Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Appl Immunohistochem Mol Morphol ; 30(2): e11-e15, 2022 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-34711739

RESUMO

Dysregulation of epigenetic mechanisms, reflected by loss of expression of 5-hydroxymethylcytosine (5-hmC) is being increasingly recognized as a marker of aggressive behavior in several neoplasms; however, the role of such epigenetic modifiers in pancreatic neuroendocrine tumors (PanNETs) has not been studied. Annotated cohort of 60 PanNETs was evaluated for 5-hmC expression using immunohistochemistry. Univariable and multivariable analyses were performed. To determine intratumor heterogeneity of 5-hmC expression, 26 additional synchronous metastatic deposits of PanNETs from 8 patients were evaluated for 5-hmC expression. 5-hmC level showed significant association with the presence of distant metastases (P=0.02), female sex (P=0.04), and Ki-67 proliferation index (P=0.002). A multivariate model created using the stepwise logistic regression analysis showed the presence of nodal metastases (odds ratio=6.15), lymphovascular invasion (odds ratio=4.07) and lack of 5-hmC expression (odds ratio=5.34) were predictive of the risk of distant metastasis in PanNETs with a c-statistic of 0.845. Epigenetic intratumoral heterogeneity of 5-hmC expression was seen in 37.5% cases (3/8). Our work provides evidence that epigenetic regulators are involved in the pathobiology of PanNETs and immunohistochemical analysis of 5-hmC may be able to refine prognostic evaluation of these tumors.


Assuntos
Tumores Neuroendócrinos , Neoplasias Pancreáticas , 5-Metilcitosina/análogos & derivados , Epigênese Genética , Feminino , Humanos , Índice Mitótico , Tumores Neuroendócrinos/metabolismo , Neoplasias Pancreáticas/metabolismo
3.
J Biol Chem ; 288(6): 3823-30, 2013 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-23269672

RESUMO

The Src-homology 2 (SH2) domain-containing tyrosine phosphatase Shp2 has been known to regulate various signaling pathways triggered by receptor and cytoplasmic tyrosine kinases. Here we describe a novel function of Shp2 in control of lipid metabolism by mediating degradation of fatty acid synthase (FASN). p38-phosphorylated COP1 accumulates in the cytoplasm and subsequently binds FASN through Shp2 here as an adapter, leading to FASN-Shp2-COP1 complex formation and FASN degradation mediated by ubiquitination pathway. By fasting p38 is activated and stimulates FASN protein degradation in mice. Consistently, the FASN protein levels are dramatically elevated in mouse liver and pancreas in which Shp2/Ptpn11 is selectively deleted. Thus, this study identifies a new activity for Shp2 in lipid metabolism.


Assuntos
Ácido Graxo Sintase Tipo I/metabolismo , Proteínas Nucleares/metabolismo , Proteína Tirosina Fosfatase não Receptora Tipo 11/metabolismo , Proteólise , Ubiquitina-Proteína Ligases/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Células 3T3 , Animais , Ácido Graxo Sintase Tipo I/genética , Células HeLa , Humanos , Metabolismo dos Lipídeos/fisiologia , Fígado/enzimologia , Camundongos , Camundongos Knockout , Proteínas Nucleares/genética , Pâncreas/enzimologia , Proteína Tirosina Fosfatase não Receptora Tipo 11/genética , Ubiquitina-Proteína Ligases/genética , Proteínas Quinases p38 Ativadas por Mitógeno/genética
4.
Mol Cell Biol ; 32(10): 1867-78, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22431513

RESUMO

In mammals, leptin regulates food intake and energy balance mainly through the activation of LepRb in the hypothalamus, and estrogen has a leptin-like effect in the hypothalamic control of metabolism. However, it remains to be elucidated how estrogen signaling is intertwined with the leptin pathway. We show here that Shp2, a nonreceptor tyrosine phosphatase, acts to integrate leptin and estrogen signals. The expression of a dominant-active mutant (Shp2(D61A)) in forebrain neurons conferred female, but not male, transgenic mice resistance to high-fat diet (HFD)-induced obesity and liver steatosis, accompanied by improved insulin sensitivity and glucose homeostasis. Fed with either HFD or regular chow food, Shp2(D61A) female mice showed dramatically enhanced leptin sensitivity. Microinjection of Shp2(D61A)-expressing adeno-associated virus into mediobasal hypothalamus elicited a similar antiobese effect in female mice. Biochemical analyses showed a physical association of Shp2 with estrogen receptor alpha, which is necessary for the synergistic and persistent activation of Erk by leptin and estrogen. Together, these results elucidate a mechanism for the direct cross talk of leptin and estrogen signaling and offer one explanation for the propensity of postmenopausal women to develop obesity.


Assuntos
Peso Corporal/genética , Metabolismo Energético/genética , Estrogênios/metabolismo , Leptina/metabolismo , Proteína Tirosina Fosfatase não Receptora Tipo 11/genética , Animais , Dieta Hiperlipídica , Receptor alfa de Estrogênio/metabolismo , Feminino , Hipotálamo/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Mutação , Neurônios/metabolismo , Obesidade/genética , Obesidade/metabolismo , Proteína Tirosina Fosfatase não Receptora Tipo 11/metabolismo , Receptores para Leptina/metabolismo , Fatores Sexuais , Transdução de Sinais
5.
Cancer Cell ; 19(5): 629-39, 2011 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-21575863

RESUMO

The human gene Ptpn11, which encodes the tyrosine phosphatase Shp2, may act as a proto-oncogene because dominantly activating mutations have been detected in several types of leukemia. Herein we report a tumor-suppressor function of Shp2. Hepatocyte-specific deletion of Shp2 promotes inflammatory signaling through the Stat3 pathway and hepatic inflammation/necrosis, resulting in regenerative hyperplasia and development of tumors in aged mice. Furthermore, Shp2 ablation dramatically enhanced diethylnitrosamine (DEN)-induced hepatocellular carcinoma (HCC) development, which was abolished by concurrent deletion of Shp2 and Stat3 in hepatocytes. Decreased Shp2 expression was detected in a subfraction of human HCC specimens. Thus, in contrast to the leukemogenic effect of dominant-active mutants, Ptpn11/Shp2 has a tumor-suppressor function in liver.


Assuntos
Adenoma de Células Hepáticas/enzimologia , Carcinoma Hepatocelular/enzimologia , Neoplasias Hepáticas/enzimologia , Fígado/enzimologia , Proteína Tirosina Fosfatase não Receptora Tipo 11/análise , Proteína Tirosina Fosfatase não Receptora Tipo 11/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Adenoma de Células Hepáticas/genética , Adenoma de Células Hepáticas/patologia , Animais , Carcinoma Hepatocelular/induzido quimicamente , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patologia , Carcinoma Hepatocelular/prevenção & controle , Citocinas/sangue , Citocinas/genética , Dietilnitrosamina , Regulação da Expressão Gênica , Hepatite/enzimologia , Hepatite/genética , Hepatite/patologia , Humanos , Hiperplasia , Mediadores da Inflamação/sangue , Interleucina-6/administração & dosagem , Lipopolissacarídeos/administração & dosagem , Fígado/efeitos dos fármacos , Fígado/patologia , Neoplasias Hepáticas/induzido quimicamente , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patologia , Neoplasias Hepáticas/prevenção & controle , Regeneração Hepática , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Necrose , Proteína Tirosina Fosfatase não Receptora Tipo 11/deficiência , Proteína Tirosina Fosfatase não Receptora Tipo 11/genética , Proto-Oncogene Mas , Fator de Transcrição STAT3/deficiência , Fator de Transcrição STAT3/genética , Transdução de Sinais , Fatores de Tempo , Proteínas Supressoras de Tumor/deficiência , Proteínas Supressoras de Tumor/genética
6.
Proc Natl Acad Sci U S A ; 106(18): 7531-6, 2009 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-19380737

RESUMO

Intracellular signaling by which pancreatic beta-cells synthesize and secrete insulin in control of glucose homeostasis is not fully understood. Here we show that Shp2, a cytoplasmic tyrosine phosphatase possessing 2 SH2 domains, coordinates signaling events required for insulin biosynthesis in beta-cells. Mice with conditional ablation of the Shp2/Ptpn11 gene in the pancreas exhibited defective glucose-stimulated insulin secretion and impaired glucose tolerance. Consistently, siRNA-mediated Shp2-knockdown in rat insulinoma INS-1 832/13 cells resulted in decreased insulin production and secretion despite an increase in cellular ATP. Shp2 modulates the strength of signals flowing through Akt/FoxO1 and Erk pathways, culminating in control of Pdx1 expression and activity on Ins1 and Ins2 promoters, and forced Pdx1 expression rescued insulin production in Shp2-knockdown beta-cells. Therefore, Shp2 acts as a signal coordinator in beta-cells, orchestrating multiple pathways controlling insulin biosynthesis to maintain glucose homeostasis.


Assuntos
Células Secretoras de Insulina/enzimologia , Insulina/biossíntese , Proteína Tirosina Fosfatase não Receptora Tipo 11/metabolismo , Animais , Linhagem Celular , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Proteína Forkhead Box O1 , Fatores de Transcrição Forkhead/metabolismo , Glucose/metabolismo , Glucose/farmacologia , Proteínas de Homeodomínio/biossíntese , Proteínas de Homeodomínio/genética , Insulina/metabolismo , Secreção de Insulina , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/metabolismo , Camundongos , Camundongos Knockout , Proteína Tirosina Fosfatase não Receptora Tipo 11/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transativadores/biossíntese , Transativadores/genética
7.
EMBO J ; 28(1): 21-33, 2009 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-19057511

RESUMO

The PTEN tumour suppressor gene is induced by the early growth response 1 (EGR1) transcription factor, which also transactivates p53, p73, and p300/CBP as well as other proapoptotic and anti-cancer genes. Here, we describe a novel Akt-EGR1-alternate reading frame (ARF)-PTEN axis, in which PTEN activation in vivo requires p14ARF-mediated sumoylation of EGR1. This modification is dependent on the phosphorylation of EGR1 at S350 and T309 by Akt, which promotes interaction of EGR1 with ARF at K272 in its repressor domain by the ARF/Ubc9/SUMO system. EGR1 sumoylation is decreased by ARF reduction, and no EGR1 sumoylation is detected in ARF(-/-) mice, which also exhibit reduced amounts of PTEN. Our model predicts that perturbation of any of the clinically important tumour suppressors, PTEN, EGR1, and ARF, will cause some degree of dysfunction of the others. These results also explain the known negative feedback regulation by PTEN on its own synthesis through PI3 kinase inhibition.


Assuntos
Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Proteína 1 de Resposta de Crescimento Precoce/metabolismo , Regulação da Expressão Gênica , Proteína Oncogênica v-akt/metabolismo , PTEN Fosfo-Hidrolase/biossíntese , Animais , Linhagem Celular , Humanos , Camundongos , Camundongos Knockout , Modelos Biológicos , Fosforilação
8.
Mol Cell Biol ; 29(2): 378-88, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19001090

RESUMO

The intracellular signaling mechanisms underlying the pathogenesis of cardiac diseases are not fully understood. We report here that selective deletion of Shp2, an SH2-containing cytoplasmic tyrosine phosphatase, in striated muscle results in severe dilated cardiomyopathy in mice, leading to heart failure and premature mortality. Development of cardiomyopathy in this mouse model is coupled with insulin resistance, glucose intolerance, and impaired glucose uptake in striated muscle cells. Shp2 deficiency leads to upregulation of leukemia inhibitory factor-stimulated phosphatidylinositol 3-kinase/Akt, Erk5, and Stat3 pathways in cardiomyocytes. Insulin resistance and impaired glucose uptake in Shp2-deficient mice are at least in part due to impaired protein kinase C-zeta/lambda and AMP-kinase activities in striated muscle. Thus, we have generated a mouse line modeling human patients suffering from cardiomyopathy and insulin resistance. This study reinforces a concept that a compound disease with multiple cardiovascular and metabolic disturbances can be caused by a defect in a single molecule such as Shp2, which modulates multiple signaling pathways initiated by cytokines and hormones.


Assuntos
Cardiomiopatia Dilatada/metabolismo , Resistência à Insulina , Músculo Esquelético/metabolismo , Miocárdio/metabolismo , Animais , Cardiomiopatia Dilatada/genética , Cardiomiopatia Dilatada/patologia , Deleção de Genes , Glucose/metabolismo , Intolerância à Glucose/genética , Intolerância à Glucose/metabolismo , Coração/fisiopatologia , Resistência à Insulina/genética , Estimativa de Kaplan-Meier , Fator Inibidor de Leucemia/metabolismo , MAP Quinase Quinase Quinases/metabolismo , Camundongos , Camundongos Knockout , Proteína Quinase 7 Ativada por Mitógeno/metabolismo , Miocárdio/patologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteína Tirosina Fosfatase não Receptora Tipo 11/deficiência , Proteína Tirosina Fosfatase não Receptora Tipo 11/genética , Proteína Tirosina Fosfatase não Receptora Tipo 11/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...