59 results on '"Whetstine JR"'
Search Results
2. Epigenetic modulators provide a path to understanding disease and therapeutic opportunity.
- Author
-
Honer MA, Ferman BI, Gray ZH, Bondarenko EA, and Whetstine JR
- Subjects
- Humans, Animals, DNA Methylation genetics, Disease genetics, Epigenesis, Genetic
- Abstract
The discovery of epigenetic modulators (writers, erasers, readers, and remodelers) has shed light on previously underappreciated biological mechanisms that promote diseases. With these insights, novel biomarkers and innovative combination therapies can be used to address challenging and difficult to treat disease states. This review highlights key mechanisms that epigenetic writers, erasers, readers, and remodelers control, as well as their connection with disease states and recent advances in associated epigenetic therapies., (© 2024 Honer et al.; Published by Cold Spring Harbor Laboratory Press.)
- Published
- 2024
- Full Text
- View/download PDF
3. Epigenetic balance ensures mechanistic control of MLL amplification and rearrangement.
- Author
-
Gray ZH, Chakraborty D, Duttweiler RR, Alekbaeva GD, Murphy SE, Chetal K, Ji F, Ferman BI, Honer MA, Wang Z, Myers C, Sun R, Kaniskan HÜ, Toma MM, Bondarenko EA, Santoro JN, Miranda C, Dillingham ME, Tang R, Gozani O, Jin J, Skorski T, Duy C, Lee H, Sadreyev RI, and Whetstine JR
- Subjects
- Adult, Animals, Humans, Infant, Mice, Doxorubicin pharmacology, Gene Rearrangement, Histocompatibility Antigens, Histone-Lysine N-Methyltransferase genetics, Histone-Lysine N-Methyltransferase metabolism, Jumonji Domain-Containing Histone Demethylases genetics, Jumonji Domain-Containing Histone Demethylases metabolism, Leukemia metabolism, Lysine metabolism, Translocation, Genetic, Epigenesis, Genetic, Myeloid-Lymphoid Leukemia Protein genetics
- Abstract
MLL/KMT2A amplifications and translocations are prevalent in infant, adult, and therapy-induced leukemia. However, the molecular contributor(s) to these alterations are unclear. Here, we demonstrate that histone H3 lysine 9 mono- and di-methylation (H3K9me1/2) balance at the MLL/KMT2A locus regulates these amplifications and rearrangements. This balance is controlled by the crosstalk between lysine demethylase KDM3B and methyltransferase G9a/EHMT2. KDM3B depletion increases H3K9me1/2 levels and reduces CTCF occupancy at the MLL/KMT2A locus, in turn promoting amplification and rearrangements. Depleting CTCF is also sufficient to generate these focal alterations. Furthermore, the chemotherapy doxorubicin (Dox), which associates with therapy-induced leukemia and promotes MLL/KMT2A amplifications and rearrangements, suppresses KDM3B and CTCF protein levels. KDM3B and CTCF overexpression rescues Dox-induced MLL/KMT2A alterations. G9a inhibition in human cells or mice also suppresses MLL/KMT2A events accompanying Dox treatment. Therefore, MLL/KMT2A amplifications and rearrangements are controlled by epigenetic regulators that are tractable drug targets, which has clinical implications., Competing Interests: Declaration of interests J.R.W. has served or is serving as a consultant or advisor for Qsonica, Salarius Pharmaceuticals, Daiichi Sankyo, Inc., Vyne Therapeutics, and Lily Asia Ventures. J.R.W. also receives funding for research from Salarius Pharmaceuticals and Oryzon Genomics. O.G. is a scientific cofounder and shareholder of EpiCypher, Inc., K36 Therapeutics, Inc., and Alternative Bio, Inc. J.J. received research funds from Celgene Corporation, Levo Therapeutics, Inc., Cullgen, Inc., and Cullinan Oncology, Inc. J.J. is a cofounder and equity shareholder in Cullgen, Inc., a scientific cofounder and scientific advisory board member of Onsero Therapeutics, Inc., and a consultant for Cullgen, Inc., EpiCypher, Inc., and Accent Therapeutics, Inc. C.D. receives research funds from Janssen outside the submitted work., (Copyright © 2023 Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
4. TET1 and TDG Suppress Inflammatory Response in Intestinal Tumorigenesis: Implications for Colorectal Tumors With the CpG Island Methylator Phenotype.
- Author
-
Tricarico R, Madzo J, Scher G, Cohen M, Jelinek J, Maegawa S, Nagarathinam R, Scher C, Chang WC, Nicolas E, Slifker M, Zhou Y, Devarajan K, Cai KQ, Kwok T, Nakajima P, Xu J, Mancuso P, Doneddu V, Bagella L, Williams R, Balachandran S, Maskalenko N, Campbell K, Ma X, Cañadas I, Viana-Errasti J, Moreno V, Valle L, Grivennikov S, Peshkova I, Kurilenko N, Mazitova A, Koltsova E, Lee H, Walsh M, Duttweiler R, Whetstine JR, Yen TJ, Issa JP, and Bellacosa A
- Subjects
- Animals, Humans, Mice, Carcinogenesis genetics, Cell Transformation, Neoplastic genetics, CpG Islands genetics, DNA Methylation, DNA-Binding Proteins genetics, Epigenesis, Genetic, Mixed Function Oxygenases genetics, Phenotype, Proto-Oncogene Proteins genetics, Adenocarcinoma genetics, Adenocarcinoma pathology, Adenoma genetics, Adenoma pathology, Colonic Neoplasms genetics, Colorectal Neoplasms genetics, Colorectal Neoplasms pathology
- Abstract
Background & Aims: Aberrant DNA methylation is frequent in colorectal cancer (CRC), but underlying mechanisms and pathologic consequences are poorly understood., Methods: We disrupted active DNA demethylation genes Tet1 and/or Tdg from Apc
Min mice and characterized the methylome and transcriptome of colonic adenomas. Data were compared to human colonic adenocarcinomas (COAD) in The Cancer Genome Atlas., Results: There were increased numbers of small intestinal adenomas in ApcMin mice expressing the TdgN151A allele, whereas Tet1-deficient and Tet1/TdgN151A -double heterozygous ApcMin colonic adenomas were larger with features of erosion and invasion. We detected reduction in global DNA hypomethylation in colonic adenomas from Tet1- and Tdg-mutant ApcMin mice and hypermethylation of CpG islands in Tet1-mutant ApcMin adenomas. Up-regulation of inflammatory, immune, and interferon response genes was present in Tet1- and Tdg-mutant colonic adenomas compared to control ApcMin adenomas. This up-regulation was also seen in murine colonic organoids and human CRC lines infected with lentiviruses expressing TET1 or TDG short hairpin RNA. A 127-gene inflammatory signature separated colonic adenocarcinomas into 4 groups, closely aligned with their microsatellite or chromosomal instability and characterized by different levels of DNA methylation and DNMT1 expression that anticorrelated with TET1 expression. Tumors with the CpG island methylator phenotype (CIMP) had concerted high DNMT1/low TET1 expression. TET1 or TDG knockdown in CRC lines enhanced killing by natural killer cells., Conclusions: Our findings reveal a novel epigenetic regulation, linked to the type of genomic instability, by which TET1/TDG-mediated DNA demethylation decreases methylation levels and inflammatory/interferon/immune responses. CIMP in CRC is triggered by an imbalance of methylating activities over demethylating activities. These mice represent a model of CIMP CRC., (Copyright © 2023 AGA Institute. Published by Elsevier Inc. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
5. Computational workflow for integrative analyses of DNA replication timing, epigenomic, and transcriptomic data.
- Author
-
Ji F, Van Rechem C, Whetstine JR, and Sadreyev RI
- Subjects
- Transcriptome, Workflow, Chromatin genetics, Epigenomics, DNA Replication Timing
- Abstract
Temporal profiling of DNA replication timing (RT) in combination with chromatin modifications, chromatin accessibility, and gene expression provides new insights into the causal relationships between chromatin and RT during cell cycle. Here, we describe a protocol for in-depth integrative computational analyses of Repli-seq, ATAC-seq, RNA-seq, and ChIP-seq or CUT&RUN data for multiple marks at various time points across cell cycle and changes in their interrelationships upon an experimental perturbation (e.g., knockdown or overexpression of a regulatory protein). For complete details on the use and execution of this protocol, please refer to Van Rechem et al. (2021)., Competing Interests: J.R.W. has or is serving as a consultant or advisor or received research support from Qsonica (consultant), Daiichi Sankyo, Inc (advisor/consultant), Vyne Therapeutics (advisor), and Salarius Pharmaceuticals (consulted and has sponsored research)., (© 2022 The Authors.)
- Published
- 2022
- Full Text
- View/download PDF
6. A cell-sorting-based protocol for cell cycle small-scale ChIP sequencing.
- Author
-
Whetstine JR and Van Rechem C
- Subjects
- Cell Cycle genetics, Chromatin Immunoprecipitation methods, Histone Code, Chromatin genetics, Chromatin Immunoprecipitation Sequencing
- Abstract
Classic approaches to characterizing cell cycle leverage chemicals or altered nucleotide pools, which could impact chromatin states at specific phases of the cell cycle. Such approaches could induce metabolic alterations and/or DNA damage, which could reshape protein recruitment and histone modifications. In this protocol, we describe ways to fix and sort cells across the cell cycle based on their DNA content. We further detail immunoprecipitation and library preparation, allowing analysis of the epigenome by chromatin immunoprecipitation sequencing (ChIP-seq) for small numbers of cells. For complete details on the use and execution of this protocol, please refer to Van Rechem et al. (2021)., Competing Interests: In the past year, J.R.W. has consulted for Qsonica (manufacturer of Q800R system). J.R.W. has served as a consultant or advisor for Salarius Pharmaceuticals, Daiichi Sankyo, Inc., and VYNE Therapeutics. J.R.W. has also received sponsored research from Salarius Pharmaceuticals. C.V.R. declares no competing interests., (© 2022 The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
7. DNA replication fork speed underlies cell fate changes and promotes reprogramming.
- Author
-
Nakatani T, Lin J, Ji F, Ettinger A, Pontabry J, Tokoro M, Altamirano-Pacheco L, Fiorentino J, Mahammadov E, Hatano Y, Van Rechem C, Chakraborty D, Ruiz-Morales ER, Arguello Pascualli PY, Scialdone A, Yamagata K, Whetstine JR, Sadreyev RI, and Torres-Padilla ME
- Subjects
- Animals, Cell Differentiation genetics, Cellular Reprogramming genetics, DNA Replication genetics, Embryonic Development genetics, Mice, Embryo, Mammalian, Pluripotent Stem Cells
- Abstract
Totipotency emerges in early embryogenesis, but its molecular underpinnings remain poorly characterized. In the present study, we employed DNA fiber analysis to investigate how pluripotent stem cells are reprogrammed into totipotent-like 2-cell-like cells (2CLCs). We show that totipotent cells of the early mouse embryo have slow DNA replication fork speed and that 2CLCs recapitulate this feature, suggesting that fork speed underlies the transition to a totipotent-like state. 2CLCs emerge concomitant with DNA replication and display changes in replication timing (RT), particularly during the early S-phase. RT changes occur prior to 2CLC emergence, suggesting that RT may predispose to gene expression changes and consequent reprogramming of cell fate. Slowing down replication fork speed experimentally induces 2CLCs. In vivo, slowing fork speed improves the reprogramming efficiency of somatic cell nuclear transfer. Our data suggest that fork speed regulates cellular plasticity and that remodeling of replication features leads to changes in cell fate and reprogramming., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
8. Protocol to isolate cells in four stages of S phase for high-resolution replication-timing sequencing.
- Author
-
Whetstine JR and Van Rechem C
- Subjects
- Bromodeoxyuridine, S Phase genetics, Sequence Analysis, DNA, DNA Replication genetics, DNA Replication Timing
- Abstract
Traditional replication timing (RT) experiments divide S phase into two phases: early and late. However, there is an increasing awareness that variation in RT can occur during the course of S phase and impact our understanding of RT patterns and regulation. Here, we describe a RT protocol in RPE-1 cells for collecting four phases within S and the library preparation that takes advantage of a commercial kit for methyl-DNA. This step allows BrdU-labeled DNA sequencing and assessment of RT genome wide. For complete details on the use and execution of this protocol, please refer to Van Rechem et al. (2021)., Competing Interests: In the past year, J.R.W. has consulted for Qsonica (manufacturer of Q800R system). J.R.W. has served as a consultant or advisor for Salarius Pharmaceuticals, Daiichi Sankyo, and VYNE Therapeutics. J.R.W. has also received sponsored research from Salarius Pharamceuticals. C.V.R. declares no competing interests., (© 2022 The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
9. Collective regulation of chromatin modifications predicts replication timing during cell cycle.
- Author
-
Van Rechem C, Ji F, Chakraborty D, Black JC, Sadreyev RI, and Whetstine JR
- Subjects
- Cell Line, Chromatin metabolism, Enhancer Elements, Genetic genetics, Genome, Histone Code genetics, Humans, Jumonji Domain-Containing Histone Demethylases genetics, Jumonji Domain-Containing Histone Demethylases metabolism, Methylation, S Phase, Chromatin chemistry, DNA Replication Timing physiology
- Abstract
Replication timing (RT) associates with genome architecture, while having a mixed relationship to histone marks. By profiling replication at high resolution and assessing broad histone marks across the cell cycle at the resolution of RT with and without genetic perturbation, we address the causal relationship between histone marks and RT. Four primary chromatin states, including an uncharacterized H3K36me2 state, emerge and define 97% of the mappable genome. RT and local replication patterns (e.g., initiation zones) quantitatively associate with chromatin states, histone mark dynamics, and spatial chromatin structure. Manipulation of broad histone marks and enhancer elements by overexpressing the histone H3 lysine 9/36 tri-demethylase KDM4A impacts RT across 11% of the genome. Broad histone modification changes were strong predictors of the observed RT alterations. Lastly, replication within H3K36me2-enriched neighborhoods is sensitive to KDM4A overexpression and is controlled at a megabase scale. These studies establish a role for collective chromatin mark regulation in modulating RT., Competing Interests: Declaration of interests In the past year, J.R.W. is or was serving as a consultant or advisor for Qsonica, Salarius Pharmaceuticals, and Daiichi Sankyo, Inc. The other authors declare no competing interests., (Copyright © 2021 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
10. Isoflurane impairs oogenesis through germ cell apoptosis in C. elegans.
- Author
-
Zhang T, Ni C, Li C, Lu P, Chen D, Dong Y, Whetstine JR, Zhang Y, and Xie Z
- Subjects
- Anesthetics, Inhalation toxicity, Animals, Caenorhabditis elegans cytology, Caenorhabditis elegans embryology, Caenorhabditis elegans genetics, Caenorhabditis elegans Proteins genetics, Caspases genetics, Embryo, Nonmammalian drug effects, Female, Hermaphroditic Organisms, Male, Oxidative Stress drug effects, Proto-Oncogene Proteins c-abl genetics, Reactive Oxygen Species metabolism, Tumor Suppressor Protein p53 genetics, Apoptosis drug effects, Caenorhabditis elegans drug effects, Isoflurane toxicity, Oogenesis drug effects
- Abstract
Anesthetic isoflurane has been reported to induce toxicity. However, the effects of isoflurane on fecundity remain largely unknown. We established a system in C. elegans to investigate the effects of isoflurane on oogenesis. Synchronized L4 stage C. elegans were treated with 7% isoflurane for 4 h. Dead cells, ROS, embryos, and unfertilized eggs laid by hermaphrodites were measured by fluorescence imaging and counting. The C. elegans with losses of ced-3, cep-1, abl-1, male C. elegans, and oxidative stress inhibitor N-acetyl-cysteine were used in the interaction studies. We found that isoflurane decreased the numbers of embryos and unfertilized eggs and increased the levels of dead cells and ROS in C. elegans. The isoflurane-induced impairment of oogenesis was associated with abl-1, ced-3, but not cep-1. N-acetyl-cysteine attenuated the isoflurane-induced impairment of oogenesis in C. elegans. Mating with male C. elegans did not attenuate the isoflurane-induced changes in oogenesis. These findings suggest that isoflurane may impair oogenesis through abl-1- and ced-3-associated, but not cep-1-associated, germ cell apoptosis and oxidative stress, pending further investigation. These studies will promote more research to determine the potential effects of anesthesia on fecundity., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
11. The lysine demethylase KDM4A controls the cell-cycle expression of replicative canonical histone genes.
- Author
-
Van Rechem C, Ji F, Mishra S, Chakraborty D, Murphy SE, Dillingham ME, Sadreyev RI, and Whetstine JR
- Subjects
- Catalysis, Epigenesis, Genetic, Gene Expression Profiling, Histones metabolism, Humans, Transcription, Genetic, DNA Replication, Gene Expression Regulation, Histones genetics, Jumonji Domain-Containing Histone Demethylases genetics
- Abstract
Chromatin modulation provides a key checkpoint for controlling cell cycle regulated gene networks. The replicative canonical histone genes are one such gene family under tight regulation during cell division. These genes are most highly expressed during S phase when histones are needed to chromatinize the new DNA template. While this fact has been known for a while, limited knowledge exists about the specific chromatin regulators controlling their temporal expression during cell cycle. Since histones and their associated mutations are emerging as major players in diseases such as cancer, identifying the chromatin factors modulating their expression is critical. The histone lysine tri-demethylase KDM4A is regulated over cell cycle and plays a direct role in DNA replication timing, site-specific rereplication, and DNA amplifications during S phase. Here, we establish an unappreciated role for the catalytically active KDM4A in directly regulating canonical replicative histone gene networks during cell cycle. Of interest, we further demonstrate that KDM4A interacts with proteins controlling histone expression and RNA processing (i.e., hnRNPUL1 and FUS/TLS). Together, this study provides a new function for KDM4A in modulating canonical histone gene expression., Competing Interests: Declaration of competing interest J.R. Whetstine has received funding from AstraZeneca, is a consultant at Qsonica, previously consulted for Celgene, and served on the Salarius Pharmaceuticals advisory board and served as a consultant. The other authors do not declare any conflict of interest.(5), (Copyright © 2020. Published by Elsevier B.V.)
- Published
- 2020
- Full Text
- View/download PDF
12. Histone Lysine Methylation Dynamics Control EGFR DNA Copy-Number Amplification.
- Author
-
Clarke TL, Tang R, Chakraborty D, Van Rechem C, Ji F, Mishra S, Ma A, Kaniskan HÜ, Jin J, Lawrence MS, Sadreyev RI, and Whetstine JR
- Subjects
- Antineoplastic Combined Chemotherapy Protocols pharmacology, Cell Hypoxia genetics, Cell Line, Tumor, DNA Copy Number Variations drug effects, DNA Methylation drug effects, Epigenesis, Genetic drug effects, ErbB Receptors antagonists & inhibitors, ErbB Receptors genetics, Gene Amplification drug effects, Gene Expression Regulation, Neoplastic drug effects, Histone-Lysine N-Methyltransferase antagonists & inhibitors, Histone-Lysine N-Methyltransferase metabolism, Humans, Jumonji Domain-Containing Histone Demethylases antagonists & inhibitors, Jumonji Domain-Containing Histone Demethylases metabolism, Lysine metabolism, Neoplasms drug therapy, Neoplasms pathology, Protein Kinase Inhibitors pharmacology, Protein Kinase Inhibitors therapeutic use, Antineoplastic Combined Chemotherapy Protocols therapeutic use, DNA Methylation genetics, Histones metabolism, Neoplasms genetics
- Abstract
Acquired chromosomal DNA copy gains are a feature of many tumors; however, the mechanisms that underpin oncogene amplification are poorly understood. Recent studies have begun to uncover the importance of epigenetic states and histone lysine methyltransferases (KMT) and demethylases (KDM) in regulating transient site-specific DNA copy-number gains (TSSG). In this study, we reveal a critical interplay between a myriad of lysine methyltransferases and demethylases in modulating H3K4/9/27 methylation balance to control extrachromosomal amplification of the EGFR oncogene. This study further establishes that cellular signals (hypoxia and EGF) are able to directly promote EGFR amplification through modulation of the enzymes controlling EGFR copy gains. Moreover, we demonstrate that chemical inhibitors targeting specific KMTs and KDMs are able to promote or block extrachromosomal EGFR amplification, which identifies potential therapeutic strategies for controlling EGFR copy-number heterogeneity in cancer, and, in turn, drug response. SIGNIFICANCE: This study identifies a network of epigenetic factors and cellular signals that directly control EGFR DNA amplification. We demonstrate that chemical inhibitors targeting enzymes controlling this amplification can be used to rheostat EGFR copy number, which uncovers therapeutic opportunities for controlling EGFR DNA amplification heterogeneity and the associated drug response. This article is highlighted in the In This Issue feature, p. 161 ., (©2019 American Association for Cancer Research.)
- Published
- 2020
- Full Text
- View/download PDF
13. PRC2 Plays Red Light, Green Light with MHC-I and CD8 + T Cells.
- Author
-
Whetstine JR
- Subjects
- CD8-Positive T-Lymphocytes immunology, Histocompatibility Antigens Class I, Humans, Immune Evasion, T-Lymphocytes, Cytotoxic immunology, Antigen Presentation, Neoplasms
- Abstract
In this issue of Cancer Cell, Burr et al. report that PRC2 plays a conserved role in silencing antigen presentation and processing genes and, in turn, CD8
+ T cell activation. Furthermore, PRC2-targeted therapeutics overcome gene silencing and promote tumor clearance by cytotoxic T cells., (Copyright © 2019 Elsevier Inc. All rights reserved.)- Published
- 2019
- Full Text
- View/download PDF
14. The Histone Deacetylase SIRT6 Restrains Transcription Elongation via Promoter-Proximal Pausing.
- Author
-
Etchegaray JP, Zhong L, Li C, Henriques T, Ablondi E, Nakadai T, Van Rechem C, Ferrer C, Ross KN, Choi JE, Samarakkody A, Ji F, Chang A, Sadreyev RI, Ramaswamy S, Nechaev S, Whetstine JR, Roeder RG, Adelman K, Goren A, and Mostoslavsky R
- Subjects
- Acetylation, Cell Cycle Proteins genetics, Cell Cycle Proteins metabolism, Cell Line, Gene Deletion, Histones genetics, Histones metabolism, Humans, Proto-Oncogene Proteins c-myc genetics, Proto-Oncogene Proteins c-myc metabolism, RNA Polymerase II genetics, Sirtuins genetics, Transcription Factors genetics, Transcription Factors metabolism, Transcriptional Elongation Factors genetics, Transcriptional Elongation Factors metabolism, Promoter Regions, Genetic, RNA Polymerase II metabolism, Sirtuins metabolism, Transcription Elongation, Genetic
- Abstract
Transcriptional regulation in eukaryotes occurs at promoter-proximal regions wherein transcriptionally engaged RNA polymerase II (Pol II) pauses before proceeding toward productive elongation. The role of chromatin in pausing remains poorly understood. Here, we demonstrate that the histone deacetylase SIRT6 binds to Pol II and prevents the release of the negative elongation factor (NELF), thus stabilizing Pol II promoter-proximal pausing. Genetic depletion of SIRT6 or its chromatin deficiency upon glucose deprivation causes intragenic enrichment of acetylated histone H3 at lysines 9 (H3K9ac) and 56 (H3K56ac), activation of cyclin-dependent kinase 9 (CDK9)-that phosphorylates NELF and the carboxyl terminal domain of Pol II-and enrichment of the positive transcription elongation factors MYC, BRD4, PAF1, and the super elongation factors AFF4 and ELL2. These events lead to increased expression of genes involved in metabolism, protein synthesis, and embryonic development. Our results identified SIRT6 as a Pol II promoter-proximal pausing-dedicated histone deacetylase., (Copyright © 2019 Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
15. Cross-talk between Lysine-Modifying Enzymes Controls Site-Specific DNA Amplifications.
- Author
-
Mishra S, Van Rechem C, Pal S, Clarke TL, Chakraborty D, Mahan SD, Black JC, Murphy SE, Lawrence MS, Daniels DL, and Whetstine JR
- Published
- 2018
- Full Text
- View/download PDF
16. E2F/DP Prevents Cell-Cycle Progression in Endocycling Fat Body Cells by Suppressing dATM Expression.
- Author
-
Guarner A, Morris R, Korenjak M, Boukhali M, Zappia MP, Van Rechem C, Whetstine JR, Ramaswamy S, Zou L, Frolov MV, Haas W, and Dyson NJ
- Subjects
- Animals, Ataxia Telangiectasia Mutated Proteins genetics, Ataxia Telangiectasia Mutated Proteins metabolism, Cell Cycle genetics, Cell Cycle Proteins metabolism, Cell Division physiology, DNA Replication, DNA-Binding Proteins metabolism, Drosophila, Drosophila Proteins biosynthesis, Drosophila Proteins genetics, Fat Body cytology, Protein Serine-Threonine Kinases, Trans-Activators genetics, Transcriptome, Ataxia Telangiectasia Mutated Proteins biosynthesis, Drosophila Proteins metabolism, E2F Transcription Factors genetics, E2F Transcription Factors metabolism, Fat Body physiology, Trans-Activators metabolism
- Abstract
To understand the consequences of the complete elimination of E2F regulation, we profiled the proteome of Drosophila dDP mutants that lack functional E2F/DP complexes. The results uncovered changes in the larval fat body, a differentiated tissue that grows via endocycles. We report an unexpected mechanism of E2F/DP action that promotes quiescence in this tissue. In the fat body, dE2F/dDP limits cell-cycle progression by suppressing DNA damage responses. Loss of dDP upregulates dATM, allowing cells to sense and repair DNA damage and increasing replication of loci that are normally under-replicated in wild-type tissues. Genetic experiments show that ectopic dATM is sufficient to promote DNA synthesis in wild-type fat body cells. Strikingly, reducing dATM levels in dDP-deficient fat bodies restores cell-cycle control, improves tissue morphology, and extends animal development. These results show that, in some cellular contexts, dE2F/dDP-dependent suppression of DNA damage signaling is key for cell-cycle control and needed for normal development., (Copyright © 2017 Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
17. Radiation Resistance in KRAS-Mutated Lung Cancer Is Enabled by Stem-like Properties Mediated by an Osteopontin-EGFR Pathway.
- Author
-
Wang M, Han J, Marcar L, Black J, Liu Q, Li X, Nagulapalli K, Sequist LV, Mak RH, Benes CH, Hong TS, Gurtner K, Krause M, Baumann M, Kang JX, Whetstine JR, and Willers H
- Subjects
- A549 Cells, Animals, Carcinoma, Non-Small-Cell Lung metabolism, Carcinoma, Non-Small-Cell Lung pathology, Female, Heterografts, Humans, Lung Neoplasms metabolism, Lung Neoplasms pathology, Male, Mice, Mice, Nude, Mutation, Neoplastic Stem Cells metabolism, Neoplastic Stem Cells pathology, Neoplastic Stem Cells radiation effects, Osteopontin biosynthesis, Osteopontin genetics, Proto-Oncogene Proteins p21(ras) metabolism, Radiation Tolerance genetics, Signal Transduction, Carcinoma, Non-Small-Cell Lung genetics, Carcinoma, Non-Small-Cell Lung radiotherapy, ErbB Receptors metabolism, Lung Neoplasms genetics, Lung Neoplasms radiotherapy, Osteopontin metabolism, Proto-Oncogene Proteins p21(ras) genetics
- Abstract
Lung cancers with activating KRAS mutations are characterized by treatment resistance and poor prognosis. In particular, the basis for their resistance to radiation therapy is poorly understood. Here, we describe a radiation resistance phenotype conferred by a stem-like subpopulation characterized by mitosis-like condensed chromatin (MLCC), high CD133 expression, invasive potential, and tumor-initiating properties. Mechanistic investigations defined a pathway involving osteopontin and the EGFR in promoting this phenotype. Osteopontin/EGFR-dependent MLCC protected cells against radiation-induced DNA double-strand breaks and repressed putative negative regulators of stem-like properties, such as CRMP1 and BIM. The MLCC-positive phenotype defined a subset of KRAS-mutated lung cancers that were enriched for co-occurring genomic alterations in TP53 and CDKN2A. Our results illuminate the basis for the radiation resistance of KRAS-mutated lung cancers, with possible implications for prognostic and therapeutic strategies. Cancer Res; 77(8); 2018-28. ©2017 AACR ., (©2017 American Association for Cancer Research.)
- Published
- 2017
- Full Text
- View/download PDF
18. Regulation of Transient Site-specific Copy Gain by MicroRNA.
- Author
-
Black JC, Zhang H, Kim J, Getz G, and Whetstine JR
- Subjects
- CDC2-CDC28 Kinases genetics, CDC2-CDC28 Kinases metabolism, Cell Line, Tumor, Female, Humans, Breast Neoplasms genetics, Gene Amplification, Gene Expression Regulation, Neoplastic, Jumonji Domain-Containing Histone Demethylases genetics, MicroRNAs genetics
- Abstract
Intra-tumor copy number heterogeneity is commonly observed in cancer; however, the molecular mechanisms that contribute to heterogeneity remain poorly understood. Up-regulation of the histone demethylase KDM4A promotes transient site-specific copy gain (TSSG) in cells; therefore, uncovering how KDM4A levels are controlled is important for understanding the regulation of copy number heterogeneity. Here, we demonstrate that KDM4A is regulated by hsa-mir-23a-3p, hsa-mir-23b-3p, and hsa-mir-137. Altering expression of these microRNAs (miRNAs) regulates KDM4A-dependent TSSG. miRNA inhibition promoted copy gains and increased expression of the drug-resistant oncogene CKS1B, which was further substantiated in primary breast tumors. Consistent with increased CKS1B expression, miRNA inhibition reduced breast cancer cell sensitivity to cisplatin. Our data identify these miRNAs as regulators of TSSG and copy gains of a drug resistance gene., (© 2016 by The American Society for Biochemistry and Molecular Biology, Inc.)
- Published
- 2016
- Full Text
- View/download PDF
19. Different Facets of Copy Number Changes: Permanent, Transient, and Adaptive.
- Author
-
Mishra S and Whetstine JR
- Subjects
- Adaptation, Biological, Animals, Drug Resistance genetics, Genetic Heterogeneity, Genomic Structural Variation, Humans, Neoplasms genetics, Chromosome Aberrations, Gene Dosage
- Abstract
Chromosomal copy number changes are frequently associated with harmful consequences and are thought of as an underlying mechanism for the development of diseases. However, changes in copy number are observed during development and occur during normal biological processes. In this review, we highlight the causes and consequences of copy number changes in normal physiologic processes as well as cover their associations with cancer and acquired drug resistance. We discuss the permanent and transient nature of copy number gains and relate these observations to a new mechanism driving transient site-specific copy gains (TSSGs). Finally, we discuss implications of TSSGs in generating intratumoral heterogeneity and tumor evolution and how TSSGs can influence the therapeutic response in cancer., (Copyright © 2016 Mishra and Whetstine.)
- Published
- 2016
- Full Text
- View/download PDF
20. RNF2 E3 or Not to E3: Dual Roles of RNF2 Overexpression in Melanoma.
- Author
-
Black JC and Whetstine JR
- Subjects
- Humans, Melanoma, Polycomb Repressive Complex 1, Ubiquitin-Protein Ligases
- Abstract
RNF2/RING1B is amplified and overexpressed in numerous tumors and contributes to tumorigenicity; however, the biologic importance is poorly understood. Surprisingly, the role of RNF2 in tumorigenesis and invasion can be separated into catalytically independent and catalytically dependent processes., (©2015 American Association for Cancer Research.)
- Published
- 2015
- Full Text
- View/download PDF
21. SETD1A modulates cell cycle progression through a miRNA network that regulates p53 target genes.
- Author
-
Tajima K, Yae T, Javaid S, Tam O, Comaills V, Morris R, Wittner BS, Liu M, Engstrom A, Takahashi F, Black JC, Ramaswamy S, Shioda T, Hammell M, Haber DA, Whetstine JR, and Maheswaran S
- Subjects
- Animals, Carcinogenesis, Cell Line, Tumor, Female, Humans, Male, Mice, Nude, Neoplasms, Experimental, Promoter Regions, Genetic, Tumor Suppressor Protein p53 metabolism, Cell Cycle, Gene Expression Regulation, Neoplastic, Histone-Lysine N-Methyltransferase metabolism, Immediate-Early Proteins metabolism, MicroRNAs metabolism, Tumor Suppressor Proteins metabolism
- Abstract
Expression of the p53-inducible antiproliferative gene BTG2 is suppressed in many cancers in the absence of inactivating gene mutations, suggesting alternative mechanisms of silencing. Using a shRNA screen targeting 43 histone lysine methyltransferases (KMTs), we show that SETD1A suppresses BTG2 expression through its induction of several BTG2-targeting miRNAs. This indirect but highly specific mechanism, by which a chromatin regulator that mediates transcriptional activating marks can lead to the downregulation of a critical effector gene, is shared with multiple genes in the p53 pathway. Through such miRNA-dependent effects, SETD1A regulates cell cycle progression in vitro and modulates tumorigenesis in mouse xenograft models. Together, these observations help explain the remarkably specific genetic consequences associated with alterations in generic chromatin modulators in cancer.
- Published
- 2015
- Full Text
- View/download PDF
22. Hypoxia drives transient site-specific copy gain and drug-resistant gene expression.
- Author
-
Black JC, Atabakhsh E, Kim J, Biette KM, Van Rechem C, Ladd B, Burrowes PD, Donado C, Mattoo H, Kleinstiver BP, Song B, Andriani G, Joung JK, Iliopoulos O, Montagna C, Pillai S, Getz G, and Whetstine JR
- Subjects
- Animals, CDC2-CDC28 Kinases genetics, Cell Hypoxia genetics, Cell Line, Cell Proliferation, Cells, Cultured, Drug Resistance, Neoplasm genetics, Humans, Zebrafish, Cell Hypoxia physiology, DNA Copy Number Variations genetics, Gene Expression Regulation
- Abstract
Copy number heterogeneity is a prominent feature within tumors. The molecular basis for this heterogeneity remains poorly characterized. Here, we demonstrate that hypoxia induces transient site-specific copy gains (TSSGs) in primary, nontransformed, and transformed human cells. Hypoxia-driven copy gains are not dependent on HIF1α or HIF2α; however, they are dependent on the KDM4A histone demethylase and are blocked by inhibition of KDM4A with a small molecule or the natural metabolite succinate. Furthermore, this response is conserved at a syntenic region in zebrafish cells. Regions with site-specific copy gain are also enriched for amplifications in hypoxic primary tumors. These tumors exhibited amplification and overexpression of the drug resistance gene CKS1B, which we recapitulated in hypoxic breast cancer cells. Our results demonstrate that hypoxia provides a biological stimulus to create transient site-specific copy alterations that could result in heterogeneity within tumors and cell populations. These findings have major implications in our understanding of copy number heterogeneity and the emergence of drug resistance genes in cancer., (© 2015 Black et al.; Published by Cold Spring Harbor Laboratory Press.)
- Published
- 2015
- Full Text
- View/download PDF
23. A coding single-nucleotide polymorphism in lysine demethylase KDM4A associates with increased sensitivity to mTOR inhibitors.
- Author
-
Van Rechem C, Black JC, Greninger P, Zhao Y, Donado C, Burrowes PD, Ladd B, Christiani DC, Benes CH, and Whetstine JR
- Subjects
- Alleles, Cell Line, Cell Line, Tumor, DNA Mutational Analysis, Gene Frequency, Humans, Jumonji Domain-Containing Histone Demethylases metabolism, Neoplasms drug therapy, Neoplasms genetics, Neoplasms mortality, Prognosis, Ubiquitination, Antineoplastic Agents pharmacology, Drug Resistance, Neoplasm genetics, Jumonji Domain-Containing Histone Demethylases genetics, Lysine genetics, Open Reading Frames, Polymorphism, Single Nucleotide, Protein Kinase Inhibitors pharmacology, TOR Serine-Threonine Kinases antagonists & inhibitors
- Abstract
Unlabelled: SNPs occur within chromatin-modulating factors; however, little is known about how these variants within the coding sequence affect cancer progression or treatment. Therefore, there is a need to establish their biochemical and/or molecular contribution, their use in subclassifying patients, and their impact on therapeutic response. In this report, we demonstrate that coding SNP-A482 within the lysine tridemethylase gene KDM4A/JMJD2A has different allelic frequencies across ethnic populations, associates with differential outcome in patients with non-small cell lung cancer (NSCLC), and promotes KDM4A protein turnover. Using an unbiased drug screen against 87 preclinical and clinical compounds, we demonstrate that homozygous SNP-A482 cells have increased mTOR inhibitor sensitivity. mTOR inhibitors significantly reduce SNP-A482 protein levels, which parallels the increased drug sensitivity observed with KDM4A depletion. Our data emphasize the importance of using variant status as candidate biomarkers and highlight the importance of studying SNPs in chromatin modifiers to achieve better targeted therapy., Significance: This report documents the first coding SNP within a lysine demethylase that associates with worse outcome in patients with NSCLC. We demonstrate that this coding SNP alters the protein turnover and associates with increased mTOR inhibitor sensitivity, which identifies a candidate biomarker for mTOR inhibitor therapy and a therapeutic target for combination therapy., (©2015 American Association for Cancer Research.)
- Published
- 2015
- Full Text
- View/download PDF
24. Lysine demethylase KDM4A associates with translation machinery and regulates protein synthesis.
- Author
-
Van Rechem C, Black JC, Boukhali M, Aryee MJ, Gräslund S, Haas W, Benes CH, and Whetstine JR
- Subjects
- Aminopyridines pharmacology, Drug Resistance genetics, Humans, Hydrazones pharmacology, Jumonji Domain-Containing Histone Demethylases antagonists & inhibitors, Methylation, Peptide Chain Initiation, Translational, Peptide Initiation Factors metabolism, Protein Binding, Protein Kinase Inhibitors pharmacology, Ribosomal Proteins metabolism, TOR Serine-Threonine Kinases antagonists & inhibitors, Jumonji Domain-Containing Histone Demethylases genetics, Jumonji Domain-Containing Histone Demethylases metabolism, Lysine metabolism, Protein Biosynthesis drug effects
- Abstract
Unlabelled: Chromatin-modifying enzymes are predominantly nuclear; however, these factors are also localized to the cytoplasm, and very little is known about their role in this compartment. In this report, we reveal a non-chromatin-linked role for the lysine-specific demethylase KDM4A. We demonstrate that KDM4A interacts with the translation initiation complex and affects the distribution of translation initiation factors within polysome fractions. Furthermore, KDM4A depletion reduced protein synthesis and enhanced the protein synthesis suppression observed with mTOR inhibitors, which paralleled an increased sensitivity to these drugs. Finally, we demonstrate that JIB-04, a JmjC demethylase inhibitor, suppresses translation initiation and enhances mTOR inhibitor sensitivity. These data highlight an unexpected cytoplasmic role for KDM4A in regulating protein synthesis and suggest novel potential therapeutic applications for this class of enzyme., Significance: This report documents an unexpected cytoplasmic role for the lysine demethylase KDM4A. We demonstrate that KDM4A interacts with the translation initiation machinery, regulates protein synthesis and, upon coinhibition with mTOR inhibitors, enhances the translation suppression and cell sensitivity to these therapeutics., (©2015 American Association for Cancer Research.)
- Published
- 2015
- Full Text
- View/download PDF
25. Too little O2 Too much gain.
- Author
-
Black JC and Whetstine JR
- Subjects
- Animals, Humans, Cell Hypoxia physiology, DNA Copy Number Variations genetics, Gene Expression Regulation
- Published
- 2015
- Full Text
- View/download PDF
26. Methylation: a multifaceted modification - looking at transcription and beyond.
- Author
-
Whetstine JR
- Subjects
- Animals, Epistasis, Genetic, Genome, Histone Methyltransferases, Humans, Protein Processing, Post-Translational, DNA Methylation physiology, DNA Modification Methylases physiology, Histone-Lysine N-Methyltransferase physiology, Lysine metabolism, Transcription, Genetic
- Published
- 2014
- Full Text
- View/download PDF
27. Examining the impact of gene variants on histone lysine methylation.
- Author
-
Van Rechem C and Whetstine JR
- Subjects
- Animals, Histone Demethylases metabolism, Histone-Lysine N-Methyltransferase metabolism, Histones metabolism, Humans, Methylation, Neoplasms genetics, Neoplasms metabolism, Histone Demethylases genetics, Histone-Lysine N-Methyltransferase genetics, Histones genetics, Lysine metabolism, Mutation, Polymorphism, Single Nucleotide
- Abstract
In recent years, there has been a boom in the amount of genome-wide sequencing data that has uncovered important and unappreciated links between certain genes, families of genes and enzymatic processes and diseases such as cancer. Such studies have highlighted the impact that chromatin modifying enzymes could have in cancer and other genetic diseases. In this review, we summarize characterized mutations and single nucleotide polymorphisms (SNPs) in histone lysine methyltransferases (KMTs), histone lysine demethylases (KDMs) and histones. We primarily focus on variants with strong disease correlations and discuss how they could impact histone lysine methylation dynamics and gene regulation., (Copyright © 2014 Elsevier B.V. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
28. Dynamic chromatin modification sustains epithelial-mesenchymal transition following inducible expression of Snail-1.
- Author
-
Javaid S, Zhang J, Anderssen E, Black JC, Wittner BS, Tajima K, Ting DT, Smolen GA, Zubrowski M, Desai R, Maheswaran S, Ramaswamy S, Whetstine JR, and Haber DA
- Subjects
- Acetylation, Carcinogenesis metabolism, Epigenesis, Genetic, Epithelial Cells drug effects, Epithelial Cells metabolism, Epithelial Cells physiology, Gene Expression Regulation, Neoplastic, Histone Deacetylase Inhibitors pharmacology, Histone Deacetylases genetics, Histone Deacetylases metabolism, Humans, MCF-7 Cells, Methylation, Promoter Regions, Genetic, Snail Family Transcription Factors, Transcription Factors genetics, Chromatin metabolism, Chromatin Assembly and Disassembly, Epithelial-Mesenchymal Transition, Protein Processing, Post-Translational, Transcription Factors metabolism
- Abstract
Epithelial-mesenchymal transition (EMT) is thought to contribute to cancer metastasis, but its underlying mechanisms are not well understood. To define early steps in this cellular transformation, we analyzed human mammary epithelial cells with tightly regulated expression of Snail-1, a master regulator of EMT. After Snail-1 induction, epithelial markers were repressed within 6 hr, and mesenchymal genes were induced at 24 hr. Snail-1 binding to its target promoters was transient (6-48 hr) despite continued protein expression, and it was followed by both transient and long-lasting chromatin changes. Pharmacological inhibition of selected histone acetylation and demethylation pathways suppressed the induction as well as the maintenance of Snail-1-mediated EMT. Thus, EMT involves an epigenetic switch that may be prevented or reversed with the use of small-molecule inhibitors of chromatin modifiers., (Copyright © 2013 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
29. Pharmacological inhibition of a microRNA family in nonhuman primates by a seed-targeting 8-mer antimiR.
- Author
-
Rottiers V, Obad S, Petri A, McGarrah R, Lindholm MW, Black JC, Sinha S, Goody RJ, Lawrence MS, deLemos AS, Hansen HF, Whittaker S, Henry S, Brookes R, Najafi-Shoushtari SH, Chung RT, Whetstine JR, Gerszten RE, Kauppinen S, and Näär AM
- Subjects
- Animals, Cholesterol, HDL blood, Female, Hep G2 Cells, Humans, Mice, Mice, Inbred C57BL, MicroRNAs genetics, Primates, Gene Silencing, MicroRNAs antagonists & inhibitors
- Abstract
MicroRNAs (miRNAs) regulate many aspects of human biology. They target mRNAs for translational repression or degradation through base pairing with 3' untranslated regions, primarily via seed sequences (nucleotides 2 to 8 in the mature miRNA sequence). A number of individual miRNAs and miRNA families share seed sequences and targets, but differ in the sequences outside of the seed. miRNAs have been implicated in the etiology of a wide variety of human diseases and therefore represent promising therapeutic targets. However, potential redundancy of different miRNAs sharing the same seed sequence and the challenge of simultaneously targeting miRNAs that differ significantly in nonseed sequences complicate therapeutic targeting approaches. We recently demonstrated effective inhibition of entire miRNA families using seed-targeting 8-mer locked nucleic acid (LNA)-modified antimiRs in short-term experiments in mammalian cells and in mice. However, the long-term efficacy and safety of this approach in higher organisms, such as humans and nonhuman primates, have not been determined. We show that pharmacological inhibition of the miR-33 family, key regulators of cholesterol/lipid homeostasis, by a subcutaneously delivered 8-mer LNA-modified antimiR in obese and insulin-resistant nonhuman primates results in derepression of miR-33 targets, such as ABCA1, increases circulating high-density lipoprotein cholesterol, and is well tolerated over 108 days of treatment. These findings demonstrate the efficacy and safety of an 8-mer LNA-antimiR against an miRNA family in a nonhuman primate metabolic disease model, suggesting that this could be a feasible approach for therapeutic targeting of miRNA families sharing the same seed sequence in human diseases.
- Published
- 2013
- Full Text
- View/download PDF
30. KDM4A lysine demethylase induces site-specific copy gain and rereplication of regions amplified in tumors.
- Author
-
Black JC, Manning AL, Van Rechem C, Kim J, Ladd B, Cho J, Pineda CM, Murphy N, Daniels DL, Montagna C, Lewis PW, Glass K, Allis CD, Dyson NJ, Getz G, and Whetstine JR
- Subjects
- Chromatin metabolism, Chromosomes, Human, Pair 1, Genomic Instability, HEK293 Cells, Humans, Jumonji Domain-Containing Histone Demethylases chemistry, Jumonji Domain-Containing Histone Demethylases genetics, Methylation, Neoplasms metabolism, Protein Structure, Tertiary, S Phase, DNA Replication, Gene Dosage, Jumonji Domain-Containing Histone Demethylases metabolism, Neoplasms genetics
- Abstract
Acquired chromosomal instability and copy number alterations are hallmarks of cancer. Enzymes capable of promoting site-specific copy number changes have yet to be identified. Here, we demonstrate that H3K9/36me3 lysine demethylase KDM4A/JMJD2A overexpression leads to localized copy gain of 1q12, 1q21, and Xq13.1 without global chromosome instability. KDM4A-amplified tumors have increased copy gains for these same regions. 1q12h copy gain occurs within a single cell cycle, requires S phase, and is not stable but is regenerated each cell division. Sites with increased copy number are rereplicated and have increased KDM4A, MCM, and DNA polymerase occupancy. Suv39h1/KMT1A or HP1γ overexpression suppresses the copy gain, whereas H3K9/K36 methylation interference promotes gain. Our results demonstrate that overexpression of a chromatin modifier results in site-specific copy gains. This begins to establish how copy number changes could originate during tumorigenesis and demonstrates that transient overexpression of specific chromatin modulators could promote these events., (Copyright © 2013 Elsevier Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
31. Detection of impaired homologous recombination repair in NSCLC cells and tissues.
- Author
-
Birkelbach M, Ferraiolo N, Gheorghiu L, Pfäffle HN, Daly B, Ebright MI, Spencer C, O'Hara C, Whetstine JR, Benes CH, Sequist LV, Zou L, Dahm-Daphi J, Kachnic LA, and Willers H
- Subjects
- Antibiotics, Antineoplastic pharmacology, BRCA1 Protein metabolism, Carcinoma, Non-Small-Cell Lung diagnosis, Carcinoma, Non-Small-Cell Lung drug therapy, Cisplatin pharmacology, DNA Damage drug effects, DNA Damage genetics, Fanconi Anemia Complementation Group D2 Protein metabolism, Humans, Immunoenzyme Techniques, Lung Neoplasms diagnosis, Lung Neoplasms drug therapy, Microscopy, Fluorescence, Mitomycin pharmacology, Phthalazines pharmacology, Piperazines pharmacology, Poly (ADP-Ribose) Polymerase-1, Rad51 Recombinase metabolism, Recombinational DNA Repair drug effects, Tumor Cells, Cultured, Tumor Stem Cell Assay, Antineoplastic Agents pharmacology, Carcinoma, Non-Small-Cell Lung genetics, Lung Neoplasms genetics, Poly(ADP-ribose) Polymerase Inhibitors, Recombination, Genetic genetics, Recombinational DNA Repair genetics
- Abstract
Introduction: Homologous recombination repair (HRR) is a critical pathway for the repair of DNA damage caused by cisplatin or poly-ADP ribose polymerase (PARP) inhibitors. HRR may be impaired by multiple mechanisms in cancer, which complicates assessing the functional HRR status in cells. Here, we monitored the ability of non-small-cell lung cancer (NSCLC) cells to form subnuclear foci of DNA repair proteins as a surrogate of HRR proficiency., Methods: We assessed clonogenic survival of 16 NSCLC cell lines in response to cisplatin, mitomycin C (MMC), and the PARP inhibitor olaparib. Thirteen tumor explants from patients with NSCLC were subjected to cisplatin ex vivo. Cells were assayed for foci of repair-associated proteins such as BRCA1, FANCD2, RAD51, and γ-H2AX., Results: Four cell lines (25%) showed an impaired RAD51 foci-forming ability in response to cisplatin. Impaired foci formation correlated with cellular sensitivity to cisplatin, MMC and olaparib. Foci responses complemented or superseded genomic information suggesting alterations in the ATM/ATR and FA/BRCA pathways. Because baseline foci in untreated cells did not predict drug sensitivity, we adapted an ex vivo biomarker assay to monitor damage-induced RAD51 foci in NSCLC explants from patients. Ex vivo cisplatin treatment of explants identified two tumors (15%) exhibiting compromised RAD51 foci induction., Conclusions: A fraction of NSCLC harbors HRR defects that may sensitize the affected tumors to DNA-damaging agents including PARP inhibitors. We propose that foci-based functional biomarker assays represent a powerful tool for prospective determination of treatment sensitivity, but will require ex vivo techniques for induction of DNA damage to unmask the underlying HRR defect.
- Published
- 2013
- Full Text
- View/download PDF
32. Tipping the lysine methylation balance in disease.
- Author
-
Black JC and Whetstine JR
- Subjects
- Histone-Lysine N-Methyltransferase genetics, Histone-Lysine N-Methyltransferase metabolism, Humans, Methylation, Mutation, Neoplasms genetics, Nervous System Diseases genetics, Lysine metabolism, Neoplasms physiopathology, Nervous System Diseases physiopathology
- Abstract
Genomic instability is a major contributing factor to the development and onset of diseases such as cancer. Emerging evidence has demonstrated that maintaining the proper balance of histone lysine methylation is critical to preserve genomic integrity. Genome-wide association studies, gene sequencing, and genome-wide mapping approaches have helped identify mutations, copy number changes, and aberrant gene regulation of lysine methyltransferases (KMTs) and demethylases (KDMs) associated with cancer and cognitive disorders. Structural analysis of KMTs and KDMs has demonstrated the drugability of these enzymes and has led to the discovery of small molecule inhibitors. Use of these inhibitors has allowed better understanding of the biochemical properties of KMTs and KDMs and demonstrated potential for therapeutic use. This review will highlight the methyl modifications, KMTs and KDMs associated with cancer and neurological disorders and how KMT and KDM and the potential for treatment of these conditions with small molecule inhibitors., (Copyright © 2012 Wiley Periodicals, Inc.)
- Published
- 2013
- Full Text
- View/download PDF
33. Histone lysine methylation dynamics: establishment, regulation, and biological impact.
- Author
-
Black JC, Van Rechem C, and Whetstine JR
- Subjects
- Animals, Histone Demethylases metabolism, Histone-Lysine N-Methyltransferase metabolism, Histones chemistry, Histones genetics, Humans, Lysine chemistry, Lysine genetics, Methylation, Histones metabolism, Lysine metabolism
- Abstract
Histone lysine methylation has emerged as a critical player in the regulation of gene expression, cell cycle, genome stability, and nuclear architecture. Over the past decade, a tremendous amount of progress has led to the characterization of methyl modifications and the lysine methyltransferases (KMTs) and lysine demethylases (KDMs) that regulate them. Here, we review the discovery and characterization of the KMTs and KDMs and the methyl modifications they regulate. We discuss the localization of the KMTs and KDMs as well as the distribution of lysine methylation throughout the genome. We highlight how these data have shaped our view of lysine methylation as a key determinant of complex chromatin states. Finally, we discuss the regulation of KMTs and KDMs by proteasomal degradation, posttranscriptional mechanisms, and metabolic status. We propose key questions for the field and highlight areas that we predict will yield exciting discoveries in the years to come., (Copyright © 2012 Elsevier Inc. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
34. RBF binding to both canonical E2F targets and noncanonical targets depends on functional dE2F/dDP complexes.
- Author
-
Korenjak M, Anderssen E, Ramaswamy S, Whetstine JR, and Dyson NJ
- Subjects
- Animals, Binding Sites, Cell Cycle Proteins genetics, Cell Cycle Proteins metabolism, Chromatin metabolism, Chromatin Immunoprecipitation, DNA-Binding Proteins genetics, DNA-Binding Proteins metabolism, Drosophila Proteins deficiency, Drosophila Proteins genetics, Drosophila melanogaster genetics, Drosophila melanogaster growth & development, E2F Transcription Factors genetics, Larva metabolism, Mutation, Promoter Regions, Genetic, Protein Binding, Retinoblastoma Protein, Trans-Activators deficiency, Trans-Activators genetics, Transcription Factors genetics, Drosophila Proteins metabolism, Drosophila melanogaster metabolism, E2F Transcription Factors metabolism, Trans-Activators metabolism, Transcription Factors metabolism
- Abstract
The retinoblastoma (RB) family of proteins regulate transcription. These proteins lack intrinsic DNA-binding activity but are recruited to specific genomic locations through interactions with sequence-specific DNA-binding factors. The best-known target of RB protein (pRB) is the E2F transcription factor; however, many other chromatin-associated proteins have been described that may allow RB family members to act at additional sites. To gain a perspective on the scale of E2F-dependent and E2F-independent functions, we generated genome-wide binding profiles of RBF1 and dE2F proteins in Drosophila larvae. RBF1 and dE2F2 associate with a large number of binding sites at genes with diverse biological functions. In contrast, dE2F1 was detected at a smaller set of promoters, suggesting that it overrides repression by RBF1/dE2F2 at a specific subset of targets. Approximately 15% of RBF1-bound regions lacked consensus E2F-binding motifs. To test whether RBF1 action at these sites is E2F independent, we examined dDP mutant larvae that lack any functional dE2F/dDP heterodimers. As measured by chromatin immunoprecipitation-microarray analysis (ChIP-chip), ChIP-quantitative PCR (qPCR), and cell fractionation, the stable association of RBF1 with chromatin was eliminated in dDP mutants. This requirement for dDP was seen at classic E2F-regulated promoters and at promoters that lacked canonical E2F-binding sites. These results suggest that E2F/DP complexes are essential for all genomic targeting of RBF1.
- Published
- 2012
- Full Text
- View/download PDF
35. LOX out, histones: a new enzyme is nipping at your tails.
- Author
-
Black JC and Whetstine JR
- Abstract
In the current issue of Molecular Cell, Herranz et al. (2012) demonstrate that LOXL2 deaminates trimethylated histone 3 lysine 4 (H3K4me3), which uncovers a new chromatin modification and a new enzymatic mechanism with the potential to regulate additional lysine residues., (Copyright © 2012 Elsevier Inc. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
36. The SKP1-Cul1-F-box and leucine-rich repeat protein 4 (SCF-FbxL4) ubiquitin ligase regulates lysine demethylase 4A (KDM4A)/Jumonji domain-containing 2A (JMJD2A) protein.
- Author
-
Van Rechem C, Black JC, Abbas T, Allen A, Rinehart CA, Yuan GC, Dutta A, and Whetstine JR
- Subjects
- Base Sequence, Binding Sites, Cell Cycle, Chromatin chemistry, Cullin Proteins chemistry, DNA Replication, Histone Demethylases chemistry, Humans, Proteasome Endopeptidase Complex chemistry, Protein Binding, Protein Structure, Tertiary, Ubiquitin chemistry, F-Box Proteins chemistry, Jumonji Domain-Containing Histone Demethylases chemistry, S-Phase Kinase-Associated Proteins chemistry, Ubiquitin-Protein Ligases chemistry
- Abstract
Chromatin-modifying enzymes play a fundamental role in regulating chromatin structure so that DNA replication is spatially and temporally coordinated. For example, the lysine demethylase 4A/Jumonji domain-containing 2A (KDM4A/JMJD2A) is tightly regulated during the cell cycle. Overexpression of JMJD2A leads to altered replication timing and faster S phase progression. In this study, we demonstrate that degradation of JMJD2A is regulated by the proteasome. JMJD2A turnover is coordinated through the SKP1-Cul1-F-box ubiquitin ligase complex that contains cullin 1 and the F-box and leucine-rich repeat protein 4 (FbxL4). This complex interacted with JMJD2A. Ubiquitin overexpression restored turnover and blocked the JMJD2A-dependent faster S phase progression in a cullin 1-dependent manner. Furthermore, increased ubiquitin levels decreased JMJD2A occupancy and BrdU incorporation at target sites. This study highlights a finely tuned mechanism for regulating histone demethylase levels and emphasizes the need to tightly regulate chromatin modifiers so that the cell cycle occurs properly.
- Published
- 2011
- Full Text
- View/download PDF
37. Chromatin landscape: methylation beyond transcription.
- Author
-
Black JC and Whetstine JR
- Subjects
- Animals, Chromatin genetics, Chromobox Protein Homolog 5, Chromosomal Proteins, Non-Histone genetics, Chromosomal Proteins, Non-Histone metabolism, DNA genetics, DNA metabolism, Gene Duplication physiology, Histones genetics, Humans, Methylation, Chromatin metabolism, Histones metabolism, Protein Processing, Post-Translational physiology, Transcription, Genetic physiology
- Abstract
The nucleus is organized and compartmentalized into a highly ordered structure that contains DNA, RNA, chromosomal and histone proteins. The dynamics associated with these various components are responsible for making sure that the DNA is properly duplicated, genes are properly transcribed, and the genome is stabilized. It is no surprise that alterations in these various components are directly associated with pathologies like cancer. This Point of View focuses on the role the chromatin modification landscape, especially histone 3 lysine 9 methylation (H3K9me), and heterochromatin proteins (HP1) play in regulating DNA-templated processes, with a particular focus on their role at non-genic regions and effects on chromatin structure. These observations will be further extended to the role that alterations in chromatin landscape will contribute to diseases. This Point of View emphasizes that alterations in histone modification landscapes are not only relevant to transcription but have broad range implications in chromatin structure, nuclear architecture, cell cycle, genome stability and disease progression.
- Published
- 2011
- Full Text
- View/download PDF
38. Conserved antagonism between JMJD2A/KDM4A and HP1γ during cell cycle progression.
- Author
-
Black JC, Allen A, Van Rechem C, Forbes E, Longworth M, Tschöp K, Rinehart C, Quiton J, Walsh R, Smallwood A, Dyson NJ, and Whetstine JR
- Subjects
- Animals, Caenorhabditis elegans genetics, Caenorhabditis elegans metabolism, Cell Cycle Proteins genetics, Cell Cycle Proteins metabolism, Cells, Cultured, Chromatin Immunoprecipitation, Chromosomal Proteins, Non-Histone antagonists & inhibitors, Chromosomal Proteins, Non-Histone genetics, DNA Replication, Flow Cytometry, HeLa Cells, Humans, Jumonji Domain-Containing Histone Demethylases antagonists & inhibitors, Transfection, Cell Cycle, Chromatin metabolism, Chromosomal Proteins, Non-Histone metabolism, Jumonji Domain-Containing Histone Demethylases metabolism
- Abstract
The KDM4/JMJD2 family of histone demethylases is amplified in human cancers. However, little is known about their physiologic or tumorigenic roles. We have identified a conserved and unappreciated role for the JMJD2A/KDM4A H3K9/36 tridemethylase in cell cycle progression. We demonstrate that JMJD2A protein levels are regulated in a cell cycle-dependent manner and that JMJD2A overexpression increased chromatin accessibility, S phase progression, and altered replication timing of specific genomic loci. These phenotypes depended on JMJD2A enzymatic activity. Strikingly, depletion of the only C. elegans homolog, JMJD-2, slowed DNA replication and increased ATR/p53-dependent apoptosis. Importantly, overexpression of HP1γ antagonized JMJD2A-dependent progression through S phase, and depletion of HPL-2 rescued the DNA replication-related phenotypes in jmjd-2(-/-) animals. Our findings describe a highly conserved model whereby JMJD2A regulates DNA replication by antagonizing HP1γ and controlling chromatin accessibility., (Copyright © 2010 Elsevier Inc. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
39. Conserved role of SIRT1 orthologs in fasting-dependent inhibition of the lipid/cholesterol regulator SREBP.
- Author
-
Walker AK, Yang F, Jiang K, Ji JY, Watts JL, Purushotham A, Boss O, Hirsch ML, Ribich S, Smith JJ, Israelian K, Westphal CH, Rodgers JT, Shioda T, Elson SL, Mulligan P, Najafi-Shoushtari H, Black JC, Thakur JK, Kadyk LC, Whetstine JR, Mostoslavsky R, Puigserver P, Li X, Dyson NJ, Hart AC, and Näär AM
- Subjects
- Acetylation, Animals, Benzamides pharmacology, Caenorhabditis elegans, Cell Line, Cholesterol biosynthesis, HeLa Cells, Heterocyclic Compounds, 4 or More Rings pharmacology, Humans, Lipids biosynthesis, Mice, Naphthols pharmacology, Niacinamide pharmacology, Protein Stability drug effects, Sirtuins antagonists & inhibitors, Down-Regulation drug effects, Fasting physiology, Sirtuin 1 metabolism, Sterol Regulatory Element Binding Protein 1 metabolism, Sterol Regulatory Element Binding Protein 2 metabolism
- Abstract
The sterol regulatory element-binding protein (SREBP) transcription factor family is a critical regulator of lipid and sterol homeostasis in eukaryotes. In mammals, SREBPs are highly active in the fed state to promote the expression of lipogenic and cholesterogenic genes and facilitate fat storage. During fasting, SREBP-dependent lipid/cholesterol synthesis is rapidly diminished in the mouse liver; however, the mechanism has remained incompletely understood. Moreover, the evolutionary conservation of fasting regulation of SREBP-dependent programs of gene expression and control of lipid homeostasis has been unclear. We demonstrate here a conserved role for orthologs of the NAD(+)-dependent deacetylase SIRT1 in metazoans in down-regulation of SREBP orthologs during fasting, resulting in inhibition of lipid synthesis and fat storage. Our data reveal that SIRT1 can directly deacetylate SREBP, and modulation of SIRT1 activity results in changes in SREBP ubiquitination, protein stability, and target gene expression. In addition, chemical activators of SIRT1 inhibit SREBP target gene expression in vitro and in vivo, correlating with decreased hepatic lipid and cholesterol levels and attenuated liver steatosis in diet-induced and genetically obese mice. We conclude that SIRT1 orthologs play a critical role in controlling SREBP-dependent gene regulation governing lipid/cholesterol homeostasis in metazoans in response to fasting cues. These findings may have important biomedical implications for the treatment of metabolic disorders associated with aberrant lipid/cholesterol homeostasis, including metabolic syndrome and atherosclerosis.
- Published
- 2010
- Full Text
- View/download PDF
40. Histone H3 methylation links DNA damage detection to activation of the tumour suppressor Tip60.
- Author
-
Sun Y, Jiang X, Xu Y, Ayrapetov MK, Moreau LA, Whetstine JR, and Price BD
- Subjects
- Chromobox Protein Homolog 5, HeLa Cells, Humans, Lysine Acetyltransferase 5, DNA Damage, DNA Methylation, Genes, Tumor Suppressor, Histone Acetyltransferases genetics, Histones metabolism
- Abstract
DNA double-strand break (DSB) repair involves complex interactions between chromatin and repair proteins, including Tip60, a tumour suppressor. Tip60 is an acetyltransferase that acetylates both histones and ATM (ataxia telangiectasia mutated) kinase. Inactivation of Tip60 leads to defective DNA repair and increased cancer risk. However, how DNA damage activates the acetyltransferase activity of Tip60 is not known. Here, we show that direct interaction between the chromodomain of Tip60 and histone H3 trimethylated on lysine 9 (H3K9me3) at DSBs activates the acetyltransferase activity of Tip60. Depletion of intracellular H3K9me3 blocks activation of the acetyltransferase activity of Tip60, resulting in defective ATM activation and widespread defects in DSB repair. In addition, the ability of Tip60 to access H3K9me3 is dependent on the DNA damage-induced displacement of HP1beta (heterochromatin protein 1beta) from H3K9me3. Finally, we demonstrate that the Mre11-Rad50-Nbs1 (MRN) complex targets Tip60 to H3K9me3, and is required to activate the acetyltransferase activity of Tip60. These results reveal a new function for H3K9me3 in coordinating activation of Tip60-dependent DNA repair pathways, and imply that aberrant patterns of histone methylation may contribute to cancer by altering the efficiency of DSB repair.
- Published
- 2009
- Full Text
- View/download PDF
41. The conserved NAD(H)-dependent corepressor CTBP-1 regulates Caenorhabditis elegans life span.
- Author
-
Chen S, Whetstine JR, Ghosh S, Hanover JA, Gali RR, Grosu P, and Shi Y
- Subjects
- Aging physiology, Animals, Caenorhabditis elegans metabolism, Caenorhabditis elegans Proteins metabolism, Forkhead Transcription Factors, Insulin metabolism, Insulin-Like Growth Factor I metabolism, RNA Interference, Signal Transduction, Sirtuins metabolism, Transcription Factors metabolism, Triglycerides metabolism, Caenorhabditis elegans physiology, Caenorhabditis elegans Proteins physiology, Longevity physiology, NAD physiology, Repressor Proteins physiology
- Abstract
CtBP (C-terminal binding protein) is an evolutionarily conserved NAD(H)-dependent transcriptional corepressor, whose activity has been shown to be regulated by the NAD/NADH ratio. Although recent studies have provided significant new insights into mechanisms by which CtBP regulates transcription, the biological function of CtBP remains incompletely understood. Here, we report that genetic inactivation of the Caenorhabditis elegans homolog, ctbp-1, results in life span extension, which is suppressed by reintroduction of the ctbp-1 genomic DNA encoding wild-type but not NAD(H)-binding defective CTBP-1 protein. We show that CTBP-1 possibly modulates aging through the insulin/IGF-1 signaling pathway, dependent on the forkhead transcription factor DAF-16, but independent of the NAD-dependent histone deacetylase SIR-2.1. Genome-wide microarray analysis identifies >200 potential CTBP-1 target genes. Importantly, RNAi inhibition of a putative triacylglycerol lipase gene lips-7(C09E8.2) but not another lipase suppresses the life span extension phenotype. Consistently, metabolic analysis shows that the triacylglycerol level is reduced in the ctbp-1 deletion mutant, which is restored to the wild-type level by RNAi inhibition of lips-7. Taken together, our data suggest that CTBP-1 controls life span probably through the regulation of lipid metabolism.
- Published
- 2009
- Full Text
- View/download PDF
42. A histone H3 lysine 27 demethylase regulates animal posterior development.
- Author
-
Lan F, Bayliss PE, Rinn JL, Whetstine JR, Wang JK, Chen S, Iwase S, Alpatov R, Issaeva I, Canaani E, Roberts TM, Chang HY, and Shi Y
- Subjects
- Animals, Cell Line, Embryo, Nonmammalian embryology, Gene Expression Regulation, Developmental, Genes, Homeobox genetics, Genome genetics, Histone Demethylases, Humans, Jumonji Domain-Containing Histone Demethylases, Methylation, Mice, Nuclear Proteins genetics, Oxidoreductases, N-Demethylating genetics, Oxidoreductases, N-Demethylating metabolism, Transcription, Genetic genetics, Zebrafish genetics, Zebrafish Proteins genetics, Body Patterning, Histones metabolism, Lysine metabolism, Nuclear Proteins metabolism, Zebrafish embryology, Zebrafish Proteins metabolism
- Abstract
The recent discovery of a large number of histone demethylases suggests a central role for these enzymes in regulating histone methylation dynamics. Histone H3K27 trimethylation (H3K27me3) has been linked to polycomb-group-protein-mediated suppression of Hox genes and animal body patterning, X-chromosome inactivation and possibly maintenance of embryonic stem cell (ESC) identity. An imbalance of H3K27 methylation owing to overexpression of the methylase EZH2 has been implicated in metastatic prostate and aggressive breast cancers. Here we show that the JmjC-domain-containing related proteins UTX and JMJD3 catalyse demethylation of H3K27me3/2. UTX is enriched around the transcription start sites of many HOX genes in primary human fibroblasts, in which HOX genes are differentially expressed, but is selectively excluded from the HOX loci in ESCs, in which HOX genes are largely silent. Consistently, RNA interference inhibition of UTX led to increased H3K27me3 levels at some HOX gene promoters. Importantly, morpholino oligonucleotide inhibition of a zebrafish UTX homologue resulted in mis-regulation of hox genes and a striking posterior developmental defect, which was partially rescued by wild-type, but not by catalytically inactive, human UTX. Taken together, these findings identify a small family of H3K27 demethylases with important, evolutionarily conserved roles in H3K27 methylation regulation and in animal anterior-posterior development.
- Published
- 2007
- Full Text
- View/download PDF
43. The X-linked mental retardation gene SMCX/JARID1C defines a family of histone H3 lysine 4 demethylases.
- Author
-
Iwase S, Lan F, Bayliss P, de la Torre-Ubieta L, Huarte M, Qi HH, Whetstine JR, Bonni A, Roberts TM, and Shi Y
- Subjects
- Animals, Cell Line, Tumor, Cell Survival, DNA, Complementary, DNA-Binding Proteins genetics, DNA-Binding Proteins metabolism, Gene Library, Histone Demethylases, Histone-Lysine N-Methyltransferase genetics, Histone-Lysine N-Methyltransferase metabolism, Histones chemistry, Humans, Intracellular Signaling Peptides and Proteins genetics, Intracellular Signaling Peptides and Proteins metabolism, Jumonji Domain-Containing Histone Demethylases, Lysine metabolism, Methylation, Mice, Minor Histocompatibility Antigens, Neoplasm Proteins genetics, Neoplasm Proteins metabolism, Neurons cytology, Neurons metabolism, Oxidoreductases, N-Demethylating metabolism, Proteins metabolism, Retinoblastoma-Binding Protein 2, Tumor Suppressor Proteins genetics, Tumor Suppressor Proteins metabolism, Histones metabolism, X-Linked Intellectual Disability genetics, Oxidoreductases, N-Demethylating genetics, Proteins genetics
- Abstract
Histone methylation regulates chromatin structure and transcription. The recently identified histone demethylase lysine-specific demethylase 1 (LSD1) is chemically restricted to demethylation of only mono- and di- but not trimethylated histone H3 lysine 4 (H3K4me3). We show that the X-linked mental retardation (XLMR) gene SMCX (JARID1C), which encodes a JmjC-domain protein, reversed H3K4me3 to di- and mono- but not unmethylated products. Other SMCX family members, including SMCY, RBP2, and PLU-1, also demethylated H3K4me3. SMCX bound H3K9me3 via its N-terminal PHD (plant homeodomain) finger, which may help coordinate H3K4 demethylation and H3K9 methylation in transcriptional repression. Significantly, several XLMR-patient point mutations reduced SMCX demethylase activity and binding to H3K9me3 peptides, respectively. Importantly, studies in zebrafish and primary mammalian neurons demonstrated a role for SMCX in neuronal survival and dendritic development and a link to the demethylase activity. Our findings thus identify a family of H3K4me3 demethylases and uncover a critical link between histone modifications and XLMR.
- Published
- 2007
- Full Text
- View/download PDF
44. Dynamic regulation of histone lysine methylation by demethylases.
- Author
-
Shi Y and Whetstine JR
- Subjects
- Animals, Chromatin metabolism, Humans, Methylation, Oxidoreductases, N-Demethylating chemistry, Protein Structure, Tertiary, Histones metabolism, Lysine metabolism, Oxidoreductases, N-Demethylating metabolism
- Abstract
Recent studies demonstrated that histone methylation is not static but is dynamically regulated by histone methyltransferases and the newly discovered histone demethylases. This review discusses the chemical mechanisms for the known and potentially new classes of demethylases, the roles of these demethylases in chromatin and transcription, and their potential biological functions and connections to human diseases.
- Published
- 2007
- Full Text
- View/download PDF
45. Reversal of histone lysine trimethylation by the JMJD2 family of histone demethylases.
- Author
-
Whetstine JR, Nottke A, Lan F, Huarte M, Smolikov S, Chen Z, Spooner E, Li E, Zhang G, Colaiacovo M, and Shi Y
- Subjects
- Animals, Caenorhabditis elegans cytology, Caenorhabditis elegans genetics, Caenorhabditis elegans Proteins genetics, Caenorhabditis elegans Proteins metabolism, Catalytic Domain, Cell Differentiation physiology, Chromosomes genetics, Chromosomes metabolism, DNA-Binding Proteins genetics, Down-Regulation physiology, Germ Cells cytology, Germ Cells metabolism, HeLa Cells, Histones chemistry, Humans, Jumonji Domain-Containing Histone Demethylases, Meiosis physiology, Mutation, Oxidoreductases, N-Demethylating, RNA Interference, Rad51 Recombinase genetics, Rad51 Recombinase metabolism, Transcription Factors genetics, Tumor Suppressor Protein p53 metabolism, Caenorhabditis elegans metabolism, DNA Methylation, DNA-Binding Proteins metabolism, Histones metabolism, Lysine metabolism, Transcription Factors metabolism
- Abstract
Histone methylation regulates chromatin structure, transcription, and epigenetic state of the cell. Histone methylation is dynamically regulated by histone methylases and demethylases such as LSD1 and JHDM1, which mediate demethylation of di- and monomethylated histones. It has been unclear whether demethylases exist that reverse lysine trimethylation. We show the JmjC domain-containing protein JMJD2A reversed trimethylated H3-K9/K36 to di- but not mono- or unmethylated products. Overexpression of JMJD2A but not a catalytically inactive mutant reduced H3-K9/K36 trimethylation levels in cultured cells. In contrast, RNAi depletion of the C. elegans JMJD2A homolog resulted in an increase in general H3-K9Me3 and localized H3-K36Me3 levels on meiotic chromosomes and triggered p53-dependent germline apoptosis. Additionally, other human JMJD2 subfamily members also functioned as trimethylation-specific demethylases, converting H3-K9Me3 to H3-K9Me2 and H3-K9Me1, respectively. Our finding that this family of demethylases generates different methylated states at the same lysine residue provides a mechanism for fine-tuning histone methylation.
- Published
- 2006
- Full Text
- View/download PDF
46. Regulation of tissue-specific and extracellular matrix-related genes by a class I histone deacetylase.
- Author
-
Whetstine JR, Ceron J, Ladd B, Dufourcq P, Reinke V, and Shi Y
- Subjects
- Animals, Caenorhabditis elegans embryology, Caenorhabditis elegans enzymology, Caenorhabditis elegans genetics, Cell Movement physiology, Extracellular Matrix genetics, Histone Deacetylases genetics, Oligonucleotide Array Sequence Analysis, Organ Specificity, RNA Interference, Reverse Transcriptase Polymerase Chain Reaction, Extracellular Matrix metabolism, Gene Expression Regulation, Developmental physiology, Histone Deacetylases physiology
- Abstract
Class I histone deacetylases (HDACs) repress transcription by deacetylating histones and have been shown to play crucial roles in mouse, Xenopus, zebrafish, and C. elegans development. To identify the molecular networks regulated by a class I HDAC in a multicellular organism, we carried out a global gene expression profiling study using C. elegans embryos, and identified tissue-specific and extracellular matrix (ECM)-related genes as major HDA-1 targets. Ectopic expression of HDA-1 or C. elegans cystatin, an HDA-1 target identified from the microarray, significantly perturbed mammalian cell invasion. Similarly, RNAi depletion or overexpression of human HDAC-1 also affected cell migration. These findings suggest that HDA-1/HDAC-1 may play a critical, evolutionarily conserved role in regulating the extracellular microenvironment. Because human HDACs are targets for cancer therapy, these findings have significant implications in cancer treatment.
- Published
- 2005
- Full Text
- View/download PDF
47. Transcriptional regulation of the human reduced folate carrier promoter C: synergistic transactivation by Sp1 and C/EBP beta and identification of a downstream repressor.
- Author
-
Payton SG, Whetstine JR, Ge Y, and Matherly LH
- Subjects
- Animals, Base Sequence, CCAAT-Binding Factor, Cells, Cultured, Chromatin Immunoprecipitation, Drosophila, Humans, Molecular Sequence Data, Promoter Regions, Genetic, Reduced Folate Carrier Protein, Transcription Factor CHOP, Transcription, Genetic, Transcriptional Activation, Transfection, Tumor Cells, Cultured, CCAAT-Enhancer-Binding Proteins physiology, Gene Expression Regulation, Membrane Transport Proteins genetics, Sp1 Transcription Factor physiology, Transcription Factors physiology
- Abstract
The human reduced folate carrier (hRFC) is ubiquitously but differentially expressed in human tissues and its levels are regulated by up to six alternatively spliced non-coding regions (designated A1/A2, A, B, C, D, and E) and by at least four promoters. By transient transfections of HepG2 human hepatoma cells with 5' and 3' deletion constructs spanning 2883 bp of upstream sequence, a transcriptionally important region was localized to within 177 bp flanking the transcriptional start sites for exon C. By gel shift and chromatin immunoprecipitation assays, Sp1 and C/EBP beta transcription factors were found to bind consensus elements (GC-box, CCAAT-box) within this region. The functional importance of these elements was confirmed by transient tranfections of HepG2 cells with hRFC-C reporter constructs in which these elements were mutated, and by co-transfections of Drosophila SL-2 cells with wild-type hRFC-C promoter and expression constructs for Sp1 and C/EBP beta. Whereas both Sp1 and C/EBP beta transactivated hRFC-C promoter activity, C/EBP alpha and gamma were transcriptionally inert. Sp1 combined with C/EBP beta resulted in a synergistic transactivation. In HepG2 cells, transfections with Sp1 and C/EBP beta both increased endogenous levels of hRFC-C transcripts. By 3' deletion analysis, a repressor sequence was localized to within 71 bp flanking the minimal promoter. On gel shifts, a novel transcriptional repressor was localized to within 30 bp. Collectively, these results identify transcriptionally important regions in the hRFC-C minimal promoter that include a GC-box and CCAAT-box, and suggest that cooperative interactions between Sp1 and C/EBP beta are essential for hRFC-C transactivation. Another possible factor in the tissue-specific regulation of the hRFC-C region involves the downstream repressor flanking the minimal promoter.
- Published
- 2005
- Full Text
- View/download PDF
48. Histone demethylation mediated by the nuclear amine oxidase homolog LSD1.
- Author
-
Shi Y, Lan F, Matson C, Mulligan P, Whetstine JR, Cole PA, Casero RA, and Shi Y
- Subjects
- Conserved Sequence genetics, Formaldehyde metabolism, Gene Expression Regulation, HeLa Cells, Histone Demethylases, Humans, Mass Spectrometry, Oxidoreductases, N-Demethylating chemistry, Oxidoreductases, N-Demethylating genetics, Oxidoreductases, N-Demethylating isolation & purification, RNA Interference, Recombinant Proteins, Repressor Proteins chemistry, Repressor Proteins genetics, Repressor Proteins metabolism, Schizosaccharomyces pombe Proteins chemistry, Schizosaccharomyces pombe Proteins genetics, Schizosaccharomyces pombe Proteins isolation & purification, Substrate Specificity, Transcription, Genetic, Histones metabolism, Lysine metabolism, Methylation, Nuclear Proteins metabolism, Oxidoreductases, N-Demethylating metabolism, Schizosaccharomyces pombe Proteins metabolism
- Abstract
Posttranslational modifications of histone N-terminal tails impact chromatin structure and gene transcription. While the extent of histone acetylation is determined by both acetyltransferases and deacetylases, it has been unclear whether histone methylation is also regulated by enzymes with opposing activities. Here, we provide evidence that LSD1 (KIAA0601), a nuclear homolog of amine oxidases, functions as a histone demethylase and transcriptional corepressor. LSD1 specifically demethylates histone H3 lysine 4, which is linked to active transcription. Lysine demethylation occurs via an oxidation reaction that generates formaldehyde. Importantly, RNAi inhibition of LSD1 causes an increase in H3 lysine 4 methylation and concomitant derepression of target genes, suggesting that LSD1 represses transcription via histone demethylation. The results thus identify a histone demethylase conserved from S. pombe to human and reveal dynamic regulation of histone methylation by both histone methylases and demethylases.
- Published
- 2004
- Full Text
- View/download PDF
49. Roles of USF, Ikaros and Sp proteins in the transcriptional regulation of the human reduced folate carrier B promoter.
- Author
-
Liu M, Whetstine JR, Payton SG, Ge Y, Flatley RM, and Matherly LH
- Subjects
- Acetylation, Animals, Base Sequence, Cell Line, Cell Line, Tumor, Chromatin Immunoprecipitation, DNA-Binding Proteins genetics, Drosophila melanogaster, Electrophoretic Mobility Shift Assay, Genes, Reporter genetics, Histones metabolism, Humans, Ikaros Transcription Factor, Molecular Sequence Data, RNA, Messenger genetics, RNA, Messenger metabolism, Reduced Folate Carrier Protein, Sequence Deletion genetics, Sp1 Transcription Factor genetics, Transcription Factors genetics, Transcriptional Activation genetics, Transfection, Upstream Stimulatory Factors, DNA-Binding Proteins metabolism, Gene Expression Regulation, Membrane Transport Proteins genetics, Promoter Regions, Genetic genetics, Response Elements genetics, Sp1 Transcription Factor metabolism, Transcription Factors metabolism, Transcription, Genetic genetics
- Abstract
The hRFC (human reduced folate carrier) is ubiquitously but differentially expressed in human tissues and its levels are regulated by up to seven non-coding regions (A1, A2, A, B, C, D and E) and at least four promoters. For the hRFC-B basal promoter, regulation involves binding of Sp (specificity protein) transcription factors to a critical GC-box. By transiently transfecting HT1080 cells with 5'- and 3'-deletion constructs spanning 1057 bp of upstream sequence, a transcriptionally important region was localized to 158 bp flanking the transcriptional start sites. By gel shift and chromatin immunoprecipitation assays, USF (upstream stimulatory factor), Sp1 and Ikaros-related proteins were bound to consensus elements (one E-box, two GC-box and three Ikaros) within this region. The functional importance of these elements was confirmed by transient tranfections of HT1080 cells with hRFC-B reporter constructs in which they were mutated, and by co-transfections of Drosophila Mel-2 cells with wild-type hRFC-B promoter and expression constructs for USF1, USF2a, Sp1 and Ikaros 2 and 8. Both USF1 and Sp1 proteins transactivated the hRFC-B promoter. Sp1 combined with USF1 resulted in a synergistic transactivation. Identical results were obtained with USF2a. Ikaros 2 was a repressor of hRFC-B promoter activity whose effects were partly reversed by the dominant-negative Ikaros 8. In HT1080 cells, transfection with Ikaros 2 decreased endogenous hRFC-B transcripts, whereas USF1 and Sp1 increased transcript levels. Ikaros 2 also decreased reporter gene activity and levels of acetylated chromatin associated with the endogenous promoter. Collectively, these results identify transcriptionally important regions in the hRFC-B promoter that include multiple GC-box, Ikaros and E-box elements. Our results also suggest that co-operative interactions between transcription factors Sp1 and USF are essential for high-level hRFC-B transactivation and imply that these effects are modulated by the family of Ikaros proteins and by histone acetylation.
- Published
- 2004
- Full Text
- View/download PDF
50. Coordinated histone modifications mediated by a CtBP co-repressor complex.
- Author
-
Shi Y, Sawada J, Sui G, Affar el B, Whetstine JR, Lan F, Ogawa H, Luke MP, Nakatani Y, and Shi Y
- Subjects
- Alcohol Oxidoreductases, Cadherins genetics, Chromatin genetics, Chromatin metabolism, DNA-Binding Proteins genetics, Histone Deacetylases metabolism, Histone Methyltransferases, Humans, Macromolecular Substances, Methyltransferases metabolism, Phosphoproteins genetics, Promoter Regions, Genetic genetics, Protein Methyltransferases, RNA Interference, Repressor Proteins genetics, DNA-Binding Proteins metabolism, Gene Expression Regulation, Histone-Lysine N-Methyltransferase, Histones metabolism, Phosphoproteins metabolism, Repressor Proteins metabolism
- Abstract
The transcriptional co-repressor CtBP (C-terminal binding protein) is implicated in tumorigenesis because it is targeted by the adenovirus E1A protein during oncogenic transformation. Genetic studies have also identified a crucial function for CtBP in animal development. CtBP is recruited to DNA by transcription factors that contain a PXDLS motif, but the detailed molecular events after the recruitment of CtBP to DNA and the mechanism of CtBP function in tumorigenesis are largely unknown. Here we report the identification of a CtBP complex that contains the essential components for both gene targeting and coordinated histone modifications, allowing for the effective repression of genes targeted by CtBP. Inhibiting the expression of CtBP and its associated histone-modifying activities by RNA-mediated interference resulted in alterations of histone modifications at the promoter of the tumour invasion suppressor gene E-cadherin and increased promoter activity in a reporter assay. These findings identify a molecular mechanism by which CtBP mediates transcriptional repression and provide insight into CtBP participation in oncogenesis.
- Published
- 2003
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.