15 results on '"Xu-Yu Yang"'
Search Results
2. Inactivation of the Dlc1 Gene Cooperates with Downregulation of p15INK4b and p16Ink4a, Leading to Neoplastic Transformation and Poor Prognosis in Human Cancer
- Author
-
Brajendra K. Tripathi, Nicholas C. Popescu, Xu-Yu Yang, Lyra B. Olson, Douglas R. Lowy, William C. Vass, Dunrui Wang, Marian E. Durkin, and Xiaolan Qian
- Subjects
Male ,Cancer Research ,MAP Kinase Kinase 4 ,Colorectal cancer ,Cell ,Down-Regulation ,Article ,law.invention ,Mice ,Downregulation and upregulation ,Cyclin-dependent kinase ,law ,Neoplasms ,medicine ,Animals ,Humans ,Neoplastic transformation ,Gene Silencing ,Cyclin-Dependent Kinase Inhibitor p16 ,Cyclin-Dependent Kinase Inhibitor p15 ,rho-Associated Kinases ,biology ,business.industry ,Genes, p16 ,Tumor Suppressor Proteins ,GTPase-Activating Proteins ,Cyclin-Dependent Kinase 4 ,Cyclin-Dependent Kinase 6 ,Fibroblasts ,Prognosis ,medicine.disease ,Gene Expression Regulation, Neoplastic ,Mice, Inbred C57BL ,Transformation (genetics) ,Cell Transformation, Neoplastic ,Genes, ras ,medicine.anatomical_structure ,Oncology ,Immunology ,biology.protein ,Cancer research ,Suppressor ,Female ,DLC1 ,business - Abstract
The tumor suppressor gene deleted in liver cancer-1 (DLC1), which encodes a protein with strong RhoGAP (GTPase activating protein) activity and weak Cdc42GAP activity, is inactivated in various human malignancies. Following Dlc1 inactivation, mouse embryo fibroblasts (MEF) with a conditional Dlc1 knockout allele reproducibly underwent neoplastic transformation. In addition to inactivation of Dlc1 and increased activity of Rho and Cdc42, transformation depended on the subsequent decreased expression of the Cdk4/6 inhibitors p15Ink4b and p16Ink4a together with increased expression and activation of Cdk4/6. The level of expression of these cell-cycle regulatory genes was relevant to human tumors with low DLC1 expression. Analysis of publicly available annotated datasets of lung and colon cancer with gene expression microarray profiles indicated that, in pairwise comparisons, low DLC1 expression occurred frequently together (P < 0.01) with downregulation of p15Ink4b or p16Ink4a or upregulation of CDK4 or CDK6. In addition, an unfavorable prognosis (P < 0.05) was associated with low DLC1 and low p15Ink4b in lung cancer and colon cancer, low DLC1 and low p16Ink4a in lung cancer, low DLC1 and high CDK4 in lung cancer, and low DLC1 and high CDK6 in colon cancer. Thus, several genes and biochemical activities collaborate with the inactivation of DLC1 to give rise to cell transformation in MEFs, and the identified genes are relevant to human tumors with low DLC1 expression. Cancer Res; 72(22); 5900–11. ©2012 AACR.
