Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Hua Xi Kou Qiang Yi Xue Za Zhi ; 37(1): 25-30, 2019 Feb 01.
Article in Chinese | MEDLINE | ID: mdl-30854814

ABSTRACT

OBJECTIVE: This study aims to establish an effective and stable periodontal ligament cell line stably expressing human telomerase reverse transcriptase (hTERT) gene by using the adenovirus method. METHODS: Polymerase chain reaction (PCR) was used to amplify the full length of hTERT gene to construct recombinant adenovirus plasmid pAd-pshuttle-cmv-hTERT. Packaged adenovirus particles were used for infection of human periodontal ligament cells. The expression levels of hTERT and osteogenic genes, such as alkaline phosphatase, Runt-related transcription factor 2, bone sialoprotein, osteocalcin, osteopontin, and collagen Ⅰ mRNA, were detected by quantitative real-time PCR (qRT-PCR). The ability of osteogenic differentiation was observed by alizarin red staining, and the cell proliferation was determined by CCK-8. RESULTS: Adenovirus particles containing the hTERT gene were successfully constructed and infected with periodontal ligament cells. The infected cells were similar to normal periodontal ligament cells. The qRT-PCR results showed that hTERT and osteogenesis-associated genes were highly expressed in the periodontal ligament cell lines constructed by adenoviruses. Alizarin red staining showed that the periodontal ligament cell line had strong osteogenic differentiation capability. CCK-8 showed that the periodontal ligament cell line had strong proliferation capability. CONCLUSIONS: The human periodontal ligament cell line with high efficiency and stable expression of hTERT was established by the adenovirus method, thereby providing an ideal cell line for studying the mechanism of periodontal regeneration.


Subject(s)
Periodontal Ligament , Telomerase , Adenoviridae , Alkaline Phosphatase , Cell Differentiation , Cell Line , Cell Proliferation , Humans , Osteogenesis
3.
PLoS One ; 12(2): e0172470, 2017.
Article in English | MEDLINE | ID: mdl-28231299

ABSTRACT

The present study was aimed to unravel the inhibitory mechanisms of curcumin for lung cancer metastasis via constructing a miRNA-transcription factor (TF)-target gene network. Differentially expressed miRNAs between human high-metastatic non-small cell lung cancer 95D cells treated with and without curcumin were identified using a TaqMan human miRNA array followed by real-time PCR, out of which, the top 6 miRNAs (miR-302b-3p, miR-335-5p, miR-338-3p, miR-34c-5p, miR-29c-3p and miR-34a-35p) with more verified target genes and TFs than other miRNAs as confirmed by a literature review were selected for further analysis. The miRecords database was utilized to predict the target genes of these 6 miRNAs, TFs of which were identified based on the TRANSFAC database. The findings of the above procedure were used to construct a miRNA-TF-target gene network, among which miR-34a-5p, miR-34c-5p and miR-302b-3p seemed to regulate CCND1, WNT1 and MYC to be involved in Wnt signaling pathway through the LEF1 transcription factor. Therefore, we suggest miR-34a-5p/miR-34c-5p/miR-302b-3p -LEF1-CCND1/WNT1/MYC axis may be a crucial mechanism in inhibition of lung cancer metastasis by curcumin.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Curcumin/pharmacology , Gene Regulatory Networks/drug effects , Lung Neoplasms/drug therapy , MicroRNAs/genetics , Neoplasm Invasiveness/prevention & control , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cell Movement/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lung/drug effects , Lung/pathology , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology
4.
Oncotarget ; 7(14): 18247-61, 2016 Apr 05.
Article in English | MEDLINE | ID: mdl-26919096

