Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Gastrointest Oncol ; 15(1): 112-124, 2024 Feb 29.
Artigo em Inglês | MEDLINE | ID: mdl-38482242

RESUMO

Background: Gastrointestinal stromal tumor (GIST) is a common mesenchymal tumor of the gastrointestinal system. They originate from the interstitial cells of Cajal located within the muscle layer and are characterized by over-expression of the tyrosine kinase receptor KIT. Methods: Data from the Surveillance Epidemiology, and End Results (SEER) database of 1,213 patients diagnosed with GIST between 2010 and 2019 were dichotomized into a modeling set and a validation set at a 2:1 ratio. For the modeling set, both univariate and multivariate Cox regression analyses were used to identify independent prognostic factors. A nomogram was then constructed based on these determinants. Model efficacy was tested using receiver operating characteristic (ROC) curves, calibration curves, clinical decision curves, and risk stratification analysis in both subsets. Results: Identified prognostic determinants included age, sex, pathological differentiation level, tumor-node-metastasis (TNM) stage, surgical intervention, radiotherapy, and marital status. The constructed nomogram showed area under the ROC curve (AUC) values of 0.822, 0.793, and 0.779 for 1-, 3-, and 5-year overall survival (OS) in the modeling set, respectively, while in the validation set, the values were 0.796, 0.823, and 0.806, respectively. Calibration plots from both sets confirmed the concordance between predicted and observed survival. Decision curve analysis (DCA) indicated significant clinical utility for the nomogram. Risk stratification of the patient data revealed distinct survival differences between high-risk and low-risk cohorts in both sets (P<0.001). Conclusions: A novel and potent nomogram for the prognosis of GIST has been introduced. This model's precision offers crucial insights for clinical decisions, yet further external validation remains essential.

2.
Int J Gen Med ; 16: 2285-2294, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37304906

RESUMO

Background: Immune checkpoint inhibitors have achieved limited clinical effectiveness in colon cancer. Stem memory T cells (TSCMs) and in-situ cytotoxic T cells are dominant contributors to host immunity. Currently, data on the correlation between TSCM and T cell abundance and clinicopathological characteristics in colon cancer are largely unavailable. Methods: In-situ cytotoxic T cells are identified based on the quantification of CD3+ and CD8+ markers using immunohistochemistry (IHC) in the core of the tumor and the invasive margin of the tumor. The expression of representative markers of TSCMs, CD27 and CD95, was assayed using IHC in colon cancer tissues. Correlations between the levels of each marker and the clinicopathological characteristics as well as prognosis were evaluated. Results: High densities of CD3+ and CD8+ T cells correlated with stage I-II tumors, whereas a lower infiltration of cytotoxic T cells correlated with advanced-stage tumors. CD27 and CD95 were both expressed in the membrane of T cells present in the tumor stroma and their levels showed a negative correlation with the TNM stage. CD3, CD8, and CD27 were expressed at the same locations simultaneously, indicating their coordinated action against cancer. In addition, cytotoxic T cell densities and CD27 and CD95 expression remained independent prognostic factors for overall survival. Conclusion: In-situ cytotoxic T cells and TSCMs play important roles in colon cancer development. TSCMs marker CD27 and CD95 were both indicators of survival in patients with colon cancer. Thus, it is believed that TSCMs represent a desirable population for future use in combination immunotherapy.

3.
J Bioenerg Biomembr ; 53(3): 343-349, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33821396

RESUMO

Non-small cell lung cancer (NSCLC) threatens human life globally with high morbidity and mortality and radiotherapy is one of the most effective methods for the treatment of NSCLC. However, it is currently reported that the angiogenesis of tumors can be induced by a low dosage of irradiation. Abiraterone is an oral anti-tumor agent for the treatment of castration-resistant prostate cancer (CRPC). In the present study, the anti-angiogenesis effect of Abiraterone against HUVECs incubated with irradiated lung cancer cell medium will be investigated. The HUVECs were incubated with a cultural medium of the NSCLC cell line-A549, Abiraterone-treated A549 cells, irradiation-treated A549 cells, and Abiraterone and irradiation co-treated A549 cells. The tolerable concentration of Abiraterone against HUVECs was determined using MTT assay. The migration and angiogenesis abilities of HUVECs were evaluated using transwell and tube formation assays, respectively. The expression levels of VEGF, MMP-2, and MMP-9 in the treated HUVECs were detected using qRT-PCR and ELISA. Western blot was used to determine the expressions of p-PI3K and p-AKT. The tolerable concentration of Abiraterone used in the present study was 50 nM. First, the migration rate and numbers of formed tubes were significantly decreased by the A549 medium treated with Abiraterone and elevated by the A549 medium treated with irradiation but greatly suppressed by the co-treatment with Abiraterone. Subsequently, VEGF, MMP-2, and MMP-9 were significantly downregulated by the A549 medium treated with Abiraterone and upregulated by the A549 medium treated with irradiation but greatly inhibited by the co-treatment with Abiraterone. Lastly, the activated PI3K/AKT signaling pathway induced by the A549 medium treated with irradiation was significantly suppressed by the A549 medium treated with both irradiation and Abiraterone. Abiraterone suppressed irradiated lung cancer cells-induced angiogenic capacities of endothelial cells.


Assuntos
Androstenos/uso terapêutico , Neoplasias Pulmonares/tratamento farmacológico , Androstenos/farmacologia , Células Endoteliais , Humanos
4.
Technol Cancer Res Treat ; 20: 1533033820964238, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33472555

RESUMO

BACKGROUND AND AIM: There are an increasing number of studies indicating the important roles served by long non-coding RNAs (lncRNAs) in the development of different types of cancer. LINC00460 is a novel identified lncRNA that was found to be upregulated in colorectal cancer. However, the biological roles of LINC00460 in colorectal cancer have yet to be fully elucidated. This study was aimed to investigate the functions and molecular mechanisms of LINC00460 on colorectal cancer metastasis. METHODS: Expression of LINC00460 and biglycan (BGN) in colorectal cancer tissues and cell lines were quantified by real time PCR or western blotting assay. Cell migration and invasion assays were performed to determine the effect of LINC00460 on tumor metastasis in vitro. The binding interaction between microRNA-149-5p and LINC00460 was revealed by luciferase reporter assay. RESULTS: In the present study, lncRNA LINC00460 was shown to be upregulated in colorectal cancer tissues, and overexpression of LINC00460 significantly promoted metastasis of colorectal cancer in vitro. Furthermore, miR-149-5p interacted with LINC00460, and they negatively regulated expression of each other. Transfection of miR-149-5p mimics partially counteracted the tumor metastasis-promoting effects induced by LINC00460 overexpression. Finally, overexpression of LINC00460 upregulated the expression levels of biglycan, a target gene of miR-149-5p, which has also been identified as an oncogenic driver in colorectal cancer. CONCLUSION: Taken together, the present study demonstrated that LINC00460 promoted metastasis of CRC by sponging miR-149-5p and thereby affecting biglycan expression levels.


Assuntos
Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Neoplasias Colorretais/genética , Neoplasias Colorretais/metabolismo , MicroRNAs/metabolismo , Idoso , Idoso de 80 Anos ou mais , Biglicano/genética , Movimento Celular/genética , Neoplasias Colorretais/patologia , Citoplasma/metabolismo , Intervalo Livre de Doença , Feminino , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Células HCT116 , Células HEK293 , Células HT29 , Humanos , Masculino , MicroRNAs/genética , Invasividade Neoplásica/genética , Metástase Neoplásica/genética , RNA Longo não Codificante , Transfecção , Regulação para Cima
5.
Oncol Lett ; 11(4): 2941-2945, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27073580

RESUMO

Liver cancer, particularly hepatocellular carcinoma (HCC), is one of leading causes of cancer-related mortality worldwide. Upregulation of the evolutionary conserved Hippo signaling pathway has been observed in HCC patients, and Yes-associated protein 1 (YAP1) has been reported to play a key role in HCC tumorigenesis. microRNAs (miRNAs) are a family of small non-coding RNAs, usually 21-25 nucleotides in length, and are essential in the regulation of gene expression. Abnormal miRNA expression has been implicated in the initiation and progression of numerous forms of cancers, including liver cancer. Here, we report the identification of a novel miRNA, miR-186, and its functions as an HCC tumor suppressor. We observed that miR-186 was downregulated in several HCC cell lines, and that it directly targets YAP1 mRNA. Overexpression of miR-186 in HCC cells significantly downregulates YAP1 mRNA and protein levels, leading to downregulation of the Hippo signaling pathway, which in turn severely inhibits HCC cell migration, invasion and proliferation. Our study is the first to report the direct involvement of miR-186 in downregulating YAP1 and, more significantly, inhibiting HCC tumorigenesis, and supports the role miR-186 as a potential therapeutic target in treating liver cancer.

6.
Gene ; 586(1): 41-7, 2016 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-27041240

RESUMO

Colon cancer is one of the major causes of cancer-related death in the world. Understanding the molecular mechanism underlying this malignancy will facilitate the diagnosis and treatment. Serine-arginine protein kinase 2 (SRPK2) has been reported to be upregulated in several cancer types. However, its expression and functions in colon cancer remains unknown. In this study, it was found that the expression of SRPK2 was up-regulated in the clinical colon cancer samples. Overexpression of SRPK2 promoted the growth and migration of colon cancer cells, while knocking down the expression of SRPK2 inhibited the growth, migration and tumorigenecity of colon cancer cells. Molecular mechanism studies revealed that SRPK2 activated ERK signaling in colon cancer cells. Taken together, our study demonstrated the tumor promoting roles of SRPK2 in colon cancer cells and SRPK2 might be a promising therapeutic target for colon cancer.


Assuntos
Neoplasias do Colo/genética , Neoplasias do Colo/patologia , Proteínas Serina-Treonina Quinases/genética , Proteínas Proto-Oncogênicas B-raf/metabolismo , Animais , Linhagem Celular Tumoral , Movimento Celular , Colo/patologia , Humanos , Sistema de Sinalização das MAP Quinases , Camundongos , Camundongos Nus
7.
Oncotarget ; 7(13): 17047-59, 2016 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-26933997

RESUMO

Here we tested anti-tumor activity of KU-0060648 in preclinical hepatocellular carcinoma (HCC) models. Our results demonstrated that KU-0060648 was anti-proliferative and pro-apoptotic in established (HepG2, Huh-7 and KYN-2 lines) and primary human HCC cells, but was non-cytotoxic to non-cancerous HL-7702 hepatocytes. DNA-PKcs (DNA-activated protein kinase catalytic subunit) is an important but not exclusive target of KU-0060648. DNA-PKcs knockdown or dominant negative mutation inhibited HCC cell proliferation. On the other hand, overexpression of wild-type DNA-PKcs enhanced HepG2 cell proliferation. Importantly, KU-0060648 was still cytotoxic to DNA-PKcs-silenced or -mutated HepG2 cells, although its activity in these cells was relatively weak. Further studies showed that KU-0060648 inhibited PI3K-AKT-mTOR activation, independent of DNA-PKcs. Introduction of constitutively-active AKT1 (CA-AKT1) restored AKT-mTOR activation after KU-0060648 treatment in HepG2 cells, and alleviated subsequent cytotoxicity. In vivo, intraperitoneal (i.p.) injection of KU-0060648 significantly inhibited HepG2 xenograft growth in nude mice. AKT-mTOR activation was also inhibited in xenografted tumors. Finally, we showed that DNA-PKcs expression was significantly upregulated in human HCC tissues. Yet miRNA-101, an anti-DNA-PKcs miRNA, was downregulated. Over-expression of miR-101 in HepG2 cells inhibited DNA-PKcs expression and cell proliferation. Together, these results indicate that KU-0060648 inhibits HCC cells through DNA-PKcs-dependent and -independent mechanisms.


Assuntos
Antineoplásicos/farmacologia , Carcinoma Hepatocelular/patologia , Cromonas/farmacologia , Neoplasias Hepáticas/patologia , Tiofenos/farmacologia , Animais , Apoptose/efeitos dos fármacos , Carcinoma Hepatocelular/metabolismo , Proliferação de Células/efeitos dos fármacos , Proteína Quinase Ativada por DNA/metabolismo , Células Hep G2 , Humanos , Neoplasias Hepáticas/metabolismo , Camundongos , Camundongos Nus , MicroRNAs/metabolismo , Proteínas Nucleares/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Tumour Biol ; 37(3): 3025-31, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26419594

RESUMO

Bone morphogenetic protein 10 (BMP10), a novel member of BMP family, has been identified as an important regulator for angiogenesis. Dysregulation of BMP has been observed in several cancer types. However, its roles in gastric cancer (GC) remain unknown. In this study, the expression of BMP10 was found to be down-regulated in GC samples. Forced expression of BMP10 in GC cells inhibited its growth and migration, while knocking down the expression of BMP10 in GC cells promoted cell growth, migration, and metastasis. BMP10 was shown to negatively regulated beta-catenin/TCF signaling by up-regulating Axin protein level. Taken together, the present study revealed the suppressive function of BMP10 in gastric cancer.


Assuntos
Proteínas Morfogenéticas Ósseas/fisiologia , Movimento Celular , Proliferação de Células , Neoplasias Gástricas/patologia , Proteínas Morfogenéticas Ósseas/análise , Linhagem Celular Tumoral , Humanos , Metástase Neoplásica , Transdução de Sinais/fisiologia , Fatores de Transcrição TCF/fisiologia , beta Catenina/fisiologia
9.
Tumour Biol ; 37(2): 2621-7, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26392113

RESUMO

Hepatocyte cell adhesion molecule (HEPACAM), a member of immunoglobulin superfamily, is an adhesion molecule. Although dysregulation of several adhesion molecules has been implicated in the progression of non-small cell lung cancer (NSCLC), the expression profile and functions of HEPACAM in NSCLC remains unknown. In this study, it was found that the expression of HEPACAM was downregulated in NSCLC tissues. Forced expression of HEPACAM in NSCLC cells inhibited the growth and migration of the cancer cells, while knocking down the expression of HEPACAM promoted cell growth, migration, and metastasis. In the molecular mechanism study, HEPACAM was found to be a negative regulator of beta-catenin/TCF signaling. Taken together, this study revealed the suppressive roles of HEPACAM in NSCLC and restoring the function of HEPACAM in NSCLC might be a promising strategy for the therapy.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/patologia , Movimento Celular/genética , Proliferação de Células/genética , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Proteínas/genética , Moléculas de Adesão Celular/genética , Proteínas de Ciclo Celular , Linhagem Celular , Linhagem Celular Tumoral , Progressão da Doença , Regulação para Baixo/genética , Regulação Neoplásica da Expressão Gênica/genética , Células HEK293 , Humanos , Transdução de Sinais/genética , beta Catenina/genética
10.
Tumour Biol ; 35(6): 5481-6, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24532432

RESUMO

Altered expression of transmembrane protease/serine 4 (TMPRSS4) is observed in various types of human cancers. However, the clinical significance of TMPRSS4 expression in gallbladder cancer (GBC) remains largely unknown. The present study aims to explore the clinicopathological significance and prognostic value of TMPRSS4 in GBC. The levels of TMPRSS4 mRNA and protein in GBC tissues and adjacent noncancerous tissues were evaluated by quantitative reverse-transcriptase polymerase chain reaction and immunohistochemistry. To investigate the correlations between TMPRSS4 and the clinicopathological features of GBC, the expression of TMPRSS4 in 97 patients with GBC were detected by immunohistochemistry. The correlation of TMPRSS4 expression with patients' survival rate was assessed by Kaplan-Meier and Cox regression. Our results showed that the expression levels of TMPRSS4 mRNA and protein in GBC tissues were both significantly higher than those in adjacent noncancerous tissues. Immunohistochemical staining revealed that high TMPRSS4 expression was closely correlated with tumor size (P=0.032), histological grade (P=0.002), pathologic T stage (P=0.005), clinical stage (P=0.013), and lymph node metastasis (P=0.003). Moreover, the results of Kaplan-Meier analysis indicated that a high expression level of TMPRSS4 resulted in a significantly poor prognosis of GBC patients. Multivariate analysis showed that the status of TMPRSS4 expression was an independent prognostic factor for GBC patients. Our results showed that TMPRSS4 plays a key role in GBC and therefore may provide an opportunity for developing a novel therapeutic target as well as a prognostic marker in GBC.


