Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 61
Filter
6.
Cancer Res ; 77(8): 1955-1967, 2017 04 15.
Article in English | MEDLINE | ID: mdl-28209618

ABSTRACT

Ovarian cancer spheroids constitute a metastatic niche for transcoelomic spread that also engenders drug resistance. Spheroid-forming cells express active STAT3 signaling and display stem cell-like properties that may contribute to ovarian tumor progression. In this study, we show that STAT3 is hyperactivated in ovarian cancer spheroids and that STAT3 disruption in this setting is sufficient to relieve chemoresistance. In an NSG murine model of human ovarian cancer, STAT3 signaling regulated spheroid formation and self-renewal properties, whereas STAT3 attenuation reduced tumorigenicity. Mechanistic investigations revealed that Wnt signaling was required for STAT3-mediated spheroid formation. Notably, the Wnt antagonist DKK1 was the most strikingly upregulated gene in response to STAT3 attenuation in ovarian cancer cells. STAT3 signaling maintained stemness and interconnected Wnt/ß-catenin signaling via the miR-92a/DKK1-regulatory pathways. Targeting STAT3 in combination with paclitaxel synergistically reduced peritoneal seeding and prolonged survival in a murine model of intraperitoneal ovarian cancer. Overall, our findings define a STAT3-miR-92a-DKK1 pathway in the generation of cancer stem-like cells in ovarian tumors, with potential therapeutic applications in blocking their progression. Cancer Res; 77(8); 1955-67. ©2017 AACR.


Subject(s)
MicroRNAs/genetics , Neoplasms, Glandular and Epithelial/metabolism , Neoplasms, Glandular and Epithelial/pathology , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , STAT3 Transcription Factor/metabolism , Wnt Signaling Pathway , Animals , Carcinoma, Ovarian Epithelial , Disease Progression , Down-Regulation , Drug Resistance, Neoplasm , Female , Heterografts , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , MicroRNAs/metabolism , Neoplasms, Glandular and Epithelial/genetics , Ovarian Neoplasms/genetics , Phosphorylation , STAT3 Transcription Factor/genetics , Spheroids, Cellular , Transcription, Genetic , Tumor Cells, Cultured , beta Catenin/metabolism
8.
Oncotarget ; 8(3): 5603-5618, 2017 Jan 17.
Article in English | MEDLINE | ID: mdl-27901498

ABSTRACT

Vascular endothelial growth factor-C (VEGF-C) has been implicated in epithelial-mesenchymal transition (EMT) processes and various human cancers, including skin cancer. Skin cancer is an aggressive human malignancy with increasing incidence worldwide; however, the underlying mechanisms involved in VEGF-C-induced skin cancer stemness and metastasis remain unclear. Here, we report that VEGF-C enhances skin cancer migration, invasion and stemness through Slug up-regulation. Oncomine database analysis indicated that the KRAS/MAPK (mitogen-activated protein kinases) pathway and YAP1 (yes-associated protein 1) expression are positively correlated with metastatic skin cancer. We show that VEGF-C triggers the activation of KRAS/MAPK signaling to increase YAP1 and downstream Slug expression, which are suppressed by an anti-VEGFR3 (VEGF receptor 3) peptide, a specific peptide targeting VEGFR3. The VEGF-C-induced migration, invasion and stemness of skin cancer cells are also abrogated by the anti-VEGFR3 peptide. Based on these data, we reveal the role of the VEGF-C/VEGFR3-mediated KRAS/MAPK-YAP1/Slug pathway in skin cancer progression and propose that the VEGF-C/VEGFR3 axis is a promising target for the anti-VEGFR3 peptide.