ABSTRACT

MiR-206 is low expression in lung cancers and associated with cancer metastasis. However, the roles of miR-206 in epithelial-mesenchymal transition (EMT) and angiogenesis in lung cancer remain unknown. In this study, we find that hepatocyte growth factor (HGF) induces EMT, invasion and migration in A549 and 95D lung cancer cells, and these processes could be markedly inhibited by miR-206 overexpression. Moreover, we demonstrate that miR-206 directly targets c-Met and inhibits its downstream PI3k/Akt/mTOR signaling pathway. In contrast, miR-206 inhibitors promote the expression of c-Met and activate the PI3k/Akt/mTOR signaling, and this effect could be attenuated by the PI3K inhibitor. Moreover, c-Met overexpression assay further confirms the significant inhibitory effect of miR-206 on HGF-induced EMT, cell migration and invasion. Notably, we also find that miR-206 effectively inhibits HGF-induced tube formation and migration of human umbilical vein endothelial cells (HUVECs), and the mechanism is also related to inhibition of PI3k/Akt/mTOR signaling. Finally, we reveal the inhibitory effect of miR-206 on EMT and angiogenesis in xenograft tumor mice model. Taken together, miR-206 inhibits HGF-induced EMT and angiogenesis in lung cancer by suppressing c-Met/PI3k/Akt/mTOR signaling. Therefore, miR-206 might be a potential target for the therapeutic strategy against EMT and angiogenesis of lung cancer.


Subject(s)
Carcinoma, Non-Small-Cell Lung/pathology , Epithelial-Mesenchymal Transition/genetics , Hepatocyte Growth Factor/metabolism , Lung Neoplasms/pathology , MicroRNAs/genetics , Neovascularization, Pathologic/pathology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-met/metabolism , TOR Serine-Threonine Kinases/metabolism , A549 Cells , Animals , Cell Line, Tumor , Cell Movement/physiology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Lung/blood supply , Lung/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/metabolism , Neoplasm Invasiveness/pathology , Neoplasm Transplantation , Phosphoinositide-3 Kinase Inhibitors , Signal Transduction , Transplantation, Heterologous
5.
Mol Biosyst ; 11(8): 2290-302, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26075299

ABSTRACT

MiRNAs associated with the metastasis of lung cancer remain largely unexplored. In this study, gene and miRNA expression profiling were performed to analyze the global expression of mRNAs and miRNAs in human high- and low-metastatic lung cancer cell strains. By developing an integrated bioinformatics analysis, six miRNAs (miR-424-3p, miR-450b-5p, miR-335-5p, miR-34a-5p, miR-302b-3p and miR-206) showed higher target gene degrees in the miRNA-gene network and might be potential metastasis-related miRNAs. Using the qRT-PCR method, the six miRNAs were further confirmed to show a significant expression difference between human lung cancer and normal tissue samples. Since miR-206 showed lower expression both in lung cancer tissues and cell lines, it was used as an example for further functional verification. The wound healing assay and transwell invasion assay showed that miR-206 mimics significantly inhibited the cell migration and invasion of the high-metastatic lung cancer 95D cell strain. One of its predicted targets in our miRNA-gene network, MET, was also obviously decreased at the protein level when miR-206 was overexpressed. Instead, miR-206 inhibitors increased MET protein expression, cell migration and invasion of the low-metastatic lung cancer 95C cell strain. Meanwhile, the luciferase assay showed that MET was a direct target of miR-206. Furthermore, MET gene silence showed a similar anti-migration and anti-invasion effect with miR-206 mimics in 95D cells and could partially attenuate the migration- and invasion-promoting effect of miR-206 inhibitors in 95C cells, suggesting that miR-206 targets MET in lung cancer metastasis. Finally, we also demonstrated that miR-206 can significantly inhibit lung cancer proliferation and metastasis in mouse models. In conclusion, our study provided a miRNA-gene regulatory network in lung cancer metastasis and further demonstrated the roles of miR-206 and MET in this process, which enhances the understanding of the regulatory mechanism in lung cancer metastasis.


Subject(s)
Gene Regulatory Networks , Lung Neoplasms/genetics , MicroRNAs/biosynthesis , Neoplasm Metastasis/genetics , Proto-Oncogene Proteins c-met/biosynthesis , Adult , Aged , Animals , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/pathology , Male , Mice , MicroRNAs/genetics , Middle Aged , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Neoplasm Metastasis/pathology , Proto-Oncogene Proteins c-met/genetics , RNA, Messenger/genetics , Xenograft Model Antitumor Assays
6.
World J Gastroenterol ; 21(19): 6044-51, 2015 May 21.
Article in English | MEDLINE | ID: mdl-26019471

