Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Am J Pathol ; 180(4): 1509-21, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22349300

ABSTRACT

Colorectal cancers (CRCs) are classified as having microsatellite instability (MSI) or chromosomal instability (CIN); herein termed microsatellite stable (MSS). MSI colon cancers frequently display a poorly differentiated histology for which the molecular basis is not well understood. Gene expression and immunohistochemical profiling of MSS and MSI CRC cell lines and tumors revealed significant down-regulation of the intestinal-specific cytoskeletal protein villin in MSI colon cancer, with complete absence in 62% and 17% of MSI cell lines and tumors, respectively. Investigation of 577 CRCs linked loss of villin expression to poorly differentiated histology in MSI and MSS tumors. Furthermore, mislocalization of villin from the membrane was prognostic for poorer outcome in MSS patients. Loss of villin expression was not due to coding sequence mutations, epigenetic inactivation, or promoter mutation. Conversely, in transient transfection assays villin promoter activity reflected endogenous villin expression, suggesting transcriptional control. A screen of gut-specific transcription factors revealed a significant correlation between expression of villin and the homeobox transcription factor Cdx-1. Cdx-1 overexpression induced villin promoter activity, Cdx-1 knockdown down-regulated endogenous villin expression, and deletion of a key Cdx-binding site within the villin promoter attenuated promoter activity. Loss of Cdx-1 expression in CRC lines was associated with Cdx-1 promoter methylation. These findings demonstrate that loss of villin expression due to Cdx-1 loss is a feature of poorly differentiated CRCs.


Subject(s)
Biomarkers, Tumor/metabolism , Colorectal Neoplasms/metabolism , Microfilament Proteins/metabolism , Animals , Biomarkers, Tumor/genetics , Cell Differentiation/physiology , Cell Membrane/metabolism , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , DNA Methylation/genetics , DNA, Neoplasm/genetics , Down-Regulation , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/metabolism , Humans , Kaplan-Meier Estimate , Mice , Mice, SCID , Microfilament Proteins/genetics , Microsatellite Instability , Microsatellite Repeats , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neoplasm Transplantation , Oligonucleotide Array Sequence Analysis/methods , Prognosis , Promoter Regions, Genetic/genetics , Real-Time Polymerase Chain Reaction/methods , Tumor Cells, Cultured
2.
Am J Physiol Gastrointest Liver Physiol ; 301(5): G856-64, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21836055

ABSTRACT

Phosphatase and tensin homolog deleted on chromosome 10 (PTEN) is a negative regulator of phosphatidylinositol 3-kinase (PI3K) signaling that is frequently inactivated in colorectal cancer through mutation, loss of heterozygosity, or epigenetic mechanisms. The aim of this study was to determine the effect of intestinal-specific PTEN inactivation on intestinal epithelial homeostasis and tumorigenesis. PTEN was deleted specifically in the intestinal epithelium, by crossing PTEN(Lox/Lox) mice with villin(Cre) mice. PTEN was robustly expressed in the intestinal epithelium and maximally in the differentiated cell compartment. Targeted inactivation of PTEN in the intestinal epithelium of PTEN(Lox/Lox)/villin(Cre) mice was confirmed by genotyping, immunohistochemistry, and qPCR. While intestinal-specific PTEN deletion did not have a major effect on cell fate determination or proliferation in the small intestine, it did increase phosphorylated (p) protein kinase B (AKT) expression in the intestinal epithelium, and 19% of animals developed small intestinal adenomas and adenocarcinomas at 12 mo of age. These tumors demonstrated pAKT and nuclear ß-catenin staining, indicating simultaneous activation of the PI3K/AKT and Wnt signaling pathways. These findings demonstrate that, while PTEN inactivation alone has a minimal effect on intestinal homeostasis, it can facilitate tumor promotion upon deregulation of ß-catenin/TCF signaling, further establishing PTEN as a bona fide tumor suppressor gene in intestinal cancer.


Subject(s)
Adenocarcinoma/metabolism , Adenoma/metabolism , Epithelial Cells/metabolism , Intestinal Neoplasms/metabolism , Intestine, Small/metabolism , PTEN Phosphohydrolase/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Adenoma/genetics , Adenoma/pathology , Animals , Epithelial Cells/pathology , Intestinal Neoplasms/genetics , Intestinal Neoplasms/pathology , Intestine, Small/pathology , Mice , Mice, Knockout , PTEN Phosphohydrolase/genetics , Phosphorylation , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/genetics , Wnt Proteins/genetics , Wnt Proteins/metabolism , beta Catenin/genetics , beta Catenin/metabolism
3.
Cancer Res ; 70(2): 609-20, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-20068171

