Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Cytokine ; 170: 156342, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37651918

RESUMO

GLIS3 is highly expressed in multiple cancers, but it has not been studied in gastric adenocarcinoma (GAC). Based on bioinformatics analysis, the prognostic significance of GLIS3 in GAC was analyzed. GAC cells were transfected with small interfering (si)-GLIS3 and GLIS3 overexpression plasmid as well as treated with SB505124 [an inhibitor for transforming growth factor beta receptor 1 (TGFßR1)] and dorsomorphin [an inhibitor for bone morphogenetic protein receptor 1 (BMPR1)]. The GLIS3 expression was detected using qRT-PCR. The impacts of GLIS3 on the proliferation, invasion and migration of GAC cells were measured using cell function assays. The activation of phosphor (p)-Smad1/5 was tested by immunofluorescence. Western blot was utilized to measure the level of transforming growth factor (TGF)-ß1/Smad1/5 signaling pathway-related proteins (TGF-ß1, p-Smad1, Smad1, p-Smad5, Smad5). GLIS3 was expressed at high levels in GAC tissues and cell lines and its high expression could indicate the poor prognosis of GAC patients. GLIS3 inhibition declined the proliferative, invasive and migratory capabilities as well as TGF-ß1 expression and phosphorylation of Smad1/5 in GAC cells. Overexpressed GLIS3 promoted proliferation, migration, invasion, TGF-ß1 expression and Smad1/5 phosphorylation in GAC cells, with SB505124 reversing the effects of overexpressed GLIS3 on proliferation, migration, invasion and Smad1/5 phosphorylation whereas dorsomorphin exhibiting no influence on GLIS3-induced effects. GLIS3 facilitated the malignant phenotype of GAC cells via regulating TGF-ß1/TGFßR1/Smad1/5 pathway, which may be a novel prognostic indicator of GAC and provided a target for GAC treatment.


Assuntos
Adenocarcinoma , Neoplasias Gástricas , Humanos , Prognóstico , Fator de Crescimento Transformador beta1 , Transdução de Sinais , Proteínas de Ligação a DNA , Proteínas Repressoras , Transativadores
2.
Cell Biochem Funct ; 41(5): 564-572, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37232472

RESUMO

Numerous signaling pathways are well-known in osteoblastic differentiation of human bone marrow mesenchymal stem cells (hBMSCs), including transforming growth factor-beta (TGF-ß) signaling pathway, which sends signals through specific type I and II serine/threonine kinase receptors. However, the key role of TGF-ß signaling during bone formation and remodeling is yet to be studied. A TGF-ß type I receptor inhibitor, SB505124, discovered through a screening of a small molecule library for their effect of osteoblast differentiation of hBMSCs. Alkaline phosphatase quantification and staining were tested as indicators of osteoblastic differentiation and Alizarin red staining was tested as an indicator of in vitro mineralization. Changes in gene expressions were assessed using qRT-PCR. SB505124 showed significant inhibition of the osteoblast differentiation of hBMSCs, as confirmed by reduced alkaline phosphatase, in vitro mineralization, and downregulation of osteoblast-associated gene expression. To further understand the molecular mechanisms involved in the inhibition of the TGF-ß type I receptor, we assessed the effects on signature genes of several signaling pathways identified in the osteoblast differentiation of hBMSCs. SB505124 downregulated gene expression of many genes linked to osteoblast-related signaling pathways including TGF-ß, insulin, focal adhesion, Notch, Vitamin D, interleukin (IL)-6, osteoblast signaling, and cytokines and inflammatory. We report TGF-ß type I receptor inhibitor (SB505124) is a potent inhibitor of osteoblastic differentiation of hBMSCs that could be a valuable innovative therapeutic tool to cure bone disorders with increased bone formation, besides its potential use to treat patients with cancer and fibrosis.


Assuntos
Células-Tronco Mesenquimais , Osteogênese , Humanos , Receptor do Fator de Crescimento Transformador beta Tipo I/metabolismo , Fosfatase Alcalina/metabolismo , Fosfatase Alcalina/farmacologia , Diferenciação Celular , Fator de Crescimento Transformador beta/metabolismo , Osteoblastos/metabolismo , Células-Tronco Mesenquimais/metabolismo , Células Cultivadas
3.
Pharmacol Res ; 85: 15-22, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24844437

RESUMO

Transforming growth factor ß (TGF-ß) belongs to a class of pleiotropic cytokines that are involved in the processes of embryonic development, wound healing, cell proliferation, and differentiation. Moreover, TGF-ß is also regarded as a central regulator in the pathogenesis and development of various liver diseases because it contributes to almost all of the stages of disease progression. A range of liver cells are considered to secrete TGF-ß ligands and express related receptors and, consequently, play a crucial role in the progression of liver disease via different signal pathways. In this manuscript, we review the role of the TGF-ß signaling pathway in liver disease and the potential of targeting the TGF-ß signaling in the pharmacological treatment of liver diseases.


