart failure. However, an association between heart failure and NETs other than carcinoid has not been established so far. In this case report, we describe a 51-year-old patient with a glucagon-producing NET of the pancreas who developed acute heart failure and even cardiogenic shock despite therapy. Heart failure eventually regressed after initialising i.v. treatment with the somatostatin analogue octreotide. Chromogranin A as a tumour marker was shown to be significantly elevated, and it decreased with clinical improvement of the patient. The effects of long-time stimulation of glucagon on the myocardium have not been studied yet; however, sarcoplasmic reticulum calcium leak can be discussed as a possible mechanism for glucagon-induced heart failure. LEARNING POINTS: Glucagonoma can be a cause for heart failure.i.v. infusion of octreotide can be successfully used to treat glucagonoma-induced acute heart failure.We suggest that cardiac function should be monitored in all NET patients.

6.
FASEB J ; 25(2): 727-36, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21084694

RESUMO

Targeted deletion of the Kcne2 potassium channel ß subunit gene ablates gastric acid secretion and predisposes to gastric neoplasia in mice. Here, we discovered that Kcne2 deletion basolaterally reroutes the Kcnq1 α subunit in vivo in parietal cells (PCs), in which the normally apical location of the Kcnq1-Kcne2 channel facilitates its essential role in gastric acid secretion. Quantitative RT-PCR and Western blotting revealed that Kcne2 deletion remodeled fundic Kcne3 (2.9±0.8-fold mRNA increase, n=10; 5.3±0.4-fold protein increase, n=7) but not Kcne1, 4, or 5, and resulted in basolateral Kcnq1-Kcne3 complex formation in Kcne2(-/-) PCs. Concomitant targeted deletion of Kcne3 (creating Kcne2(-/-)Kcne3(-/-) mice) restored PC apical Kcnq1 localization without Kcne1, 4, or 5 remodeling (assessed by quantitative RT-PCR; n=5-10), indicating Kcne3 actively, basolaterally rerouted Kcnq1 in Kcne2(-/-) PCs. Despite this, Kcne3 deletion exacerbated gastric hyperplasia in Kcne2(-/-) mice, and both hypochlorhydria and hyperplasia in Kcne2(+/-) mice, suggesting that Kcne3 up-regulation was beneficial in Kcne2-depleted PCs. The findings reveal, in vivo, Kcne-dependent α subunit polarized trafficking and the existence and consequences of potassium channel ß subunit remodeling.


Assuntos
Regulação da Expressão Gênica/fisiologia , Canal de Potássio KCNQ1/metabolismo , Transporte Proteico/fisiologia , Animais , Feminino , Deleção de Genes , Hiperplasia/genética , Hiperplasia/patologia , Canal de Potássio KCNQ1/genética , Masculino , Camundongos , Células Parietais Gástricas/metabolismo , Subunidades Proteicas , Estômago/patologia , Gastropatias/genética , Gastropatias/patologia
7.
PLoS One ; 5(7): e11451, 2010 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-20625512

RESUMO

Gastric cancer is the second leading cause of cancer death worldwide. Predisposing factors include achlorhydria, Helicobacter pylori infection, oxyntic atrophy and TFF2-expressing metaplasia. In parietal cells, apical potassium channels comprising the KCNQ1 alpha subunit and the KCNE2 beta subunit provide a K(+) efflux current to facilitate gastric acid secretion by the apical H(+)K(+)ATPase. Accordingly, genetic deletion of murine Kcnq1 or Kcne2 impairs gastric acid secretion. Other evidence has suggested a role for KCNE2 in human gastric cancer cell proliferation, independent of its role in gastric acidification. Here, we demonstrate that 1-year-old Kcne2(-/-) mice in a pathogen-free environment all exhibit a severe gastric preneoplastic phenotype comprising gastritis cystica profunda, 6-fold increased stomach mass, increased Ki67 and nuclear Cyclin D1 expression, and TFF2- and cytokeratin 7-expressing metaplasia. Some Kcne2(-/-) mice also exhibited pyloric polypoid adenomas extending into the duodenum, and neoplastic invasion of thin walled vessels in the sub-mucosa. Finally, analysis of human gastric cancer tissue indicated reduced parietal cell KCNE2 expression. Together with previous findings, the results suggest KCNE2 disruption as a possible risk factor for gastric neoplasia.


Assuntos
Gastrite/etiologia , Gastrite/genética , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Neoplasias Gástricas/etiologia , Neoplasias Gástricas/genética , Animais , Western Blotting , Linhagem Celular , Linhagem Celular Tumoral , Ciclina D1/genética , Ciclina D1/metabolismo , Imunofluorescência , Mucosa Gástrica/metabolismo , Mucosa Gástrica/patologia , Deleção de Genes , ATPase Trocadora de Hidrogênio-Potássio/genética , ATPase Trocadora de Hidrogênio-Potássio/metabolismo , Humanos , Imuno-Histoquímica , Canal de Potássio KCNQ1/genética , Canal de Potássio KCNQ1/metabolismo , Antígeno Ki-67/genética , Antígeno Ki-67/metabolismo , Metaplasia/genética , Metaplasia/metabolismo , Camundongos , Camundongos Mutantes , Peptídeos/genética , Peptídeos/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Fator Trefoil-2
9.
Nature ; 453(7194): 524-8, 2008 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-18432194

RESUMO

The functional heart is comprised of distinct mesoderm-derived lineages including cardiomyocytes, endothelial cells and vascular smooth muscle cells. Studies in the mouse embryo and the mouse embryonic stem cell differentiation model have provided evidence indicating that these three lineages develop from a common Flk-1(+) (kinase insert domain protein receptor, also known as Kdr) cardiovascular progenitor that represents one of the earliest stages in mesoderm specification to the cardiovascular lineages. To determine whether a comparable progenitor is present during human cardiogenesis, we analysed the development of the cardiovascular lineages in human embryonic stem cell differentiation cultures. Here we show that after induction with combinations of activin A, bone morphogenetic protein 4 (BMP4), basic fibroblast growth factor (bFGF, also known as FGF2), vascular endothelial growth factor (VEGF, also known as VEGFA) and dickkopf homolog 1 (DKK1) in serum-free media, human embryonic-stem-cell-derived embryoid bodies generate a KDR(low)/C-KIT(CD117)(neg) population that displays cardiac, endothelial and vascular smooth muscle potential in vitro and, after transplantation, in vivo. When plated in monolayer cultures, these KDR(low)/C-KIT(neg) cells differentiate to generate populations consisting of greater than 50% contracting cardiomyocytes. Populations derived from the KDR(low)/C-KIT(neg) fraction give rise to colonies that contain all three lineages when plated in methylcellulose cultures. Results from limiting dilution studies and cell-mixing experiments support the interpretation that these colonies are clones, indicating that they develop from a cardiovascular colony-forming cell. Together, these findings identify a human cardiovascular progenitor that defines one of the earliest stages of human cardiac development.


Assuntos
Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Miócitos Cardíacos/citologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Ativinas/farmacologia , Proteína Morfogenética Óssea 4 , Proteínas Morfogenéticas Ósseas/farmacologia , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Linhagem da Célula/efeitos dos fármacos , Células-Tronco Embrionárias/efeitos dos fármacos , Células-Tronco Embrionárias/transplante , Fator 2 de Crescimento de Fibroblastos/farmacologia , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Técnicas de Patch-Clamp , Proteínas Proto-Oncogênicas c-kit/genética , Fator A de Crescimento do Endotélio Vascular/farmacologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/deficiência , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética
10.
Cardiovasc Res ; 77(1): 98-106, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18006462

RESUMO

AIMS: Auditory stimulus-induced long QT syndrome (LQTS) is almost exclusively linked to mutations in the hERG potassium channel, which generates the I Kr ventricular repolarization current. Here, a young woman with prior episodes of auditory stimulus-induced syncope presented with LQTS and ventricular fibrillation (VF) with hypomagnesaemia and hypocalcaemia after completing a marathon, followed by subsequent VF with hypokalaemia. The patient was found to harbour a KCNE2 gene mutation encoding a T10M amino acid substitution in MiRP1, an ancillary subunit that co-assembles with and functionally modulates hERG. Other family members with the mutation were asymptomatic, and the proband had no mutations in hERG or other LQTS-linked cardiac ion channel genes. The T10M mutation was absent from 578 unrelated, ethnically matched control chromosomes analysed here and was previously described only once-in an LQTS patient-but not functionally characterized. METHODS AND RESULTS: T10M-MiRP1-hERG currents were assessed using whole-cell voltage clamp of transfected Chinese Hamster ovary cells. T10M-MiRP1-hERG channels showed

Assuntos
Estimulação Acústica , Mutação , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Fibrilação Ventricular/genética , Desequilíbrio Hidroeletrolítico/complicações , Adulto , Animais , Células CHO , Cálcio/sangue , Cricetinae , Cricetulus , Canais de Potássio Éter-A-Go-Go/fisiologia , Feminino , Humanos , Canal de Potássio KCNQ1/fisiologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/fisiologia , Fibrilação Ventricular/etiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA