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










Base de dados
Intervalo de ano de publicação
1.
Oncotarget ; 9(5): 5716-5735, 2018 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-29464029

RESUMO

The multifunctional cytokine transforming growth factor ß (TGFß) controls homeostasis and disease during embryonic and adult life. TGFß alters epithelial cell differentiation by inducing epithelial-mesenchymal transition (EMT), which involves downregulation of several cell-cell junctional constituents. Little is understood about the mechanism of tight junction disassembly by TGFß. We found that one of the newly identified gene targets of TGFß, encoding the serine/threonine kinase salt-inducible kinase 1 (SIK), controls tight junction dynamics. We provide bioinformatic and biochemical evidence that SIK can potentially phosphorylate the polarity complex protein Par3, an established regulator of tight junction assembly. SIK associates with Par3, and induces degradation of Par3 that can be prevented by proteasomal and lysosomal inhibition or by mutation of Ser885, a putative phosphorylation site on Par3. Functionally, this mechanism impacts on tight junction downregulation. Furthermore, SIK contributes to the loss of epithelial polarity and examination of advanced and invasive human cancers of diverse origin displayed high levels of SIK expression and a corresponding low expression of Par3 protein. High SIK mRNA expression also correlates with lower chance for survival in various carcinomas. In specific human breast cancer samples, aneuploidy of tumor cells best correlated with cytoplasmic SIK distribution, and SIK expression correlated with TGFß/Smad signaling activity and low or undetectable expression of Par3. Our model suggests that SIK can act directly on the polarity protein Par3 to regulate tight junction assembly.

2.
Am J Physiol Regul Integr Comp Physiol ; 314(4): R523-R532, 2018 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-29212808

RESUMO

The preterm diaphragm is functionally immature compared with its term counterpart. In utero inflammation further exacerbates preterm diaphragm dysfunction. We hypothesized that preterm lambs are more vulnerable to in utero inflammation-induced diaphragm dysfunction compared with term lambs. Pregnant ewes received intra-amniotic (IA) injections of saline or 10 mg lipopolysaccharide (LPS) 2 or 7 days before delivery at 121 days (preterm) or ∼145 days (term) of gestation. Diaphragm contractile function was assessed in vitro. Plasma cytokines, diaphragm myosin heavy chain (MHC) isoforms, and oxidative stress were evaluated. Maximum diaphragm force in preterm control lambs was significantly lower (22%) than in term control lambs ( P < 0.001). Despite similar inflammatory cytokine responses to in utero LPS exposure, diaphragm function in preterm and term lambs was affected differentially. In term lambs, maximum force after a 2-day LPS exposure was significantly lower than in controls (by ~20%, P < 0.05). In preterm lambs, maximum forces after 2-day and 7-day LPS exposures were significantly lower than in controls (by ~30%, P < 0.05). Peak twitch force after LPS exposure was significantly lower in preterm than in controls, but not in term lambs. In term lambs, LPS exposure increased the proportion of MHC-I fibers, increased twitch contraction times, and increased fatigue resistance relative to controls. In preterm diaphragm, the cross-sectional area of embryonic MHC fibers was significantly lower after 7-day versus 2-day LPS exposures. We conclude that preterm lambs are more vulnerable to IA LPS-induced diaphragm dysfunction than term lambs. In utero inflammation exacerbates diaphragm dysfunction and may increase susceptibility to postnatal respiratory failure.


Assuntos
Corioamnionite/fisiopatologia , Diafragma/fisiopatologia , Lipopolissacarídeos , Contração Muscular , Força Muscular , Debilidade Muscular/induzido quimicamente , Efeitos Tardios da Exposição Pré-Natal , Animais , Animais Recém-Nascidos , Corioamnionite/sangue , Corioamnionite/induzido quimicamente , Citocinas/sangue , Diafragma/metabolismo , Modelos Animais de Doenças , Feminino , Idade Gestacional , Mediadores da Inflamação/sangue , Debilidade Muscular/sangue , Debilidade Muscular/fisiopatologia , Cadeias Pesadas de Miosina/metabolismo , Estresse Oxidativo , Gravidez , Nascimento Prematuro , Índice de Gravidade de Doença , Carneiro Doméstico
3.
Pediatr Res ; 82(3): 509-517, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28388600

RESUMO

BackgroundPregnant women at a high risk of preterm delivery receive glucocorticoids to accelerate fetal lung maturation and surfactant synthesis. However, the effect of antenatal steroids on the developing diaphragm remains unclear. We hypothesized that maternal betamethasone impairs the fetal diaphragm, and the magnitude of the detrimental effect increases with longer duration of exposure. We aimed to determine how different durations of fetal exposure to maternal betamethasone treatment influence the fetal diaphragm at the functional and molecular levels.MethodsDate-mated merino ewes received intramuscular injections of saline (control) or two doses of betamethasone (5.7 mg) at an interval of 24 h commencing either 2 or 14 days before delivery. Preterm lambs were killed after cesarean delivery at 121-day gestational age. In vitro contractile measurements were performed on the right hemidiaphragm, whereas molecular/cellular analyses used the left costal diaphragm.ResultsDifferent durations of fetal exposure to maternal betamethasone had no consistent effect on the protein metabolic pathway, expression of glucocorticoid receptor and its target genes, cellular oxidative status, or contractile properties of the fetal lamb diaphragm.ConclusionThese data suggest that the potential benefits of betamethasone exposure on preterm respiratory function are not compromised by impaired diaphragm function after low-dose maternal intramuscular glucocorticoid exposure.


Assuntos
Betametasona/administração & dosagem , Diafragma/efeitos dos fármacos , Idade Gestacional , Glucocorticoides/administração & dosagem , Exposição Materna , Ovinos/embriologia , Animais , Western Blotting , Cesárea , Diafragma/metabolismo , Diafragma/fisiologia , Relação Dose-Resposta a Droga , Feminino , Regulação da Expressão Gênica , Complexo Principal de Histocompatibilidade/genética , Contração Muscular/efeitos dos fármacos , Proteólise , RNA/isolamento & purificação , Receptores de Glucocorticoides/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...