Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Cell Rep ; 37(8): 110036, 2021 11 23.
Artigo em Inglês | MEDLINE | ID: mdl-34818550

RESUMO

Balance between the hematopoietic stem cell (HSC) duality to either possess self-renewal capacity or differentiate into multipotency progenitors (MPPs) is crucial for maintaining homeostasis of the hematopoietic stem/progenitor cell (HSPC) compartment. To retain the HSC self-renewal activity, KIT, a receptor tyrosine kinase, in HSCs is activated by its cognate ligand KITLG originating from niche cells. Here, we show that AT-rich interaction domain 4B (ARID4B) interferes with KITLG/KIT signaling, consequently allowing HSC differentiation. Conditional Arid4b knockout in mouse hematopoietic cells blocks fetal HSC differentiation, preventing hematopoiesis. Mechanistically, ARID4B-deficient HSCs self-express KITLG and overexpress KIT. As to downstream pathways of KITLG/KIT signaling, inhibition of Src family kinases rescues the HSC differentiation defect elicited by ARID4B loss. In summary, the intrinsic ARID4B-KITLG/KIT-Src axis is an HSPC regulatory program that enables the differentiation state, while KIT stimulation by KITLG from niche cells preserves the HSPC undifferentiated pool.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Proteínas Proto-Oncogênicas c-kit/metabolismo , Animais , Comunicação Autócrina , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Proliferação de Células/fisiologia , Autorrenovação Celular/fisiologia , Proteínas de Ligação a DNA/fisiologia , Feminino , Hematopoese/fisiologia , Células-Tronco Hematopoéticas/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Proto-Oncogênicas c-kit/genética , Transdução de Sinais/fisiologia , Fator de Células-Tronco/metabolismo , Nicho de Células-Tronco/fisiologia , Fatores de Transcrição/metabolismo , Quinases da Família src/metabolismo
2.
Nat Commun ; 10(1): 4332, 2019 09 24.
Artigo em Inglês | MEDLINE | ID: mdl-31551414

RESUMO

PTEN is frequently mutated in prostate cancer. The tumor suppressor function of PTEN is attributed to its lipid phosphatase activity that counters PI3K action. Here, we report a PTEN-ARID4B-PI3K axis in which PTEN inhibits expression of ARID4B, while ARID4B is a transcriptional activator of the PI3K subunit genes PIK3CA and PIK3R2 that are crucial for activation of the PI3K/AKT pathway. Reciprocal binding of ARID4B and histone H1 to the PIK3CA and PIK3R2 promoters modulates chromatin condensation, suggesting a mechanism by which ARID4B activates these promoters. Functional analyses reveals that ARID4B is required for prostate tumorigenesis when PTEN is deficient. The biological significance is further substantiated by the existence of a PTEN/ARID4B/PIK3CA three-gene signature that improves the predictive power for prostate cancer recurrence in patients. In summary, we identify ARID4B as a master regulator in the PTEN-PI3K pathway, thus providing a potential therapeutic target for prostate cancer carrying PTEN mutations.


Assuntos
Antígenos de Neoplasias/metabolismo , Proteínas de Neoplasias/metabolismo , PTEN Fosfo-Hidrolase/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Neoplasias da Próstata/genética , Animais , Antígenos de Neoplasias/genética , Histonas/metabolismo , Humanos , Masculino , Camundongos Knockout , Proteínas de Neoplasias/genética , PTEN Fosfo-Hidrolase/genética , Neoplasias da Próstata/metabolismo , Transdução de Sinais
3.
BMC Complement Altern Med ; 17(1): 91, 2017 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-28153003

RESUMO

BACKGROUND: During the onset of osteoarthritis (OA), certain biochemical events have been shown to accelerate cartilage degradation, including the dysregulation of cartilage ECM anabolism, abnormal generation of reactive oxygen species (ROS) and overproduction of proteolytic enzymes and inflammatory cytokines. The potency of aucubin in protecting cellular components against oxidative stress, inflammation and apoptosis effects are well documented, which makes it a potential candidate for OA treatment. In this study, we aimed to evaluate the protective benefits of aucubin against OA using H2O2 and compression induced OA-like chondrocyte models. METHODS: The effects of aucubin were studied in porcine chondrocytes after 1 mM H2O2 stimulation for 30 min or sustained compression for 24 h. Effects of aucubin on cell proliferation and cytotoxicity of chondrocytes were measured with WST-1 and LDH assays. ROS production was evaluated by the Total ROS/Superoxide Detection Kit. Caspase-3 activity was evaluated by the CaspACE assay system. The levels of apoptosis were evaluated by the Annexin V-FITC apoptosis detection kit. OA-related gene expression was measured by reverse transcription quantitative polymerase chain reaction (RT-qPCR). Total DNA quantification was evaluated by the DNeasy Blood and Tissue kit. Sulfated-glycosaminoglycans (sGAGs) production and content were evaluated by DMMB assay and Alcian blue staining. RESULTS: The results showed that the ROS scavenge effects of aucubin appeared after 1 h of pretreatment. Aucubin could reduce the caspase-3 activity induced by H2O2, and reduced the apoptosis cell population in flowcytometry. In RT-qPCR results, aucubin could maintain ACAN and COL2A1 gene expressions, and prevent IL6 and MMP13 gene up-regulation induced by H2O2 and compression stimulations. In the DMMB assay and Alcian blue staining, aucubin could maintain the sGAG content and protect chondrocytes against compressive stress, but not oxidative stress from H2O2. CONCLUSIONS: These results indicated that aucubin has protective effects in an osteoarthritic chondrocyte model induced by H2O2 and mechanical stimulus.


Assuntos
Condrócitos/efeitos dos fármacos , Glucosídeos Iridoides/uso terapêutico , Osteoartrite/tratamento farmacológico , Agrecanas/genética , Animais , Apoptose/efeitos dos fármacos , Caspase 3/metabolismo , Proliferação de Células/efeitos dos fármacos , Colágeno Tipo II/genética , Modelos Animais de Doenças , Expressão Gênica/efeitos dos fármacos , Peróxido de Hidrogênio , Técnicas In Vitro , Interleucina-6/genética , Glucosídeos Iridoides/toxicidade , Metaloproteinase 13 da Matriz/genética , Osteoartrite/genética , Estimulação Física , Espécies Reativas de Oxigênio/metabolismo , Suínos
4.
Mol Nutr Food Res ; 61(9)2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28181403

RESUMO

SCOPE: Caffeic acid phenethyl ester (CAPE), a bioactive component of propolis, is considered as a new anti-cancer agent. Oral squamous cell carcinoma (OSCC) is the most common oral cancer with unsatisfying survival. N-myc downstream regulated family genes (NDRGs) involve in numerous physiological processes. We investigated the anti-cancer effect of CAPE on OSCC and related mechanisms. METHODS AND RESULTS: Cell proliferation assay, western blot, gene transfection and knockdown, and reporter assay were applied. We showed that CAPE attenuated OSCC cell proliferation and invasion in vitro, and safely and effectively inhibited OSCC cell growth in a xenograft animal model. CAPE treatment induced NDRG1, but not NDRG2 and NDRG3, expression in OSCC cells as determined by western blot, RT-qPCR, and reporter assay. The 5'-deletion assay demonstrated that CAPE increased NDRG1 promoter activity depending on the region of -128 to +46 of the 5'-flanking of NDRG1 gene. NDRG1 gene knockdown attenuated CAPE anti-growth effect on OSCC cells. CAPE activated mitogen-activated protein kinase (MAPK) signaling pathway. The extracellular signal regulated kinase (ERK) inhibitor (PD0325901) and ERK1 knockdown blocked CAPE-induced NDRG1 expression in OSCC cells. CONCLUSION: CAPE activated MAPK signaling pathway and increased NDRG1 expression through phosphorylation of ERK1/2 to repress OSCC cells growth.


Assuntos
Ácidos Cafeicos/farmacologia , Carcinoma de Células Escamosas/tratamento farmacológico , Proteínas de Ciclo Celular/genética , MAP Quinases Reguladas por Sinal Extracelular/fisiologia , Neoplasias de Cabeça e Pescoço/tratamento farmacológico , Peptídeos e Proteínas de Sinalização Intracelular/genética , Sistema de Sinalização das MAP Quinases/fisiologia , Neoplasias Bucais/tratamento farmacológico , Álcool Feniletílico/análogos & derivados , Animais , Carcinoma de Células Escamosas/patologia , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Neoplasias de Cabeça e Pescoço/patologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Neoplasias Bucais/patologia , Álcool Feniletílico/farmacologia , Regiões Promotoras Genéticas , Carcinoma de Células Escamosas de Cabeça e Pescoço , Regulação para Cima , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Int J Mol Sci ; 17(9)2016 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-27589737

RESUMO

Oral squamous cell carcinoma (OSCC) is a common malignancy with a growing worldwide incidence and prevalence. The N-myc downstream regulated gene (NDRG) family of NDRG1, 2, 3, and mammary serine protease inhibitor (Maspin) gene are well-known modulators in the neoplasia process. Current research has considered iron chelators as new anti-cancer agents; however, the anticancer activities of iron chelators and their target genes in OSCC have not been well investigated. We showed that iron chelators (Dp44mT, desferrioxamine (DFO), and deferasirox) all significantly inhibit SAS cell growth. Flow cytometry further indicated that Dp44mT inhibition of SAS cells growth was partly due to induction of G1 cell cycle arrest. Iron chelators enhanced expressions of NDRG1 and NDRG3 while repressing cyclin D1 expression in OSCC cells. The in vivo antitumor effect on OSCC and safety of Dp44mT were further confirmed through a xenograft animal model. The Dp44mT treatment also increased Maspin protein levels in SAS and OECM-1 cells. NDRG3 knockdown enhanced the growth of OECM-1 cells in vitro and in vivo. Our results indicated that NDRG3 is a tumor suppressor gene in OSCC cells, and Dp44mT could be a promising therapeutic agent for OSCC treatment.


Assuntos
Antineoplásicos/farmacologia , Carcinoma de Células Escamosas/tratamento farmacológico , Proliferação de Células/efeitos dos fármacos , Quelantes de Ferro/farmacologia , Neoplasias Bucais/tratamento farmacológico , Tiossemicarbazonas/farmacologia , Animais , Antineoplásicos/uso terapêutico , Carcinoma de Células Escamosas/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Ciclina D1/genética , Ciclina D1/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Quelantes de Ferro/uso terapêutico , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias Bucais/metabolismo , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Serpinas/genética , Serpinas/metabolismo , Tiossemicarbazonas/uso terapêutico
6.
Int J Biol Macromol ; 93(Pt A): 852-859, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27642127

RESUMO

During the progression of osteoarthritis (OA), dysregulation of extracellular matrix anabolism, abnormal generation of reactive oxygen species (ROS) and inflammatory cytokines have been shown to accelerate the degradation process of cartilage. The potency of c-phycocyanin (C-PC) to protect cellular components against oxidative stress, along with its anti-inflammation and anti-apoptosis effects, are well documented; however, effects of C-PC on OA are still unclear. In this study, we aimed to investigate the effects of C-PC on OA using H2O2 or compression-stimulated OA-like porcine chondrocyte models. The results showed that C-PC had the ability to inhibit ROS production, reverse caspase-3 activity, and reduce apoptosis cell population. C-PC also reversed aggrecan and type II collagen gene expressions after stimulation with 1mM H2O2 or 60psi of compression. Inhibition of IL-6 and MMP-13 genes was observed in compression-stimulated chondrocytes but not in H2O2-treated cells. In dimethylmethylene blue assay and alcian blue staining, C-PC maintained the sulfated-glycosaminoglycan (sGAG) content after stimulation with compression. We concluded that C-PC can prevent early signs of OA caused by compressive stress and attenuate H2O2-induced oxidative stress. Therefore, we suggest that C-PC can be used as a potential drug candidate for chronic OA treatment.