Subject(s)
Neoplasm Invasiveness/pathology , Neoplastic Stem Cells/pathology , Signal Transduction/physiology , Skin Neoplasms/pathology , Cell Movement/physiology , Humans , Neoplastic Stem Cells/metabolism , Skin Neoplasms/metabolism , Snail Family Transcription Factors/metabolism , Vascular Endothelial Growth Factor C/metabolism , Vascular Endothelial Growth Factor Receptor-3/metabolism
9.
Oncotarget ; 7(39): 63924-63936, 2016 Sep 27.
Article in English | MEDLINE | ID: mdl-27590506

ABSTRACT

Epidermal growth factor receptor (EGFR) is commonly overexpressed in breast cancer and is associated with poor clinical outcomes; however, an increasing number of patients have shown a poor effective response to EGFR tyrosine kinase inhibitors (EGFR-TKI). Here, we found that AXL expression was positively correlated with poor progression in breast cancer patients. Suppression of AXL by an anti-tumor protein, E1A, enhanced EGFR-TKI (gefitinib, erlotinib and lapatinib) sensitization, resulting in significant inhibition of tumor growth in breast cancer cells. Additionally, AXL overexpression dramatically impaired E1A-mediated EGFR-TKI sensitization. These findings show that downregulation of AXL expression by E1A contributes to sensitization to EGFR-TKI in breast cancer, suggesting that combinatorial therapy of AXL inhibitors or E1A gene therapy with EGFR-TKI may be a potential therapeutic strategy for treatment of breast cancer patients.


Subject(s)
Adenovirus E1A Proteins/pharmacology , Breast Neoplasms/drug therapy , ErbB Receptors/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Animals , Apoptosis , Cell Line, Tumor , Cell Survival , Disease Progression , Down-Regulation , Drug Resistance, Neoplasm , Erlotinib Hydrochloride/administration & dosage , Female , Gefitinib , Genetic Therapy , Humans , Kaplan-Meier Estimate , Lapatinib , Mice , Mice, SCID , Neoplasm Transplantation , Prognosis , Quinazolines/administration & dosage , Axl Receptor Tyrosine Kinase
10.
Hepatology ; 64(5): 1637-1651, 2016 11.
Article in English | MEDLINE | ID: mdl-27530187

ABSTRACT

Angiopoietin-like protein 1 (ANGPTL1) has been shown to act as a tumor suppressor by inhibiting angiogenesis, cancer invasion, and metastasis. However, little is known about the effects of ANGPTL1 on sorafenib resistance and cancer stem cell properties in hepatocellular carcinoma (HCC) and the mechanism underlying these effects. Here, we show that ANGPTL1 expression positively correlates with sorafenib sensitivity in HCC cells and human HCC tissues. ANGPTL1 significantly decreases epithelial-mesenchymal transition (EMT)-driven sorafenib resistance, cancer stemness, and tumor growth of HCC cells by repressing Slug expression. ANGPTL1 directly interacts with and inactivates MET receptor, which contributes to Slug suppression through inhibition of the extracellular receptor kinase/protein kinase B (ERK/AKT)-dependent early growth response protein 1 (Egr-1) pathway. ANGPTL1 expression inversely correlates with Slug expression, poor sorafenib responsiveness, and poor clinical outcomes in HCC patients. CONCLUSION: ANGPTL1 inhibits sorafenib resistance and cancer stemness in HCC cells by repressing EMT through inhibition of the MET receptor-AKT/ERK-Egr-1-Slug signaling cascade. ANGPTL1 may serve as a novel MET receptor inhibitor for advanced HCC therapy. (Hepatology 2016;64:1637-1651).


Subject(s)
Angiopoietins/physiology , Antineoplastic Agents/therapeutic use , Carcinoma, Hepatocellular/drug therapy , Drug Resistance, Neoplasm , Liver Neoplasms/drug therapy , Niacinamide/analogs & derivatives , Phenylurea Compounds/therapeutic use , Proto-Oncogene Proteins c-met/physiology , Angiopoietin-Like Protein 1 , Angiopoietin-like Proteins , Animals , Female , Humans , Male , Mice , Middle Aged , Neoplastic Stem Cells , Niacinamide/therapeutic use , Sorafenib
11.
Cancer Res ; 76(13): 3916-28, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27216190

ABSTRACT

Paclitaxel is a standard-of-care chemotherapy for breast cancer, despite the increasing recognition of its poor effectiveness in the treatment of patients with advanced disease. Here, we report that adenovirus-type 5 E1A-mediated elevation of the miRNA-processing enzyme Dicer is sufficient to enhance paclitaxel sensitization and reduce cancer stem-like cell properties in this setting. Elevating Dicer expression increased levels of the AXL kinase targeting miRNA miR-494, thereby repressing AXL expression to increase paclitaxel sensitivity. We found that Dicer expression was regulated at the transcription level by E1A, through activation of an MAPK14/CEBPα pathway. Our findings define a mechanism of E1A-mediated chemosensitization for paclitaxel, which is based upon the suppression of breast cancer stem-like cells, with potential implications for the diagnosis and treatment of breast cancer patients. Cancer Res; 76(13); 3916-28. ©2016 AACR.


Subject(s)
Biomarkers, Tumor/metabolism , Breast Neoplasms/drug therapy , DEAD-box RNA Helicases/metabolism , Neoplastic Stem Cells/drug effects , Paclitaxel/pharmacology , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Ribonuclease III/metabolism , Adenoviridae/genetics , Adenovirus E1A Proteins/metabolism , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , CCAAT-Enhancer-Binding Proteins/metabolism , Female , Humans , Mice , Mice, Inbred NOD , Mice, SCID , MicroRNAs/genetics , Mitogen-Activated Protein Kinase 14/metabolism , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , Axl Receptor Tyrosine Kinase
12.
Proc Natl Acad Sci U S A ; 113(18): E2526-35, 2016 May 03.
Article in English | MEDLINE | ID: mdl-27091996

ABSTRACT

Therapy with epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (EGFR-TKIs, such as gefitinib or erlotinib) significantly prolongs survival time for patients with tumors harboring an activated mutation on EGFR; however, up to 40% of lung cancer patients exhibit acquired resistance to EGFR-TKIs with an unknown mechanism. FOXO3a, a transcription factor of the forkhead family, triggers apoptosis, but the mechanistic details involved in EGFR-TKI resistance and cancer stemness remain largely unclear. Here, we observed that a high level of FOXO3a was correlated with EGFR mutation-independent EGFR-TKI sensitivity, the suppression of cancer stemness, and better progression-free survival in lung cancer patients. The suppression of FOXO3a obviously increased gefitinib resistance and enhanced the stem-like properties of lung cancer cells; consistent overexpression of FOXO3a in gefitinib-resistant lung cancer cells reduced these effects. Moreover, we identified that miR-155 targeted the 3'UTR of FOXO3a and was transcriptionally regulated by NF-κB, leading to repressed FOXO3a expression and increased gefitinib resistance, as well as enhanced cancer stemness of lung cancer in vitro and in vivo. Our findings indicate that FOXO3a is a significant factor in EGFR mutation-independent gefitinib resistance and the stemness of lung cancer, and suggest that targeting the NF-κB/miR-155/FOXO3a pathway has potential therapeutic value in lung cancer with the acquisition of resistance to EGFR-TKIs.


Subject(s)
ErbB Receptors/metabolism , Forkhead Box Protein O3/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , NF-kappa B/metabolism , Quinazolines/administration & dosage , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/administration & dosage , Dose-Response Relationship, Drug , Down-Regulation/drug effects , Drug Resistance, Neoplasm , ErbB Receptors/genetics , Female , Gefitinib , Humans , Lung Neoplasms/pathology , Male , Middle Aged , Mutation , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Protein Kinase Inhibitors/administration & dosage , Treatment Outcome , Tumor Cells, Cultured
13.
Oncotarget ; 6(30): 28755-73, 2015 Oct 06.
Article in English | MEDLINE | ID: mdl-26356813

ABSTRACT

Lung adenocarcinoma is one of the most deadly human diseases. However, the molecular mechanisms underlying this disease, particularly RNA splicing, have remained underexplored. Here, we report a triple-level (gene-, transcript-, and exon-level) analysis of lung adenocarcinoma transcriptomes from 77 paired tumor and normal tissues, as well as an analysis pipeline to overcome genetic variability for accurate differentiation between tumor and normal tissues. We report three major results. First, more than 5,000 differentially expressed transcripts/exonic regions occur repeatedly in lung adenocarcinoma patients. These transcripts/exonic regions are enriched in nicotine metabolism and ribosomal functions in addition to the pathways enriched for differentially expressed genes (cell cycle, extracellular matrix receptor interaction, and axon guidance). Second, classification models based on rationally selected transcripts or exonic regions can reach accuracies of 0.93 to 1.00 in differentiating tumor from normal tissues. Of the 28 selected exonic regions, 26 regions correspond to alternative exons located in such regulators as tumor suppressor (GDF10), signal receptor (LYVE1), vascular-specific regulator (RASIP1), ubiquitination mediator (RNF5), and transcriptional repressor (TRIM27). Third, classification systems based on 13 to 14 differentially expressed genes yield accuracies near 100%. Genes selected by both detection methods include C16orf59, DAP3, ETV4, GABARAPL1, PPAR, RADIL, RSPO1, SERTM1, SRPK1, ST6GALNAC6, and TNXB. Our findings imply a multilayered lung adenocarcinoma regulome in which transcript-/exon-level regulation may be dissociated from gene-level regulation. Our described method may be used to identify potentially important genes/transcripts/exonic regions for the tumorigenesis of lung adenocarcinoma and to construct accurate tumor vs. normal classification systems for this disease.


Subject(s)
Adenocarcinoma/genetics , Biomarkers, Tumor/genetics , Exons , Lung Neoplasms/genetics , RNA, Messenger/genetics , Signal Transduction/genetics , Transcriptome/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Adenovirus E1A Proteins/genetics , Adenovirus E1A Proteins/metabolism , Aldehyde Dehydrogenase/genetics , Aldehyde Dehydrogenase/metabolism , Biomarkers, Tumor/metabolism , Cell Proliferation , Databases, Genetic , Gene Expression Profiling/methods , Genetic Association Studies , Genetic Predisposition to Disease , HEK293 Cells , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Phenotype , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-ets , RNA Interference , RNA, Messenger/metabolism , Time Factors , Transfection
14.
Oncotarget ; 6(13): 11434-46, 2015 May 10.
Article in English | MEDLINE | ID: mdl-25825984

ABSTRACT

Chondrosarcoma is a malignant tumor that produces cartilage matrix. The most lethal aspect is its metastatic property. We demonstrated that amphiregulin (AR) is significantly upregulated in highly aggressive cells. AR silencing markedly suppressed cell migration. Exogenous AR markedly increased cell migration by transactivation of α6ß1 integrin expression. A neutralizing α6ß1 integrin antibody can abolish AR-induced cell motility. Knockdown of AR inhibits metastasis of cells to the lung in vivo. Furthermore, elevated AR expression is positively correlated with α6ß1 integrin levels and higher grades in patients. These findings can potentially serve as biomarker and therapeutic approach for controlling chondrosarcoma metastasis.


Subject(s)
Biomarkers, Tumor/metabolism , Bone Neoplasms/enzymology , Cell Movement/drug effects , Chondrosarcoma/enzymology , EGF Family of Proteins/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Integrin alpha6beta1/metabolism , MAP Kinase Kinase 1/metabolism , Transcription Factor AP-1/metabolism , raf Kinases/metabolism , ras Proteins/metabolism , Amphiregulin , Animals , Antibodies/pharmacology , Antineoplastic Agents/pharmacology , Biomarkers, Tumor/genetics , Bone Neoplasms/drug therapy , Bone Neoplasms/genetics , Bone Neoplasms/pathology , Cell Line, Tumor , Chondrosarcoma/drug therapy , Chondrosarcoma/genetics , Chondrosarcoma/secondary , Dose-Response Relationship, Drug , EGF Family of Proteins/genetics , EGF Family of Proteins/pharmacology , Gene Expression Regulation, Neoplastic , Humans , Integrin alpha6beta1/antagonists & inhibitors , Integrin alpha6beta1/genetics , Lung Neoplasms/enzymology , Lung Neoplasms/prevention & control , Lung Neoplasms/secondary , MAP Kinase Kinase 1/genetics , Male , Mice, Nude , Neoplasm Grading , Neoplasm Invasiveness , Phosphorylation , RNA Interference , Signal Transduction , Time Factors , Transcription Factor AP-1/genetics , Transfection , Xenograft Model Antitumor Assays , raf Kinases/genetics , ras Proteins/genetics
15.
Am J Respir Crit Care Med ; 192(1): 64-75, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-25906011

ABSTRACT

RATIONALE: CARD-recruited membrane-associated protein 3 (CARMA3) is a novel scaffold protein that regulates nuclear factor (NF)-κB activation; however, the underlying mechanism of CARMA3 in lung cancer stemness and metastasis remains largely unknown. OBJECTIVES: To investigate the molecular mechanisms underlying the involvement of CARMA3 in non-small cell lung cancer progression. METHODS: The expression levels of CARMA3 and NME2 in a cohort of patients with lung cancer (n = 91) were examined by immunohistochemistry staining and assessed by Kaplan-Meier survival analysis. The effects of CARMA3, microRNA-182 (miR-182), and NME2 on cancer stemness and metastasis were measured in vitro and in vivo. Chromatin immunoprecipitation and luciferase reporter assays were performed to determine the mechanisms of NF-κB-driven miR-182 expression and NME2 regulation. MEASUREMENTS AND MAIN RESULTS: We observed that CARMA3 inversely correlated with NME2 expression in patients with lung cancer (Pearson correlation coefficient: R = -0.24; P = 0.022). NME2 levels were significantly decreased in tumor tissues compared with adjacent normal lung tissues (P < 0.001), and patients with lung cancer with higher levels of NME2 had longer survival outcomes (overall survival, P < 0.01; disease-free survival, P < 0.01). Mechanistically, CARMA3 promoted cell motility by reducing the level of NME2 through the NF-κB/miR-182 pathway and by increasing cancer stem cell properties and metastasis in lung cancer. CONCLUSIONS: We identified a novel mechanism of CARMA3 in lung cancer stemness and metastasis through the negative regulation of NME2 by NF-κB-dependent induction of miR-182. Our findings provide an attractive strategy for targeting the CARMA3/NF-κB/miR-182 pathway as a potential treatment for lung cancer.


Subject(s)
Biomarkers, Tumor/metabolism , CARD Signaling Adaptor Proteins/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Lung Neoplasms/metabolism , NM23 Nucleoside Diphosphate Kinases/metabolism , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Non-Small-Cell Lung/pathology , Case-Control Studies , Cell Line, Tumor , Cell Transformation, Neoplastic , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/genetics , Lung Neoplasms/mortality , Lung Neoplasms/pathology , MicroRNAs/metabolism , NF-kappa B/metabolism , Neoplasm Metastasis , Survival Analysis
16.
Ann Surg Oncol ; 22(3): 889-98, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25212833

ABSTRACT

BACKGROUND: Triple-negative breast cancer (TNBC) is defined by reduced expression of the estrogen receptor, progesterone receptor, and HER2. TNBC is an especially aggressive group of breast cancers with poor prognosis. There are currently no validated molecular targets to effectively treat this disease. Thus, it is necessary to identify effective molecular targets and therapeutic strategies for TNBC patients. METHODS: The expression of HSPA5 in patients with breast cancer was examined by immunohistochemistry. The association of HSPA5 expression with tumor grade and metastatic events in TNBC patients was analyzed using the Oncomine database. The knockdown and overexpression of HSPA5 protein were performed to investigate the effects on E1A-suppressed cell migration/invasion of TNBC using in vitro transwell assays and tumor growth/experimental metastasis studies in animal models. RESULTS: The expression of HSPA5 was positively correlated with high-grade tumors, metastatic events, and poor overall survival in breast cancer patients with TNBC. E1A-inhibited HSPA5 expression suppressed cell migration/invasive ability of TNBC cell lines. Moreover, E1A significantly abolished lung metastases from breast cancer cells by inhibiting HSPA5 expression in a xenograft tumor model. CONCLUSIONS: The overexpression of HSPA5 is critical for high-risk metastasis of breast cancer and TNBC. The results of our study suggest that HSPA5 may be a crucial mediator of E1A-suppressed metastatic ability of breast cancer cells. Thus, E1A may be a potential target for diagnosis and individualized treatment in clinical practice.


Subject(s)
Adenovirus E1A Proteins/genetics , Cell Movement , Cell Proliferation , Heat-Shock Proteins/antagonists & inhibitors , Lung Neoplasms/prevention & control , Triple Negative Breast Neoplasms/prevention & control , Animals , Apoptosis , Endoplasmic Reticulum Chaperone BiP , Female , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Humans , Immunoenzyme Techniques , Lung Neoplasms/metabolism , Lung Neoplasms/mortality , Lung Neoplasms/secondary , Mice , Mice, SCID , Neoplasm Grading , Neoplasm Invasiveness , Neoplasm Staging , Prognosis , Survival Rate , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/mortality , Triple Negative Breast Neoplasms/pathology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
17.
PLoS One ; 9(11): e112636, 2014.
Article in English | MEDLINE | ID: mdl-25390068

ABSTRACT

Bone morphogenic protein (BMP)-7 is a member of the transforming growth factor (TGF)-beta superfamily, which is originally identified based on its ability to induce cartilage and bone formation. In recent years, BMP-7 is also defined as a potent promoter of cell motility, invasion, and metastasis. However, there is little knowledge of the role of BMP-7 and its cellular function in chondrosarcoma cells. In the present study, we investigated the biological impact of BMP-7 on cell motility using transwell assay. In addition, the intracellular signaling pathways were also investigated by pharmacological and genetic approaches. Our results demonstrated that treatment with exogenous BMP-7 markedly increased cell migration by activating c-Src/PI3K/Akt/IKK/NF-κB signaling pathway, resulting in the transactivation of αvß3 integrin expression. Indeed, abrogation of signaling activation, by chemical inhibition or expression of a kinase dead form of the protein attenuated BMP-7-induced expression of integrin αvß3 and cell migration. These findings may provide a useful tool for diagnostic/prognostic purposes and even therapeutically in late-stage chondrosarcoma as an anti-metastatic agent.


Subject(s)
Bone Morphogenetic Protein 7/pharmacology , Bone Neoplasms/metabolism , Cell Movement/drug effects , Chondrosarcoma/metabolism , Genes, src/physiology , Integrin alphaVbeta3/metabolism , Signal Transduction/drug effects , Cell Line, Tumor , Humans , NF-kappa B/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Up-Regulation/drug effects
18.
Oncotarget ; 5(21): 10558-70, 2014 Nov 15.
Article in English | MEDLINE | ID: mdl-25301734

ABSTRACT

Elevated expression of heat shock protein 5 (HSPA5) promotes drug resistance and metastasis and is a marker of poor prognosis in breast cancer patients. Adenovirus type 5 E1A gene therapy has demonstrated antitumor efficacy but the mechanisms of metastasis-inhibition are unclear. Here, we report that E1A interacts with p300 histone acetyltransferase (HAT) and blocks p300-mediated HSPA5 acetylation at K353, which in turn promotes HSPA5 ubiquitination by GP78 (E3 ubiquitin ligase) and subsequent proteasome-mediated degradation. Our findings point out the Ying-Yang regulation of two different post-translational modifications (ubiquitination and acetylation) of HSPA5 in tumor metastasis.


Subject(s)
Adenovirus E1A Proteins/metabolism , Breast Neoplasms/prevention & control , Cell Movement , E1A-Associated p300 Protein/metabolism , Heat-Shock Proteins/metabolism , Lung Neoplasms/prevention & control , Acetylation , Adenovirus E1A Proteins/genetics , Animals , Apoptosis , Blotting, Western , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Proliferation , E1A-Associated p300 Protein/genetics , Endoplasmic Reticulum Chaperone BiP , Female , Heat-Shock Proteins/genetics , Humans , Immunoenzyme Techniques , Immunoprecipitation , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Mice , Mice, SCID , Protein Binding , Protein Processing, Post-Translational , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , Ubiquitination , Xenograft Model Antitumor Assays
19.
Ann Surg Oncol ; 21 Suppl 4: S767-75, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25212831

ABSTRACT

BACKGROUND: Vascular endothelial growth factor-C (VEGF-C) plays an important role during cancer progression and metastasis through activation of VEGF receptors. However, the role of VEGF-C in esophageal squamous cell carcinoma (ESCC) remains unclear. METHODS: The expression of VEGF-C in advanced stages of esophageal cancer was examined by immunohistochemistry and its expression was correlated with the protein level of cortactin (CTTN) by Western blot. Knockdown and overexpression of the CTTN protein were respectively performed to investigate the effects on VEGF-C-enhanced ESCC migration/invasion by in vitro transwell assay, cell tracing assay, and tumor growth/experimental metastasis in animal models. RESULTS: The expression of VEGF-C was positively correlated with tumor status and poor clinical prognosis in patient with esophageal cancer. VEGF-C-upregulated CTTN expression contributed the migration/invasive abilities of ESCC cell lines through Src-mediated downregulation of miR-326. Moreover, knockdown of CTTN expression significantly abolished VEGF-C-induced tumor growth and experimental lung metastasis in vivo. CONCLUSIONS: Upregulation of CTTN is critical for VEGF-C-mediated tumor growth and metastasis of ESCC. These finding suggest that VEGF-C upregulated CTTN expression through Src-mediated downregulation of miR-326. CTTN may be a crucial mediator of VEGF-C-involved ESCC metastasis, which provides a potential target for diagnosis and individualized treatment in clinical practice.


Subject(s)
Carcinoma, Squamous Cell/chemistry , Carcinoma, Squamous Cell/genetics , Cortactin/analysis , Cortactin/genetics , Esophageal Neoplasms/chemistry , Esophageal Neoplasms/genetics , Lung Neoplasms/genetics , Vascular Endothelial Growth Factor C/analysis , Animals , Carcinoma, Squamous Cell/secondary , Cell Line, Tumor , Cell Movement , Cell Tracking , Cortactin/metabolism , Down-Regulation , Esophageal Neoplasms/pathology , Gene Knockdown Techniques , Humans , Lung Neoplasms/secondary , Mice, SCID , MicroRNAs/metabolism , Proto-Oncogene Proteins pp60(c-src)/genetics , Proto-Oncogene Proteins pp60(c-src)/metabolism , Signal Transduction/genetics , Transfection , Up-Regulation , Vascular Endothelial Growth Factor C/metabolism
20.
Cancer Res ; 74(21): 6280-90, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-25205106

ABSTRACT

Esophageal cancer is an aggressive human malignancy with increasing incidence in the developed world. VEGF-C makes crucial contributions to esophageal cancer progression that are not well understood. Here, we report the discovery of regulatory relationship in esophageal cancers between the expression of VEGF-C and cortactin (CTTN), a regulator of the cortical actin cytoskeleton. Upregulation of CTTN expression by VEGF-C enhanced the invasive properties of esophageal squamous cell carcinoma in vitro and tumor metastasis in vivo. Mechanistic investigations showed that VEGF-C increased CTTN expression by downregulating Dicer-mediated maturation of miR326, thereby relieving the suppressive effect of miR326 on CTTN expression. Clinically, expression of Dicer and miR326 correlated with poor prognosis in patients with esophageal cancer. Our findings offer insights into how VEGF-C enhances the robust invasive and metastatic properties of esophageal cancer, which has potential implications for the development of new biomarkers or therapies in this setting.


Subject(s)
Carcinoma, Squamous Cell/genetics , Cortactin/metabolism , Esophageal Neoplasms/genetics , MicroRNAs/genetics , Vascular Endothelial Growth Factor C/genetics , Biomarkers, Tumor , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Cortactin/genetics , Disease Progression , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma , Gene Expression Regulation, Neoplastic , Humans , Prognosis , Vascular Endothelial Growth Factor C/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...