31 results on '"Rochelle L. Tiedemann"'
Search Results
2. The histone and non-histone methyllysine reader activities of the UHRF1 tandem Tudor domain are dispensable for the propagation of aberrant DNA methylation patterning in cancer cells
- Author
-
Robert M. Vaughan, Ariana Kupai, Caroline A. Foley, Cari A. Sagum, Bailey M. Tibben, Hope E. Eden, Rochelle L. Tiedemann, Christine A. Berryhill, Varun Patel, Kevin M. Shaw, Krzysztof Krajewski, Brian D. Strahl, Mark T. Bedford, Stephen V. Frye, Bradley M. Dickson, and Scott B. Rothbart
- Subjects
Genetics ,QH426-470 - Abstract
Abstract The chromatin-binding E3 ubiquitin ligase ubiquitin-like with PHD and RING finger domains 1 (UHRF1) contributes to the maintenance of aberrant DNA methylation patterning in cancer cells through multivalent histone and DNA recognition. The tandem Tudor domain (TTD) of UHRF1 is well-characterized as a reader of lysine 9 di- and tri-methylation on histone H3 (H3K9me2/me3) and, more recently, lysine 126 di- and tri-methylation on DNA ligase 1 (LIG1K126me2/me3). However, the functional significance and selectivity of these interactions remain unclear. In this study, we used protein domain microarrays to search for additional readers of LIG1K126me2, the preferred methyl state bound by the UHRF1 TTD. We show that the UHRF1 TTD binds LIG1K126me2 with high affinity and selectivity compared to other known methyllysine readers. Notably, and unlike H3K9me2/me3, the UHRF1 plant homeodomain (PHD) and its N-terminal linker (L2) do not contribute to multivalent LIG1K126me2 recognition along with the TTD. To test the functional significance of this interaction, we designed a LIG1K126me2 cell-penetrating peptide (CPP). Consistent with LIG1 knockdown, uptake of the CPP had no significant effect on the propagation of DNA methylation patterning across the genomes of bulk populations from high-resolution analysis of several cancer cell lines. Further, we did not detect significant changes in DNA methylation patterning from bulk cell populations after chemical or genetic disruption of lysine methyltransferase activity associated with LIG1K126me2 and H3K9me2. Collectively, these studies identify UHRF1 as a selective reader of LIG1K126me2 in vitro and further implicate the histone and non-histone methyllysine reader activity of the UHRF1 TTD as a dispensable domain function for cancer cell DNA methylation maintenance.
- Published
- 2020
- Full Text
- View/download PDF
3. Acute Depletion Redefines the Division of Labor among DNA Methyltransferases in Methylating the Human Genome
- Author
-
Rochelle L. Tiedemann, Emily L. Putiri, Jeong-Heon Lee, Ryan A. Hlady, Katsunobu Kashiwagi, Tamas Ordog, Zhiguo Zhang, Chen Liu, Jeong-Hyeon Choi, and Keith D. Robertson
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Global patterns of DNA methylation, mediated by the DNA methyltransferases (DNMTs), are disrupted in all cancers by mechanisms that remain largely unknown, hampering their development as therapeutic targets. Combinatorial acute depletion of all DNMTs in a pluripotent human tumor cell line, followed by epigenome and transcriptome analysis, revealed DNMT functions in fine detail. DNMT3B occupancy regulates methylation during differentiation, whereas an unexpected interplay was discovered in which DNMT1 and DNMT3B antithetically regulate methylation and hydroxymethylation in gene bodies, a finding confirmed in other cell types. DNMT3B mediated non-CpG methylation, whereas DNMT3L influenced the activity of DNMT3B toward non-CpG versus CpG site methylation. Altogether, these data reveal functional targets of each DNMT, suggesting that isoform selective inhibition would be therapeutically advantageous.
- Published
- 2014
- Full Text
- View/download PDF
4. Linking DNA Methyltransferases to Epigenetic Marks and Nucleosome Structure Genome-wide in Human Tumor Cells
- Author
-
Bilian Jin, Jason Ernst, Rochelle L. Tiedemann, Hongyan Xu, Suhas Sureshchandra, Manolis Kellis, Stephen Dalton, Chen Liu, Jeong-Hyeon Choi, and Keith D. Robertson
- Subjects
Biology (General) ,QH301-705.5 - Abstract
DNA methylation, mediated by the combined action of three DNA methyltransferases (DNMT1, DNMT3A, and DNMT3B), is essential for mammalian development and is a major contributor to cellular transformation. To elucidate how DNA methylation is targeted, we mapped the genome-wide localization of all DNMTs and methylation, and examined the relationships among these markers, histone modifications, and nucleosome structure in a pluripotent human tumor cell line in its undifferentiated and differentiated states. Our findings reveal a strong link between DNMTs and transcribed loci, and that DNA methylation is not a simple sum of DNMT localization patterns. A comparison of the epigenomes of normal and cancerous stem cells, and pluripotent and differentiated states shows that the presence of at least two DNMTs is strongly associated with loci targeted for DNA hypermethylation. Taken together, these results shed important light on the determinants of DNA methylation and how it may become disrupted in cancer cells.
- Published
- 2012
- Full Text
- View/download PDF
5. A physical basis for quantitative ChIP-sequencing
- Author
-
Robert M. Vaughan, Evan M. Cornett, Scott B. Rothbart, Rochelle L. Tiedemann, Bradley M. Dickson, and Alison A. Chomiak
- Subjects
0301 basic medicine ,Normalization (statistics) ,Chromatin Immunoprecipitation ,quantitative ChIP ,Computer science ,spike-in ,genetic processes ,Computational biology ,quantitative ChIP-Seq ,Biochemistry ,Epigenesis, Genetic ,03 medical and health sciences ,ChIP-Seq ,biophysics ,natural sciences ,Editors' Picks ,chromatin immunoprecipitation (ChiP) ,Molecular Biology ,030102 biochemistry & molecular biology ,Basis (linear algebra) ,epigenetics ,Scale (chemistry) ,antibody specificity ,ChIP normalization ,mathematical modeling ,Cell Biology ,ChIP-sequencing ,Sequence Analysis, DNA ,Chromatin ,030104 developmental biology ,Trustworthiness ,Chromatin Immunoprecipitation Sequencing - Abstract
ChIP followed by next-generation sequencing (ChIP-Seq) is a key technique for mapping the distribution of histone posttranslational modifications (PTMs) and chromatin-associated factors across genomes. There is a perceived challenge to define a quantitative scale for ChIP-Seq data, and as such, several approaches making use of exogenous additives, or "spike-ins," have recently been developed. Herein, we report on the development of a quantitative, physical model defining ChIP-Seq. The quantitative scale on which ChIP-Seq results should be compared emerges from the model. To test the model and demonstrate the quantitative scale, we examine the impacts of an EZH2 inhibitor through the lens of ChIP-Seq. We report a significant increase in immunoprecipitation of presumed off-target histone PTMs after inhibitor treatment, a trend predicted by the model but contrary to spike-in–based indications. Our work also identifies a sensitivity issue in spike-in normalization that has not been considered in the literature, placing limitations on its utility and trustworthiness. We call our new approach the sans-spike-in method for quantitative ChIP-sequencing (siQ-ChIP). A number of changes in community practice of ChIP-Seq, data reporting, and analysis are motivated by this work.
- Published
- 2020
6. The Human Epigenome
- Author
-
Rochelle L. Tiedemann, Gangning Liang, and Peter A. Jones
- Published
- 2022
- Full Text
- View/download PDF
7. Chromosome-specific retention of cancer-associated DNA hypermethylation following pharmacological inhibition of DNMT1
- Author
-
Ashley K. Wiseman, Rochelle L. Tiedemann, Huihui Fan, Hui Shen, Zachary Madaj, Michael T. McCabe, Melissa B. Pappalardi, and Peter A. Jones
- Subjects
Male ,X Chromosome ,Neoplasms ,Medicine (miscellaneous) ,Humans ,CpG Islands ,Female ,DNA ,DNA Methylation ,General Agricultural and Biological Sciences ,General Biochemistry, Genetics and Molecular Biology - Abstract
The DNA methylation status of the X-chromosome in cancer cells is often overlooked because of computational difficulties. Most of the CpG islands on the X-chromosome are mono-allelically methylated in normal female cells and only present as a single copy in male cells. We treated two colorectal cancer cell lines from a male (HCT116) and a female (RKO) with increasing doses of a DNA methyltransferase 1 (DNMT1)-specific inhibitor (GSK3685032/GSK5032) over several months to remove as much non-essential CpG methylation as possible. Profiling of the remaining DNA methylome revealed an unexpected, enriched retention of DNA methylation on the X-chromosome. Strikingly, the identified retained X-chromosome DNA methylation patterns accurately predicted de novo DNA hypermethylation in colon cancer patient methylomes in the TCGA COAD/READ cohort. These results suggest that a re-examination of tumors for X-linked DNA methylation changes may enable greater understanding of the importance of epigenetic silencing of cancer related genes.
- Published
- 2021
8. Oocyte age and preconceptual alcohol use are highly correlated with epigenetic imprinting of a noncoding RNA ( nc886 )
- Author
-
Peter A. Jones, Hein J. Odendaal, Brittany L. Carpenter, Lucy Brink, Stacey L. Thomas, Zachary Madaj, Tanaka K Remba, and Rochelle L. Tiedemann
- Subjects
RNA, Untranslated ,Alcohol Drinking ,Population ,Biology ,Epigenesis, Genetic ,Genomic Imprinting ,nc886 ,Pregnancy ,Humans ,Epigenetics ,Allele ,Imprinting (psychology) ,oocyte ,education ,Gene ,Alleles ,Genetics ,education.field_of_study ,Multidisciplinary ,Population Biology ,High-Throughput Nucleotide Sequencing ,Epigenome ,Biological Sciences ,DNA Methylation ,Maternal Exposure ,DNA methylation ,Oocytes ,CpG Islands ,Female ,imprinting ,Genomic imprinting ,Maternal Age - Abstract
Significance Genomic imprinting is essential for human development and occurs in germ cells before fertilization. The noncoding RNA, nc886, is the only known example of more than 100 such human genes which shows variable frequencies of maternal imprinting. Here, we show that the DNA methylation imprint is present in oocytes and that the probability of imprinting increases as a function of maternal age. Importantly, we demonstrate that alcohol consumption but not cigarette smoking is associated with a lower frequency of imprinting. While most studies focus on the postconceptional developmental time, our work indicates that maternal age and exposures the year prior to pregnancy may alter the epigenome and therefore the developing child., Genomic imprinting occurs before fertilization, impacts every cell of the developing child, and may be sensitive to environmental perturbations. The noncoding RNA, nc886 (also called VTRNA2-1) is the only known example of the ∼100 human genes imprinted by DNA methylation, that shows polymorphic imprinting in the population. The nc886 gene is part of an ∼1.6-kb differentially methylated region (DMR) that is methylated in the oocyte and silenced on the maternal allele in about 75% of humans worldwide. Here, we show that the presence or absence of imprinting at the nc886 DMR in an individual is consistent across different tissues, confirming that the imprint is established before cellular differentiation and is maintained into adulthood. We investigated the relationships between the frequency of imprinting in newborns and maternal age, alcohol consumption and cigarette smoking before conception in more than 1,100 mother/child pairs from South Africa. The probability of imprinting in newborns was increased in older mothers and decreased in mothers who drank alcohol before conception. On the other hand, cigarette smoking had no apparent relationship with the frequency of imprinting. These data show an epigenetic change during oocyte maturation which is potentially subject to environmental influence. Much focus has been placed on avoiding alcohol consumption during pregnancy, but our data suggest that drinking before conception may affect the epigenome of the newborn.
- Published
- 2021
- Full Text
- View/download PDF
9. Distinguishing Active Versus Passive DNA Demethylation Using Illumina MethylationEPIC BeadChip Microarrays
- Author
-
Hope E. Eden, Zhijun Huang, Keith D. Robertson, Rochelle L. Tiedemann, and Scott B. Rothbart
- Subjects
Bisulfite ,genomic DNA ,Cytosine nucleotide ,DNA demethylation ,CpG site ,Chemistry ,DNA methylation ,Computational biology ,Epigenetics ,DNA microarray ,Article - Abstract
The 5-carbon positions on cytosine nucleotides preceding guanines in genomic DNA (CpG) are common targets for DNA methylation (5mC). DNA methylation removal can occur through both active and passive mechanisms. Ten-eleven translocation enzymes (TETs) oxidize 5mC in a stepwise manner to 5-hydroxymethylcytosine (5hmC), 5-formylcytosine (5fC), and 5-carboxylcytosine (5caC). 5mC can also be removed passively through sequential cell divisions in the absence of DNA methylation maintenance. In this chapter, we describe approaches that couple TET-assisted bisulfite (TAB) and oxidative bisulfite (OxBS) conversion to the Illumina MethylationEPIC BeadChIP (EPIC array) and show how these technologies can be used to distinguish active versus passive DNA demethylation. We also describe integrative bioinformatics pipelines to facilitate this analysis.
- Published
- 2021
- Full Text
- View/download PDF
10. In silico APC/C substrate discovery reveals cell cycle-dependent degradation of UHRF1 and other chromatin regulators
- Author
-
Derek L. Bolhuis, Aussie Suzuki, Taylor P. Enrico, Rajarshi Choudhury, Thomas Bonacci, Ryan D. Mouery, Jennifer L. Franks, Jeffrey S. Damrauer, Michael J. Emanuele, Feng Yan, M. Ben Major, Xianxi Wang, Nicholas G. Brown, Katherine A. Hoadley, Rochelle L. Tiedemann, Raquel C. Martinez-Chacin, Scott B. Rothbart, and Joseph S. Harrison
- Subjects
0301 basic medicine ,Cell Cycle Proteins ,Biochemistry ,0302 clinical medicine ,Ubiquitin ,Cell Cycle and Cell Division ,Biology (General) ,Cyclin ,DNA methylation ,biology ,General Neuroscience ,Cell Cycle ,Cell cycle ,Small interfering RNA ,Chromatin ,Ubiquitin ligase ,Cell biology ,Nucleic acids ,Cell Processes ,Epigenetics ,General Agricultural and Biological Sciences ,DNA modification ,Chromatin modification ,Research Article ,Chromosome biology ,QH301-705.5 ,Ubiquitin-Protein Ligases ,Immunoblotting ,Mitosis ,Molecular Probe Techniques ,Research and Analysis Methods ,Transfection ,General Biochemistry, Genetics and Molecular Biology ,Anaphase-Promoting Complex-Cyclosome ,Cell Line ,03 medical and health sciences ,Cyclins ,Genetics ,Humans ,Computer Simulation ,Molecular Biology Techniques ,Non-coding RNA ,Molecular Biology ,General Immunology and Microbiology ,Biology and life sciences ,Ubiquitination ,DNA ,Gene regulation ,030104 developmental biology ,HEK293 Cells ,Mitotic exit ,biology.protein ,CCAAT-Enhancer-Binding Proteins ,RNA ,Gene expression ,Protein Processing, Post-Translational ,030217 neurology & neurosurgery ,HeLa Cells ,Transcription Factors - Abstract
The anaphase-promoting complex/cyclosome (APC/C) is an E3 ubiquitin ligase and critical regulator of cell cycle progression. Despite its vital role, it has remained challenging to globally map APC/C substrates. By combining orthogonal features of known substrates, we predicted APC/C substrates in silico. This analysis identified many known substrates and suggested numerous candidates. Unexpectedly, chromatin regulatory proteins are enriched among putative substrates, and we show experimentally that several chromatin proteins bind APC/C, oscillate during the cell cycle, and are degraded following APC/C activation, consistent with being direct APC/C substrates. Additional analysis revealed detailed mechanisms of ubiquitylation for UHRF1, a key chromatin regulator involved in histone ubiquitylation and DNA methylation maintenance. Disrupting UHRF1 degradation at mitotic exit accelerates G1-phase cell cycle progression and perturbs global DNA methylation patterning in the genome. We conclude that APC/C coordinates crosstalk between cell cycle and chromatin regulatory proteins. This has potential consequences in normal cell physiology, where the chromatin environment changes depending on proliferative state, as well as in disease., This study shows that the cell cycle E3 ubiquitin ligase APC/C is a regulator of several chromatin regulatory proteins, including the multivalent epigenetic reader and writer UHRF1. Perturbing UHRF1 ubiquitylation and degradation alters cell cycle and DNA methylation patterning, pointing to a key role for cell cycle degradation in shaping chromatin environments.
- Published
- 2020
11. The histone and non-histone methyllysine reader activities of the UHRF1 tandem Tudor domain are dispensable for the propagation of aberrant DNA methylation patterning in cancer cells
- Author
-
Varun Patel, Bailey M. Tibben, Stephen V. Frye, Rochelle L. Tiedemann, Robert M. Vaughan, Christine A. Berryhill, Krzysztof Krajewski, Cari A. Sagum, Caroline A. Foley, Brian D. Strahl, Ariana Kupai, Scott B. Rothbart, Mark T. Bedford, Kevin M. Shaw, Hope E. Eden, and Bradley M. Dickson
- Subjects
Tudor domain ,Methyltransferase ,lcsh:QH426-470 ,Ubiquitin-Protein Ligases ,Methylation ,Epigenesis, Genetic ,Histones ,03 medical and health sciences ,Methyllysine ,chemistry.chemical_compound ,Histone H3 ,0302 clinical medicine ,Genetics ,Humans ,Molecular Biology ,030304 developmental biology ,chemistry.chemical_classification ,0303 health sciences ,DNA ligase ,biology ,Tudor Domain ,Research ,Lysine ,DNA Methylation ,HCT116 Cells ,Ubiquitin ligase ,Cell biology ,Gene Expression Regulation, Neoplastic ,Histone Code ,lcsh:Genetics ,Histone ,chemistry ,DNA methylation ,biology.protein ,CCAAT-Enhancer-Binding Proteins ,Protein Processing, Post-Translational ,030217 neurology & neurosurgery ,HeLa Cells - Abstract
The chromatin-binding E3 ubiquitin ligase ubiquitin-like with PHD and RING finger domains 1 (UHRF1) contributes to the maintenance of aberrant DNA methylation patterning in cancer cells through multivalent histone and DNA recognition. The tandem Tudor domain (TTD) of UHRF1 is well-characterized as a reader of lysine 9 di- and tri-methylation on histone H3 (H3K9me2/me3) and, more recently, lysine 126 di- and tri-methylation on DNA ligase 1 (LIG1K126me2/me3). However, the functional significance and selectivity of these interactions remain unclear. In this study, we used protein domain microarrays to search for additional readers of LIG1K126me2, the preferred methyl state bound by the UHRF1 TTD. We show that the UHRF1 TTD binds LIG1K126me2 with high affinity and selectivity compared to other known methyllysine readers. Notably, and unlike H3K9me2/me3, the UHRF1 plant homeodomain (PHD) and its N-terminal linker (L2) do not contribute to multivalent LIG1K126me2 recognition along with the TTD. To test the functional significance of this interaction, we designed a LIG1K126me2 cell-penetrating peptide (CPP). Consistent with LIG1 knockdown, uptake of the CPP had no significant effect on the propagation of DNA methylation patterning across the genomes of bulk populations from high-resolution analysis of several cancer cell lines. Further, we did not detect significant changes in DNA methylation patterning from bulk cell populations after chemical or genetic disruption of lysine methyltransferase activity associated with LIG1K126me2 and H3K9me2. Collectively, these studies identify UHRF1 as a selective reader of LIG1K126me2 in vitro and further implicate the histone and non-histone methyllysine reader activity of the UHRF1 TTD as a dispensable domain function for cancer cell DNA methylation maintenance.
- Published
- 2020
12. In silicoAPC/C substrate discovery reveals cell cycle degradation of chromatin regulators including UHRF1
- Author
-
Michael B. Major, Feng Yan, Derek L. Bolhuis, Rajarshi Choudhury, Thomas Bonacci, Michael J. Emanuele, Raquel C. Martinez-Chacin, Scott B. Rothbart, Rochelle L. Tiedemann, Nicholas G. Brown, Joseph S. Harrison, Jennifer L. Kernan, Xianxi Wang, Katherine A. Hoadley, Jeffrey S. Damrauer, and Aussie Suzuki
- Subjects
Ubiquitin ,biology ,Mitotic exit ,Chemistry ,In silico ,DNA methylation ,biology.protein ,Regulator ,Cell cycle ,Ubiquitin ligase ,Cell biology ,Chromatin - Abstract
The Anaphase-Promoting Complex/Cyclosome (APC/C) is an E3 ubiquitin ligase and critical regulator of cell cycle progression. Despite its vital role, it has remained challenging to globally map APC/C substrates. By combining orthogonal features of known substrates, we predicted APC/C substratesin silico. This analysis identified many known substrates and suggested numerous candidates. Unexpectedly, chromatin regulatory proteins are enriched among putative substrates and we show that several chromatin proteins bind APC/C, oscillate during the cell cycle and are degraded following APC/C activation, consistent with being direct APC/C substrates. Additional analysis revealed detailed mechanisms of ubiquitylation for UHRF1, a key chromatin regulator involved in histone ubiquitylation and DNA methylation maintenance. Disrupting UHRF1 degradation at mitotic exit accelerates G1-phase cell cycle progression and perturbs global DNA methylation patterning in the genome. We conclude that APC/C coordinates crosstalk between cell cycle and chromatin regulatory proteins. This has potential consequences in normal cell physiology, where the chromatin environment changes depending on proliferative state, as well as in disease.
- Published
- 2020
- Full Text
- View/download PDF
13. siQ-ChIP: A reverse-engineered quantitative framework for ChIP-sequencing
- Author
-
Robert M. Vaughan, Bradley M. Dickson, Scott B. Rothbart, Rochelle L. Tiedemann, Alison A. Chomiak, and Evan M. Cornett
- Subjects
Reverse engineering ,0303 health sciences ,biology ,010405 organic chemistry ,Computer science ,genetic processes ,Computational biology ,Chip ,computer.software_genre ,01 natural sciences ,Genome ,Plot (graphics) ,0104 chemical sciences ,ChIP-sequencing ,03 medical and health sciences ,Histone ,biology.protein ,natural sciences ,Chromatin immunoprecipitation ,computer ,030304 developmental biology - Abstract
Chromatin immunoprecipitation followed by next-generation sequencing (ChIP-seq) is a key technique for mapping the distribution and relative abundance of histone posttranslational modifications (PTMs) and chromatin-associated factors across genomes. There is a perceived challenge regarding the ability to quantitatively plot ChIP-seq data, and as such, approaches making use of exogenous additives, or “spike-ins” have recently been developed. Relying on the fact that the IP step of ChIP-seq is a competitive binding reaction, we present a quantitative framework for ChIP-seq analysis that circumvents the need to modify standard sample preparation pipelines with spike-in reagents. We also introduce a visualization technique that, when paired with our formal developments, produces a much more rich characterization of sequencing data.
- Published
- 2019
- Full Text
- View/download PDF
14. Defining UHRF1 Domains That Support Maintenance of Human Colon Cancer DNA Methylation and Oncogenic Properties
- Author
-
Michael J. Topper, Daiming Fan, Wenbing Xie, Cynthia A. Zahnow, Jie Chen, Xiangqian Kong, Ray Whay Chiu Yen, Rochelle L. Tiedemann, Srinivasan Yegnasubramanian, Hariharan Easwaran, Stephen M. Brown, Scott B. Rothbart, Limin Xia, Kaichun Wu, Stephen B. Baylin, Yongzhan Nie, Yi Cai, and Yong Tao
- Subjects
0301 basic medicine ,Male ,Cancer Research ,Time Factors ,Colorectal cancer ,Mice, SCID ,law.invention ,Epigenesis, Genetic ,Histones ,chemistry.chemical_compound ,0302 clinical medicine ,law ,Mice, Inbred NOD ,Neoplasm Metastasis ,Mice, Inbred BALB C ,Prognosis ,Chromatin ,Ubiquitin ligase ,Gene Expression Regulation, Neoplastic ,Histone ,Oncology ,030220 oncology & carcinogenesis ,DNA methylation ,Female ,Colorectal Neoplasms ,HT29 Cells ,Ubiquitin-Protein Ligases ,Mice, Nude ,Biology ,Article ,03 medical and health sciences ,medicine ,Animals ,Humans ,Gene ,DNA Methylation ,medicine.disease ,HCT116 Cells ,Mice, Inbred C57BL ,030104 developmental biology ,chemistry ,Mutation ,biology.protein ,Cancer research ,CCAAT-Enhancer-Binding Proteins ,Suppressor ,CpG Islands ,Caco-2 Cells ,PHD Zinc Fingers ,DNA - Abstract
Summary UHRF1 facilitates the establishment and maintenance of DNA methylation patterns in mammalian cells. The establishment domains are defined, including E3 ligase function, but the maintenance domains are poorly characterized. Here, we demonstrate that UHRF1 histone- and hemimethylated DNA binding functions, but not E3 ligase activity, maintain cancer-specific DNA methylation in human colorectal cancer (CRC) cells. Disrupting either chromatin reader activity reverses DNA hypermethylation, reactivates epigenetically silenced tumor suppressor genes (TSGs), and reduces CRC oncogenic properties. Moreover, an inverse correlation between high UHRF1 and low TSG expression tracks with CRC progression and reduced patient survival. Defining critical UHRF1 domain functions and its relationship with CRC prognosis suggests directions for, and value of, targeting this protein to develop therapeutic DNA demethylating agents.
- Published
- 2019
15. High‐resolution Epigenome Mapping Reveals Distinct and Divergent Roles for UHRF1 in the Maintenance of DNA Methylation
- Author
-
Alison A. Chomiak, Peter A. Jones, Bradley M. Dickson, Stephen B. Baylin, Wanding Zhou, Susan J. Clark, Qian Du, Benjamin K. Johnson, Xiangqian Kong, Rochelle L. Tiedemann, and Scott B. Rothbart
- Subjects
DNA methylation ,Genetics ,High resolution ,Epigenome ,Computational biology ,Biology ,Molecular Biology ,Biochemistry ,Biotechnology - Published
- 2020
- Full Text
- View/download PDF
16. Epigenomic Reprogramming toward Mesenchymal-Epithelial Transition in Ovarian-Cancer-Associated Mesenchymal Stem Cells Drives Metastasis
- Author
-
Huda Atiya, Huihui Fan, Kelly K. Foy, Thomas R. Pisanic, Ronald J. Buckanovich, Leonard Frisbie, Alison A. Chomiak, Ie Ming Shih, Scott B. Rothbart, Chelsea Chandler, Tza-Huei Wang, Rochelle L. Tiedemann, Hui Shen, Yeh Wang, and Lan G. Coffman
- Subjects
0301 basic medicine ,Epigenomics ,Epithelial-Mesenchymal Transition ,Primary Cell Culture ,Gene Expression ,Biology ,Carcinoma, Ovarian Epithelial ,General Biochemistry, Genetics and Molecular Biology ,Article ,Metastasis ,03 medical and health sciences ,Epigenome ,Mice ,0302 clinical medicine ,Cell Movement ,Mice, Inbred NOD ,Cell Line, Tumor ,medicine ,Mesenchymal–epithelial transition ,Animals ,Humans ,Enhancer of Zeste Homolog 2 Protein ,Neoplasm Metastasis ,WT1 Proteins ,Cell Proliferation ,Ovarian Neoplasms ,Tumor microenvironment ,Mesenchymal stem cell ,Cell Differentiation ,Mesenchymal Stem Cells ,medicine.disease ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,Cancer cell ,Cancer research ,Female ,Ovarian cancer ,Reprogramming ,030217 neurology & neurosurgery ,Signal Transduction - Abstract
SUMMARY A role for cancer cell epithelial-to-mesenchymal transition (EMT) in cancer is well established. Here, we show that, in addition to cancer cell EMT, ovarian cancer cell metastasis relies on an epigenomic mesenchymal-to-epithelial transition (MET) in host mesenchymal stem cells (MSCs). These reprogrammed MSCs, termed carcinoma-associated MSCs (CA-MSCs), acquire pro-tumorigenic functions and directly bind cancer cells to serve as a metastatic driver/chaperone. Cancer cells induce this epigenomic MET characterized by enhancer-enriched DNA hypermethylation, altered chromatin accessibility, and differential histone modifications. This phenomenon appears clinically relevant, as CA-MSC MET is highly correlated with patient survival. Mechanistically, mirroring MET observed in development, MET in CA-MSCs is mediated by WT1 and EZH2. Importantly, EZH2 inhibitors, which are clinically available, significantly inhibited CA-MSC-mediated metastasis in mouse models of ovarian cancer., Graphical Abstract, In Brief Fan et al. demonstrate that ovarian cancer reprograms the epigenome of stromal cells, inducing a mesenchymal-to-epithelial transition (MET) to form carcinoma-associated mesenchymal stem cells (CA-MSCs). This MET, mediated by WT1 and EZH2, enables CA-MSC:cancer cell co-metastasis. EZH2 inhibition decreases CA-MSC-mediated metastasis, presenting a potential therapeutic opportunity in ovarian cancer.
- Published
- 2020
17. Examining the Roles of H3K4 Methylation States with Systematically Characterized Antibodies
- Author
-
Alexander J. Ruthenburg, Brandon A. Boone, Rohan N. Shah, Neha Arora, Martis W. Cowles, Zu-Wen Sun, Robert M. Vaughan, Danielle Maryanski, Andrea L. Johnstone, Rochelle L. Tiedemann, Michael-Christopher Keogh, Adrian T. Grzybowski, Marcus A. Cheek, Bradley M. Dickson, Scott B. Rothbart, Matthew J. Meiners, and Evan M. Cornett
- Subjects
0301 basic medicine ,Chromatin Immunoprecipitation ,Computational biology ,Biology ,Methylation ,Antibodies ,Article ,Histones ,03 medical and health sciences ,chemistry.chemical_compound ,Transcription (biology) ,Antibody Specificity ,Heterochromatin ,Nucleosome ,Humans ,Enhancer ,Promoter Regions, Genetic ,Molecular Biology ,Cell Biology ,Chromatin ,Nucleosomes ,Histone Code ,030104 developmental biology ,Histone ,chemistry ,biology.protein ,Antibody ,Chromatin immunoprecipitation ,Protein Processing, Post-Translational ,DNA - Abstract
Histone post-translational modifications (PTMs) are important genomic regulators often studied by chromatin immunoprecipitation (ChIP), whereby their locations and relative abundance are inferred by antibody capture of nucleosomes and associated DNA. However, the specificity of antibodies within these experiments have not been systematically studied. Here, we use histone peptide arrays and internally calibrated ChIP (ICeChIP) to characterize 52 commercial antibodies purported to distinguish the H3K4 methylforms (me1, me2, and me3, with each ascribed distinct biological functions). We find that many widely-used antibodies poorly distinguish the methylforms and that high- and low-specificity reagents can yield dramatically different biological interpretations, resulting in substantial divergence from the literature for numerous H3K4 methylform paradigms. Using ICeChIP, we also discern quantitative relationships between enhancer H3K4 methylation and promoter transcriptional output and can measure global PTM abundance changes. Our results illustrate how poor antibody specificity contributes to the “reproducibility crisis,” demonstrating the need for rigorous, platform-appropriate validation.
- Published
- 2018
18. Epigenetic signatures of alcohol abuse and hepatitis infection during human hepatocarcinogenesis
- Author
-
Ryan A. Hlady, Lewis R. Roberts, Jeong Hyeon Choi, Rochelle L. Tiedemann, Ivan Zendejas, William Puszyk, Chen Liu, and Keith D. Robertson
- Subjects
Liver Cirrhosis ,Male ,Carcinoma, Hepatocellular ,Carcinogenesis ,etiology ,medicine.disease_cause ,Epigenesis, Genetic ,Hepatitis ,03 medical and health sciences ,0302 clinical medicine ,Cell Line, Tumor ,medicine ,Humans ,Epigenetics ,030304 developmental biology ,0303 health sciences ,DNA methylation ,epigenetics ,business.industry ,cirrhosis ,Liver Neoplasms ,Cancer ,hepatocellular carcinoma ,Epigenome ,Hepatitis B ,medicine.disease ,Hepatitis C ,digestive system diseases ,3. Good health ,Alcoholism ,Liver ,Oncology ,030220 oncology & carcinogenesis ,Hepatocellular carcinoma ,Immunology ,Cancer research ,Female ,business ,Research Paper - Abstract
// Ryan A. Hlady 1 , Rochelle L. Tiedemann 1,2 , William Puszyk 3 , Ivan Zendejas 4 , Lewis R. Roberts 5 , Jeong-Hyeon Choi 2 , Chen Liu 3 and Keith D. Robertson 1 1 Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA 2 Cancer Center, Georgia Regents University, Augusta, GA, USA 3 Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL, USA 4 Department of Surgery, University of Florida, Gainesville, FL, USA 5 Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA Correspondence: Keith D. Robertson, email: // Chen Liu, email: // Keywords : hepatocellular carcinoma, cirrhosis, etiology, epigenetics, DNA methylation Received : August 28, 2014 Accepted : September 07, 2014 Published : September 08, 2014 Abstract Hepatocellular carcinoma (HCC) is the second most common cause of cancer deaths worldwide. Deregulated DNA methylation landscapes are ubiquitous in human cancers. Interpretation of epigenetic aberrations in HCC is confounded by multiple etiologic drivers and underlying cirrhosis. We globally profiled the DNA methylome of 34 normal and 122 liver disease tissues arising in settings of hepatitis B (HBV) or C (HCV) viral infection, alcoholism (EtOH), and other causes to examine how these environmental agents impact DNA methylation in a manner that contributes to liver disease. Our results demonstrate that each ‘exposure’ leaves unique and overlapping signatures on the methylome. CpGs aberrantly methylated in cirrhosis-HCV and conserved in HCC were enriched for cancer driver genes, suggesting a pathogenic role for HCV-induced methylation changes. Additionally, large genomic regions displaying stepwise hypermethylation or hypomethylation during disease progression were identified. HCC-HCV/EtOH methylomes overlap highly with cryptogenic HCC, suggesting shared epigenetically deregulated pathways for hepatocarcinogenesis. Finally, overlapping methylation abnormalities between primary and cultured tumors unveil conserved epigenetic signatures in HCC. Taken together, this study reveals profound epigenome deregulation in HCC beginning during cirrhosis and influenced by common environmental agents. These results lay the foundation for defining epigenetic drivers and clinically useful methylation markers for HCC.
- Published
- 2014
- Full Text
- View/download PDF
19. Impact of human MLL/COMPASS and polycomb complexes on the DNA methylome
- Author
-
Rochelle L. Tiedemann, Emily L. Putiri, Jeong Hyeon Choi, Chunsheng Liu, and Keith D. Robertson
- Subjects
Gene Expression ,Cell Cycle Proteins ,macromolecular substances ,Biology ,Cell Line, Tumor ,Histone methylation ,Histone H2A ,Humans ,cancer ,Histone code ,histone modification ,histone methylation ,Cancer epigenetics ,RNA-Directed DNA Methylation ,Epigenomics ,Genetics ,DNA methylation ,epigenetics ,Polycomb Repressive Complex 2 ,Histone-Lysine N-Methyltransferase ,3. Good health ,Oncology ,Histone methyltransferase ,Protein Processing, Post-Translational ,Myeloid-Lymphoid Leukemia Protein ,Protein Binding ,Research Paper - Abstract
// Emily L. Putiri 1 , Rochelle L. Tiedemann 1,2 , Chunsheng Liu 1 , Jeong-Hyeon Choi 2 and Keith D. Robertson 1 1 Department of Molecular Pharmacology and Experimental Therapeutics and Center for Individualized Medicine, Mayo Clinic, Rochester, MN 2 Cancer Center, Georgia Regents University, Augusta, GA Correspondence: Keith D. Robertson, email: // Keywords : DNA methylation, epigenetics, cancer, histone modification, histone methylation Received : May 05, 2014 Accepted : July 13, 2014 Published : July 14, 2014 Abstract The correlation between DNA methylation and a subset of histone post-translational modifications (positive and negative) has hinted at an underlying regulatory crosstalk between histone marks and DNA methylation in patterning the human DNA methylome, an idea further supported by corresponding alterations to both histone marks and DNA methylation during malignant transformation. This study investigated the framework by which histone marks influence DNA methylation at a genome-wide level. Using RNAi in a pluripotent human embryonic carcinoma cell line we depleted essential components of the MLL/COMPASS, polycomb repressive complex 2 (PRC2), and PRC1 histone modifying complexes that establish, respectively, the post-translational modifications H3K4me3, H3K27me3, and H2AK119ub, and assayed the impact of the subsequent depletion of these marks on the DNA methylome. Absence of H2AK119ub resulted predominantly in hypomethylation across the genome. Depletion of H3K4me3 and, surprisingly, H3K27me3 caused CpG island hypermethylation at a subset of loci. Intriguingly, many promoters were co-regulated by all three histone marks, becoming hypermethylated with loss of H3K4me3 or H3K27me3 and hypomethylated with depletion of H2AK119ub, and many of these co-regulated loci were among those commonly targeted for aberrant hypermethylation in cancer. Taken together, our results elucidate novel roles for polycomb and MLL/COMPASS in regulating DNA methylation and define targets of this regulation.
- Published
- 2014
- Full Text
- View/download PDF
20. Abstract 2628: Non-canonical Wnt signaling in late-stage PDAC
- Author
-
Payton D. Stevens, Adam Racette, Rochelle L. Tiedemann, and Bart O. Williams
- Subjects
Cancer Research ,Cell signaling ,Frizzled ,Microarray analysis techniques ,Cell ,Wnt signaling pathway ,Cancer ,Biology ,medicine.disease ,medicine.anatomical_structure ,Oncology ,Pancreatic cancer ,Cancer research ,medicine ,Receptor - Abstract
Pancreatic cancer has a disproportionate death-to-incidence rate largely resultant from late stage diagnoses, leading to advanced cancers which have metastasized and have increased resistance to common chemotherapeutics. This malignant progression is characterized by the increase of many molecular markers, and developing novel therapeutics targeting these late stage markers is essential for improving patient prognoses. We have identified several proteins within the non-canonical Wnt pathway that are upregulated in advanced pancreatic ductal adenocarcinomas (PDAC), and we are exploring the mechanisms driving upregulation and the resulting alterations to cell signaling. Analysis of microarray data from human derived organoids (from 6 normal pancreas samples and 38 tumor samples) revealed that two receptors in the Wnt pathway, Frizzled 2 (FZD2) and Frizzled 6 (FZD6), have increased expression levels in late-stage PDAC. The temporal increase of these non-canonical, planar cell polarity altering receptors suggests a previously underappreciated role for Wnt signaling in the progression of pancreatic cancer. The increase of these two receptors in tumor tissue was confirmed through analysis of RNA-sequencing of patient samples, including 179 tumor and 171 normal samples. Interestingly, after analyzing patient overall survival and progression free survival we found that, out of the ten members of the Frizzled receptor family, higher levels of FZD2 and FZD6 result in the worst patient prognoses. Additionally, retrospective analysis of gene expression data from Moffitt et al. suggests that FZD2 and FZD6 are higher in the aggressive “basal-like” sub-category of pancreatic cancer. To begin to understand the mechanism for these changes we have used a panel of PDAC cell lines, grown in both traditional and 3-dimensional culture. We have found that decreasing levels of FZD2 and FZD6, through stable sh-RNA knockdown, results in cells which are less motile (observed via Transwell and scratch assays) and are also less capable of anchorage independent growth. Also, cell lines with increased levels of FZD2 result in cells migrating through 3D-matrix as small cell clusters rather than as single cells, which has also been observed in partial-epithelial to mesenchymal transition cell lines. Taken together, non-canonical Wnt signaling in PDAC, mediated by FZD2 and/or FZD6 receptors, results in phenotypic alterations which are consistent with those often observed during cancer progression. These changes may help explain why patients with increased expression of FZD2 and FZD6 have worse prognoses and may also warrant more exploration into specific Frizzled inhibitors. Citation Format: Payton D. Stevens, Adam Racette, Rochelle L. Tiedemann, Bart O. Williams. Non-canonical Wnt signaling in late-stage PDAC [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 2628.
- Published
- 2019
- Full Text
- View/download PDF
21. Nucleosome positioning changes during human embryonic stem cell differentiation
- Author
-
Shaying Zhao, Rochelle L. Tiedemann, Michael Kulik, Keith D. Robertson, Wenjuan Zhang, Yaping Li, and Stephen Dalton
- Subjects
0301 basic medicine ,Cancer Research ,Cellular differentiation ,Myocytes, Smooth Muscle ,Biology ,Cell Line ,03 medical and health sciences ,Nucleosome ,Humans ,Promoter Regions, Genetic ,Molecular Biology ,Gene ,Embryonic Stem Cells ,Genetics ,Base Composition ,Intron ,Gene Expression Regulation, Developmental ,Promoter ,Cell Differentiation ,Chromatin Assembly and Disassembly ,Embryonic stem cell ,Chromatin ,Nucleosomes ,030104 developmental biology ,embryonic structures ,Human genome ,Research Paper - Abstract
Nucleosomes are the basic unit of chromatin. Nucleosome positioning (NP) plays a key role in transcriptional regulation and other biological processes. To better understand NP we used MNase-seq to investigate changes that occur as human embryonic stem cells (hESCs) transition to nascent mesoderm and then to smooth muscle cells (SMCs). Compared to differentiated cell derivatives, nucleosome occupancy at promoters and other notable genic sites, such as exon/intron junctions and adjacent regions, in hESCs shows a stronger correlation with transcript abundance and is less influenced by sequence content. Upon hESC differentiation, genes being silenced, but not genes being activated, display a substantial change in nucleosome occupancy at their promoters. Genome-wide, we detected a shift of NP to regions of higher G+C content as hESCs differentiate to SMCs. Notably, genomic regions with higher nucleosome occupancy harbor twice as many G↔C changes but fewer than half A↔T changes, compared to regions with lower nucleosome occupancy. Finally, our analysis indicates that the hESC genome is not rearranged and has a sequence mutation rate resembling normal human genomes. Our study reveals another unique feature of hESC chromatin, and sheds light on the relationship between nucleosome occupancy and sequence G+C content.
- Published
- 2016
22. Linking DNA Methyltransferases to Epigenetic Marks and Nucleosome Structure Genome-wide in Human Tumor Cells
- Author
-
Stephen Dalton, Suhas Sureshchandra, Jason Ernst, Jeong Hyeon Choi, Rochelle L. Tiedemann, Bilian Jin, Chen Liu, Hongyan Xu, Manolis Kellis, Keith D. Robertson, Massachusetts Institute of Technology. Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology. Department of Electrical Engineering and Computer Science, Ernst, Jason, and Kellis, Manolis
- Subjects
DNA (Cytosine-5-)-Methyltransferase 1 ,Epigenomics ,Male ,Transcription, Genetic ,Biology ,Article ,General Biochemistry, Genetics and Molecular Biology ,Cell Line ,DNA Methyltransferase 3A ,Histones ,03 medical and health sciences ,0302 clinical medicine ,Testicular Neoplasms ,Carcinoma, Embryonal ,Histone methylation ,Genes, Overlapping ,Humans ,Nucleosome ,DNA (Cytosine-5-)-Methyltransferases ,Epigenetics ,Cancer epigenetics ,RNA-Directed DNA Methylation ,lcsh:QH301-705.5 ,Embryonic Stem Cells ,030304 developmental biology ,Genetics ,0303 health sciences ,Genome, Human ,1. No poverty ,DNA ,DNA Methylation ,HCT116 Cells ,Chromatin ,Nucleosomes ,3. Good health ,Histone ,lcsh:Biology (General) ,Genetic Loci ,030220 oncology & carcinogenesis ,DNA methylation ,biology.protein ,CpG Islands ,Protein Binding - Abstract
DNA methylation, mediated by the combined action of three DNA methyltransferases (DNMT1, DNMT3A, and DNMT3B), is essential for mammalian development and is a major contributor to cellular transformation. To elucidate how DNA methylation is targeted, we mapped the genome-wide localization of all DNMTs and methylation, and examined the relationships among these markers, histone modifications, and nucleosome structure in a pluripotent human tumor cell line in its undifferentiated and differentiated states. Our findings reveal a strong link between DNMTs and transcribed loci, and that DNA methylation is not a simple sum of DNMT localization patterns. A comparison of the epigenomes of normal and cancerous stem cells, and pluripotent and differentiated states shows that the presence of at least two DNMTs is strongly associated with loci targeted for DNA hypermethylation. Taken together, these results shed important light on the determinants of DNA methylation and how it may become disrupted in cancer cells., National Institutes of Health (U.S.) (Grant RC1HG005334), National Science Foundation (U.S.) (Postdoctoral Fellowship 0905968)
- Published
- 2012
23. Abstract 2305: Acute depletion reveals novel co-regulation of DNA methylation at conserved loci by DNMT1 and DNMT3B
- Author
-
Jeong Hyeon Choi, Rochelle L. Tiedemann, and Keith D. Robertson
- Subjects
Cancer Research ,Methyltransferase ,Promoter ,Methylation ,Biology ,Molecular biology ,Epigenetics of physical exercise ,Oncology ,CpG site ,embryonic structures ,DNA methylation ,Cancer research ,RNA-Directed DNA Methylation ,Epigenomics - Abstract
DNA methyltransferases (DNMTs) are responsible for establishing (DNMT3A, 3B, 3L) and maintaining (DNMT1) DNA methylation genome-wide. Aberrant DNA methylation is frequently observed in cancer; however, little is known about how regulation of this modification goes awry. In this study, we aim to understand how DNA methylation is regulated by the DNMTs throughout the genome by identifying specific and broad changes in methylation patterning upon depletion of the DNMTs. We utilize siRNA technology to acutely deplete NCCIT embryonal carcinoma cells of DNMT mRNA (individually and in combination), and then assay the impact on genome-wide DNA methylation patterns using the HumanMethylation450 Bead Chip (450K array). Depletion of DNMT1 (individual/combination) resulted in widespread hypomethylation, most notably in gene bodies, 3′UTRs, and intergenic sequences. DNMT3 knockdown resulted in more specific changes in DNA methylation, but surprisingly, more hypermethylation (predominately in gene bodies) than hypomethylation events occurred. These specific hypermethylation events, particularly in samples with DNMT3B KD, significantly overlapped with sites hypomethylated in DNMT1 KD conditions, indicating a potential cross-regulatory role for DNMT1 and DNMT3B in regulating DNA methylation across gene bodies. To gain a more comprehensive genome-wide view of DNA methylation in the absence of DNMT3B, we performed Methyl-CpG-Binding-Domain(MBD)-seq on DNMT3B KD cells. MBD-seq revealed dynamic changes in methylation with minor hypermethylation in promoters and subtle hypomethylation across gene bodies; however, analysis of only the most significant methylation changes (> 4-fold) revealed that more hypermethylation events occur in intronic sequences, consistent with results obtained using the 450K array. To further investigate the overlap between DNMT1 hypomethylated and DNMT3B hypermethylated sites, we examined DNA methylation in HCT116 colorectal carcinoma cells lacking (KO) or over-expressing (KI) DNMT1/DNMT3B. Interestingly, a marked number of CpG sites that gained methylation in the DNMT3B KO overlapped significantly with sites that became hypermethylated in DNMT1 and DNMT3B KI, and hypomethylated in DNMT1 KO. Additionally, these HCT116 hypermethylated CpG sites gained methylation in NCCIT DNMT3B KD (individual/combination) and lost methylation in DNMT1 KD. Taken together, these results suggest that DNMT1 and DNMT3B co-regulate DNA methylation at conserved loci across cell types in an opposing fashion, providing novel insight into a potential regulatory mechanism for DNA methylation patterning. Further elucidation of this DNMT1 and DNMT3B co-regulation holds the potential to yield novel therapeutic strategies for correcting aberrant methylation events in cancer. Citation Format: Rochelle Tiedemann, Jeong-Hyeon Choi, Keith Robertson. Acute depletion reveals novel co-regulation of DNA methylation at conserved loci by DNMT1 and DNMT3B. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 2305. doi:10.1158/1538-7445.AM2014-2305
- Published
- 2014
- Full Text
- View/download PDF
24. Abstract 2319: Dynamics of TET methylcytosine dioxygenases in 5-methylcytosine and 5-hydroxymethylcytosine patterning in human cancer cells
- Author
-
Emily L. Putiri, Jeong Hyeon Choi, Rochelle L. Tiedemann, and Keith D. Robertson
- Subjects
5-Hydroxymethylcytosine ,Cancer Research ,biology ,Epigenome ,Molecular biology ,Chromatin ,5-Methylcytosine ,chemistry.chemical_compound ,Histone ,Oncology ,chemistry ,CpG site ,DNA methylation ,biology.protein ,Epigenetics - Abstract
The Ten-eleven translocation (TET) family of dioxygenases hydroxylate 5-methylcytosine (5mC) to 5-hydroxymethylcytosine (5hmC) in DNA. 5mC provides important epigenetic instructions during development, and its aberrant control is a major contributor to cellular transformation; however TET functions in regulating the epigenome, particularly in cancer, remain largely unknown. We targeted TET1, TET2, and TET3 for siRNA-mediated depletion in pluripotent human embryonic carcinoma cells and examined the impact on 5mC and 5hmC genome-wide localization. TET1, TET2, and TET3 co-regulate 5hmC at many sites, and depletion of only one of the TETs is sufficient to reduce 5hmC at these co-regulated sites, suggesting a functional co-dependence for TETs. Depletion of TET1 and TET2 had the greatest impact on 5hmC levels at high and low CpG density promoters, respectively, indicating that TETs exhibit DNA sequence-based functional specificity. All TETs prevent hypermethylation throughout the genome, especially in CpG island shores, where TET depletion resulted in prolific hypermethylation. Promoter hypermethylation resulting from TET depletion was associated with histone H2AK119 monoubiquitination, DNMT1, and DNMT3B occupancy. Surprisingly, TETs also promote cytosine methylation, as many loci became hypomethylated following TET depletion. Induction of differentiation generally caused 5hmC reduction, except at transcriptionally activated genes, which become enriched for 5hmC. Importantly, genes prone to promoter hypermethylation in cancer become depleted of intragenic 5hmC and 5mC with TET deficiency. This study highlights the multi-dimensional functions of TETs in mediating DNA methylation, hydroxymethylation, and gene expression patterns, and the results reveal that chromatin landscape and DNA sequence composition are regulators of TET function. Citation Format: Emily L. Putiri, Rochelle L. Tiedemann, Jeong-Hyeon Choi, Keith D. Robertson. Dynamics of TET methylcytosine dioxygenases in 5-methylcytosine and 5-hydroxymethylcytosine patterning in human cancer cells. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 2319. doi:10.1158/1538-7445.AM2014-2319
- Published
- 2014
- Full Text
- View/download PDF
25. Abstract 2309: Identification of common and unique epigenetic signatures of chronic hepatitis infection and alcohol abuse in human liver disease
- Author
-
Ryan A. Hlady, Chen Liu, Jeong Hyeon Choi, Rochelle L. Tiedemann, William Puszyk, and Keith D. Robertson
- Subjects
Hepatitis B virus ,Cancer Research ,Methylation ,Epigenome ,Biology ,medicine.disease ,medicine.disease_cause ,Oncology ,Hepatocellular carcinoma ,Immunology ,DNA methylation ,medicine ,Cancer research ,Epigenetics ,Carcinogenesis ,Liver cancer - Abstract
Dysregulation of the intrinsic DNA methylation landscape is a ubiquitous feature of human carcinogenesis, manifested by global hypomethylation and promoter-specific hypermethylation, ultimately resulting in genome instability and tumor suppressor gene silencing, respectively. Alterations in DNA methylation are particularly apparent in hepatocellular carcinoma (HCC) which afflicts roughly 750,000 new patients each year [1]. Indeed, it has been demonstrated that a variety of tumor suppressor genes (e.g. p53, E-cadherin) are hypermethylated and silenced in HCC [2-4]. Importantly, HCC is accompanied by the premalignant stage of cirrhosis in 80% of cases. One major roadblock to understanding the methylome in HCC is the presence of multiple etiologies such as Hepatitis C virus (HCV), Hepatitis B virus (HBV), and chronic alcoholism. Therefore, we performed genome-wide methylation profiling to dissect the methylation patterns of more than 170 primary liver samples to stratify etiologic and stage-specific changes in the DNA methylation landscape. Our results profile the DNA methylation landscape across normal, cirrhotic, and HCC livers in the largest study of its kind to date. We unveil distinct locus-specific and large-scale effects of HCV, HBV, and chronic alcoholism in hepatocarcinogenesis. Furthermore, analysis indicates a specific methylation profile for individual etiologies as well as conserved patterns throughout cirrhosis and hepatocellular carcinoma. Our study demonstrates that each etiology contains potential biomarkers and targets for downstream clinical therapeutics. This study is our first step toward defining the epigenome in cirrhosis and hepatocellular carcinoma and will be combined with future genomic and epimutational data (e.g. transcription, histone modifications, miRNA) to determine the true extent and interplay between epigenetic marks across different stages of liver cancer. Overall, this research has the potential to improve our understanding of epigenetics and result in diagnostic, prognostic, and therapeutic epigenetic signatures in cirrhosis and hepatocellular carcinoma, which are expected to allow for more timely and efficient detection of disease. 1. Jemal, A., et al., Global cancer statistics. CA: a cancer journal for clinicians, 2011. 61(2): p. 69-90. 2. Tischoff, I. and A. Tannapfe, DNA methylation in hepatocellular carcinoma. World journal of gastroenterology : WJG, 2008. 14(11): p. 1741-8. 3. Calvisi, D.F., et al., Mechanistic and prognostic significance of aberrant methylation in the molecular pathogenesis of human hepatocellular carcinoma. The Journal of clinical investigation, 2007. 117(9): p. 2713-22. 4. Lambert, M.P., et al., Aberrant DNA methylation distinguishes hepatocellular carcinoma associated with HBV and HCV infection and alcohol intake. Journal of hepatology, 2011. 54(4): p. 705-15. Citation Format: Ryan A. Hlady, Rochelle Tiedemann, William Puszyk, Chen Liu, Jeong-Hyeon Choi, Keith D. Robertson. Identification of common and unique epigenetic signatures of chronic hepatitis infection and alcohol abuse in human liver disease. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 2309. doi:10.1158/1538-7445.AM2014-2309
- Published
- 2014
- Full Text
- View/download PDF
26. Distinct and overlapping control of 5-methylcytosine and 5-hydroxymethylcytosine by the TET proteins in human cancer cells
- Author
-
Keith D. Robertson, Rochelle L. Tiedemann, Joyce J. Thompson, Emily L. Putiri, Jeong Hyeon Choi, Chunsheng Liu, and Thai H. Ho
- Subjects
Biology ,Dioxygenases ,Epigenesis, Genetic ,Mixed Function Oxygenases ,Cytosine ,chemistry.chemical_compound ,Cytosine nucleotide ,Cell Line, Tumor ,Proto-Oncogene Proteins ,Humans ,Epigenetics ,Promoter Regions, Genetic ,Genetics ,5-Hydroxymethylcytosine ,Regulation of gene expression ,Research ,Cell Differentiation ,Sequence Analysis, DNA ,Methylation ,biochemical phenomena, metabolism, and nutrition ,DNA Methylation ,Chromatin ,DNA-Binding Proteins ,Gene Expression Regulation, Neoplastic ,chemistry ,CpG site ,Gene Knockdown Techniques ,DNA methylation ,5-Methylcytosine - Abstract
Background: The TET family of dioxygenases catalyze conversion of 5-methylcytosine (5mC) to 5-hydroxymethylcytosine (5hmC), but their involvement in establishing normal 5mC patterns during mammalian development and their contributions to aberrant control of 5mC during cellular transformation remain largely unknown. We depleted TET1, TET2, and TET3 in a pluripotent embryonic carcinoma cell model and examined the impact on genome-wide 5mC, 5hmC, and transcriptional patterns. Results: TET1 depletion yields widespread reduction of 5hmC, while depletion of TET2 and TET3 reduces 5hmC at a subset of TET1 targets suggesting functional co-dependence. TET2 or TET3 depletion also causes increased 5hmC, suggesting these proteins play a major role in 5hmC removal. All TETs prevent hypermethylation throughout the genome, a finding dramatically illustrated in CpG island shores, where TET depletion results in prolific hypermethylation. Surprisingly, TETs also promote methylation, as hypomethylation was associated with 5hmC reduction. TET function is highly specific to chromatin environment: 5hmC maintenance by all TETs occurs at polycomb-marked chromatin and genes expressed at moderate levels; 5hmC removal by TET2 is associated with highly transcribed genes enriched for H3K4me3 and H3K36me3. Importantly, genes prone to hypermethylation in cancer become depleted of 5hmC with TET deficiency, suggesting that TETs normally promote 5hmC at these loci. Finally, all three TETs, but especially TET2, are required for 5hmC enrichment at enhancers, a condition necessary for expression of adjacent genes. Conclusions: These results provide novel insight into the division of labor among TET proteins and reveal important connections between TET activity, the chromatin landscape, and gene expression. Background Vertebrate cellular identity arises through intricate differentiation events orchestrated by epigenetic regulation of gene expression. One key epigenetic mechanism is methylation of DNA. DNA is covalently modified by methylation of the carbon-5 position within cytosine nucleotides (5mC), an epigenetic mark that, when occurring in gene promoters, is associated with transcriptional repression. DNA methylation primarily occurs in the context of cytosine followed by guanine (CpG), and normal CpG methylation patterns have been extensively characterized
- Published
- 2014
- Full Text
- View/download PDF
27. Abstract B34: Acute depletion reveals novel divisions of labor among human DNA methyltransferases in cancer
- Author
-
Rochelle L. Tiedemann, Jeong Hyeon Choi, and Keith D. Robertson
- Subjects
Genetics ,Cancer Research ,DNA Methylation Regulation ,Epigenetics of physical exercise ,Methyltransferase ,Oncology ,DNA methylation ,Cancer research ,Epigenetics ,Methylation ,Biology ,RNA-Directed DNA Methylation ,Epigenomics - Abstract
DNA methyltransferases (DNMTs) are responsible for establishing (DNMT3A, DNMT3B, DNMT3L) and maintaining (DNMT1) DNA methylation genome-wide. Hypomethylation of repetitive sequences and transposable elements coupled with gene-specific promoter hypermethylation events contribute to the genomic instability and loss of tumor suppressor gene transcription observed in cancer. Regulation of aberrant methylation in cancer remains poorly understood. The aim of our study was to identify unique and overlapping target sites for each of the DNMTs to better understand regulation of normal and aberrant DNA methylation. We hypothesized that acute depletion of the DNMTs (individual and combination) mediated by siRNA technology in NCCIT human embryonic carcinoma cells would result in both distinct and broad changes in DNA methylation patterning. Genome-wide methylation was assayed using the HumanMethylation450 Bead Chip (450K array) allowing for specific CpG site methylation status determination. Select target sites were verified by bisulfite genomic sequencing. DNMT1 knockdown samples (individual/combination) revealed genome-wide hypomethylation, with the strongest demethylation occurring in gene bodies, 3′UTR, and intergenic sequences. Interestingly, only the DNMT1 individual knockdown showed significant hypermethylation in gene promoters. DNMT3 knockdowns showed more specific changes in DNA methylation, but surprisingly, more hypermethylation events occurred than hypomethylation at CpG dinucleotides. Hypermethylation observed in DNMT3 knockdown occurred primarily in gene bodies and 3′UTR, and overlapped significantly with those genes that become hypomethylated in DNMT1 knockdown, indicating a potential cross-regulatory role for the DNMTs to maintain proper regulation of DNA methylation at specific gene termini. Conversely, gene promoters were targeted for hypomethylation in DNMT3 knockdown, and did not significantly overlap with genes that become hypermethylated in DNMT1 knockdown. Of particular interest was that DNMT3B knockdown resulted in widespread non-CpG hypomethylation. In contrast, DNMT3L knockdown showed non-CpG hypermethylation, indicating a potential mechanism for regulation of non-CpG methylation where DNMT3B is responsible for non-CpG methylation, and DNMT3L acts to restrict DNMT3B's activity at non-CpG dinucleotides. Our results reveal a complex view of DNA methylation regulation, in which DNMTs not only target specific sites for methylation, but also cooperate to establish and maintain proper levels of DNA methylation at CpG and non-CpG dinucleotides. Moving forward, we believe our results will provide the framework needed to define the regulatory mechanisms by which DNA methylation is conferred and ultimately develop therapeutic strategies to correct aberrant methylation events that occur in cancer. Citation Format: Rochelle L. Tiedemann, Jeong-Hyeon Choi, Keith D. Robertson. Acute depletion reveals novel divisions of labor among human DNA methyltransferases in cancer. [abstract]. In: Proceedings of the AACR Special Conference on Chromatin and Epigenetics in Cancer; Jun 19-22, 2013; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2013;73(13 Suppl):Abstract nr B34.
- Published
- 2013
- Full Text
- View/download PDF
28. Abstract 2970: Acute depletion of DNA methyltransferases reveals unique and overlapping target sites in cancer
- Author
-
Jeong Hyeon Choi, Keith D. Robertson, and Rochelle L. Tiedemann
- Subjects
Genetics ,Cancer Research ,Methyltransferase ,Oncology ,CpG site ,DNA methylation ,Bisulfite sequencing ,DNMT1 ,Illumina Methylation Assay ,Promoter ,Methylation ,Biology ,Molecular biology - Abstract
DNA methyltransferases (DNMTs) are responsible for establishing (DNMT3A, DNMT3B, DNMT3L) and maintaining (DNMT1) DNA methylation to regulate gene transcription and promote overall genome stability. Global changes in DNA methylation, such as hypomethylation of repetitive sequences and hypermethylation of tumor supressor gene promoters, are commonly observed in various types of cancer; however, the DNMTs’ contribution to this aberrant methylation remains largely unknown. In this study, we aim to identify unique and overlapping target sites for each of the DNMTs in order to better understand aberrant DNA methylation in cancer that will ultimately permit development of new therapeutic strategies. We utilize siRNA-mediated knockdown technology to acutely deplete the mRNA for each of the DNMTs in both individual and combinatorial fashion in NCCIT embryonal carcinoma cells. Subsequently, DNA methylation is observed genome-wide in each DNA sample using two different methodologies: (1) Methyl-CpG Binding Domain (MBD)-seq, which identifies regions of the genome that are enriched for DNA methylation, and (2) Illumina Infinium HumanMethylation450 BeadChip (450K array) allowing for specific CpG site methylation status determination. Aligned MBD-seq sequences for individual DNMT knockdowns are analyzed by read coverage nomalization within 100Kbp windows and using various peak-calling algorithms (e.g. MACS, BALM). For the 450K array, DNA samples for both individual and combination knockdowns undergo bisulfite conversion and array processing; β-values for each CpG site are derived from the array signal intensities using GenomeStudio and R/Bioconductor (minfi). Genome-wide DNA methylation analysis reveals that DNMT1 depletion, both individual and in combination with knockdown of other DNMTs, results in global demethylation among all genomic features. Interestingly, a small population of genes exhibit hypermethylation in gene-promoter CpG islands for DNMT1 (individual only) depletion. In contrast, de novo methyltransferase depletion (individual and combination) shows more specific demethylation effects. In particular, hypomethylation events resulting from DNMT3B depletion (individual and combination (not 3B+3L)) occur within gene bodies and largely outside of CpG islands and flanking regions (shores, shelves). Additionally, a number of hypermethylation events occurring within the 3’UTR region of genes are observed in de novo methyltransferase depleted samples. We anticipate further analysis will reveal unique and overlapping target sites for each of the DNMTs that will lay the ground-work necessary to characterize and understand DNMT recruitment both in normal and cancerous tissues. Citation Format: Rochelle L. Tiedemann, Jeong-Hyeon Choi, Keith D. Robertson. Acute depletion of DNA methyltransferases reveals unique and overlapping target sites in cancer. [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr 2970. doi:10.1158/1538-7445.AM2013-2970
- Published
- 2013
- Full Text
- View/download PDF
29. Abstract 5009: Identification of DNA methyltransferase target sites in cancer cells
- Author
-
Rochelle L. Tiedemann and Keith D. Robertson
- Subjects
Genetics ,Genome instability ,Cancer Research ,Methyltransferase ,Genomics ,Biology ,DNA methyltransferase ,DNA sequencing ,chemistry.chemical_compound ,Oncology ,chemistry ,DNA methylation ,DNMT1 ,DNA - Abstract
Aberrant DNA methylation is commonly observed in cancer and is characterized by genome-wide hypomethylation and gene-specific hypermethylation, which is thought to contribute to genomic instability and tumor suppressor gene silencing, respectively. The DNA methyltransferases (DNMTs) are responsible for the establishment (DNMT3A, DNMT3B, DNMT3L) and maintenance (DNMT1) of DNA methylation patterns genome-wide. The mechanism by which DNA methylation patterns are altered in cancer is not well understood. Genome-wide unique and overlapping target sites for each of the DNMTs are also unknown both in normal and cancer states. Identification of DNMT target loci is essential in order to better understand how aberrant DNA methylation occurs in cancer. To study this process, DNMT mRNA levels were depleted both individually and in a combinatorial fashion via RNAi-based techniques in embryonic carcinoma cells. Following reduction in DNMT expression, the resulting DNA and RNA was analyzed for genome-wide DNA methylation and gene expression patterns, respectively. An affinity purification method, Methyl-Binding Domain (MBD)-seq, was used to capture and enrich methylated regions of the genome by utilizing the methyl-binding domain of MBD2b. The enriched methylated DNA was then used to construct an Illumina sequencing library. Global gene expression patterns were analyzed by microarray for each RNA sample. Next generation sequencing and microarray data were analyzed using several available algorithms (e.g. MACS) and high-throughput data software packages (e.g. Partek Genomics Suite) in order to construct and evaluate the DNA methylation profiles and subsequent gene expression changes that result from DNMT depletion. Our data show that distinct changes in DNA methylation profiles occur among the various DNMT knockdown samples that permit us to identify unique and cooperative target loci for each DNMT. Further elucidation of DNMT target sites holds great promise for enhancing our understanding of mechanisms that control aberrant DNA methylation that is observed in cancer as well as provide insight and rationale for targeting specific DNMTs in cancer therapies. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr 5009. doi:1538-7445.AM2012-5009
- Published
- 2012
- Full Text
- View/download PDF
30. Acute Depletion Redefines the Division of Labor among DNA Methyltransferases in Methylating the Human Genome
- Author
-
Jeong Heon Lee, Tamas Ordog, Ryan A. Hlady, Keith D. Robertson, Zhiguo H Zhang, Rochelle L. Tiedemann, Katsunobu Kashiwagi, Chen Liu, Jeong Hyeon Choi, and Emily L. Putiri
- Subjects
DNA (Cytosine-5-)-Methyltransferase 1 ,Methyltransferase ,Biology ,Article ,General Biochemistry, Genetics and Molecular Biology ,DNA Methyltransferase 3A ,Epigenesis, Genetic ,Transcriptome ,Gene Knockout Techniques ,03 medical and health sciences ,0302 clinical medicine ,Cell Line, Tumor ,Humans ,DNA (Cytosine-5-)-Methyltransferases ,RNA, Small Interfering ,lcsh:QH301-705.5 ,RNA-Directed DNA Methylation ,030304 developmental biology ,Genetics ,0303 health sciences ,Genome, Human ,Methylation ,Epigenome ,DNA Methylation ,Gene Expression Regulation, Neoplastic ,lcsh:Biology (General) ,CpG site ,Genetic Loci ,030220 oncology & carcinogenesis ,DNA methylation ,embryonic structures ,5-Methylcytosine ,DNMT1 ,CpG Islands - Abstract
Global patterns of DNA methylation, mediated by the DNA methyltransferases (DNMTs), are disrupted in all cancers by mechanisms that remain largely unknown, hampering their development as therapeutic targets. Combinatorial acute depletion of all DNMTs in a pluripotent human tumor cell line, followed by epigenome and transcriptome analysis, revealed DNMT functions in unprecedented detail. DNMT3B occupancy regulates methylation during differentiation, while an unexpected interplay was discovered in which DNMT1 and DNMT3B antithetically regulate methylation and hydroxymethylation in gene bodies, a finding confirmed in other cell types. DNMT3B mediated nonCpG methylation, while DNMT3L influenced the activity of DNMT3B toward nonCpG versus CpG site methylation. Taken together, these data reveal new functional targets of each DNMT suggesting that isoform selective inhibition would be therapeutically advantageous.
- Full Text
- View/download PDF
31. In silico APC/C substrate discovery reveals cell cycle-dependent degradation of UHRF1 and other chromatin regulators.
- Author
-
Jennifer L Franks, Raquel C Martinez-Chacin, Xianxi Wang, Rochelle L Tiedemann, Thomas Bonacci, Rajarshi Choudhury, Derek L Bolhuis, Taylor P Enrico, Ryan D Mouery, Jeffrey S Damrauer, Feng Yan, Joseph S Harrison, M Ben Major, Katherine A Hoadley, Aussie Suzuki, Scott B Rothbart, Nicholas G Brown, and Michael J Emanuele
- Subjects
Biology (General) ,QH301-705.5 - Abstract
The anaphase-promoting complex/cyclosome (APC/C) is an E3 ubiquitin ligase and critical regulator of cell cycle progression. Despite its vital role, it has remained challenging to globally map APC/C substrates. By combining orthogonal features of known substrates, we predicted APC/C substrates in silico. This analysis identified many known substrates and suggested numerous candidates. Unexpectedly, chromatin regulatory proteins are enriched among putative substrates, and we show experimentally that several chromatin proteins bind APC/C, oscillate during the cell cycle, and are degraded following APC/C activation, consistent with being direct APC/C substrates. Additional analysis revealed detailed mechanisms of ubiquitylation for UHRF1, a key chromatin regulator involved in histone ubiquitylation and DNA methylation maintenance. Disrupting UHRF1 degradation at mitotic exit accelerates G1-phase cell cycle progression and perturbs global DNA methylation patterning in the genome. We conclude that APC/C coordinates crosstalk between cell cycle and chromatin regulatory proteins. This has potential consequences in normal cell physiology, where the chromatin environment changes depending on proliferative state, as well as in disease.
- Published
- 2020
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.