47 results on '"Estecio MR"'
Search Results
2. CCAT2, a novel noncoding RNA mapping to 8q24, underlies metastatic progression and chromosomal instability in colon cancer
- Author
-
Ling, H, Spizzo, R, Atlasi, Yaser, Nicoloso, M, Shinnizu, M, Redis, RS, Nishida, N, Gafa, R, Song, J, Guo, ZY, Ivan, C, Barbarotto, E, de Vries, I, Zhang, XN, Ferracin, M, Churchman, M, Galen, Janneke, Beverloo, BH, Shariati, M, Haderk, F, Estecio, MR, Garcia-Manero, G, Patijn, GA, Gotley, DC, Bhardwaj, V, Shureiqi, I, Sen, S, Multani, AS, Welsh, J, Yamamoto, K, Taniguchi, I, Song, MA, Gallinger, S, Casey, G, Thibodeau, SN, Le Marchand, L, Tiirikainen, M, Mani, SA, Zhang, W, Davuluri, RV, Mimori, K, Mori, M, Sieuwerts, Anieta, Martens, John, Tomlinson, I, Negrini, M, Berindan-Neagoe, I, Foekens, John, Hamilton, SR, Lanza, G, Kopetz, S, Fodde, Riccardo, Calin, GA, Ling, H, Spizzo, R, Atlasi, Yaser, Nicoloso, M, Shinnizu, M, Redis, RS, Nishida, N, Gafa, R, Song, J, Guo, ZY, Ivan, C, Barbarotto, E, de Vries, I, Zhang, XN, Ferracin, M, Churchman, M, Galen, Janneke, Beverloo, BH, Shariati, M, Haderk, F, Estecio, MR, Garcia-Manero, G, Patijn, GA, Gotley, DC, Bhardwaj, V, Shureiqi, I, Sen, S, Multani, AS, Welsh, J, Yamamoto, K, Taniguchi, I, Song, MA, Gallinger, S, Casey, G, Thibodeau, SN, Le Marchand, L, Tiirikainen, M, Mani, SA, Zhang, W, Davuluri, RV, Mimori, K, Mori, M, Sieuwerts, Anieta, Martens, John, Tomlinson, I, Negrini, M, Berindan-Neagoe, I, Foekens, John, Hamilton, SR, Lanza, G, Kopetz, S, Fodde, Riccardo, and Calin, GA
- Abstract
The functional roles of SNPs within the 8q24 gene desert in the cancer phenotype are not yet well understood. Here, we report that CCAT2, a novel long noncoding RNA transcript (IncRNA) encompassing the rs6983267 SNP, is highly over-expressed in microsatellite-stable colorectal cancer and promotes tumor growth, metastasis, and chromosomal instability. We demonstrate that MY, miR-17-5p, and miR-20a are up-regulated by CCAT2 through TCF7L2-mediated transcriptional regulation. We further identify the physical interaction between CC4T2 and TCF7L2 resulting in an enhancement of WNT signaling activity. We show that CCAT2 is itself a WNT downstream target, which suggests the existence of a feedback loop. Finally, we demonstrate that the SNP status affects CC4T2 expression and the risk allele G produces more CCAT2 transcript. Our results support a new mechanism of MYC and WNT regulation by the novel IncRNA CCAT2 in colorectal cancer pathogenesis, and provide an alternative explanation of the SNP-conferred cancer risk.
- Published
- 2013
3. Immune evolution from preneoplasia to invasive lung adenocarcinomas and underlying molecular features
- Author
-
Dejima, H, primary, Hu, X, additional, Chen, R, additional, Zhang, J, additional, Fujimoto, J, additional, Parra, ER, additional, Haymaker, C, additional, Hubert, SM, additional, Douse, D, additional, Solis, LM, additional, Su, D, additional, Fukuoda, J, additional, Tabata, K, additional, Pham, HHN, additional, Mcgranahan, N, additional, Zhang, B, additional, Ye, J, additional, Ying, L, additional, Little, L, additional, Gumbs, C, additional, Chow, C, additional, Estecio, MR, additional, Godoy, MCB, additional, Antonoff, MB, additional, Sepesi, B, additional, Pass, HI, additional, Behrens, C, additional, Vaporciyan, AA, additional, Heymach, JV, additional, Scheet, P, additional, Lee, JJ, additional, Wu, J, additional, Futreal, PA, additional, Reuben, A, additional, Kadara, H, additional, and Wistuba, II, additional
- Full Text
- View/download PDF
4. Genetic variation drives cancer cell adaptation to ECM stiffness.
- Author
-
Wang TC, Sawhney S, Morgan D, Bennett RL, Rashmi R, Estecio MR, Brock A, Singh I, Baer CF, Licht JD, and Lele TP
- Subjects
- Humans, Cell Line, Tumor, Neoplasms genetics, Neoplasms pathology, Adaptation, Physiological genetics, Cell Proliferation genetics, Cell Movement genetics, Extracellular Matrix metabolism, Extracellular Matrix genetics, Genetic Variation
- Abstract
The progression of many solid tumors is accompanied by temporal and spatial changes in the stiffness of the extracellular matrix (ECM). Cancer cells adapt to soft and stiff ECM through mechanisms that are not fully understood. It is well known that there is significant genetic heterogeneity from cell to cell in tumors, but how ECM stiffness as a parameter might interact with that genetic variation is not known. Here, we employed experimental evolution to study the response of genetically variable and clonal populations of tumor cells to variable ECM stiffness. Proliferation rates of genetically variable populations cultured on soft ECM increased over a period of several weeks, whereas clonal populations did not evolve. Tracking of DNA barcoded cell lineages revealed that soft ECM consistently selected for the same few variants. These data provide evidence that ECM stiffness exerts natural selection on genetically variable tumor populations. Soft-selected cells were highly migratory, with enriched oncogenic signatures and unusual behaviors such as spreading and traction force generation on ECMs with stiffness as low as 1 kPa. Rho-regulated cell spreading was found to be the directly selected trait, with yes-associated protein 1 translocation to the nucleus mediating fitness on soft ECM. Overall, these data show that genetic variation can drive cancer cell adaptation to ECM stiffness., Competing Interests: Competing interests statement:J.D.L. serves as a consultant for AstraZeneca.
- Published
- 2024
- Full Text
- View/download PDF
5. Tumor- and circulating-free DNA methylation identifies clinically relevant small cell lung cancer subtypes.
- Author
-
Heeke S, Gay CM, Estecio MR, Tran H, Morris BB, Zhang B, Tang X, Raso MG, Rocha P, Lai S, Arriola E, Hofman P, Hofman V, Kopparapu P, Lovly CM, Concannon K, De Sousa LG, Lewis WE, Kondo K, Hu X, Tanimoto A, Vokes NI, Nilsson MB, Stewart A, Jansen M, Horváth I, Gaga M, Panagoulias V, Raviv Y, Frumkin D, Wasserstrom A, Shuali A, Schnabel CA, Xi Y, Diao L, Wang Q, Zhang J, Van Loo P, Wang J, Wistuba II, Byers LA, and Heymach JV
- Subjects
- Humans, DNA Methylation, Epigenesis, Genetic, Biomarkers, Tumor genetics, Small Cell Lung Carcinoma genetics, Small Cell Lung Carcinoma pathology, Lung Neoplasms genetics, Lung Neoplasms pathology, Cell-Free Nucleic Acids genetics
- Abstract
Small cell lung cancer (SCLC) is an aggressive malignancy composed of distinct transcriptional subtypes, but implementing subtyping in the clinic has remained challenging, particularly due to limited tissue availability. Given the known epigenetic regulation of critical SCLC transcriptional programs, we hypothesized that subtype-specific patterns of DNA methylation could be detected in tumor or blood from SCLC patients. Using genomic-wide reduced-representation bisulfite sequencing (RRBS) in two cohorts totaling 179 SCLC patients and using machine learning approaches, we report a highly accurate DNA methylation-based classifier (SCLC-DMC) that can distinguish SCLC subtypes. We further adjust the classifier for circulating-free DNA (cfDNA) to subtype SCLC from plasma. Using the cfDNA classifier (cfDMC), we demonstrate that SCLC phenotypes can evolve during disease progression, highlighting the need for longitudinal tracking of SCLC during clinical treatment. These data establish that tumor and cfDNA methylation can be used to identify SCLC subtypes and might guide precision SCLC therapy., Competing Interests: Declaration of interests S.H., C.M.G., L.A.B., and J.V.H. own intellectual property on the classification of SCLC from DNA methylation and gene expression. D.F., A.W., A.S., and C.A.S. are full time employees of Nucleix and own stocks and stock options of Nucleix. Furthermore, S.H. reports consulting fees from Guardant Health, AstraZeneca, Boehringer Ingelheim, and Qiagen. C.M.G. is a member of the advisory board at Jazz Pharmaceuticals, AstraZeneca, and Bristol Myers Squibb and served as speaker for AstraZeneca and BeiGene. P.R. received travel support from AstraZeneca, BMS, and MSD. E.A. reports consulting fees from Eli Lilly, AstraZeneca, BMS, Boehringer Ingelheim, Takeda, Roche, and MSD, speaker’s fees from AstraZeneca, BMS, Boehringer Ingelheim, Roche, and MSD, research funding from Roche and AstraZeneca and travel support from AstraZeneca and Takeda. P.H. reports research grants from Thermo Fisher Scientific and Biocartis, and speakers’ fees from AstraZeneca, Roche, Novartis, Bristol-Myers Squibb, Pfizer, Bayer, Illumina, Biocartis, Thermo Fisher Scientific, AbbVie, Amgen, Janssen, Eli Lilly, Daiichi Sankyo, Pierre Fabre, and Guardant. V.H. reports speakers’ fees from BMS. C.M.L. reports personal fees from Amgen, Arrivent, AstraZeneca, Blueprints Medicine, Cepheid, D2G Oncology, Daiichi Sankyo, Eli Lilly, EMD Serono, Foundation Medicine, Genentech, Janssen, Medscape, Novartis, Pfizer, Puma, Syros, and Takeda. N.V. receives consulting fees from Sanofi, Regeneron, Oncocyte, and Eli Lilly, and research funding from Mirati. M.B.N. receives royalties and licensing fees from Spectrum Pharmaceuticals. I.H. received personal as well as institutional funding from Nucleix. J.Z. served on advisory board for AstraZeneca and Geneplus and received speaker’s fees from BMS, Geneplus, OrigMed, Innovent and grants from Merck, Johnson and Johnson. L.A.B received consulting fees and research funding from AstraZeneca, GenMab, Sierra Oncology, research funding from ToleroPharmaceuticals and served as advisor or consultant for PharmaMar, AbbVie, Bristol-Myers Squibb, Alethia, Merck, Pfizer, Jazz Pharmaceuticals, Genentech, and Debiopharm Group. J.V.H. served as advisor for AstraZeneca, EMD Serono, Boehringer-Ingelheim, Catalyst, Genentech, GlaxoSmithKline, Guardant Health, Foundation medicine, Hengrui Therapeutics, Eli Lilly, Novartis, Spectrum, Sanofi, Takeda, Mirati Therapeutics, BMS, BrightPath Biotherapeutics, Janssen Global Services, Nexus Health Systems, Pneuma Respiratory, Kairos Venture Investments, Roche, Leads Biolabs, RefleXion, Chugai Pharmaceuticals, received research support from AstraZeneca, GlaxoSmithKline, Spectrum as well as royalties and licensing fees from Spectrum., (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
6. miR-543 regulates the epigenetic landscape of myelofibrosis by targeting TET1 and TET2.
- Author
-
Fuentes-Mattei E, Bayraktar R, Manshouri T, Silva AM, Ivan C, Gulei D, Fabris L, Soares do Amaral N, Mur P, Perez C, Torres-Claudio E, Dragomir MP, Badillo-Perez A, Knutsen E, Narayanan P, Golfman L, Shimizu M, Zhang X, Zhao W, Ho WT, Estecio MR, Bartholomeusz G, Tomuleasa C, Berindan-Neagoe I, Zweidler-McKay PA, Estrov Z, Zhao ZJ, Verstovsek S, Calin GA, and Redis RS
- Published
- 2024
- Full Text
- View/download PDF
7. Cigarette smoke exposure accelerates AML progression in FLT3-ITD models.
- Author
-
Figueroa M, Ma H, Alfayez M, Morales-Mantilla DE, Wang F, Lu Y, Estecio MR, King KY, Kleinerman E, Moghaddam SJ, Daver N, Andreeff M, Konopleva M, DiNardo C, and Chandra J
- Subjects
- Humans, Mutation, fms-Like Tyrosine Kinase 3 genetics, Cigarette Smoking adverse effects, Leukemia, Myeloid, Acute etiology
- Published
- 2023
- Full Text
- View/download PDF
8. GSK-3484862 targets DNMT1 for degradation in cells.
- Author
-
Chen Q, Liu B, Zeng Y, Hwang JW, Dai N, Corrêa IR Jr, Estecio MR, Zhang X, Santos MA, Chen T, and Cheng X
- Abstract
Maintenance of genomic methylation patterns at DNA replication forks by DNMT1 is the key to faithful mitotic inheritance. DNMT1 is often overexpressed in cancer cells and the DNA hypomethylating agents azacytidine and decitabine are currently used in the treatment of hematologic malignancies. However, the toxicity of these cytidine analogs and their ineffectiveness in treating solid tumors have limited wider clinical use. GSK-3484862 is a newly-developed, dicyanopyridine containing, non-nucleoside DNMT1-selective inhibitor with low cellular toxicity. Here, we show that GSK-3484862 targets DNMT1 for protein degradation in both cancer cell lines and murine embryonic stem cells (mESCs). DNMT1 depletion was rapid, taking effect within hours following GSK-3484862 treatment, leading to global hypomethylation. Inhibitor-induced DNMT1 degradation was proteasome-dependent, with no discernible loss of DNMT1 mRNA. In mESCs, GSK-3484862-induced Dnmt1 degradation requires the Dnmt1 accessory factor Uhrf1 and its E3 ubiquitin ligase activity. We also show that Dnmt1 depletion and DNA hypomethylation induced by the compound are reversible after its removal. Together, these results indicate that this DNMT1-selective degrader/inhibitor will be a valuable tool for dissecting coordinated events linking DNA methylation to gene expression and identifying downstream effectors that ultimately regulate cellular response to altered DNA methylation patterns in a tissue/cell-specific manner., (© The Author(s) 2023. Published by Oxford University Press on behalf of NAR Cancer.)
- Published
- 2023
- Full Text
- View/download PDF
9. Genotype-to-Phenotype Associations in the Aggressive Variant Prostate Cancer Molecular Profile (AVPC-m) Components.
- Author
-
Soundararajan R, Viscuse P, Pilie P, Liu J, Logotheti S, Laberiano Fernández C, Lorenzini D, Hoang A, Lu W, Soto LMS, Wistuba II, Xu M, Song X, Shepherd PDA, Navone NM, Tidwell RSS, Lozano G, Logothetis C, Zhang J, Long JP, Estecio MR, Tzelepi V, and Aparicio AM
- Abstract
The aggressive variant prostate cancer molecular profile (AVPC-m), composed of combined defects in TP53, RB1 and PTEN, characterizes a subset of prostate cancers linked to androgen indifference and platinum sensitivity. To contribute to the optimization of the AVPC-m assessment for inclusion in prospective clinical trials, we investigated the status of the AVPC-m components in 28 patient tumor-derived xenografts (PDXs) developed at MDACC. We subjected single formalin-fixed, paraffin-embedded (FFPE) blocks from each PDX to immunohistochemistry (IHC), targeted next-generation genomic sequencing (NGS) and Clariom-S Affymetrix human microarray expression profiling. Standard validated IHC assays and a 10% labeling index cutoff resulted in high reproducibility across three separate laboratories and three independent readers for all tumor suppressors, as well as strong correlations with loss-of-function transcriptional scores (LOF-TS). Adding intensity assessment to labeling indices strengthened the association between IHC results and LOF-TS for TP53 and RB1, but not for PTEN. For TP53, genomic alterations determined by NGS had slightly higher agreement scores with LOF-TS than aberrant IHC, while for RB1 and PTEN, NGS and IHC determinations resulted in similar agreement scores with LOF-TS. Nonetheless, our results indicate that the AVPC-m components can be assessed reproducibly by IHC using various widely available standardized assays.
- Published
- 2022
- Full Text
- View/download PDF
10. Stem Cell Theory of Cancer: Rude Awakening or Bad Dream from Cancer Dormancy?
- Author
-
Tu SM, Estecio MR, Lin SH, and Zacharias NM
- Abstract
To be dormant or not depends on the origin and nature of both the cell and its niche. Similar to other cancer hallmarks, dormancy is ingrained with stemness, and stemness is embedded within dormancy. After all, cancer dormancy is dependent on multiple factors such as cell cycle arrest, metabolic inactivity, and the microenvironment. It is the net results and sum effects of a myriad of cellular interactions, interconnections, and interplays. When we unite all cancer networks and integrate all cancer hallmarks, we practice and preach a unified theory of cancer. From this perspective, we review cancer dormancy in the context of a stem cell theory of cancer. We revisit the seed and soil hypothesis of cancer. We reexamine its implications in both primary tumors and metastatic lesions. We reassess its roles in cell cycle arrest, metabolic inactivity, and stemness property. Cancer dormancy is particularly revealing when it informs us about the mysteries of late relapse, prolonged remission, and second malignancy. It is paradoxically rewarding when it delivers us the promises and power of cancer prevention and maintenance therapy in patient care.
- Published
- 2022
- Full Text
- View/download PDF
11. Myotonic dystrophy type 1 (DM1) clinical subtypes and CTCF site methylation status flanking the CTG expansion are mutant allele length-dependent.
- Author
-
Morales F, Corrales E, Zhang B, Vásquez M, Santamaría-Ulloa C, Quesada H, Sirito M, Estecio MR, Monckton DG, and Krahe R
- Subjects
- Alleles, CCCTC-Binding Factor, DNA Methylation genetics, Humans, Myotonin-Protein Kinase genetics, Trinucleotide Repeat Expansion genetics, Myotonic Dystrophy genetics
- Abstract
Myotonic dystrophy type 1 (DM1) is a complex disease with a wide spectrum of symptoms. The exact relationship between mutant CTG repeat expansion size and clinical outcome remains unclear. DM1 congenital patients (CDM) inherit the largest expanded alleles, which are associated with abnormal and increased DNA methylation flanking the CTG repeat. However, DNA methylation at the DMPK locus remains understudied. Its relationship to DM1 clinical subtypes, expansion size and age-at-onset is not yet completely understood. Using pyrosequencing-based methylation analysis on 225 blood DNA samples from Costa Rican DM1 patients, we determined that the size of the estimated progenitor allele length (ePAL) is not only a good discriminator between CDM and non-CDM cases (with an estimated threshold at 653 CTG repeats), but also for all DM1 clinical subtypes. Secondly, increased methylation at both CTCF sites upstream and downstream of the expansion was almost exclusively present in CDM cases. Thirdly, levels of abnormal methylation were associated with clinical subtype, age and ePAL, with strong correlations between these variables. Fourthly, both ePAL and the intergenerational expansion size were significantly associated with methylation status. Finally, methylation status was associated with ePAL and maternal inheritance, with almost exclusively maternal transmission of CDM. In conclusion, increased DNA methylation at the CTCF sites flanking the DM1 expansion could be linked to ePAL, and both increased methylation and the ePAL could be considered biomarkers for the CDM phenotype., (© The Author(s) 2021. Published by Oxford University Press. All rights reserved. For Permissions, please email: journals.permissions@oup.com.)
- Published
- 2021
- Full Text
- View/download PDF
12. Immune evolution from preneoplasia to invasive lung adenocarcinomas and underlying molecular features.
- Author
-
Dejima H, Hu X, Chen R, Zhang J, Fujimoto J, Parra ER, Haymaker C, Hubert SM, Duose D, Solis LM, Su D, Fukuoka J, Tabata K, Pham HHN, Mcgranahan N, Zhang B, Ye J, Ying L, Little L, Gumbs C, Chow CW, Estecio MR, Godoy MCB, Antonoff MB, Sepesi B, Pass HI, Behrens C, Zhang J, Vaporciyan AA, Heymach JV, Scheet P, Lee JJ, Wu J, Futreal PA, Reuben A, Kadara H, Wistuba II, and Zhang J
- Subjects
- Adenocarcinoma in Situ immunology, Adenocarcinoma in Situ pathology, Adenocarcinoma of Lung immunology, Adenocarcinoma of Lung pathology, Carcinogenesis immunology, Carcinogenesis pathology, Chromosome Aberrations, Clone Cells, DNA Copy Number Variations, DNA Methylation, Gene Expression Profiling, Gene Expression Regulation, Neoplastic, Humans, Immunity, Innate, Lung immunology, Lung metabolism, Lung pathology, Lung Neoplasms immunology, Lung Neoplasms pathology, Neoplasm Proteins immunology, Precancerous Conditions immunology, Precancerous Conditions pathology, Signal Transduction, T-Lymphocytes, Cytotoxic immunology, T-Lymphocytes, Cytotoxic pathology, T-Lymphocytes, Helper-Inducer immunology, T-Lymphocytes, Helper-Inducer pathology, T-Lymphocytes, Regulatory immunology, T-Lymphocytes, Regulatory pathology, Tumor Escape genetics, Tumor Escape immunology, Tumor Microenvironment genetics, Tumor Microenvironment immunology, Adenocarcinoma in Situ genetics, Adenocarcinoma of Lung genetics, Carcinogenesis genetics, Lung Neoplasms genetics, Neoplasm Proteins genetics, Precancerous Conditions genetics, Transcriptome
- Abstract
The mechanism by which anti-cancer immunity shapes early carcinogenesis of lung adenocarcinoma (ADC) is unknown. In this study, we characterize the immune contexture of invasive lung ADC and its precursors by transcriptomic immune profiling, T cell receptor (TCR) sequencing and multiplex immunofluorescence (mIF). Our results demonstrate that anti-tumor immunity evolved as a continuum from lung preneoplasia, to preinvasive ADC, minimally-invasive ADC and frankly invasive lung ADC with a gradually less effective and more intensively regulated immune response including down-regulation of immune-activation pathways, up-regulation of immunosuppressive pathways, lower infiltration of cytotoxic T cells (CTLs) and anti-tumor helper T cells (Th), higher infiltration of regulatory T cells (Tregs), decreased T cell clonality, and lower frequencies of top T cell clones in later-stages. Driver mutations, chromosomal copy number aberrations (CNAs) and aberrant DNA methylation may collectively impinge host immune responses and facilitate immune evasion, promoting the outgrowth of fit subclones in preneoplasia into dominant clones in invasive ADC.
- Published
- 2021
- Full Text
- View/download PDF
13. Evolution of DNA methylome from precancerous lesions to invasive lung adenocarcinomas.
- Author
-
Hu X, Estecio MR, Chen R, Reuben A, Wang L, Fujimoto J, Carrot-Zhang J, McGranahan N, Ying L, Fukuoka J, Chow CW, Pham HHN, Godoy MCB, Carter BW, Behrens C, Zhang J, Antonoff MB, Sepesi B, Lu Y, Pass HI, Kadara H, Scheet P, Vaporciyan AA, Heymach JV, Wistuba II, Lee JJ, Futreal PA, Su D, Issa JJ, and Zhang J
- Subjects
- Adenocarcinoma of Lung mortality, Adenocarcinoma of Lung pathology, Adult, Aged, Aged, 80 and over, Carcinogenesis genetics, Chromosomal Instability, DNA Copy Number Variations, Disease Progression, Female, Genetic Heterogeneity, Humans, Hyperplasia genetics, Hyperplasia pathology, Lung pathology, Lung Neoplasms mortality, Lung Neoplasms pathology, Male, Middle Aged, Mutagenesis, Mutation Rate, Precancerous Conditions pathology, Survival Analysis, Tumor Microenvironment genetics, Adenocarcinoma of Lung genetics, DNA Methylation genetics, Epigenome genetics, Lung Neoplasms genetics, Precancerous Conditions genetics
- Abstract
The evolution of DNA methylome and methylation intra-tumor heterogeneity (ITH) during early carcinogenesis of lung adenocarcinoma has not been systematically studied. We perform reduced representation bisulfite sequencing of invasive lung adenocarcinoma and its precursors, atypical adenomatous hyperplasia, adenocarcinoma in situ and minimally invasive adenocarcinoma. We observe gradual increase of methylation aberrations and significantly higher level of methylation ITH in later-stage lesions. The phylogenetic patterns inferred from methylation aberrations resemble those based on somatic mutations suggesting parallel methylation and genetic evolution. De-convolution reveal higher ratio of T regulatory cells (Tregs) versus CD8 + T cells in later-stage diseases, implying progressive immunosuppression with neoplastic progression. Furthermore, increased global hypomethylation is associated with higher mutation burden, copy number variation burden and AI burden as well as higher Treg/CD8 ratio, highlighting the potential impact of methylation on chromosomal instability, mutagenesis and tumor immune microenvironment during early carcinogenesis of lung adenocarcinomas.
- Published
- 2021
- Full Text
- View/download PDF
14. Author Correction: Bone morphogenetic protein 7 promotes resistance to immunotherapy.
- Author
-
Cortez MA, Masrorpour F, Ivan C, Zhang J, Younes AI, Lu Y, Estecio MR, Barsoumian HB, Menon H, da Silva Caetano M, Ramapriyan R, Schoenhals JE, Wang X, Skoulidis F, Wasley MD, Calin G, Hwu P, and Welsh JW
- Abstract
An amendment to this paper has been published and can be accessed via a link at the top of the paper.
- Published
- 2020
- Full Text
- View/download PDF
15. Bone morphogenetic protein 7 promotes resistance to immunotherapy.
- Author
-
Cortez MA, Masrorpour F, Ivan C, Zhang J, Younes AI, Lu Y, Estecio MR, Barsoumian HB, Menon H, Caetano MDS, Ramapriyan R, Schoenhals JE, Wang X, Skoulidis F, Wasley MD, Calin G, Hwu P, and Welsh JW
- Subjects
- Animals, Antibodies, Monoclonal, Humanized pharmacology, CD4-Positive T-Lymphocytes, Cell Line, Tumor, Female, Follistatin metabolism, Gene Expression Regulation, Neoplastic, Gene Knockdown Techniques, Humans, Mice, Mitogen-Activated Protein Kinase 14 genetics, Mitogen-Activated Protein Kinase 14 metabolism, Programmed Cell Death 1 Receptor drug effects, RAW 264.7 Cells, Smad1 Protein metabolism, Transcriptome, Tumor Microenvironment drug effects, Bone Morphogenetic Protein 7 genetics, Bone Morphogenetic Protein 7 metabolism, Drug Resistance, Neoplasm drug effects, Immunotherapy methods, Neoplasms metabolism
- Abstract
Immunotherapies revolutionized cancer treatment by harnessing the immune system to target cancer cells. However, most patients are resistant to immunotherapies and the mechanisms underlying this resistant is still poorly understood. Here, we report that overexpression of BMP7, a member of the TGFB superfamily, represents a mechanism for resistance to anti-PD1 therapy in preclinical models and in patients with disease progression while on immunotherapies. BMP7 secreted by tumor cells acts on macrophages and CD4
+ T cells in the tumor microenvironment, inhibiting MAPK14 expression and impairing pro-inflammatory responses. Knockdown of BMP7 or its neutralization via follistatin in combination with anti-PD1 re-sensitizes resistant tumors to immunotherapies. Thus, we identify the BMP7 signaling pathway as a potential immunotherapeutic target in cancer.- Published
- 2020
- Full Text
- View/download PDF
16. Longitudinal increases in somatic mosaicism of the expanded CTG repeat in myotonic dystrophy type 1 are associated with variation in age-at-onset.
- Author
-
Morales F, Vásquez M, Corrales E, Vindas-Smith R, Santamaría-Ulloa C, Zhang B, Sirito M, Estecio MR, Krahe R, and Monckton DG
- Subjects
- Adolescent, Age Factors, Age of Onset, Alleles, Child, Child, Preschool, Female, Humans, Male, Myotonic Dystrophy pathology, Phenotype, Trinucleotide Repeat Expansion, DNA genetics, Mosaicism, Myotonic Dystrophy genetics, Trinucleotide Repeats genetics
- Abstract
In myotonic dystrophy type 1 (DM1), somatic mosaicism of the (CTG)n repeat expansion is age-dependent, tissue-specific and expansion-biased. These features contribute toward variation in disease severity and confound genotype-to-phenotype analyses. To investigate how the (CTG)n repeat expansion changes over time, we collected three longitudinal blood DNA samples separated by 8-15 years and used small pool and single-molecule PCR in 43 DM1 patients. We used the lower boundary of the allele length distribution as the best estimate for the inherited progenitor allele length (ePAL), which is itself the best predictor of disease severity. Although in most patients the lower boundary of the allele length distribution was conserved over time, in many this estimate also increased with age, suggesting samples for research studies and clinical trials should be obtained as early as possible. As expected, the modal allele length increased over time, driven primarily by ePAL, age-at-sampling and the time interval. As expected, small expansions <100 repeats did not expand as rapidly as larger alleles. However, the rate of expansion of very large alleles was not obviously proportionally higher. This may, at least in part, be a result of the allele length-dependent increase in large contractions that we also observed. We also determined that individual-specific variation in the increase of modal allele length over time not accounted for by ePAL, age-at-sampling and time was inversely associated with individual-specific variation in age-at-onset not accounted for by ePAL, further highlighting somatic expansion as a therapeutic target in DM1., (© The Author(s) 2020. Published by Oxford University Press. All rights reserved. For Permissions, please email: journals.permissions@oup.com.)
- Published
- 2020
- Full Text
- View/download PDF
17. miR-543 regulates the epigenetic landscape of myelofibrosis by targeting TET1 and TET2.
- Author
-
Fuentes-Mattei E, Bayraktar R, Manshouri T, Silva AM, Ivan C, Gulei D, Fabris L, Soares do Amaral N, Mur P, Perez C, Torres-Claudio E, Dragomir MP, Badillo-Perez A, Knutsen E, Narayanan P, Golfman L, Shimizu M, Zhang X, Zhao W, Ho WT, Estecio MR, Bartholomeusz G, Tomuleasa C, Berindan-Neagoe I, Zweidler-McKay PA, Estrov Z, Zhao ZJ, Verstovsek S, Calin GA, and Redis RS
- Subjects
- Animals, Cytokines metabolism, DNA-Binding Proteins genetics, Dioxygenases, Disease Models, Animal, Histones, Humans, Janus Kinase Inhibitors therapeutic use, Janus Kinases metabolism, Mice, MicroRNAs genetics, Mixed Function Oxygenases, Mutation, Myeloproliferative Disorders, Nitriles, Primary Myelofibrosis genetics, Protein Kinase Inhibitors pharmacology, Proto-Oncogene Proteins genetics, Pyrazoles therapeutic use, Pyrimidines, STAT3 Transcription Factor, Transcriptome, United States, DNA-Binding Proteins drug effects, Epigenesis, Genetic drug effects, MicroRNAs metabolism, MicroRNAs pharmacology, Primary Myelofibrosis drug therapy, Proto-Oncogene Proteins drug effects
- Abstract
Myelofibrosis (MF) is a myeloproliferative neoplasm characterized by cytopenia and extramedullary hematopoiesis, resulting in splenomegaly. Multiple pathological mechanisms (e.g., circulating cytokines and genetic alterations, such as JAKV617F mutation) have been implicated in the etiology of MF, but the molecular mechanism causing resistance to JAK2V617F inhibitor therapy remains unknown. Among MF patients who were treated with the JAK inhibitor ruxolitinib, we compared noncoding RNA profiles of ruxolitinib therapy responders versus nonresponders and found miR-543 was significantly upregulated in nonresponders. We validated these findings by reverse transcription-quantitative PCR. in this same cohort, in 2 additional independent MF patient cohorts from the United States and Romania, and in a JAK2V617F mouse model of MF. Both in vitro and in vivo models were used to determine the underlying molecular mechanism of miR-543 in MF. Here, we demonstrate that miR-543 targets the dioxygenases ten-eleven translocation 1 (TET1) and 2 (TET2) in patients and in vitro, causing increased levels of global 5-methylcytosine, while decreasing the acetylation of histone 3, STAT3, and tumor protein p53. Mechanistically, we found that activation of STAT3 by JAKs epigenetically controls miR-543 expression via binding the promoter region of miR-543. Furthermore, miR-543 upregulation promotes the expression of genes related to drug metabolism, including CYP3A4, which is involved in ruxolitinib metabolism. Our findings suggest miR-543 as a potentially novel biomarker for the prognosis of MF patients with a high risk of treatment resistance and as a potentially new target for the development of new treatment options.
- Published
- 2020
- Full Text
- View/download PDF
18. DNA methylation patterns in bladder tumors of African American patients point to distinct alterations in xenobiotic metabolism.
- Author
-
Vantaku V, Amara CS, Piyarathna DWB, Donepudi SR, Ambati CR, Putluri V, Tang W, Rajapakshe K, Estecio MR, Terris MK, Castro PD, Ittmann MM, Williams SB, Lerner SP, Sreekumar A, Bollag R, Coarfa C, Kornberg MD, Lotan Y, Ambs S, and Putluri N
- Subjects
- Black or African American genetics, Chromatography, Liquid, Epigenesis, Genetic, Gene Expression Regulation, Neoplastic, Glucuronic Acid analysis, Glucuronic Acid metabolism, Humans, Metabolomics, Promoter Regions, Genetic, S-Adenosylhomocysteine analysis, S-Adenosylhomocysteine metabolism, S-Adenosylmethionine analysis, S-Adenosylmethionine metabolism, Tandem Mass Spectrometry, Urinary Bladder Neoplasms metabolism, White People genetics, CpG Islands, DNA Methylation, Inactivation, Metabolic genetics, Urinary Bladder Neoplasms genetics
- Abstract
Racial/ethnic disparities have a significant impact on bladder cancer outcomes with African American patients demonstrating inferior survival over European-American patients. We hypothesized that epigenetic difference in methylation of tumor DNA is an underlying cause of this survival health disparity. We analyzed bladder tumors from African American and European-American patients using reduced representation bisulfite sequencing (RRBS) to annotate differentially methylated DNA regions. Liquid chromatography-mass spectrometry (LC-MS/MS) based metabolomics and flux studies were performed to examine metabolic pathways that showed significant association to the discovered DNA methylation patterns. RRBS analysis showed frequent hypermethylated CpG islands in African American patients. Further analysis showed that these hypermethylated CpG islands in patients are commonly located in the promoter regions of xenobiotic enzymes that are involved in bladder cancer progression. On follow-up, LC-MS/MS revealed accumulation of glucuronic acid, S-adenosylhomocysteine, and a decrease in S-adenosylmethionine, corroborating findings from the RRBS and mRNA expression analysis indicating increased glucuronidation and methylation capacities in African American patients. Flux analysis experiments with 13C-labeled glucose in cultured African American bladder cancer cells confirmed these findings. Collectively, our studies revealed robust differences in methylation-related metabolism and expression of enzymes regulating xenobiotic metabolism in African American patients indicate that race/ethnic differences in tumor biology may exist in bladder cancer., (© The Author(s) 2019. Published by Oxford University Press. All rights reserved. For Permissions, please email: journals.permissions@oup.com.)
- Published
- 2019
- Full Text
- View/download PDF
19. DNMT3L facilitates DNA methylation partly by maintaining DNMT3A stability in mouse embryonic stem cells.
- Author
-
Veland N, Lu Y, Hardikar S, Gaddis S, Zeng Y, Liu B, Estecio MR, Takata Y, Lin K, Tomida MW, Shen J, Saha D, Gowher H, Zhao H, and Chen T
- Subjects
- Animals, DNA Methyltransferase 3A, Enzyme Stability genetics, Mice, Mouse Embryonic Stem Cells metabolism, Protein Binding genetics, DNA (Cytosine-5-)-Methyltransferases genetics, DNA Methylation genetics, Embryonic Development genetics
- Abstract
DNMT3L (DNMT3-like), a member of the DNMT3 family, has no DNA methyltransferase activity but regulates de novo DNA methylation. While biochemical studies show that DNMT3L is capable of interacting with both DNMT3A and DNMT3B and stimulating their enzymatic activities, genetic evidence suggests that DNMT3L is essential for DNMT3A-mediated de novo methylation in germ cells but is dispensable for de novo methylation during embryogenesis, which is mainly mediated by DNMT3B. How DNMT3L regulates DNA methylation and what determines its functional specificity are not well understood. Here we show that DNMT3L-deficient mouse embryonic stem cells (mESCs) exhibit downregulation of DNMT3A, especially DNMT3A2, the predominant DNMT3A isoform in mESCs. DNA methylation analysis of DNMT3L-deficient mESCs reveals hypomethylation at many DNMT3A target regions. These results confirm that DNMT3L is a positive regulator of DNA methylation, contrary to a previous report that, in mESCs, DNMT3L regulates DNA methylation positively or negatively, depending on genomic regions. Mechanistically, DNMT3L forms a complex with DNMT3A2 and prevents DNMT3A2 from being degraded. Restoring the DNMT3A protein level in DNMT3L-deficient mESCs partially recovers DNA methylation. Thus, our work uncovers a role for DNMT3L in maintaining DNMT3A stability, which contributes to the effect of DNMT3L on DNMT3A-dependent DNA methylation.
- Published
- 2019
- Full Text
- View/download PDF
20. Cancer-associated rs6983267 SNP and its accompanying long noncoding RNA CCAT2 induce myeloid malignancies via unique SNP-specific RNA mutations.
- Author
-
Shah MY, Ferracin M, Pileczki V, Chen B, Redis R, Fabris L, Zhang X, Ivan C, Shimizu M, Rodriguez-Aguayo C, Dragomir M, Van Roosbroeck K, Almeida MI, Ciccone M, Nedelcu D, Cortez MA, Manshouri T, Calin S, Muftuoglu M, Banerjee PP, Badiwi MH, Parker-Thornburg J, Multani A, Welsh JW, Estecio MR, Ling H, Tomuleasa C, Dima D, Yang H, Alvarez H, You MJ, Radovich M, Shpall E, Fabbri M, Rezvani K, Girnita L, Berindan-Neagoe I, Maitra A, Verstovsek S, Fodde R, Bueso-Ramos C, Gagea M, Manero GG, and Calin GA
- Subjects
- Adult, Aged, Aged, 80 and over, Animals, Cell Line, Tumor, Female, Gene Expression Regulation, Neoplastic, Humans, Male, Mice, Mice, Transgenic, Middle Aged, Myelodysplastic-Myeloproliferative Diseases pathology, Polymorphism, Single Nucleotide genetics, RNA Editing genetics, Cell Proliferation genetics, Myelodysplastic-Myeloproliferative Diseases genetics, RNA, Long Noncoding genetics
- Abstract
The cancer-risk-associated rs6983267 single nucleotide polymorphism (SNP) and the accompanying long noncoding RNA CCAT2 in the highly amplified 8q24.21 region have been implicated in cancer predisposition, although causality has not been established. Here, using allele-specific CCAT2 transgenic mice, we demonstrate that CCAT2 overexpression leads to spontaneous myeloid malignancies. We further identified that CCAT2 is overexpressed in bone marrow and peripheral blood of myelodysplastic/myeloproliferative neoplasms (MDS/MPN) patients. CCAT2 induces global deregulation of gene expression by down-regulating EZH2 in vitro and in vivo in an allele-specific manner. We also identified a novel non-APOBEC, non-ADAR, RNA editing at the SNP locus in MDS/MPN patients and CCAT2 -transgenic mice. The RNA transcribed from the SNP locus in malignant hematopoietic cells have different allelic composition from the corresponding genomic DNA, a phenomenon rarely observed in normal cells. Our findings provide fundamental insights into the functional role of rs6983267 SNP and CCAT2 in myeloid malignancies., (© 2018 Shah et al.; Published by Cold Spring Harbor Laboratory Press.)
- Published
- 2018
- Full Text
- View/download PDF
21. Energy Balance Modulation Impacts Epigenetic Reprogramming, ERα and ERβ Expression, and Mammary Tumor Development in MMTV-neu Transgenic Mice.
- Author
-
Rossi EL, Dunlap SM, Bowers LW, Khatib SA, Doerstling SS, Smith LA, Ford NA, Holley D, Brown PH, Estecio MR, Kusewitt DF, deGraffenried LA, Bultman SJ, and Hursting SD
- Subjects
- Animals, Breast Neoplasms physiopathology, Caloric Restriction, Carcinogenesis genetics, DNA Methylation genetics, Energy Metabolism genetics, Epigenesis, Genetic genetics, Female, Gene Expression Regulation, Neoplastic, Humans, Mammary Neoplasms, Animal etiology, Mammary Neoplasms, Animal physiopathology, Mice, Mice, Transgenic, Obesity complications, Obesity physiopathology, Receptor, ErbB-2 genetics, Risk Factors, Breast Neoplasms genetics, Estrogen Receptor alpha genetics, Estrogen Receptor beta genetics, Mammary Neoplasms, Animal genetics, Obesity genetics
- Abstract
The association between obesity and breast cancer risk and prognosis is well established in estrogen receptor (ER)-positive disease but less clear in HER2-positive disease. Here, we report preclinical evidence suggesting weight maintenance through calorie restriction (CR) may limit risk of HER2-positive breast cancer. In female MMTV-HER2/neu transgenic mice, we found that ERα and ERβ expression, mammary tumorigenesis, and survival are energy balance dependent in association with epigenetic reprogramming. Mice were randomized to receive a CR, overweight-inducing, or diet-induced obesity regimen ( n = 27/group). Subsets of mice ( n = 4/group/time point) were euthanized after 1, 3, and 5 months to characterize diet-dependent metabolic, transcriptional, and epigenetic perturbations. Remaining mice were followed up to 22 months. Relative to the overweight and diet-induced obesity regimens, CR decreased body weight, adiposity, and serum metabolic hormones as expected and also elicited an increase in mammary ERα and ERβ expression. Increased DNA methylation accompanied this pattern, particularly at CpG dinucleotides located within binding or flanking regions for the transcriptional regulator CCCTC-binding factor of ESR1 and ESR2, consistent with sustained transcriptional activation of ERα and ERβ. Mammary expression of the DNA methylation enzyme DNMT1 was stable in CR mice but increased over time in overweight and diet-induced obesity mice, suggesting CR obviates epigenetic alterations concurrent with chronic excess energy intake. In the survival study, CR elicited a significant suppression in spontaneous mammary tumorigenesis. Overall, our findings suggest a mechanistic rationale to prevent or reverse excess body weight as a strategy to reduce HER2-positive breast cancer risk. Cancer Res; 77(9); 2500-11. ©2017 AACR ., (©2017 American Association for Cancer Research.)
- Published
- 2017
- Full Text
- View/download PDF
22. In Vivo Functional Platform Targeting Patient-Derived Xenografts Identifies WDR5-Myc Association as a Critical Determinant of Pancreatic Cancer.
- Author
-
Carugo A, Genovese G, Seth S, Nezi L, Rose JL, Bossi D, Cicalese A, Shah PK, Viale A, Pettazzoni PF, Akdemir KC, Bristow CA, Robinson FS, Tepper J, Sanchez N, Gupta S, Estecio MR, Giuliani V, Dellino GI, Riva L, Yao W, Di Francesco ME, Green T, D'Alesio C, Corti D, Kang Y, Jones P, Wang H, Fleming JB, Maitra A, Pelicci PG, Chin L, DePinho RA, Lanfrancone L, Heffernan TP, and Draetta GF
- Subjects
- Adenocarcinoma genetics, Adenocarcinoma metabolism, Adenocarcinoma pathology, Animals, Carcinogenesis metabolism, Carcinogenesis pathology, Carcinoma, Pancreatic Ductal genetics, Carcinoma, Pancreatic Ductal metabolism, Carcinoma, Pancreatic Ductal pathology, Cell Line, Tumor, Cell Proliferation, DNA Damage, Disease Progression, Epigenesis, Genetic, Humans, Intracellular Signaling Peptides and Proteins, Lentivirus metabolism, Mice, Models, Biological, Multiprotein Complexes metabolism, Pancreatic Neoplasms genetics, Pancreatic Neoplasms pathology, Protein Binding, Protein Subunits metabolism, RNA, Small Interfering metabolism, Stress, Physiological, Histone-Lysine N-Methyltransferase metabolism, Pancreatic Neoplasms metabolism, Proto-Oncogene Proteins c-myc metabolism, Xenograft Model Antitumor Assays
- Abstract
Current treatment regimens for pancreatic ductal adenocarcinoma (PDAC) yield poor 5-year survival, emphasizing the critical need to identify druggable targets essential for PDAC maintenance. We developed an unbiased and in vivo target discovery approach to identify molecular vulnerabilities in low-passage and patient-derived PDAC xenografts or genetically engineered mouse model-derived allografts. Focusing on epigenetic regulators, we identified WDR5, a core member of the COMPASS histone H3 Lys4 (H3K4) MLL (1-4) methyltransferase complex, as a top tumor maintenance hit required across multiple human and mouse tumors. Mechanistically, WDR5 functions to sustain proper execution of DNA replication in PDAC cells, as previously suggested by replication stress studies involving MLL1, and c-Myc, also found to interact with WDR5. We indeed demonstrate that interaction with c-Myc is critical for this function. By showing that ATR inhibition mimicked the effects of WDR5 suppression, these data provide rationale to test ATR and WDR5 inhibitors for activity in this disease., (Copyright © 2016 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
23. Obesity-Associated Alterations in Inflammation, Epigenetics, and Mammary Tumor Growth Persist in Formerly Obese Mice.
- Author
-
Rossi EL, de Angel RE, Bowers LW, Khatib SA, Smith LA, Van Buren E, Bhardwaj P, Giri D, Estecio MR, Troester MA, Hair BY, Kirk EL, Gong T, Shen J, Dannenberg AJ, and Hursting SD
- Subjects
- Animals, Carcinoma, Ductal, Breast genetics, Carcinoma, Ductal, Breast pathology, DNA Methylation genetics, Disease Models, Animal, Female, Humans, Inflammation complications, Mice, Mice, Inbred C57BL, Oligonucleotide Array Sequence Analysis, Polymerase Chain Reaction, Epigenesis, Genetic, Mammary Neoplasms, Experimental etiology, Mammary Neoplasms, Experimental genetics, Mammary Neoplasms, Experimental pathology, Obesity complications
- Abstract
Using a murine model of basal-like breast cancer, we tested the hypothesis that chronic obesity, an established breast cancer risk and progression factor in women, induces mammary gland epigenetic reprogramming and increases mammary tumor growth. Moreover, we assessed whether the obesity-induced epigenetic and protumor effects are reversed by weight normalization. Ovariectomized female C57BL/6 mice were fed a control diet or diet-induced obesity (DIO) regimen for 17 weeks, resulting in a normal weight or obese phenotype, respectively. Mice on the DIO regimen were then randomized to continue the DIO diet or were switched to the control diet, resulting in formerly obese (FOb) mice with weights comparable with control mice. At week 24, all mice were orthotopically injected with MMTV-Wnt-1 mouse mammary tumor cells. Mean tumor volume, serum IL6 levels, expression of proinflammatory genes in the mammary fat pad, and mammary DNA methylation profiles were similar in DIO and FOb mice and higher than in controls. Many of the genes found to have obesity-associated hypermethylation in mice were also found to be hypermethylated in the normal breast tissue of obese versus nonobese human subjects, and nearly all of these concordant genes remained hypermethylated after significant weight loss in the FOb mice. Our findings suggest that weight normalization may not be sufficient to reverse the effects of chronic obesity on epigenetic reprogramming and inflammatory signals in the microenvironment that are associated with breast cancer progression. Cancer Prev Res; 9(5); 339-48. ©2016 AACR., (©2016 American Association for Cancer Research.)
- Published
- 2016
- Full Text
- View/download PDF
24. A Molecular Portrait of High-Grade Ductal Carcinoma In Situ.
- Author
-
Abba MC, Gong T, Lu Y, Lee J, Zhong Y, Lacunza E, Butti M, Takata Y, Gaddis S, Shen J, Estecio MR, Sahin AA, and Aldaz CM
- Subjects
- Antigens, Differentiation, T-Lymphocyte analysis, Breast chemistry, Breast Neoplasms chemistry, Breast Neoplasms immunology, CTLA-4 Antigen analysis, Carcinoma, Intraductal, Noninfiltrating classification, Carcinoma, Intraductal, Noninfiltrating genetics, Carcinoma, Intraductal, Noninfiltrating immunology, Female, Gene Expression Regulation, Neoplastic, Genes, Neoplasm, Humans, Lymphocytes, Tumor-Infiltrating chemistry, Lymphocytes, Tumor-Infiltrating immunology, Mutation, Neoplasm Invasiveness genetics, Neoplasm Proteins analysis, RNA, Messenger genetics, RNA, Neoplasm genetics, RNA, Untranslated genetics, T-Lymphocytes, Regulatory immunology, Breast Neoplasms genetics, Carcinoma, Intraductal, Noninfiltrating chemistry, DNA Methylation, DNA, Neoplasm genetics, Gene Expression Profiling, Neoplasm Proteins genetics, Transcriptome
- Abstract
Ductal carcinoma in situ (DCIS) is a noninvasive precursor lesion to invasive breast carcinoma. We still have no understanding on why only some DCIS lesions evolve to invasive cancer whereas others appear not to do so during the life span of the patient. Here, we performed full exome (tumor vs. matching normal), transcriptome, and methylome analysis of 30 pure high-grade DCIS (HG-DCIS) and 10 normal breast epithelial samples. Sixty-two percent of HG-DCIS cases displayed mutations affecting cancer driver genes or potential drivers. Mutations were observed affecting PIK3CA (21% of cases), TP53 (17%), GATA3 (7%), MLL3 (7%) and single cases of mutations affecting CDH1, MAP2K4, TBX3, NF1, ATM, and ARID1A. Significantly, 83% of lesions displayed numerous large chromosomal copy number alterations, suggesting they might precede selection of cancer driver mutations. Integrated pathway-based modeling analysis of RNA-seq data allowed us to identify two DCIS subgroups (DCIS-C1 and DCIS-C2) based on their tumor-intrinsic subtypes, proliferative, immune scores, and in the activity of specific signaling pathways. The more aggressive DCIS-C1 (highly proliferative, basal-like, or ERBB2(+)) displayed signatures characteristic of activated Treg cells (CD4(+)/CD25(+)/FOXP3(+)) and CTLA4(+)/CD86(+) complexes indicative of a tumor-associated immunosuppressive phenotype. Strikingly, all lesions showed evidence of TP53 pathway inactivation. Similarly, ncRNA and methylation profiles reproduce changes observed postinvasion. Among the most significant findings, we observed upregulation of lncRNA HOTAIR in DCIS-C1 lesions and hypermethylation of HOXA5 and SOX genes. We conclude that most HG-DCIS lesions, in spite of representing a preinvasive stage of tumor progression, displayed molecular profiles indistinguishable from invasive breast cancer., (©2015 American Association for Cancer Research.)
- Published
- 2015
- Full Text
- View/download PDF
25. Epigenetic synergy between decitabine and platinum derivatives.
- Author
-
Qin T, Si J, Raynal NJ, Wang X, Gharibyan V, Ahmed S, Hu X, Jin C, Lu Y, Shu J, Estecio MR, Jelinek J, and Issa JJ
- Abstract
Background: Aberrant epigenetic silencing of tumor suppressor genes has been recognized as a driving force in cancer. Epigenetic drugs such as the DNA methylation inhibitor decitabine reactivate genes and are effective in myeloid leukemia, but resistance often develops and efficacy in solid tumors is limited. To improve their clinical efficacy, we searched among approved anti-cancer drugs for an epigenetic synergistic combination with decitabine., Results: We used the YB5 cell line, a clonal derivative of the SW48 colon cancer cell line that contains a single copy of a hypermethylated cytomegalovirus (CMV) promoter driving green fluorescent protein (GFP) to screen for drug-induced gene reactivation and synergy with decitabine. None of the 16 anti-cancer drugs tested had effects on their own. However, in combination with decitabine, platinum compounds showed striking synergy in activating GFP. This was dose dependent, observed both in concurrent and sequential combinations, and also seen with other alkylating agents. Clinically achievable concentrations of carboplatin at (25 μM) and decitabine reactivated GFP in 28 % of the YB5 cells as compared to 15 % with decitabine alone. Epigenetic synergy was also seen at endogenously hypermethylated tumor suppressor genes such as MLH1 and PDLIM4. Genome-wide studies showed that reactivation of hypermethylated genes by the combination was significantly better than that induced by decitabine alone or carboplatin alone. Platinum compounds did not enhance decitabine-induced hypomethylation. Rather, we found significantly inhibited HP1α expression by carboplatin and the combination. This was accompanied by increased histone H3 lysine 4 (H3K4) trimethylation and histone H3 lysine 9 (H3K9) acetylation at reactivated genes (P < 0.0001) and reduced occupancy by methyl-binding proteins including MeCP2 and methyl-CpG-binding domain protein 2 (MBD2) (P < 0.0001)., Conclusions: Our results suggest that the combination of decitabine with platinum analogs shows epigenetic synergy that might be exploited in the treatment of different cancers.
- Published
- 2015
- Full Text
- View/download PDF
26. Regulation of AURKC expression by CpG island methylation in human cancer cells.
- Author
-
Fujii S, Srivastava V, Hegde A, Kondo Y, Shen L, Hoshino K, Gonzalez Y, Wang J, Sasai K, Ma X, Katayama H, Estecio MR, Hamilton SR, Wistuba I, Issa JP, and Sen S
- Subjects
- Cell Transformation, Neoplastic, Humans, Male, Neoplasms pathology, Promoter Regions, Genetic genetics, Real-Time Polymerase Chain Reaction, Aurora Kinase C genetics, CpG Islands genetics, DNA Methylation, Gene Expression Regulation, Neoplastic, Neoplasms genetics, Testis metabolism
- Abstract
AURKC, a member of the Aurora kinase gene family, is highly expressed in testis but is either moderately expressed or repressed in most somatic cells. Varying expression of AURKC has been observed in human cancers, but the underlying mechanisms of differential expression have been investigated only to a limited extent. We investigated the role of promoter CpG methylation in the regulation of AURKC gene expression in human cancer cells, in relation to a recently reported AURKC transcription repressor PLZF/ZBTB16, implicated in transformation and tumorigenesis. AURKC and PLZF/ZBTB16 expression profiles were investigated in reference to CpG methylation status on the AURKC promoter experimentally, and also in The Cancer Genome Atlas (TCGA) dataset involving multiple cancer types. AURKC promoter showed dense to moderate hypermethylation correlating with low to moderate expression of the gene in normal somatic cells and cancer cell lines, while testis with high expression revealed marked hypo-methylation. Treatment with the demethylating agent, 5-aza-dC, but not the histone deacetylase (HDAC) inhibitor, TSA, led to elevated expression in cancer cell lines, indicating that promoter DNA methylation negatively regulates AURKC expression. High expression of PLZF in PLZF-transfected cells treated with 5-aza-dC only partially repressed expression of AURKC despite 5-aza-dC also inducing elevated PLZF expression. Analyses of the TCGA data showed differential expression of AURKC in multiple cancer types and stronger correlation of AURKC expression with CpG methylation compared to PLZF levels. These findings demonstrate that differential promoter CpG methylation is an important mechanism regulating AURKC expression in cancer cells.
- Published
- 2015
- Full Text
- View/download PDF
27. TET2 Mutations Affect Non-CpG Island DNA Methylation at Enhancers and Transcription Factor-Binding Sites in Chronic Myelomonocytic Leukemia.
- Author
-
Yamazaki J, Jelinek J, Lu Y, Cesaroni M, Madzo J, Neumann F, He R, Taby R, Vasanthakumar A, Macrae T, Ostler KR, Kantarjian HM, Liang S, Estecio MR, Godley LA, and Issa JP
- Subjects
- Binding Sites genetics, Case-Control Studies, CpG Islands, Dioxygenases, Gene Expression Profiling, Gene Expression Regulation, Leukemic, Genome, Human, Hematopoiesis genetics, High-Throughput Nucleotide Sequencing, Humans, Microarray Analysis, Promoter Regions, Genetic genetics, DNA Methylation genetics, DNA-Binding Proteins genetics, Enhancer Elements, Genetic genetics, Leukemia, Myelomonocytic, Chronic genetics, Mutation, Missense, Proto-Oncogene Proteins genetics, Transcription Factors metabolism
- Abstract
TET2 enzymatically converts 5-methylcytosine to 5-hydroxymethylcytosine as well as other covalently modified cytosines and its mutations are common in myeloid leukemia. However, the exact mechanism and the extent to which TET2 mutations affect DNA methylation remain in question. Here, we report on DNA methylomes in TET2 wild-type (TET2-WT) and mutant (TET2-MT) cases of chronic myelomonocytic leukemia (CMML). We analyzed 85,134 CpG sites [28,114 sites in CpG islands (CGI) and 57,020 in non-CpG islands (NCGI)]. TET2 mutations do not explain genome-wide differences in DNA methylation in CMML, and we found few and inconsistent differences at CGIs between TET2-WT and TET2-MT cases. In contrast, we identified 409 (0.71%) TET2-specific differentially methylated CpGs (tet2-DMCs) in NCGIs, 86% of which were hypermethylated in TET2-MT cases, suggesting a strikingly different biology of the effects of TET2 mutations at CGIs and NCGIs. DNA methylation of tet2-DMCs at promoters and nonpromoters repressed gene expression. Tet2-DMCs showed significant enrichment at hematopoietic-specific enhancers marked by H3K4me1 and at binding sites for the transcription factor p300. Tet2-DMCs showed significantly lower 5-hydroxymethylcytosine in TET2-MT cases. We conclude that leukemia-associated TET2 mutations affect DNA methylation at NCGI regions containing hematopoietic-specific enhancers and transcription factor-binding sites., (©2015 American Association for Cancer Research.)
- Published
- 2015
- Full Text
- View/download PDF
28. BM-SNP: A Bayesian Model for SNP Calling Using High Throughput Sequencing Data.
- Author
-
Xu Y, Zheng X, Yuan Y, Estecio MR, Issa JP, Qiu P, Ji Y, and Liang S
- Subjects
- Bayes Theorem, Cell Line, Cell Line, Tumor, Humans, Markov Chains, Stem Cells, High-Throughput Nucleotide Sequencing methods, Polymorphism, Single Nucleotide genetics, Sequence Analysis, DNA methods
- Abstract
A single-nucleotide polymorphism (SNP) is a sole base change in the DNA sequence and is the most common polymorphism. Detection and annotation of SNPs are among the central topics in biomedical research as SNPs are believed to play important roles on the manifestation of phenotypic events, such as disease susceptibility. To take full advantage of the next-generation sequencing (NGS) technology, we propose a Bayesian approach, BM-SNP, to identify SNPs based on the posterior inference using NGS data. In particular, BM-SNP computes the posterior probability of nucleotide variation at each covered genomic position using the contents and frequency of the mapped short reads. The position with a high posterior probability of nucleotide variation is flagged as a potential SNP. We apply BM-SNP to two cell-line NGS data, and the results show a high ratio of overlap ( >95 percent) with the dbSNP database. Compared with MAQ, BM-SNP identifies more SNPs that are in dbSNP, with higher quality. The SNPs that are called only by BM-SNP but not in dbSNP may serve as new discoveries. The proposed BM-SNP method integrates information from multiple aspects of NGS data, and therefore achieves high detection power. BM-SNP is fast, capable of processing whole genome data at 20-fold average coverage in a short amount of time.
- Published
- 2014
- Full Text
- View/download PDF
29. Expression of PD-L1, PD-L2, PD-1 and CTLA4 in myelodysplastic syndromes is enhanced by treatment with hypomethylating agents.
- Author
-
Yang H, Bueso-Ramos C, DiNardo C, Estecio MR, Davanlou M, Geng QR, Fang Z, Nguyen M, Pierce S, Wei Y, Parmar S, Cortes J, Kantarjian H, and Garcia-Manero G
- Subjects
- Adult, Aged, Aged, 80 and over, Antimetabolites, Antineoplastic pharmacology, Azacitidine pharmacology, B7-H1 Antigen metabolism, Biomarkers, Tumor genetics, Biomarkers, Tumor metabolism, CTLA-4 Antigen metabolism, Cohort Studies, DNA Methylation, Decitabine, Female, Flow Cytometry, Humans, Immunoenzyme Techniques, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute mortality, Leukemia, Myelomonocytic, Chronic drug therapy, Leukemia, Myelomonocytic, Chronic mortality, Leukocytes, Mononuclear drug effects, Leukocytes, Mononuclear metabolism, Male, Middle Aged, Myelodysplastic Syndromes drug therapy, Myelodysplastic Syndromes mortality, Prognosis, Programmed Cell Death 1 Ligand 2 Protein metabolism, Programmed Cell Death 1 Receptor metabolism, RNA, Messenger genetics, Real-Time Polymerase Chain Reaction, Reverse Transcriptase Polymerase Chain Reaction, Survival Rate, Tumor Cells, Cultured, Azacitidine analogs & derivatives, B7-H1 Antigen genetics, CTLA-4 Antigen genetics, Leukemia, Myeloid, Acute genetics, Leukemia, Myelomonocytic, Chronic genetics, Myelodysplastic Syndromes genetics, Programmed Cell Death 1 Ligand 2 Protein genetics, Programmed Cell Death 1 Receptor genetics
- Abstract
Blockade of immune checkpoints is emerging as a new form of anticancer therapy. We studied the expression of programmed death ligand 1 (PD-L1), PD-L2, programmed death 1 (PD-1) and cytotoxic T lymphocyte-associated antigen 4 (CTLA4) mRNA in CD34+ cells from myelodysplastic syndrome (MDS), chronic myelomonocytic leukemia (CMML) and acute myeloid leukemia (AML) patients (N=124). Aberrant upregulation (⩾2-fold) was observed in 34, 14, 15 and 8% of the patients. Increased expression of these four genes was also observed in peripheral blood mononuclear cells (PBMNCs) (N=61). The relative expression of PD-L1 from PBMNC was significantly higher in MDS (P=0.018) and CMML (P=0.0128) compared with AML. By immunohistochemical analysis, PD-L1 protein expression was observed in MDS CD34+ cells, whereas stroma/non-blast cellular compartment was positive for PD-1. In a cohort of patients treated with epigenetic therapy, PD-L1, PD-L2, PD-1 and CTLA4 expression was upregulated. Patients resistant to therapy had relative higher increments in gene expression compared with patients who achieved response. Treatment of leukemia cells with decitabine resulted in a dose-dependent upregulation of above genes. Exposure to decitabine resulted in partial demethylation of PD-1 in leukemia cell lines and human samples. This study suggests that PD-1 signaling may be involved in MDS pathogenesis and resistance mechanisms to hypomethylating agents. Blockade of this pathway can be a potential therapy in MDS and AML.
- Published
- 2014
- Full Text
- View/download PDF
30. TET1 is a maintenance DNA demethylase that prevents methylation spreading in differentiated cells.
- Author
-
Jin C, Lu Y, Jelinek J, Liang S, Estecio MR, Barton MC, and Issa JP
- Subjects
- 5-Methylcytosine analogs & derivatives, Catalytic Domain, Cell Differentiation genetics, CpG Islands, Cytosine analogs & derivatives, Cytosine analysis, Cytosine metabolism, DNA-Binding Proteins chemistry, Dioxygenases chemistry, HEK293 Cells, Humans, Mixed Function Oxygenases, Proto-Oncogene Proteins chemistry, Transcription, Genetic, DNA Methylation, DNA-Binding Proteins metabolism, Dioxygenases metabolism, Proto-Oncogene Proteins metabolism
- Abstract
TET1 is a 5-methylcytosine dioxygenase and its DNA demethylating activity has been implicated in pluripotency and reprogramming. However, the precise role of TET1 in DNA methylation regulation outside of developmental reprogramming is still unclear. Here, we show that overexpression of the TET1 catalytic domain but not full length TET1 (TET1-FL) induces massive global DNA demethylation in differentiated cells. Genome-wide mapping reveals that 5-hydroxymethylcytosine production by TET1-FL is inhibited as DNA methylation increases, which can be explained by the preferential binding of TET1-FL to unmethylated CpG islands (CGIs) through its CXXC domain. TET1-FL specifically accumulates 5-hydroxymethylcytosine at the edges of hypomethylated CGIs, while knockdown of endogenous TET1 induces methylation spreading from methylated edges into hypomethylated CGIs. We also found that gene expression changes after TET1-FL overexpression are relatively small and independent of its dioxygenase function. Thus, our results identify TET1 as a maintenance DNA demethylase that does not purposely decrease methylation levels, but specifically prevents aberrant methylation spreading into CGIs in differentiated cells., (© The Author(s) 2014. Published by Oxford University Press on behalf of Nucleic Acids Research.)
- Published
- 2014
- Full Text
- View/download PDF
31. Age-related epigenetic drift in the pathogenesis of MDS and AML.
- Author
-
Maegawa S, Gough SM, Watanabe-Okochi N, Lu Y, Zhang N, Castoro RJ, Estecio MR, Jelinek J, Liang S, Kitamura T, Aplan PD, and Issa JP
- Subjects
- Animals, Bone Marrow Cells, Core Binding Factor Alpha 2 Subunit genetics, Disease Models, Animal, Humans, Leukemia, Myeloid, Acute pathology, Mice, Myelodysplastic Syndromes pathology, Nuclear Pore Complex Proteins genetics, DNA Methylation genetics, Epigenesis, Genetic genetics, Leukemia, Myeloid, Acute genetics, Myelodysplastic Syndromes genetics
- Abstract
The myelodysplastic syndrome (MDS) is a clonal hematologic disorder that frequently evolves to acute myeloid leukemia (AML). Its pathogenesis remains unclear, but mutations in epigenetic modifiers are common and the disease often responds to DNA methylation inhibitors. We analyzed DNA methylation in the bone marrow and spleen in two mouse models of MDS/AML, the NUP98-HOXD13 (NHD13) mouse and the RUNX1 mutant mouse model. Methylation array analysis showed an average of 512/3445 (14.9%) genes hypermethylated in NHD13 MDS, and 331 (9.6%) genes hypermethylated in RUNX1 MDS. Thirty-two percent of genes in common between the two models (2/3 NHD13 mice and 2/3 RUNX1 mice) were also hypermethylated in at least two of 19 human MDS samples. Detailed analysis of 41 genes in mice showed progressive drift in DNA methylation from young to old normal bone marrow and spleen; to MDS, where we detected accelerated age-related methylation; and finally to AML, which markedly extends DNA methylation abnormalities. Most of these genes showed similar patterns in human MDS and AML. Repeat element hypomethylation was rare in MDS but marked the transition to AML in some cases. Our data show consistency in patterns of aberrant DNA methylation in human and mouse MDS and suggest that epigenetically, MDS displays an accelerated aging phenotype.
- Published
- 2014
- Full Text
- View/download PDF
32. Architecture of epigenetic reprogramming following Twist1-mediated epithelial-mesenchymal transition.
- Author
-
Malouf GG, Taube JH, Lu Y, Roysarkar T, Panjarian S, Estecio MR, Jelinek J, Yamazaki J, Raynal NJ, Long H, Tahara T, Tinnirello A, Ramachandran P, Zhang XY, Liang S, Mani SA, and Issa JP
- Subjects
- Cells, Cultured, Epithelial Cells metabolism, Female, Gene Expression Regulation, Genome, Human, Histones metabolism, Humans, Mammary Glands, Human metabolism, Molecular Sequence Data, Promoter Regions, Genetic, Sequence Analysis, RNA methods, Epigenesis, Genetic, Epithelial-Mesenchymal Transition, Mammary Glands, Human cytology, Nuclear Proteins genetics, Twist-Related Protein 1 genetics
- Abstract
Background: Epithelial-mesenchymal transition (EMT) is known to impart metastasis and stemness characteristics in breast cancer. To characterize the epigenetic reprogramming following Twist1-induced EMT, we characterized the epigenetic and transcriptome landscapes using whole-genome transcriptome analysis by RNA-seq, DNA methylation by digital restriction enzyme analysis of methylation (DREAM) and histone modifications by CHIP-seq of H3K4me3 and H3K27me3 in immortalized human mammary epithelial cells relative to cells induced to undergo EMT by Twist1., Results: EMT is accompanied by focal hypermethylation and widespread global DNA hypomethylation, predominantly within transcriptionally repressed gene bodies. At the chromatin level, the number of gene promoters marked by H3K4me3 increases by more than one fifth; H3K27me3 undergoes dynamic genomic redistribution characterized by loss at half of gene promoters and overall reduction of peak size by almost half. This is paralleled by increased phosphorylation of EZH2 at serine 21. Among genes with highly altered mRNA expression, 23.1% switch between H3K4me3 and H3K27me3 marks, and those point to the master EMT targets and regulators CDH1, PDGFRα and ESRP1. Strikingly, Twist1 increases the number of bivalent genes by more than two fold. Inhibition of the H3K27 methyltransferases EZH2 and EZH1, which form part of the Polycomb repressive complex 2 (PRC2), blocks EMT and stemness properties., Conclusions: Our findings demonstrate that the EMT program requires epigenetic remodeling by the Polycomb and Trithorax complexes leading to increased cellular plasticity. This suggests that inhibiting epigenetic remodeling and thus decrease plasticity will prevent EMT, and the associated breast cancer metastasis.
- Published
- 2013
- Full Text
- View/download PDF
33. CCAT2, a novel noncoding RNA mapping to 8q24, underlies metastatic progression and chromosomal instability in colon cancer.
- Author
-
Ling H, Spizzo R, Atlasi Y, Nicoloso M, Shimizu M, Redis RS, Nishida N, Gafà R, Song J, Guo Z, Ivan C, Barbarotto E, De Vries I, Zhang X, Ferracin M, Churchman M, van Galen JF, Beverloo BH, Shariati M, Haderk F, Estecio MR, Garcia-Manero G, Patijn GA, Gotley DC, Bhardwaj V, Shureiqi I, Sen S, Multani AS, Welsh J, Yamamoto K, Taniguchi I, Song MA, Gallinger S, Casey G, Thibodeau SN, Le Marchand L, Tiirikainen M, Mani SA, Zhang W, Davuluri RV, Mimori K, Mori M, Sieuwerts AM, Martens JW, Tomlinson I, Negrini M, Berindan-Neagoe I, Foekens JA, Hamilton SR, Lanza G, Kopetz S, Fodde R, and Calin GA
- Subjects
- Animals, Breast Neoplasms genetics, Breast Neoplasms metabolism, Case-Control Studies, Cell Line, Tumor, Colonic Neoplasms metabolism, Colonic Neoplasms pathology, Female, Gene Expression Regulation, Neoplastic, Humans, Male, Mice, MicroRNAs genetics, MicroRNAs metabolism, Neoplasm Metastasis genetics, Polymorphism, Single Nucleotide, Proto-Oncogene Proteins c-myc genetics, Proto-Oncogene Proteins c-myc metabolism, Transcription Factor 7-Like 1 Protein genetics, Transcription Factor 7-Like 1 Protein metabolism, Transcription, Genetic, Wnt Signaling Pathway, Chromosomal Instability, Chromosomes, Human, Pair 8 genetics, Colonic Neoplasms genetics, RNA, Long Noncoding genetics, RNA, Long Noncoding metabolism
- Abstract
The functional roles of SNPs within the 8q24 gene desert in the cancer phenotype are not yet well understood. Here, we report that CCAT2, a novel long noncoding RNA transcript (lncRNA) encompassing the rs6983267 SNP, is highly overexpressed in microsatellite-stable colorectal cancer and promotes tumor growth, metastasis, and chromosomal instability. We demonstrate that MYC, miR-17-5p, and miR-20a are up-regulated by CCAT2 through TCF7L2-mediated transcriptional regulation. We further identify the physical interaction between CCAT2 and TCF7L2 resulting in an enhancement of WNT signaling activity. We show that CCAT2 is itself a WNT downstream target, which suggests the existence of a feedback loop. Finally, we demonstrate that the SNP status affects CCAT2 expression and the risk allele G produces more CCAT2 transcript. Our results support a new mechanism of MYC and WNT regulation by the novel lncRNA CCAT2 in colorectal cancer pathogenesis, and provide an alternative explanation of the SNP-conferred cancer risk.
- Published
- 2013
- Full Text
- View/download PDF
34. The epigenome of AML stem and progenitor cells.
- Author
-
Yamazaki J, Estecio MR, Lu Y, Long H, Malouf GG, Graber D, Huo Y, Ramagli L, Liang S, Kornblau SM, Jelinek J, and Issa JP
- Subjects
- Cell Differentiation genetics, Chromatin metabolism, Chromatin Immunoprecipitation, CpG Islands genetics, DNA Methylation genetics, Gene Expression Regulation, Leukemic, Histones metabolism, Humans, Oligonucleotide Array Sequence Analysis, Protein Processing, Post-Translational genetics, Sequence Analysis, DNA, Signal Transduction genetics, Epigenesis, Genetic, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute pathology, Neoplastic Stem Cells metabolism, Neoplastic Stem Cells pathology
- Abstract
Acute myeloid leukemia (AML) is sustained by a population of cancer stem cells (CSCs or cancer-initiating cell). The mechanisms underlying switches from CSCs to non-CSCs in vivo remain to be understood. We address this issue in AML from the aspect of epigenetics using genome-wide screening for DNA methylation and selected histone modifications. We found no major differences in DNA methylation, especially in promoter CpG islands, between CSCs and non-CSCs. By contrast, we found thousands of genes that change H3K4me3 and/or H3K27me3 status between stem and progenitor cells as well as between progenitor and mature cells. Stem cell related pathways and proliferation or metabolism related pathways characterize genes differentially enriched for H3K4me3/H3K27me3 in stem and progenitor populations. Bivalent genes in stem cells are more plastic during differentiation and are more likely to lose H3K4me3 than to lose H3K27me3, consistent with increasingly closed chromatin state with differentiation. Our data indicates that histone modifications but not promoter DNA methylation are involved in switches from CSCs to non-CSCs in AML.
- Published
- 2013
- Full Text
- View/download PDF
35. A Bayesian Model for SNP Discovery Based on Next-Generation Sequencing Data.
- Author
-
Xu Y, Zheng X, Yuan Y, Estecio MR, Issa JP, Ji Y, and Liang S
- Abstract
A single-nucleotide polymorphism (SNP) is a single base change in the DNA sequence and is the most common polymorphism. Since some SNPs have a major influence on disease susceptibility, detecting SNPs plays an important role in biomedical research. To take fully advantage of the next-generation sequencing (NGS) technology and detect SNP more effectively, we propose a Bayesian approach that computes a posterior probability of hidden nucleotide variations at each covered genomic position. The position with higher posterior probability of hidden nucleotide variation has a higher chance to be a SNP. We apply the proposed method to detect SNPs in two cell lines: the prostate cancer cell line PC3 and the embryonic stem cell line H1. A comparison between our results with dbSNP database shows a high ratio of overlap (>95%). The positions that are called only under our model but not in dbSNP may serve as candidates for new SNPs.
- Published
- 2012
- Full Text
- View/download PDF
36. Conserved DNA methylation patterns in healthy blood cells and extensive changes in leukemia measured by a new quantitative technique.
- Author
-
Jelinek J, Liang S, Lu Y, He R, Ramagli LS, Shpall EJ, Estecio MR, and Issa JP
- Subjects
- Base Sequence, Cell Line, Tumor, Chromosomes, Human, X, Conserved Sequence, CpG Islands, Deoxyribonucleases, Type II Site-Specific metabolism, Female, Genomic Imprinting, Humans, Leukemia pathology, Leukemia, Myeloid, Acute genetics, Male, Reference Values, Repetitive Sequences, Nucleic Acid, Sequence Analysis, DNA economics, DNA Methylation, Leukemia genetics, Leukocytes physiology, Sequence Analysis, DNA methods
- Abstract
Genome wide analysis of DNA methylation provides important information in a variety of diseases, including cancer. Here, we describe a simple method, Digital Restriction Enzyme Analysis of Methylation (DREAM), based on next generation sequencing analysis of methylation-specific signatures created by sequential digestion of genomic DNA with SmaI and XmaI enzymes. DREAM provides information on 150,000 unique CpG sites, of which 39,000 are in CpG islands and 30,000 are at transcription start sites of 13,000 RefSeq genes. We analyzed DNA methylation in healthy white blood cells and found methylation patterns to be remarkably uniform. Inter individual differences > 30% were observed only at 227 of 28,331 (0.8%) of autosomal CpG sites. Similarly, > 30% differences were observed at only 59 sites when we comparing the cord and adult blood. These conserved methylation patterns contrasted with extensive changes affecting 18-40% of CpG sites in a patient with acute myeloid leukemia and in two leukemia cell lines. The method is cost effective, quantitative (r ( 2) = 0.93 when compared with bisulfite pyrosequencing) and reproducible (r ( 2) = 0.997). Using 100-fold coverage, DREAM can detect differences in methylation greater than 10% or 30% with a false positive rate below 0.05 or 0.001, respectively. DREAM can be useful in quantifying epigenetic effects of environment and nutrition, correlating developmental epigenetic variation with phenotypes, understanding epigenetics of cancer and chronic diseases, measuring the effects of drugs on DNA methylation or deriving new biological insights into mammalian genomes.
- Published
- 2012
- Full Text
- View/download PDF
37. Repetitive elements and enforced transcriptional repression co-operate to enhance DNA methylation spreading into a promoter CpG-island.
- Author
-
Zhang Y, Shu J, Si J, Shen L, Estecio MR, and Issa JP
- Subjects
- Cell Line, Tumor, Chromatin metabolism, DNA chemistry, Humans, Intercellular Signaling Peptides and Proteins genetics, Transgenes, CpG Islands, DNA Methylation, Gene Silencing, Promoter Regions, Genetic, Repetitive Sequences, Nucleic Acid, Transcription, Genetic
- Abstract
Repression of many tumor suppressor genes in cancer is concurrent with aberrantly increased DNA methylation levels at promoter CpG islands (CGIs). About one-fourth of empirically defined human promoters are surrounded by or contain clustered repetitive elements. It was previously observed that a sharp transition of methylation exists between highly methylated repetitive elements and unmethylated promoter-CGIs in normal tissues. The factors that lead to aberrant CGI hypermethylation in cancer remain poorly understood. Here, we established a site-specific integration system with enforced local transcriptional repression in colorectal cancer cells and monitored the occurrence of initial de novo methylation at specific CG sites adjacent to the CGI of the INSL6 promoter, which could be accelerated by binding a KRAB-containing transcriptional factor. Additional repetitive elements from P16 and RIL (PDLIM4), if situated adjacent to the promoter of INSL6, could confer DNA methylation spreading into the CGI particularly in the setting of KRAB-factor binding. However, a repressive chromatin alone was not sufficient to initiate DNA methylation, which required specific DNA sequences and was integration-site (and/or cell-line) specific. Overall, these results demonstrate a requirement for specific DNA sequences to trigger de novo DNA methylation, and repetitive elements as cis-regulatory factors to cooperate with advanced transcriptional repression in promoting methylation spreading.
- Published
- 2012
- Full Text
- View/download PDF
38. Effects of TET2 mutations on DNA methylation in chronic myelomonocytic leukemia.
- Author
-
Yamazaki J, Taby R, Vasanthakumar A, Macrae T, Ostler KR, Shen L, Kantarjian HM, Estecio MR, Jelinek J, Godley LA, and Issa JP
- Subjects
- 5-Methylcytosine metabolism, Adult, Aged, Aged, 80 and over, Antigens, Nuclear genetics, Dioxygenases, Female, Humans, Male, Middle Aged, Nuclear Proteins genetics, Transcription Factors genetics, DNA Methylation genetics, DNA-Binding Proteins genetics, Leukemia, Myelomonocytic, Chronic genetics, Mutation genetics, Proto-Oncogene Proteins genetics
- Abstract
TET2 enzymatically converts 5-methyl-cytosine to 5-hydroxymethyl-cytosine, possibly leading to loss of DNA methylation. TET2 mutations are common in myeloid leukemia and were proposed to contribute to leukemogenesis through DNA methylation. To expand on this concept, we studied chronic myelomonocytic leukemia (CMML) samples. TET2 missense or nonsense mutations were detected in 53% (16/30) of patients. In contrast, only 1/30 patient had a mutation in IDH1 or IDH2, and none of them had a mutation in DNMT3A in the sites most frequently mutated in leukemia. Using bisulfite pyrosequencing, global methylation measured by the LINE-1 assay and DNA methylation levels of 10 promoter CpG islands frequently abnormal in myeloid leukemia were not different between TET2 mutants and wild-type CMML cases. This was also true for 9 out of 11 gene promoters reported by others as differentially methylated by TET2 mutations. We found that two non-CpG island promoters, AIM2 and SP140, were hypermethylated in patients with mutant TET2. These were the only two gene promoters (out of 14,475 genes) previously found to be hypermethylated in TET2 mutant cases. However, total 5-methyl-cytosine levels in TET2 mutant cases were significantly higher than TET2 wild-type cases (median = 14.0% and 9.8%, respectively) (p = 0.016). Thus, TET2 mutations affect global methylation in CMML but most of the changes are likely to be outside gene promoters.
- Published
- 2012
- Full Text
- View/download PDF
39. Frequent alteration of MLL3 frameshift mutations in microsatellite deficient colorectal cancer.
- Author
-
Watanabe Y, Castoro RJ, Kim HS, North B, Oikawa R, Hiraishi T, Ahmed SS, Chung W, Cho MY, Toyota M, Itoh F, Estecio MR, Shen L, Jelinek J, and Issa JP
- Subjects
- Base Sequence, Cell Line, Tumor, Colorectal Neoplasms pathology, DNA Methylation genetics, DNA Mutational Analysis, DNA-Binding Proteins metabolism, Female, Gene Expression Regulation, Neoplastic, Genome, Human genetics, Humans, Male, Middle Aged, Molecular Sequence Data, Reproducibility of Results, Colorectal Neoplasms genetics, DNA-Binding Proteins genetics, Frameshift Mutation genetics, Microsatellite Repeats genetics
- Abstract
Background: MLL3 is a histone 3-lysine 4 methyltransferase with tumor-suppressor properties that belongs to a family of chromatin regulator genes potentially altered in neoplasia. Mutations in MLL3 were found in a whole genome analysis of colorectal cancer but have not been confirmed by a separate study., Methods and Results: We analyzed mutations of coding region and promoter methylation in MLL3 using 126 cases of colorectal cancer. We found two isoforms of MLL3 and DNA sequencing revealed frameshift and other mutations affecting both isoforms of MLL3 in colorectal cancer cells and 19 of 134 (14%) primary colorectal samples analyzed. Moreover, frameshift mutations were more common in cases with microsatellite instability (31%) both in CRC cell lines and primary tumors. The largest isoform of MLL3 is transcribed from a CpG island-associated promoter that has highly homology with a pseudo-gene on chromosome 22 (psiTPTE22). Using an assay which measured both loci simultaneously we found prominent age related methylation in normal colon (from 21% in individuals less than 25 years old to 56% in individuals older than 70, R = 0.88, p<0.001) and frequent hypermethylation (83%) in both CRC cell lines and primary tumors. We next studied the two loci separately and found that age and cancer related methylation was solely a property of the pseudogene CpG island and that the MLL3 loci was unmethylated., Conclusions: We found that frameshift mutations of MLL3 in both CRC cells and primary tumor that were more common in cases with microsatellite instability. Moreover, we have shown CpG island-associated promoter of MLL3 gene has no DNA methylation in CRC cells but also primary tumor and normal colon, and this region has a highly homologous of pseudo gene (psiTPTE22) that was age relate DNA methylation.
- Published
- 2011
- Full Text
- View/download PDF
40. DNA methylation profiles of primary colorectal carcinoma and matched liver metastasis.
- Author
-
Konishi K, Watanabe Y, Shen L, Guo Y, Castoro RJ, Kondo K, Chung W, Ahmed S, Jelinek J, Boumber YA, Estecio MR, Maegawa S, Kondo Y, Itoh F, Imawari M, Hamilton SR, and Issa JP
- Subjects
- Adult, Aged, Aged, 80 and over, Cell Line, Tumor, CpG Islands genetics, DNA Mutational Analysis, Female, Gene Expression Regulation, Neoplastic, Genome, Human genetics, Humans, Male, Middle Aged, Oligonucleotide Array Sequence Analysis, Colorectal Neoplasms genetics, DNA Methylation genetics, Liver Neoplasms genetics, Liver Neoplasms secondary
- Abstract
Background: The contribution of DNA methylation to the metastatic process in colorectal cancers (CRCs) is unclear., Methods: We evaluated the methylation status of 13 genes (MINT1, MINT2, MINT31, MLH1, p16, p14, TIMP3, CDH1, CDH13, THBS1, MGMT, HPP1 and ERα) by bisulfite-pyrosequencing in 79 CRCs comprising 36 CRCs without liver metastasis and 43 CRCs with liver metastasis, including 16 paired primary CRCs and liver metastasis. We also performed methylated CpG island amplification microarrays (MCAM) in three paired primary and metastatic cancers., Results: Methylation of p14, TIMP3 and HPP1 in primary CRCs progressively decreased from absence to presence of liver metastasis (13.1% vs. 4.3%; 14.8% vs. 3.7%; 43.9% vs. 35.8%, respectively) (P<.05). When paired primary and metastatic tumors were compared, only MGMT methylation was significantly higher in metastatic cancers (27.4% vs. 13.4%, P = .013), and this difference was due to an increase in methylation density rather than frequency in the majority of cases. MCAM showed an average 7.4% increase in DNA methylated genes in the metastatic samples. The numbers of differentially hypermethylated genes in the liver metastases increased with increasing time between resection of the primary and resection of the liver metastasis. Bisulfite-pyrosequencing validation in 12 paired samples showed that most of these increases were not conserved, and could be explained by differences in methylation density rather than frequency., Conclusions: Most DNA methylation differences between primary CRCs and matched liver metastasis are due to random variation and an increase in DNA methylation density rather than de-novo inactivation and silencing. Thus, DNA methylation changes occur for the most part before progression to liver metastasis.
- Published
- 2011
- Full Text
- View/download PDF
41. Aberrant DNA methylation is associated with disease progression, resistance to imatinib and shortened survival in chronic myelogenous leukemia.
- Author
-
Jelinek J, Gharibyan V, Estecio MR, Kondo K, He R, Chung W, Lu Y, Zhang N, Liang S, Kantarjian HM, Cortes JE, and Issa JP
- Subjects
- Adolescent, Adult, Aged, Benzamides, Cell Line, Tumor, Female, Gene Expression Regulation, Leukemic drug effects, Genes, Neoplasm genetics, Genome, Human genetics, Humans, Imatinib Mesylate, Leukemia, Myelogenous, Chronic, BCR-ABL Positive drug therapy, Male, Middle Aged, Multivariate Analysis, Neoplasm Staging, Oligonucleotide Array Sequence Analysis, Piperazines adverse effects, Piperazines therapeutic use, Pyrimidines adverse effects, Pyrimidines therapeutic use, Sequence Analysis, DNA, Survival Analysis, Treatment Outcome, Young Adult, DNA Methylation drug effects, DNA Methylation genetics, Disease Progression, Drug Resistance, Neoplasm drug effects, Drug Resistance, Neoplasm genetics, Leukemia, Myelogenous, Chronic, BCR-ABL Positive genetics, Piperazines pharmacology, Pyrimidines pharmacology
- Abstract
The epigenetic impact of DNA methylation in chronic myelogenous leukemia (CML) is not completely understood. To elucidate its role we analyzed 120 patients with CML for methylation of promoter-associated CpG islands of 10 genes. Five genes were identified by DNA methylation screening in the K562 cell line and 3 genes in patients with myeloproliferative neoplasms. The CDKN2B gene was selected for its frequent methylation in myeloid malignancies and ABL1 as the target of BCR-ABL translocation. Thirty patients were imatinib-naïve (mostly treated by interferon-alpha before the imatinib era), 30 were imatinib-responsive, 50 were imatinib-resistant, and 10 were imatinib-intolerant. We quantified DNA methylation by bisulfite pyrosequencing. The average number of methylated genes was 4.5 per patient in the chronic phase, increasing significantly to 6.2 in the accelerated and 6.4 in the blastic phase. Higher numbers of methylated genes were also observed in patients resistant or intolerant to imatinib. These patients also showed almost exclusive methylation of a putative transporter OSCP1. Abnormal methylation of a Src suppressor gene PDLIM4 was associated with shortened survival independently of CML stage and imatinib responsiveness. We conclude that aberrant DNA methylation is associated with CML progression and that DNA methylation could be a marker associated with imatinib resistance. Finally, DNA methylation of PDLIM4 may help identify a subset of CML patients that would benefit from treatment with Src/Abl inhibitors.
- Published
- 2011
- Full Text
- View/download PDF
42. Sensitive and specific detection of early gastric cancer with DNA methylation analysis of gastric washes.
- Author
-
Watanabe Y, Kim HS, Castoro RJ, Chung W, Estecio MR, Kondo K, Guo Y, Ahmed SS, Toyota M, Itoh F, Suk KT, Cho MY, Shen L, Jelinek J, and Issa JP
- Subjects
- Aged, Analysis of Variance, Biomarkers, Tumor analysis, Biomarkers, Tumor genetics, Cell Line, Tumor, DNA, Neoplasm analysis, Early Detection of Cancer, Epigenesis, Genetic, Female, Gene Expression Regulation, Neoplastic, Humans, Male, Middle Aged, Neoplasm Staging, Precancerous Conditions pathology, Probability, ROC Curve, Reverse Transcriptase Polymerase Chain Reaction, Sensitivity and Specificity, Tumor Suppressor Proteins metabolism, DNA Methylation, Genetic Predisposition to Disease, Precancerous Conditions genetics, Stomach Neoplasms genetics, Stomach Neoplasms pathology, Tumor Suppressor Proteins genetics
- Abstract
Background & Aims: Aberrant DNA methylation is an early and frequent process in gastric carcinogenesis and could be useful for detection of gastric neoplasia. We hypothesized that methylation analysis of DNA recovered from gastric washes could be used to detect gastric cancer., Methods: We studied 51 candidate genes in 7 gastric cancer cell lines and 24 samples (training set) and identified 6 for further studies. We examined the methylation status of these genes in a test set consisting of 131 gastric neoplasias at various stages. Finally, we validated the 6 candidate genes in a different population of 40 primary gastric cancer samples and 113 nonneoplastic gastric mucosa samples., Results: Six genes (MINT25, RORA, GDNF, ADAM23, PRDM5, MLF1) showed frequent differential methylation between gastric cancer and normal mucosa in the training, test, and validation sets. GDNF and MINT25 were most sensitive molecular markers of early stage gastric cancer, whereas PRDM5 and MLF1 were markers of a field defect. There was a close correlation (r = 0.5-0.9, P = .03-.001) between methylation levels in tumor biopsy and gastric washes. MINT25 methylation had the best sensitivity (90%), specificity (96%), and area under the receiver operating characteristic curve (0.961) in terms of tumor detection in gastric washes., Conclusions: These findings suggest MINT25 is a sensitive and specific marker for screening in gastric cancer. Additionally, we have developed a new method for gastric cancer detection by DNA methylation in gastric washes.
- Published
- 2009
- Full Text
- View/download PDF
43. Hypomethylation of LINE-1 and Alu in well-differentiated neuroendocrine tumors (pancreatic endocrine tumors and carcinoid tumors).
- Author
-
Choi IS, Estecio MR, Nagano Y, Kim DH, White JA, Yao JC, Issa JP, and Rashid A
- Subjects
- Carcinoid Tumor genetics, Carcinoid Tumor pathology, Cell Line, Tumor, Chromosome Deletion, Chromosomes, Human, Pair 11 genetics, Chromosomes, Human, Pair 16 genetics, Chromosomes, Human, Pair 18 genetics, Cyclin-Dependent Kinase Inhibitor p16 biosynthesis, Cyclin-Dependent Kinase Inhibitor p16 genetics, Female, Humans, Lymphatic Metastasis, Male, Middle Aged, Neuroendocrine Tumors genetics, O(6)-Methylguanine-DNA Methyltransferase genetics, Pancreatic Neoplasms genetics, Pancreatic Neoplasms pathology, Sequence Analysis, DNA methods, Tumor Suppressor Protein p14ARF genetics, Tumor Suppressor Proteins genetics, Alu Elements genetics, DNA Methylation, Long Interspersed Nucleotide Elements genetics, Neuroendocrine Tumors pathology
- Abstract
Neuroendocrine tumors including carcinoid tumors and pancreatic endocrine tumors are uncommon, and the genetic alterations in these indolent tumors are not well characterized. We studied global hypomethylation by analyzing long interspersed nucleotide elements (LINE)-1 and Alu methylation using pyrosequencing in 35 neuroendocrine tumors and corresponding normal tissue. The tumor samples were less methylated than normal tissue at LINE-1 (P=0.04) and Alu (P=0.001). The mean relative tumor hypomethylation (difference in methylation between normal tissue and in tumor) was 11.5+/-10.0 for LINE-1 and 5.8+/-6.4 for Alu, and were correlated with each other (correlation coefficient 0.6, P=0.001). Relative tumor hypomethylation of LINE-1 was higher in ileal carcinoid tumors than in non-ileal carcinoid tumors and pancreatic endocrine tumors (P=0.047), and tumors with lymph node metastasis (P=0.02), chromosome 18 loss (P=0.001) and RAS-association domain family 1, isoform A gene methylation (P=0.02). Alu methylation in tumors was inversely correlated with methylation of O(6)-methyl-guanine methyltransferase gene (P=0.02). Our study shows that hypomethylation is more common in carcinoid tumors than in pancreatic endocrine tumors and is associated with clinicopathologic features, and genetic and epigenetic alterations in these tumors, including lymph node metastasis.
- Published
- 2007
- Full Text
- View/download PDF
44. DNA methylation changes after 5-aza-2'-deoxycytidine therapy in patients with leukemia.
- Author
-
Yang AS, Doshi KD, Choi SW, Mason JB, Mannari RK, Gharybian V, Luna R, Rashid A, Shen L, Estecio MR, Kantarjian HM, Garcia-Manero G, and Issa JP
- Subjects
- Adult, Aged, Aged, 80 and over, Antimetabolites, Antineoplastic therapeutic use, Azacitidine pharmacology, Azacitidine therapeutic use, Decitabine, Dose-Response Relationship, Drug, Female, Genes, p53 drug effects, Homeodomain Proteins genetics, Humans, Leukemia, Myelogenous, Chronic, BCR-ABL Positive metabolism, Leukemia, Myeloid, Acute metabolism, Male, Middle Aged, Mutation, RNA, Long Noncoding, RNA, Untranslated genetics, RNA, Untranslated metabolism, Antimetabolites, Antineoplastic pharmacology, Azacitidine analogs & derivatives, DNA Methylation drug effects, Leukemia, Myelogenous, Chronic, BCR-ABL Positive drug therapy, Leukemia, Myelogenous, Chronic, BCR-ABL Positive genetics, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute genetics
- Abstract
5-Aza-2'-deoxycytidine (decitabine) is postulated to have clinical activity in myeloid leukemias via its ability to inhibit DNA methylation. To study this, we examined DNA methylation in patients with leukemia treated with decitabine. Five days after the treatment, total genomic 5-methylcytosine/cytosine decreased on average by 14% (from 4.3% to 3.7%), whereas methylation of repetitive DNA elements showed a mean decrease of 9% and 16% for Alu and long interspersed nucleotide elements, respectively. Methylation decreased linearly with increasing doses between 5 and 20 mg/m(2)/d (r = 0.88; P = 0.05) but showed a plateau above that. Hypomethylation correlated with response in patients with acute myelogenous leukemia treated with low doses (5-20 mg/m(2)/d), but patients with chronic myelogenous leukemia treated with high doses (100-180 mg/m(2)/d) showed no such correlation. Aberrant methylation of p15 (>10%) was found in 27% of patients, and 80% of these showed a decrease by at least one third, but this did not correlate with response. The imprinted gene H19 showed little change in methylation after decitabine. In conclusion, we show dose-dependent hypomethylation after decitabine at low doses. Increasing the dose, which has been shown previously to result in a reduced response rate, was not accompanied by further hypomethylation.
- Published
- 2006
- Full Text
- View/download PDF
45. Methylation and regulation of expression of different retinoic acid receptor beta isoforms in human colon cancer.
- Author
-
Youssef EM, Estecio MR, and Issa JP
- Subjects
- Base Sequence, Cell Division genetics, Cell Line, Tumor, DNA Methylation, DNA Primers, DNA, Neoplasm isolation & purification, Humans, Polymerase Chain Reaction, Promoter Regions, Genetic genetics, Colonic Neoplasms genetics, DNA, Neoplasm genetics, Gene Expression Regulation, Neoplastic genetics, Receptors, Retinoic Acid genetics
- Abstract
Tumor suppressor genes can become inactivated in cancer via hypermethylation of their promoter. The retinoic acid receptor beta (RARbeta) gene is expressed from two distinct promoters, both of which have CpG islands. RARbeta1 is expressed primarily during embryogenesis, whereas RARbeta2 is expressed in adult tissues and hypermethylated in a number of cancer cells. We used combined bisulfite restriction analysis to evaluate their methylation in colorectal mucosa and tumors. Methylation of RARbeta1 was detected, with a mean of 2% in normal colon tissues in young subjects (< 32 years), and 16% in older subjects (> 75 years) (P < 0.001). Using paired normal/tumor tissue samples, we found higher mean methylation rate in tumors than in adjacent normal tissue (mean, 46% versus 16%; P < 0.001) and hypermethylation of RARbeta1 in all eight cell lines examined. By RT-PCR, RARbeta1 was not expressed in normal adult colon tissues and its expression could not be efficiently activated in most cell lines by the DNA methyltransferase inhibitor 5-aza-2'-deoxycytidine (5-Aza-CdR). RARbeta2 methylation was also observed in normal colon tissues and was lower in young individuals than in older ones (mean, 11% versus 23%; P < 0.05). Among paired samples, RARbeta2 methylation was higher in tumor tissue than in normal tissue in 14 cases, vice versa in 7 cases, and equal in 6 cases. All eight cell lines were hypermethylated and did not express RARbeta2, but RARbeta2 expression could be reactivated easily by 5-Aza-CdR. We suggest that the embryonic RARbeta1 isoform is readily hypermethylated in aging colon mucosa and all colorectal cancers because of its lack of expression in normal tissues. The adult RARbeta2 isoform also shows age-related methylation in normal tissues but more variable methylation in colorectal cancer, perhaps because its expression offers continued protection against methylation or its silencing does not provide a selective advantage in the early stages of the disease.
- Published
- 2004
- Full Text
- View/download PDF
46. Comment on "Chromosomal instability and tumors promoted by DNA hypomethylation" and "Induction of tumors in nice by genomic hypomethylation".
- Author
-
Yang AS, Estecio MR, Garcia-Manero G, Kantarjian HM, and Issa JP
- Subjects
- Animals, Azacitidine adverse effects, Azacitidine therapeutic use, DNA (Cytosine-5-)-Methyltransferase 1, DNA (Cytosine-5-)-Methyltransferases antagonists & inhibitors, DNA (Cytosine-5-)-Methyltransferases metabolism, Decitabine, Genes, APC, Humans, Incidence, Intestinal Neoplasms epidemiology, Intestinal Neoplasms prevention & control, Mice, Neoplasms etiology, Neoplasms prevention & control, Point Mutation, Time Factors, Azacitidine analogs & derivatives, Chromosomal Instability, DNA Methylation, Neoplasms genetics
- Published
- 2003
- Full Text
- View/download PDF
47. [Chromosome abnormalities caused by computer video display monitors' radiation].
- Author
-
Estecio MR and Silva AE
- Subjects
- Adult, Cell Cycle physiology, Cell Cycle radiation effects, Chromosome Breakage, Female, Humans, Male, Chromosome Aberrations, Computer Terminals, Electromagnetic Fields adverse effects, Occupational Exposure adverse effects
- Abstract
Objective: Concerns were raised about the potential damaging effects of electromagnetic field (EMF) radiation emissions of ELF (extremely low frequency) and VLF (very low frequency) computer video display monitors (VDM), it was assessed the frequency of structural chromosome abnormalities and investigated the cell cycle kinetics in individuals occupationally exposed to VDM radiation., Methods: Chromosome aberrations were investigated in 2,000 first cell cycle metaphases obtained after 48-hour cultures of peripheral blood lymphocytes drawn from 10 individuals occupationally exposed to VDM radiation (group E) and 10 controls (group C). Cell cycle kinetics was studied using the mitotic index (MI) and cellular proliferation index (CPI)., Results: Statistical analysis showed significantly higher frequencies of anomalous metaphases (E=5.9%; C=3.7%) and anomaly/cell (E=0.066+/-0.026; C= 0.040+/-0.026) in individuals exposed to VDM radiation. The most common cytogenetic alterations seen were chromatid breaks at frequencies of 0.034+/-0.016 in group E and 0.016+/-0.015 in group C. There was no significant difference between MI and CPI frequencies in both groups., Conclusions: The study findings suggest genotoxic effects of EMF emissions revealed by the higher frequency of chromatid breaks in individuals exposed to VDM radiation. However, there is a need of further studies on EMF genetic effects using other investigation methods.
- Published
- 2002
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.