Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Cell Transplant ; 28(9-10): 1289-1298, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31161784

RESUMO

Pancreatic cancer (PC) is a highly lethal cancer that has a strong ability for invasion and metastasis, poor prognosis, and a stubbornly high death rate due to late diagnosis and early metastasis. Therefore, a better understanding of the mechanisms of metastasis should provide novel opportunities for therapeutic purposes. As a route of metastasis in PC, perineural invasion (PNI) occurs frequently; however, the molecular mechanism of PNI is still poorly understood. In this study, we show that the hepatocyte growth factor (HGF)/c-Met pathway plays a vital role in the PNI of PC. We found that HGF promotes PC cell migration and invasion by activating the HGF/c-Met pathway, and enhances the expression of nerve growth factor (NGF) and matrix metalloproteinase-9 (MMP9) in vitro. Furthermore, HGF significantly increased PC cell invasion of the dorsal root ganglia (DRG) and promoted the outgrowth of DRG in cocultured models of PC cells and DRG. In contrast, the capacity for invasion and the phenomenon of PNI in PC cells were reduced when the HGF/c-Met pathway was blocked by siRNA. In conclusion, PSCs facilitate PC cell PNI via the HGF/c-Met pathway. Targeting the HGF/c-Met signaling pathway could be a promising therapeutic strategy for PC.


Assuntos
Fator de Crescimento de Hepatócito/metabolismo , Neoplasias Pancreáticas/metabolismo , Células Estreladas do Pâncreas/metabolismo , Proteínas Proto-Oncogênicas c-met/metabolismo , Transdução de Sinais , Linhagem Celular Tumoral , Humanos , Invasividade Neoplásica , Neoplasias Pancreáticas/patologia , Neoplasias Pancreáticas/terapia , Células Estreladas do Pâncreas/patologia
2.
Oxid Med Cell Longev ; 2016: 1616781, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26881012

RESUMO

Pancreatic cancer is the fourth leading cause of cancer-related death in the United States. Reactive oxygen species (ROS) are generally increased in pancreatic cancer cells compared with normal cells. ROS plays a vital role in various cellular biological activities including proliferation, growth, apoptosis, and invasion. Besides, ROS participates in tumor microenvironment orchestration. The role of ROS is a doubled-edged sword in pancreatic cancer. The dual roles of ROS depend on the concentration. ROS facilitates carcinogenesis and cancer progression with mild-to-moderate elevated levels, while excessive ROS damages cancer cells dramatically and leads to cell death. Based on the recent knowledge, either promoting ROS generation to increase the concentration of ROS with extremely high levels or enhancing ROS scavenging ability to decrease ROS levels may benefit the treatment of pancreatic cancer. However, when faced with oxidative stress, the antioxidant programs of cancer cells have been activated to help cancer cells to survive in the adverse condition. Furthermore, ROS signaling and antioxidant programs play the vital roles in the progression of pancreatic cancer and in the response to cancer treatment. Eventually, it may be the novel target for various strategies and drugs to modulate ROS levels in pancreatic cancer therapy.


Assuntos
Antioxidantes/metabolismo , Estresse Oxidativo/fisiologia , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Antineoplásicos/química , Apoptose , Carcinogênese , Dano ao DNA , Progressão da Doença , Humanos , Transdução de Sinais
3.
Oncotarget ; 7(14): 17760-72, 2016 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-26497365