Assuntos
Condrócitos , Peróxido de Hidrogênio/toxicidade , Osteoartrite , Ficocianina/farmacologia , Estresse Mecânico , Animais , Apoptose/efeitos dos fármacos , Caspase 3/metabolismo , Condrócitos/metabolismo , Condrócitos/patologia , Força Compressiva , Interleucina-6/metabolismo , Metaloproteinase 13 da Matriz/metabolismo , Osteoartrite/induzido quimicamente , Osteoartrite/tratamento farmacológico , Osteoartrite/metabolismo , Osteoartrite/patologia , Suínos
7.
Cancer Lett ; 375(1): 142-151, 2016 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-26965996

RESUMO

Prostate-derived Ets (E-twenty six) factor (PDEF), an epithelium-specific member of the Ets family of transcription factors, has been shown to play a role in suppressing the development of many epithelium-derived cancers such as prostate and breast cancer. It is not clear, however, whether PDEF is involved in the development or progression of bladder cancer. In a comparison between normal urothelium and bladder tumor tissue, we identified significant decreases of PDEF in the tumor tissue. Further, the immunohistochemistry assays indicated a significantly higher immunostaining of PDEF in low-grade bladder tumors. Additionally, the highly differentiated transitional-cell bladder carcinoma RT-4 cells expressed significantly more PDEF levels than the bladder carcinoma HT1376 and the T24 cells. Ectopic overexpression of PDEF attenuated proliferation, invasion, and tumorigenesis of bladder carcinoma cells in vitro and in vivo. PDEF enhanced the expression levels of mammary serine protease inhibitor (MASPIN), N-myc downstream regulated gene 1 (NDRG1), KAI1, and B-cell translocation gene 2 (BTG2). PDEF modulated epithelial-mesenchymal-transition (EMT) by upregulating E-cadherin expression and downregulating the expression of N-cadherin, SNAIL, SLUG, and vimentin, leading to lower migration and invasion abilities of bladder carcinoma cells. Filamentous actin (F-actin) polarization and remodeling were observed in PDEF-knockdown RT-4 cells. Our results suggest that PDEF gene expression is associated with the extent of bladder neoplasia and PDEF modulated the expressions of EMT-related genes. The induction of BTG2, NDRG1, MASPIN, and KAI1 gene expressions by PDEF may explain the inhibitory functions of PDEF on the proliferation, invasion, and tumorigenesis in bladder carcinoma cells.


Assuntos
Carcinogênese/metabolismo , Transição Epitelial-Mesenquimal , Proteínas Proto-Oncogênicas c-ets/metabolismo , Neoplasias da Bexiga Urinária/metabolismo , Animais , Linhagem Celular Tumoral , Proliferação de Células , Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Camundongos Endogâmicos BALB C , Camundongos Nus , Invasividade Neoplásica , Transplante de Neoplasias , Proteínas Proto-Oncogênicas c-ets/genética , Carga Tumoral , Neoplasias da Bexiga Urinária/patologia
8.
Sci Rep ; 4: 5814, 2014 Jul 24.
Artigo em Inglês | MEDLINE | ID: mdl-25056456

RESUMO

Accumulating evidence suggests that renal tubulointerstitial fibrosis is a main cause of end-stage renal disease. Clinically, there are no beneficial treatments that can effectively reverse the progressive loss of renal functions. Caffeic acid phenethyl ester is a natural phenolic antifibrotic agent, but rapid decomposition by an esterase leads to its low bioavailability. In this study, we evaluated the effects of KS370G, a caffeic acid phenylethyl amide, on murine renal fibrosis induced by unilateral renal ischemia-reperfusion injury (IRI) and in TGF-ß1 stimulated renal tubular epithelial cells (NRK52E and HK-2). In the animal model, renal fibrosis was evaluated at 14 days post-operation. Immediately following the operation, KS370G (10 mg/kg) was administered by oral gavage once a day. Our results show that KS370G markedly attenuates collagen deposition and inhibits an IRI-induced increase of fibronectin, vimentin, α-SMA and TGF-ß1 expression and plasma TGF-ß1 levels in the mouse kidney. Furthermore, KS370G reverses TGF-ß1-induced downregulation of E-cadherin and upregulation of α-SMA and also decreases the expression of fibronectin, collagen I and PAI-1 and inhibits TGF-ß1-induced phosphorylation of Smad2/3. These findings show the beneficial effects of KS370G on renal fibrosis in vivo and in vitro with the possible mechanism being the inhibition of the Smad2/3 signaling pathway.


