Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Cancer Discov ; 12(1): 186-203, 2022 01.
Article in English | MEDLINE | ID: mdl-34417224

ABSTRACT

Mutations in epigenetic regulators are common in relapsed pediatric acute lymphoblastic leukemia (ALL). Here, we uncovered the mechanism underlying the relapse of ALL driven by an activating mutation of the NSD2 histone methyltransferase (p.E1099K). Using high-throughput drug screening, we found that NSD2-mutant cells were specifically resistant to glucocorticoids. Correction of this mutation restored glucocorticoid sensitivity. The transcriptional response to glucocorticoids was blocked in NSD2-mutant cells due to depressed glucocorticoid receptor (GR) levels and the failure of glucocorticoids to autoactivate GR expression. Although H3K27me3 was globally decreased by NSD2 p.E1099K, H3K27me3 accumulated at the NR3C1 (GR) promoter. Pretreatment of NSD2 p.E1099K cell lines and patient-derived xenograft samples with PRC2 inhibitors reversed glucocorticoid resistance in vitro and in vivo. PRC2 inhibitors restored NR3C1 autoactivation by glucocorticoids, increasing GR levels and allowing GR binding and activation of proapoptotic genes. These findings suggest a new therapeutic approach to relapsed ALL associated with NSD2 mutation. SIGNIFICANCE: NSD2 histone methyltransferase mutations observed in relapsed pediatric ALL drove glucocorticoid resistance by repression of the GR and abrogation of GR gene autoactivation due to accumulation of K3K27me3 at its promoter. Pretreatment with PRC2 inhibitors reversed resistance, suggesting a new therapeutic approach to these patients with ALL.This article is highlighted in the In This Issue feature, p. 1.


Subject(s)
Enzyme Inhibitors/therapeutic use , Glucocorticoids/therapeutic use , Histone Methyltransferases/antagonists & inhibitors , Histone-Lysine N-Methyltransferase/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Repressor Proteins/genetics , Cell Line, Tumor/drug effects , Cell Survival , Child , Drug Resistance, Neoplasm , Enzyme Inhibitors/pharmacology , Female , Glucocorticoids/pharmacology , Humans , Male , Mutation , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology
2.
Cancer Discov ; 9(10): 1438-1451, 2019 10.
Article in English | MEDLINE | ID: mdl-31337617

ABSTRACT

By examination of the cancer genomics database, we identified a new set of mutations in core histones that frequently recur in cancer patient samples and are predicted to disrupt nucleosome stability. In support of this idea, we characterized a glutamate to lysine mutation of histone H2B at amino acid 76 (H2B-E76K), found particularly in bladder and head and neck cancers, that disrupts the interaction between H2B and H4. Although H2B-E76K forms dimers with H2A, it does not form stable histone octamers with H3 and H4 in vitro, and when reconstituted with DNA forms unstable nucleosomes with increased sensitivity to nuclease. Expression of the equivalent H2B mutant in yeast restricted growth at high temperature and led to defective nucleosome-mediated gene repression. Significantly, H2B-E76K expression in the normal mammary epithelial cell line MCF10A increased cellular proliferation, cooperated with mutant PIK3CA to promote colony formation, and caused a significant drift in gene expression and fundamental changes in chromatin accessibility, particularly at gene regulatory elements. Taken together, these data demonstrate that mutations in the globular domains of core histones may give rise to an oncogenic program due to nucleosome dysfunction and deregulation of gene expression. SIGNIFICANCE: Mutations in the core histones frequently occur in cancer and represent a new mechanism of epigenetic dysfunction that involves destabilization of the nucleosome, deregulation of chromatin accessibility, and alteration of gene expression to drive cellular transformation.See related commentary by Sarthy and Henikoff, p. 1346.This article is highlighted in the In This Issue feature, p. 1325.