ABSTRACT

Histone deacetylase inhibitors (HDACi) induce growth arrest and apoptosis in colon cancer cells and are being considered for colon cancer therapy. The underlying mechanism of action of these effects is poorly defined with both transcription-dependent and -independent mechanisms implicated. We screened a panel of 30 colon cancer cell lines for sensitivity to HDACi-induced apoptosis and correlated the differences with gene expression patterns induced by HDACi in the five most sensitive and resistant lines. A robust and reproducible transcriptional response involving coordinate induction of multiple immediate-early (fos, jun, egr1, egr3, atf3, arc, nr4a1) and stress response genes (Ndrg4, Mt1B, Mt1E, Mt1F, Mt1H) was selectively induced in HDACi sensitive cells. Notably, a significant percentage of these genes were basally repressed in colon tumors. Bioinformatics analysis revealed that the promoter regions of the HDACi-induced genes were enriched for KLF4/Sp1/Sp3 transcription factor binding sites. Altering KLF4 levels failed to modulate apoptosis or transcriptional responses to HDACi treatment. In contrast, HDACi preferentially stimulated the activity of Spl/Sp3 and blocking their action attenuated both the transcriptional and apoptotic responses to HDACi treatment. Our findings link HDACi-induced apoptosis to activation of a Spl/Sp3-mediated response that involves derepression of a transcriptional network basally repressed in colon cancer.


Subject(s)
Apoptosis/drug effects , Colonic Neoplasms/drug therapy , Colonic Neoplasms/genetics , Gene Expression Regulation, Neoplastic/drug effects , Genes, Immediate-Early/drug effects , Histone Deacetylase Inhibitors/pharmacology , Sp1 Transcription Factor/genetics , Sp3 Transcription Factor/genetics , Apoptosis/genetics , Apoptosis/physiology , Binding Sites , Butyrates/pharmacology , Caco-2 Cells , Colonic Neoplasms/enzymology , Colonic Neoplasms/pathology , Dactinomycin/pharmacology , HCT116 Cells , HT29 Cells , Humans , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Promoter Regions, Genetic , Sp1 Transcription Factor/metabolism , Sp3 Transcription Factor/metabolism , Transcriptional Activation
4.
J Proteomics ; 71(5): 530-46, 2008 Dec 02.
Article in English | MEDLINE | ID: mdl-18824147

ABSTRACT

Intestinal epithelial cells undergo progressive cell maturation as they migrate along the crypt-villus axis. To determine molecular signatures that define this process, proteins differentially expressed between the crypt and villus were identified by 2D-DIGE and MALDI-MS. Forty-six differentially expressed proteins were identified, several of which were validated by immunohistochemistry. Proteins upregulated in the villus were enriched for those involved in brush border assembly and lipid uptake, established features of differentiated intestinal epithelial cells. Multiple proteins involved in glycolysis were also upregulated in the villus, suggesting increased glycolysis is a feature of intestinal cell differentiation. Conversely, proteins involved in nucleotide metabolism, and protein processing and folding were increased in the crypt, consistent with functions associated with cell proliferation. Three novel paneth cell markers, AGR2, HSPA5 and RRBP1 were also identified. Notably, significant correlation was observed between overall proteomic changes and corresponding gene expression changes along the crypt-villus axis, indicating intestinal cell maturation is primarily regulated at the transcriptional level. This proteomic profiling analysis identified several novel proteins and functional processes differentially induced during intestinal cell maturation in vivo. Integration of proteomic, immunohistochemical, and parallel gene expression datasets demonstrate the coordinated manner in which intestinal cell maturation is regulated.


Subject(s)
Intestinal Mucosa/physiology , Intestine, Small/physiology , Proteomics , Animals , Coloring Agents , Electrophoresis, Gel, Two-Dimensional , Endoplasmic Reticulum Chaperone BiP , Enzymes/chemistry , Enzymes/genetics , Enzymes/isolation & purification , Gene Expression Regulation , Intestinal Mucosa/chemistry , Intestinal Mucosa/cytology , Intestine, Small/chemistry , Intestine, Small/cytology , Lipids/physiology , Mice , Proteins/chemistry , Proteins/genetics , Proteins/isolation & purification , Rats , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Steroids/metabolism
5.
Gastroenterology ; 135(6): 2030-42, 2042.e1-3, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18824170

