Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
EMBO Rep ; 23(8): e54265, 2022 08 03.
Article in English | MEDLINE | ID: mdl-35766227

ABSTRACT

The aggressive nature and poor prognosis of lung cancer led us to explore the mechanisms driving disease progression. Utilizing our invasive cell-based model, we identified methylthioadenosine phosphorylase (MTAP) and confirmed its suppressive effects on tumorigenesis and metastasis. Patients with low MTAP expression display worse overall and progression-free survival. Mechanistically, accumulation of methylthioadenosine substrate in MTAP-deficient cells reduce the level of protein arginine methyltransferase 5 (PRMT5)-mediated symmetric dimethylarginine (sDMA) modification on proteins. We identify vimentin as a dimethyl-protein whose dimethylation levels drop in response to MTAP deficiency. The sDMA modification on vimentin reduces its protein abundance but trivially affects its filamentous structure. In MTAP-deficient cells, lower sDMA modification prevents ubiquitination-mediated vimentin degradation, thereby stabilizing vimentin and contributing to cell invasion. MTAP and PRMT5 negatively correlate with vimentin in lung cancer samples. Taken together, we propose a mechanism for metastasis involving vimentin post-translational regulation.


Subject(s)
Lung Neoplasms , Purine-Nucleoside Phosphorylase , Humans , Lung Neoplasms/genetics , Protein-Arginine N-Methyltransferases/genetics , Protein-Arginine N-Methyltransferases/metabolism , Purine-Nucleoside Phosphorylase/metabolism , Vimentin/genetics
2.
Clin Cancer Res ; 24(4): 916-926, 2018 02 15.
Article in English | MEDLINE | ID: mdl-29217529

ABSTRACT

Purpose: The comprehensive understanding of mechanisms involved in the tumor metastasis is urgently needed for discovering novel metastasis-related genes for developing effective diagnoses and treatments for lung cancer.Experimental Design: FAM198B was identified from an isogenic lung cancer metastasis cell model by microarray analysis. To investigate the clinical relevance of FAM198B, the FAM198B expression of 95 Taiwan lung adenocarcinoma patients was analyzed by quantitative real-time PCR and correlated to patients' survivals. The impact of FAM198B on cell invasion, metastasis, and tumor growth was examined by in vitro cellular assays and in vivo mouse models. In addition, the N-glycosylation-defective FAM198B mutants generated by site-directed mutagenesis were used to study protein stability and subcellular localization of FAM198B. Finally, the microarray and pathway analyses were used to elucidate the underlying mechanisms of FAM198B-mediated tumor suppression.Results: We found that the high expression of FAM198B was associated with favorable survival in Taiwan lung adenocarcinoma patients and in a lung cancer public database. Enforced expression of FAM198B inhibited cell invasion, migration, mobility, proliferation, and anchorage-independent growth, and FAM198B silencing exhibited opposite activities in vitro FAM198B also attenuated tumor growth and metastasis in vivo We further identified MMP-1 as a critical downstream target of FAM198B. The FAM198B-mediated MMP-1 downregulation was via inhibition of the phosphorylation of ERK. Interestingly deglycosylation nearly eliminated the metastasis suppression activity of FAM198B due to a decrease of protein stability.Conclusions: Our results implicate FAM198B as a potential tumor suppressor and to be a prognostic marker in lung adenocarcinoma. Clin Cancer Res; 24(4); 916-26. ©2017 AACR.


Subject(s)
Adenocarcinoma/genetics , Gene Expression Regulation, Neoplastic , Lung Neoplasms/genetics , Matrix Metalloproteinase 1/genetics , Membrane Glycoproteins/genetics , Membrane Proteins/genetics , Adenocarcinoma/ethnology , Adenocarcinoma/metabolism , Animals , Asian People/genetics , Cell Line, Tumor , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Kaplan-Meier Estimate , Lung Neoplasms/ethnology , Lung Neoplasms/metabolism , Matrix Metalloproteinase 1/metabolism , Membrane Glycoproteins/metabolism , Membrane Proteins/metabolism , Mice, SCID , Neoplasm Metastasis , Oligonucleotide Array Sequence Analysis/methods , RNA Interference , Taiwan , Transplantation, Heterologous , Tumor Burden/genetics
3.
Am J Cancer Res ; 6(6): 1317-30, 2016.
Article in English | MEDLINE | ID: mdl-27429846

ABSTRACT

Although targeted therapy can prolong the survival of non-small cell lung cancer (NSCLC) patients with EGFR mutations, chemotherapy still is the choice for patients with wild-type EGFR or failure in targeted therapy. However, most of the patients will eventually develop chemoresistance. Our previous study showed that miR-137 is a risky microRNA and is associated with poor prognosis in NSCLC patients. Here we investigated the role of miR-137 in cisplatin resistance in lung adenocarcinoma patients. Our data indicated that miR-137 overexpression increases the survival of lung cancer cells exposed to cisplatin and decreases cisplatin-induced apoptosis. Through computational prediction and microarray, we identified caspase-3 (CASP3) as a potential target of miR-137. Luciferase reporter and site-directed mutagenesis assays demonstrated that miR-137 downregulates CASP3 through binding to its 3'-UTR. Moreover, the endogenous CASP3 can be modulated by overexpressing or silencing miR-137 in lung adenocarcinoma cell lines regardless of EGFR status. Suppression of CASP3 by miR-137 provides cancer cells with anti-apoptotic ability, leading to cisplatin resistance. Immunohistochemistry results revealed an inverse correlation between miR-137 and CASP3 expressions in lung adenocarcinoma patients. Together, our data provide a new chemoresistance mechanism in lung adenocarcinoma and a possible target to control chemoresistance in lung adenocarcinoma patients.

4.
Oncotarget ; 7(28): 44417-44429, 2016 Jul 12.
Article in English | MEDLINE | ID: mdl-27323831

ABSTRACT

SPANXA (Sperm Protein Associated with the Nucleus on the X-chromosome, family members A1/A2) acts as a cancer-testis antigen expressed in normal testes, but dysregulated in various tumors. We found that SPANXA is highly expressed in low-invasive CL1-0 cells compared with isogenous high-invasive CL1-5 cells. SPANXA was preferably expressed in tumor tissues and associated with the prolonged survival of lung adenocarcinomas. SPANXA suppressed the invasion and metastasis of lung cancer cells in vitro and in vivo. By the expression microarray and pathway analysis, we found that the SPANXA-altered genes were enriched in the epithelial-mesenchymal transition (EMT) pathway. SPANXA reduced SNAI2 expression resulted in up-regulating E-cadherin. c-JUN acts as the positive-regulator of EMT. Silencing SPANXA increased c-JUN mRNA expression and blockage of c-JUN led to SNAI2 down-regulation. Our results clearly characterized SPANXA as an EMT inhibitor by suppressing c-JUN-SNAI2 axis in lung adenocarcinoma.


Subject(s)
Adenocarcinoma/metabolism , Lung Neoplasms/metabolism , Nuclear Proteins/metabolism , Proto-Oncogene Proteins c-jun/antagonists & inhibitors , Snail Family Transcription Factors/antagonists & inhibitors , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Animals , Cell Line, Tumor , Epithelial-Mesenchymal Transition , Heterografts , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mice , Mice, SCID , Nuclear Proteins/genetics , Proto-Oncogene Proteins c-jun/metabolism , Signal Transduction , Snail Family Transcription Factors/metabolism , Transfection , Up-Regulation
5.
PLoS One ; 10(4): e0124191, 2015.
Article in English | MEDLINE | ID: mdl-25875824

ABSTRACT

Homeobox genes comprise a family of regulatory genes that contain a common homeobox domain and act as transcription factors. Recent studies indicate that homeobox A5 (HOXA5) may serve as a tumour suppressor gene in breast cancers. However, the precise role and the underlying mechanism of HOXA5 in lung cancer remain unclear. Oligonucleotide microarrays and an invasion/metastasis lung adenocarcinoma cell line model were used to determine the correlation between HOXA5 expression and cancer cell invasion ability. We found that ectopic expression of HOXA5 in highly invasive cancer cells suppressed cell migration, invasion, and filopodia formation in vitro and inhibited metastatic potential in vivo. Knockdown of HOXA5 promoted the invasiveness of lung cancer cells. In addition, HOXA5 expression was associated with better clinical outcome in non-small cell lung cancer patients with wild-type EGFR. Furthermore, genome-wide transcriptomic and pathway analyses were performed to identify the potential molecular mechanisms. Our data showed that HOXA5 may bind to the promoters of the cytoskeleton-related genes and downregulate their mRNA and protein expression levels. Our studies provide new insights into how HOXA5 may contribute to the suppression of metastasis in lung cancer via cytoskeleton remodelling regulation. Therefore, targeted induction of HOXA5 may represent a promising approach for non-small-cell lung cancer therapy.


Subject(s)
Carcinoma, Non-Small-Cell Lung/pathology , Cytoskeleton/metabolism , Homeodomain Proteins/metabolism , Lung Neoplasms/pathology , Animals , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/mortality , Cell Line, Tumor , Cell Movement , Cell Proliferation , Disease Models, Animal , Disease-Free Survival , ErbB Receptors/metabolism , Homeodomain Proteins/antagonists & inhibitors , Homeodomain Proteins/genetics , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/mortality , Mice , Mice, SCID , Microscopy, Fluorescence , Neoplasm Metastasis , Oligonucleotide Array Sequence Analysis , Promoter Regions, Genetic , Protein Binding , RNA Interference , RNA, Small Interfering/metabolism , Survival Rate , Transplantation, Heterologous
6.
PLoS One ; 10(3): e0118530, 2015.
Article in English | MEDLINE | ID: mdl-25738875

ABSTRACT

α-parvin (PARVA) is known to be involved in the linkage of integrins, regulation of actin cytoskeleton dynamics and cell survival. However, the role that PARVA plays in cancer progression remains unclear. Here, using a lung cancer invasion cell line model and expression microarrays, we identify PARVA as a potential oncogene. The overexpression of PARVA increased cell invasion, colony-forming ability and endothelial cell tube formation. By contrast, knockdown of PARVA inhibited invasion and tube formation in vitro. Overexpression of PARVA also promoted tumorigenicity, angiogenesis and metastasis in in vivo mouse models. To explore the underlying mechanism, we compared the expression microarray profiles of PARVA-overexpressing cells with those of control cells to identify the PARVA-regulated signalling pathways. Pathway analysis showed that eight of the top 10 pathways are involved in invasion, angiogenesis and cell death. Next, to identify the direct downstream signalling pathway of PARVA, 371 significantly PARVA-altered genes were analysed further using a transcription factor target model. Seven of the top 10 PARVA-altered transcription factors shared a common upstream mediator, ILK. Lastly, we found that PARVA forms a complex with SGK1 and ILK to enhance the phosphorylation of ILK, which led to the phosphorylation of Akt and GSK3ß. Notably, the inactivation of ILK reversed PARVA-induced invasion. Taken together, our findings imply that PARVA acts as an oncogene by activating ILK, and that this activation is followed by the activation of Akt and inhibition of GSK3ß. To our knowledge, this is the first study to characterize the role of PARVA in lung cancer progression.


Subject(s)
Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Microfilament Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Adenocarcinoma/blood supply , Adenocarcinoma of Lung , Animals , Carcinogenesis , Cell Line, Tumor , Cell Movement , Cell Proliferation , Disease Progression , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Humans , Lung Neoplasms/blood supply , Mice , Neoplasm Invasiveness , Neoplasm Metastasis , Neovascularization, Pathologic , Phosphorylation
7.
Am J Respir Crit Care Med ; 190(4): 433-44, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-25036006

ABSTRACT

RATIONALE: Despite advances in treatment and prognosis of non-small cell lung cancer (NSCLC), patient outcomes are still unsatisfactory. OBJECTIVES: To reduce the morbidity and mortality of patients with NSCLC, a more comprehensive understanding of mechanisms involved in cancer progression is urgently needed. METHODS: By comparison of gene expression profiles in the cell line pair with differential invasion ability, CL1-0 and CL1-5, we found that Shisa3 was highly expressed in the low invasive cells. The effect of Shisa3 on invasion, migration, proliferation, apoptosis, epithelial-mesenchymal transition, and anchorage-independent growth activities in vitro and on tumor growth and metastasis in mice models were examined. The underlying mechanism of Shisa3 was explored by microarray and pathway analysis. Finally, the correlation of Shisa3 expression and clinical outcome was also calculated. MEASUREMENTS AND MAIN RESULTS: We identified Shisa3 as a novel tumor suppressor, which induces ß-catenin degradation resulting in suppression of tumorigenesis and invasion in vitro. Shisa3 decreased the tumor growth in mice with subcutaneous implantation and reduced the number of metastatic nodules in mice with tail vein injection and orthotopic implantation. Shisa3 performs the tumor suppression activity through WNT signaling predicted by microarray analysis. Our data found that Shisa3 accelerates ß-catenin degradation and was positively associated with overall survival and progression-free survival of NSCLC. CONCLUSIONS: Our results reveal that Shisa3 acts as a tumor suppressor by acceleration of ß-catenin degradation and provide new insight for cancer prognosis and therapy.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Lung Neoplasms/metabolism , Membrane Proteins/metabolism , beta Catenin/metabolism , Aged , Animals , Apoptosis/genetics , Blotting, Western/methods , Carcinoma, Non-Small-Cell Lung/genetics , Cell Movement/genetics , Cell Proliferation , Cell Survival/genetics , Disease Models, Animal , Humans , In Vitro Techniques , Lung Neoplasms/genetics , Membrane Proteins/genetics , Mice , Mice, SCID , Microarray Analysis/methods , Polymerase Chain Reaction/methods , Signal Transduction/genetics , Taiwan , Tumor Cells, Cultured , beta Catenin/genetics
8.
Article in English | MEDLINE | ID: mdl-23970932

ABSTRACT

Colorectal cancer (CRC) is a serious public health problem that results due to changes of diet and various environmental stress factors in the world. Curcumin is a traditional medicine used for treatment of a wide variety of tumors. However, antimetastasis mechanism of curcumin on CRC has not yet been completely investigated. Here, we explored the underlying molecular mechanisms of curcumin on metastasis of CRC cells in vitro and in vivo. Curcumin significantly inhibits cell migration, invasion, and colony formation in vitro and reduces tumor growth and liver metastasis in vivo. We found that curcumin suppresses Sp-1 transcriptional activity and Sp-1 regulated genes including ADEM10, calmodulin, EPHB2, HDAC4, and SEPP1 in CRC cells. Curcumin inhibits focal adhesion kinase (FAK) phosphorylation and enhances the expressions of several extracellular matrix components which play a critical role in invasion and metastasis. Curcumin reduces CD24 expression in a dose-dependent manner in CRC cells. Moreover, E-cadherin expression is upregulated by curcumin and serves as an inhibitor of EMT. These results suggest that curcumin executes its antimetastasis function through downregulation of Sp-1, FAK, and CD24 and by promoting E-cadherin expression in CRC cells.

9.
Nano Lett ; 13(10): 4632-41, 2013 Oct 09.
Article in English | MEDLINE | ID: mdl-23984819

ABSTRACT

Multiple-walled carbon nanotubes (MWCNTs) may cause carcinogenesis. We found that long-term exposure to MWCNTs can induce irreversible oncogenic transformation of human bronchial epithelial cells and tumorigenicity in vivo. A genome-wide array-comparative genomic hybridization (aCGH) analysis revealed global chromosomal aberration in MWCNTs-treated clones, predominantly at chromosome 2q31-32, where the potential oncogenes HOXD9 and HOXD13 are located. Functional assays confirmed that this variation can modulate oncogenic signaling and plays a part in MWCNTs-induced tumorigenesis, suggesting that MWCNTs are carcinogens that act by altering genomic stability and oncogenic copy numbers.


Subject(s)
Carcinogenesis , Chromosomes/drug effects , Homeodomain Proteins/genetics , Nanotubes, Carbon/toxicity , Neoplasm Proteins/genetics , Transcription Factors/genetics , Bronchi/cytology , Bronchi/drug effects , Cell Transformation, Neoplastic/drug effects , Chromosomes/genetics , Comparative Genomic Hybridization , Epithelial Cells/cytology , Epithelial Cells/drug effects , Genome, Human , Genomic Instability/drug effects , Humans , Nanotubes, Carbon/chemistry
10.
Breast Cancer Res ; 13(6): R116, 2011.
Article in English | MEDLINE | ID: mdl-22113133

ABSTRACT

INTRODUCTION: Estrogen is involved in several physiological and pathological processes through estrogen receptor (ER)-mediated transcriptional gene regulation. miRNAs (miRs), which are noncoding RNA genes, may respond to estrogen and serve as posttranscriptional regulators in tumorigenic progression, especially in breast cancer; however, only limited information about this possibility is available. In the present study, we identified the estrogen-regulated miR-34b and investigated its functional role in breast cancer progression. METHODS: Estrogen-regulated miRNAs were identified by using a TaqMan low density array. Our in vivo Tet-On system orthotopic model revealed the tumor-suppressive ability of miR-34b. Luciferase reporter assays and chromatin immunoprecipitation assay demonstrated miR-34b were regulated by p53-ER interaction. RESULTS: In this study, we identified one such estrogen downregulated miRNA, miR-34b, as an oncosuppressor that targets cyclin D1 and Jagged-1 (JAG1) in an ER+/wild-type p53 breast cancer cell line (MCF-7), as well as in ovarian and endometrial cells, but not in ER-negative or mutant p53 breast cancer cell lines (T47D, MBA-MB-361 and MDA-MB-435). There is a negative association between ERα and miR-34b expression levels in ER+ breast cancer patients. Tet-On induction of miR-34b can cause inhibition of tumor growth and cell proliferation. Also, the overexpression of miR-34b inhibited ER+ breast tumor growth in an orthotopic mammary fat pad xenograft mouse model. Further validation indicated that estrogen's inhibition of miR-34b expression was mediated by interactions between ERα and p53, not by DNA methylation regulation. The xenoestrogens diethylstilbestrol and zeranol also showed similar estrogenic effects by inhibiting miR-34b expression and by restoring the protein levels of the miR-34b targets cyclin D1 and JAG1 in MCF-7 cells. CONCLUSIONS: These findings reveal that miR-34b is an oncosuppressor miRNA requiring both ER+ and wild-type p53 phenotypes in breast cancer cells. These results improve our ability to develop new therapeutic strategies to target the complex estrogenic pathway in human breast cancer progression through miRNA regulation.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Estrogens/metabolism , Genes, Tumor Suppressor , MicroRNAs/metabolism , Adult , Aged , Animals , Antineoplastic Agents, Hormonal/pharmacology , Breast Neoplasms/pathology , Calcium-Binding Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin D1/metabolism , Estrogens/pharmacology , Female , Gene Expression/drug effects , Gene Expression Profiling , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Jagged-1 Protein , Membrane Proteins/metabolism , Mice , Mice, Nude , Mice, SCID , Middle Aged , Models, Biological , Neoplasm Staging , Receptors, Estrogen/genetics , Serrate-Jagged Proteins , Tamoxifen/pharmacology , Tumor Suppressor Protein p53/metabolism , Xenograft Model Antitumor Assays
11.
Cell Host Microbe ; 9(1): 58-69, 2011 Jan 20.
Article in English | MEDLINE | ID: mdl-21238947

ABSTRACT

Viruses rely on the host translation machinery to complete their life cycles. Picornaviruses use an internal ribosome entry site to initiate cap-independent protein translation and in parallel host cap-dependent translation is shut off. This process is thought to occur primarily via cleavage of host translation initiation factors eIF4GI and eIF4GII by viral proteases. Here we describe another mechanism whereby miR-141 induced upon enterovirus infection targets the cap-dependent translation initiation factor, eIF4E, for shutoff of host protein synthesis. Knockdown of miR-141 reduces viral propagation, and silencing of eIF4E can completely reverse the inhibitory effect of the miR-141 antagomiR on viral propagation. Ectopic expression of miR-141 promotes the switch from cap-dependent to cap-independent translation. Moreover, we identified a transcription factor, EGR1, which is partly responsible for miR-141 induction in response to enterovirus infection. Our results suggest that upregulation of miR-141 upon enterovirus infection can facilitate viral propagation by expediting the translational switch.


Subject(s)
Enterovirus/pathogenicity , Eukaryotic Initiation Factor-4E/antagonists & inhibitors , MicroRNAs/biosynthesis , Protein Biosynthesis , Cell Line , Humans , Models, Biological
12.
Cancer Cell ; 13(1): 48-57, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18167339

ABSTRACT

We investigated whether microRNA expression profiles can predict clinical outcome of NSCLC patients. Using real-time RT-PCR, we obtained microRNA expressions in 112 NSCLC patients, which were divided into the training and testing sets. Using Cox regression and risk-score analysis, we identified a five-microRNA signature for the prediction of treatment outcome of NSCLC in the training set. This microRNA signature was validated by the testing set and an independent cohort. Patients with high-risk scores in their microRNA signatures had poor overall and disease-free survivals compared to the low-risk-score patients. This microRNA signature is an independent predictor of the cancer relapse and survival of NSCLC patients.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Gene Expression Regulation, Neoplastic , Lung Neoplasms/diagnosis , Lung Neoplasms/genetics , MicroRNAs/genetics , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , Aged , Carcinoma, Non-Small-Cell Lung/classification , Carcinoma, Non-Small-Cell Lung/pathology , Cohort Studies , Disease-Free Survival , Female , Humans , Kaplan-Meier Estimate , Lung Neoplasms/classification , Lung Neoplasms/pathology , Male , Neoplasm Invasiveness , Neoplasm Staging , Prognosis , Regression Analysis , Reproducibility of Results
13.
Nano Lett ; 8(2): 437-45, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18225938

ABSTRACT

Carbon nanotubes are a nanomaterial that is extensively used in industry. The potential health risk of chronic carbon nanotubes exposure has been raised as of great public concern. In the present study, we have demonstrated that intratracheal instillation of 0.5 mg of single-walled carbon nanotubes (SWCNT) into male ICR mice (8 weeks old) induced alveolar macrophage activation, various chronic inflammatory responses, and severe pulmonary granuloma formation. We then used Affymetrix microarrays to investigate the molecular effects on the macrophages when exposed to SWCNT. A biological pathway analysis, a literature survey, and experimental validation suggest that the uptake of SWCNT into the macrophages is able to activate various transcription factors such as nuclear factor kappaB (NF-kappaB) and activator protein 1 (AP-1), and this leads to oxidative stress, the release of proinflammatory cytokines, the recruitment of leukocytes, the induction of protective and antiapoptotic gene expression, and the activation of T cells. The resulting innate and adaptive immune responses may explain the chronic pulmonary inflammation and granuloma formation in vivo caused by SWCNT.


Subject(s)
Cytokines/immunology , Disease Models, Animal , Lung Diseases/chemically induced , Lung Diseases/immunology , Lung/immunology , Macrophages/immunology , Nanotubes, Carbon/toxicity , Animals , Cells, Cultured , Lung/drug effects , Lung/pathology , Lung Diseases/pathology , Macrophages/drug effects , Male , Mice , Mice, Inbred ICR , Nanotubes, Carbon/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL
...