Assuntos
Ácidos Cafeicos/farmacologia , Nefropatias/tratamento farmacológico , Traumatismo por Reperfusão/tratamento farmacológico , Animais , Ácidos Cafeicos/uso terapêutico , Proteínas Cdh1/metabolismo , Linhagem Celular , Colágeno/metabolismo , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/fisiologia , Transição Epitelial-Mesenquimal , Fibronectinas/metabolismo , Fibrose , Humanos , Isquemia/tratamento farmacológico , Isquemia/patologia , Nefropatias/metabolismo , Túbulos Renais/irrigação sanguínea , Túbulos Renais/metabolismo , Túbulos Renais/patologia , Masculino , Camundongos Endogâmicos ICR , Fosforilação , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Traumatismo por Reperfusão/metabolismo , Serpina E2/metabolismo , Proteína Smad2/metabolismo , Proteína Smad3/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Vimentina/metabolismo
9.
Eur J Pharmacol ; 684(1-3): 108-15, 2012 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-22484506

RESUMO

Cardiac hypertrophy is an important compensatory mechanism in response to a pressure overload, but a sustained excessive cardiac workload may deteriorate to maladaptive hypertrophy and to increased risk of heart failure. In this study, we evaluated the effects of KS370G on left ventricular hypertrophy and function. Abdominal aortic banding was performed by constricting the abdominal aorta. Hypertrophied heart was studied at 8 weeks after the operation. After the operation, KS370G 1mg/kg (K1 group) was administered by oral gavage once a day. Left ventricular function was measured by a 1.2F pressure-volume catheter (Scisense, Canada). The levels of protein for α-SMA (smooth muscle actin), p-AKT (protein kinase B), p-GSK3ß (glycogen synthase kinase 3ß) and p-ERKs (extracellular signal-regulated kinases) in myocardium were analyzed by Western blot. Plasma levels of angiotensin II, atrial natriuretic peptide and lactate dehydrogenase were analyzed by commercial kits. H.E. staining and M.T. staining methods were also used to observe diameter of cardiomyocytes and collagen accumulation. Chronic oral treatment with 1mg/kg KS370G inhibited cardiac hypertrophy and improved cardiac function induced by pressure overload. KS370G also decreased the plasma levels of atrial natriuretic peptide and lactate dehydrogenase. Besides, pressure overload-induced increase of α-SMA and phosphorylation of ERK, AKT and GSK3ß were significantly reduced by chronic oral treatment with KS370G. We also found that chronic oral treatment with KS370G reduced cardiac collagen accumulation. KS370G improved left ventricular function and inhibited cardiac hypertrophy through the decrease of the phosphorylation of ERK, AKT and GSK3ß in pressure-overload mice heart.


Assuntos
Ácidos Cafeicos/farmacologia , Cardiotônicos/farmacologia , Ventrículos do Coração/patologia , Ventrículos do Coração/fisiopatologia , Pressão/efeitos adversos , Actinas/metabolismo , Angiotensina II/sangue , Animais , Aorta/efeitos dos fármacos , Aorta/patologia , Aorta/fisiopatologia , Fator Natriurético Atrial/sangue , Ácidos Cafeicos/uso terapêutico , Cardiotônicos/uso terapêutico , Colágeno/metabolismo , Constrição , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Ventrículos do Coração/efeitos dos fármacos , Hipertrofia/tratamento farmacológico , Hipertrofia/etiologia , Hipertrofia/metabolismo , Hipertrofia/fisiopatologia , L-Lactato Desidrogenase/sangue , Masculino , Camundongos , Camundongos Endogâmicos ICR , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Fosfoproteínas/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA