Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Methods Mol Biol ; 2534: 197-213, 2022.
Article in English | MEDLINE | ID: mdl-35670977

ABSTRACT

Whole-slide imaging (WSI) has wide spectrum of application in histopathology, especially in the study of cancer including papillary thyroid carcinoma. The main applications of WSI system include research, teaching, and assessment and recently pathology practices. The other major advantages of WSI over histological sections on glass slides are easier storage and sharing of information as well as adaptation of use in artificial intelligence. The applications of WSI depend on factors such as volume of services requiring WSI, physical factors (computer server, bandwidth limitation of networks, storages requirements for data), adaption of the WSI images with the laboratory workflow, personnel (IT expert, pathologist, technicians) adaptation to the WSI workflow, validation studies, ethics, and cost efficiency of the application(s).


Subject(s)
Artificial Intelligence , Thyroid Neoplasms , Humans , Laboratories , Thyroid Cancer, Papillary , Thyroid Neoplasms/diagnostic imaging , Workflow
2.
Genome Biol ; 17: 58, 2016 Mar 30.
Article in English | MEDLINE | ID: mdl-27029486

ABSTRACT

BACKGROUND: The IL23/Th17 pathway is essential for the onset of inflammatory bowel disease (IBD), yet the specific mechanism by which this pathway initiates the disease remains unknown. In this study, we identify the mechanisms that mediate cross-talk between the IL23 pathway and the intestinal barrier in IBD. RESULTS: The downstream targets of the IL23 pathway were identified by RNA array profiling and confirmed by immunohistochemical staining. The role of miRNAs that interact with IL23 was explored in mice with TNBS-induced colitis. Claudin-8 (CLDN8), a multigene family protein that constitutes the backbone of tight junctions, was identified as a novel target of IL23 in IBD. CLDN8 was significantly downregulated in IBD patients with inflamed colonic mucosa, and in trinitrobenzene sulphonic acid (TNBS) induced colitis in mice. Therapeutic treatment of colitis in mice using an IL23 antibody restored CLDN8 abundance, in parallel with recovery from colitis. In addition, we identify miR-223 as a novel mediator of the crosstalk between the IL23 signal pathway and CLDN8 in the development of IBD. MiR-223 was upregulated in IBD, and its activity was regulated through the IL23 pathway. Antagomir inhibition of miR-223 reactivated CLDN8 and improved a number of signs associated with TNBS-induced colitis in mice. CONCLUSIONS: Our study characterizes a new mechanistic pathway in IBD, in which miR-223 interacts with the IL23 pathway by targeting CLDN8. Strategies designed to disrupt this interaction may provide novel therapeutic agents for the management of IBD.


Subject(s)
Claudins/genetics , Gene Expression Profiling/methods , Inflammatory Bowel Diseases/genetics , MicroRNAs/genetics , Oligonucleotide Array Sequence Analysis/methods , Animals , Antagomirs , Caco-2 Cells , Cell Line , Claudins/metabolism , Disease Models, Animal , Gene Expression Regulation , Humans , Inflammatory Bowel Diseases/chemically induced , Inflammatory Bowel Diseases/drug therapy , Inflammatory Bowel Diseases/metabolism , Interleukin-23/administration & dosage , Interleukin-23/genetics , Interleukin-23/pharmacology , Mice , MicroRNAs/metabolism , Oligonucleotides/administration & dosage , Oligonucleotides/therapeutic use , Signal Transduction , Trinitrobenzenesulfonic Acid/adverse effects
3.
J Crohns Colitis ; 10(6): 726-34, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26802082

ABSTRACT

BACKGROUND AND AIMS: Accumulating evidence supports epigenetic modifications in mediating intestinal immunity in inflammatory bowel disease [IBD] pathogenesis. This study aimed to identify key dysregulated epigenetic modulators and the molecular downstream pathways in IBD. METHODS: Expression of 116 well-defined epigenetic modulators was profiled and validated in 96 intestinal tissues from patients with Crohn's disease [CD], ulcerative colitis [UC], and healthy controls using quantitative reverse transcriptase polymerase chain reaction [QRT-PCR], western blot, and immunohistochemistry. Dysregulation of histone modifications and IBD-related cytokines were examined by chromatin immunoprecipitation, luciferase activity, and gene expression analyses in normal colonic epithelial cell line, NCM460, upon small-molecule inhibition or RNA interference, followed by validation in primary colonic tissues. RESULTS: Targeted expression profiling uncovered seven differentially expressed epigenetic modulators, of which the down-regulation of lysine acetyltransferase 2B [KAT2B] mRNA and protein was the most significant and was consequently validated in inflamed CD and UC compared with healthy colonic tissues. KAT2B protein localised abundantly in nuclei of normal colonic epithelium but diminished in paired inflamed CD and UC tissues. Pharmacological inhibition of KAT2B by anacardic acid in NCM460 cells reduced the levels of histone H4 lysine 5 acetylation [H4K5ac] and interleukin-10 [IL-10] in a dose-dependent manner. Knockdown of KAT2B reduced the IL-10 promoter occupancy of KAT2B and H4K5ac, resulting in transcriptional silencing. IL-10 level was also diminished in inflamed IBD tissues. CONCLUSIONS: Our findings demonstrated a novel epigenetic mechanism of IL-10 dysregulation in IBD. Down-regulation of KAT2B may disrupt the innate and adaptive inflammatory responses due to the suppression of this crucial anti-inflammatory cytokine.


Subject(s)
Colon/metabolism , Epigenesis, Genetic , Inflammatory Bowel Diseases/genetics , Interleukin-10/metabolism , p300-CBP Transcription Factors/metabolism , Biomarkers/metabolism , Blotting, Western , Case-Control Studies , Cell Line , Colon/immunology , Down-Regulation , Gene Expression Profiling , Histone Code , Humans , Immunohistochemistry , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/metabolism , Interleukin-10/genetics , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction , p300-CBP Transcription Factors/genetics
4.
Nat Med ; 19(7): 901-908, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23793099

ABSTRACT

Here we show that glioblastoma express high levels of branched-chain amino acid transaminase 1 (BCAT1), the enzyme that initiates the catabolism of branched-chain amino acids (BCAAs). Expression of BCAT1 was exclusive to tumors carrying wild-type isocitrate dehydrogenase 1 (IDH1) and IDH2 genes and was highly correlated with methylation patterns in the BCAT1 promoter region. BCAT1 expression was dependent on the concentration of α-ketoglutarate substrate in glioma cell lines and could be suppressed by ectopic overexpression of mutant IDH1 in immortalized human astrocytes, providing a link between IDH1 function and BCAT1 expression. Suppression of BCAT1 in glioma cell lines blocked the excretion of glutamate and led to reduced proliferation and invasiveness in vitro, as well as significant decreases in tumor growth in a glioblastoma xenograft model. These findings suggest a central role for BCAT1 in glioma pathogenesis, making BCAT1 and BCAA metabolism attractive targets for the development of targeted therapeutic approaches to treat patients with glioblastoma.


Subject(s)
Amino Acids, Branched-Chain/metabolism , Brain Neoplasms/metabolism , Cell Proliferation , Glioma/metabolism , Transaminases/physiology , Animals , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Line, Tumor , Female , Glioma/genetics , Glioma/pathology , HEK293 Cells , Humans , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/physiology , Metabolism/genetics , Mice , Mice, Nude , Models, Biological , Transaminases/genetics , Transaminases/metabolism
5.
Int J Mol Sci ; 14(4): 7492-505, 2013 Apr 08.
Article in English | MEDLINE | ID: mdl-23567267

ABSTRACT

Medulloblastoma (MB) is a malignant pediatric brain tumor arising in the cerebellum consisting of four distinct subgroups: WNT, SHH, Group 3 and Group 4, which exhibit different molecular phenotypes. We studied the expression of Dickkopf (DKK) 1-4 family genes, inhibitors of the Wnt signaling cascade, in MB by screening 355 expression profiles derived from four independent datasets. Upregulation of DKK1, DKK2 and DKK4 mRNA was observed in the WNT subgroup, whereas DKK3 was downregulated in 80% MBs across subgroups with respect to the normal cerebellum (p < 0.001). Since copy number aberrations targeting the DKK3 locus (11p15.3) are rare events, we hypothesized that epigenetic factors could play a role in DKK3 regulation. Accordingly, we studied 77 miRNAs predicting to repress DKK3; however, no significant inverse correlation between miRNA/mRNA expression was observed. Moreover, the low methylation levels in the DKK3 promoters (median: 3%, 5% and 5% for promoter 1, 2 and 3, respectively) excluded the downregulation of gene expression by methylation. On the other hand, the treatment of MB cells with Trichostatin A (TSA), a potent inhibitor of histone deacetylases (HDAC), was able to restore both DKK3 mRNA and protein. In conclusion, DKK3 downregulation across all MB subgroups may be due to epigenetic mechanisms, in particular, through chromatin condensation.


Subject(s)
Chromatin Assembly and Disassembly , Gene Expression Regulation, Neoplastic , Gene Silencing , Intercellular Signaling Peptides and Proteins/biosynthesis , Medulloblastoma/metabolism , Neoplasm Proteins/biosynthesis , Adaptor Proteins, Signal Transducing , Adolescent , Adult , Aged , Cell Line, Tumor , Chemokines , Child , Child, Preschool , Databases, Genetic , Female , Gene Expression Profiling , Humans , Infant , Intercellular Signaling Peptides and Proteins/genetics , Male , Medulloblastoma/genetics , Medulloblastoma/pathology , Methylation , Middle Aged , Neoplasm Proteins/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Neoplasm/biosynthesis
7.
Acta Neuropathol ; 123(4): 539-52, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22402744

ABSTRACT

Medulloblastomas are the most common malignant brain tumors in children. Several large-scale genomic studies have detailed their heterogeneity, defining multiple subtypes with unique molecular profiles and clinical behavior. Increased expression of the miR-183~96~182 cluster of microRNAs has been noted in several subgroups, including the most clinically aggressive subgroup associated with genetic amplification of MYC. To understand the contribution of miR-183~96~182 to the pathogenesis of this aggressive subtype of medulloblastoma, we analyzed global gene expression and proteomic changes that occur upon modulation of miRNAs in this cluster individually and as a group in MYC-amplified medulloblastoma cells. Knockdown of the full miR-183~96~182 cluster results in enrichment of genes associated with apoptosis and dysregulation of the PI3K/AKT/mTOR signaling axis. Conversely, there is a relative enrichment of pathways associated with migration, metastasis and epithelial to mesenchymal transition, as well as pathways associated with dysfunction of DNA repair in cells with preserved miR-183 cluster expression. Immunocytochemistry and FACS analysis confirm induction of apoptosis upon knockdown of the miR-183 cluster. Importantly, cell-based migration and invasion assays verify the positive regulation of cell motility/migration by the miR-183 cluster, which is largely mediated by miR-182. We show that the effects on cell migration induced by the miR-183 cluster are coupled to the PI3K/AKT/mTOR pathway through differential regulation of AKT1 and AKT2 isoforms. Furthermore, we show that rapamycin inhibits cell motility/migration in medulloblastoma cells and phenocopies miR-183 cluster knockdown. Thus, the miR-183 cluster regulates multiple biological programs that converge to support the maintenance and metastatic potential of medulloblastoma.


Subject(s)
Apoptosis/genetics , Cell Movement/genetics , Cell Proliferation , MicroRNAs/genetics , Signal Transduction/genetics , Cell Cycle/genetics , Cell Line, Tumor , Cell Migration Assays , Cerebellar Neoplasms/pathology , Comet Assay , Computational Biology , Gene Expression Regulation, Neoplastic/genetics , Gene Knockdown Techniques , Humans , Medulloblastoma/pathology , Proteomics , Proto-Oncogene Proteins c-myc/genetics , Transfection
8.
Acta Neuropathol ; 123(4): 529-38, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22134538

ABSTRACT

The contribution of microRNAs to the initiation, progression, and metastasis of medulloblastoma (MB) remains poorly understood. Metastatic dissemination at diagnosis is present in about 30% of MB patients, and is associated with a dismal prognosis. Using microRNA expression profiling, we demonstrate that the retinal miR-183-96-182 cluster on chromosome 7q32 is highly overexpressed in non-sonic hedgehog MBs (non-SHH-MBs). Expression of miR-182 and miR-183 is associated with cerebellar midline localization, and miR-182 is significantly overexpressed in metastatic MB as compared to non-metastatic tumors. Overexpression of miR-182 in non-SHH-MB increases and knockdown of miR-182 decreases cell migration in vitro. Xenografts overexpressing miR-182 invaded adjacent normal tissue and spread to the leptomeninges, phenotypically reminiscent of clinically highly aggressive large cell anaplastic MB. Hence, our study provides strong in vitro and in vivo evidence that miR-182 contributes to leptomeningeal metastatic dissemination in non-SHH-MB. We therefore reason that targeted inhibition of miR-182 may prevent leptomeningeal spread in patients with non-SHH-MB.


Subject(s)
Cerebellar Neoplasms/genetics , Cerebellar Neoplasms/pathology , Medulloblastoma/genetics , Medulloblastoma/pathology , Meningeal Neoplasms/secondary , MicroRNAs/genetics , Adolescent , Animals , Cell Migration Assays , Cell Proliferation , Child , Child, Preschool , Cohort Studies , Computational Biology , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Hedgehog Proteins/genetics , Humans , Male , Meningeal Neoplasms/genetics , Mice , Mice, SCID , Microarray Analysis , Transplantation, Heterologous/methods , Tumor Cells, Cultured , Wnt Proteins/genetics , Young Adult
9.
Acta Neuropathol ; 117(4): 457-64, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19057917

ABSTRACT

Embryonal tumors with abundant neuropil and true rosettes (ETANTR) comprise a rare variant of embryonal brain tumors usually occurring in infants. Only 13 cases have been reported in the literature to date and little is known about the molecular pathogenesis of these tumors. Here, we describe a case of ETANTR in a 2-year-old girl presenting with a large tumor in the vermis of the cerebellum. Histological examination showed clusters of small-undifferentiated cells including ependymoblastic-like rosettes admixed with large fibrillar and paucicellular neuropil-like areas indicative for ETANTR. Genomic imbalances were detected by using array-based comparative genomic hybridization. In addition to trisomy of chromosome 2, which has been previously described in ETANTR, array-CGH revealed high-level genomic amplification of 0.89 Mb at chromosome band 19q13.42 covering a microRNA cluster and several protein-coding genes. This aberration has not been described in any other brain tumor to date, indicating a specific aberration in ETANTR. MicroRNAs contained in the microRNA cluster at 19q13.42 including oncomirs miRNA-372 and miRNA-373 were highly up-regulated in the tumor when compared to normal cerebellum or whole brain. In summary, this is the first report on a potentially specific genetic aberration in ETANTR, supporting the hypothesis of a distinct tumor entity.


Subject(s)
Cerebellar Neoplasms/genetics , Chromosomes, Human, Pair 19/genetics , Gene Amplification , MicroRNAs/genetics , Neoplasms, Germ Cell and Embryonal/genetics , Cerebellar Neoplasms/pathology , Child, Preschool , Comparative Genomic Hybridization , Female , Gene Expression Profiling , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Microarray Analysis , Neoplasms, Germ Cell and Embryonal/pathology , Polymerase Chain Reaction , RNA, Messenger/metabolism
10.
Hepatology ; 46(2): 436-45, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17559150

ABSTRACT

UNLABELLED: Metastasis is a major cause of cancer morbidity and mortality in individuals with hepatocellular carcinoma (HCC), yet little is known about the underlying molecular basis. Using genetic information derived from chromosome-based comparative genomic hybridization, we have reported previously on regional chromosome 7q21-q22 gains in close association with HCC progression. In this study, we undertook cDNA microarray-based comparative genomic hybridization, to examine the 7q21-q22 region for the involved gene(s) in HCC. High-resolution mapping analysis highlighted 7 candidates, namely PFTAIRE protein kinase 1 (PFTK1), ODAG, CDK6, CAS1, PEX1, SLC25A, and PEG10, within the region. Quantitative reverse transcription (RT)-PCR evaluation further indicated upregulation of a single candidate gene, PFTK1, that correlated significantly with both advanced metastatic HCCs (P = 0.032) and tumor microvascular invasion (P = 0.012). Given that little is known about the function(s) of PFTK1, which is a novel cell division cycle (Cdc)2-related gene, we examined its potential role in the motile phenotype of HCC cells by both ectopic expression and knockdown investigations. RNA-interference knockdown of PFTK1 in invasive Hep3B cells resulted in a significant reduction in cell invasion, chemotactic migration, and cell motility (P < 0.001). Conversely, ectopic expression of PFTK1 in noninvasive HKCI-C3 cells induced substantial cellular invasion and migration (P < or = 0.007). In neither cell line was there any effect on cell viability. Immunofluorescence showed marked filamentous actin polymerizations in PFTK1-expressing cells. CONCLUSION: In this study, we have thus provided preliminary evidence that overexpression of PFTK1 may confer a motile phenotype in malignant hepatocytes that accounts for the association of upregulation of this gene in metastatic HCC.


Subject(s)
CDC2 Protein Kinase/genetics , Carcinoma, Hepatocellular/genetics , Liver Neoplasms/genetics , Adult , Aged , Aged, 80 and over , CDC2 Protein Kinase/analysis , Cyclin-Dependent Kinases , Female , Gene Dosage , Humans , Male , Middle Aged , Nucleic Acid Hybridization , Oligonucleotide Array Sequence Analysis , Phenotype , RNA, Messenger/analysis
11.
Cancer Lett ; 243(2): 246-54, 2006 Nov 18.
Article in English | MEDLINE | ID: mdl-16412567

ABSTRACT

Peroxisome proliferator-activated receptor gamma (PPARgamma) ligands have been shown to inhibit angiogenesis. We showed that treatment with 15d-PGJ(2), a PPARgamma ligand, downregulate the expressions of angiopoietin-1 (Ang-1) in gastric cancer cells MKN45. The medium of MKN45 cells treated with 15d-PGJ(2) significantly inhibited the migration and tube formation of human umbilical vein endothelial cells (HUVECs). Moreover, Matrigel plug assay revealed that 15d-PGJ(2) reduced in vivo angiogenesis induced by MKN45 cells. These modulations were restored by the addition of recombinant Ang-1. Our findings supported that 15d-PGJ(2) suppressed angiogenesis of gastric cancer cells by downregulation of Ang-1.


Subject(s)
Angiopoietin-1/genetics , Neovascularization, Pathologic/prevention & control , Prostaglandin D2/analogs & derivatives , Stomach Neoplasms/blood supply , Angiogenesis Inhibitors/pharmacology , Animals , Cell Line , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Down-Regulation/drug effects , Down-Regulation/genetics , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/physiology , Female , Gene Expression Profiling , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Physiologic/drug effects , Oligonucleotide Array Sequence Analysis , Prostaglandin D2/pharmacology , Stomach Neoplasms/pathology , Vascular Endothelial Growth Factor A/metabolism
12.
Am J Gastroenterol ; 100(10): 2274-9, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16181380

ABSTRACT

OBJECTIVES: While promoter hypermethylation is a common molecular alteration of human colorectal cancer that could be detected in the bloodstream, we tested the feasibility of quantitative detection of aberrant DNA methylation in multiple genes in the serum samples of colorectal cancer patients. METHODS: The pre-therapeutic serum samples of 49 colorectal cancer patients and 41 age-matched controls with normal colonoscopy were examined. The presence of methylated DNA in APC (adenomatous polyposis coli), hMLH1 (human MutL homolog 1), and HLTF (helicase-like transcription factor) was detected by quantitative methylation-specific PCR (MethyLight). RESULTS: There was a significant difference in the concentration of methylated serum DNA between cancer patients and controls for HLTF (p= 0.015) and hMLH1 (p= 0.0001) genes, but not for APC gene (p= 0.21). In total, 28 patients with colorectal cancer and 4 controls had methylated DNA detected in at least one marker, which gave a sensitivity of 57% and specificity of 90%. All patients with methylation in two methylation markers had advanced (stage III/IV) cancer (p= 0.006) and patients with methylation in at least one marker tended to have a lower probability of survival (p= 0.08). CONCLUSION: The quantitative detection of aberrant DNA methylation in serum may be a promising high-throughput approach for the noninvasive screening and monitoring of colorectal cancer.


Subject(s)
Colorectal Neoplasms/blood , DNA Methylation , DNA-Binding Proteins/blood , Genes, APC/physiology , Neoplasm Proteins/blood , Nuclear Proteins/blood , Promoter Regions, Genetic/physiology , Transcription Factors/blood , Adaptor Proteins, Signal Transducing , Aged , Aged, 80 and over , Carrier Proteins , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , DNA-Binding Proteins/genetics , Feasibility Studies , Female , Glutathione S-Transferase pi , Glutathione Transferase/blood , Glutathione Transferase/genetics , Humans , Isoenzymes/blood , Isoenzymes/genetics , Male , Middle Aged , MutL Protein Homolog 1 , Neoplasm Proteins/genetics , Nuclear Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sensitivity and Specificity , Transcription Factors/genetics
13.
World J Gastroenterol ; 11(1): 41-5, 2005 Jan 07.
Article in English | MEDLINE | ID: mdl-15609394

ABSTRACT

AIM: Mechanisms underlying the chemopreventive effects of cyclooxygenase (COX) inhibitors remain elusive. We have previously shown that celecoxib but not indomethacin could prevent carcinogen-induced gastric cancer development in Wistar rats. This chemopreventive effect appeared to be independent of COX-2 and prostaglandin (PG) E2 suppression since the lowest PGE2 was obtained in indomethacin group. This study compared the cell kinetic changes in stomachs of rats after treatment with celecoxib (5, 10, 20 mg/(kg.d)) or indomethacin (3 mg/(kg.d)) to gain more insights into the chemopreventive mechanism. METHODS: The apoptosis and proliferation indexes in gastric tumor, adjacent non-cancer tissues and normal gastric tissues were determined. Apoptosis was quantified by apoptotic nuclei counting and TUNEL, whereas proliferation was determined by Ki67 immunostaining. RESULTS: Treatment with either celecoxib or indomethacin inhibited gastric tumor proliferation by more than 65% (P<0.02). However, celecoxib caused a dose-dependent increase in apoptosis (P<0.05) which was not seen in indomethacin-treated tumors (P = 0.54). The highest apoptosis to proliferation ratio was seen in tumors treated with celecoxib at 10 mg/(kg.d). Treatment with this dose of celecoxib was associated with the lowest incidence of gastric cancer development. CONCLUSION: Our findings suggest that the difference in chemopreventive effects of indomethacin and celecoxib in this animal model of gastric carcinogenesis is largely due to the differential cell kinetic changes, which does not correlate with the degree of COX-2 and PG suppression.


Subject(s)
Adenocarcinoma/prevention & control , Cyclooxygenase Inhibitors/pharmacology , Indomethacin/pharmacology , Pyrazoles/pharmacology , Stomach Neoplasms/prevention & control , Sulfonamides/pharmacology , Adenocarcinoma/pathology , Animals , Apoptosis/drug effects , Celecoxib , Cell Division/drug effects , Cyclooxygenase 2 , Cyclooxygenase 2 Inhibitors , Epithelial Cells/drug effects , Epithelial Cells/enzymology , Epithelial Cells/pathology , Isoenzymes/metabolism , Male , Prostaglandin-Endoperoxide Synthases/metabolism , Rats , Rats, Wistar , Stomach Neoplasms/pathology
15.
Int J Cancer ; 112(5): 846-53, 2004 Dec 10.
Article in English | MEDLINE | ID: mdl-15386372

ABSTRACT

Gene promoter hypermethylation is increasingly recognized to play an important role in cancer development through silencing of gene transcription. This study determined the methylation profiles of primary colorectal cancers and adenomas to elucidate the role of epigenetic changes in different stages of colorectal carcinogenesis. We examined the methylation profiles of 47 sporadic colorectal cancers, 36 colonic adenomas from patients without cancer and 34 colonic biopsies from patients without colonic lesions. Paired adjacent dysplasia tissues obtained from 17 cancer patients were also examined. Promoter hypermethylation in 10 tumor-related genes (APC, ATM, GSTP1, HLTF, MGMT, hMLH1, p14, p15, SOCS-1 and TIMP-3) were studied by methylation-specific PCR. Promoter hypermethylation was frequently detected in more than 40% of colonic cancers and adenomas in APC, ATM, HLTF, MGMT and hMLH1 genes (p < 0.0001 vs. normal). While low level of methylation was detected in p14, p15 and TIMP-3, there was no methylation detected in GSTP1 and SOCS-1. The frequencies of methylation were comparable between tumors and adenomas, and advanced and nonadvanced adenoma. In contrast, K-ras mutation was only detected in advanced adenomas and cancers. Concurrent methylation in >/= 3 genes was found in 66.7% adenomas and 68.1% cancers but not in normal colonic tissues. Methylation was associated with reduced protein expressions in colorectal adenomas and cancers. Moreover, methylation in ATM was more common in older cancer patients (p = 0.002), but there was no significant association between promoter hypermethylation and other clinicopathologic characteristics of cancer. Our study demonstrated the early and specific involvement of promoter hypermethylation in the colorectal adenoma-carcinoma sequence.


Subject(s)
Adenoma/genetics , Adenoma/pathology , Carcinoma/genetics , Carcinoma/pathology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , DNA Methylation , Gene Silencing , Biopsy , Case-Control Studies , Cell Transformation, Neoplastic , Disease Progression , Humans , Promoter Regions, Genetic , Transcription, Genetic
16.
Mol Carcinog ; 37(2): 91-7, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12766908

ABSTRACT

Helicase-like transcription factor (HLTF), a member of the SWI/SNF (mating type switching/sucrose nonfermenting) chromatin-remodeling complex, is recently found to be inactivated by promoter hypermethylation in human colorectal cancer. However, the role of this putative tumor suppressor gene in other tumors has not been determined. We evaluated the role of HLTF promoter hypermethylation in gastric cancer. Expression of HLTF was examined by reverse-transcription (RT)-polymerase chain reaction (PCR), and promoter hypermethylation in HLTF was determined by methylation-specific PCR. Bisulfite DNA sequencing was performed to determine the detailed methylation profiles of the promoter region. HLTF expression was lost in two of five gastric cell lines and in 13 (28%) of 46 primary gastric cancers. Accordingly, promoter hypermethylation was detected in the two cell lines and in nine of 13 gastric cancer samples. Of the ten normal gastric specimens and ten paired adjacent nonneoplastic tissues, methylation was detected in only one adjacent nonneoplastic tissue. Bisulfite DNA sequencing of the promoter region of HLTF showed that the CpG island was densely methylated in cell lines and cancer samples; this also appeared to correlate with expression level. Treatment of gastric cell lines that lacked HLTF expression with the demethylating agent 5-azacytidine (5-azaDC) restored HLTF expression. These results suggest that HLTF promoter hypermethylation is frequently demonstrated in human gastric cancer, and inactivation of HLTF or the chromatin-remodeling complex may play a crucial role in gastric carcinogenesis.


Subject(s)
DNA Methylation , DNA-Binding Proteins/genetics , Promoter Regions, Genetic/physiology , Stomach Neoplasms/genetics , Transcription Factors/genetics , Adaptor Proteins, Signal Transducing , Antimetabolites, Antineoplastic/pharmacology , Azacitidine/pharmacology , Carrier Proteins , CpG Islands , DNA Methylation/drug effects , DNA-Binding Proteins/metabolism , Gastric Mucosa/cytology , Gastric Mucosa/metabolism , Gene Expression Regulation, Neoplastic , Gene Silencing , Humans , MutL Protein Homolog 1 , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Nuclear Proteins , Promoter Regions, Genetic/drug effects , Sequence Analysis, DNA/methods , Stomach Neoplasms/metabolism , Sulfites/chemistry , Transcription Factors/metabolism , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...