Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
4.
Nat Cell Biol ; 18(9): 993-1005, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27525719

ABSTRACT

Epigenetic reprogramming has been associated with the functional plasticity of cancer-initiating cells (CICs); however, the regulatory pathway has yet to be elucidated. A siRNA screen targeting known epigenetic genes revealed that G9a profoundly impairs the chemo-resistance, self-renewal and metastasis of CICs obtained from patients with colorectal cancer (CRC). Patients with elevated G9a were shown to face a high risk of relapse and poor survival rates. From a mechanistic perspective, G9a binds with and stabilizes RelB, thereby recruiting DNA methyltransferase 3 on the Let-7b promoter and repressing its expression. This leads to the activation of the K-RAS/ß-catenin pathway and regulates self-renewal and function of CICs. These findings indicate that the G9a/RelB/Let-7b axis acts as a critical regulator in the maintenance of CIC phenotypes and is strongly associated with negative clinical outcomes. Thus, these findings may have diagnostic as well as therapeutic implications for the treatment of chemotherapy-resistant or metastatic CRC.


Subject(s)
Colorectal Neoplasms/metabolism , Drug Resistance, Neoplasm/genetics , Histocompatibility Antigens/genetics , Histone-Lysine N-Methyltransferase/genetics , MicroRNAs/genetics , Transcription Factor RelB/genetics , Animals , Colon/metabolism , Colorectal Neoplasms/pathology , Gene Silencing/physiology , Genes, ras/genetics , Humans , Mice , Neoplastic Stem Cells/metabolism , Phenotype , Wnt Signaling Pathway/genetics , Wnt Signaling Pathway/physiology , beta Catenin/genetics , beta Catenin/metabolism
5.
J Invest Dermatol ; 135(4): 1065-1073, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25380351

ABSTRACT

IGF II mRNA-binding protein 3 (IMP-3) has been reported to be a marker of melanoma progression. However, the mechanisms by which it impacts melanoma are incompletely understood. In this study, we investigate the clinical significance of IMP-3 in melanoma progression and also its underlying mechanisms. We found that IMP-3 expression was much higher in advanced-stage/metastatic melanomas and that it was associated with a poor prognosis (P=0.001). Univariate analysis showed that IMP-3 expression was associated with stage III/IV melanomas (odds ratio=5.40, P=0.031) and the acral lentiginous subtype (odds ratio=3.93, P=0.0034). MeWo cells with overexpression of IMP-3 showed enhanced proliferation and migration and significantly increased tumorigenesis and metastatic ability in nude mice. We further demonstrated that IMP-3 could bind and enhance the stability of the mRNA of high mobility group AT-hook 2 (HMGA2). It was also confirmed that IMP-3 had an important role in melanoma invasion and metastasis through regulating HMGA2 mRNA expression. IMP-3 expression was positively correlated with HMGA2 expression in melanoma cells and also in melanoma tissues. Our results show that IMP-3 expression is a strong prognostic factor for melanoma, especially acral lentiginous melanoma.


Subject(s)
Gene Expression Regulation, Neoplastic , HMGA2 Protein/metabolism , Melanoma/metabolism , RNA-Binding Proteins/metabolism , Skin Neoplasms/metabolism , Biomarkers, Tumor , Cell Line, Tumor , Cell Movement , Disease Progression , Humans , Neoplasm Invasiveness , Neoplasm Metastasis , Odds Ratio , Oligonucleotide Array Sequence Analysis , Prognosis , Melanoma, Cutaneous Malignant
6.
J Dermatol Sci ; 72(2): 123-33, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23856244

ABSTRACT

BACKGROUND: Higher CXCR4 expression enhances basal cell carcinoma (BCC) invasion and angiogenesis. The underlying mechanism of increased CXCR4 expression in invasive BCC is still not well understood. OBJECTIVE: To investigate the mechanisms involved in the regulation of CXCR4 expression in invasive BCC. METHODS: We used qRT-PCR, RT-PCR, Western blot, and flow cytometric analyses to examine different CXCR4 levels among the clinical samples, co-cultured BCC cells and BCC cells treated with recombinant transforming growth factor-ß1 (TGF-ß1) and connective tissue growth factor (CTGF). Immunohistochemical studies were used to demonstrate the correlation between TGF-ß1 and CXCR4 expressions. The signal transduction pathway and transcriptional regulation were confirmed by treatments with chemical inhibitors, neutralizing antibodies, or short interfering RNAs, as well as luciferase reporter activity. RESULTS: Invasive BCC has higher TGF-ß1 and CTGF levels compared to non-invasive BCC. Non-contact dermal fibroblasts co-culture with human BCC cells also increases the expression of CXCR4 in BCC cells. Treatment with recombinant human TGF-ß1, but not CTGF, enhanced the CXCR4 levels in time- and dose-dependent manners. The protein level and surface expression of CXCR4 in human BCC cells was increased by TGF-ß1 treatment. TGF-ß1 was intensely expressed in the surrounding fibroblasts of invasive BCC and was positively correlated with the CXCR4 expression of BCC cells. The transcriptional regulation of CXCR4 by TGF-ß1 is mediated by its binding to the TGF-ß receptor II and phosphorylation of the extracellular signal-related kinase 1/2 (ERK1/2)-ETS-1 pathway. CONCLUSION: TGF-ß1 induces upregulation of CXCR4 in human BCC cells by phosphorylation of ERK1/2-ETS-1 pathway.


Subject(s)
Carcinoma, Basal Cell/metabolism , Gene Expression Regulation, Neoplastic , Receptors, CXCR4/metabolism , Skin Neoplasms/metabolism , Transforming Growth Factor beta1/pharmacology , Cell Line, Tumor , Coculture Techniques , Connective Tissue Growth Factor/metabolism , Dose-Response Relationship, Drug , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibroblasts/cytology , Genes, Dominant , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Neoplasm Invasiveness , Neovascularization, Pathologic , Phosphorylation , Proto-Oncogene Protein c-ets-1/metabolism , Recombinant Proteins/pharmacology , Time Factors
7.
Oral Oncol ; 48(6): 507-15, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22366438

ABSTRACT

Stromal cell-derived factor-1 (SDF-1) (CXCL12) has been observed to promote laryngeal and hypopharyngeal squamous cell carcinomas (LHSCCs) invasion through cooperation with its receptor CXCR4. Here, we further explore the angiogenesis mechanism induced by SDF-1/CXCR4 interaction in LHSCCs. Immunohistochemistry (IHC) reveals the significant correlation between CXCR4 and angiogenesis in tumors. After blocking the function of CXCR4 by specific inhibitor AMD3100 and neutralized antibody 12G5 or inhibiting the expression by siRNA, we were able to disrupt the HUVECs tube formation, demonstrating that SDF-1/CXCR4 indeed regulated the angiogenesis mechanism. The angiogenesis profiling from angiogenesis array and reverse transcription polymerase chain reaction indicates that IL-8 can be significantly triggered by SDF-1/CXCR4 interaction in LHSCCs. We also demonstrate that IL-8 secretion mechanism is regulated by Akt phosphorylation after SDF-1 stimulation. These results point out the importance of SDF-1/CXCR4 interaction in LHSCCs angiogenesis. The angiogenic factor IL-8 would be triggered by the cooperation of SDF-1 and CXCR4 through an Akt-dependent pathway. This provides a new targeting therapy utility, disrupting SDF-1/CXCR4 interaction combined with downstream-induced angiogenic factors in LHSCCs would be beneficial to improve clinical outcome.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Hypopharyngeal Neoplasms/metabolism , Interleukin-8/metabolism , Laryngeal Neoplasms/metabolism , Blotting, Western , Cell Line, Tumor , Chemokine CXCL12/metabolism , Humans , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Real-Time Polymerase Chain Reaction , Receptors, CXCR4/metabolism , Reverse Transcriptase Polymerase Chain Reaction
8.
J Clin Invest ; 121(9): 3442-55, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21841313

ABSTRACT

MicroRNAs (miRNAs) influence many biological processes, including cancer. They do so by posttranscriptionally repressing target mRNAs to which they have sequence complementarity. Although it has been postulated that miRNAs can regulate other miRNAs, this has never been shown experimentally to our knowledge. Here, we demonstrate that miR-107 negatively regulates the tumor suppressor miRNA let-7 via a direct interaction. miR-107 was found to be highly expressed in malignant tissue from patients with advanced breast cancer, and its expression was inversely correlated with let-7 expression in tumors and in cancer cell lines. Ectopic expression of miR-107 in human cancer cell lines led to destabilization of mature let-7, increased expression of let-7 targets, and increased malignant phenotypes. In contrast, depletion of endogenous miR-107 dramatically increased the stability of mature let-7 and led to downregulation of let-7 targets. Accordingly, miR-107 expression increased the tumorigenic and metastatic potential of a human breast cancer cell line in mice via inhibition of let-7 and upregulation of let-7 targets. By mutating individual sites within miR-107 and let-7, we found that miR-107 directly interacts with let-7 and that the internal loop of the let-7/miR-107 duplex is critical for repression of let-7 expression. Altogether, we have identified an oncogenic role for miR-107 and provide evidence of a transregulational interaction among miRNAs in human cancer development.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , Gene Expression Regulation, Neoplastic , MicroRNAs/metabolism , Animals , Base Pairing , Base Sequence , Cell Line, Tumor , Disease Progression , Female , Humans , Mice , MicroRNAs/genetics , Middle Aged , Neoplasm Transplantation , Survival Rate
9.
Cancer Res ; 70(7): 2675-85, 2010 Apr 01.
Article in English | MEDLINE | ID: mdl-20233879

ABSTRACT

Hypoxia-inducible factor-1alpha (HIF-1alpha) is widely considered to be one of the key regulators of tumor angiogenesis. The upstream regulation is complex and involves several growth factors, cytokines, and hypoxia. Herein, we have identified miR-519c as a hypoxia-independent regulator of HIF-1alpha, acting through direct binding to the HIF-1alpha 3' untranslated region and leading to reduced tumor angiogenesis. Overexpression of miR-519c resulted in a significant decrease of HIF-1alpha protein levels and reduced the tube formation of human umbilical vein endothelial cells; similarly, antagomir inhibition of miR-519c increased the level of HIF-1alpha protein and enhanced angiogenic activity, suggesting an important role of miR-519c in HIF-1alpha-mediated angiogenesis. Consistent with the overexpression of miR-519c in cancer patients with better prognosis, mice injected with miR-519c-overexpressing cells exhibited dramatically reduced HIF-1alpha levels, followed by suppressed tumor angiogenesis, growth, and metastasis. In addition, we found that hepatocyte growth factor (HGF), a known HIF-1alpha inducer, reduced the miR-519c levels through an Akt-dependent pathway. This regulation was posttranscriptional and may be mediated by suppression of miR-519c maturation. Taken together, our findings provide the first evidence that miR-519c is a pivotal regulator of tumor angiogenesis and that microenvironmental HGF contributes to regulating miR-519c biogenesis in cancer cells.


Subject(s)
Adenocarcinoma/metabolism , Breast Neoplasms/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Lung Neoplasms/metabolism , MicroRNAs/metabolism , 3' Untranslated Regions , Adenocarcinoma/blood supply , Adenocarcinoma/genetics , Animals , Breast Neoplasms/blood supply , Breast Neoplasms/genetics , Cell Line, Tumor , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Lung Neoplasms/blood supply , Lung Neoplasms/genetics , Male , Mice , Mice, Inbred BALB C , MicroRNAs/biosynthesis , MicroRNAs/genetics , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , RNA Processing, Post-Transcriptional
10.
Carcinogenesis ; 30(2): 205-13, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18849299

ABSTRACT

Stromal cell-derived factor 1alpha (SDF-1alpha) (CXCL12) has been observed to enhance tumor angiogenesis. However, the comprehensive role of SDF-1alpha (CXCL12)-CXCR4 interaction, exerted during angiogenesis, has not been well understood. We have previously demonstrated that human basal cell carcinoma (BCC) tissues and a BCC cell line (BCC-1/KMC) had significant expression of CXCR4, whose level was higher in invasive than in the non-invasive BCC types. Here, we observed that human BCC tissues with high expression levels of CXCR4 had higher vascularity. Further, among the 71 BCCs diagnosed between the years 2004-2005, BCCs with high CXCR4 expression had concomitantly higher microvessel density, as compared with those with low CXCR4 expression (P < 0.001). We found that SDF-1alpha induced angiogenic activity in human BCC cells, both in vitro and in vivo. SDF-1alpha significantly upregulated several angiogenesis-associated genes such as interferon-alpha-inducible protein 27, interleukin (IL)-6, bone morphogenetic protein (BMP)-6, SOCS2 and cyclooxygenase 2 (COX)-2 in human BCC cells. Among them, IL-6 was the earliest and highest upregulated gene whose induction was observed within 6 h of the commencement of SDF-1alpha-CXCR4 interaction. The mechanisms behind the SDF-1alpha-induced time and dose-dependent upregulation of messenger RNA expression and protein secretion of IL-6 were investigated. The transcriptional regulation of IL-6 by SDF-1alpha was mediated by phosphorylation of extracellular signal-related kinase 1/2 and activation of the nuclear factor-kappaB complex. The identification of the angiogenic profiles induced through SDF-1alpha-CXCR4 interactions in human BCC cells may contribute further insights into the mechanisms involved in the angiogenic potential of SDF-1alpha (CXCL12).


Subject(s)
Carcinoma, Basal Cell/metabolism , Chemokine CXCL12/physiology , Interleukin-6/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , NF-kappa B/metabolism , Neovascularization, Pathologic/metabolism , Skin Neoplasms/metabolism , Biomarkers, Tumor/metabolism , Carcinoma, Basal Cell/blood supply , Carcinoma, Basal Cell/pathology , Cell Line, Tumor , Humans , Neovascularization, Pathologic/pathology , Phosphorylation , Receptors, CXCR4/metabolism , Skin Neoplasms/blood supply , Skin Neoplasms/pathology , Up-Regulation
11.
Carcinogenesis ; 29(8): 1519-27, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18487224

ABSTRACT

Laryngeal and hypopharyngeal squamous cell carcinomas (LHSCCs) are common head and neck cancers with a high propensity for lymph node (LN) and lung metastasis. Here, we report that LHSCCs express high levels of functional CXCR4 receptors, native for chemokine stromal cell-derived factor-1 (SDF-1/CXCL12). Primary tumor immunohistochemistry from LHSCC patients has revealed significant expression of CXCR4 and CXCL12. Greater expression of CXCR4 but not that of CXCL12 is correlated with LN and distant metastasis. Reverse transcription-polymerase chain reaction and western blots have demonstrated that CXCR4 messenger RNA (mRNA) and protein were expressed in LHSCC cell lines as well, but failed to detect CXCL12 mRNA expression. CXCL12 treatment enhanced extracellular signal-regulated kinase (ERK) pathway activation and the motility/invasiveness of LHSCC cell lines, which were blocked by treatment with a CXCR4 antagonist (AMD3100) and a specific MEK inhibitor (U0126). Results show that the mRNA and protein levels of matrix metalloproteinase (MMP)-13, but not MMP-2 or MMP-9, were elevated in HEp-2 cells in response to CXCL12. Again, U0126 almost inhibited the induction of MMP-13 in HEp-2 cells by stimulating CXCL12. The transcriptional factor, c-Jun, a downstream factor of ERK pathway, was found to be readily phosphorylated and translocated to the nucleus after 10 min of exposure to CXCL12. Blockage of c-Jun activity by transfection with c-jun antisense oligodeoxynucleotide significantly decreased CXCL12-induced MMP-13 expression and cell invasion. CXCL12 seems to enhance LHSCC cell invasion through paracrine-activated CXCR4, which triggers ERK/c-Jun-dependent MMP-13 upregulation.


Subject(s)
Carcinoma, Squamous Cell/pathology , Chemokine CXCL12/genetics , Hypopharyngeal Neoplasms/pathology , Laryngeal Neoplasms/pathology , Matrix Metalloproteinase 13/genetics , Receptors, CXCR4/genetics , Cell Line, Tumor , Cell Movement , Humans , Immunohistochemistry , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Neoplasm Invasiveness , Neoplasm Metastasis , Phosphorylation , Polymerase Chain Reaction , Transcription Factor AP-1/metabolism
12.
J Cell Sci ; 120(Pt 12): 2053-65, 2007 Jun 15.
Article in English | MEDLINE | ID: mdl-17550972

ABSTRACT

Connective tissue growth factor (CTGF) expression is elevated in advanced stages of breast cancer, but the regulatory role of CTGF in invasive breast cancer cell phenotypes is unclear. Presently, overexpression of CTGF in MCF-7 cells (MCF-7/CTGF cells) enhanced cellular migratory ability and spindle-like morphological alterations, as evidenced by actin polymerization and focal-adhesion-complex aggregation. Reducing the CTGF level in MDA-MB-231 (MDA231) cells by antisense CTGF cDNA (MDA231/AS cells) impaired cellular migration and promoted a change to an epithelial-like morphology. A neutralizing antibody against integrin alphavbeta3 significantly attenuated CTGF-mediated ERK1/2 activation and cellular migration, indicating that the integrin-alphavbeta3-ERK1/2 signaling pathway is crucial in mediating CTGF function. Moreover, the cDNA microarray analysis revealed CTGF-mediated regulation of the prometastatic gene S100A4. Transfection of MCF-7/CTGF cells with AS-S100A4 reversed the CTGF-induced cellular migratory ability, whereas overexpression of S100A4 in MDA231/AS cells restored their high migratory ability. Genetic and pharmacological manipulations suggested that the CTGF-mediated S100A4 upregulation was dependent on ERK1/2 activation, with expression levels of CTGF and S100A4 being closely correlated with human breast tumors. We conclude that CTGF plays a crucial role in migratory/invasive processes in human breast cancer by a mechanism involving activation of the integrin-alphavbeta3-ERK1/2-S100A4 pathway.


Subject(s)
Breast Neoplasms , Cell Movement/physiology , Immediate-Early Proteins/metabolism , Integrin alphaVbeta3/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , S100 Proteins/metabolism , Actins/metabolism , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Shape , Connective Tissue Growth Factor , Enzyme Activation , Female , Focal Adhesions/metabolism , Gene Expression Regulation, Neoplastic , Humans , Immediate-Early Proteins/genetics , Integrin alphaVbeta3/genetics , Intercellular Signaling Peptides and Proteins/genetics , Mice , Mice, Nude , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 3/genetics , Neoplasm Metastasis , Oligonucleotides, Antisense/genetics , Oligonucleotides, Antisense/metabolism , Paxillin/metabolism , S100 Calcium-Binding Protein A4 , S100 Proteins/genetics , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...