RESUMO

Cigarette smoking is a risk factor for pancreatic cancer. It is suggested that 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK), a tobacco-specific nitrosamine, mediates the carcinogenic action of cigarette smoking by promoting cancer growth. In the present study, we show that smoking, HIF-1α expression and ß2-adrenogenic receptor (ß2-AR) expression are negatively correlated with the overall survival of pancreatic cancer patients. Moreover, HIF-1α expression and ß2-AR expression are positively correlated with smoking status, different histological differentiation and among the tumor node metastasis (TNM) stages in pancreatic cancer patients. NNK increases HIF-1α expression in pancreatic cancer in vitro and in vivo. Furthermore, knockdown of HIF-1α and ICI118, 551 (a ß2-AR selective antagonist) abrogates NNK-induced pancreatic cancer proliferation and invasion in vitro and inhibits NNK-induced pancreatic cancer growth in vivo. However, using CoCl2 (a HIF-1α stabilizing agent which decreases HIF-1α degradation under normoxia conditions) reverses ICI118, 551 induced effects under NNK exposure. Thus, our data indicate that ß2-AR signaling regulates NNK-induced pancreatic cancer progression via upregulation of HIF-1α. Taken together, ß2-AR signaling and HIF-1α may represent promising therapeutic targets for preventing smoking induced pancreatic cancer progression.


Assuntos
Carcinógenos/farmacologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Nitrosaminas/farmacologia , Neoplasias Pancreáticas/etiologia , Neoplasias Pancreáticas/patologia , Receptores Adrenérgicos beta 2/metabolismo , Idoso , Animais , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Progressão da Doença , Feminino , Técnicas de Silenciamento de Genes , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/biossíntese , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Camundongos , Camundongos Nus , Pessoa de Meia-Idade , Neoplasias Pancreáticas/induzido quimicamente , Neoplasias Pancreáticas/metabolismo , Receptores Adrenérgicos beta 2/biossíntese , Transdução de Sinais , Fumar/metabolismo , Ativação Transcricional , Regulação para Cima
4.
Oncotarget ; 6(16): 14153-64, 2015 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-25883224

RESUMO

MicroRNAs are involved in the initiation and progression of pancreatic cancer. In this study, we showed that miR-221/222 is overexpressed in pancreatic cancer. MiR-221/222 overexpression significantly promoted pancreatic cancer cell proliferation and invasion while inhibiting apoptosis. The expression of the matrix metalloproteinases (MMPs) MMP-2 and MMP-9 was increased in miR-221/222 mimic-transfected pancreatic cancer cells. Validation experiments identified TIMP-2 as a direct target of miR-221/222. These data indicate that overexpressed miR-221/222 may play an oncogenic role in pancreatic cancer by inducing the expression of MMP-2 and MMP-9, thus leading to cancer cell invasion.


Assuntos
Metaloproteinases da Matriz/metabolismo , MicroRNAs/genética , Neoplasias Pancreáticas/enzimologia , Neoplasias Pancreáticas/genética , Linhagem Celular Tumoral , Proliferação de Células/genética , Progressão da Doença , Células HEK293 , Humanos , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , MicroRNAs/biossíntese , Oncogenes , Neoplasias Pancreáticas/patologia , Transfecção , Regulação para Cima
5.
Ann N Y Acad Sci ; 1348(1): 10-9, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26284849

RESUMO

Pancreatic cancer (PCa), which is now the fourth most frequent cause of cancer-related death, has a median survival of less than 6 months and a 5-year survival rate of <6%. The hallmarks of this cancer include poor outcome, short survival duration, and resistance to therapy. The poor prognosis of PCa is related to its local recurrence, lymph node and liver metastases, and peritoneal dissemination. Recent studies have indicated that resveratrol has cancer-chemopreventive and anticancer activities. In this short review we summarize the chemopreventive and treatment effects of resveratrol in PCa, as follows: resveratrol inhibits the proliferation of pancreatic cancer cells; induces apoptosis and cell cycle arrest; inhibits metastasis and invasion of PCa cells; inhibits the proliferation and viability of PCa stem cells; enhances the chemoradiosensitization of PCa cells; and can affect diabetes mellitus in addition to PCa. On the basis of these data, resveratrol may be considered a potential anticancer agent for the treatment of PCa.


