801
|
Takahashi T, Shivapurkar N, Reddy J, Shigematsu H, Miyajima K, Suzuki M, Toyooka S, Zöchbauer-Müller S, Drach J, Parikh G, Zheng Y, Feng Z, Kroft SH, Timmons C, McKenna RW, Gazdar AF. DNA Methylation Profiles of Lymphoid and Hematopoietic Malignancies. Clin Cancer Res 2004; 10:2928-35. [PMID: 15131027 DOI: 10.1158/1078-0432.ccr-03-0716] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Aberrant methylation of the 5' gene promoter regions is an epigenetic phenomenon that is the major mechanism for silencing of tumor suppressor genes in many cancer types. The aims of our study were (a) to compare the methylation profiles of the major forms of hematological malignancies and (b) to determine the methylation profile of monoclonal gammopathy of undetermined significance (MGUS) and compare it with that of multiple myeloma (MM). EXPERIMENTAL DESIGN We compared the aberrant promoter methylation profile of 14 known or suspected tumor suppressor genes in leukemias (n = 48), lymphomas (n = 42), and MMs (n = 40). We also examined the methylation profile of MGUS (n = 20), a premalignant plasma cell dyscrasia. The genes studied represent five of the six "hallmarks of cancer." RESULTS Peripheral blood lymphocytes (n = 14) from healthy volunteers were negative for methylation of all genes, and methylation percentages in 41 nonmalignant tissues (peripheral blood mononuclear cells, bone marrows, and lymph nodes) from hematological patients were low (0-9%) for all 14 genes, confirming that methylation was tumor specific. Ten of the genes were methylated at frequencies of 29-68% in one or more tumor types, and the methylation indices (an indicator of overall methylation) varied from 0.25 to 0.34. With two exceptions, the methylation patterns of leukemias and lymphomas were similar. However, the pattern of MMs varied from the other tumor types for six genes. In general, the methylation pattern of MGUS was similar to that of MM, although the methylation frequencies were lower (the methylation index of MGUS was 0.15, and that of MM was 0.3). However, the methylation frequencies of six genes were significantly higher in MGUS than in control tissues. The relatively high frequencies of methylation in MGUS are consistent with it being a premalignant condition. CONCLUSIONS The three major forms of lymphoid/hematopoietic malignancies show overlapping but individual patterns of methylation.
Collapse
Affiliation(s)
- Takao Takahashi
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
802
|
Noble M, Dietrich J. The complex identity of brain tumors: emerging concerns regarding origin, diversity and plasticity. Trends Neurosci 2004; 27:148-54. [PMID: 15036880 DOI: 10.1016/j.tins.2003.12.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Elucidation of genetic and epigenetic mechanisms underlying neoplasia is one of the great success stories of modern science, but this success has not been associated with parallel improvements in the treatment of malignant tumors. One possible explanation for this failure is that the most important variables that support growth of malignancies are not yet identified. Another possible explanation, however, is that multiple variables important in neoplastic progression combine to create a level of disease complexity not taken into account by current therapeutic approaches. The study of development and neoplasia in the CNS provides some of the strongest support for the latter view--a view that, if correct, would suggest that a radical rethinking of the biology of malignancy is required if we are to make progress in the treatment of this important medical condition.
Collapse
Affiliation(s)
- Mark Noble
- Department of Biomedical Genetics, University of Rochester Medical Center, 601 Elmwood Avenue, Box 633, Rochester, NY 14642, USA.
| | | |
Collapse
|
803
|
Sathyanarayana UG, Maruyama R, Padar A, Suzuki M, Bondaruk J, Sagalowsky A, Minna JD, Frenkel EP, Grossman HB, Czerniak B, Gazdar AF. Molecular detection of noninvasive and invasive bladder tumor tissues and exfoliated cells by aberrant promoter methylation of laminin-5 encoding genes. Cancer Res 2004; 64:1425-30. [PMID: 14973053 DOI: 10.1158/0008-5472.can-03-0701] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Laminin-5 (LN5) anchors epithelial cells to the underlying basement membrane, and it is encoded by three distinct genes: LAMA3, LAMB3, and LAMC2. To metastasize and grow, cancer cells must invade and destroy the basement membrane. Our previous work has shown that epigenetic inactivation is a major mechanism of silencing LN5 genes in lung cancers. We extended our methylation studies to resected bladder tumors (n = 128) and exfoliated cell samples (bladder washes and voided urine; n = 71) and correlated the data with clinicopathologic findings. Nonmalignant urothelium had uniform expression of LN5 genes and lacked methylation. The methylation frequencies for LN5 genes in tumors were 21-45%, and there was excellent concordance between methylation in tumors and corresponding exfoliated cells. Methylation of LAMA3 and LAMB3 and the methylation index were correlated significantly with several parameters of poor prognosis (tumor grade, growth pattern, muscle invasion, tumor stage, and ploidy pattern), whereas methylation of LAMC2 and methylation index were associated with shortened patient survival. Of particular interest, methylation frequencies of LAMA3 helped to distinguish invasive (72%) from noninvasive (12%) tumors. These results suggest that methylation of LN5 genes has potential clinical applications in bladder cancers.
Collapse
Affiliation(s)
- Ubaradka G Sathyanarayana
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, 6000 Harry Hines Boulevard, Dallas, TX 75390-8593, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
804
|
Garnis C, Buys TPH, Lam WL. Genetic alteration and gene expression modulation during cancer progression. Mol Cancer 2004; 3:9. [PMID: 15035667 PMCID: PMC408463 DOI: 10.1186/1476-4598-3-9] [Citation(s) in RCA: 90] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2004] [Accepted: 03/22/2004] [Indexed: 02/06/2023] Open
Abstract
Cancer progresses through a series of histopathological stages. Progression is thought to be driven by the accumulation of genetic alterations and consequently gene expression pattern changes. The identification of genes and pathways involved will not only enhance our understanding of the biology of this process, it will also provide new targets for early diagnosis and facilitate treatment design. Genomic approaches have proven to be effective in detecting chromosomal alterations and identifying genes disrupted in cancer. Gene expression profiling has led to the subclassification of tumors. In this article, we will describe the current technologies used in cancer gene discovery, the model systems used to validate the significance of the genes and pathways, and some of the genes and pathways implicated in the progression of preneoplastic and early stage cancer.
Collapse
Affiliation(s)
- Cathie Garnis
- Cancer Genetics and Developmental Biology, British Columbia Cancer Research Centre; 601 West 10th Ave, Vancouver, BC, Canada V5Z 1L3
| | - Timon PH Buys
- Cancer Genetics and Developmental Biology, British Columbia Cancer Research Centre; 601 West 10th Ave, Vancouver, BC, Canada V5Z 1L3
| | - Wan L Lam
- Cancer Genetics and Developmental Biology, British Columbia Cancer Research Centre; 601 West 10th Ave, Vancouver, BC, Canada V5Z 1L3
| |
Collapse
|
805
|
Sato N, Fukushima N, Matsubayashi H, Goggins M. Identification of maspin and S100P as novel hypomethylation targets in pancreatic cancer using global gene expression profiling. Oncogene 2004; 23:1531-8. [PMID: 14716296 DOI: 10.1038/sj.onc.1207269] [Citation(s) in RCA: 109] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
DNA hypomethylation is one of the major epigenetic alterations in human cancers. We have previously shown that genes identified as hypomethylated in pancreatic cancer are expressed in pancreatic cancer cell lines, but not in normal pancreatic ductal epithelium and can be reexpressed in nonexpressing cells using 'epigenetic modifying agents' such as DNA methyltransferase inhibitors. To identify additional targets for aberrant hypomethylation in pancreatic cancer, we used oligonucleotide microarrays to screen for genes that displayed expression patterns associated with hypomethylation. This analysis identified a substantial number of candidates including previously reported hypomethylated genes. A subset of eight genes were selected for further methylation analysis, and two cancer-related genes, maspin and S100P, were found to be aberrantly hypomethylated in a large fraction of pancreatic cancer cell lines and primary pancreatic carcinomas. Combined treatment with 5-aza-2'-deoxycytidie and trichostatin A resulted in synergistic induction of maspin and S100P mRNA in MiaPaCa2 cells where both genes were methylated. Furthermore, there was an inverse correlation between methylation and mRNA expression level for maspin and S100P in a large panel of pancreatic cancer cell lines. We also found a significant difference in the methylation patterns of maspin and two previously identified hypomethylated genes (trefoil factor 2 and lipocalin 2) between pancreatic and breast cancer cell lines, suggesting cancer-type specificity for some hypomethylation patterns. Thus, our present results confirm that DNA hypomethylation is a frequent epigenetic event in pancreatic cancer, and suggest that gene expression profiling may help to identify potential targets affected by this epigenetic alteration.
Collapse
Affiliation(s)
- Norihiro Sato
- 1Department of Pathology, The Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | | | | | | |
Collapse
|
806
|
Ebinger M, Senf L, Wachowski O, Scheurlen W. Promoter methylation pattern of caspase-8, P16INK4A, MGMT, TIMP-3, and E-cadherin in medulloblastoma. Pathol Oncol Res 2004; 10:17-21. [PMID: 15029256 DOI: 10.1007/bf02893403] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2004] [Accepted: 03/01/2004] [Indexed: 12/31/2022]
Abstract
Methylation of promoter regions of CpG-rich sites is an important mechanism for silencing of tumor suppressor genes (TSG). To evaluate the role of tumor suppressor genes caspase-8 (CASP8), TIMP-3, E-cadherin (CDH1), p16INK4A, and MGMT in medulloblastoma tumorigenesis, 51 medulloblastomas (46 primary tumor specimens, 5 cell lines) were screened for methylation of promoter linked CpG-islands. For CASP8, we examined the 5' UTR region that has been shown to be associated with expression of CASP8. As detected by methylation specific PCR, methylation rate was low for TIMP-3 (3% of tumor samples; 1/5 cell lines), for MGMT (0% of tumor samples; 1/5 cell lines), for p16INK4A (2% of tumor samples; 2/5 cell lines) and for CDH1 (8% of tumor samples; 1/4 cell lines). CASP8, however, was methylated in 90% of tumor samples and 4/5 cell lines examined. Screening other tumor entities for CASP8 methylation, we found a similarly high level in 6 neuroblastoma cell lines in contrast to 5 osteosarcoma-, 4 Ewing's sarcoma- and 6 non-embryonic tumor cell lines without any increased promoter methylation. From our results we conclude that methylation of the CASP8 5' UTR region may play a role in inactivation of CASP8 in neural crest tumors.
Collapse
Affiliation(s)
- Martin Ebinger
- Department of Molecular Pathology, Institute of Pathology, Tübingen, Germany
| | | | | | | |
Collapse
|
807
|
Neumann S, Schuchardt K, Reske A, Reske A, Emmrich P, Paschke R. Lack of correlation for sodium iodide symporter mRNA and protein expression and analysis of sodium iodide symporter promoter methylation in benign cold thyroid nodules. Thyroid 2004; 14:99-111. [PMID: 15068624 DOI: 10.1089/105072504322880337] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Cold thyroid nodules (CTNs) are characterized by a reduced iodide uptake in comparison to normal thyroid tissue. The sodium iodide symporter (NIS) is the first step in thyroid hormone synthesis and mediates the active iodide transport in the thyroid cells suggesting that decreased iodide uptake could be a result of changes in NIS expression or molecular defects in the NIS gene. In contrast to previous studies, an intraindividual comparison of NIS mRNA expression in CTNs and their corresponding surrounding tissue was performed using direct detection of NIS mRNA. A significant reduction in NIS mRNA expression was detected in 86% of the 14 investigated CTNs. We hypothesized that human sodium iodide symporter (hNIS) transcriptional failure could be caused by primary molecular NIS gene defects and/or methylation of DNA in the NIS promoter. However, no mutation in the NIS cDNA nor in the NIS promoter region upstream up to-443 bp from the ATG start codon was detected. Therefore, primary molecular NIS gene defects were excluded. However, in 50% of CTNs with reduced NIS mRNA expression, the promoter region was hypermethylated. NIS mRNA expression in these hypermethylated CTNs only reached a maximum of 30% of the corresponding surrounding tissue. Hence, methylation of CpG islands in the NIS promotor could be a regulatory mechanism of NIS transcription in CTNs. Immunoblot revealed absent hNIS protein expression in the total cell membrane fraction in 45% of investigated nodules. In the majority of the remaining CTNs NIS protein expression was decreased in the nodule tissue compared to the corresponding surrounding tissue. For investigating protein expression immunhistochemistry has two advantages. First, the whole nodule area can be investigated, and second, NIS expression can be detected in areas where an immunoblot of a cell membrane fraction is negative. Interestingly, immunhistochemistry revealed higher NIS expression in 50% of CTNs compared to their corresponding surrounding tissues and NIS staining was predominantly intracellular. These data demonstrate that NIS protein expression does not reflect NIS mRNA expression. Therefore, factors that affect targeting of NIS to the plasma membrane are likely to be affected.
Collapse
Affiliation(s)
- Susanne Neumann
- III. Medical Department, University of Leipzig, Leipzig, Germany
| | | | | | | | | | | |
Collapse
|
808
|
Cottrell SE, Distler J, Goodman NS, Mooney SH, Kluth A, Olek A, Schwope I, Tetzner R, Ziebarth H, Berlin K. A real-time PCR assay for DNA-methylation using methylation-specific blockers. Nucleic Acids Res 2004; 32:e10. [PMID: 14722226 PMCID: PMC373310 DOI: 10.1093/nar/gnh008] [Citation(s) in RCA: 109] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
DNA methylation-based biomarkers have been discovered that could potentially be used for the diagnosis of cancer by detection of circulating, tumor-derived DNA in bodily fluids. Any methylation detection assay that would be applied to these samples must be capable of detecting small amounts of tumor DNA in the presence of background normal DNA. We have developed a real-time PCR assay, called HeavyMethyl, that is well suited for this application. HeavyMethyl uses methylation-specific oligonucleotide blockers and a methylation-specific probe to achieve methylation-specific amplification and detection. We tested the assays on unmethylated and artificially methylated DNA in order to determine the limit of detection. After careful optimization, our glutathione-S-transferase pi1 and Calcitonin assays can amplify as little as 30 and 60 pg of methylated DNA, respectively, and neither assay amplifies unmethylated DNA. The Calcitonin assay showed a highly significant methylation difference between normal colon and colon adenocarcinomas, and methylation was also detected in serum DNA from colon cancer patients. These assays show that HeavyMethyl technology can be successfully employed for the analysis of very low concentrations of methylated DNA, e.g. in serum of patients with tumors.
Collapse
Affiliation(s)
- Susan E Cottrell
- Epigenomics Inc, 1000 Seneca Street, Suite 300 Seattle, WA 98101, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
809
|
Liu TC, Lin SF, Chang JG, Yang MY, Hung SY, Chang CS. Epigenetic alteration of the SOCS1 gene in chronic myeloid leukaemia. Br J Haematol 2004; 123:654-61. [PMID: 14616969 DOI: 10.1046/j.1365-2141.2003.04660.x] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The expression of the suppressor of cytokine signalling-1 (SOCS1) protein is induced in response to stimulation by several cytokines. The induced SOCS1 inhibits the signalling pathway through the association with a variety of tyrosine kinase proteins. In this study, the mutation analyses, CpG island methylation status, and the expression of the SOCS1 gene in 112 chronic myeloid leukaemia (CML) samples, five leukaemia cell lines, and 30 normal controls were analysed. No genetic mutations of SOCS1 gene were noted in the CML samples. The SOCS1 gene was hypermethylated in 67% and 46% of the blastic and chronic phase CML samples respectively (P < 0.0001). However, there was no methylation of the SOCS1 gene in normal controls or CML in molecular remission. The methylation status of the SOCS1 gene is consistent with the results of the real-time quantitative reverse transcription polymerase chain reaction and immunocytochemistry staining. Our results demonstrate that the SOCS1 gene silencing is caused by the methylation of CpG islands in CML and is reversed to an unmethylated status in molecular remission. As SOCS1 has universal activity to negatively regulate several cytokine signalling pathways, the loss of the negative regulation of cytokine signalling by the SOCS1 may play a role in the pathogenesis of CML progression.
Collapse
Affiliation(s)
- Ta-Chih Liu
- Division of Hematology-Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan.
| | | | | | | | | | | |
Collapse
|
810
|
Kawakami T, Okamoto K, Ogawa O, Okada Y. XIST unmethylated DNA fragments in male-derived plasma as a tumour marker for testicular cancer. Lancet 2004; 363:40-2. [PMID: 14723995 DOI: 10.1016/s0140-6736(03)15170-7] [Citation(s) in RCA: 91] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Testicular germ-cell tumours (TGCTs) are the most common malignant diseases among men aged 20-40 years. We developed a DNA tumour marker for TGCTs based on the unmethylated DNA profile of a neoplasm. The 5' end of the XIST gene is mainly hypomethylated in TGCTs irrespective of XIST expression. Male somatic cells, however, show complete methylation through the CpG sites, including the minimum promoter and XIST-conserved repeats. Identification of a XIST unmethylated fragment in male plasma might be diagnostic for TGCTs.
Collapse
MESH Headings
- Biomarkers, Tumor/blood
- Chromosomes, Human, X/genetics
- Chromosomes, Human, X/metabolism
- DNA Methylation
- DNA, Neoplasm/analysis
- DNA, Neoplasm/blood
- DNA, Neoplasm/metabolism
- Female
- Gene Expression Regulation, Neoplastic/genetics
- Germinoma/blood
- Germinoma/diagnosis
- Germinoma/genetics
- Humans
- Male
- Methylation
- RNA, Long Noncoding
- RNA, Untranslated/blood
- RNA, Untranslated/genetics
- RNA, Untranslated/metabolism
- Testicular Neoplasms/blood
- Testicular Neoplasms/diagnosis
- Testicular Neoplasms/genetics
Collapse
Affiliation(s)
- Takahiro Kawakami
- Department of Urology, Shiga University of Medical Science, Otsu, 520-2192, Shiga, Japan.
| | | | | | | |
Collapse
|
811
|
Maruyama R, Sugio K, Yoshino I, Maehara Y, Gazdar AF. Hypermethylation ofFHIT as a prognostic marker in nonsmall cell lung carcinoma. Cancer 2004; 100:1472-7. [PMID: 15042681 DOI: 10.1002/cncr.20144] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
BACKGROUND Methylation of CpG islands in the promoter and upstream coding regions has been identified as a mechanism for transcriptional inactivation of tumor suppressor genes. The purpose of the current study was to determine the correlation between the aberrant promoter methylation of multiple genes and survival in patients with nonsmall cell lung carcinoma (NSCLC). METHODS The methylation status of nine genes was determined in 124 surgically resected NSCLC cases using methylation-specific polymerase chain reaction. RESULTS The methylation frequencies of the genes tested in NSCLC specimens were 52% for E-cadherin (CDH1), 41% for RAS association domain family protein (RASSF1A), 38% for fragile histidine triad (FHIT) and adenomatous polyposis coli (APC), 27% for retinoic acid receptor beta (RARbeta) and H-cadherin (CDH13), 20% for p16INK4A, 0.8% for O6-methylguanine-DNA-methyltransferase (MGMT), and 0% for glutathione S-transferase P1 (GSTP1). The survival of the patients with FHIT methylation-positive tumors was found to be significantly shorter than that for those patients with methylation-negative tumors (P=0.03), even in those patients with International Union Against Cancer TNM Stage I or Stage II disease (P=0.007). In contrast, there were no significant survival differences noted between the methylation-positive and methylation-negative tumors for the other genes tested. In addition, based on multivariate analyses, FHIT methylation-positive status was found to be independently associated with poor survival (P=0.046) and disease stage (P<0.0001). CONCLUSIONS The results of the current study suggest that methylation of FHIT is a useful biomarker of biologically aggressive disease in patients with NSCLC.
Collapse
Affiliation(s)
- Riichiroh Maruyama
- Hamon Center for Therapeutic Oncology Research, the University of Texas Southwestern Medical Center, Dallas, Texas, USA.
| | | | | | | | | |
Collapse
|
812
|
Azhikina T, Gainetdinov I, Skvortsova Y, Batrak A, Dmitrieva N, Sverdlov E. Non-methylated Genomic Sites Coincidence Cloning (NGSCC): an approach to large scale analysis of hypomethylated CpG patterns at predetermined genomic loci. Mol Genet Genomics 2003; 271:22-32. [PMID: 14666421 DOI: 10.1007/s00438-003-0959-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2003] [Accepted: 11/07/2003] [Indexed: 10/26/2022]
Abstract
We have developed a new approach to the analysis of hypomethylated CpG patterns within predetermined, megabase long, genome regions. The approach, which we term Non-methylated Genomic Sites Coincidence Cloning (NGSCC), includes three main steps. First, total genomic DNA is digested with a methylation sensitive restriction endonuclease, such as HpaII or HhaI. Then the fragments corresponding to the genomic area of interest are selected. To this end the fragmented genome DNA is hybridized with a mixture of clones (BACs, cosmids etc.) representing a given region and digested with the same restriction enzyme(s). A special version of the coincidence cloning procedure was developed to make this hybridization selection highly efficient and specific. Finally, fragments of the locus under study are mapped and sequenced. The technique proved to be efficient and specific. As a test, it was applied to the analysis of hypomethylated CpG patterns along the 1-Mb D19S208-COX7A1 (Chr 19q13.12) locus, on human chromosome 19, in normal testis and in seminoma tissues. Some differences in the distribution of hypomethylated CpGs between the two tissues were demonstrated. The methylation profiles in both tissues revealed a clear trend to clustering of non-methylated sites. We also analyzed the expression of genes located within hypomethylated clusters in both tissues. It was shown that, whereas the expression of some of the genes investigated was correlated with hypomethylation of the region, other genes were expressed regardless of their methylation status. NGSCC thus promises to be a useful approach for the analysis of the role of dynamic epigenetic factors in genome function.
Collapse
Affiliation(s)
- T Azhikina
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117871 Moscow, Russia.
| | | | | | | | | | | |
Collapse
|
813
|
Blanchard F, Tracy E, Smith J, Chattopadhyay S, Wang Y, Held WA, Baumann H. DNA methylation controls the responsiveness of hepatoma cells to leukemia inhibitory factor. Hepatology 2003; 38:1516-28. [PMID: 14647063 DOI: 10.1016/j.hep.2003.09.029] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The related members of the interleukin 6 (IL-6) family of cytokines, IL-6, leukemia inhibitory factor (LIF), and oncostatin M, act as major inflammatory mediators and induce the hepatic acute phase reaction. Normal parenchymal liver cells express the receptors for these cytokines, and these receptors activate, to a comparable level, the intracellular signaling through signal transducer and activator of transcription (STAT) proteins and extracellular-regulated kinase (ERK). In contrast, hepatoma cell lines show attenuated responsiveness to some of these cytokines that is correlated with lower expression of the corresponding ligand-binding receptor subunits. This study tests the hypothesis that the reduced expression of LIF receptor (LIFR) observed in hepatoma cells is mediated by altered DNA methylation. H-35 rat hepatoma cells that have a greatly reduced LIF responsiveness were treated with 5-aza-2'-deoxycytidine, an inhibitor of DNA methyltransferase. Surviving and proliferating cells showed reestablished expression of LIFR protein and function. Restriction landmark genomic scanning (RLGS) demonstrated genome-wide drug-induced alterations in DNA methylation status, with striking similarities in the demethylation pattern among independently derived clonal lines. Upon extended growth in the absence of 5-aza-2'-deoxycytidine, the cells exhibit partial reversion to pretreatment patterns. Demethylation and remethylation of the CpG island within the LIFR promoter that is active in normal liver cells correlate with increased and decreased usage of this promoter in H-35 cells. In conclusion, these results indicate that transformed liver cells frequently undergo epigenetic alterations that suppress LIFR gene expression and modify the responsiveness to this IL-6 type cytokine.
Collapse
Affiliation(s)
- Frédéric Blanchard
- Department of Molecular and Cellular Biology, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | | | | | | | | | | | | |
Collapse
|
814
|
Affiliation(s)
- James G Herman
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Johns Hopkins Medical Institutions, Baltimore, USA.
| | | |
Collapse
|
815
|
Dominguez G, Silva J, Garcia JM, Silva JM, Rodriguez R, Muñoz C, Chacón I, Sanchez R, Carballido J, Colás A, España P, Bonilla F. Prevalence of aberrant methylation of p14ARF over p16INK4a in some human primary tumors. Mutat Res 2003; 530:9-17. [PMID: 14563526 DOI: 10.1016/s0027-5107(03)00133-7] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The INK4a/ARF locus encodes two unrelated tumor suppressor proteins, p16INK4a and p14ARF, which participate in the two main cell-cycle control pathways, p16-Rb and p14-p53. Methylation of CpG promoter islands has been described as a mechanism of gene silencing. Exon 1 of the p16INK4a gene and the p14ARF promoter gene reside within CpG islands. Therefore, both can become methylated de novo and silenced. It has recently been proposed that the methylation changes in certain genes could be used as molecular markers for the detection of almost all forms of human cancer. Here, we analyzed concomitantly in each tumor sample and normal tissue the methylation status of p16INK4a and p14ARF by methylation-specific PCR (MSP) in 100 breast, 95 colon and 27 bladder carcinomas. A series of clinicopathological parameter were obtained from the medical records of the patients, p14ARF showed a higher rate of hypermethylation than p16INK4a in all three tumor types. p16INK4a and p14ARF aberrant methylation was significantly correlated with poor prognosis clinicopathological parameters of the three tumor types. We conclude that both p16INKa and p14ARF hypermethylation may be involved in breast, colon and bladder carcinogenesis, with special emphasis on the role of the lesser studied p14ARF gene, and that tumors with aberrant methylation in the two genes were associated with worse prognosis.
Collapse
Affiliation(s)
- Gemma Dominguez
- Department of Medical Oncology, Hospital Universitario Puerta de Hierro, C/San Martin de Porres 4, E-28035, Madrid, Spain
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
816
|
Frühwald MC. DNA methylation patterns in cancer: novel prognostic indicators? AMERICAN JOURNAL OF PHARMACOGENOMICS : GENOMICS-RELATED RESEARCH IN DRUG DEVELOPMENT AND CLINICAL PRACTICE 2003; 3:245-60. [PMID: 12930158 DOI: 10.2165/00129785-200303040-00003] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
DNA methylation is an epigenetic phenomenon influencing the normal function of DNA and its scaffolding proteins. Especially in cancer, aberrant methylation patterns may contribute to the disease process by the induction of point mutations, activation of inactive genes through hypomethylation of promoters, and transcriptional inactivation through a complex interplay with histone acetylation and other inhibitory mechanisms. Aberrant methylation patterns have been evaluated as tools in the management of patients with cancer. The predictive value, the therapeutic manipulation and the prognostic significance of aberrantly methylated gene loci have been tested in hematological as well as in solid neoplasias in adults and children. A seemingly insurmountable wealth of data has been generated, however, data on clinical associations are sometimes presented in an almost incautious fashion. Nevertheless, some genes like p15INK4B in myelodysplastic syndrome (MDS) and p16INK4A in some lung cancer subtypes have been shown to confer a certain prognosis. In selected cases the data have been confirmed by independent studies. Assays have been developed that can be used by almost any clinical laboratory (e.g. methylation-specific PCR) for the rapid and affordable screening of tumors for aberrant methylation. The study of aberrant methylation patterns has successfully entered the arena of relevant clinical applications. Importantly, methylation does not only hold the potential for being 'just another' biomarker, but also, as it can be reverted chemically, it is a phenomenon that holds great promise for therapeutic exploitation.
Collapse
Affiliation(s)
- Michael C Frühwald
- Department of Pediatric Hematology and Oncology, University Children's Hospital Muenster, Albert-Schweitzer-Strasse 33, Muenster, Germany.
| |
Collapse
|
817
|
Abstract
Aberrant DNA methylation of the promoter region is a key mechanism for inactivation of genes that suppress tumorigenesis. Genes that are involved in every step of tumor formation can be silenced by this mechanism. Inhibitors of DNA methylation, such as 5-azadeoxycytidine (5AZA), can reverse this epigenetic event suggesting a potential use in cancer therapy. The structure of chromatin can also play an important role with respect to the regulation of gene expression. Chromatin containing hypoacetylated lysines in histones has a compact structure that is repressive for transcription. Inhibitors of histone deacetylase (HDAC) can convert chromatin to an open structure and activate certain genes that inhibit tumor growth. These HDAC inhibitors also have potential in cancer therapy. A 'cross-talk' between DNA methylation and histone deacetylation can occur and work in concert to silence gene expression. The molecular mechanism involves the attachment of a methylated CpG binding protein (MBP) to the methylated promoters and its recruitment of HDAC to form a complex that suppresses transcription. These two epigenetic modifications represent an interesting target for therapeutic intervention using 5AZA and HDAC inhibitors. These agents in combination have been shown to produce a synergistic reactivation of tumor suppressor genes and an enhanced antineoplastic effect against tumor cells, and should be investigated as a novel form of epigenetic therapy for cancer.
Collapse
Affiliation(s)
- Richard L Momparler
- Département de pharmacology, Université de Montréal, Centre de recherché, Hôpital Sainte-Justine, 3175 Côte Ste-Catherine Montreal, Que H3T 1C5, Canada.
| |
Collapse
|
818
|
Virmani A, Rathi A, Heda S, Sugio K, Lewis C, Tonk V, Takahashi T, Roth JA, Minna JD, Euhus DM, Gazdar AF. Aberrant methylation of the cyclin D2 promoter in primary small cell, nonsmall cell lung and breast cancers. Int J Cancer 2003; 107:341-5. [PMID: 14506731 DOI: 10.1002/ijc.11393] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
DNA methylation alteration of several genes contributes to human tumorigenesis. Cyclin D2, a member of the D-type cyclins, is implicated in cell cycle regulation and malignant transformation. In our study, we examined the methylation status of the cyclin D2 promoter in small cell lung cancer (SCLC), nonsmall cell lung cancer (NSCLC), breast tumors and tumor cell lines. We observed that aberrant methylation of cyclin D2 was present in 32 of 56 (57%) SCLC cell lines, 7 of 32 (22%) SCLC tumor tissues; 25 of 61 (47%) NSCLC cell lines, 19 of 48 (40%) NSCLC tumor tissues; 18 of 30 (60%) breast tumor cell lines and 19 of 63 (30%) breast tumor tissues. Methylation was more frequent in the tumor cell lines compared to the primary breast and SCLC tumors (p = 0.007 and p = 0.001, respectively). Methylation was rare in the control tissue samples; 0 of 12 peripheral blood lymphocytes; 0 of 12 buccal epithelial cells; 0 of 18 nonmalignant lung tissues and 3 of 28 (11%) nonmalignant breast tissues. Promoter methylation correlated with loss of transcript by reverse transcription PCR (RT-PCR) in 9 of 11 (6 lung, 5 breast) tumor cell lines tested. Two cell lines that were not methylated also lacked expression, suggesting that other mechanisms of inactivation may be involved. Expression was restored by treatment with the demethylating agent, 5 aza 2' deoxycytidine, in all 9 methylated cell lines. Our results confirm earlier reports in breast cancer and indicate that aberrant methylation of cyclin D2 may contribute to the pathogenesis of the 2 major types of lung cancers.
Collapse
Affiliation(s)
- Arvind Virmani
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX 75390-8593, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
819
|
Schuster JM, Longo M, Nelson PS. Differential expression of bikunin (HAI-2/PB), a proposed mediator of glioma invasion, by demethylation treatment. J Neurooncol 2003; 64:219-25. [PMID: 14558597 DOI: 10.1023/a:1025674227750] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Effective therapies for primary brain tumors continue to be elusive. Successful adjuvant therapies for CNS tumors will require a better understanding of their basic biology. Hepatocyte growth factor activator inhibitor type-2/placental bikunin (HAI-2/PB) is a serine proteinase inhibitor that has a broad inhibitory spectra against various serine proteinases. HAI-2/PB has anti-invasive effects thought to be mediated primarily by the inhibitory activity against serine proteinase-dependent matrix degradation. It has been previously demonstrated that the expression of HAI-2/PB is inversely related to degree of malignancy and possibly involved in the progression and invasion of human gliomas. Aberrant methylation patterns are an early change in glioma tumorigenesis, earlier than genetic changes. Methylation within 5' regulatory CpG islands by DNA methyltransferase is one of the most common epigenetic modifications. 5-Aza-2'-deoxycytidine (azacytidine) inhibits DNA methyltransferase and has been used in vitro to induce the expression of genes silenced by methylation. We have utilized azacytidine treatment and a micro-array system to investigate methylation influenced gene expression across several tumor cell lines of different lineage (brain, breast, prostate, liver). Using this system we have demonstrated that the expression of HAI-2/PB is under methylation control to a variable extent in glioma cell lines, in comparison to the other tested cell lines. Because the expression of HAI-2/B is inversely related to glioma invasiveness and degree of malignancy, this finding may provide insight into glioma initiation and progression as well as potentially providing new therapeutic targets.
Collapse
Affiliation(s)
- James M Schuster
- Department of Neurosurgery, University of Pennsylvania, Philadelphia 19107, USA.
| | | | | |
Collapse
|
820
|
Rousseau E, Ruchoux MM, Scaravilli F, Chapon F, Vinchon M, De Smet C, Godfraind C, Vikkula M. CDKN2A
, CDKN2B
and p14 ARF
are frequently and differentially methylated in ependymal tumours. Neuropathol Appl Neurobiol 2003; 29:574-83. [PMID: 14636164 DOI: 10.1046/j.0305-1846.2003.00505.x] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Ependymal tumours are histologically and clinically varied lesions. Numerical abnormalities of chromosome 9 are frequently associated with these tumours. Nevertheless, the three important tumour suppressor genes located in this chromosome, CDKN2A, CDKN2B and p14 ARF, have not been reported to be commonly altered in them. We studied promoter methylation of these genes, an important mechanism associated with gene silencing in a series of 152 ependymal tumours of WHO grades I to III. Methylation status of the CDKN2A, CDKN2B and p14 ARF promoters was assessed by methylation-specific polymerase chain reaction and the genetic results were correlated to clinicopathological features. We observed promoter methylation for CDKN2A in 21% (26/123) of tumours, for CDKN2B in 32% (23/71) and p14 ARF in 21% (23/108). For all three genes, posterior fossa ependymomas were less frequently methylated in paediatric patients than in adults. For CDKN2B, extracranial tumours were more frequently methylated than intracranial ones. For CDKN2B and p14 ARF, methylation was more frequent in low-grade tumours; the reverse was observed for CDKN2A. CDKN2A, CDKN2B and p14 ARF promoters were methylated in 21-32% of the tumours. Frequencies of methylation varied according to clinicopathological features. This suggests a role for these genes in ependymoma tumorigenesis.
Collapse
Affiliation(s)
- E Rousseau
- Laboratory of Neuropathology, Université Catholique de Louvain, Brussels, Belgium
| | | | | | | | | | | | | | | |
Collapse
|
821
|
Waha A, Waha A, Koch A, Meyer-Puttlitz B, Weggen S, Sörensen N, Tonn JC, Albrecht S, Goodyer CG, Berthold F, Wiestler OD, Pietsch T. Epigenetic Silencing of theHIC-1Gene in Human Medulloblastomas. J Neuropathol Exp Neurol 2003; 62:1192-201. [PMID: 14656076 DOI: 10.1093/jnen/62.11.1192] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The HIC-1 (hypermethylated in cancer) candidate tumor suppressor gene is located on chromosome 17p13.3, a region frequently deleted in medulloblastomas (MBs). MBs arising in the cerebellum represent the most common malignant brain tumors in children. In this study we have analyzed the sequence, methylation, and expression status of the HIC-1 gene in MBs. Hypermethylation of the 5'UTR and/or second exon of HIC-1 was detected in 33/39 (85%) of MB biopsies and in 7/8 (88%) of MB cell lines by methylation-specific PCR. There was a significant correlation (p < 0.001) between HIC-1 methylation and lack of transcription as determined by competitive RT-PCR. Treatment of the MB cell lines Daoy and MEB-MED-8A with 5-aza-2'deoxycytidine led to re-expression of HIC-1 transcripts, indicating a silencing of HIC-1 by CpG island methylation. Mutation analysis of the coding region of HIC-1 revealed a single deletion leading to an in-frame deletion of 4 amino acids in the second exon of HIC-1 (1/68, 1.5%). Our data indicate that a significant number of MBs exhibit strikingly reduced HIC-1 expression caused by altered CpG island methylation. These data suggest that epigenetic silencing of HIC-1 may well contribute to the pathogenesis in the majority of MBs.
Collapse
Affiliation(s)
- Anke Waha
- Department of Neuropathology, University of Bonn, Bonn, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
822
|
Taylor MD, Raffel C, Bayani J, Marrano P, Mainprize TG, Squire J. Advanced cancer genetics in neurosurgical research. Neurosurgery 2003; 53:1168-78; discussion 1178. [PMID: 14580285 DOI: 10.1227/01.neu.0000088807.95566.9b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2002] [Accepted: 07/11/2003] [Indexed: 11/18/2022] Open
Abstract
RAPID ADVANCES IN the technology used to study nucleic acids have revealed a great deal regarding the underlying biology of cancer. Most cancers arise as a result of chromosomal rearrangements and deoxyribonucleic acid mutations that lead to the activation of proto-oncogenes and loss of function of tumor suppressor genes. There are a number of different molecular routes that lead to these common goals, necessitating several different techniques of mutational analysis. Although many of these techniques can be difficult in practice, most are conceptually simple. We discuss several of the current techniques in cytogenetics and molecular genetics that are widely used in cancer biology laboratories. Understanding the molecular events that lead to cancer should allow the future development of targeted, nontoxic therapeutics similar to modern-day antibiotics. These technologies are being progressively applied in clinical neurosurgery, where they will be used to detect, diagnose, stratify, and treat cancers of the nervous system. High demand from an increasingly educated patient population means that neurosurgeons will need to be familiar with many of these techniques.
Collapse
Affiliation(s)
- Michael D Taylor
- Division of Neurosurgery, Hospital for Sick Children, Arthur and Sonia Labatt Brain Tumor Research Center, University of Toronto, Toronto, Ontario, Canada.
| | | | | | | | | | | |
Collapse
|
823
|
Ward RL, Cheong K, Ku SL, Meagher A, O'Connor T, Hawkins NJ. Adverse prognostic effect of methylation in colorectal cancer is reversed by microsatellite instability. J Clin Oncol 2003; 21:3729-36. [PMID: 14551292 DOI: 10.1200/jco.2003.03.123] [Citation(s) in RCA: 147] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
PURPOSE DNA methylation is an important biologic event in colorectal cancer and in some cases is associated with the development of microsatellite instability (MSI). In this study, we sought to determine the prognostic significance of DNA methylation, both in univariate analysis and in concert with other clinicopathologic factors known to influence outcome. PATIENTS AND METHODS Fresh tissue (625 cancers) was obtained from 605 individuals (age range, 29 to 99 years) undergoing curative surgery for colorectal cancer at one institution during a period of 8 years. Clinicopathologic details were recorded for all tumors, including stage, grade, type, vascular space invasion, and clinical follow-up to 5 years. Microsatellite status was assessed using standard markers. Methylation of p16 and hMLH1 promoters was determined by methylation-specific polymerase chain reaction (PCR), whereas methylation at methylated-in-tumor loci (MINT)1, MINT2, MINT12, and MINT31 loci were assessed by bisulfite-PCR. RESULTS Patients with microsatellite unstable tumors (12%) had better disease-specific survival than those with microsatellite stable (MSS) tumors (univariate analysis: hazard ratio [HR], 0.53; 95% CI, 0.27 to 1.0). Overall survival of individuals with MSS tumors was influenced by three independently significant factors: tumor stage (HR, 7.3; 95% CI, 5.1 to 10.4), heavy tumor methylation (HR, 2.1; 95% CI, 1.1 to 4.0), and vascular space invasion (HR, 1.9; 95% CI, 1.3 to 2.9). In MSS tumors, methylation at any single site was not independently predictive of survival. Neither methylation nor microsatellite status predicted a favorable response to chemotherapy. CONCLUSION DNA methylation is associated with a worse outcome in colorectal cancer, but this adverse prognostic influence is lost in those methylated tumors showing MSI. The mechanisms of these events warrant additional investigation.
Collapse
Affiliation(s)
- Robyn Lynne Ward
- Department of Medical Oncology and Colorectal Surgery, St Vincent's Hospital, Darlinghurst, Australia.
| | | | | | | | | | | |
Collapse
|
824
|
Feltus FA, Lee EK, Costello JF, Plass C, Vertino PM. Predicting aberrant CpG island methylation. Proc Natl Acad Sci U S A 2003; 100:12253-8. [PMID: 14519846 PMCID: PMC218745 DOI: 10.1073/pnas.2037852100] [Citation(s) in RCA: 171] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Epigenetic silencing associated with aberrant methylation of promoter region CpG islands is one mechanism leading to loss of tumor suppressor function in human cancer. Profiling of CpG island methylation indicates that some genes are more frequently methylated than others, and that each tumor type is associated with a unique set of methylated genes. However, little is known about why certain genes succumb to this aberrant event. To address this question, we used Restriction Landmark Genome Scanning to analyze the susceptibility of 1,749 unselected CpG islands to de novo methylation driven by overexpression of DNA cytosine-5-methyltransferase 1 (DNMT1). We found that although the overall incidence of CpG island methylation was increased in cells overexpressing DNMT1, not all loci were equally affected. The majority of CpG islands (69.9%) were resistant to de novo methylation, regardless of DNMT1 overexpression. In contrast, we identified a subset of methylation-prone CpG islands (3.8%) that were consistently hypermethylated in multiple DNMT1 overexpressing clones. Methylation-prone and methylation-resistant CpG islands were not significantly different with respect to size, C+G content, CpG frequency, chromosomal location, or promoter association. We used DNA pattern recognition and supervised learning techniques to derive a classification function based on the frequency of seven novel sequence patterns that was capable of discriminating methylation-prone from methylation-resistant CpG islands with 82% accuracy. The data indicate that CpG islands differ in their intrinsic susceptibility to de novo methylation, and suggest that the propensity for a CpG island to become aberrantly methylated can be predicted based on its sequence context.
Collapse
Affiliation(s)
- F A Feltus
- Department of Radiation Oncology and Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, USA
| | | | | | | | | |
Collapse
|
825
|
Motiwala T, Ghoshal K, Das A, Majumder S, Weichenhan D, Wu YZ, Holman K, James SJ, Jacob ST, Plass C. Suppression of the protein tyrosine phosphatase receptor type O gene (PTPRO) by methylation in hepatocellular carcinomas. Oncogene 2003; 22:6319-31. [PMID: 14508512 PMCID: PMC3020652 DOI: 10.1038/sj.onc.1206750] [Citation(s) in RCA: 91] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2003] [Revised: 04/23/2003] [Accepted: 04/26/2003] [Indexed: 11/09/2022]
Abstract
A diet lacking folic acid and choline and low in methionine (folate/methyl deficient diet, FMD diet) fed to rats is known to produce preneoplastic nodules (PNNs) after 36 weeks and hepatocellular carcinomas (tumors) after 54 weeks. FMD diet-induced tumors exhibit global hypomethylation and regional hypermethylation. Restriction landmark genome scanning analysis with methylation-sensitive enzyme NotI (RLGS-M) of genomic DNA isolated from control livers, PNNs and tumor tissues was performed to identify the genes that are differentially methylated or amplified during multistage hepatocarcinogenesis. Out of the 1250 genes analysed, 2 to 5 genes were methylated in the PNNs, whereas 5 to 45 genes were partially or completely methylated in the tumors. This analysis also showed amplification of 3 to 12 genes in the primary tumors. As a first step towards identifying the genes methylated in the PNNs and primary hepatomas, we generated a rat NotI-EcoRV genomic library in the pBluescriptKS vector. Here, we describe identification of one methylated and downregulated gene as the rat protein tyrosine phosphatase receptor type O (PTPRO) and one amplified gene as rat C-MYC. Methylation of PTPRO at the NotI site located immediate upstream of the trancription start site in the PNNs and tumors, and amplification of C-MYC gene in the tumors were confirmed by Southern blot analyses. Bisulfite genomic sequencing of the CpG island encompassing exon 1 of the PTPRO gene revealed dense methylation in the PNNs and tumors, whereas it was methylation free in the livers of animals on normal diet. Reverse transcription-polymerase chain reaction (RT-PCR) analysis showed significant decrease in the expression of PTPRO in the tumors and in a transplanted rat hepatoma. The expression of PTPRO mRNA in the transplanted hepatoma after demethylation with 5-azacytidine, a potent inhibitor of DNA methyltransferases, further confirmed the role of methylation in PTPRO gene expression. These results demonstrate alteration in methylation profile and expression of specific genes during tumor progression in the livers of rats in response to folate/methyl deficiency, and further implicate the potential role of PTPRO as a novel growth regulatory gene at least in the hepatocellular carcinomas.
Collapse
Affiliation(s)
- Tasneem Motiwala
- Department of Molecular and Cellular Biochemistry, The Ohio State University, Columbus, OH 43210, USA
| | - Kalpana Ghoshal
- Department of Molecular and Cellular Biochemistry, The Ohio State University, Columbus, OH 43210, USA
| | - Anindita Das
- Department of Molecular and Cellular Biochemistry, The Ohio State University, Columbus, OH 43210, USA
| | - Sarmila Majumder
- Department of Molecular and Cellular Biochemistry, The Ohio State University, Columbus, OH 43210, USA
| | - Dieter Weichenhan
- Medizinische Universität zu Lübeck, Institut für Biologie, Ratzeburger Allee 160, 23538 Lübeck, Germany
| | - Yue-Zhong Wu
- Division of Human Cancer Genetics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Kristen Holman
- Division of Human Cancer Genetics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - S Jill James
- Division of Biochemical Toxicology, Food and Drug administration, National Center for Toxicological Research, Jefferson, AR 72079, USA
| | - Samson T Jacob
- Department of Molecular and Cellular Biochemistry, The Ohio State University, Columbus, OH 43210, USA
| | - Christoph Plass
- Division of Human Cancer Genetics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
826
|
Gutierrez MI, Siraj AK, Bhargava M, Ozbek U, Banavali S, Chaudhary MA, El Solh H, Bhatia K. Concurrent methylation of multiple genes in childhood ALL: Correlation with phenotype and molecular subgroup. Leukemia 2003; 17:1845-50. [PMID: 12970785 DOI: 10.1038/sj.leu.2403060] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Multiple genes have been shown to be independently hypermethylated in lymphoid malignancies. We report here on the extent of concurrent methylation of E-cadherin, Dap-kinase, O(6)MGMT, p73, p16, p15 and p14 in 129 pediatric ALL cases. While most of these genes demonstrated methylation in a proportion of cases, O(6)MGMT, p16 and p14 were infrequently methylated (11, 7 and 3%, respectively). Methylation of at least one gene was found in the vast majority (83%) of cases. To determine the extent and concordance of methylation we calculated a methylation index (MI=number of methylated genes/number of studied genes) for each sample. The average MI was 0.28, corresponding to 2/7 methylated genes. MI was correlated with standard prognostic factors, including immunophenotype, age, sex, WBC and presence of specific translocations (TEL-AML1, BCR-ABL, E2A-PBX1 or MLL-AF4). We determined that children >/=10 years old and children presenting with high WBC (>/=50 x 10(9)/l) both associated with a higher MI (P<0.01 and <0.05, respectively). T-ALLs demonstrated a lower MI (median=0.17) than precursor B ALLs (median=0.28). Among the different molecular subgroups, MLL-ALLs had the highest MI (mean=0.35), while ALLs carrying the t(1;19) had the lowest MI (mean=0.07). The most common epigenetic lesion in childhood ALL was methylation of E-cadherin (72%) independent of the molecular subtype or other clinicopathological factors.
Collapse
Affiliation(s)
- M I Gutierrez
- King Fahad National Centre for Children's Cancer and Research, Riyadh, Saudi Arabia
| | | | | | | | | | | | | | | |
Collapse
|
827
|
Uhlmann K, Rohde K, Zeller C, Szymas J, Vogel S, Marczinek K, Thiel G, Nürnberg P, Laird PW. Distinct methylation profiles of glioma subtypes. Int J Cancer 2003; 106:52-9. [PMID: 12794756 DOI: 10.1002/ijc.11175] [Citation(s) in RCA: 78] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Gliomas are tumors of the central nervous system with a wide spectrum of different tumor types. They range from pilocytic astrocytoma, with a generally good prognosis, to the extremely aggressive malignant glioblastoma. In addition to these 2 types of contrasting neoplasms, several other subtypes can be distinguished, each characterized by specific phenotypic, as well as genotypic features. Recently, the epigenotype, as evident from differentially methylated DNA loci, has been proposed to be useful as a further criterion to distinguish between tumor types. In our study, we screened 139 tissue samples, including 33 pilocytic astrocytomas, 46 astrocytomas of different grades, 7 oligoastrocytomas, 10 oligodendrogliomas, 10 glioblastoma multiforme samples and 33 control tissues, for methylation at CpG islands of 15 different gene loci. We used the semiquantitative high throughput method MethyLight to analyze a gene panel comprising ARF, CDKN2B, RB1, APC, CDH1, ESR1, GSTP1, TGFBR2, THBS1, TIMP3, PTGS2, CTNNB1, CALCA, MYOD1 and HIC1. Seven of these loci showed tumor specific methylation changes. We found tissue as well as grade specific methylation profiles. Interestingly, pilocytic astrocytomas showed no evidence of CpG island hypermethylation, but were significantly hypomethylated, relative to control tissues, at MYOD1. Our results show that glioma subtypes have characteristic methylation profiles and, with the exception of pilocytic astrocytomas, show both locus specific hyper- as well as hypomethylation.
Collapse
Affiliation(s)
- Karen Uhlmann
- Institut für Medizinische Genetik, Universitätsklinikum Charité, Humboldt-Universität, Berlin, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
828
|
Chim CS, Wong SY, Kwong YL. Aberrant gene promoter methylation in acute promyelocytic leukaemia: profile and prognostic significance. Br J Haematol 2003; 122:571-8. [PMID: 12899712 DOI: 10.1046/j.1365-2141.2003.04462.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Acute promyelocytic leukaemia (APL) has distinct clinicopathological and molecular features. However, the profile of aberrant gene promoter methylation is undefined. In this study, methylation-specific polymerase chain reaction (MSP) was used to define the methylation status of a panel of nine genes, comprising p15, p16, RARbeta, oestrogen receptor (ER), E-cadherin (E-CAD), p73, caspase 8 (CASP8), VHL and MGMT, in 29 patients with APL. Aberrant methylation of p15, ER, RARbeta, p16 and E-CAD occurred, respectively, in 23 (79%), 14 (48%), six (21%), six (21%) and two (7%) patients at diagnosis, but p73, VHL, CASP8 and MGMT were not methylated in any patients. There was methylation of one gene in 13 patients (45%), two genes in four patients (14%), three genes in six patients (21%) and four genes in three patients (10%). Concurrent methylation of two or more genes occurred in 13 patients (45%). No association was identified between gene methylation and presenting clinicopathological features. However, p15 methylation was significantly associated with an inferior disease-free survival (DFS, P = 0.008), and remained the only poor prognostic factor in multivariate analysis (P = 0.019). In APL, p15, p16, ER and RARbeta were most frequently methylated. This profile is distinct from other types of myeloid leukaemias. p15 methylation has a poor prognostic impact on DFS.
Collapse
Affiliation(s)
- Chor S Chim
- University Department of Medicine, Queen Mary Hospital, Pokfulam Road, Hong Kong
| | | | | |
Collapse
|
829
|
Matsuyama T, Kimura MT, Koike K, Abe T, Nakano T, Asami T, Ebisuzaki T, Held WA, Yoshida S, Nagase H. Global methylation screening in the Arabidopsis thaliana and Mus musculus genome: applications of virtual image restriction landmark genomic scanning (Vi-RLGS). Nucleic Acids Res 2003; 31:4490-6. [PMID: 12888509 PMCID: PMC169878 DOI: 10.1093/nar/gkg488] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Understanding the role of 'epigenetic' changes such as DNA methylation and chromatin remodeling has now become critical in understanding many biological processes. In order to delineate the global methylation pattern in a given genomic DNA, computer software has been developed to create a virtual image of restriction landmark genomic scanning (Vi-RLGS). When using a methylation- sensitive enzyme such as NotI as the restriction landmark, the comparison between real and in silico RLGS profiles of the genome provides a methylation map of genomic NotI sites. A methylation map of the Arabidopsis genome was created that could be confirmed by a methylation-sensitive PCR assay. The method has also been applied to the mouse genome. Although a complete methylation map has not been completed, a region of methylation difference between two tissues has been tested and confirmed by bisulfite sequencing. Vi-RLGS in conjunction with real RLGS will make it possible to develop a more complete map of genomic sites that are methylated or demethylated as a consequence of normal or abnormal development.
Collapse
Affiliation(s)
- Tomoki Matsuyama
- Plant Functions Laboratory, RIKEN (The Institute of Physical and Chemical Research), Wako, Saitama 351-0198, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
830
|
Li H, Myeroff L, Smiraglia D, Romero MF, Pretlow TP, Kasturi L, Lutterbaugh J, Rerko RM, Casey G, Issa JP, Willis J, Willson JKV, Plass C, Markowitz SD. SLC5A8, a sodium transporter, is a tumor suppressor gene silenced by methylation in human colon aberrant crypt foci and cancers. Proc Natl Acad Sci U S A 2003; 100:8412-7. [PMID: 12829793 PMCID: PMC166243 DOI: 10.1073/pnas.1430846100] [Citation(s) in RCA: 231] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
We identify a gene, SLC5A8, and show it is a candidate tumor suppressor gene whose silencing by aberrant methylation is a common and early event in human colon neoplasia. Aberrant DNA methylation has been implicated as a component of an epigenetic mechanism that silences genes in human cancers. Using restriction landmark genome scanning, we performed a global search to identify genes that would be aberrantly methylated at high frequency in human colon cancer. From among 1,231 genomic NotI sites assayed, site 3D41 was identified as methylated in 11 of 12 colon cancers profiled. Site 3D41 mapped to exon 1 of SLC5A8, a transcript that we assembled. In normal colon mucosa we found that SLC5A8 exon 1 is unmethylated and SLC5A8 transcript is expressed. In contrast, SLC5A8 exon 1 proved to be aberrantly methylated in 59% of primary colon cancers and 52% of colon cancer cell lines. SLC5A8 exon 1 methylated cells were uniformly silenced for SLC5A8 expression, but reactivated expression on treatment with a demethylating drug, 5-azacytidine. Transfection of SLC5A8 suppressed colony growth in each of three SLC5A8-deficient cell lines, but showed no suppressive effect in any of three SLC5A8-proficient cell lines. SLC5A8 exon 1 methylation is an early event, detectable in colon adenomas, and in even earlier microscopic colonic aberrant crypt foci. Structural homology and functional testing demonstrated that SLC5A8 is a member of the family of sodium solute symporters, which are now added as a class of candidate colon cancer suppressor genes.
Collapse
Affiliation(s)
- Hui Li
- Cancer Center and Department of Medicine, and
Ireland Cancer Center, Department of Medicine, and Research Institute,
University Hospitals of Cleveland and Case Western Reserve University,
Cleveland, OH 44106; Departments of Molecular
Biology and Microbiology, and Physiology and
Biophysics, and Department of Pathology and
Cancer Center, Case Western Reserve University, Cleveland, OH 44106;
Division of Human Cancer Genetics, Ohio State
University, Columbus, OH 43210; Howard Hughes
Medical Institute, Cleveland, OH 44106; Lerner
Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195;
Department of Leukemia, University of
Texas M. D. Anderson Cancer Center, Houston, TX 77030; and
Department of Pathology and Ireland
Cancer Center, University Hospitals of Cleveland and Case Western Reserve
University, Cleveland, OH 44106
| | - Lois Myeroff
- Cancer Center and Department of Medicine, and
Ireland Cancer Center, Department of Medicine, and Research Institute,
University Hospitals of Cleveland and Case Western Reserve University,
Cleveland, OH 44106; Departments of Molecular
Biology and Microbiology, and Physiology and
Biophysics, and Department of Pathology and
Cancer Center, Case Western Reserve University, Cleveland, OH 44106;
Division of Human Cancer Genetics, Ohio State
University, Columbus, OH 43210; Howard Hughes
Medical Institute, Cleveland, OH 44106; Lerner
Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195;
Department of Leukemia, University of
Texas M. D. Anderson Cancer Center, Houston, TX 77030; and
Department of Pathology and Ireland
Cancer Center, University Hospitals of Cleveland and Case Western Reserve
University, Cleveland, OH 44106
| | - Dominic Smiraglia
- Cancer Center and Department of Medicine, and
Ireland Cancer Center, Department of Medicine, and Research Institute,
University Hospitals of Cleveland and Case Western Reserve University,
Cleveland, OH 44106; Departments of Molecular
Biology and Microbiology, and Physiology and
Biophysics, and Department of Pathology and
Cancer Center, Case Western Reserve University, Cleveland, OH 44106;
Division of Human Cancer Genetics, Ohio State
University, Columbus, OH 43210; Howard Hughes
Medical Institute, Cleveland, OH 44106; Lerner
Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195;
Department of Leukemia, University of
Texas M. D. Anderson Cancer Center, Houston, TX 77030; and
Department of Pathology and Ireland
Cancer Center, University Hospitals of Cleveland and Case Western Reserve
University, Cleveland, OH 44106
| | - Michael F. Romero
- Cancer Center and Department of Medicine, and
Ireland Cancer Center, Department of Medicine, and Research Institute,
University Hospitals of Cleveland and Case Western Reserve University,
Cleveland, OH 44106; Departments of Molecular
Biology and Microbiology, and Physiology and
Biophysics, and Department of Pathology and
Cancer Center, Case Western Reserve University, Cleveland, OH 44106;
Division of Human Cancer Genetics, Ohio State
University, Columbus, OH 43210; Howard Hughes
Medical Institute, Cleveland, OH 44106; Lerner
Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195;
Department of Leukemia, University of
Texas M. D. Anderson Cancer Center, Houston, TX 77030; and
Department of Pathology and Ireland
Cancer Center, University Hospitals of Cleveland and Case Western Reserve
University, Cleveland, OH 44106
| | - Theresa P. Pretlow
- Cancer Center and Department of Medicine, and
Ireland Cancer Center, Department of Medicine, and Research Institute,
University Hospitals of Cleveland and Case Western Reserve University,
Cleveland, OH 44106; Departments of Molecular
Biology and Microbiology, and Physiology and
Biophysics, and Department of Pathology and
Cancer Center, Case Western Reserve University, Cleveland, OH 44106;
Division of Human Cancer Genetics, Ohio State
University, Columbus, OH 43210; Howard Hughes
Medical Institute, Cleveland, OH 44106; Lerner
Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195;
Department of Leukemia, University of
Texas M. D. Anderson Cancer Center, Houston, TX 77030; and
Department of Pathology and Ireland
Cancer Center, University Hospitals of Cleveland and Case Western Reserve
University, Cleveland, OH 44106
| | - Lakshmi Kasturi
- Cancer Center and Department of Medicine, and
Ireland Cancer Center, Department of Medicine, and Research Institute,
University Hospitals of Cleveland and Case Western Reserve University,
Cleveland, OH 44106; Departments of Molecular
Biology and Microbiology, and Physiology and
Biophysics, and Department of Pathology and
Cancer Center, Case Western Reserve University, Cleveland, OH 44106;
Division of Human Cancer Genetics, Ohio State
University, Columbus, OH 43210; Howard Hughes
Medical Institute, Cleveland, OH 44106; Lerner
Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195;
Department of Leukemia, University of
Texas M. D. Anderson Cancer Center, Houston, TX 77030; and
Department of Pathology and Ireland
Cancer Center, University Hospitals of Cleveland and Case Western Reserve
University, Cleveland, OH 44106
| | - James Lutterbaugh
- Cancer Center and Department of Medicine, and
Ireland Cancer Center, Department of Medicine, and Research Institute,
University Hospitals of Cleveland and Case Western Reserve University,
Cleveland, OH 44106; Departments of Molecular
Biology and Microbiology, and Physiology and
Biophysics, and Department of Pathology and
Cancer Center, Case Western Reserve University, Cleveland, OH 44106;
Division of Human Cancer Genetics, Ohio State
University, Columbus, OH 43210; Howard Hughes
Medical Institute, Cleveland, OH 44106; Lerner
Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195;
Department of Leukemia, University of
Texas M. D. Anderson Cancer Center, Houston, TX 77030; and
Department of Pathology and Ireland
Cancer Center, University Hospitals of Cleveland and Case Western Reserve
University, Cleveland, OH 44106
| | - Ronald M. Rerko
- Cancer Center and Department of Medicine, and
Ireland Cancer Center, Department of Medicine, and Research Institute,
University Hospitals of Cleveland and Case Western Reserve University,
Cleveland, OH 44106; Departments of Molecular
Biology and Microbiology, and Physiology and
Biophysics, and Department of Pathology and
Cancer Center, Case Western Reserve University, Cleveland, OH 44106;
Division of Human Cancer Genetics, Ohio State
University, Columbus, OH 43210; Howard Hughes
Medical Institute, Cleveland, OH 44106; Lerner
Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195;
Department of Leukemia, University of
Texas M. D. Anderson Cancer Center, Houston, TX 77030; and
Department of Pathology and Ireland
Cancer Center, University Hospitals of Cleveland and Case Western Reserve
University, Cleveland, OH 44106
| | - Graham Casey
- Cancer Center and Department of Medicine, and
Ireland Cancer Center, Department of Medicine, and Research Institute,
University Hospitals of Cleveland and Case Western Reserve University,
Cleveland, OH 44106; Departments of Molecular
Biology and Microbiology, and Physiology and
Biophysics, and Department of Pathology and
Cancer Center, Case Western Reserve University, Cleveland, OH 44106;
Division of Human Cancer Genetics, Ohio State
University, Columbus, OH 43210; Howard Hughes
Medical Institute, Cleveland, OH 44106; Lerner
Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195;
Department of Leukemia, University of
Texas M. D. Anderson Cancer Center, Houston, TX 77030; and
Department of Pathology and Ireland
Cancer Center, University Hospitals of Cleveland and Case Western Reserve
University, Cleveland, OH 44106
| | - Jean-Pierre Issa
- Cancer Center and Department of Medicine, and
Ireland Cancer Center, Department of Medicine, and Research Institute,
University Hospitals of Cleveland and Case Western Reserve University,
Cleveland, OH 44106; Departments of Molecular
Biology and Microbiology, and Physiology and
Biophysics, and Department of Pathology and
Cancer Center, Case Western Reserve University, Cleveland, OH 44106;
Division of Human Cancer Genetics, Ohio State
University, Columbus, OH 43210; Howard Hughes
Medical Institute, Cleveland, OH 44106; Lerner
Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195;
Department of Leukemia, University of
Texas M. D. Anderson Cancer Center, Houston, TX 77030; and
Department of Pathology and Ireland
Cancer Center, University Hospitals of Cleveland and Case Western Reserve
University, Cleveland, OH 44106
| | - Joseph Willis
- Cancer Center and Department of Medicine, and
Ireland Cancer Center, Department of Medicine, and Research Institute,
University Hospitals of Cleveland and Case Western Reserve University,
Cleveland, OH 44106; Departments of Molecular
Biology and Microbiology, and Physiology and
Biophysics, and Department of Pathology and
Cancer Center, Case Western Reserve University, Cleveland, OH 44106;
Division of Human Cancer Genetics, Ohio State
University, Columbus, OH 43210; Howard Hughes
Medical Institute, Cleveland, OH 44106; Lerner
Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195;
Department of Leukemia, University of
Texas M. D. Anderson Cancer Center, Houston, TX 77030; and
Department of Pathology and Ireland
Cancer Center, University Hospitals of Cleveland and Case Western Reserve
University, Cleveland, OH 44106
| | - James K. V. Willson
- Cancer Center and Department of Medicine, and
Ireland Cancer Center, Department of Medicine, and Research Institute,
University Hospitals of Cleveland and Case Western Reserve University,
Cleveland, OH 44106; Departments of Molecular
Biology and Microbiology, and Physiology and
Biophysics, and Department of Pathology and
Cancer Center, Case Western Reserve University, Cleveland, OH 44106;
Division of Human Cancer Genetics, Ohio State
University, Columbus, OH 43210; Howard Hughes
Medical Institute, Cleveland, OH 44106; Lerner
Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195;
Department of Leukemia, University of
Texas M. D. Anderson Cancer Center, Houston, TX 77030; and
Department of Pathology and Ireland
Cancer Center, University Hospitals of Cleveland and Case Western Reserve
University, Cleveland, OH 44106
| | - Christoph Plass
- Cancer Center and Department of Medicine, and
Ireland Cancer Center, Department of Medicine, and Research Institute,
University Hospitals of Cleveland and Case Western Reserve University,
Cleveland, OH 44106; Departments of Molecular
Biology and Microbiology, and Physiology and
Biophysics, and Department of Pathology and
Cancer Center, Case Western Reserve University, Cleveland, OH 44106;
Division of Human Cancer Genetics, Ohio State
University, Columbus, OH 43210; Howard Hughes
Medical Institute, Cleveland, OH 44106; Lerner
Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195;
Department of Leukemia, University of
Texas M. D. Anderson Cancer Center, Houston, TX 77030; and
Department of Pathology and Ireland
Cancer Center, University Hospitals of Cleveland and Case Western Reserve
University, Cleveland, OH 44106
| | - Sanford D. Markowitz
- Cancer Center and Department of Medicine, and
Ireland Cancer Center, Department of Medicine, and Research Institute,
University Hospitals of Cleveland and Case Western Reserve University,
Cleveland, OH 44106; Departments of Molecular
Biology and Microbiology, and Physiology and
Biophysics, and Department of Pathology and
Cancer Center, Case Western Reserve University, Cleveland, OH 44106;
Division of Human Cancer Genetics, Ohio State
University, Columbus, OH 43210; Howard Hughes
Medical Institute, Cleveland, OH 44106; Lerner
Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195;
Department of Leukemia, University of
Texas M. D. Anderson Cancer Center, Houston, TX 77030; and
Department of Pathology and Ireland
Cancer Center, University Hospitals of Cleveland and Case Western Reserve
University, Cleveland, OH 44106
- To whom correspondence should be addressed. E-mail:
| |
Collapse
|
831
|
Chen CM, Chen HL, Hsiau THC, Hsiau AHA, Shi H, Brock GJR, Wei SH, Caldwell CW, Yan PS, Huang THM. Methylation target array for rapid analysis of CpG island hypermethylation in multiple tissue genomes. THE AMERICAN JOURNAL OF PATHOLOGY 2003; 163:37-45. [PMID: 12819009 PMCID: PMC1868173 DOI: 10.1016/s0002-9440(10)63628-0] [Citation(s) in RCA: 77] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Hypermethylation of multiple CpG islands is a common event in cancer. To assess the prognostic values of this epigenetic alteration, we developed Methylation Target Array (MTA), derived from the concept of tissue microarray, for simultaneous analysis of DNA hypermethylation in hundreds of tissue genomes. In MTA, linker-ligated CpG island fragments were digested with methylation-sensitive endonucleases and amplified with flanking primers. A panel of 468 MTA amplicons, which represented the whole repertoire of methylated CpG islands in 93 breast tumors, 20 normal breast tissues, and 4 breast cancer cell lines, were arrayed on nylon membrane for probe hybridization. Positive hybridization signals detected in tumor amplicons, but not in normal amplicons, were indicative of aberrant hypermethylation in tumor samples. This is attributed to aberrant sites that were protected from methylation-sensitive restriction and were amplified by PCR in tumor samples, while the same sites were restricted and could not be amplified in normal samples. Hypermethylation frequencies of the 10 genes tested in breast tumors and cancer cell lines were 60% for GPC3, 58% for RASSF1A, 32% for 3OST3B, 30% for HOXA5, 28% for uPA, 25% for WT1, 23% for BRCA1, 9% for DAPK1, and 0% for KL. Furthermore, hypermethylation of 5 to 7 loci of these genes was significantly correlated with hormone receptor status, clinical stages, and ages at diagnosis of the patients analyzed. This novel approach thus provides an additional avenue for assessing clinicopathological consequences of DNA hypermethylation in breast cancer.
Collapse
Affiliation(s)
- Chuan-Mu Chen
- Department of Life Sciences, National Chung Hsing University, Taiwan, Republic of China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
832
|
Yue CM, Deng DJ, Bi MX, Guo LP, Lu SH. Expression of ECRG4, a novel esophageal cancer-related gene, downregulated by CpG island hypermethylation in human esophageal squamous cell carcinoma. World J Gastroenterol 2003; 9:1174-8. [PMID: 12800218 PMCID: PMC4611778 DOI: 10.3748/wjg.v9.i6.1174] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To study the mechanisms responsible for inactivation of a novel esophageal cancer related gene 4 (ECRG4) in esophageal squamous cell carcinoma (ESCC).
METHODS: A pair of primers was designed to amplify a 220 bp fragment, which contains 16 CpG sites in the core promoter region of the ECRG 4 gene. PCR products of bisulfite-modified CpG islands were analyzed by denaturing high-performance liquid chromatography (DHPLC), which were confirmed by DNA sequencing. The methylation status of ECRG 4 promoter in 20 cases of esophageal cancer and the adjacent normal tissues, 5 human tumor cell lines (esophageal cancer cell line-NEC, EC109, EC9706; gastric cancer cell line- GLC; human embryo kidney cell line-Hek293) and 2 normal esophagus tissues were detected. The expression level of the ECRG 4 gene in these samples was examined by RT-PCR.
RESULTS: The expression level of ECRG 4 gene was varied. Of 20 esophageal cancer tissues, nine were unexpressed, six were lowly expressed and five were highly expressed compared with the adjacent tissues and the 2 normal esophageal epithelia. In addition, 4 out of the 5 human cell lines were also unexpressed. A high frequency of methylation was revealed in 12 (8 unexpressed and 4 lowly expressed) of the 15 (80%) downregulated cancer tissues and 3 of the 4 unexpressed cell lines. No methylation peak was observed in the two highly expressed normal esophageal epithelia and the methylation frequency was low (3/20) among the 20 cases in the highly expressed adjacent tissues. The methylation status of the samples was consistent with the result of DNA sequencing.
CONCLUSION: These results indicate that the inactivation of ECRG 4 gene by hypermethylation is a frequent molecular event in ESCC and may be involved in the carcinogenesis of this cancer.
Collapse
Affiliation(s)
- Chun-Mei Yue
- Department of Etiology and Carcinogenesis, Cancer Institute, Chinese Academy of Medical Sciences, Beijing 100021, China
| | | | | | | | | |
Collapse
|
833
|
Narayan G, Arias-Pulido H, Koul S, Vargas H, Zhang FF, Villella J, Schneider A, Terry MB, Mansukhani M, Murty VV. Frequent promoter methylation of CDH1, DAPK, RARB, and HIC1 genes in carcinoma of cervix uteri: its relationship to clinical outcome. Mol Cancer 2003; 2:24. [PMID: 12773202 PMCID: PMC156646 DOI: 10.1186/1476-4598-2-24] [Citation(s) in RCA: 180] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2003] [Accepted: 05/13/2003] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Cervical cancer (CC), a leading cause of cancer-related deaths in women worldwide, has been causally linked to genital human papillomavirus (HPV) infection. Although a host of genetic alterations have been identified, molecular basis of CC development is still poorly understood. RESULTS We examined the role of promoter hypermethylation, an epigenetic alteration that is associated with the silencing tumor suppressor genes in human cancer, by studying 16 gene promoters in 90 CC cases. We found a high frequency of promoter methylation in CDH1, DAPK, RARB, and HIC1 genes. Correlation of promoter methylation with clinical characteristics and other genetic changes revealed the following: a) overall promoter methylation was higher in more advanced stage of the disease, b) promoter methylation of RARB and BRCA1 predicted worse prognosis, and c) the HIC1 promoter methylation was frequently seen in association with microsatellite instability. Promoter methylation was associated with gene silencing in CC cell lines. Treatment with methylation or histone deacetylation-inhibiting agents resulted in profound reactivation of gene expression. CONCLUSIONS These results may have implications in understanding the underlying epigenetic mechanisms in CC development, provide prognostic indicators, and identify important gene targets for treatment.
Collapse
Affiliation(s)
- Gopeshwar Narayan
- Department of Pathology, Columbia University, New York, New York 10032, USA
| | - Hugo Arias-Pulido
- Department of Molecular Genetics and Microbiology, University of New Mexico, Albuquerque, New Mexico, USA
| | - Sanjay Koul
- Department of Pathology, Columbia University, New York, New York 10032, USA
| | - Hernan Vargas
- Department of Tumor Molecular Biology, Instituto Nacional de Cancerología, Bogotá, Colombia
| | - Fang F Zhang
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, New York, 10032, USA
| | - Jeannine Villella
- Department of Obstetrics & Gynecology, Columbia University, New York, New York 10032, USA
| | - Achim Schneider
- Department of Obstetrics & Gynecology, Friedrich Schiller University, Jena, 07740, Germany
| | - Mary B Terry
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, New York, 10032, USA
| | - Mahesh Mansukhani
- Department of Pathology, Columbia University, New York, New York 10032, USA
| | - Vundavalli V Murty
- Department of Pathology, Columbia University, New York, New York 10032, USA
- Institute for Cancer Genetics, College of Physicians & Surgeons of Columbia University, New York, New York 10032
| |
Collapse
|
834
|
Ushijima T, Watanabe N, Okochi E, Kaneda A, Sugimura T, Miyamoto K. Fidelity of the methylation pattern and its variation in the genome. Genome Res 2003; 13:868-74. [PMID: 12727906 PMCID: PMC430912 DOI: 10.1101/gr.969603] [Citation(s) in RCA: 125] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2002] [Accepted: 02/26/2003] [Indexed: 12/31/2022]
Abstract
The methylated or unmethylated status of a CpG site is copied faithfully from parental DNA to daughter DNA, and functions as a cellular memory. However, no information is available for the fidelity of methylation pattern in unmethylated CpG islands (CGIs) or its variation in the genome. Here, we determined the methylation status of each CpG site on each DNA molecule obtained from clonal populations of normal human mammary epithelial cells. Methylation pattern error rates (MPERs) were calculated based upon the deviation from the methylation patterns that should be obtained if the cells had 100% fidelity in replicating the methylation pattern. Unmethylated CGIs in the promoter regions of five genes showed MPERs of 0.018-0.032 errors/site/21.6 generations, and the fidelity of methylation pattern was calculated as 99.85%-99.92%/site/generation. In contrast, unmethylated CGIs outside the promoter regions showed MPERs more than twice as high (P < 0.01). Methylated regions, including a CGI in the MAGE-A3 promoter and DMR of the H19 gene, showed much lower MPERs than unmethylated CGIs. These showed that errors in methylation pattern were mainly due to de novo methylations in unmethylated regions. The differential MPERs even among unmethylated CGIs indicated that a promoter-specific protection mechanism(s) from de novo methylation was present.
Collapse
Affiliation(s)
- Toshikazu Ushijima
- Carcinogenesis Division, National Cancer Center Research Institute, Chuo-ku, Tokyo 104-0045, Japan.
| | | | | | | | | | | |
Collapse
|
835
|
Hou P, Ji M, Liu Z, Shen J, Cheng LU, He N, Lu Z. A microarray to analyze methylation patterns of p16(Ink4a) gene 5'-CpG islands. Clin Biochem 2003; 36:197-202. [PMID: 12726928 DOI: 10.1016/s0009-9120(02)00445-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
OBJECTIVES Aberrant DNA methylation of the CpG site is among the earliest and most frequent alterations in cancer. Several studies suggest that aberrant methylation on the CpG sites of the tumor suppressor gene is closely associated with carcinogenesis. However, large-scale analysis of candidate genes has so far been hampered by the lack of high throughput approach for analyzing methylation patterns. The aim of this study was to develop a new method to analyze methylation patterns of p16(Ink4a) gene. DESIGN AND METHODS We selected a 336 bp segment of the 5' untranslated region and the first exon of the p16(Ink4a) gene, as the target sequence, which include the most densely packed CpG fragment of the islands containing 32 CpG sites. A set of oligonucleotide probes was designed to assemble a DNA microarray to discriminate the methylation patterns of several adjacent CpG sites. RESULTS Methylation patterns of human p16(Ink4a) gene were mapped and the results were validated by bisulphite DNA sequencing. A good reproducibility was observed in several parallel experiments. CONCLUSIONS The methylation oligonucleotide microarray can be applied as a useful and powerful tool to map methylation patterns in multiple CpG island sites.
Collapse
Affiliation(s)
- Peng Hou
- Department of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | | | | | | | | | | | | |
Collapse
|
836
|
Tost J, Schatz P, Schuster M, Berlin K, Gut IG. Analysis and accurate quantification of CpG methylation by MALDI mass spectrometry. Nucleic Acids Res 2003; 31:e50. [PMID: 12711695 PMCID: PMC154238 DOI: 10.1093/nar/gng050] [Citation(s) in RCA: 100] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2003] [Revised: 03/01/2003] [Accepted: 03/01/2003] [Indexed: 12/31/2022] Open
Abstract
As the DNA sequence of the human genome is now nearly finished, the main task of genome research is to elucidate gene function and regulation. DNA methylation is of particular importance for gene regulation and is strongly implicated in the development of cancer. Even minor changes in the degree of methylation can have severe consequences. An accurate quantification of the methylation status at any given position of the genome is a powerful diagnostic indicator. Here we present the first assay for the analysis and precise quantification of methylation on CpG positions in simplex and multiplex reactions based on matrix-assisted laser desorption/ ionisation mass spectrometry detection. Calibration curves for CpGs in two genes were established and an algorithm was developed to account for systematic fluctuations. Regression analysis gave R(2) >or= 0.99 and standard deviation around 2% for the different positions. The limit of detection was approximately 5% for the minor isomer. Calibrations showed no significant differences when carried out as simplex or multiplex analyses. All variable parameters were thoroughly investigated, several paraffin-embedded tissue biopsies were analysed and results were verified by established methods like analysis of cloned material. Mass spectrometric results were also compared to chip hybridisation.
Collapse
Affiliation(s)
- Jörg Tost
- Centre National de Génotypage, Bâtiment G2, 2 Rue Gaston Crémieux, CP 5721, 91057 Evry Cedex, France
| | | | | | | | | |
Collapse
|
837
|
Abstract
The past few years have seen an explosion of interest in the epigenetics of cancer. This has been a consequence of both the exciting coalescence of the chromatin and DNA methylation fields, and the realization that DNA methylation changes are involved in human malignancies. The ubiquity of DNA methylation changes has opened the way to a host of innovative diagnostic and therapeutic strategies. Recent advances attest to the great promise of DNA methylation markers as powerful future tools in the clinic.
Collapse
Affiliation(s)
- Peter W Laird
- University of Southern California, Norris Comprehensive Cancer Center, Room 6418, 1441 Eastlake Avenue, Los Angeles, California 90089-9176, USA.
| |
Collapse
|
838
|
|
839
|
Smiraglia DJ, Plass C. The development of CpG island methylation biomarkers using restriction landmark genomic scanning. Ann N Y Acad Sci 2003; 983:110-9. [PMID: 12724216 DOI: 10.1111/j.1749-6632.2003.tb05966.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
CpG island hypermethylation is a common occurrence in cancer. Because this is a stable molecular alteration of the DNA, which can be detected easily from very small amounts, DNA methylation is an attractive candidate to use as a molecular biomarker. Recent studies have used DNA methylation of genes known to be targets of genetic disruption in cancer as biomarkers for early detection of cancer, classification of malignancies, response to drug treatment, and as markers predictive of outcome. Since many of the currently used targets of methylation are methylated at rather low frequencies in various cancer types even though the gene may be frequently disrupted by other mechanisms, it would be useful to develop additional markers that are methylated at high frequency in the cancer being studied. Restriction landmark genomic scanning has been used for the identification of frequent targets of methylation in multiple malignancies. These markers, which can be either cancer type-specific or nonspecific, may prove to be effective biomarkers for diagnostic or prognostic purposes, or for midpoint analysis of intervention strategies.
Collapse
Affiliation(s)
- Dominic J Smiraglia
- Division of Human Cancer Genetics, Department of Molecular Virology, Immunology and Medical Genetics, The Ohio State University, Columbus, Ohio 43210, USA.
| | | |
Collapse
|
840
|
Danesi R, de Braud F, Fogli S, de Pas TM, Di Paolo A, Curigliano G, Del Tacca M. Pharmacogenetics of anticancer drug sensitivity in non-small cell lung cancer. Pharmacol Rev 2003; 55:57-103. [PMID: 12615954 DOI: 10.1124/pr.55.1.4] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
In mammalian cells, the process of malignant transformation is characterized by the loss or down-regulation of tumor-suppressor genes and/or the mutation or overexpression of proto-oncogenes, whose products promote dysregulated proliferation of cells and extend their life span. Deregulation in intracellular transduction pathways generates mitogenic signals that promote abnormal cell growth and the acquisition of an undifferentiated phenotype. Genetic abnormalities in cancer have been widely studied to identify those factors predictive of tumor progression, survival, and response to chemotherapeutic agents. Pharmacogenetics has been founded as a science to examine the genetic basis of interindividual variation in drug metabolism, drug targets, and transporters, which result in differences in the efficacy and safety of many therapeutic agents. The traditional pharmacogenetic approach relies on studying sequence variations in candidate genes suspected of affecting drug response. However, these studies have yielded contradictory results because of the small number of molecular determinants of drug response examined, and in several cases this approach was revealed to be reductionistic. This limitation is now being overcome by the use of novel techniques, i.e., high-density DNA and protein arrays, which allow genome- and proteome-wide tumor profiling. Pharmacogenomics represents the natural evolution of pharmacogenetics since it addresses, on a genome-wide basis, the effect of the sum of genetic variants on drug responses of individuals. Development of pharmacogenomics as a new field has accelerated the progress in drug discovery by the identification of novel therapeutic targets by expression profiling at the genomic or proteomic levels. In addition to this, pharmacogenetics and pharmacogenomics provide an important opportunity to select patients who may benefit from the administration of specific agents that best match the genetic profile of the disease, thus allowing maximum activity.
Collapse
Affiliation(s)
- Romano Danesi
- Division of Pharmacology and Chemotherapy, Department of Oncology, Transplants and Advanced Technologies in Medicine, University of Pisa, Pisa, Italy.
| | | | | | | | | | | | | |
Collapse
|
841
|
Abstract
Human cancers are caused by multiple mechanisms. Research in the last 30 years has firmly established the roles of a group of genes including oncogenes, tumor suppressor genes, and DNA repair genes in human cancers. The activation and inactivation of these cancer genes can be caused by genetic mutations or epigenetic alterations. The epigenetic changes in cancers include methylation of CpG islands, loss of imprinting, and chromatin modification. The completion of the genome sequences of many organisms including the human has transformed the traditional approach to molecular biology research into an era of functional genome research. Traditional research usually involves the study of one or a few genes (proteins) in a particular biological process in normal physiology or disease. Functional genome research takes advantage of newly available genome sequences and high-throughput genome technologies to study genes and/or proteins to inform the perspective of entire biological processes. I will focus on recent progress in the identification of imprinted genes and methylation of CpG islands through genome-wide analysis.
Collapse
Affiliation(s)
- Maxwell P Lee
- Laboratory of Population Genetics, National Cancer Institute, 41 Library Drive D702C, Bethesda, Maryland 20892, USA.
| |
Collapse
|
842
|
Abstract
Recent advances in human genome research have resulted in novel approaches for the identification of epigenetic modifications associated with cancer. Modulators of DNA methylation and chromatin structure have a dramatic effect on gene expression, cellular proliferation, differentiation, and apoptosis. Molecular pathways regulating epigenetic events that occur during tumorigenesis have been exploited as new targets for therapeutic intervention. Clinical studies exploring the effectiveness of therapeutic agents targeting DNA methylation and acetylation of histones have yielded promising results. Molecular profiles of epigenetic alterations in cancer cells could allow better stratification of patients who may show responsiveness to specific treatments.
Collapse
Affiliation(s)
- Thea Kalebic
- Lung and Upper Aerodigestive Cancer Research Group, Division of Cancer Prevention, National Cancer Institute, Bethesda, Maryland 20892, USA.
| |
Collapse
|
843
|
Abstract
Hypomethylation signifies one end of a spectrum of DNA methylation states. In most cases hypomethylation refers to a relative state that represents a change from the "normal" methylation level. Hypomethylation, when approached from a topographical perspective, has been used to describe either overall decreases in the methylation status of the entire genome (global hypomethylation) or more localized relative demethylation of specific subsets of the genome, such as the promoter regions of protooncogenes or normally highly methylated repetitive sequences. Global hypomethylation accompanied by gene-specific hypermethylation is observed in at least two important settings: cancer and aging. Global hypomethylation is generally reflective of decreased methylation in CpGs dispersed throughout repetitive sequences as well as the bodies of genes. Hypomethylation of repetitive and parasitic DNA sequences correlates with a number of adverse outcomes. For example, decreased methylation of repetitive sequences in the satellite DNA of the pericentric region of chromosomes is associated with increased chromosomal rearrangements, a hallmark of cancer. Decreased methylation of proviral sequences can lead to reactivation and increased infectivity. However, hypomethylation in cancer can also affect the CpGs in the promoters of specific genes-namely, protooncogenes-leading to their overexpression and resulting in the functional outcome of increased cell proliferation. Thus, hypomethylation, in a variety of settings in which it represents a deviation from "normal," appears to correlate with progression to cancer and offers potential mechanisms to explain the carcinogenic process.
Collapse
Affiliation(s)
- Barbara K Dunn
- Basic Prevention Sciences Research Group, Division of Cancer Prevention, National Cancer Institute, Bethesda, Maryland 20892, USA.
| |
Collapse
|
844
|
Jubb AM, Quirke P, Oates AJ. DNA methylation, a biomarker for colorectal cancer: implications for screening and pathological utility. Ann N Y Acad Sci 2003; 983:251-67. [PMID: 12724230 DOI: 10.1111/j.1749-6632.2003.tb05980.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Currently up to one-third of colorectal cancer patients present with locally advanced or metastatic disease that precludes a surgical cure. Performance limitations and low uptake of current screening tools have fueled research to develop minimally invasive approaches that can detect early-stage neoplasms. The observation that altered DNA can be amplified from the stool or circulation has stimulated research on its use as a biomarker of occult neoplasia. De novo methylation of CpG islands 5' to certain tumor suppressor genes has been associated with epigenetic silencing. At certain loci this phenomenon is specific for neoplastic populations, and it is frequently detected at early stages in colorectal tumorigenesis. Accordingly, hypermethylation events have been proposed by researchers as ideal targets for the basis of a screening panel to detect peripheral tumor DNA. This critique reviews research findings on the use of epigenetic biomarkers in screening for occult neoplasia. In addition, the authors consider the pathological utility of epigenetic testing in refining tumor staging and predicting disease recurrence.
Collapse
Affiliation(s)
- Adrian M Jubb
- Academic Unit of Pathology, Leeds University, Leeds, UK.
| | | | | |
Collapse
|
845
|
Abstract
In recent years, many molecular biomarkers have been discovered that are capable of distinguishing tumors from normal tissue. Among the different types of markers, DNA methylation markers stand out for their potential to provide a unique combination of specificity, sensitivity, high information content, and applicability to a wide variety of clinical specimens. Methylation markers are particularly suited for situations where sensitive detection is necessary, such as when tumor DNA is either scarce or diluted by excess normal DNA. One of the most widely used methods for measuring methylation levels, methylation-specific PCR (MSP), has been proved to be a very effective tool in situations requiring sensitive detection. The addition of fluorogenic probes makes these assays more informative, quantitative, and suitable for a clinical format. The field of sensitive detection is not limited to MSP; hence, an alternative methylation-sensitive amplification is discussed. PCR-based methylation assays have been applied to the detection of tumor DNA in a variety of body fluids, including serum, plasma, urine, sputum, and lavage fluids. In many cases, the sensitivity and specificity of these detection assays has been impressive, but important technological issues remain in areas such as sample preparation, assay design, and marker selection. Once these technical concerns have been addressed, the sensitive detection of methylation will provide a powerful diagnostic and prognostic tool, especially for the early detection of preneoplastic and neoplastic lesions.
Collapse
|
846
|
Abstract
Hypermethylation of CpG islands, an epigenetic event that is not accompanied by changes in DNA sequence, represents an alternative mechanism to deletions or mutations to inactivate tumor suppressor genes. Recent evidence supports the notion that CpG island hypermethylation, by silencing key cancer-related genes, plays a major causal role in cancer. However, a long-standing issue in the field is the sequence of molecular events leading to epigenetic gene silencing. A new model has been proposed that chromatin remodeling, as a result of histone deacetylation and methylation, is the primary event in abrogating transcriptional initiation; subsequently, CpG island hypermethylation establishes a permanent state of gene silencing. Accumulating evidence indicates that CpG island hypermethylation is an early event in cancer development and, in some cases, may precede the neoplastic process. Because of their heritable nature, hypermethylated CpG islands leave 'molecular footprints' in evolving cancer cells and can be used as molecular markers to reconstruct epigenetic progression during tumorigenesis. Furthermore, hypermethylated CpG islands are proving to be useful for molecular classification of different cancer types.
Collapse
Affiliation(s)
- Kenneth P Nephew
- Medical Sciences, School of Medicine, Indiana University, Bloomington, IN 47405-4401, USA
| | | |
Collapse
|
847
|
Zvara A, Hackler L, Nagy ZB, Micsik T, Puskás LG. New molecular methods for classification, diagnosis and therapy prediction of hematological malignancies. Pathol Oncol Res 2003; 8:231-240. [PMID: 12579208 DOI: 10.1007/bf03036737] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2002] [Accepted: 12/15/2002] [Indexed: 10/20/2022]
Abstract
Normal functions of the cell are based on the precise regulation of various genes. If this strict regulation and the hierarchy of genes becomes upset due to flaws in this system, the result will be cellular dysfunction which eventually may lead to carcinogenic transformation. Two basic challenges of the classification of cancers are the discovery of new molecular markers characteristic to defined disease groups and the classification of already diagnosed or new cases into existing groups. This precise classification may open the door to tailored treatment or project the expected outcome of the disease. Today there is unlimited access available to the databases containing sequences and localization of the genes within the confines of Human Genome project. It provides significant help for the discovery of chromosome abnormalities and systematic analysis of gene expression patterns. This is important not only to understand normal functions of the cells, but it also contributes to the identification of new genes that are characteristic to given disease groups as markers and that are potential drug targets. Until the second half of the twentieth century the study of the function and regulation of genes was based on step-by-step investigation of individual genes. Regarding the fact, that the genomes of an increasing number of organisms have become known in whole or in part, numerous new techniques have been developed that facilitated the systematic analysis of gene functions. The aim of this study is to summarize the new, molecular based possibilities for classification, diagnosis and prognosis of hematological malignancies, as well as to summarize the main results of these areas.
Collapse
Affiliation(s)
- Agnes Zvara
- Biological Research Center, Hungarian Academy of Sciences, Szeged, H-6726, Hungary
| | | | | | | | | |
Collapse
|
848
|
Davis AJ, Gelmon KA, Siu LL, Moore MJ, Britten CD, Mistry N, Klamut H, D'Aloisio S, MacLean M, Wainman N, Ayers D, Firby P, Besterman JM, Reid GK, Eisenhauer EA. Phase I and pharmacologic study of the human DNA methyltransferase antisense oligodeoxynucleotide MG98 given as a 21-day continuous infusion every 4 weeks. Invest New Drugs 2003; 21:85-97. [PMID: 12795533 DOI: 10.1023/a:1022976528441] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
PURPOSE MG98 is a second generation phosphorothioate antisense oligodeoxynucleotide which is a highly specific inhibitor of translation of the mRNA for human DNA MeTase I (DNMT 1). This phase I study examined the toxicity and pharmacologic profile of MG98 administered as a continuous 21-day intravenous infusion every 4 weeks. PATIENTS AND METHODS Fourteen patients with solid cancers received a total of 25 cycles of MG98 at doses ranging from 40 to 240 mg/m2/day. Steady-state concentrations of MG98 were measured as were several pharmacodynamic assessments including mRNA of the target gene, DNMT1, in PBMC. In addition, other potential surrogate markers of drug effects were explored, including hemoglobin F, Vimentin and GADD45. RESULTS Dose limiting effects were drug-related reversible transaminase elevation and fatigue seen at doses of 240, 200 and 160 mg/m2/day. The dose level of 80 mg/m2/day was felt to be safe and tolerable when delivered on this schedule. No evidence of antitumor activity was observed. Although pharmacokinetic analysis revealed that at the higher dose levels, mean Css values of MG98 were approximately 10-fold times the IC50 values associated with target inhibition in vitro, the extent of MG98 penetration into target tumors in this trial was not determined. No consistent, dose-related changes in correlative markers including DNMT1 mRNA, hemoglobin F, Vimentin and GADD45, were observed. CONCLUSIONS This schedule of MG98 given as a 21-day continuous intravenous infusion every 4 weeks was poorly tolerated in the highest doses; therefore, further disease-site specific evaluation of the efficacy of this agent will utilize a more favorable, intermittent dosing schedule. Pharmacodynamic evaluations undertaken in an attempt to explore and validate the biological mechanisms of MG98 did not show dose-related effects.
Collapse
|
849
|
Yuan BZ, Durkin ME, Popescu NC. Promoter hypermethylation of DLC-1, a candidate tumor suppressor gene, in several common human cancers. CANCER GENETICS AND CYTOGENETICS 2003; 140:113-7. [PMID: 12645648 DOI: 10.1016/s0165-4608(02)00674-x] [Citation(s) in RCA: 77] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Aberrant methylation of CpG islands within the promoter regions of tumor suppressor or cancer-related genes is a common mechanism leading to the silencing of gene expression. To determine whether aberrant methylation is a contributing factor to transcriptional inactivation of DLC-1 (deleted in liver cancer-1), a candidate tumor suppressor gene, we examined its methylation status in twelve hepatocellular carcinoma. breast, colon, and prostate tumor cell lines with low or undetectable expression of DLC-1. By Southern blot analysis of DNA digested with the methylation sensitive enzyme HpaII, we found a different degree of promoter hypermethylation in all cell lines with aberrant DLC-1 expression. The hypermethylation status was reversed by the addition of 5-aza-2'-deoxycytidine, a demethylating agent, in one human hepatocellular carcinoma line. These observations suggest that hypermethylation is responsible for abrogating the function of the DLC-1 gene in a subset of liver, breast, colon, and prostate cancers.
Collapse
MESH Headings
- Adenocarcinoma/genetics
- Adenocarcinoma/pathology
- Azacitidine/analogs & derivatives
- Azacitidine/pharmacology
- Base Sequence
- Breast Neoplasms/genetics
- Breast Neoplasms/pathology
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/pathology
- Colonic Neoplasms/genetics
- Colonic Neoplasms/pathology
- CpG Islands
- DNA Methylation/drug effects
- DNA, Neoplasm/chemistry
- DNA, Neoplasm/genetics
- Decitabine
- Female
- GTPase-Activating Proteins
- Gene Expression Regulation, Neoplastic/drug effects
- Genes, Tumor Suppressor
- Humans
- Liver Neoplasms/genetics
- Liver Neoplasms/pathology
- Male
- Molecular Sequence Data
- Neoplasm Proteins/deficiency
- Neoplasm Proteins/genetics
- Neoplasms/genetics
- Promoter Regions, Genetic/drug effects
- Promoter Regions, Genetic/genetics
- Prostatic Neoplasms/genetics
- Prostatic Neoplasms/pathology
- Tumor Cells, Cultured/chemistry
- Tumor Cells, Cultured/pathology
- Tumor Suppressor Proteins/deficiency
- Tumor Suppressor Proteins/genetics
Collapse
Affiliation(s)
- Bao-Zhu Yuan
- Health Effects Laboratory Division, Toxicology and Molecular Biology Branch, National Institute for Occupational Safely and Health, Morgantown, WV 26505, USA
| | | | | |
Collapse
|
850
|
Stach D, Schmitz OJ, Stilgenbauer S, Benner A, Döhner H, Wiessler M, Lyko F. Capillary electrophoretic analysis of genomic DNA methylation levels. Nucleic Acids Res 2003; 31:E2. [PMID: 12527791 PMCID: PMC140527 DOI: 10.1093/nar/gng002] [Citation(s) in RCA: 102] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Changes in DNA methylation have been found in the large majority of tumors. This phenomenon includes both genome-wide hypomethylation and gene- specific hypermethylation. However, the clinical relevance of either mechanism has remained contentious. In order to determine DNA methylation levels from a large number of clinical samples, we have established a method for accurate high-throughput quantification of 5-methylcytosine in genomic DNA. Our protocol requires a small amount (<1 micro g) of DNA that is enzymatically hydrolyzed to single nucleotides. Single nucleotides are then derivatized with a fluorescent marker and separated by capillary electrophoresis. After calibration of the method, we have determined cytosine methylation levels from tumor samples of 81 patients that had been diagnosed with chronic lymphocytic leukemia (CLL). These patients showed a high variability in their methylation levels with a general trend towards hypomethylation. Because of its high accuracy and throughput our method will be useful in determining the role of genomic DNA methylation levels in tumorigenesis.
Collapse
Affiliation(s)
- Dirk Stach
- Division of Molecular Toxicology, Deutsches Krebsforschungszentrum, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | | | | | | | | | | | | |
Collapse
|