- Published
- 2012
3. Preclinical evaluation of combined antineoplastic effect of DLC1 tumor suppressor protein and suberoylanilide hydroxamic acid on prostate cancer cells
- Author
-
Nicholas C. Popescu, Xu-Yu Yang, and Xiaoling Zhou
- Subjects
Male ,BALB 3T3 Cells ,Tumor suppressor gene ,Drug Evaluation, Preclinical ,Biophysics ,Antineoplastic Agents ,Biology ,Hydroxamic Acids ,Biochemistry ,Article ,Mice ,Transduction, Genetic ,Cell Line, Tumor ,LNCaP ,medicine ,Animals ,Humans ,Molecular Biology ,Vorinostat ,Tumor Suppressor Proteins ,GTPase-Activating Proteins ,Prostatic Neoplasms ,Cell Biology ,DNA Methylation ,Combined Modality Therapy ,Molecular biology ,Histone Deacetylase Inhibitors ,Trichostatin A ,Acetylation ,DNA methylation ,Cancer research ,Histone deacetylase ,DLC1 ,medicine.drug - Abstract
Deleted in liver cancer (DLC1), a tumor suppressor gene in multiple cancers, is recurrently down regulated or inactivated by epigenetic mechanisms in primary prostate carcinomas (PCAs). In this study the methylation and acetylation profile of the DLC1 promoter region was examined in three PCA cell lines with low or undetectable DLC1 expression: LNCaP, its derivative C4-2B-2, and 22Rv1. Two histone deacetylase inhibitors (HDAC), suberoylanilide hydroxamic acid (SAHA) and trichostatin A (TSA) induced histone acetylation of the DLC1 promoter in all three lines. DLC1 promoter methylation and deacetylation were detected in LNCaP and C4-2B-2 cells while in 22Rv1 cells DLC1 is silenced by deacetylation. Treatment with SAHA or TSA efficiently increased DLC1 expression in all lines, particularly in 22Rv1 cells, and activated the DLC1 promoter through the same Sp1 sites. The 22Rv1 cell line was selected to evaluate the efficacy of combined DLC1 transduction and SAHA treatment on tumor growth in athymic mice. Individually, DLC1 transduction and SAHA exposure reduced the tumor size by 75-80% compared to controls and in combination almost completely inhibited tumor growth. The antitumor effect was associated with the induction of apoptosis and inhibition of RhoA activity. SAHA alone significantly reduced RhoA activity, showing that this RhoGTPase is a target for SAHA. These results, obtained with a reliable preclinical in vivo test, predict that combined therapeutic agents targeting the pathways governing DLC1 function and HDAC inhibitors may be beneficial in management of prostate cancer.
- Published
- 2012
4. Cooperative antiproliferative effect of coordinated ectopic expression of DLC1 tumor suppressor protein and silencing of MYC oncogene expression in liver cancer cells: Therapeutic implications
- Author
-
Marian E. Durkin, Xiaoling Zhou, Nicholas C. Popescu, Xu-Yu Yang, and Paul Tone
- Subjects
0301 basic medicine ,Cancer Research ,Gene knockdown ,RHOA ,Oncogene ,biology ,Liver cell ,Articles ,medicine.disease_cause ,Molecular biology ,Small hairpin RNA ,03 medical and health sciences ,030104 developmental biology ,Oncology ,biology.protein ,medicine ,Ectopic expression ,DLC1 ,Carcinogenesis - Abstract
Human hepatocellular carcinoma (HCC) is one of the most common types of cancer and has a very poor prognosis; thus, the development of effective therapies for the treatment of advanced HCC is of high clinical priority. In the present study, the anti-oncogenic effect of combined knockdown of c-Myc expression and ectopic restoration of deleted in liver cancer 1 (DLC1) expression was investigated in human liver cancer cells. Expression of c-Myc in human HCC cells was knocked down by stable transfection with a Myc-specific short hairpin (sh) RNA vector. DLC1 expression in Huh7 cells was restored by adenovirus transduction, and the effects of DLC1 expression and c-Myc knockdown on Ras homolog gene family, member A (RhoA) levels, cell proliferation, soft agar colony formation and cell invasion were measured. Downregulation of c-Myc or re-expression of DLC1 led to a marked reduction in RhoA levels, which was associated with decreases in cell proliferation, soft agar colony formation and invasiveness; this inhibitory effect was augmented with a combination of DLC1 transduction and c-Myc suppression. To determine whether liver cell-specific delivery of DLC1 was able to enhance the inhibitory effect of c-Myc knockdown on tumor growth in vivo, DLC1 vector DNA complexed with galactosylated polyethylene glycol-linear polyethyleneimine was administered by tail vein injection to mice bearing subcutaneous xenografts of Huh7 cells transfected with shMyc or control shRNA. A cooperative inhibitory effect of DLC1 expression and c-Myc knockdown on the growth of Huh7-derived tumors was observed, suggesting that targeted liver cell delivery of DLC1 and c-Myc shRNA may serve as a possible gene therapy modality for the treatment of human HCC.
- Published
- 2015
5. DLC1 Interaction with S100A10 Mediates Inhibition of In Vitro Cell Invasion and Tumorigenicity of Lung Cancer Cells through a RhoGAP-Independent Mechanism
- Author
-
Nicholas C. Popescu, Xu-Yu Yang, and Drazen B. Zimonjic
- Subjects
Cancer Research ,Lung Neoplasms ,GTPase-activating protein ,Tumor suppressor gene ,Amino Acid Motifs ,Down-Regulation ,Breast Neoplasms ,Cell Growth Processes ,Article ,Cell surface receptor ,Carcinoma, Non-Small-Cell Lung ,Cell Line, Tumor ,Humans ,Medicine ,Annexin A2 ,Binding Sites ,biology ,business.industry ,Tumor Suppressor Proteins ,GTPase-Activating Proteins ,S100 Proteins ,HEK 293 cells ,Ubiquitination ,S100A10 ,Plasminogen ,Cell migration ,Cell biology ,HEK293 Cells ,Oncology ,Immunology ,biology.protein ,DLC1 ,business ,Protein Binding - Abstract
The DLC1 gene encodes a Rho GTPase-activating protein (RhoGAP) that functions as a tumor suppressor in several common human cancers. The multidomain structure of DLC1 enables interaction with a number of other proteins. Here we report that the proinflammatory protein S100A10 (also known as p11), a key cell surface receptor for plasminogen which regulates pericellular proteolysis and tumor cell invasion, is a new binding partner of DLC1 in human cells. We determined that the 2 proteins colocalize in the cell cytoplasm and that their binding is mediated by central sequences in the central domain of DLC1 and the C-terminus of S100A10. Because the same S100A10 sequence also mediates binding to Annexin 2, we found that DLC1 competed with Annexin 2 for interaction with S100A10. DLC1 binding to S100A10 did not affect DLC1's RhoGAP activity, but it decreased the steady-state level of S100A10 expression in a dose-dependent manner by displacing it from Annexin 2 and making it accessible to ubiquitin-dependent degradation. This process attenuated plasminogen activation and resulted in inhibition of in vitro cell migration, invasion, colony formation, and anchorage-independent growth of aggressive lung cancer cells. These results suggest that a novel GAP-independent mechanism contributes to the tumor suppressive activity of DLC1, and highlight the importance and complexity of protein–protein interactions involving DLC1 in certain cancers. Cancer Res; 71(8); 2916–25. ©2011 AACR.
- Published
- 2011
6. DLC1 tumor suppressor gene inhibits migration and invasion of multiple myeloma cells through RhoA GTPase pathway
- Author
-
Drazen B. Zimonjic, Xu-Yu Yang, Xiaoling Zhou, Veenu Tripathi, Veronika Ullmannova-Benson, Ming Guan, and Nicholas C. Popescu
- Subjects
Adult ,Male ,Cancer Research ,RHOA ,Tumor suppressor gene ,Biology ,Article ,Young Adult ,Cell Movement ,Cell Line, Tumor ,Humans ,Gene silencing ,Neoplasm Invasiveness ,Gene Silencing ,Promoter Regions, Genetic ,Aged ,Aged, 80 and over ,Cell growth ,Tumor Suppressor Proteins ,GTPase-Activating Proteins ,Cell migration ,Hematology ,DNA Methylation ,Middle Aged ,Actin cytoskeleton ,Oncology ,DNA methylation ,Cancer research ,biology.protein ,Female ,DLC1 ,Multiple Myeloma ,rhoA GTP-Binding Protein - Abstract
DLC1 (deleted in liver cancer 1), a tumor suppressor gene that encodes a RhoGTPase-activating protein, is recurrently downregulated or silenced in various solid tumors and hematological malignancies because of epigenetic modifications or genomic deletion. Here, we identified DLC1 promoter hypermethylation in 43 out of 44 multiple myeloma (MM) cell lines, which resulted in downregulation or silencing of DLC1 in 41 samples. High frequency of tumor-specific methylation and attenuation or silencing of DLC1 expression could serve as an independent diagnostic marker for MM. Combined treatment with demethylating and acetylating agents significantly elevated the expression of DLC1 and suppressed MM cell proliferation. Two cell lines exhibiting complete promoter methylation and the absence of DLC1 expression were transduced by an adenoviral vector containing DLC1 cDNA. In both cell lines, the reexpression of DLC1 inhibited myeloma cell invasion and migration, reduced RhoA activity and resulted in the reorganization of actin cytoskeleton. These results provide the first evidence for the antiproliferative effect of DLC1 in a hematological cancer and implicate RhoA pathway in suppression of MM migration and invasion. Given the myeloma cells sensitivity to the reactivation of DLC1 function, the potential for molecular targeted therapy of DLC1-mediated pathways as well as epigenetic therapies hold prospects.
- Published
- 2008
7. Flotillin-2 promotes metastasis of nasopharyngeal carcinoma by activating NF-κB and PI3K/Akt3 signaling pathways
- Author
-
Qiuyuan Wen, Lei Wang, Yuxiang Chen, Jie Liu, Bin Zhu, Xiangling Feng, Huan Chen, Chang Zhang, Caiping Ren, Xu-Yu Yang, Liang Zeng, Weidong Liu, Wei Jia, Wei Huang, Lihua Zhang, and Xiaomeng Xia
- Subjects
Pathology ,medicine.medical_specialty ,Nasopharyngeal neoplasm ,Mice, Nude ,Biology ,Article ,Metastasis ,Mice ,Phosphatidylinositol 3-Kinases ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Neoplasm Metastasis ,Author Correction ,Lipid raft ,PI3K/AKT/mTOR pathway ,Multidisciplinary ,Nasopharyngeal Carcinoma ,Carcinoma ,NF-kappa B ,Membrane Proteins ,Nasopharyngeal Neoplasms ,medicine.disease ,Nasopharyngeal carcinoma ,Tumor progression ,Cell culture ,Cancer research ,Heterografts ,Signal transduction ,Proto-Oncogene Proteins c-akt ,Neoplasm Transplantation ,Signal Transduction - Abstract
Lipid raft proteins have been confirmed to be important in cell signal transduction. Some reports have shown that the aberrant expression of lipid raft proteins is associated with malignant phenotypes in some cancers. However, the role of the lipid raft protein flotillin-2 (Flot-2) in nasopharyngeal carcinoma (NPC) remains to be comprehensively characterized. Here, overexpression of Flot-2 in NPC tissues and cell lines was detected by immunostaining and Flot-2 expression was found to be positively associated with NPC metastasis. Furthermore, inhibiting Flot-2 expression impaired the malignancy of the highly metastatic NPC cell line 5-8F by constraining its growth and proliferation, mobility and migration and decreasing the capacity of 5-8F cells to metastasize in nude mice. In contrast, forced overexpression of Flot-2 increased the malignancy of 6-10B, a non-metastatic NPC cell line that weakly expresses Flot-2. Moreover, in 5-8F-shFlot-2 cells, which have inhibited Flot-2 expression, the NF-κB and PI3K/Akt3 pathways were inactivated. Subsequently, MMPs expression were decreased and Foxo1 activity was increased. In addition, enhanced NF-κB and PI3K/Akt3 activities were observed in Flot-2 overexpressing 6-10B cells. Thus, Flot-2 exerts a pro-neoplastic role in NPC and is involved in tumor progression and metastasis. Moreover, Flot-2 exerts its role through NF-κB and PI3K/Akt3 signaling.
- Published
- 2014
8. Suppression of bcl-2 Gene by RNA Interference Increases Chemosensitivity to Cisplatin in Nasopharyngeal Carcinoma Cell Line CNE1
- Author
-
Ming Zhao, Zhi-Hua Yin, Feng Li, Kaitai Yao, Caiping Ren, Hui Li, and Xu-Yu Yang
- Subjects
Cisplatin ,Cell growth ,Genetic enhancement ,Biophysics ,General Medicine ,Transfection ,Biology ,medicine.disease ,Biochemistry ,Molecular biology ,Nasopharyngeal carcinoma ,Cell culture ,RNA interference ,Gene expression ,Cancer research ,medicine ,medicine.drug - Abstract
To explore the effect of suppressing BCL-2 expression using RNA interference (RNAi) technique in nasopharyngeal carcinoma cell line CNE1. CNE1 cell lines stably expressing shRNAs targeted bcl-2 and GL3 gene were established and gene expression inhibition was assessed by Western blotting analysis. The effect of suppressing bcl-2 by RNAi on cell growth was studied, the apoptosis induction and the sensitization of CNE1 cells to cisplatin were quantified by MTT assay and flow cytometry. The results showed that: stable transfection of CNE1 cells with vectors expressing shRNAs against bcl-2 decreased the expression of BCL-2 protein; suppression of BCL-2 expression did not affect cell proliferation but could increase the chemosensitivity to cisplatin in CNE1 cells. This will help physicians to make some clinical trials of gene therapy on nasopharyngeal carcinoma by RNAi.
- Published
- 2004
9. Synergistic antineoplastic effect of DLC1 tumor suppressor protein and histone deacetylase inhibitor, suberoylanilide hydroxamic acid (SAHA), on prostate and liver cancer cells: perspectives for therapeutics
- Author
-
Nicholas C. Popescu, Xiaoling Zhou, and Xu-Yu Yang
- Subjects
Male ,Cancer Research ,Time Factors ,Tumor suppressor gene ,medicine.drug_class ,Cell Survival ,Genetic Vectors ,Apoptosis ,Biology ,medicine.disease_cause ,Hydroxamic Acids ,Adenoviridae ,Cell Movement ,Transduction, Genetic ,Cell Line, Tumor ,medicine ,Humans ,Cancer epigenetics ,RNA, Messenger ,Promoter Regions, Genetic ,Vorinostat ,Cell Shape ,Cell Proliferation ,Caspase 3 ,Tumor Suppressor Proteins ,Histone deacetylase inhibitor ,GTPase-Activating Proteins ,Liver Neoplasms ,Cancer ,Prostatic Neoplasms ,Genetic Therapy ,medicine.disease ,Enzyme Activation ,Histone Deacetylase Inhibitors ,Oncology ,Immunology ,Cancer research ,DLC1 ,Carcinogenesis ,Epigenetic therapy ,medicine.drug - Abstract
Inactivation of tumor suppressor genes is a major contributing alteration in the initiation or progression of cancer. The human tumor suppressor gene DLC1 (deleted in liver cancer 1) is frequently downregulated or silenced in multiple cancers, predominantly by epigenetic mechanisms. With the current considerable interest and progress in epigenetic therapy, a number of promising antineoplastic agents, particularly histone deacetylase (HDAC) inhibitors, have been developed and used successfully in clinical trials. Both DLC1 and HDAC inhibitors exert antineoplastic functions, and their combined action could be exploited for a more effective cancer therapy. To evaluate the potential benefits of this approach, we examined the antineoplastic effects of adenoviral (Ad)-DLC1-mediated transduction and exposure to suberoylanilide hydroxamic acid (SAHA), a powerful HDAC inhibitor, in two human cancer cell lines that lack intrinsic DLC1 expression, 22Rv1 prostate cancer cells and 7703K human hepatocellular carcinoma cells. Consistent with the oncosuppressive function of DLC1 in several cancers, including prostate and liver cancer, transduction of 22Rv1 and 7703K cells with an Ad-DLC1 expression vector resulted in alterations of cell morphology, induction of apoptosis, and inhibition of cell proliferation, migration, and anchorage-independent growth. A low concentration of SAHA (5 microM) efficiently restored the expression of DLC1 in 22Rv1 cells that lack DLC1 expression due to histone deacetylation but had a minimal effect in 7703K cells in which silencing of the DLC1 gene is due mainly to promoter hypermethylation. Regardless of the epigenetic mechanism of DLC1 inactivation, SAHA treatment of DLC1-transduced cells had a synergistic inhibitory effect on tumor cell proliferation and tumorigenesis in both cell lines. In 22Rv1 cells, this combination regimen nearly abolished the formation of colonies in semisolid media as a measure of tumorigenicity in vitro. Current in vitro results validate this protocol as a potentially new therapeutic option in certain cancers.
- Published
- 2010
10. DLC1 suppresses distant dissemination of human hepatocellular carcinoma cells in nude mice through reduction of RhoA GTPase activity, actin cytoskeletal disruption and down-regulation of genes involved in metastasis
- Author
-
Drazen B. Zimonjic, Marian E. Durkin, Nicholas C. Popescu, Sang Won Park, Xiaoling Zhou, and Xu-Yu Yang
- Subjects
Cancer Research ,RHOA ,Carcinoma, Hepatocellular ,Lung Neoplasms ,Skin Neoplasms ,Tumor suppressor gene ,Down-Regulation ,Mice, Nude ,Apoptosis ,Biology ,Matrix Metalloproteinase Inhibitors ,Article ,Metastasis ,Mice ,medicine ,Tumor Cells, Cultured ,Animals ,Humans ,Neoplasm Invasiveness ,Cytoskeleton ,Cell Proliferation ,Matrigel ,Cell growth ,Tumor Suppressor Proteins ,GTPase-Activating Proteins ,Liver Neoplasms ,Cancer ,medicine.disease ,Actins ,Drug Combinations ,Oncology ,Matrix Metalloproteinase 9 ,Cancer cell ,Cancer research ,biology.protein ,Osteopontin ,Proteoglycans ,DLC1 ,Collagen ,Laminin ,rhoA GTP-Binding Protein - Abstract
The process of cell dissemination from the primary tumors to distant sites is the most harmful event during cancer progression, and the leading cause of cancer death. We have previously demonstrated that restoration of DLC1 tumor suppressor gene expression in the DLC1-negative Focus and 7703K human hepatocellular carcinoma (HCC) cell lines induced caspase-3 mediated apoptosis, reduced cell growth in vitro and tumorigenicity in vivo and diminished the ability to migrate through Matrigel, a property suggestive of metastatic potential in vivo. We now show that subcutaneous tumors developing after inoculation of Focus and 7703K cells into nude mice disseminate cells to liver and lung, and this process is markedly suppressed by restoration of DLC1 expression. Inhibition of tumor cell dissemination was associated with lower levels of RhoA activity, an increase in rounded cells and a reduction in actin stress fibers and focal adhesion molecules that are of critical importance in cancer cell invasion and metastasis. In addition, DLC1 down-regulated the expression of osteopontin and matrix metalloproteinase-9, which are highly up-regulated in most primary HCC with associated metastases. These observations implicate the DLC1 gene in suppression of HCC cell dissemination and identify novel cellular and genetic alterations that contribute to prevention of metastasis, a life-threatening event in cancer progression.
- Published
- 2008
11. DLC1 suppresses distant dissemination of human hepatocellular carcinoma cells in nude mice through reduction of RhoA GTPase activity, actin cytoskeletal disruption and down-regulation of genes involved in metastasis
- Author
-
Xiaoling Zhou, Drazen Zimonjic, Sang-Won Park, Xu-Yu Yang, Marian Durkin, and Nicholas Popescu
- Subjects
Cancer Research ,Oncology - Published
- 2008
12. Reexploring the possible roles of some genes associated with nasopharyngeal carcinoma using microarray-based detection
- Author
-
Kai tai Yao, Yan Qing Ding, Zhong xi Huang, Qiu Zhen Liu, Cai Ping Ren, Xin Li, Wei Yi Fang, Xin Deng, Wei Bing Xie, Teng-Fei Liu, Xu Yu Yang, and Shuang Wang
- Subjects
Cell type ,Microarray ,Biophysics ,Nasopharyngeal neoplasm ,Cathepsin D ,Biology ,Biochemistry ,otorhinolaryngologic diseases ,medicine ,Biomarkers, Tumor ,Tumor Cells, Cultured ,Humans ,Genetic Predisposition to Disease ,Genetic Testing ,Gene ,Oligonucleotide Array Sequence Analysis ,Gene Expression Profiling ,Carcinoma ,Nasopharyngeal Neoplasms ,General Medicine ,medicine.disease ,Neoplasm Proteins ,Gene expression profiling ,Gene Expression Regulation, Neoplastic ,stomatognathic diseases ,Nasopharyngeal carcinoma ,Cancer research ,DNA microarray - Abstract
In gene expression profiling, nasopharyngeal carcinoma (NPC) 5-8F cells differ from 6-10B cells in terms of their high tumorigenicity and metastatic ability. Differentially expressed genes from the two cell types were analyzed by combining with MILANO (the automatic custom annotation of microarray results which is based on all the available published work in PubMed). The results showed that five genes, including CTSD, P63, CSE1L, BPAG1 and EGR1, have been studied or mentioned in published work on NPC. Subsequently, we reevaluated the roles of these genes in the pathogenesis of NPC by combining the data of gene chips from NPCs versus NPs and pooled cells from 5-8F, 6-10B and CNE2 versus NPs. The results suggested that the roles of BPAG1 and EGR1 are possibly different from those reported in previous NPC studies. These five genes are likely to be involved in the proliferation, apoptosis, invasion and metastasis of NPC. A reexploration of the genes will further define their roles in the pathogenesis of NPC.
- Published
- 2005
13. Identification of Differentially Expressed Genes in Metastatic and Non-Metastatic Nasopharyngeal Carcinoma Cells by Suppression Subtractive Hybridization
- Author
-
Hui Li, Lei Wang, Xu-Yu Yang, Chun-Jie Jiang, Hong-Bo Zhang, Caiping Ren, Ming Zhao, and Kai-Tai Yao
- Subjects
reverse northern blot ,Cancer Research ,DNA, Complementary ,Sequence analysis ,Biology ,lcsh:RC254-282 ,Pathology and Forensic Medicine ,Nasopharyngeal carcinoma (NPC) ,Sequence Homology, Nucleic Acid ,Complementary DNA ,Tumor Cells, Cultured ,medicine ,Humans ,Genomic library ,RNA, Messenger ,Neoplasm Metastasis ,lcsh:QH573-671 ,Deoxyribonucleases, Type II Site-Specific ,Gene ,Gene Library ,tumor metastasis ,lcsh:Cytology ,Gene Expression Profiling ,Glyceraldehyde-3-Phosphate Dehydrogenases ,Nucleic Acid Hybridization ,Nasopharyngeal Neoplasms ,Sequence Analysis, DNA ,Cell Biology ,General Medicine ,suppression subtractive hybridization (SSH) ,Reverse northern blot ,medicine.disease ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Molecular biology ,Gene Expression Regulation, Neoplastic ,Gene expression profiling ,Nasopharyngeal carcinoma ,Suppression subtractive hybridization ,Molecular Medicine ,Other - Abstract
Background & Objective: Nasopharyngeal carcinoma (NPC) is an epithelial neoplasm with high occurrence rates in southern China. The disease often metastasizes to regional lymphnodes at a very early stage. Local recurrences and metastasis occur frequently in patients with NPC and are a leading cause of death, despite improvements on treatment modalities. The molecular mechanism underlying the metastasis of nasopharyngeal carcinoma remains poorly understood, however, and requires additional elucidation. The aim of this study was to explore possible NPC gene candidates that may play key roles in NPC metastasis. Methods: Subtractive suppression hybridization (SSH) was performed to isolate differentially expressed clones between the metastatic 5-8F and non-metastatic 6-10B nasopharyngeal carcinoma cell lines. Differentially expressed clones were screened and confirmed by reverse Northern blotting. The sequences of cDNA fragments were subsequently analyzed and compared to known sequences in Genbank. Results & Discussion: The SSH library contained thousands of positive clones. Random analysis of 300 clones by PCR demonstrated that 269 clones contained inserted fragments. Reverse Northern blot confirmed that 20 out of 192 clones examined were significantly up-regulated in the 5-8F cell line. Among these 20 clones, 16 were previously identified genes (flotilin-2, ezrin, pim-3, fli-1, mel, neugrin, znf216, ASB1, raly, UBE2A, keratin6A, TMED7, EIF3S9, FTL, two ribosomal proteins RPL21 and RPL16), two were predicted genes (c9orf74 and MDS006), and two sequences shared no homology with known genes listed in GenBank and may represent novel genes. The proposed functions of the genes identified in this study include cell signal transduction, cell survival, transcription regulation, cell mobility, protein synthesis, and DNA damage repair. Flotillin-2, fli-1, pim-3 and ezrin have previously been reported to be associated with tumor metastasis and progression. The remaining up-regulated genes identified in this study have not been reported to be markers of metastasis and may represent new candidates of NPC metastasis-related genes. The Results of this study may provide novel points of therapeutic intervention for NPC.
- Published
- 2005
- Full Text
- View/download PDF
14. Abstract 4254: ESE1 binds to beta catenin and modulates its expression
- Author
-
Seong-Ho Lee, Kui Jin Kim, Xu-yu Yang, and Hong Jun Shao
- Subjects
Cancer Research ,Gene knockdown ,Small interfering RNA ,Beta-catenin ,Oncology ,Apoptosis ,Cytoplasm ,HEK 293 cells ,biology.protein ,Biology ,Binding site ,Molecular biology ,Transcription factor - Abstract
ESE-1 is an epithelium-specific transcription factor that functions in the differentiation of intestinal epithelial cells. ESE-1's structure predicts its interaction with a number of other proteins with various cellular functions. Previously we have shown that tolfenamic acid (TA) could induce nuclear translocation of ESE1 in colorectal cancer cells, and plays a critical role in the activation of apoptosis. Here we show that β-catenin, an important protein regulating cell-cell adhesion and gene transcription, binds and colocalizes with ESE-1 in cell cytoplasm. The binding sites were mapped to carboxy terminus ESE-1, which includes the ETS domain, and N-terminus of β-catenin, which contains several important phosphorylation sites. C-terminus of ESE1 also binds to α-catenin, and ESE1/ β-catenin interaction was abrogated with suppression of either β-catenin or α-catenin in HEK293 cells by si RNA treatment; moreover, suppression of both α-catenin and β-catenin further impaired ESE1/ β-catenin binding. Furthermore, knockdown of endogenous ESE1 significantly decreased β-catenin level in HCT116 cells. Current data support a role of ESE1 in the regulation of β-catenin function in colon cancer. Citation Format: Xu-yu Yang, Kui Jin Kim, Hong Jun Shao, Seong-Ho Lee. ESE1 binds to beta catenin and modulates its expression. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 4254. doi:10.1158/1538-7445.AM2014-4254
- Published
- 2014
15. Abstract 2349: DLC1 interaction with S100A10 mediates inhibition of in vitro cell invasion and tumorigenicity of lung cancer cells
- Author
-
Drazen B. Zimonjic, Nicolae C. Popescu, and Xu-Yu Yang
- Subjects
Cancer Research ,biology ,Plasmin ,Chemistry ,S100A10 ,Cell migration ,Cell membrane ,medicine.anatomical_structure ,Oncology ,Cytoplasm ,Cancer research ,biology.protein ,medicine ,DLC1 ,Receptor ,Annexin A2 ,medicine.drug - Abstract
Deleted in liver cancer 1 (DLC1) is a multidomain Rho-GTPase-activating protein (RhoGAP) and a tumor suppressor in several human common cancers. DLC1's structure predicts its interaction with a number of other proteins with various cellular functions. Previously we have shown that S100A10 (also known as p11), a key plasminogen receptor on the surface of cell membrane, colocalizes and binds with DLC1 in cell cytoplasm. The binding sites were mapped to the central domain of the DLC1 and at the end of C-terminus of S100A10. The same terminus is also a binding site for Annexin A2, which is believed to regulate availability of extracellular S100A10 and, therefore, dynamics of plasmin formation. Now we demonstrate that DLC1 competes with Annexin A2 for interaction with S100A10. DLC1 binding to S100A10 did not affect DLC1's RhoGAP activity but did reduce availability of S100A10 for binding by Annexin A2. Furthermore, the expression of DLC1 also affected S100A10 expression in a dose dependent manner, and increased S100A10 ubiquitin-dependent degradation. All of the above interactions attenuated plasminogen activation and resulted in inhibition of in vitro cell migration, invasion, colony formation and anchorage independent growth of aggressive and tumorigenic lung tumor cells. Transduction of DLC1-negative breast tumor cells significantly reduced the ability of tumor cells to form pulmonary metastases as well as inhibited distant dissemination of liver tumor cells in athymic mice. Current observations provide a plausible mechanism for DLC1 antimetastatic function by negatively affecting plasminogen activation, which in turn lead to a significant reduction in invasiveness of tumor cells. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 2349.
- Published
- 2010
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.