ABSTRACT

BACKGROUND & AIMS: HuR is a RNA-binding factor whose expression is commonly upregulated in some human tumor types. We explored the molecular mechanism underlying HuR elevation and its role in gastric cancer tumorigenesis. METHODS: HuR expression and subcellular localization were determined by polymerase chain reaction, immunoblot, and immunohistochemical analyses. Its effect on tumor growth was characterized using flow cytometry, terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick-end labeling, and soft agar analyses. Luciferase reporter, chromatin immunoprecipitation, and electrophoretic mobility shift assays were used to measure transcriptional activation by nuclear factor kappaB (NF-kappaB) signaling. RESULTS: Compared with normal gastric tissues, HuR was expressed at higher levels in gastric tumors, particularly in advanced versus early tumors; this increase was associated with enhanced cytoplasmic translocation of HuR. HuR overexpression increased proliferation of tumor cells, activating the G(1) to S transition of the cell cycle, DNA synthesis, and anchorage-independent growth. Small interfering RNA-mediated knockdown of HuR expression reduced tumor cell proliferation and response to apoptotic stimuli. No genetic or epigenetic alterations of HuR were observed in gastric tumor cell lines or primary tumors; overexpression depended on phosphatidylinositol 3-kinase/AKT signaling and NF-kappaB activity. AKT activation increased p65/RelA binding to a putative NF-kappaB binding site in the HuR promoter, the stability of HuR target transcripts, and the cytoplasmic import of HuR. CONCLUSIONS: HuR is a direct transcription target of NF-kappaB; its activation in gastric cancer cell lines depends on phosphatidylinositol 3-kinase/AKT signaling. HuR activation by this pathway has proliferative and antiapoptotic effects on gastric cancer cells.


Subject(s)
Antigens, Surface/genetics , NF-kappa B/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA, Neoplasm/genetics , RNA-Binding Proteins/genetics , Stomach Neoplasms/genetics , Transcription, Genetic , Cell Line, Tumor , Cell Proliferation , ELAV Proteins , ELAV-Like Protein 1 , Gene Expression Regulation, Neoplastic , Humans , Immunoblotting , Immunohistochemistry , Immunoprecipitation , NF-kappa B/metabolism , Phosphatidylinositol 3-Kinases/genetics , Polymerase Chain Reaction , Proto-Oncogene Proteins c-akt/genetics , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology
6.
Mol Biol Cell ; 19(10): 4062-75, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18632985

ABSTRACT

The class II Histone deacetylase (HDAC), HDAC4, is expressed in a tissue-specific manner, and it represses differentiation of specific cell types. We demonstrate here that HDAC4 is expressed in the proliferative zone in small intestine and colon and that its expression is down-regulated during intestinal differentiation in vivo and in vitro. Subcellular localization studies demonstrated HDAC4 expression was predominantly nuclear in proliferating HCT116 cells and relocalized to the cytoplasm after cell cycle arrest. Down-regulating HDAC4 expression by small interfering RNA (siRNA) in HCT116 cells induced growth inhibition and apoptosis in vitro, reduced xenograft tumor growth, and increased p21 transcription. Conversely, overexpression of HDAC4 repressed p21 promoter activity. p21 was likely a direct target of HDAC4, because HDAC4 down-regulation increased p21 mRNA when protein synthesis was inhibited by cycloheximide. The importance of p21 repression in HDAC4-mediated growth promotion was demonstrated by the failure of HDAC4 down-regulation to induce growth arrest in HCT116 p21-null cells. HDAC4 down-regulation failed to induce p21 when Sp1 was functionally inhibited by mithramycin or siRNA-mediated down-regulation. HDAC4 expression overlapped with that of Sp1, and a physical interaction was demonstrated by coimmunoprecipitation. Chromatin immunoprecipitation (ChIP) and sequential ChIP analyses demonstrated Sp1-dependent binding of HDAC4 to the proximal p21 promoter, likely directed through the HDAC4-HDAC3-N-CoR/SMRT corepressor complex. Consistent with increased transcription, HDAC4 or SMRT down-regulation resulted in increased histone H3 acetylation at the proximal p21 promoter locus. These studies identify HDAC4 as a novel regulator of colon cell proliferation through repression of p21.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21/antagonists & inhibitors , Gene Expression Regulation, Neoplastic , Histone Deacetylases/physiology , Repressor Proteins/physiology , Animals , Apoptosis , Cell Line, Tumor , Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Down-Regulation , Humans , Male , Mice , Mice, SCID , Models, Biological , Neoplasm Transplantation , RNA, Small Interfering/metabolism
7.
Mol Nutr Food Res ; 52(11): 1289-99, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18435490

ABSTRACT

To identify candidate genes involved in the development of colorectal cancer, we used cDNA microarrays to analyze gene expression differences between human colorectal tumors and paired adjacent normal mucosa. We identified approximately 3.5-fold significant downregulation of selenium-binding protein 1 (SBP1) in colorectal tumors compared to normal mucosa (p = 0.003). Importantly, stage III colorectal cancer patients with low tumor-SBP1 expression had significantly shorter disease-free and overall survival as compared with those patients with high tumor-SBP1 expression (p = 0.04 and 0.03, respectively). We further characterized the role of SBP1 in colorectal cancer in vivo and in vitro. In normal tissue, SBP1 was maximally expressed in terminally differentiated epithelial cells on the luminal surface of crypts in the large intestine. Consistent with this in vivo localization, SBP1 was upregulated during in vitro colonic cell differentiation along the absorptive (Caco-2) and secretory (HT29 Clones 16E and 19A) cell lineages. Downregulation (approximately 50%) of SBP1 expression by small interfering RNA in colonic cancer cells was associated with reduced expression of another epithelial differentiation marker, carcinoembryonic antigen (CEA), although PCNA and p21(WAF1/cip1 )expression were not altered. These data demonstrate that higher expression of SBP1 is associated with differentiation of the normal colonic epithelia and may be a positive prognostic factor for survival in stage III colorectal carcinoma.


Subject(s)
Colorectal Neoplasms/genetics , Intestines/physiopathology , Selenium-Binding Proteins/genetics , Adult , Aged , Aged, 80 and over , Cell Line, Tumor , Colonic Neoplasms/genetics , Colorectal Neoplasms/pathology , Humans , Intestinal Mucosa/pathology , Intestinal Mucosa/physiopathology , Intestines/pathology , Middle Aged , Neoplasm Staging , Oligonucleotide Array Sequence Analysis , Prognosis , RNA, Small Interfering/genetics , Rectal Neoplasms/genetics , Selenium-Binding Proteins/metabolism , Survivors
8.
Cancer Res ; 68(6): 1953-61, 2008 Mar 15.
Article in English | MEDLINE | ID: mdl-18339877

ABSTRACT

Cetuximab is a monoclonal antibody that targets the human epidermal growth factor receptor (EGFR). Although approved for use in EGFR-overexpressing advanced colorectal cancer, recent studies have shown a lack of association between EGFR overexpression and cetuximab response, requiring the identification of novel biomarkers predictive of response to this agent. To do so, 22 colon cancer cell lines were screened for cetuximab response in vitro and sensitive and resistant lines were identified. In sensitive cell lines, cetuximab induced a G(0)-G(1) arrest without inducing apoptosis. Notably, cetuximab-sensitive but not cetuximab-resistant cell lines were preferentially responsive to EGF-stimulated growth. Whereas neither EGFR protein/mRNA expression nor gene copy number correlated with cetuximab response, examination of the mutation status of signaling components downstream of EGFR showed that cell lines with activating PIK3CA mutations or loss of PTEN expression (PTEN null) were more resistant to cetuximab than PIK3CA wild type (WT)/PTEN-expressing cell lines (14 +/- 5.0% versus 38.5 +/- 6.4% growth inhibition, mean +/- SE; P = 0.008). Consistently, PIK3CA mutant isogenic HCT116 cells showed increased resistance to cetuximab compared with PIK3CA WT controls. Furthermore, cell lines that were PIK3CA mutant/PTEN null and Ras/BRAF mutant were highly resistant to cetuximab compared with those without dual mutations/PTEN loss (10.8 +/- 4.3% versus 38.8 +/- 5.9% growth inhibition, respectively; P = 0.002), indicating that constitutive and simultaneous activation of the Ras and PIK3CA pathways confers maximal resistance to this agent. A priori screening of colon tumors for PTEN expression status and PIK3CA and Ras/BRAF mutation status could help stratify patients likely to benefit from this therapy.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Colonic Neoplasms/drug therapy , Mutation , PTEN Phosphohydrolase/biosynthesis , Phosphatidylinositol 3-Kinases/genetics , Antibodies, Monoclonal, Humanized , Cell Line, Tumor , Cetuximab , Class I Phosphatidylinositol 3-Kinases , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Epidermal Growth Factor/pharmacology , ErbB Receptors/biosynthesis , ErbB Receptors/genetics , ErbB Receptors/metabolism , Erlotinib Hydrochloride , G1 Phase/drug effects , Gene Dosage , Genes, ras , HCT116 Cells , Humans , MAP Kinase Signaling System , Proto-Oncogene Proteins B-raf/genetics , Quinazolines/pharmacology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Resting Phase, Cell Cycle/drug effects , ras Proteins/genetics
9.
Int J Cancer ; 122(7): 1573-84, 2008 Apr 01.
Article in English | MEDLINE | ID: mdl-18059034

ABSTRACT

hSRBC is a putative tumor suppressor located at 11p15.4, at which frequent genomic loss has been observed in several human malignancies. To explore the candidacy of hSRBC as a suppressor of gastric tumorigenesis, we analyzed the expression and mutation status of hSRBC in gastric tissues and cell lines. hSRBC transcript was expressed in all normal and benign tumor tissues examined, but undetectable or very low in 73% (11/15) cancer cell lines and 41% (46/111) primary tumors. Loss or reduction of hSRBC expression was tumor-specific and correlated with stage and grade of tumors. While allelic loss or somatic mutations of the gene were infrequent, its expression was restored in tumor cells by 5-aza-2'-deoxycytidine treatment and aberrant hypermethylation of 23 CpG sites in the promoter region showed a tight association with altered expression. Transient or stable expression of hSRBC led to a G(1) cell cycle arrest and apoptosis of tumor cells, and strongly suppresses colony forming ability and xenograft tumor growth. In addition, hSRBC elevated apoptotic sensitivity of tumor cells to genotoxic agents, such as 5-FU, etoposide and ultraviolet. Interestingly, hSRBC increased the protein stability of p53 and expression of p53 target genes, such as p21(Waf1), PUMA and NOXA, while hSRBC-mediated cell cycle arrest and apoptosis were abolished by blockade of p53 function. Our findings suggest that hSRBC is a novel tumor suppressor whose epigenetic inactivation contributes to the malignant progression of gastric tumors, in part, through attenuated p53 response to stresses.


Subject(s)
Gene Silencing , Intracellular Signaling Peptides and Proteins/genetics , Stomach Neoplasms/genetics , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/genetics , Adenocarcinoma/genetics , Adenoma/genetics , Apoptosis , Blotting, Northern , Cell Line, Tumor , CpG Islands/genetics , DNA Methylation , Down-Regulation , Flow Cytometry , Fluorescent Antibody Technique , Hamartoma/genetics , Humans , Immunoblotting , In Situ Nick-End Labeling , Intracellular Signaling Peptides and Proteins/metabolism , Neoplastic Stem Cells , Polyps/genetics , Promoter Regions, Genetic/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transplantation, Heterologous , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Proteins/metabolism
10.
Gastroenterology ; 132(7): 2459-77, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17570219

ABSTRACT

BACKGROUND & AIMS: X-linked inhibitor of apoptosis protein-associated factor 1 (XAF1) is a candidate tumor suppressor located at the chromosome 17p13 region, but the molecular basis underlying its inactivation in human tumors and growth-inhibiting function has not been well defined. We explored the candidacy of XAF1 as a suppressor in colorectal tumorigenesis. METHODS: XAF1 expression was characterized by polymerase chain reaction-based cloning, isoform-specific polymerase chain reaction, ribonuclease protection, and immunoblot assays. Allelic loss of the gene was evaluated by loss of heterozygosity (LOH) assay, and promoter CG dinucleotide (CpG) site methylation was determined using bisulfite sequencing. The effect of XAF1 on tumor growth was examined using flow cytometry, terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick-end labeling, colony formation, and viability assays. RESULTS: Expression of 5 XAF1 variants including 2 novel transcripts was down-regulated concomitantly in 11 of 20 (55%) cell lines and 26 of 65 (40%) primary tumors. XAF1 reduction was tumor-specific and showed a correlation with advanced stage and high grade of tumor. LOH of the gene was found in 12 of 33 (36%) tumors. Promoter CpG site methylation was observed frequently in both cell lines and tumor tissues including many LOH tumors, suggesting that biallelic inactivation of XAF1 might be common in colorectal cancers. XAF1 expression suppressed tumor cell growth and enhanced cellular response to various apoptotic stimuli, such as 5-fluorouracil, etoposide, H(2)O(2), gamma-irradiation, ultraviolet, and tumor necrosis factor-alpha, whereas knockdown of its expression protected cells from the stresses. CONCLUSIONS: Genetic and epigenetic alteration of XAF1 is a common event in colorectal tumorigenesis and contributes to the malignant tumor progression by providing survival advantages for tumor cells under various stress conditions.


Subject(s)
Apoptosis , Colorectal Neoplasms/physiopathology , Neoplasm Proteins/metabolism , Adaptor Proteins, Signal Transducing , Amino Acid Sequence , Apoptosis Regulatory Proteins , Cell Line, Tumor , Cell Survival , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , CpG Islands/genetics , DNA Methylation , Down-Regulation , Gene Expression , Gene Silencing , Genetic Variation , Humans , Intracellular Signaling Peptides and Proteins , Loss of Heterozygosity , Molecular Sequence Data , Neoplasm Proteins/genetics , Neoplasm Staging , Promoter Regions, Genetic , Stimulation, Chemical , Transcription, Genetic
11.
J Biol Chem ; 281(19): 13548-13558, 2006 May 12.
Article in English | MEDLINE | ID: mdl-16533812

ABSTRACT

Inhibitors of histone deacetylases (HDACs) induce growth arrest, differentiation, and apoptosis of colon cancer cell lines in vitro and have demonstrated anti-cancer efficacy in clinical trials. Whereas a role for HDAC1 and -2 in mediating components of the HDAC inhibitor response has been reported, the role of HDAC3 is unknown. Here we demonstrate increased protein expression of HDAC3 in human colon tumors and in duodenal adenomas from Apc1638(N/+) mice. HDAC3 was also maximally expressed in proliferating crypt cells in normal intestine. Silencing of HDAC3 expression in colon cancer cell lines resulted in growth inhibition, a decrease in cell survival, and increased apoptosis. Similar effects were observed for HDAC2 and, to a lesser extent, for HDAC1. HDAC3 silencing also selectively induced expression of alkaline phosphatase, a marker of colon cell maturation. Concurrent with its effect on cell growth, overexpression of HDAC3 and other Class I HDACs inhibited basal and butyrate-induced p21 transcription in a Sp1/Sp3-dependent manner, whereas silencing of HDAC3 stimulated p21 promoter activity and expression. However, the magnitude of the effects elicited by silencing of individual Class I HDACs was significantly less than that induced by HDAC inhibitors. These findings identify HDAC3 as a gene deregulated in human colon cancer and as a novel regulator of colon cell maturation and p21 expression. These findings also demonstrate that multiple Class I HDACs are involved in repressing p21 and suggest that the growth-inhibitory and apoptotic effects induced by HDAC inhibitors are probably mediated through the inhibition of multiple HDACs.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21/metabolism , Histone Deacetylases/metabolism , Apoptosis , Caco-2 Cells , Cell Differentiation , Cell Proliferation , Colon/cytology , Colonic Neoplasms , Cyclin-Dependent Kinase Inhibitor p21/genetics , Down-Regulation , Gene Expression Regulation, Enzymologic , Gene Silencing , HCT116 Cells , Histone Deacetylase Inhibitors , Humans , Time Factors , Up-Regulation
12.
Cancer Res ; 63(21): 7068-75, 2003 Nov 01.
Article in English | MEDLINE | ID: mdl-14612497

ABSTRACT

X-linked inhibitor of apoptosis (XIAP) is the most potent member of the IAP family that exerts antiapoptotic effects by interfering with the activities of caspases. Recently, XIAP-associated factor 1 (XAF1) and two mitochondrial proteins, Smac/DIABLO and HtrA2, have been identified to negatively regulate the caspase-inhibiting activity of XIAP. To explore the candidacy of XAF1, Smac/DIABLO, and HtrA2 as a tumor suppressor in gastric tumorigenesis, we investigated the expression and mutation status of the genes in 123 gastric tissues and 15 cancer cell lines. Whereas Smac/DIABLO and HtrA2 transcripts were normally expressed in all cancer specimens we examined, XAF1 transcript was not expressed or present at extremely low levels in 40% (6 of 15) of cancer cell lines and in 23% (20 of 87) of primary carcinomas. Abnormal reduction of XAF1 expression showed a strong correlation with stage and grade of tumors, and a tumor-specific down-regulation of XAF1 was observed in 45% (9 of 20) of matched sets. Unlike XAF1, XIAP expression exhibited no detectable alteration in cancers. Whereas loss of heterozygosity within the XAF1 region or somatic mutations of the gene was not detected, expression of XAF1 transcript was reactivated in all nonexpressor cell lines after 5-aza-2-deoxycytidine treatment. The 5' upstream region of the XAF1 gene encompasses no gastric cell-rich region that rigorously satisfies the formal criteria for CpG islands. However, bisulfite DNA sequencing analysis for 34 CpG sites in the promoter region revealed a strong association between hypermethylation and gene silencing. Moreover, transcriptional silencing of XAF1 was tightly associated with hypermethylation of seven CpGs located in the 5' proximal region (nucleotides -23 to -234). Additionally, loss or abnormal reduction of XAF1 expression was found to inversely correlate with p53 mutations, suggesting that epigenetic inactivation of XAF1 and mutational alteration of p53 might be mutually exclusive events in gastric tumorigenesis. Collectively, our study suggests that epigenetic silencing of XAF1 by aberrant promoter methylation may contribute to the malignant progression of human gastric tumors.


Subject(s)
Adenocarcinoma/genetics , Chromosomes, Human, Pair 17/genetics , DNA Methylation , Genes, Tumor Suppressor , Neoplasm Proteins/genetics , Stomach Neoplasms/genetics , Adaptor Proteins, Signal Transducing , Adenocarcinoma/metabolism , Alleles , Apoptosis Regulatory Proteins , Carrier Proteins/biosynthesis , Carrier Proteins/genetics , Cell Line, Tumor , Down-Regulation , Gene Deletion , Gene Expression Regulation, Neoplastic , Gene Silencing , High-Temperature Requirement A Serine Peptidase 2 , Humans , Intracellular Signaling Peptides and Proteins , Mitochondrial Proteins/biosynthesis , Mitochondrial Proteins/genetics , Mutation , Neoplasm Proteins/biosynthesis , Promoter Regions, Genetic , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Serine Endopeptidases/biosynthesis , Serine Endopeptidases/genetics , Stomach Neoplasms/metabolism , Tumor Suppressor Protein p53/genetics
13.
Oncogene ; 22(28): 4314-32, 2003 Jul 10.
Article in English | MEDLINE | ID: mdl-12853969

ABSTRACT

Transforming growth factor (TGF)-beta1 acts as a potent growth inhibitor of prostate epithelial cells, and aberrant function of its receptor type I and II correlates with tumor aggressiveness. However, intracellular and serum TGF-beta1 levels are elevated in prostate cancer patients and further increased in patients with metastatic carcinoma, suggesting the oncogenic switch of TGF-beta1 role in prostate tumorigenesis. Recently, we reported the mitogenic conversion of TGF-beta1 effect by oncogenic Ha-Ras in prostate cancer cells. Here, we show that TGF-beta1 activates interleukin (IL)-6, which has been implicated in the malignant progression of prostate cancers, via multiple signaling pathways including Smad2, nuclear factor-kappaB (NF-kappaB), JNK, and Ras. TGF-beta1-induced IL-6 gene expression was strongly inhibited by DN-Smad2 but not by DN-Smad3 while it was further activated by wild-type Smad2 transfection. IL-6 activation by TGF-beta1 was accompanied by nuclear translocation of NF-kappaB, which was blocked by the p38 inhibitors SB202190 and SB203580 or by IkappaBalphaDeltaN transfection, indicating the crucial role for the p38-NF-kappaB signaling in TGF-beta1 induction of IL-6. TGF-beta1 activated c-Jun phosphorylation, and IL-6 induction by TGF-beta1 was severely impeded by DN-c-Jun and DN-JNK or AP-1 inhibitor curcumin, showing that the JNK-c-Jun-AP-1 signaling plays a pivotal role in TGF-beta1 stimulation of IL-6. It was also found that the Ras-Raf-MEK1 cascade is activated by TGF-beta1 and participates in the TGF-beta1 induction of IL-6 in an AP-1-dependent manner. Cotransfection assays demonstrated that TGF-beta1 stimulation of IL-6 results from the synergistic collaboration of the Smad2, p38-NF-kappaB, JNK-c-Jun-AP-1, or Ras-Raf-MEK1 cascades. In addition, a time course IL-6 decay revealed that mRNA stability of IL-6 is modestly increased by TGF-beta1, indicating that TGF-beta1 also regulates IL-6 at the post-transcriptional level. Intriguingly, IL-6 inactivation restored the sensitivity to TGF-beta1-mediated growth arrest and apoptosis, suggesting that elevated IL-6 in advanced prostate tumors might act as a resistance factor against TGF-beta1. Collectively, our data demonstrate that IL-6 expression is stimulated by tumor-producing TGF-beta1 in human prostate cancer cells through multiple signaling pathways including Smad2, p38, JNK, and Ras, and enhanced expression of IL-6 could contribute to the oncogenic switch of TGF-beta1 role for prostate tumorigenesis, in part by counteracting its growth suppression function.


Subject(s)
DNA-Binding Proteins/physiology , Interleukin-6/genetics , Mitogen-Activated Protein Kinases/physiology , NF-kappa B/physiology , Prostatic Neoplasms/etiology , Trans-Activators/physiology , Transforming Growth Factor beta/pharmacology , Apoptosis , Humans , JNK Mitogen-Activated Protein Kinases , Male , RNA, Messenger/analysis , Smad2 Protein , Transcription Factor AP-1/physiology , Transforming Growth Factor beta1 , Tumor Cells, Cultured , p38 Mitogen-Activated Protein Kinases
14.
Int J Cancer ; 104(3): 318-27, 2003 Apr 10.
Article in English | MEDLINE | ID: mdl-12569555

ABSTRACT

Mutational alterations of PTEN and PIK3CA, which negatively and positively regulate PI3-kinase activity, respectively, have been observed in many types of human cancer. To explore the implication of PTEN and PIK3CA mutations in gastric tumorigenesis, we characterized the expression and mutation status of the genes in 126 gastric tissues and 15 cell lines. Expression of PTEN transcript was abnormally low in 5 of 15 (33%) cell lines and 20 of 55 (36%) primary carcinomas, whereas 0 of 71 noncancerous tissues including 16 benign tumors showed altered expression. Allelotyping analysis using an intragenic polymorphism (IVS4+109) revealed that 14 of 30 (47%) informative cases carried LOH of the gene, which is closely linked to low expression. The LOH rate was significantly higher in advanced tumors [12 of 19 (63%)] compared to early-stage tumors [2 of 11 (18%)] and more frequent in poorly differentiated tumors [9 of 13 (69%)] than well- or moderately differentiated tumors [5 of 17 (29%)]. Interestingly, however, none of the LOH tumors carried mutational disruption of the remaining allele, suggesting haploinsufficiency of PTEN in gastric tumorigenesis. Methylation studies revealed that PTEN pseudogene, but not PTEN, is methylated in cell lines and primary tumors, indicating that PTEN is not a target of epigenetic silencing in gastric cancers and that the pseudogene should be considered more carefully in methylation analysis of the PTEN promoter. Genomic amplification of PIK3CA was found in 9 of 15 (60%) cell lines and 20 of 55 (36.4%) primary tumors but in no noncancerous tissues. Furthermore, PIK3CA amplification was predominantly detected in tumors with no PTEN alterations, suggesting that mutations of PTEN and PIK3CA are mutually exclusive events in gastric tumorigenesis. Amplification of PIK3CA was strongly associated with increased expression of PIK3CA transcript and elevated levels of phospho-AKT. Collectively, our data reveal that 13 of 15 (87%) gastric cell lines and 31 of 55 (56%) primary carcinomas harbored either amplification of PIK3CA or abnormal reduction of PTEN. Mutually exclusive alterations of PTEN and PIK3CA also suggest that mutations of either gene could activate the PI3-kinase/AKT signaling pathway, which is directly linked to the malignant progression of gastric tumor cells.


Subject(s)
Adenocarcinoma/genetics , Adenoma/genetics , Phosphatidylinositol 3-Kinases/genetics , Phosphoric Monoester Hydrolases/genetics , Protein Serine-Threonine Kinases , Stomach Neoplasms/genetics , Tumor Suppressor Proteins/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Adenoma/metabolism , Adenoma/pathology , DNA Methylation , DNA Mutational Analysis , DNA Primers/chemistry , DNA, Neoplasm/analysis , Gene Amplification , Gene Silencing , Hamartoma/genetics , Hamartoma/metabolism , Hamartoma/pathology , Humans , Loss of Heterozygosity , Male , Mutation , Neoplasm Staging , PTEN Phosphohydrolase , Phosphatidylinositol 3-Kinases/metabolism , Phosphoric Monoester Hydrolases/metabolism , Phosphorylation , Polymerase Chain Reaction , Polymorphism, Single-Stranded Conformational , Promoter Regions, Genetic , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Pseudogenes/genetics , RNA, Messenger/metabolism , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Tumor Cells, Cultured , Tumor Suppressor Proteins/metabolism
15.
Int J Cancer ; 98(5): 718-23, 2002 Apr 10.
Article in English | MEDLINE | ID: mdl-11920641

ABSTRACT

p53R2, a recently identified putative tumor suppressor located at 8q23.1, encodes a protein with striking similarity to a small subunit of ribonucleotide reductase. p53R2 is directly induced by wild-type p53 and involved in the p53 checkpoint for repair of damaged DNA, raising the possibility that mutational inactivation of p53R2 may contribute to the development and progression of human malignancies. To explore the p53R2's candidacy for a suppressor in gastric tumorigenesis, we examined the expression and mutation status of p53R2 in 166 gastric specimens including 90 primary adenocarcinomas and 15 cell lines. In response to genotoxic damages, p53R2 transcription was clearly activated in wild-type but not mutant-type p53-carrying cells while basal expression of p53R2 in undamaged cells showed no association with the mutational status of p53. Host cell reactivation assay revealed that p53R2 enhances DNA repair efficiency and plays a role in the p53-mediated repair of damaged DNA, whereas no significant effect of p53R2 on cell growth and apoptosis was detected in flow cytometry and [(3)H]thymidine incorporation assays. p53R2 transcript was expressed in all normal and tumor tissues and its expression levels were not significantly different between normal and malignant carcinoma tissues. p53R2 expression showed no correlation with stage, grade and histological types of tumors. Moreover, no tumor-specific reduction of p53R2 was detected in 30 matched sets. Mutational analysis of p53R2 in 105 carcinomas including 15 cell lines also failed to detect any evidences for genomic deletion or somatic mutations leading to amino acid substitutions or frameshift whereas 31% (28 of 90) of the same primary tumors showed p53 alterations. Whereas 82% (23 of 28) of the mutant p53-carrying primary tumors expressed abnormally low p21(Waf1), no association of p53R2 expression with the p53 status was recognized, suggesting that basal transcription of p53R2 is regulated through the p53-independent mechanism. Collectively, our study indicates that although p53R2 is induced in a p53-dependent manner and involved the p53-mediated DNA repair in gastric epithelial cells, it is not a critical target of genetic inactivation in gastric tumorigenesis.


Subject(s)
Adenocarcinoma/genetics , Adenoma/genetics , DNA Repair , Ribonucleotide Reductases/genetics , Stomach Neoplasms/genetics , Tumor Suppressor Protein p53/genetics , Adenocarcinoma/pathology , Adenoma/pathology , Antineoplastic Agents/pharmacology , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Division , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/genetics , Cyclins/metabolism , DNA Damage , DNA Mutational Analysis , DNA Primers/chemistry , DNA, Neoplasm/analysis , Doxorubicin/pharmacology , Humans , Polymerase Chain Reaction , RNA, Messenger/metabolism , Ribonucleotide Reductases/metabolism , Stomach Neoplasms/pathology , Transfection , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...