Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Clin Cancer Res ; 20(7): 1856-64, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24486589

ABSTRACT

PURPOSE: Non-small cell lung cancer (NSCLC) is the leading cause of cancer mortality in the world. Novel diagnostic biomarkers may augment both existing NSCLC screening methods as well as molecular diagnostic tests of surgical specimens to more accurately stratify and stage candidates for adjuvant chemotherapy. Hypermethylation of CpG islands is a common and important alteration in the transition from normal tissue to cancer. EXPERIMENTAL DESIGN: Following previously validated methods for the discovery of cancer-specific hypermethylation changes, we treated eight NSCLC cell lines with the hypomethylating agent deoxyazacitidine or trichostatin A. We validated the findings using a large publicly available database and two independent cohorts of primary samples. RESULTS: We identified >300 candidate genes. Using The Cancer Genome Atlas (TCGA) and extensive filtering to refine our candidate genes for the greatest ability to distinguish tumor from normal, we define a three-gene panel, CDO1, HOXA9, and TAC1, which we subsequently validate in two independent cohorts of primary NSCLC samples. This three-gene panel is 100% specific, showing no methylation in 75 TCGA normal and seven primary normal samples and is 83% to 99% sensitive for NSCLC depending on the cohort. CONCLUSION: This degree of sensitivity and specificity may be of high value to diagnose the earliest stages of NSCLC. Addition of this three-gene panel to other previously validated methylation biomarkers holds great promise in both early diagnosis and molecular staging of NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/diagnosis , Cysteine Dioxygenase/genetics , Homeodomain Proteins/genetics , Tachykinins/genetics , Aged , Biomarkers, Tumor/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , CpG Islands/genetics , DNA Methylation/genetics , Female , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Neoplasm Staging , Proportional Hazards Models
2.
Epigenetics ; 7(7): 701-9, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22647880

ABSTRACT

Breast cancer (BC) is a disease with diverse tumor heterogeneity, which challenges conventional approaches to develop biomarkers for early detection and prognosis. To identify effective biomarkers, we performed a genome-wide screen for functional methylation changes in BC, i.e., genes silenced by promoter hypermethylation, using a functionally proven gene expression approach. A subset of candidate hypermethylated genes were validated in primary BCs and tested as markers for detection and prognosis prediction of BC. We identified 33 cancer specific methylated genes and, among these, two categories of genes: (1) highly frequent methylated genes that detect early stages of BC. Within that category, we have identified the combination of NDRG2 and HOXD1 as the most sensitive (94%) and specific (90%) gene combination for detection of BC; (2) genes that show stage dependent methylation frequency pattern, which are candidates to help delineate BC prognostic signatures. For this category, we found that methylation of CDO1, CKM, CRIP1, KL and TAC1 correlated with clinical prognostic variables and was a significant prognosticator for poor overall survival in BC patients. CKM [Hazard ratio (HR) = 2.68] and TAC1 (HR = 7.73) were the strongest single markers and the combination of both (TAC1 and CKM) was associated with poor overall survival independent of age and stage in our training (HR = 1.92) and validation cohort (HR = 2.87). Our study demonstrates an efficient method to utilize functional methylation changes in BC for the development of effective biomarkers for detection and prognosis prediction of BC.


Subject(s)
Biomarkers, Tumor/genetics , Breast Neoplasms/diagnosis , DNA Methylation , Homeodomain Proteins/genetics , Tumor Suppressor Proteins/genetics , Adult , Aged , Breast Neoplasms/mortality , Early Detection of Cancer/methods , Female , Gene Silencing , Genetic Loci , Homeodomain Proteins/metabolism , Humans , Middle Aged , Prognosis , Sensitivity and Specificity , Tumor Suppressor Proteins/metabolism
3.
Clin Cancer Res ; 17(6): 1535-45, 2011 Mar 15.
Article in English | MEDLINE | ID: mdl-21278247

ABSTRACT

PURPOSE: The importance of genetic and epigenetic alterations maybe in their aggregate role in altering core pathways in tumorigenesis. EXPERIMENTAL DESIGN: Merging genome-wide genomic and epigenomic alterations, we identify key genes and pathways altered in colorectal cancers (CRC). DNA methylation analysis was tested for predicting survival in CRC patients using Cox proportional hazard model. RESULTS: We identified 29 low frequency-mutated genes that are also inactivated by epigenetic mechanisms in CRC. Pathway analysis showed the extracellular matrix (ECM) remodeling pathway is silenced in CRC. Six ECM pathway genes were tested for their prognostic potential in large CRC cohorts (n = 777). DNA methylation of IGFBP3 and EVL predicted for poor survival (IGFBP3: HR = 2.58, 95% CI: 1.37-4.87, P = 0.004; EVL: HR = 2.48, 95% CI: 1.07-5.74, P = 0.034) and simultaneous methylation of multiple genes predicted significantly worse survival (HR = 8.61, 95% CI: 2.16-34.36, P < 0.001 for methylation of IGFBP3, EVL, CD109, and FLNC). DNA methylation of IGFBP3 and EVL was validated as a prognostic marker in an independent contemporary-matched cohort (IGFBP3 HR = 2.06, 95% CI: 1.04-4.09, P = 0.038; EVL HR = 2.23, 95% CI: 1.00-5.0, P = 0.05) and EVL DNA methylation remained significant in a secondary historical validation cohort (HR = 1.41, 95% CI: 1.05-1.89, P = 0.022). Moreover, DNA methylation of selected ECM genes helps to stratify the high-risk stage 2 colon cancers patients who would benefit from adjuvant chemotherapy (HR: 5.85, 95% CI: 2.03-16.83, P = 0.001 for simultaneous methylation of IGFBP3, EVL, and CD109). CONCLUSIONS: CRC that have silenced genes in ECM pathway components show worse survival suggesting that our finding provides novel prognostic biomarkers for CRC and reflects the high importance of integrative analyses linking genetic and epigenetic abnormalities with pathway disruption in cancer.


Subject(s)
Colonic Neoplasms/diagnosis , Colonic Neoplasms/genetics , Epigenomics , Genomics , Aged , Biomarkers, Tumor , Cell Line, Tumor , Chemotherapy, Adjuvant/methods , DNA Methylation , Epigenesis, Genetic , Female , Humans , Male , Middle Aged , Prognosis , Proportional Hazards Models , Treatment Outcome
4.
Int J Cancer ; 129(8): 1889-98, 2011 Oct 15.
Article in English | MEDLINE | ID: mdl-21154739

ABSTRACT

The diagnosis of sessile serrated adenomas (SSAs) is challenging, and there is a great deal of interobserver variability amongst pathologists in differentiating SSAs from hyperplastic polyps (HPPs). The aim of this study was (i) to assess the utility of epigenetic changes such as DNA methylation in differentiating SSAs from HPPs and (ii) to identify common methylation based molecular markers potentially useful for early detection of premalignant neoplastic lesions of gastrointestinal tract. A total of 97 primary patient adenoma samples were obtained from The Johns Hopkins Hospital pathology archive with IRB approval and HIPAA compliance. We analyzed the promoter associated CpG island methylation status of 17 genes using nested multiplex methylation specific PCR (MSP). Methylation of CDX2, hMLH1 and TLR2 was detected in SSAs and SSAs with dysplasia but not in HPPs. A subset of genes including EVL, GATAs (4 and 5), HIN-1, SFRPs (1, 2, 4 and 5), SOX17 and SYNE1 were methylated frequently in all premalignant gastrointestinal adenomas including tubular adenomas, villous adenomas, SSAs and SSAs with dysplasia but infrequently in non-premalignant polyps such as HPPs. Methylation of CDX2, hMLH1 and TLR2 may be of diagnostic utility in differentiating, histologically challenging cases of SSAs from HPPs. Genes such as EVL, GATAs, HIN-1, SFRPs, SOX17 and SYNE1, which are frequently methylated in all types of tested premalignant adenomas, may be useful as biomarkers in stool-based strategies for early detection of these adenomas and CRCs in future.


Subject(s)
Adenoma/genetics , DNA Methylation , Gastrointestinal Neoplasms/diagnosis , Gastrointestinal Neoplasms/genetics , Precancerous Conditions/diagnosis , Precancerous Conditions/genetics , Adaptor Proteins, Signal Transducing/genetics , Adenoma/diagnosis , Aged , Aged, 80 and over , CDX2 Transcription Factor , Colonic Polyps/diagnosis , Colonic Polyps/genetics , CpG Islands , Diagnosis, Differential , Female , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/genetics , Humans , Hyperplasia , Male , Middle Aged , MutL Protein Homolog 1 , Nuclear Proteins/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins p21(ras) , Toll-Like Receptor 2/genetics , ras Proteins/genetics
5.
Cancer Res ; 69(11): 4691-9, 2009 Jun 01.
Article in English | MEDLINE | ID: mdl-19435926

ABSTRACT

We have used a gene expression array-based strategy to identify the methylation of tissue factor pathway inhibitor 2 (TFPI2), a potential tumor suppressor gene, as a frequent event in human colorectal cancers (CRC). TFPI2 belongs to the recently described group of embryonic cell Polycomb group (PcG)-marked genes that may be predisposed to aberrant DNA methylation in early stages of colorectal carcinogenesis. Aberrant methylation of TFPI2 was detected in almost all CRC adenomas (97%, n = 56) and stages I to IV CRCs (99%, n = 115). We further explored the potential of TFPI2 as a biomarker for the early detection of CRC using stool DNA-based assays in patients with nonmetastatic CRC and average-risk noncancer controls who were candidates for screening. TFPI2 methylation was detected in stool DNA from stage I to III CRC patients with a sensitivity of 76% to 89% and a specificity of 79% to 93%. Detection of TFPI2 methylation in stool DNA may act as a useful adjunct to the noninvasive strategies for screening of CRCs in the future.


Subject(s)
Biomarkers, Tumor/genetics , Carcinoma/diagnosis , Colorectal Neoplasms/diagnosis , DNA Methylation , Feces/chemistry , Glycoproteins/genetics , Aged , Aged, 80 and over , Algorithms , Biomarkers, Tumor/analysis , Caco-2 Cells , Carcinoma/genetics , Carcinoma/pathology , Case-Control Studies , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , DNA Methylation/physiology , DNA Mutational Analysis/methods , Early Detection of Cancer , Female , Glycoproteins/analysis , HCT116 Cells , HT29 Cells , Humans , Male , Middle Aged , Pilot Projects , Tumor Cells, Cultured
6.
Cancer Res ; 68(19): 8094-103, 2008 Oct 01.
Article in English | MEDLINE | ID: mdl-18829568

ABSTRACT

Much recent effort has focused on identifying and characterizing cellular markers that distinguish tumor propagating cells (TPC) from more differentiated progeny. We report here an unusual promoter DNA methylation pattern for one such marker, the cell surface antigen CD133 (Prominin 1). This protein has been extensively used to enrich putative cancer propagating stem-like cell populations in epithelial tumors and, especially, glioblastomas. We find that, within individual cell lines of cultured colon cancers and glioblastomas, the promoter CpG island of CD133 is DNA methylated, primarily, in cells with absent or low expression of the marker protein, whereas lack of such methylation is evident in purely CD133+ cells. Differential histone modification marks of active versus repressed genes accompany these DNA methylation changes. This heterogeneous CpG island DNA methylation status in the tumors is unusual in that other DNA hypermethylated genes tested in such cultures preserve their methylation patterns between separated CD133+ and CD133- cell populations. Furthermore, the CD133 DNA methylation seems to constitute an abnormal promoter signature because it is not found in normal brain and colon but only in cultured and primary tumors. Thus, the DNA methylation is imposed on the transition between the active versus repressed transcription state for CD133 only in tumors. Our findings provide additional insight for the dynamics of aberrant DNA methylation associated with aberrant gene silencing in human tumors.


Subject(s)
Antigens, CD/genetics , Brain Neoplasms/genetics , Carcinoma/genetics , Colorectal Neoplasms/genetics , DNA Methylation , Glioblastoma/genetics , Glycoproteins/genetics , Peptides/genetics , AC133 Antigen , Animals , Antigens, CD/metabolism , Antineoplastic Agents/therapeutic use , Azacitidine/analogs & derivatives , Azacitidine/therapeutic use , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Caco-2 Cells , Carcinoma/drug therapy , Carcinoma/metabolism , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , DNA (Cytosine-5-)-Methyltransferases/antagonists & inhibitors , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methylation/drug effects , Decitabine , Female , Gene Deletion , Gene Expression Regulation, Neoplastic , Glioblastoma/drug therapy , Glioblastoma/metabolism , Glycoproteins/metabolism , HCT116 Cells , HT29 Cells , Humans , Mice , Mice, Nude , Peptides/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
7.
J Gastrointest Surg ; 12(10): 1745-53, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18716850

ABSTRACT

INTRODUCTION: WNT signaling pathway dysregulation is an important event in the pathogenesis of colorectal cancer (CRC) with APC mutations seen in more than 80% of sporadic CRC. However, such mutations in the WNT signaling pathway genes are rare in inflammatory bowel disease (IBD) associated neoplasia (dysplasia and cancer). This study examined the role of epigenetic silencing of WNT signaling pathway genes in the pathogenesis of IBD-associated neoplasia. METHODS: Paraffin-embedded tissue samples were obtained and methylation of ten WNT signaling pathway genes, including APC1A, APC2, SFRP1, SFRP2, SFRP4, SFRP5, DKK1, DKK3, WIF1 and LKB1, was analyzed. Methylation analysis was performed on 41 IBD samples, 27 normal colon samples (NCs), and 24 sporadic CRC samples. RESULTS: Methylation of WNT signaling pathway genes is a frequent and early event in IBD and IBD-associated neoplasia. A progressive increase in the percentage of methylated genes in the WNT signaling pathway from NCs (4.2%) to IBD colitis (39.7%) to IBD-associated neoplasia (63.4%) was seen (NCs vs. IBD colitis, p < 0.01; IBD colitis vs. IBD-associated neoplasia, p = 0.01). In the univariate logistic regression model, methylation of APC2 (OR 4.7, 95% CI: 1.1-20.63, p = 0.04), SFRP1 (OR 5.1, 95% CI: 1.1-31.9, p = 0.04), and SFRP2 (OR 5.1, 95% CI: 1.1-32.3, p = 0.04) was associated with progression from IBD colitis to IBD-associated neoplasia, while APC1A methylation was borderline significant (OR 4.1, 95% CI: 0.95-17.5, p = 0.06). In the multivariate logistic regression model, methylation of APC1A and APC2 was more likely to be associated with IBD-associated neoplasia than IBD colitis. (OR APC1A: 6.4, 95% CI: 1.1-37.7 p = 0.04; OR APC2 9.1, 95% CI: 1.3-61.7, p = 0.02). SUMMARY: Methylation of the WNT signaling genes is an early event seen in patients with IBD colitis and there is a progressive increase in methylation of the WNT signaling genes during development of IBD-associated neoplasia. Moreover, methylation of APC1A, APC2, SFRP1, and SFRP2 appears to mark progression from IBD colitis to IBD-associated neoplasia, and these genes may serve as biomarkers for IBD-associated neoplasia.


Subject(s)
Colorectal Neoplasms/genetics , Inflammatory Bowel Diseases/genetics , Wnt Proteins/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , DNA Methylation , Disease Progression , Epigenesis, Genetic , Humans , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/pathology , Middle Aged , Signal Transduction , Wnt Proteins/metabolism
8.
PLoS Med ; 5(5): e114, 2008 May 27.
Article in English | MEDLINE | ID: mdl-18507500

ABSTRACT

BACKGROUND: The identification and characterization of tumor suppressor genes has enhanced our understanding of the biology of cancer and enabled the development of new diagnostic and therapeutic modalities. Whereas in past decades, a handful of tumor suppressors have been slowly identified using techniques such as linkage analysis, large-scale sequencing of the cancer genome has enabled the rapid identification of a large number of genes that are mutated in cancer. However, determining which of these many genes play key roles in cancer development has proven challenging. Specifically, recent sequencing of human breast and colon cancers has revealed a large number of somatic gene mutations, but virtually all are heterozygous, occur at low frequency, and are tumor-type specific. We hypothesize that key tumor suppressor genes in cancer may be subject to mutation or hypermethylation. METHODS AND FINDINGS: Here, we show that combined genetic and epigenetic analysis of these genes reveals many with a higher putative tumor suppressor status than would otherwise be appreciated. At least 36 of the 189 genes newly recognized to be mutated are targets of promoter CpG island hypermethylation, often in both colon and breast cancer cell lines. Analyses of primary tumors show that 18 of these genes are hypermethylated strictly in primary cancers and often with an incidence that is much higher than for the mutations and which is not restricted to a single tumor-type. In the identical breast cancer cell lines in which the mutations were identified, hypermethylation is usually, but not always, mutually exclusive from genetic changes for a given tumor, and there is a high incidence of concomitant loss of expression. Sixteen out of 18 (89%) of these genes map to loci deleted in human cancers. Lastly, and most importantly, the reduced expression of a subset of these genes strongly correlates with poor clinical outcome. CONCLUSIONS: Using an unbiased genome-wide approach, our analysis has enabled the discovery of a number of clinically significant genes targeted by multiple modes of inactivation in breast and colon cancer. Importantly, we demonstrate that a subset of these genes predict strongly for poor clinical outcome. Our data define a set of genes that are targeted by both genetic and epigenetic events, predict for clinical prognosis, and are likely fundamentally important for cancer initiation or progression.


Subject(s)
Breast Neoplasms/diagnosis , Breast Neoplasms/genetics , Colonic Neoplasms/diagnosis , Colonic Neoplasms/genetics , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Mutation , Cell Line, Tumor , CpG Islands , DNA Methylation , Heterozygote , Homozygote , Humans , Models, Genetic , Prognosis , Sequence Analysis, DNA , Treatment Outcome
9.
Cancer Res ; 68(8): 2764-72, 2008 Apr 15.
Article in English | MEDLINE | ID: mdl-18413743

ABSTRACT

SRY-box containing gene 17 (Sox17) is a member of the high mobility group (HMG) transcription factor superfamily, which plays critical roles in the regulation of development and stem/precursor cell function, at least partly through repression of Wnt pathway activity. Modulators controlling aberrant Wnt signaling activation are frequently disrupted in human cancers through complementary effects of epigenetic and genetic changes. Our recent global analysis of CpG island hypermethylation and gene expression in colorectal cancer (CRC) cell lines revealed that SOX17 gene silencing is associated with DNA hypermethylation of a CpG island in the promoter region. Here, we report that CpG island methylation-dependent silencing of SOX17 occurs in 100% of CRC cell lines, 86% of colorectal adenomas, 100% of stage I and II CRC, 89% of stage III CRC, 89% of primary esophageal cancer, and 50% of non-small cell lung cancer. Overexpression of SOX17 in HCT116 CRC cells inhibits colony growth and beta-catenin/T-cell factor-dependent transcription. Structure-based deletion analysis further shows the presence of a Wnt signaling repression domain in the SOX17 HMG box. Together, our studies suggest that SOX17 is a negative modulator of canonical Wnt signaling, and that SOX17 silencing due to promoter hypermethylation is an early event during tumorigenesis and may contribute to aberrant activation of Wnt signaling in CRC.


Subject(s)
Colorectal Neoplasms/genetics , DNA-Binding Proteins/genetics , Gene Silencing , Genes, sry , High Mobility Group Proteins/genetics , Transcription Factors/genetics , Cell Line , Cell Line, Tumor , Colony-Forming Units Assay , DNA Primers , Genes, Reporter , Genetic Vectors , Humans , Kidney/embryology , Polymerase Chain Reaction , RNA, Neoplasm/genetics , RNA, Neoplasm/isolation & purification , SOXF Transcription Factors
10.
N Engl J Med ; 358(11): 1118-28, 2008 Mar 13.
Article in English | MEDLINE | ID: mdl-18337602

ABSTRACT

BACKGROUND: Despite optimal and early surgical treatment of non-small-cell lung cancer (NSCLC), many patients die of recurrent NSCLC. We investigated the association between gene methylation and recurrence of the tumor. METHODS: Fifty-one patients with stage I NSCLC who underwent curative resection but who had a recurrence within 40 months after resection (case patients) were matched on the basis of age, NSCLC stage, sex, and date of surgery to 116 patients with stage I NSCLC who underwent curative resection but who did not have a recurrence within 40 months after resection (controls). We investigated whether the methylation of seven genes in tumor and lymph nodes was associated with tumor recurrence. RESULTS: In a multivariate model, promoter methylation of the cyclin-dependent kinase inhibitor 2A gene p16, the H-cadherin gene CDH13, the Ras association domain family 1 gene RASSF1A, and the adenomatous polyposis coli gene APC in tumors and in histologically tumor-negative lymph nodes was associated with tumor recurrence, independently of NSCLC stage, age, sex, race, smoking history, and histologic characteristics of the tumor. Methylation of the promoter regions of p16 and CDH13 in both tumor and mediastinal lymph nodes was associated with an odds ratio of recurrent cancer of 15.50 in the original cohort and an odds ratio of 25.25 when the original cohort was combined with an independent validation cohort of 20 patients with stage I NSCLC. CONCLUSIONS: Methylation of the promoter region of the four genes in patients with stage I NSCLC treated with curative intent by means of surgery is associated with early recurrence.


Subject(s)
Cadherins/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Genes, APC , Genes, p16 , Lung Neoplasms/genetics , Neoplasm Recurrence, Local/genetics , Tumor Suppressor Proteins/genetics , Aged , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/surgery , Case-Control Studies , DNA Methylation , Female , Follow-Up Studies , Genes, Tumor Suppressor , Genetic Markers , Humans , Kaplan-Meier Estimate , Logistic Models , Lung Neoplasms/pathology , Lung Neoplasms/surgery , Male , Middle Aged , Neoplasm Staging , Odds Ratio , Polymerase Chain Reaction , Promoter Regions, Genetic
11.
Cancer Biol Ther ; 6(11): 1710-6, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17986858

ABSTRACT

The hypermethylation of tumor-suppressor gene promoter regions has been shown to result in the epigenetic inactivation of many genes. ASC/TMS1 is a pro-apoptotic gene that has been shown to be methylated in many different human neoplasms. The methylation status of ASC/TMS1 was analyzed in a series of colorectal cancer (CRC) cell lines, adenomas and primary colorectal cancers and normal colorectal tissue samples using methylation-specific PCR (MSP). The gene expression of ASC/TMS1 in the CRC cell lines was analyzed using reverse-transcriptase PCR (RT-PCR). Methylation analysis showed complete methylation of ASC/TMS1 in 5 of 7 (71%) CRC cell lines. RT-PCR showed absence of mRNA expression in these same cell lines, and expression was restored after treatment with the demethylating drug 5-aza-2'-deoxyazacytidine. The two unmethylated cell lines showed ASC/TMS1 mRNA expression both before and after treatment with 5-aza-2'-deoxyazacytidine. Methylation was seen in 20 of 115 (17%) of primary colorectal cancer specimens, but no methylation was seen in 30 colorectal adenomas and 11 normal colorectal tissue samples. Methylation status of ASC/TMS1 was correlated with a series of clinicopathological variables using multivariate analysis. Methylation of ASC/TMS1 was more common in right-sided tumors (p = 0.02), concordant with hMLH1 methylation (p = 0.03) and is a late stage event, occurring in 0 of 18 tubular adenomas, 0 of 12 villous adenomas, 2 of 44 (5%) Stage 1 cancers, 8 of 31 (26%) Stage 2 cancers, 8 of 21 (38%) Stage 3 cancers and 2 of 19 (11%) Stage 4 cancers. The ASC/TMS1 gene is frequently silenced in CRC due to promoter hypermethylation. Methylation of ASC/TMS1 appears to be a late-stage event in colorectal carcinogenesis associated with invasive carcinomas but not with normal colorectal tissue or colorectal adenomas. Methylation of ASC/TMS1 may have implications for cancer prognosis.


Subject(s)
Colorectal Neoplasms/genetics , Cytoskeletal Proteins/genetics , DNA Methylation , Adult , Aged , CARD Signaling Adaptor Proteins , Cell Line, Tumor , Colorectal Neoplasms/pathology , Female , Gene Silencing , Humans , Male , Middle Aged , Neoplasm Staging , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction , Tumor Necrosis Factor-alpha/pharmacology
12.
PLoS Genet ; 3(9): 1709-23, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17892325

ABSTRACT

We have developed a transcriptome-wide approach to identify genes affected by promoter CpG island DNA hypermethylation and transcriptional silencing in colorectal cancer. By screening cell lines and validating tumor-specific hypermethylation in a panel of primary human colorectal cancer samples, we estimate that nearly 5% or more of all known genes may be promoter methylated in an individual tumor. When directly compared to gene mutations, we find larger numbers of genes hypermethylated in individual tumors, and a higher frequency of hypermethylation within individual genes harboring either genetic or epigenetic changes. Thus, to enumerate the full spectrum of alterations in the human cancer genome, and to facilitate the most efficacious grouping of tumors to identify cancer biomarkers and tailor therapeutic approaches, both genetic and epigenetic screens should be undertaken.


Subject(s)
Colorectal Neoplasms/genetics , DNA Methylation , Mutation , Base Sequence , CpG Islands , DNA Primers , Humans , Promoter Regions, Genetic , Reverse Transcriptase Polymerase Chain Reaction
13.
Am J Pathol ; 162(5): 1487-94, 2003 May.
Article in English | MEDLINE | ID: mdl-12707031

ABSTRACT

Chimerism on the parenchymal level has been shown for several human allografts, including liver, heart, and kidney, with the integrated recipient-derived cells most likely originating from multipotent bone marrow precursors. We investigated whether chimerism also occurs within epithelial structures of the lung. For this purpose archival tissue biopsies from seven explanted human lung allografts were obtained. We performed laser microdissection of the target structures with subsequent short tandem repeat analysis to detect chimerism within the isolated cells. We found integration of recipient-derived cells in the bronchial epithelium, in type II pneumocytes and in seromucous glands lying adjacent to larger bronchi in all lung allografts studied. Quantitative analysis revealed that the epithelial structures displaying signs of chronic injury, such as squamous metaplasia, showed a markedly higher degree of chimerism (24% versus 9.5%). We therefore conclude that in human lungs, epithelial chimerism occurs at least within bronchi, type II pneumocytes, and seromucous peribronchial glands. Although a bone marrow origin of immigrating host-derived stem cells has been suggested by previous studies in rodents, analysis of lung biopsies from bone marrow-transplanted patients (n = 3) could not prove such delineation in this study. The observation of an enhanced integration of recipient cells into chronically damaged epithelial structures suggests that extrapulmonary precursor cells are able to contribute to pulmonary regeneration.


Subject(s)
Bronchi/pathology , Chromosomes, Human, Y , Lung Transplantation/pathology , Pulmonary Alveoli/pathology , Respiratory Mucosa/pathology , Transplantation Chimera , Adolescent , Adult , Bone Marrow Transplantation/immunology , Child, Preschool , Humans , Immunosuppression Therapy/methods , Male , Middle Aged
14.
Lab Invest ; 82(10): 1419-26, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12379776

ABSTRACT

Intratumoral heterogeneity mirrors subclonal diversity and might affect treatment response. To investigate molecular heterogeneity of primary breast cancer specimens, we determined the amplification status of growth regulatory genes (c-erbB2, topoisomerase IIalpha, c-myc, and cyclinD1) in macroscopically and microscopically separate areas of individual tumors (n = 21). Using laser-assisted microdissection and quantitative PCR, we found marked intratumoral heterogeneity with different patterns for each gene. Molecular heterogeneity in amplification pattern could be demonstrated between both macroscopically (0.5 to several centimeters) and microscopically (10 to several hundred micrometers) distant tumor areas. C-erbB2 amplification proved to be the most stable amplification in individual tumors, with heterogeneity occurring in only 36% of amplified cases. By contrast, amplification of c-myc and cyclinD1 revealed varying patterns in the vast majority of amplified cases (100% and 83%). The constancy of c-erbB2 amplification underlines its presumed importance in breast cancer biology. We conclude that the molecular heterogeneity of breast cancer as evidenced in this study requires thorough and representative sampling of different tumor areas when the biologic significance of somatic mutations is considered. Patterns of heterogeneity can be used to trace the clonal evolution within different compartments of an individual tumor.


Subject(s)
Breast Neoplasms/genetics , Cyclin D1/genetics , Genes, myc , Receptor, ErbB-2/genetics , Breast Neoplasms/pathology , Female , Gene Amplification , Humans , Polymerase Chain Reaction
15.
Am J Pathol ; 160(2): 605-12, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11839581

ABSTRACT

The aberrant methylation of cytosine residues in the promoter region of growth regulatory genes is now widely recognized as an additional mechanism for gene inactivation in cancer cells. In this study we analyzed the methylation status of four growth regulatory genes (p16, RASSF1A, cyclinD2, 14-3-3zeta) during breast cancer progression. For this purpose invasive and noninvasive tumor cell populations as well as hyperplastic cell proliferations were isolated from a series of archival breast tissue specimens (n = 57) using laser-assisted microdissection. A new real-time polymerase chain reaction-based assay was used for the sensitive and quantitative determination of the cell-specific methylation status. We found that aberrant promoter methylation was already prevalent in pure intraductal carcinoma with different frequencies and different methylation levels for the four genes analyzed. For RASSF1A and 14-3-3zeta promoter methylation was also demonstrated in epithelial hyperplasia and intraductal papillomas. By contrast, aberrant methylation of cyclinD2 and p16 was restricted to cancerous epithelium. Increased methylation of the cyclinD2 gene was significantly associated with a higher van Nuys grade. Furthermore, when intraductal and invasive tumor cells were compared, significant quantitative changes in the methylation level were detected primarily within the cyclinD2 gene. These results demonstrate that promoter methylation is an early and frequent event in breast cancer development, but displays great quantitative and gene-specific differences, and changes in a gene-specific manner during tumor progression.


Subject(s)
Biomarkers, Tumor , Breast Neoplasms/pathology , Carcinoma, Intraductal, Noninfiltrating/pathology , DNA Methylation , Exonucleases , Promoter Regions, Genetic/genetics , Tumor Suppressor Proteins , 14-3-3 Proteins , Breast/metabolism , Breast/pathology , Breast Neoplasms/genetics , Carcinoma, Intraductal, Noninfiltrating/genetics , Cyclin D2 , Cyclins/genetics , Cyclins/metabolism , Disease Progression , Exoribonucleases , Female , Genes, Tumor Suppressor , Genes, p16 , Humans , Hyperplasia/physiopathology , Lymphocytes/metabolism , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Proteins/genetics , Proteins/metabolism , Reproducibility of Results
16.
Hepatology ; 35(1): 110-6, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11786966

ABSTRACT

It has recently been shown that epithelial cells derived from stem cells originating outside the liver are integrated into liver allografts. Whether epithelial intragraft chimerism protects transplants from rejection or chronic transplant dysfunction, and whether it interferes with recurrence of primary liver disease, is not known. Twenty-seven sequential biopsies derived from 9 liver-transplant recipients were studied for chimerism of hepatocytes and cholangiocytes. The target cells were isolated by laser microdissection after cytokeratin immunolabeling and genotyped using DNA analysis of a highly polymorphic short tandem repeat. Irrespective of whether early (up to 4 weeks) or late (more than 12 months) posttransplantation biopsies were studied, cholangiocyte chimerism was almost constantly found in 91% of the samples. No significant differences occurred between samples derived from patients with chronic organ dysfunction (n = 3), recurrent hepatitis (n = 3), or mild, unspecific changes (n = 3). By contrast, hepatocyte chimerism tended to occur later (55% vs. 22%) and appeared to be associated with recurrent hepatitis (67% vs. 27%). In this respect, chronic organ dysfunction did not differ from mild, unspecific changes. While cholangiocyte chimerism represents a constant and early phenomenon in liver transplantations, an enhanced chimeric integration of recipient-derived hepatocytes can be observed in recurrent hepatitis, supporting the concept of an increased recruitment of extrahepatic progenitor cells to the liver in chronic hepatitis.


Subject(s)
Liver Transplantation , Tandem Repeat Sequences , Transplantation Chimera , Bile Ducts/chemistry , Bile Ducts/pathology , Biopsy , DNA/analysis , Epithelial Cells/chemistry , Genotype , Graft Rejection , Hepatocytes/chemistry , Humans , Immunohistochemistry , Keratins/analysis , Liver/chemistry , Liver/pathology , Polymerase Chain Reaction , Polymorphism, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...