Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Oncol Rep ; 33(1): 239-49, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25370413

ABSTRACT

The concurrent administration of chemotherapy and epidermal growth factor receptor­tyrosine kinase inhibitors (EGFR­TKIs) has previously produced a negative interaction and failed to confer a survival benefit to non­small cell lung cancer (NSCLC) patients compared with first­line cytotoxic chemotherapy. The present study aimed to investigate the optimal schedule of the combined treatment of cisplatin/paclitaxel and icotinib in NSCLC cell lines and clarify the underlying mechanisms. HCC827, H1975, H1299 and A549 human NSCLC cell lines with wild­type and mutant EGFR genes were used as in vitro models to define the differential effects of various schedules of cisplatin/paclitaxel with icotinib treatments on cell growth, proliferation, cell cycle distribution, apoptosis, and EGFR signaling pathway. Sequence­dependent antiproliferative effects differed among the four NSCLC cell lines, and were not associated with EGFR mutation, constitutive expression levels of EGFR or downstream signaling molecules. The antiproliferative effect of cisplatin plus paclitaxel followed by icotinib was superior to that of cisplatin or paclitaxel followed by icotinib in the HCC827, H1975, H1299 and A549 cell lines, and induced more cell apoptosis and G0/G1 phase arrest. Cisplatin and paclitaxel significantly increased the expression of EGFR phosphorylation in the HCC827 cell line. However, only paclitaxel increased the expression of EGFR phosphorylation in the H1975 cell line. Cisplatin/paclitaxel followed by icotinib influenced the expression of p­EGFR and p­AKT, although the expression of p­ERK1/2 remained unchanged. The results suggest that the optimal schedule of the combined treatment of cisplatin/paclitaxel and icotinib differed among the NSCLC cell lines. The results also provide molecular evidence to support clinical treatment strategies for NSCLC patients.


Subject(s)
Antineoplastic Agents/pharmacology , Cisplatin/pharmacology , Crown Ethers/pharmacology , Paclitaxel/pharmacology , Quinazolines/pharmacology , Apoptosis , Base Sequence , Carcinoma, Non-Small-Cell Lung , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , DNA Mutational Analysis , Drug Resistance, Neoplasm , Drug Screening Assays, Antitumor , Drug Synergism , ErbB Receptors/genetics , ErbB Receptors/metabolism , Humans , Inhibitory Concentration 50 , Lung Neoplasms , Resting Phase, Cell Cycle , Signal Transduction
2.
Oncol Rep ; 31(6): 2569-78, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24700316

ABSTRACT

Recent studies have demonstrated that SMG-1, a newly characterized member of the family of phosphatidylinositol 3-kinase-related protein kinases (PIKKs), is involved in tumorigenesis as a new tumor suppressor. However, its expression and significance in hepatocellular carcinoma (HCC) remain obscure. The present study investigated SMG-1 expression in HCC tissue specimens, aimed at defining the association with clinicopathological significance. Both immunohistochemistry and qRT-PCR were employed to analyze SMG-1 expression in 157 HCC and corresponding distant normal tissue specimens. The results revealed that expression of SMG-1 was significantly lower in the HCC tissue specimens than that in the distant normal tissues. Moreover, a lower expression level of SMG-1 was significantly correlated with serum α-fetoprotein level (P=0.001), poorly differentiated tumors (P=0.009) and more advanced TNM stage (P<0.001). Further study showed that SMG-1 expression was exactly associated with tumor differentiation and clinical stage in HCC. Kaplan-Meier analysis indicated that low SMG-1 expression was related to poor overall survival, and the prognostic impact of SMG-1 was further confirmed by stratified survival analysis. Importantly, multivariate analysis revealed that low SMG-1 expression was an independent prognostic marker for an unfavorable overall survival. We conclude that SMG-1 is downregulated in HCC and may represent a promising biomarker for predicting the prognosis of HCC, including the prognosis of early-stage patients.


Subject(s)
Carcinoma, Hepatocellular/genetics , Liver Neoplasms/genetics , Phosphatidylinositol 3-Kinases/biosynthesis , Tumor Suppressor Proteins/biosynthesis , Adult , Aged , Carcinoma, Hepatocellular/pathology , Female , Gene Expression Regulation, Neoplastic , Humans , Kaplan-Meier Estimate , Liver Neoplasms/pathology , Male , Middle Aged , Neoplasm Staging , Phosphatidylinositol 3-Kinases/genetics , Prognosis , Protein Serine-Threonine Kinases , Tumor Suppressor Proteins/genetics
3.
Onco Targets Ther ; 7: 353-63, 2014.
Article in English | MEDLINE | ID: mdl-24591842

ABSTRACT

PURPOSE: Celecoxib, an inhibitor of cyclooxygenase-2 (COX2), was investigated for enhancement of chemotherapeutic efficacy in cancer clinical trials. This study aimed to determine whether celecoxib combined with 5-fluorouracil or sorafenib or gefitinib is beneficial in HepG2 multicellular spheroids (MCSs), as well as elucidate the underlying mechanisms. METHODS: The human hepatocellular carcinoma cell line HepG2 MCSs were used as in vitro models to investigate the effects of celecoxib combined with 5-fluorouracil or sorafenib or gefitinib treatment on cell growth, apoptosis, and signaling pathway. RESULTS: MCSs showed resistance to drugs compared with monolayer cells. Celecoxib combined with 5-fluorouracil or sorafenib exhibited a synergistic action. Exposure to celecoxib (21.8 µmol/L) plus 5-fluorouracil (8.1 × 10(-3) g/L) or sorafenib (4.4 µmol/L) increased apoptosis but exerted no effect on COX2, phosphorylated epidermal growth-factor receptor (p-EGFR) and phosphorylated (p)-AKT expression. Gefitinib (5 µmol/L), which exhibits no growth-inhibition activity as a single agent, increased the inhibitory effect of celecoxib. Gefitinib (5 µmol/L) plus celecoxib (21.8 µmol/L) increased apoptosis. COX2, p-EGFR, and p-AKT were inhibited. CONCLUSION: Celecoxib combined with 5-fluorouracil or sorafenib or gefitinib may be superior to single-agent therapy in HepG2 MCSs. Our results provided molecular evidence to support celecoxib combination-treatment strategies for patients with human hepatocellular carcinoma. MCSs provided a good model to evaluate the interaction of anticancer drugs.

4.
Dis Markers ; 35(5): 395-405, 2013.
Article in English | MEDLINE | ID: mdl-24223455

ABSTRACT

BACKGROUND/OBJECTIVE: We aimed to examine the expression of lymphoid enhancer factor 1 (LEF1) and Notch2 in colorectal cancer (CRC) and their association with clinicopathologic variables and CRC patients' prognosis. METHODS: Immunohistochemistry, quantitative real-time polymerase chain reaction (qRT-PCR), and Western blot analysis were performed to assess the expression of LEF1 and Notch2 in 184 patients with CRC. RESULTS: We observed a strong negative correlation between LEF1 expression and Notch2 expression (P < 0.001). Both LEF1 mRNA and protein expression increased while the Notch2 mRNA and protein expression decreased in tumor specimens compared with the matched paratumorous normal tissue (P < 0.001). An increase in LEF1 protein expression was significantly associated with lymph node metastases, distant metastasis, advanced TNM (tumor-node-metastasis) stage, and shorter overall survival. A decrease in Notch2 protein expression was associated with poorly differentiated tumors, lymph node metastases, distant metastasis, advanced TNM stage, and shorter overall survival of patients. In the multivariate Cox regression analysis, the LEF1 protein expression (P < 0.001), Notch2 protein expression (P < 0.001), TNM stage (P < 0.001), and the combination of increased LEF1 protein coexpression and decreased Notch2 protein coexpression (P < 0.001) were found to be independent prognostic indicators in CRC. CONCLUSION: Our results suggest that increased LEF1 coexpression and decreased Notch2 coexpression represent a risk factor for poor overall survival of CRC patients.


Subject(s)
Biomarkers, Tumor/genetics , Carcinoma/genetics , Colorectal Neoplasms/genetics , Lymphoid Enhancer-Binding Factor 1/genetics , RNA, Messenger/genetics , Receptor, Notch2/genetics , Adult , Aged , Biomarkers, Tumor/metabolism , Carcinoma/diagnosis , Carcinoma/metabolism , Colorectal Neoplasms/diagnosis , Colorectal Neoplasms/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , Lymphatic Metastasis , Lymphoid Enhancer-Binding Factor 1/metabolism , Male , Middle Aged , Prognosis , RNA, Messenger/metabolism , Receptor, Notch2/metabolism , Transcription, Genetic
5.
PLoS One ; 8(10): e76596, 2013.
Article in English | MEDLINE | ID: mdl-24098538

ABSTRACT

Expression of lymphoid enhancer factor 1 (LEF1) is frequently altered in different human cancers. This study aimed to assess LEF1 expression in colon cancer tissues and to explore changed phenotypes, gene expressions, and the possible mechanism after knocked down LEF1 expression in colon cancer cell lines. A total of 106 colon cancer and matched paratumorous normal tissues were used to assess LEF1 expression using immunohistochemistry and qRT-PCR. LEF1 lentivirus was used to knockdown LEF1 expression for the assessment of cell viability, cell cycle distribution, apoptosis, and gene expressions. The nude mouse xenograft assay was performed to detect the effects of LEF1 knockdown in vivo. The data showed that the levels of LEF1 mRNA and protein were significantly increased in human colon cancer tissues compared to the matched paratumorous normal tissues and were associated with infiltration depth, lymph node and distant metastases, advanced TNM (tumor-node-metastasis) stages, and shorter overall survival. Furthermore, LEF1 knockdown reduced tumor cell viability, invasion capacity, MMP2 and MMP-9 expression, but induced apoptosis. Nude mouse xenograft assay showed that LEF1 knockdown suppressed tumor formation and growth in vivo. In addition, the expression of Notch pathway-related proteins RBP-jκ and Hes1 was reduced in LEF1 knockdown cells. Taken together, LEF1 protein was overexpressed in colon cancer tissues and knockdown of LEF1 expression inhibited colon cancer growth in vitro and in vivo. These data suggest that targeting of LEF1 expression should be further evaluated for colon cancer prevention and therapy.


Subject(s)
Colonic Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Lymphoid Enhancer-Binding Factor 1/genetics , Adult , Aged , Aged, 80 and over , Animals , Apoptosis/genetics , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Female , Genetic Vectors , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Lentivirus/genetics , Lymphatic Metastasis , Lymphoid Enhancer-Binding Factor 1/antagonists & inhibitors , Lymphoid Enhancer-Binding Factor 1/metabolism , Male , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Nude , Middle Aged , Neoplasm Transplantation , Signal Transduction , Transcription Factor HES-1
6.
Int J Biochem Cell Biol ; 45(11): 2666-78, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24004834

ABSTRACT

Infection with Helicobacter pylori is important in the development and progression of gastric cancer. However, the mechanisms that regulate this activation in gastric tumors remain elusive. CACUL1 has been cloned and identified as a novel gene that is expressed in many types of cancer and is involved in cell cycle regulation and tumor growth. The current study aimed to examine the expression of CACUL1 in gastric cancer samples and analyze its correlation with H. pylori infection. We found that CACUL1 was highly expressed in gastric cancer tissues and negatively correlated with gastric cancer differentiation and TNM stage. In addition, CACUL1 expression was high in H. pylori-infected tissues compared with H. pylori non-infected tissue. We found that H. pylori could up-regulate CACUL1 expression through activating protein 1. The up-regulation of CACUL1 expression could promote matrix metalloproteinase 9 and Slug expression to increase invasion and metastasis of tumor cells. These results suggested that H. pylori-triggered CACUL1 production occurred in an activating protein 1-dependent manner and regulated matrix metalloproteinase 9 and Slug expression to affect the invasion and metastasis of tumor cells. Therefore, CACUL1 is a potential therapeutic target for the treatment of aggressive gastric cancer.


Subject(s)
Cullin Proteins/genetics , Helicobacter pylori/physiology , Stomach Neoplasms/microbiology , Stomach Neoplasms/pathology , Transcription Factor AP-1/metabolism , Up-Regulation , Adult , Aged , Aged, 80 and over , Animals , Anthracenes/pharmacology , Cell Line, Tumor , Cullin Proteins/metabolism , Curcumin/pharmacology , Female , Gastric Mucosa/metabolism , Gene Expression Regulation, Neoplastic , Humans , Male , Matrix Metalloproteinase 9/metabolism , Mice , Middle Aged , Neoplasm Invasiveness , Neoplasm Metastasis , Phosphorylation , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins c-fos/metabolism , Stomach/pathology , Stomach Neoplasms/enzymology , Stomach Neoplasms/genetics , Transcriptional Activation/genetics , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...