ABSTRACT

AIM: To compare the efficacy and safety of biological agents for the treatment of active ulcerative colitis (UC). METHODS: PubMed, MEDLINE, EMBASE and the Cochrane library were searched to screen relevant articles from January 1996 to August 2014. The mixed treatment comparison meta-analysis within a Bayesian framework was performed using WinBUGS14 software. The proportions of patients reaching clinical response, clinical remission and mucosal healing in induction and maintenance phases were analyzed as efficacy indicators. Serious adverse events in maintenance phase were analyzed as safety indicators. RESULTS: The meta-analysis results showed that biological agents achieved better clinical response, clinical remission and mucosal healing than placebo. Indirect comparison indicated that in induction phase, infliximab was more effective than adalimumab in inducing clinical response (OR = 0.41, 95%CI: 0.29-0.57), clinical remission (OR = 0.33, 95%CI: 0.19-0.56) and mucosal healing (OR = 0.33, 95%CI: 0.19-0.56), and golimumab in inducing clinical response (OR = 0.66, 95%CI: 0.39-2.33) and mucosal healing (OR = 2.15, 95%CI: 1.18-4.22). No significant difference was found between placebo and biological agents regarding their safety. CONCLUSION: All biological agents were superior to placebo for UC treatment in both induction and maintenance phases with a similar safety profile, and infliximab had a better clinical effect than the other biological agents.


Subject(s)
Biological Products/therapeutic use , Colitis, Ulcerative/drug therapy , Colon/drug effects , Immunosuppressive Agents/therapeutic use , Infliximab/therapeutic use , Intestinal Mucosa/drug effects , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Adult , Biological Products/adverse effects , Colitis, Ulcerative/diagnosis , Colitis, Ulcerative/immunology , Colon/immunology , Colon/pathology , Humans , Immunosuppressive Agents/adverse effects , Infliximab/adverse effects , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Middle Aged , Odds Ratio , Remission Induction , Risk Factors , Time Factors , Treatment Outcome , Wound Healing/drug effects
7.
Mol Biosyst ; 11(3): 859-68, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25578635

ABSTRACT

A microarray analysis of differential genes by curcumin treatment was performed and the crucial pathways associated with non-small cell lung cancer (NSCLC) were investigated. Total RNAs from 0, 10 or 20 µM curcumin treated NSCLC 95D cells were used to prepare microarray chips. The differentially expressed genes (DEGs) were identified using the RankProducts package and their function was predicted by DAVID and gene set enrichment analysis. The pathway crosstalk was analyzed by mapping the gene expression profiles into protein-protein interaction databases. Validation of the microarray results was performed by cell viability, cell migration and western blot analyses. A total of 486 (10 µM) and 264 (20 µM) DEGs were screened between the 95D cells in the presence and absence of curcumin. Function enrichment analysis indicated the DEGs were mainly involved in the steroid biosynthetic process and regulation of autophagy. Pathway crosstalk analysis suggested there was a significant interaction between NSCLC and adherens junctions (or Wnt signaling pathways, which are important for cancer cell proliferation and invasion) in both 10 µM and 20 µM curcumin treated 95D cells. Furthermore, early growth response (EGR-1) was demonstrated to regulate the crosstalk between adherens junctions and Wnt signaling pathways, indicating that EGR-1 may also regulate cell proliferation and migration. This hypothesis was validated by in vitro experiments: EGR-1 was decreased after curcumin treatment. Curcumin exhibited a significant anti-proliferation and anti-migration activity in NSCLC 95D cells, possibly by steering the crosstalk between the Wnt signaling pathway and adherens junction via EGR-1.


Subject(s)
Adherens Junctions/metabolism , Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/metabolism , Curcumin/pharmacology , Early Growth Response Protein 1/metabolism , Lung Neoplasms/metabolism , Wnt Signaling Pathway/drug effects , Carcinoma, Non-Small-Cell Lung/genetics , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Computational Biology , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Gene Regulatory Networks , Humans , Lung Neoplasms/genetics
8.
J Nutr Biochem ; 25(2): 177-85, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24445042

ABSTRACT

Curcumin, a natural and crystalline compound isolated from the plant Curcuma longa with low toxicity in normal cells, has been shown to protect against carcinogenesis and prevent tumor development. However, little is known about antimetastasis effects and mechanism of curcumin in lung cancer. Rac1 is an important small Rho GTPases family protein and has been widely implicated in cytoskeleton rearrangements and cancer cell migration, invasion and metastasis. In this study, we examined the influence of curcumin on in vitro invasiveness of human lung cancer cells and the expressions of Rac1. The results indicate that curcumin at 10 µM slightly reduced the proliferation of 801D lung cancer cells but showed an obvious inhibitory effect on epidermal growth factor or transforming growth factor ß1-induced lung cancer cell migration and invasion. Meanwhile, we demonstrated that the suppression of invasiveness correlated with inhibition of Rac1/PAK1 signaling pathways and matrix metalloproteinase (MMP) 2 and 9 protein expression by combining curcumin treatment with the methods of Rac1 gene silence and overexpression in lung cancer cells. Laser confocal microscope also showed that Rac1-regulated actin cytoskeleton rearrangement may be involved in anti-invasion effect of curcumin on lung cancer cell. At last, through xenograft experiments, we confirmed the connection between Rac1 and the growth and metastasis inhibitory effect of curcumin in vivo. In summary, these data demonstrated that low-toxic levels of curcumin could efficiently inhibit migration and invasion of lung cancer cells through inhibition of Rac1/PAK1 signaling pathway and MMP-2 and MMP-9 expression, which provided a novel insight into the molecular mechanism of curcumin against lung cancer.


Subject(s)
Cell Movement , Curcumin/pharmacology , Lung Neoplasms/pathology , Neoplasm Invasiveness , Signal Transduction , rac1 GTP-Binding Protein/metabolism , Cell Line, Tumor , Humans , Phosphorylation , p21-Activated Kinases/metabolism
9.
Mol Biosyst ; 9(12): 3080-90, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24077187

ABSTRACT

Metastasis is a common feature of lung cancer, involving relationships between genes, proteins and miRNAs. However, lack of early detection and limited options for targeted therapies are weaknesses that cantribute to the dismal statistics observed in lung cancer metastasis. In this paper, gene expression profiling analysis for genes differentially expressed between high- (95D) and low-metastatic lung cancer cell lines (95C) was performed using gene annotation, pathway analysis, literature mining, and the integrated regulatory network as well as motif analysis of miRNA-DEG and TF-DEG. In addition, the expression of EGR-1 (early growth reponse-1) in surgically resected lung squamous carcinomas, adenocarcinomas and normal lung tissue was detected by immunohistochemistry to reveal the relationships between EGR-1 and lung cancer metastasis. A total of 570 different expressed genes (DEGs) were screened, the vast majority of up-regulated DEGs were connected to cell adhesion and focal adhesion. EGR-1 was observed in the center node of the regulatory network, which seems to play a role in the process of cancer metastasis, and further immunohistochemistry detection confirmed this reasoning. Besides EGR-1, several significant module-related DEGs were enriched in the pathway within cancer and focal adhesion according to KEGG pathway enrichment analysis of network modules. The construction of an integrated regulatory network and the functional prediction of EGR-1 provided us with the cytological basis of lung cancer metastasis research and an understanding of the mechanism of metastasis in lung cancer. EGR-1 should be considered as a potential target gene in therapeutic agent for lung cancer metastasis.


Subject(s)
Early Growth Response Protein 1/genetics , Early Growth Response Protein 1/metabolism , Giant Cell Tumors/genetics , Lung Neoplasms/genetics , Neoplasm Metastasis/genetics , Cell Line, Tumor , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Humans , MicroRNAs/genetics , Signal Transduction
10.
Zhonghua Zhong Liu Za Zhi ; 34(6): 436-40, 2012 Jun.
Article in Chinese | MEDLINE | ID: mdl-22967445

ABSTRACT

OBJECTIVE: To explore the expression of ezrin protein in human non-small cell lung cancer (NSCLC) tissues and lung cancer cell lines, and the association between the expression of ezrin protein and the expression of E-cadherin and CD44V6 proteins. METHODS: The expression of ezrin protein and mRNA in lung cancer cell lines was detected by RT-PCR and Western blotting. Ezrin, E-cadherin and CD44V6 were detected by immunohistochemical SP staining in tumor tissues from 150 lung cancer cases and in adjacent normal lung tissues from 30 patients. Furthermore, the expression of ezrin in 30 freshly-taken NSCLC tissues was also detected by Western blot. RESULTS: The expression of ezrin protein and mRNA was up-regulated in highly metastatic human lung cancer. The positive rate of ezrin, E-cadherin and CD44V6 expression in the lung cancer was 61.3%, 54.0% and 58.7%, respectively. The up-regulation of ezrin expression was significantly correlated with lymph node metastasis, but not correlated with age, sex, tumor size, histological type, clinical TNM system and pathological grade. Western blot analysis showed that the level of ezrin in the NSCLC tissues was significantly higher than that in the normal tissues (t = 5.013, P < 0.01). Survival analysis showed that the 5-year survival rate of patients with negative ezrin expression was 29.3%, significantly higher than that of patients with positive ezrin expression (15.2%, χ(2) = 4.128, P = 0.042). Multivariate Cox regression analysis showed that ezrin expression (RR = 3.012, P = 0.047) and lymph node metastasis (RR = 4.827, P = 0.035) were significantly independent prognostic factors for patients with lung cancer. Furthermore, a negative correlation was observed between the expressions of ezrin and E-cadherin in lung cancer, and a positive correlation between the expressions of ezrin and CD44V6 in lung cancer. CONCLUSIONS: Ezrin, E-cadherin and CD44V6 play an important role in the regulation of growth and meastasis of lung cancer. Combined detection of ezrin, E-cadherin and CD44V6 expression is helpful in evaluating the metastasis and prognosis of non-small cell lung cancer.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cytoskeletal Proteins/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Adult , Aged , Antigens, CD , Cadherins/metabolism , Cell Line, Tumor , Cytoskeletal Proteins/genetics , Female , Humans , Hyaluronan Receptors/metabolism , Lymphatic Metastasis , Male , Middle Aged , RNA, Messenger/metabolism , Survival Rate , Up-Regulation
11.
Zhonghua Zhong Liu Za Zhi ; 34(1): 21-5, 2012 Jan.
Article in Chinese | MEDLINE | ID: mdl-22490850

ABSTRACT

OBJECTIVE: To investigate the relationship between p16 expression and cell proliferation and prognosis in gastric cancer patients. METHODS: Gastric cancer cell lines SGC-7901, MKN45, MKN28, human embryonic kidney cell line HEK293, human fibroblast cell line MRC-5, and surgical specimens of gastric carcinoma and adjacent normal gastric mucosa from 65 patients were included in this study. RT-PCR, MTT and FCM assays were used to detect p16 expression in gastric cancer cell lines and surgical specimens of gastric cancer. MTT assay was used to determine cancer cell viability and FCM to detect cell cycle. Kaplan-Meier survival curve and Log-Rank statistics were used to analyze the relationship between p16 expression and survival of petients with gastric cancer. RESULTS: Gastric cancer cell lines were mostly negative for p16 expression, and p16 was re-expressed after the cells transfected with p16 gene by adenovirus AdCMV-p16. p16 re-expression resulted in the decrease of cancer cell viability and cancer cell cycle arrest with increased G(1) phase and decreased S phase. p16 expression in cancer specimens was 32.3% (21/65), significantly lower than the 81.5% (53/65) in normal mucosa (χ(2) = 32.124, P < 0.001). The disease-free survival was significantly shorter in p16-negative patients than that in p16-positive patients (P < 0.01), but not the overall survival (P > 0.05). p16 expression was significantly correlated with differentiation and lymph node metastasis, but not significantly correlated with sex, age, tumor size or invasion depth of the gastric cancer. CONCLUSIONS: p16 gene is important for cancer cell proliferation. The inactivation gives cancer cells a high activity for proliferation and metastasis, and then influences the disease-free survival of gastric cancer patients.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p16/metabolism , Genes, p16 , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Adenoviridae/genetics , Adult , Aged , Cell Cycle Checkpoints , Cell Line, Tumor , Cell Proliferation , Cell Survival , Cyclin-Dependent Kinase Inhibitor p16/genetics , Disease-Free Survival , Female , Gene Expression Regulation, Neoplastic , Humans , Lymphatic Metastasis , Male , Middle Aged , Neoplasm Invasiveness , RNA, Messenger/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Survival Rate , Transfection
12.
Mol Cell Biochem ; 359(1-2): 389-98, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21874542

ABSTRACT

We previously reported that curcumin inhibited lung cancer A549 cells growth and promoted cell apoptosis in vitro. In this study, we further examined the apoptosis-related parameters, including lysosomal damage and cathepsin activation, in A549 cells exposed to curcumin. We found that curcumin caused lysosomal membrane permeabilization (LMP) and cytosolic relocation of cathepsin B (cath B) and cathepsin D (cath D). However, only Z-FA-fmk (a cath B inhibitor) but not pepstatin A (a cath D inhibitor) inhibited curcumin-induced cell apoptosis, mitochondrial membrane potential loss, and cytochrome c release. The antioxidant N-acetylcysteine and glutathione attenuated LMP, suggesting that lysosomal destabilization was dependent on the elevation of reactive oxygen species and which precedes mitochondrial dysfunction. These findings indicated a novel pathway for curcumin regulation of ROS-lysosomal-mitochondrial pathway and provided the key mechanism of regulation of LMP in cell apoptosis, which may be exploited for cancer treatment.


Subject(s)
Apoptosis/drug effects , Curcumin/pharmacology , Intracellular Membranes/drug effects , Lung Neoplasms/pathology , Lysosomes/ultrastructure , Antineoplastic Agents , Cathepsin B/metabolism , Cathepsin D/metabolism , Cell Line, Tumor , Curcumin/therapeutic use , Cytochromes c , Humans , Lung Neoplasms/drug therapy , Membrane Potential, Mitochondrial/drug effects , Mitochondria/metabolism , Permeability/drug effects , Reactive Oxygen Species
13.
Int J Mol Med ; 28(5): 769-76, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21837360

ABSTRACT

Rac1, an intracellular signal transducer, regulates a variety of cell functions, including the organization of the cytoskeleton, cell migration, and invasion. Overexpression of Rac1 has been reported in several human cancers. However, the underlying mechanisms are not well understood. In the present study, we evaluated the possibility of Rac1 as an appropriate molecular target for cancer gene therapy. The expression of Rac1 in 150 primary non-small cell lung cancer tissues (NSCLC) and 30 normal paraneoplastic lung tissues was determined by immunohistochemical staining, and the correlation of Rac1 overexpression with clinicopathological factors was evaluated. Overexpression of Rac1 was detected in 94 of 150 lung cancer specimens, the incidence rate being higher than that in normal lung tissue specimens. In addition, overexpression of Rac1 was also associated with poor differentiation, high TNM stage, and lymph node metastasis in NSCLC patients. Moreover, RNAi-mediated suppression of Rac1 expression reduced lamellipodia formation, migration and invasion potential of a lung cancer cell carcinoma cell line, 801D. Down-regulation of Rac1 expression also reduced the expression of Pak1. NSC23766, an inhibitor of Rac1 activity, could also inhibit lung cancer cell migration, invasion and induce rearrangements of the actin cytoskeleton. Furthermore, the suppression of Rac1 expression also sensitized cells to antitumor drugs. These results indicate that the overexpression of Rac1 is tightly associated with an aggressive phenotype of lung cancer cells. Therefore, we proposed that Rac1 could be a potential molecular target of gene therapy by RNAi-targeting in lung cancer cells.


Subject(s)
Actin Cytoskeleton/metabolism , Antineoplastic Agents/pharmacology , Cell Movement/drug effects , Lung Neoplasms/metabolism , rac1 GTP-Binding Protein/metabolism , Adult , Aged , Blotting, Western , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/drug effects , Female , Humans , Immunohistochemistry , In Vitro Techniques , Lung Neoplasms/genetics , Male , Microscopy, Confocal , Middle Aged , Organoplatinum Compounds/pharmacology , Young Adult , rac1 GTP-Binding Protein/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...