Subject(s)
Histones/genetics , Mutation , Neoplasms/genetics , Oncogenes , Alleles , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Gene Expression , Gene Expression Profiling , Histones/chemistry , Histones/metabolism , Humans , Mutation, Missense , Neoplasms/metabolism , Nucleosomes/metabolism , Protein Multimerization , Yeasts/genetics , Yeasts/metabolism
3.
Oncogene ; 38(5): 671-686, 2019 01.
Article in English | MEDLINE | ID: mdl-30171259

ABSTRACT

NSD2, a histone methyltransferase specific for methylation of histone 3 lysine 36 (H3K36), exhibits a glutamic acid to lysine mutation at residue 1099 (E1099K) in childhood acute lymphocytic leukemia (ALL), and cells harboring this mutation can become the predominant clone in relapsing disease. We studied the effects of this mutant enzyme in silico, in vitro, and in vivo using gene edited cell lines. The E1099K mutation altered enzyme/substrate binding and enhanced the rate of H3K36 methylation. As a result, cell lines harboring E1099K exhibit increased H3K36 dimethylation and reduced H3K27 trimethylation, particularly on nucleosomes containing histone H3.1. Mutant NSD2 cells exhibit reduced apoptosis and enhanced proliferation, clonogenicity, adhesion, and migration. In mouse xenografts, mutant NSD2 cells are more lethal and brain invasive than wildtype cells. Transcriptional profiling demonstrates that mutant NSD2 aberrantly activates factors commonly associated with neural and stromal lineages in addition to signaling and adhesion genes. Identification of these pathways provides new avenues for therapeutic interventions in NSD2 dysregulated malignancies.


Subject(s)
Cellular Reprogramming , Histone-Lysine N-Methyltransferase , Mutation, Missense , Neoplasm Proteins , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Repressor Proteins , Amino Acid Substitution , HeLa Cells , Heterografts , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Humans , Neoplasm Invasiveness , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neoplasm Transplantation , Precursor Cell Lymphoblastic Leukemia-Lymphoma/enzymology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Repressor Proteins/genetics , Repressor Proteins/metabolism
4.
Cell Rep ; 21(3): 628-640, 2017 Oct 17.
Article in English | MEDLINE | ID: mdl-29045832

ABSTRACT

Loss or inactivation of the histone H3K27 demethylase UTX occurs in several malignancies, including multiple myeloma (MM). Using an isogenic cell system, we found that loss of UTX leads to deactivation of gene expression ultimately promoting the proliferation, clonogenicity, adhesion, and tumorigenicity of MM cells. Moreover, UTX mutant cells showed increased in vitro and in vivo sensitivity to inhibition of EZH2, a histone methyltransferase that generates H3K27me3. Such sensitivity was related to a decrease in the levels of IRF4 and c-MYC and an activation of repressors of IRF4 characteristic of germinal center B cells such as BCL6 and IRF1. Rebalance of H3K27me3 levels at specific genes through EZH2 inhibitors may be a therapeutic strategy in MM cases harboring UTX mutations.


Subject(s)
Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Histone Demethylases/deficiency , Multiple Myeloma/pathology , Nuclear Proteins/deficiency , Animals , Carcinogenesis/drug effects , Carcinogenesis/genetics , Carcinogenesis/pathology , Cell Adhesion/drug effects , Cell Adhesion/genetics , Cell Dedifferentiation/drug effects , Cell Dedifferentiation/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cell Survival/drug effects , Cell Survival/genetics , Clone Cells , Enhancer of Zeste Homolog 2 Protein/metabolism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Histone Demethylases/metabolism , Histones/metabolism , Indazoles/pharmacology , Interferon Regulatory Factors/metabolism , Lysine/metabolism , Methylation , Mice , Mice, Inbred NOD , Mice, SCID , Multiple Myeloma/genetics , Mutation/genetics , Nuclear Proteins/metabolism , Phenotype , Pyridones/pharmacology , Transcription, Genetic/drug effects
5.
Cell Rep ; 12(8): 1300-13, 2015 Aug 25.
Article in English | MEDLINE | ID: mdl-26279576

ABSTRACT

Unrestrained receptor tyrosine kinase (RTK) signaling and epigenetic deregulation are root causes of tumorigenesis. We establish linkage between these processes by demonstrating that aberrant RTK signaling unleashed by oncogenic HRas(G12V) or loss of negative feedback through Sprouty gene deletion remodels histone modifications associated with active typical and super-enhancers. However, although both lesions disrupt the Ras-Erk axis, the expression programs, enhancer signatures, and transcription factor networks modulated upon HRas(G12V) transformation or Sprouty deletion are largely distinct. Oncogenic HRas(G12V) elevates histone 3 lysine 27 acetylation (H3K27ac) levels at enhancers near the transcription factor Gata4 and the kinase Prkcb, as well as their expression levels. We show that Gata4 is necessary for the aberrant gene expression and H3K27ac marking at enhancers, and Prkcb is required for the oncogenic effects of HRas(G12V)-driven cells. Taken together, our findings demonstrate that dynamic reprogramming of the cellular enhancer landscape is a major effect of oncogenic RTK signaling.


Subject(s)
Carcinogenesis/genetics , Enhancer Elements, Genetic , Gene Expression Regulation, Neoplastic , MAP Kinase Signaling System , Proto-Oncogene Proteins p21(ras)/metabolism , Acetylation , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Carcinogenesis/metabolism , Cell Line , Extracellular Signal-Regulated MAP Kinases/metabolism , GATA4 Transcription Factor/genetics , GATA4 Transcription Factor/metabolism , Histones/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Phosphoproteins/genetics , Phosphoproteins/metabolism , Protein Kinase C beta/genetics , Protein Kinase C beta/metabolism , Protein Processing, Post-Translational , Proto-Oncogene Proteins p21(ras)/genetics , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism
6.
PLoS Genet ; 10(9): e1004566, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25188243

ABSTRACT

Overexpression of the histone methyltransferase MMSET in t(4;14)+ multiple myeloma patients is believed to be the driving factor in the pathogenesis of this subtype of myeloma. MMSET catalyzes dimethylation of lysine 36 on histone H3 (H3K36me2), and its overexpression causes a global increase in H3K36me2, redistributing this mark in a broad, elevated level across the genome. Here, we demonstrate that an increased level of MMSET also induces a global reduction of lysine 27 trimethylation on histone H3 (H3K27me3). Despite the net decrease in H3K27 methylation, specific genomic loci exhibit enhanced recruitment of the EZH2 histone methyltransferase and become hypermethylated on this residue. These effects likely contribute to the myeloma phenotype since MMSET-overexpressing cells displayed increased sensitivity to EZH2 inhibition. Furthermore, we demonstrate that such MMSET-mediated epigenetic changes require a number of functional domains within the protein, including PHD domains that mediate MMSET recruitment to chromatin. In vivo, targeting of MMSET by an inducible shRNA reversed histone methylation changes and led to regression of established tumors in athymic mice. Together, our work elucidates previously unrecognized interplay between MMSET and EZH2 in myeloma oncogenesis and identifies domains to be considered when designing inhibitors of MMSET function.


Subject(s)
DNA Methylation/genetics , Epigenesis, Genetic/genetics , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Multiple Myeloma/genetics , Polycomb Repressive Complex 2/metabolism , Protein Binding/genetics , Animals , Cell Line , Cell Transformation, Neoplastic/genetics , Chromatin/genetics , Female , HEK293 Cells , Histone-Lysine N-Methyltransferase/genetics , Histones/genetics , Humans , Lysine/genetics , Mice , Mice, Inbred C57BL , Multiple Myeloma/metabolism , Polycomb Repressive Complex 2/genetics , RNA, Small Interfering/genetics
SELECTION OF CITATIONS
SEARCH DETAIL