Assuntos
Antineoplásicos Fitogênicos/uso terapêutico , Neoplasias Pancreáticas/tratamento farmacológico , Estilbenos/uso terapêutico , Animais , Antineoplásicos Fitogênicos/farmacologia , Apoptose/efeitos dos fármacos , Pontos de Checagem do Ciclo Celular , Proliferação de Células/efeitos dos fármacos , Humanos , Neoplasias Pancreáticas/patologia , Resveratrol , Estilbenos/farmacologia
6.
Oncotarget ; 6(25): 20993-1003, 2015 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-25895130

RESUMO

Ginkgolic acid (GA) is a botanical drug extracted from the seed coat of Ginkgo biloba L. with a wide range of bioactive properties, including anti-tumor effect. However, whether GA has antitumor effect on pancreatic cancer cells and the underlying mechanisms have yet to be investigated. In this study, we show that GA suppressed the viability of cancer cells but has little toxicity on normal cells, e.g, HUVEC cells. Furthermore, treatment of GA resulted in impaired colony formation, migration, and invasion ability and increased apoptosis of cancer cells. In addition, GA inhibited the de novo lipogenesis of cancer cells through inducing activation of AMP-activated protein kinase (AMPK) signaling and downregulated the expression of key enzymes (e.g. acetyl-CoA carboxylase [ACC], fatty acid synthase [FASN]) involved in lipogenesis. Moreover, the in vivo experiment showed that GA reduced the expression of the key enzymes involved in lipogenesis and restrained the tumor growth. Taken together, our results suggest that GA may serve as a new candidate against tumor growth of pancreatic cancer partially through targeting pathway driving lipogenesis.


Assuntos
Lipogênese/fisiologia , Neoplasias Pancreáticas/metabolismo , Salicilatos/química , Proteínas Quinases Ativadas por AMP/metabolismo , Acetil-CoA Carboxilase/metabolismo , Animais , Apoptose , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Sobrevivência Celular , Ácido Graxo Sintases/metabolismo , Ginkgo biloba/química , Células Hep G2 , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Humanos , Lipogênese/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Invasividade Neoplásica , Neoplasias/patologia , Neoplasias Pancreáticas/tratamento farmacológico , Interferência de RNA , Transdução de Sinais
7.
Oncotarget ; 6(3): 1490-506, 2015 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-25557170

RESUMO

Metastasis is the major cause for the high mortality rate of pancreatic cancer. Human embryonic stem cell (hESC) associated genes frequently correlate with malignant disease progression. Recent studies have demonstrated that the embryonic protein Nodal, which plays a critical role during embryonic development, is re-expressed in several types of tumors and promotes cancers progression. However, little is known about the role of Nodal in pancreatic cancer. Here, we show that Nodal expression is upregulated in human pancreatic cancer tissues. Moreover, Nodal expression levels correlate well with the grade of pancreatic cancer differentiation. In addition, we present clear evidence that Nodal induces signal transduction through the Smad2/3-dependent pathway in vitro. Furthermore, we show that Nodal promotes pancreatic cancer cell migration and invasion, induces epithelial-mesenchymal transition (EMT) and enhances the expression of matrix metalloproteinase-2 (MMP2) and CXC chemokine receptor 4 (CXCR4). Using an in vivo liver metastasis model of pancreatic cancer, we observed that blocking Nodal signaling activity with the small-molecule inhibitor SB431542 decreases the number and size of liver metastases. Taken together, our results suggest that Nodal overexpression induces a metastatic phenotype in pancreatic cancer cells, and that targeting Nodal signaling may be a promising therapeutic strategy for pancreatic cancer.


Assuntos
Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patologia , Proteína Nodal/biossíntese , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Proteína Smad2/metabolismo , Proteína Smad3/metabolismo , Animais , Carcinoma Ductal Pancreático/genética , Diferenciação Celular/fisiologia , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Feminino , Técnicas de Silenciamento de Genes , Xenoenxertos , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Metástase Neoplásica , Proteína Nodal/genética , Proteína Nodal/metabolismo , Neoplasias Pancreáticas/genética , Fenótipo , Transdução de Sinais , Proteína Smad2/genética , Proteína Smad3/genética , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA