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.
Diabetes Res Clin Pract ; 140: 107-117, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29601913

RESUMO

AIMS: We evaluated specific alterations in amino acids (AAs) profile in patients with type 2 diabetes mellitus (T2DM) and impaired fasting glucose (IFG) compared with healthy controls. In addition, we tried to find the mechanisms behind these AA alterations. METHODS: Twenty AAs, TNF-α, and IL-6 were analyzed in fasting serum samples from a total of 198 individuals (56 drug-naïve patients with T2DM, 69 patients IFG, and 73 healthy controls). The C2C12 mouse myoblast cell lines were used to examine the changes of MAFbx and MuRF1 expressions, which are muscle specific E3 ligases acting as major mediators of skeletal muscle proteolysis, after development of insulin resistance induced by palmitate treatment. RESULTS: In addition to branched chain amino acids BCAAs, fasting serum AAs such as glutamic acid, lysine, phenylalanine, arginine, alanine, tyrosine, aspartic acid, were higher in patients with T2DM and intermediately elevated in patients with IFG compared with normoglycemic controls. These serum AA concentrations positively correlated with fasting glucose, homeostasis model assessment of insulin resistance (HOMA-IR), and pro-inflammatory cytokines. In addition, HOMA-IR and pro-inflammatory cytokines were two important independent predictors of serum AA levels. In vitro experiments showed that palmitate treatment in C2C12 myotubes induced insulin resistance, increased pro-inflammatory cytokine gene expression, and increased MAFbx gene and protein expression. CONCLUSIONS: The increase in fasting serum AAs can be an early manifestation of insulin resistance. Increased muscle proteolysis induced by insulin resistance and inflammatory cytokines can be a possible mechanism for the rise in serum AA levels.


Assuntos
Aminoácidos de Cadeia Ramificada/sangue , Citocinas/sangue , Diabetes Mellitus Tipo 2/sangue , Resistência à Insulina , Adulto , Jejum , Feminino , Glucose , Humanos , Masculino , Pessoa de Meia-Idade , Estado Pré-Diabético/complicações , Adulto Jovem
2.
Oncotarget ; 7(31): 49902-49916, 2016 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-27363019

RESUMO

Wee1 is a member of the Serine/Threonine protein kinase family and is a key regulator of cell cycle progression. It has been known that WEE1 is highly expressed and has oncogenic functions in various cancers, but it is not yet studied in gastric cancers. In this study, we investigated the oncogenic role and therapeutic potency of targeting WEE1 in gastric cancer. At first, higher expression levels of WEE1 with lower survival probability were determined in stage 4 gastric cancer patients or male patients with accompanied lymph node metastasis. To determine the function of WEE1 in gastric cancer cells, we determined that WEE1 ablation decreased the proliferation, migration, and invasion, while overexpression of WEE1 increased these effects in gastric cancer cells. We also validated the clinical application of WEE1 targeting by a small molecule, AZD1775 (MK-1775), which is a WEE1 specific inhibitor undergoing clinical trials. AZD1775 significantly inhibited cell proliferation and induced apoptosis and cell cycle arrest in gastric cancer cells, which was more effective in WEE1 high-expressing gastric cancer cells. Moreover, we performed combination treatments with AZD1775 and anti-cancer agents, 5- fluorouracil or Paclitaxel in gastric cancer cells and in gastric cancer orthotopic-transplanted mice to maximize the therapeutic effect and safety of AZD1775. The combination treatments dramatically inhibited the proliferation of gastric cancer cells and tumor burdens in stomach orthotopic-transplanted mice. Taken together, we propose that WEE1 is over-expressed and could enhance gastric cancer cell proliferation and metastasis. Therefore, we suggest that WEE1 is a potent target for gastric cancer therapy.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Terapia de Alvo Molecular , Proteínas Nucleares/metabolismo , Proteínas Tirosina Quinases/metabolismo , Neoplasias Gástricas/genética , Neoplasias Gástricas/terapia , Animais , Antineoplásicos/uso terapêutico , Apoptose , Ciclo Celular , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Feminino , Fluoruracila/uso terapêutico , Humanos , Estimativa de Kaplan-Meier , Metástase Linfática , Masculino , Camundongos , Camundongos Nus , Invasividade Neoplásica , Transplante de Neoplasias , Paclitaxel/uso terapêutico , Fenótipo , Prognóstico , Pirazóis/uso terapêutico , Pirimidinas/uso terapêutico , Pirimidinonas
3.
J Biol Chem ; 290(15): 9863-73, 2015 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-25691573

RESUMO

Autophagy is a conserved process that contributes to cell homeostasis. It is well known that induction mainly occurs in response to nutrient starvation, such as starvation of amino acids and insulin, and its mechanisms have been extensively characterized. However, the mechanisms behind cellular glucose deprivation-induced autophagy are as of now poorly understood. In the present study, we determined a mechanism by which glucose deprivation induced the PKC-dependent proteasomal degradation of ß-catenin, leading to autophagy. Glucose deprivation was shown to cause a sub-G1 transition and enhancement of the LC3-II protein levels, whereas ß-catenin protein underwent degradation in a proteasome-dependent manner. Moreover, the inhibition of GSK3ß was unable to abolish the glucose deprivation-mediated ß-catenin degradation or up-regulation of LC3-II protein levels, which suggested GSK3ß-independent protein degradation. Intriguingly, the inhibition of PKCα using a pharmacological inhibitor and transfection of siRNA for PKCα was observed to effectively block glucose deprivation-induced ß-catenin degradation as well as the increase in LC3-II levels and the accumulation of a sub-G1 population. Together, our results demonstrated a molecular mechanism by which glucose deprivation can induce the GSK3ß-independent protein degradation of ß-catenin, leading to autophagy.


Assuntos
Glucose/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteína Quinase C-alfa/metabolismo , beta Catenina/metabolismo , Autofagia/efeitos dos fármacos , Autofagia/genética , Carbazóis/farmacologia , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/genética , Linhagem Celular , Glucose/farmacologia , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Quinase 3 da Glicogênio Sintase/genética , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Células HEK293 , Humanos , Immunoblotting , Cloreto de Lítio/farmacologia , Proteína Quinase C-alfa/antagonistas & inibidores , Proteína Quinase C-alfa/genética , Proteólise , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , beta Catenina/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...