Assuntos
Neoplasias da Vesícula Biliar/patologia , Proteínas de Membrana/fisiologia , Serina Endopeptidases/fisiologia , Adulto , Idoso , Feminino , Neoplasias da Vesícula Biliar/mortalidade , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Proteínas de Membrana/análise , Proteínas de Membrana/genética , Pessoa de Meia-Idade , Estadiamento de Neoplasias , RNA Mensageiro/análise , Serina Endopeptidases/análise , Serina Endopeptidases/genética , Taxa de Sobrevida
11.
Lab Invest ; 93(7): 844-54, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23752129

RESUMO

M2-polarized tumor-associated macrophages (TAMs) are key regulators of the link between inflammation and cancer. A negative correlation between infiltration intensity of M2-polarized TAMs and prognosis of pancreatic cancer has been reported. Epithelial-mesenchymal transition (EMT) is an important biological process in the progression of primary tumors toward metastasis. Inflammation-induced EMT has been previously shown, therefore, we hypothesized M2-polarized TAMs could induce EMT in pancreatic cancer. Toll-like receptor 4 (TLR4) signaling has an active role in tumor progression during chronic inflammation and the receptor is primarily expressed on macrophages. Activation of TLR4 on M2-polarized TAMs stimulates an increase in the cytokine interleukin-10 (IL-10); consequently, another aim was to investigate the potential role of TLR4/IL-10 signaling in the EMT of pancreatic cancer. Treatment with IL-4 (20 ng/ml) for 24 h successfully induced the polarization of macrophage cell line RAW 264.7 to M2 phenotype, IL-10(high), IL-12(low), and IL-23(low), and high expression of CD204 and CD206. A coculture system allowed investigation of the roles of M2-polarized TAMs and TLR4/IL-10 signaling in the EMT of Panc-1 and BxPC-3 pancreatic cancer cell lines. Our results showed that coculture with M2-polarized TAMs increased fibroblastic morphology, upregulated mesenchymal markers vimentin and snail at the mRNA and protein levels, and increased proliferation, migration, and metalloproteinase (MMP)2 and MMP9 proteolytic activity in pancreatic cancer cells. Simultaneously, coculture with M2-polarized TAMs decreased the expression of the epithelial marker E-cadherin. Coculture with pancreatic cancer cells increased TLR4 mRNA and protein expression in M2-polarized TAMs. Application of TLR4 siRNA and neutralizing antibodies against TLR4 and IL-10 markedly inhibited E-cadherin reduction and the upregulation of snail and vimentin. Furthermore, activation of TLR4 signaling by lipopolysaccharide profoundly increased the EMT of pancreatic cancer cells. In conclusion, M2-polarized TAMs promoted EMT in pancreatic cancer cells partially through TLR4/IL-10 signaling, suggesting novel therapeutic strategies and enhancing our understanding of M2-polarized TAMs.


Assuntos
Adenocarcinoma/patologia , Transição Epitelial-Mesenquimal , Interleucina-10/metabolismo , Macrófagos/fisiologia , Neoplasias Pancreáticas/patologia , Receptor 4 Toll-Like/metabolismo , Adenocarcinoma/imunologia , Adenocarcinoma/metabolismo , Animais , Biomarcadores/metabolismo , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Técnicas de Cocultura , Fibroblastos/patologia , Humanos , Neoplasias Pancreáticas/imunologia , Neoplasias Pancreáticas/metabolismo , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA