Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Oncotarget ; 5(14): 5725-35, 2014 Jul 30.
Article in English | MEDLINE | ID: mdl-25015418

ABSTRACT

Tumor relapse after therapy typifies hepatocellular carcinoma (HCC) and is believed to be attributable to residual cancer stem cells (CSCs) that survive treatment. We have previously identified a CSC population derived from HCC that is characterized by CD133. Despite our growing knowledge of the importance of this subset of cells in driving HCC, the regulatory mechanism of CD133 is not known. Epigenetic changes are believed to be essential in the control of cancer and stem cells. Here, we report the epigenetic regulation of CD133 by miR-142-3p. The interaction between CD133 and miR-142-3p was identified by in silico prediction and substantiated by luciferase reporter analysis. Expression of CD133 was found to be inversely correlated with miR-142-3p in HCC clinical samples as well as in cell lines. Importantly, lower miR-142-3p expression in HCC was significantly associated with worst survival. Functional studies with miR-142-3p stably transduced in HCC cells demonstrated a diminished ability to self-renew, initiate tumor growth, invade, migrate, induce angiogenesis and resist chemotherapy. Rescue experiments whereby CD133 and miR-142-3p is simultaneously overexpressed compensated the deregulated ability of the cells to confer these features. Thus, miR-142-3p directly targets CD133 to regulate its ability to confer cancer and stem cell-like features in HCC.


Subject(s)
Antigens, CD/metabolism , Carcinoma, Hepatocellular/metabolism , Glycoproteins/metabolism , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , MicroRNAs/metabolism , Neoplastic Stem Cells/metabolism , Peptides/metabolism , AC133 Antigen , Animals , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Hep G2 Cells , Heterografts , Humans , Liver Neoplasms/genetics , Mice , Mice, Nude , MicroRNAs/genetics , Neoplastic Stem Cells/pathology , Transfection
2.
Cancer Res ; 73(7): 2322-32, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23382045

ABSTRACT

Tumor-initiating cells (TIC), also known as cancer stem cells, are regarded widely as a specific subpopulation of cells needed for cancer initiation and progression. TICs have yet to be identified in esophageal tumors that have an increasing incidence in developed countries. Here, we report a CD90(+) cell population found in esophageal squamous cell carcinoma (ESCC), which is endowed with stem cell-like properties and high tumorigenic and metastatic potential. mRNA profiling of these cells suggested pathways through which they drive tumor growth and metastasis, with deregulation of an Ets-1/MMP signaling pathway and epithelial-mesenchymal transition figuring prominently. These cells possessed higher self-renewal activity and were sufficient for tumor growth, differentiation, metastasis, and chemotherapeutic resistance. CD90(+) TICs were isolated and characterized from ESCC clinical specimens as well as ESCC cell lines. In freshly resected clinical specimens, they represented a rare cell population, the levels of which correlated with strong family histories and lymph node metastasis. Our results prompt further study of this CD90(+) population of esophageal TICs as potential therapeutic targets.


Subject(s)
Carcinoma, Squamous Cell/secondary , Cell Movement , Cell Proliferation , Epithelial-Mesenchymal Transition , Esophageal Neoplasms/pathology , Neoplastic Stem Cells/pathology , Thy-1 Antigens/metabolism , Animals , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Blotting, Western , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Cell Adhesion , Cell Differentiation , Disease Progression , Esophageal Neoplasms/genetics , Esophageal Neoplasms/metabolism , Female , Flow Cytometry , Gene Expression Profiling , Humans , Immunoenzyme Techniques , Liver Neoplasms, Experimental/genetics , Liver Neoplasms, Experimental/metabolism , Liver Neoplasms, Experimental/secondary , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Lymphatic Metastasis , Male , Mice , Mice, Inbred NOD , Mice, SCID , Middle Aged , Neoplasm Staging , Neoplastic Stem Cells/metabolism , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Thy-1 Antigens/chemistry , Thy-1 Antigens/genetics
3.
Carcinogenesis ; 34(4): 750-9, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23239745

ABSTRACT

Mitotic progression of mammalian cells is tightly regulated by the E3 ubiquitin ligase anaphase promoting complex (APC)/C. Deregulation of APC/C is frequently observed in cancer cells and is suggested to contribute to chromosome instability and cancer predisposition. In this study, we identified Daxx as a novel APC/C inhibitor frequently overexpressed in prostate cancer. Daxx interacts with the APC/C coactivators Cdc20 and Cdh1 in vivo, with the binding of Cdc20 dependent on the consensus destruction boxes near the N-terminal of the Daxx protein. Ectopic expression of Daxx, but not the D-box deleted mutant (DaxxΔD-box), inhibited the degradation of APC/Cdc20 and APC/Cdh1 substrates, leading to a transient delay in mitotic progression. Daxx is frequently upregulated in prostate cancer tissues; the expression level positively correlated with the Gleason score and disease metastasis (P = 0.027 and 0.032, respectively). Furthermore, ectopic expression of Daxx in a non-malignant prostate epithelial cell line induced polyploidy under mitotic stress. Our data suggest that Daxx may function as a novel APC/C inhibitor, which promotes chromosome instability during prostate cancer development.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Adenomatous Polyposis Coli Protein/antagonists & inhibitors , Cadherins/metabolism , Cell Cycle Proteins/metabolism , Chromosomal Instability , Mitosis , Nuclear Proteins/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adenomatous Polyposis Coli Protein/metabolism , Antigens, CD , Cdc20 Proteins , Cell Cycle , Cell Cycle Checkpoints , Cell Cycle Proteins/genetics , Cell Line, Tumor , Co-Repressor Proteins , HEK293 Cells , HeLa Cells , Humans , Male , Molecular Chaperones , Mutation , Neoplasm Grading , Neoplasm Metastasis , Nuclear Proteins/genetics , RNA Interference , RNA, Small Interfering
4.
Cancer Res ; 72(22): 6024-35, 2012 Nov 15.
Article in English | MEDLINE | ID: mdl-22991305

ABSTRACT

Esophageal squamous cell carcinoma (ESCC), the major histologic subtype of esophageal cancer, is a devastating disease characterized by distinctly high incidences and mortality rates. However, there remains limited understanding of molecular events leading to development and progression of the disease, which are of paramount importance to defining biomarkers for diagnosis, prognosis, and personalized treatment. By high-throughout transcriptome sequence profiling of nontumor and ESCC clinical samples, we identified a subset of significantly differentially expressed genes involved in integrin signaling. The Rab25 gene implicated in endocytic recycling of integrins was the only gene in this group significantly downregulated, and its downregulation was confirmed as a frequent event in a second larger cohort of ESCC tumor specimens by quantitative real-time PCR and immunohistochemical analyses. Reduced expression of Rab25 correlated with decreased overall survival and was also documented in ESCC cell lines compared with pooled normal tissues. Demethylation treatment and bisulfite genomic sequencing analyses revealed that downregulation of Rab25 expression in both ESCC cell lines and clinical samples was associated with promoter hypermethylation. Functional studies using lentiviral-based overexpression and suppression systems lent direct support of Rab25 to function as an important tumor suppressor with both anti-invasive and -angiogenic abilities, through a deregulated FAK-Raf-MEK1/2-ERK signaling pathway. Further characterization of Rab25 may provide a prognostic biomarker for ESCC outcome prediction and a novel therapeutic target in ESCC treatment.


Subject(s)
Carcinoma, Squamous Cell/genetics , Esophageal Neoplasms/genetics , Genes, Tumor Suppressor , rab GTP-Binding Proteins/genetics , Animals , Base Sequence , Carcinoma, Squamous Cell/blood supply , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cell Movement/genetics , DNA Methylation , Down-Regulation , Esophageal Neoplasms/blood supply , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/pathology , Gene Expression Profiling , Humans , MAP Kinase Signaling System , Mice , Mice, Nude , Molecular Sequence Data , Neoplasm Invasiveness , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Promoter Regions, Genetic , rab GTP-Binding Proteins/biosynthesis
5.
Gastroenterology ; 143(3): 675-686.e12, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22705009

ABSTRACT

BACKGROUND & AIMS: Esophageal squamous cell carcinoma (ESCC) is the most commonly observed histologic subtype of esophageal cancer. ESCC is believed to develop via accumulation of numerous genetic alterations, including inactivation of tumor suppressor genes and activation of oncogenes. We searched for transcripts that were altered in human ESCC samples compared with nontumor tissues. METHODS: We performed integrative transcriptome sequencing (RNA-Seq) analysis using ESCC samples from 3 patients and adjacent nontumor tissues to identify transcripts that were altered in ESCC tissue. We performed molecular and functional studies of the transcripts identified and investigated the mechanisms of alteration. RESULTS: We identified protein tyrosine kinase 6 (PTK6) as a transcript that was significantly down-regulated in ESCC tissues and cell lines compared with nontumor tissues or immortalized normal esophageal cell lines. The promoter of the PTK6 gene was inactivated in ESCC tissues at least in part via hypermethylation and histone deacetylation. Knockdown of PTK6 in KYSE30 ESCC cells using small hairpin RNAs increased their ability to form foci, migrate, and invade extracellular matrix in culture and form tumors in nude mice. Overexpression of PTK6 in these cells reduced their proliferation in culture and tumor formation in mice. PTK6 reduced phosphorylation of Akt and glycogen synthase kinase (GSK)3ß, leading to activation of ß-catenin. CONCLUSIONS: PTK6 was identified as a transcript that is down-regulated in human ESCC tissues via epigenetic modification at the PTK6 locus. Its product appears to regulate cell proliferation by reducing phosphorylation of Akt and GSK3ß, leading to activation of ß-catenin. Reduced levels of PTK6 promote growth of xenograft tumors in mice; it might be developed as a marker of ESCC.


Subject(s)
Carcinoma, Squamous Cell/enzymology , Esophageal Neoplasms/enzymology , Neoplasm Proteins/metabolism , Protein-Tyrosine Kinases/metabolism , Tumor Suppressor Proteins/metabolism , Acetylation , Adult , Animals , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cell Movement , Cell Proliferation , DNA Methylation , Epigenesis, Genetic , Esophageal Neoplasms/genetics , Esophageal Neoplasms/pathology , Extracellular Matrix/metabolism , Female , G1 Phase Cell Cycle Checkpoints , Gene Expression Profiling , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Histones/metabolism , Humans , Male , Mice , Mice, Nude , Middle Aged , Neoplasm Invasiveness , Neoplasm Proteins/genetics , Phosphorylation , Promoter Regions, Genetic , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , RNA, Messenger/metabolism , Sequence Analysis, RNA , Signal Transduction , Transcription, Genetic , Transfection , Tumor Suppressor Proteins/genetics , beta Catenin/metabolism
6.
Hepatology ; 55(3): 807-20, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21994122

ABSTRACT

UNLABELLED: A novel theory in the field of tumor biology postulates that cancer growth is driven by a population of stem-like cells, called tumor-initiating cells (TICs). We previously identified a TIC population derived from hepatocellular carcinoma (HCC) that is characterized by membrane expression of CD133. Here, we describe a novel mechanism by which these cells mediate tumor growth and angiogenesis by systematic comparison of the gene expression profiles between sorted CD133 liver subpopulations through genome-wide microarray analysis. A significantly dysregulated interleukin-8 (IL-8) signaling network was identified in CD133(+) liver TICs obtained from HCC clinical samples and cell lines. IL-8 was found to be overexpressed at both the genomic and proteomic levels in CD133(+) cells isolated from HCC cell lines or clinical samples. Functional studies found enhanced IL-8 secretion in CD133(+) liver TICs to exhibit a greater ability to self-renew, induce tumor angiogenesis, and initiate tumors. In further support of these observations, IL-8 repression in CD133(+) liver TICs by knockdown or neutralizing antibody abolished these effects. Subsequent studies of the IL-8 functional network identified neurotensin (NTS) and CXCL1 to be preferentially expressed in CD133(+) liver TICs. Addition of exogenous NTS resulted in concomitant up-regulation of IL-8 and CXCL1 with simultaneous activation of p-ERK1/2 and RAF-1, both key components of the mitogen-activated protein kinase (MAPK) pathway. Enhanced IL-8 secretion by CD133(+) liver TICs can in turn activate an IL-8-dependent feedback loop that signals through the MAPK pathway. Further, in its role as a liver TIC marker CD133 also plays a functional part in regulating tumorigenesis of liver TICs by way of regulating NTS, IL-8, CXCL1, and MAPK signaling. CONCLUSION: CD133(+) liver TICs promote angiogenesis, tumorigenesis, and self-renewal through NTS-induced activation of the IL-8 signaling cascade.


Subject(s)
Antigens, CD/metabolism , Cell Proliferation , Chemokine CXCL1/physiology , Glycoproteins/metabolism , Interleukin-8/physiology , Liver Neoplasms/pathology , Neoplastic Stem Cells/physiology , Neovascularization, Pathologic/physiopathology , Neurotensin/physiology , Peptides/metabolism , Signal Transduction/physiology , AC133 Antigen , Animals , Antigens, CD/genetics , Carcinoma, Hepatocellular/blood supply , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/surgery , Cell Line, Tumor , Cells, Cultured , Feedback, Physiological , Glycoproteins/deficiency , Glycoproteins/genetics , Hepatectomy , Humans , Interleukin-8/deficiency , Interleukin-8/genetics , Liver/blood supply , Liver/pathology , Liver/surgery , Liver Neoplasms/blood supply , Liver Neoplasms/surgery , Mice , Mice, Nude , Mitogen-Activated Protein Kinase Kinases , Neoplastic Stem Cells/immunology , Neurotensin/pharmacology , Peptides/deficiency , Peptides/genetics , Xenograft Model Antitumor Assays
7.
Cancer Res ; 71(2): 583-92, 2011 Jan 15.
Article in English | MEDLINE | ID: mdl-21224345

ABSTRACT

Expression of microRNA genes is profoundly altered in cancer but their role in the development of androgen-independent prostate cancer has received limited attention as yet. In this study, we report a functional impact in prostate cancer cells for overexpression of the microRNA miR-616, which occurred consistently in cells that were androgen-independent (AI) versus androgen-dependent (AD). miR-616 overexpression was confirmed in malignant prostate tissues as opposed to benign prostate specimens. Stable miR-616 overexpression in LNCaP cells by a lentiviral-based approach stimulated AI prostate cancer cell proliferation in vitro whereas concomitantly reducing androgen-induced cell growth. More importantly, miR-616 overexpressing LNCaP cells overcame castration resistance as shown by an enhanced ability to proliferate in vivo after bilateral orchiectomy. Conversely, antagonizing miR-616 in AI prostate cancer cells yielded opposite effects. Microarray profiling and bioinformatics analysis identified the tissue factor pathway inhibitor TFPI-2 mRNA as a candidate downstream target of miR-616. In support of this candidacy, we documented interactions between miR-616 and the 3'UTR of TFPI-2 and determined TFPI-2 expression to be inversely correlated to miR-616 in a series of prostate cell lines and clinical specimens. Notably, reexpression of TFPI-2 in LNCaP cells with stable miR-616 overexpression rescued the AD phenotype, as shown by a restoration of androgen dependence and cell growth inhibition. Taken together, our findings define a functional involvement for miR-616 and TFPI-2 in the development and maintenance of androgen-independent prostate cancer.


Subject(s)
Glycoproteins/biosynthesis , MicroRNAs/biosynthesis , Prostatic Neoplasms/genetics , Androgens/physiology , Animals , Cell Growth Processes/physiology , Cell Line, Tumor , Humans , Male , Mice , Mice, Nude , MicroRNAs/genetics , Neoplasms, Hormone-Dependent/genetics , Neoplasms, Hormone-Dependent/metabolism , Neoplasms, Hormone-Dependent/pathology , Orchiectomy , Prostatic Hyperplasia/genetics , Prostatic Hyperplasia/metabolism , Prostatic Hyperplasia/pathology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology
8.
Cell Stem Cell ; 7(6): 694-707, 2010 Dec 03.
Article in English | MEDLINE | ID: mdl-21112564

ABSTRACT

A novel paradigm in tumor biology suggests that cancer growth is driven by stem-like cells within a tumor, called tumor-initiating cells (TICs) or cancer stem cells (CSCs). Here we describe the identification and characterization of such cells from hepatocellular carcinoma (HCC) using the marker CD133. CD133 accounts for approximately 1.3%-13.6% of the cells in the bulk tumor of human primary HCC samples. When compared with their CD133⁻ counterparts, CD133(+) cells not only possess the preferential ability to form undifferentiated tumor spheroids in vitro but also express an enhanced level of stem cell-associated genes, have a greater ability to form tumors when implanted orthotopically in immunodeficient mice, and can be serially passaged into secondary animal recipients. Xenografts resemble the original human tumor and maintain a similar percentage of tumorigenic CD133(+) cells. Quantitative PCR analysis of 41 separate HCC tissue specimens with follow-up data found that CD133(+) tumor cells were frequently detected at low quantities in HCC, and their presence was also associated with worse overall survival and higher recurrence rates. Subsequent differential microRNA expression profiling of CD133(+) and CD133⁻ cells from human HCC clinical specimens and cell lines identified an overexpression of miR-130b in CD133(+) TICs. Functional studies on miR-130b lentiviral-transduced CD133⁻ cells demonstrated superior resistance to chemotherapeutic agents, enhanced tumorigenicity in vivo, and a greater potential for self renewal. Conversely, antagonizing miR-130b in CD133(+) TICs yielded an opposing effect. The increased miR-130b paralleled the reduced TP53INP1, a known miR-130b target. Silencing TP53INP1 in CD133⁻ cells enhanced both self renewal and tumorigenicity in vivo. Collectively, miR-130b regulates CD133(+) liver TICs, in part, via silencing TP53INP1.


Subject(s)
Antigens, CD/metabolism , Carcinoma, Hepatocellular/metabolism , Carrier Proteins/metabolism , Glycoproteins/metabolism , Heat-Shock Proteins/metabolism , Liver Neoplasms/metabolism , MicroRNAs/metabolism , Neoplastic Stem Cells/metabolism , Peptides/metabolism , AC133 Antigen , Adult , Aged , Aged, 80 and over , Animals , Antigens, CD/genetics , Carcinoma, Hepatocellular/genetics , Cell Line, Tumor , Cell Proliferation , Female , Glycoproteins/genetics , Humans , Liver/metabolism , Liver/pathology , Liver Neoplasms/genetics , Male , Mice , Mice, SCID , Middle Aged , Nuclear Proteins/metabolism , Peptides/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...