Assuntos
Hepatopatias/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Humanos , Hepatopatias/tratamento farmacológico , Receptores de Fatores de Crescimento Transformadores beta/metabolismo
4.
Stem Cell Res Ther ; 12(1): 65, 2021 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-33461597

RESUMO

BACKGROUND: Cancer cachexia is a wasting syndrome that is quite common in terminal-stage cancer patients. Cancer-related anemia is one of the main features of cancer cachexia and mostly results in a poor prognosis. The disadvantages of the current therapies are obvious, but few new treatments have been developed because the pathological mechanism remains unclear. METHODS: C57BL/6 mice were subcutaneously injected with Lewis lung carcinoma cells to generate a cancer-related anemia model. The treated group received daily intraperitoneal injections of SB505124. Blood parameters were determined with a routine blood counting analyzer. Erythroid cells and hematopoietic stem/progenitor cells were analyzed by flow cytometry. The microarchitecture changes of the femurs were determined by micro-computed tomography scans. Smad2/3 phosphorylation was analyzed by immunofluorescence and Western blotting. The changes in the hematopoietic stem cell niche were revealed by qPCR analysis of both fibrosis-related genes and hematopoietic genes, fibroblastic colony-forming unit assays, and lineage differentiation of mesenchymal stromal cells. RESULTS: The mouse model exhibited hematopoietic suppression, marked by a decrease of erythrocytes in the peripheral blood, as well as an increase of immature erythroblasts and reduced differentiation of multipotent progenitors in the bone marrow. The ratio of bone volume/total volume, trabecular number, and cortical wall thickness all appeared to decrease, and the increased osteoclast number has led to the release of latent TGFß and TGFß signaling over-activation. Excessive TGFß deteriorated the hematopoietic stem cell niche, inducing fibrosis of the bone marrow as well as the transition of mesenchymal stromal cells. Treatment with SB505124, a small-molecule inhibitor of TGFß signaling, significantly attenuated the symptoms of cancer-related anemia in this model, as evidenced by the increase of erythrocytes in the peripheral blood and the normalized proportion of erythroblast cell clusters. Meanwhile, hindered hematopoiesis and deteriorated hematopoietic stem cell niche were also shown to be restored with SB505124 treatment. CONCLUSION: This study investigated the role of TGFß released by bone remodeling in the progression of cancer-related anemia and revealed a potential therapeutic approach for relieving defects in hematopoiesis.


Assuntos
Anemia , Neoplasias , Anemia/tratamento farmacológico , Animais , Diferenciação Celular , Hematopoese , Células-Tronco Hematopoéticas , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Nicho de Células-Tronco , Fator de Crescimento Transformador beta/genética , Microtomografia por Raio-X
5.
J Pharm Biomed Anal ; 117: 205-9, 2016 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-26363490

RESUMO

A sensitive, selective and rapid liquid chromatography-electrospray ionization-tandem mass spectrometry (LC-MS/MS) method has been developed for the quantification of the novel transforming growth factor-ß (TGF-ß) inhibitor SB-505124 in rat plasma and then validated. Plasma samples were prepared by simple protein precipitation. Separation was performed on a Diamonsil ODS chromatography column using a mobile phase of acetonitrile and 0.1% (v/v) aqueous formic acid. SB-505124 and the internal standard doxorubicin were detected in the positive ion mode using multiple reaction monitoring of the transitions at m/z 336.2→320.1 and 544.2→397.2, respectively. Calibration curve was linear (r>0.9996) over a concentration range of 10-5000 ng/mL with the lower quantification limit of 10 ng/mL. Both intra- and inter-day precision were within 6.5% and trueness were not more than 3.1%. Extraction recovery and matrix effect were within acceptable limits. Stability tests showed that SB-505124 and the IS remained stable throughout the analytical procedure. The validated LC-MS/MS method was then used to analyze the pharmacokinetics of SB-505124 administered to rats intravenously (8 mg/kg) or orally (10 mg/kg). Oral bioavailability of SB-505124 was calculated as 76.4%, indicating the potential of SB-505124 as an orally administered drug.


Assuntos
Benzodioxóis/sangue , Benzodioxóis/farmacocinética , Imidazóis/sangue , Imidazóis/farmacocinética , Piridinas/sangue , Piridinas/farmacocinética , Receptores de Fatores de Crescimento Transformadores beta/antagonistas & inibidores , Espectrometria de Massas em Tandem/métodos , Administração Oral , Animais , Benzodioxóis/administração & dosagem , Cromatografia Líquida/métodos , Imidazóis/administração & dosagem , Masculino , Piridinas/administração & dosagem , Ratos , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa