Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
Add more filters










Publication year range
1.
Genes Dev ; 2024 Jun 24.
Article in English | MEDLINE | ID: mdl-38914477

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.

2.
Cell ; 186(21): 4528-4545.e18, 2023 10 12.
Article in English | MEDLINE | ID: mdl-37788669

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.


Subject(s)
Epigenesis, Genetic , Myeloid-Lymphoid Leukemia Protein , 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 , Myeloid-Lymphoid Leukemia Protein/genetics , Translocation, Genetic
3.
Gastroenterology ; 164(6): 921-936.e1, 2023 05.
Article in English | MEDLINE | ID: mdl-36764492

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 ApcMin 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.


Subject(s)
Adenocarcinoma , Adenoma , Colonic Neoplasms , Colorectal Neoplasms , Animals , Humans , Mice , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Adenoma/genetics , Adenoma/pathology , Carcinogenesis/genetics , Cell Transformation, Neoplastic/genetics , Colonic Neoplasms/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , CpG Islands/genetics , DNA Methylation , DNA-Binding Proteins/genetics , Epigenesis, Genetic , Mixed Function Oxygenases/genetics , Phenotype , Proto-Oncogene Proteins/genetics
4.
STAR Protoc ; 3(4): 101827, 2022 12 16.
Article in English | MEDLINE | ID: mdl-36386876

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).


Subject(s)
DNA Replication Timing , Epigenomics , Transcriptome , Workflow , Chromatin/genetics
5.
Nat Genet ; 54(3): 318-327, 2022 03.
Article in English | MEDLINE | ID: mdl-35256805

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.


Subject(s)
Embryo, Mammalian , Pluripotent Stem Cells , Animals , Cell Differentiation/genetics , Cellular Reprogramming/genetics , DNA Replication/genetics , Embryonic Development/genetics , Mice
6.
STAR Protoc ; 3(1): 101209, 2022 03 18.
Article in English | MEDLINE | ID: mdl-35243385

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).


Subject(s)
DNA Replication Timing , DNA Replication , Bromodeoxyuridine , DNA Replication/genetics , S Phase/genetics , Sequence Analysis, DNA
7.
STAR Protoc ; 3(2): 101243, 2022 06 17.
Article in English | MEDLINE | ID: mdl-35310076

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).


Subject(s)
Chromatin Immunoprecipitation Sequencing , Chromatin , Cell Cycle/genetics , Chromatin/genetics , Chromatin Immunoprecipitation/methods , Histone Code
8.
Cell Rep ; 37(1): 109799, 2021 10 05.
Article in English | MEDLINE | ID: mdl-34610305

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.


Subject(s)
Chromatin/chemistry , DNA Replication Timing/physiology , 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
9.
Sci Rep ; 11(1): 14481, 2021 07 14.
Article in English | MEDLINE | ID: mdl-34262087

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.


Subject(s)
Apoptosis/drug effects , Caenorhabditis elegans/drug effects , Isoflurane/toxicity , Oogenesis/drug effects , 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
10.
Biochim Biophys Acta Gene Regul Mech ; 1863(10): 194624, 2020 10.
Article in English | MEDLINE | ID: mdl-32798738

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.


Subject(s)
DNA Replication , Gene Expression Regulation , Histones/genetics , Jumonji Domain-Containing Histone Demethylases/genetics , Catalysis , Epigenesis, Genetic , Gene Expression Profiling , Histones/metabolism , Humans , Transcription, Genetic
11.
Cancer Discov ; 10(2): 306-325, 2020 02.
Article in English | MEDLINE | ID: mdl-31776131

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.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , DNA Methylation/genetics , Histones/metabolism , Neoplasms/genetics , 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
12.
Cancer Cell ; 36(4): 343-345, 2019 10 14.
Article in English | MEDLINE | ID: mdl-31614112

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.


Subject(s)
Antigen Presentation , Neoplasms , CD8-Positive T-Lymphocytes/immunology , Histocompatibility Antigens Class I , Humans , Immune Evasion , T-Lymphocytes, Cytotoxic/immunology
13.
Mol Cell ; 75(4): 683-699.e7, 2019 08 22.
Article in English | MEDLINE | ID: mdl-31399344

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.


Subject(s)
Promoter Regions, Genetic , RNA Polymerase II/metabolism , Sirtuins/metabolism , Transcription Elongation, Genetic , 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
15.
Cell ; 174(4): 803-817.e16, 2018 08 09.
Article in English | MEDLINE | ID: mdl-30057114

ABSTRACT

Acquired chromosomal DNA amplifications are features of many tumors. Although overexpression and stabilization of the histone H3 lysine 9/36 (H3K9/36) tri-demethylase KDM4A generates transient site-specific copy number gains (TSSGs), additional mechanisms directly controlling site-specific DNA copy gains are not well defined. In this study, we uncover a collection of H3K4-modifying chromatin regulators that function with H3K9 and H3K36 regulators to orchestrate TSSGs. Specifically, the H3K4 tri-demethylase KDM5A and specific COMPASS/KMT2 H3K4 methyltransferases modulate different TSSG loci through H3K4 methylation states and KDM4A recruitment. Furthermore, a distinct chromatin modifier network, MLL1-KDM4B-KDM5B, controls copy number regulation at a specific genomic locus in a KDM4A-independent manner. These pathways comprise an epigenetic addressing system for defining site-specific DNA rereplication and amplifications.


Subject(s)
Chromatin/metabolism , DNA Copy Number Variations , DNA Methylation , Histones/metabolism , Lysine/metabolism , Retinoblastoma-Binding Protein 2/metabolism , Cell Cycle , HEK293 Cells , Humans , Retinoblastoma-Binding Protein 2/genetics
16.
Dev Cell ; 43(6): 689-703.e5, 2017 12 18.
Article in English | MEDLINE | ID: mdl-29233476

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.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/biosynthesis , Drosophila Proteins/metabolism , E2F Transcription Factors/genetics , E2F Transcription Factors/metabolism , Fat Body/physiology , Trans-Activators/metabolism , 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
17.
Cancer Res ; 77(8): 2018-2028, 2017 04 15.
Article in English | MEDLINE | ID: mdl-28202526

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.


Subject(s)
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 , 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
18.
Mol Cell Biol ; 36(7): 1050-63, 2016 Jan 11.
Article in English | MEDLINE | ID: mdl-26755558

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.


Subject(s)
Chromosome Aberrations , Gene Dosage , Adaptation, Biological , Animals , Drug Resistance/genetics , Genetic Heterogeneity , Genomic Structural Variation , Humans , Neoplasms/genetics
19.
J Biol Chem ; 291(10): 4862-71, 2016 Mar 04.
Article in English | MEDLINE | ID: mdl-26755726

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.


Subject(s)
Breast Neoplasms/genetics , Gene Amplification , Gene Expression Regulation, Neoplastic , Jumonji Domain-Containing Histone Demethylases/genetics , MicroRNAs/genetics , CDC2-CDC28 Kinases/genetics , CDC2-CDC28 Kinases/metabolism , Cell Line, Tumor , Female , Humans
20.
Cancer Discov ; 5(12): 1241-3, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26637658

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.


Subject(s)
Polycomb Repressive Complex 1 , Ubiquitin-Protein Ligases , Humans , Melanoma
SELECTION OF CITATIONS
SEARCH DETAIL
...