Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Clin Cancer Res ; 27(11): 3178-3189, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33731366

ABSTRACT

PURPOSE: Multiple myeloma is a malignancy of plasma cells. Extensive genetic and transcriptional characterization of myeloma has identified subtypes with prognostic and therapeutic implications. In contrast, relatively little is known about the myeloma epigenome. EXPERIMENTAL DESIGN: CD138+CD38+ myeloma cells were isolated from fresh bone marrow aspirate or the same aspirate after freezing for 1-6 months. Gene expression and chromatin accessibility were compared between fresh and frozen samples by RNA sequencing (RNA-seq) and assay for transpose accessible chromatin sequencing (ATAC-seq). Chromatin accessible regions were used to identify regulatory RNA expression in more than 700 samples from newly diagnosed patients in the Multiple Myeloma Research Foundation CoMMpass trial (NCT01454297). RESULTS: Gene expression and chromatin accessibility of cryopreserved myeloma recapitulated that of freshly isolated samples. ATAC-seq performed on a series of biobanked specimens identified thousands of chromatin accessible regions with hundreds being highly coordinated with gene expression. More than 4,700 of these chromatin accessible regions were transcribed in newly diagnosed myelomas from the CoMMpass trial. Regulatory element activity alone recapitulated myeloma gene expression subtypes, and in particular myeloma subtypes with immunoglobulin heavy chain translocations were defined by transcription of distal regulatory elements. Moreover, enhancer activity predicted oncogene expression implicating gene regulatory mechanisms in aggressive myeloma. CONCLUSIONS: These data demonstrate the feasibility of using biobanked specimens for retrospective studies of the myeloma epigenome and illustrate the unique enhancer landscapes of myeloma subtypes that are coupled to gene expression and disease progression.


Subject(s)
Chromatin/genetics , Gene Expression Regulation, Neoplastic/genetics , Gene Expression , Multiple Myeloma/genetics , RNA/genetics , Disease Progression , Epigenome , Feasibility Studies , Humans , Prognosis , Sequence Analysis, RNA
2.
Am J Pathol ; 185(8): 2206-18, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26216285

ABSTRACT

The intestinal epithelium is a dynamic barrier that maintains the distinct environments of intestinal tissue and lumen. Epithelial barrier function is defined principally by tight junctions, which, in turn, depend on the regulated expression of claudin family proteins. Claudins are expressed differentially during intestinal epithelial cell (IEC) differentiation. However, regulatory mechanisms governing claudin expression during epithelial differentiation are incompletely understood. We investigated the molecular mechanisms regulating claudin-7 during IEC differentiation. Claudin-7 expression is increased as epithelial cells differentiate along the intestinal crypt-luminal axis. By using model IECs we observed increased claudin-7 mRNA and nascent heteronuclear RNA levels during differentiation. A screen for potential regulators of the CLDN7 gene during IEC differentiation was performed using a transcription factor/DNA binding array, CLDN7 luciferase reporters, and in silico promoter analysis. We identified hepatocyte nuclear factor 4α as a regulatory factor that bound endogenous CLDN7 promoter in differentiating IECs and stimulated CLDN7 promoter activity. These findings support a role of hepatocyte nuclear factor 4α in controlling claudin-7 expression during IEC differentiation.


Subject(s)
Cell Differentiation/genetics , Claudins/metabolism , Epithelial Cells/metabolism , Hepatocyte Nuclear Factor 4/metabolism , Intestinal Mucosa/metabolism , Caco-2 Cells , Claudins/genetics , Epithelial Cells/cytology , Gene Expression Regulation , HT29 Cells , Hepatocyte Nuclear Factor 4/genetics , Humans , Intestinal Mucosa/cytology , Promoter Regions, Genetic
3.
PLoS One ; 8(9): e75069, 2013.
Article in English | MEDLINE | ID: mdl-24069380

ABSTRACT

Though clinicians can predict which patients are at risk for developing metastases, traditional therapies often prove ineffective and metastatic disease is the primary cause of cancer patient death; therefore, there is a need to develop anti-metastatic therapies that can be administered over long durations to specifically inhibit the motility of cancer cells. Withaniasomnifera root extracts (WRE) have anti-proliferative activity and the active component, Withaferin A, inhibits the pro-metastatic protein, vimentin. Vimentin is an intermediate filament protein and is part of the epithelial to mesenchymal transition (EMT) program to promote metastasis. Here, we determined whether WRE standardized to Withaferin A (sWRE) possesses anti-metastatic activity and whether it inhibits cancer motility via inhibition of vimentin and the EMT program. Several formulations of sWRE were created to enrich for Withaferin A and a stock solution of sWRE in EtOH could recover over 90% of the Withaferin A found in the original extract powder. This sWRE formulation inhibited breast cancer cell motility and invasion at concentrations less than 1µM while having negligible cytotoxicity at this dose. sWRE treatment disrupted vimentin morphology in cell lines, confirming its vimentin inhibitory activity. To determine if sWRE inhibited EMT, TGF-ß was used to induce EMT in MCF10A human mammary epithelial cells. In this case, sWRE prevented EMT induction and inhibited 3-D spheroid invasion. These studies were taken into a human xenograft and mouse mammary carcinoma model. In both models, sWRE and Withaferin A showed dose-dependent inhibition of tumor growth and metastatic lung nodule formation with minimal systemic toxicity. Taken together, these data support the hypothesis that low concentrations of sWRE inhibit cancer metastasis potentially through EMT inhibition. Moreover, these doses of sWRE have nearly no toxicity in normal mouse organs, suggesting the potential for clinical use of orally administered WRE capsules.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/pathology , Epithelial-Mesenchymal Transition/drug effects , Plant Extracts/pharmacology , Plant Roots/chemistry , Withania/chemistry , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/toxicity , Breast Neoplasms/drug therapy , Cell Line, Tumor , Cell Movement/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Humans , Inhibitory Concentration 50 , Mice , Neoplasm Metastasis , Plant Extracts/chemistry , Plant Extracts/toxicity , Solubility , Transforming Growth Factor beta/pharmacology , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
4.
Genome Res ; 22(12): 2339-55, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22899282

ABSTRACT

Monoallelic point mutations of the NADP(+)-dependent isocitrate dehydrogenases IDH1 and IDH2 occur frequently in gliomas, acute myeloid leukemias, and chondromas, and display robust association with specific DNA hypermethylation signatures. Here we show that heterozygous expression of the IDH1(R132H) allele is sufficient to induce the genome-wide alterations in DNA methylation characteristic of these tumors. Using a gene-targeting approach, we knocked-in a single copy of the most frequently observed IDH1 mutation, R132H, into a human cancer cell line and profiled changes in DNA methylation at over 27,000 CpG dinucleotides relative to wild-type parental cells. We find that IDH1(R132H/WT) mutation induces widespread alterations in DNA methylation, including hypermethylation of 2010 and hypomethylation of 842 CpG loci. We demonstrate that many of these alterations are consistent with those observed in IDH1-mutant and G-CIMP+ primary gliomas and can segregate IDH wild-type and mutated tumors as well as those exhibiting the G-CIMP phenotype in unsupervised analysis of two primary glioma cohorts. Further, we show that the direction of IDH1(R132H/WT)-mediated DNA methylation change is largely dependent upon preexisting DNA methylation levels, resulting in depletion of moderately methylated loci. Additionally, whereas the levels of multiple histone H3 and H4 methylation modifications were globally increased, consistent with broad inhibition of histone demethylation, hypermethylation at H3K9 in particular accompanied locus-specific DNA hypermethylation at several genes down-regulated in IDH1(R132H/WT) knock-in cells. These data provide insight on epigenetic alterations induced by IDH1 mutations and support a causal role for IDH1(R132H/WT) mutants in driving epigenetic instability in human cancer cells.


Subject(s)
DNA Methylation , Heterozygote , Isocitrate Dehydrogenase/genetics , Mutation , Alleles , Blotting, Western , Chromatin Immunoprecipitation/methods , Down-Regulation , Epigenomics , Gene Expression Profiling , Gene Expression Regulation , Gene Silencing , Genetic Loci , HCT116 Cells , Histones/genetics , Histones/metabolism , Humans , Isocitrate Dehydrogenase/metabolism , Leukemia, Myeloid, Acute/genetics , Phenotype , Real-Time Polymerase Chain Reaction , Sequence Analysis, DNA
5.
Mol Cancer Res ; 8(7): 1048-59, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20587535

ABSTRACT

DNA methyltransferase inhibitors are currently the standard of care for myelodysplastic syndrome and are in clinical trials for leukemias and solid tumors. However, the molecular basis underlying their activity remains poorly understood. Here, we studied the induction and long-term stability of gene reactivation at three methylated tumor suppressor loci in response to the DNA methyltransferase inhibitor 5-aza-2'-deoxycytidine (5-azaCdR) in human breast cancer cells. At the TMS1/ASC locus, treatment with 5-azaCdR resulted in partial DNA demethylation, the reengagement of RNA polymerase II (Pol II), and a shift from a repressive chromatin profile marked with H3K9me2 and H4K20me3 to an active profile enriched in H3ac and H3K4me2. Using a single-molecule approach coupling chromatin immunoprecipitation with bisulfite sequencing, we show that H3ac, H3K4me2, and Pol II selectively associated with the demethylated alleles, whereas H3K9me2 preferentially marked alleles resistant to demethylation. H4K20me3 was unaffected by DNA demethylation and associated with both unmethylated and methylated alleles. After drug removal, TMS1 underwent partial remethylation, yet a subset of alleles remained stably demethylated for over 3 months. These alleles remained selectively associated with H3K4me2, H3ac, and Pol II and correlated with a sustained low level of gene expression. TMS1 alleles reacquired H3K9me2 over time, and those alleles that became remethylated retained H3ac. In contrast, CDH1 and ESR1 were remethylated and completely silenced within approximately 1 week of drug removal, and failed to maintain stably unmethylated alleles. Our data suggest that the ability to maintain Pol II occupancy is a critical factor in the long-term stability of drug-induced CpG island demethylation.


Subject(s)
Azacitidine/analogs & derivatives , DNA Methylation/drug effects , RNA Polymerase II/metabolism , Azacitidine/pharmacology , Cell Line, Tumor , CpG Islands , Decitabine , Gene Expression Regulation, Neoplastic , Gene Silencing , Genes, Tumor Suppressor , Humans , Promoter Regions, Genetic , RNA Polymerase II/genetics
6.
Cancer Genet Cytogenet ; 197(2): 130-41, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20193846

ABSTRACT

The STK11/LKB1 gene encodes a ubiquitously expressed serine/threonine kinase that is mutated in multiple sporadic cancers including non-small cell lung carcinomas, pancreatic cancers, and melanomas. LKB1 plays a role in multiple cellular functions including cell growth, cell cycle progression, metabolism, cell polarity, and migration. To date, only a limited number of studies have assessed the status of LKB1 in cervical cancers. Herein, we investigate DNA methylation, DNA mutation, and transcription at the LKB1 locus in cervical cancer cell lines. We identified homozygous deletions of 25-85kb in the HeLa and SiHa cell lines. Deletion breakpoint analysis in HeLa cells revealed that the deletion resulted from an Alu-recombination-mediated deletion (ARMD) and generated a novel LKB1 fusion transcript driven by an uncharacterized CpG island promoter located approximately 11kb upstream of LKB1. Although the homozygous deletion in SiHa cells removes the entire LKB1 gene and portions of the neighboring genes SBNO2 and c19orf26, this deletion also generates a fusion transcript driven by the c19orf26 promoter and composed of both c19orf26 and SBNO2 sequences. Further analyses of public gene expression and mutation databases suggest that LKB1 and its neighboring genes are frequently dysregulated in primary cervical cancers. Thus, homozygous deletions affecting LKB1 in cervical cancers may generate multiple fusion transcripts involving LKB1, SBNO2, and c19orf26.


Subject(s)
Gene Deletion , Protein Serine-Threonine Kinases/genetics , Uterine Cervical Neoplasms/genetics , AMP-Activated Protein Kinase Kinases , Alu Elements/genetics , Base Sequence , Cell Line, Tumor , DNA Methylation , Female , Gene Expression Regulation, Neoplastic , HeLa Cells , Humans , Molecular Sequence Data , Mutation , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Protein Serine-Threonine Kinases/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sequence Alignment , Transcription, Genetic , Uterine Cervical Neoplasms/enzymology
7.
J Biol Chem ; 284(22): 14698-709, 2009 May 29.
Article in English | MEDLINE | ID: mdl-19324871

ABSTRACT

Epigenetic silencing involving the aberrant DNA methylation of promoter-associated CpG islands is one mechanism leading to the inactivation of tumor suppressor genes in human cancers. However, the molecular mechanisms underlying this event remains poorly understood. TMS1/ASC is a novel proapoptotic signaling factor that is subject to epigenetic silencing in human breast and other cancers. The TMS1 promoter is embedded within a CpG island that is unmethylated in normal cells and is spanned by three DNase I-hypersensitive sites (HS). Silencing of TMS1 in cancer cells is accompanied by local alterations in histone modification, remodeling of the HS, and hypermethylation of DNA. In this study, we probed the functional significance of the CpG island-specific HS. We identified a methylation-sensitive complex that bound a 55-bp intronic element corresponding to HS2. Affinity chromatography and mass spectrometry identified a component of this complex to be the GA-binding protein (GABP) alpha. Supershift analysis indicated that the GABPalpha binding partner, GABPbeta1, was also present in the complex. The HS2 element conferred a 3-fold enhancement in TMS1 promoter activity, which was dependent on both intact tandem ets binding sites and the presence of GABPalpha/beta1 in trans. GABPalpha was selectively enriched at HS2 in human cells, and its occupancy was inversely correlated with CpG island methylation. Down-regulation of GABPalpha led to a concomitant decrease in TMS1 expression. These data indicate that the intronic HS2 element acts in cis to maintain transcriptional competency at the TMS1 locus and that this activity is mediated by the ets transcription factor, GABPalpha.


Subject(s)
Cytoskeletal Proteins/metabolism , DNA Methylation , GA-Binding Protein Transcription Factor/metabolism , Proto-Oncogene Proteins c-ets/metabolism , Base Pairing , Base Sequence , CARD Signaling Adaptor Proteins , Cell Line, Tumor , Cytoskeletal Proteins/genetics , DNA/metabolism , DNA Footprinting , Deoxyribonuclease I/metabolism , Gene Knockdown Techniques , Humans , Molecular Sequence Data , Promoter Regions, Genetic/genetics , Protein Binding
8.
Cancer Res ; 68(16): 6810-21, 2008 Aug 15.
Article in English | MEDLINE | ID: mdl-18701507

ABSTRACT

Epigenetic silencing of tumor suppressor genes in human cancers is associated with aberrant methylation of promoter region CpG islands and local alterations in histone modifications. However, the mechanisms that drive these events remain unclear. Here, we establish an important role for histone H4 lysine 16 acetylation (H4K16Ac) and the histone acetyltransferase hMOF in the regulation of TMS1/ASC, a proapoptotic gene that undergoes epigenetic silencing in human cancers. In the unmethylated and active state, the TMS1 CpG island is spanned by positioned nucleosomes and marked by histone H3K4 methylation. H4K16Ac was uniquely localized to two sharp peaks that flanked the unmethylated CpG island and corresponded to strongly positioned nucleosomes. Aberrant methylation and silencing of TMS1 was accompanied by loss of the H4K16Ac peaks, loss of nucleosome positioning, hypomethylation of H3K4, and hypermethylation of H3K9. In addition, a single peak of histone H4 lysine 20 trimethylation was observed near the transcription start site. Down-regulation of hMOF or another component of the MSL complex resulted in a gene-specific decrease in H4K16Ac, loss of nucleosome positioning, and silencing of TMS1. Gene silencing induced by H4K16 deacetylation occurred independently of changes in histone methylation and DNA methylation and was reversed on hMOF reexpression. These results indicate that the selective marking of nucleosomes flanking the CpG island by hMOF is required to maintain TMS1 gene activity and suggest that the loss of H4K16Ac, mobilization of nucleosomes, and transcriptional down-regulation may be important events in the epigenetic silencing of certain tumor suppressor genes in cancer.


Subject(s)
Cytoskeletal Proteins/genetics , Gene Expression Regulation, Neoplastic , Histone Acetyltransferases/physiology , Histones/physiology , Lysine/chemistry , Acetylation , Antigens, CD , Apoptosis/physiology , Blotting, Western , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , CARD Signaling Adaptor Proteins , Cadherins/genetics , Cadherins/metabolism , Cells, Cultured , Chromatin Immunoprecipitation , CpG Islands , Cytoskeletal Proteins/metabolism , Epigenesis, Genetic , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Kidney/metabolism , Kidney/pathology , Methylation , Nucleosomes/physiology , Polymerase Chain Reaction , RNA, Small Interfering/pharmacology
9.
Mol Cell ; 30(3): 336-47, 2008 May 09.
Article in English | MEDLINE | ID: mdl-18471979

ABSTRACT

Estrogen receptor alpha (ER) is a ligand-dependent transcription factor. Upon binding estrogen, ER recruits coactivator complexes with histone acetyltransferase or methyltransferase activities to activate downstream target genes. In addition to histones, coactivators can modify ER itself and other proteins in the transactivation complex. Here, we show that ER is directly methylated at lysine 302 (K302) by the SET7 methyltransferase. SET7-mediated methylation stabilizes ER and is necessary for the efficient recruitment of ER to its target genes and for their transactivation. The SET7-ER complex structure reveals the molecular basis for ER peptide recognition and predicts that modifications or mutations of nearby residues would affect K302 methylation. Indeed, a breast cancer-associated mutation at K303 (K303R) alters methylation at K302 in vitro and in vivo. These findings raise the possibility that generation, recognition, and removal of modifications within the ER hinge region generate "ER modification cassettes" that yield distinct patterns for signaling downstream events.


Subject(s)
Estrogen Receptor alpha/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Lysine/metabolism , Amino Acid Sequence , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line , Crystallography, X-Ray , Estrogen Receptor alpha/chemistry , Estrogen Receptor alpha/genetics , Estrogens/metabolism , Female , Gene Expression Regulation , Histone Methyltransferases , Histone-Lysine N-Methyltransferase/chemistry , Histone-Lysine N-Methyltransferase/genetics , Humans , Methylation , Models, Molecular , Molecular Sequence Data , Mutation , Peptides/chemistry , Peptides/genetics , Peptides/metabolism , Protein Conformation , Protein Methyltransferases , Sequence Alignment
10.
Eur J Immunol ; 35(4): 1267-74, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15739164

ABSTRACT

Several GATA-binding sites have been identified in regions both distal to and within the murine IL-4 gene locus, yet their relative role in IL-4 expression is unknown. Chromatin immunoprecipitation assays were used to demonstrate that GATA-1 and GATA-2 are associated with a regulatory element within the second intron of the IL-4 gene in murine mast cells in vivo. Furthermore, although expression from a stably integrated wild-type IL-4 minigene parallels endogenous IL-4 gene expression, mutation of the GATA-binding element, but not an SP-1-binding site, virtually abolishes transcription in mast cells, an observation that correlates with the local loss of H3 and H4 histone acetylation in the intron. Treatment with the chromatin remodeling agents 5 azacytidine and trichostatin A can restore this defect in transcription. These results define an essential site of GATA influence on IL-4 expression in mast cells and directly support the idea that GATA factors have a profound impact on locus accessibility.


Subject(s)
Chromatin Assembly and Disassembly/physiology , DNA/metabolism , Interleukin-4/genetics , Introns/physiology , Mast Cells/metabolism , Transcription Factors/metabolism , Binding Sites/genetics , Gene Expression Regulation/physiology , Histones/metabolism , Interleukin-4/biosynthesis , Introns/genetics , Mutation
11.
J Immunol ; 169(7): 3811-8, 2002 Oct 01.
Article in English | MEDLINE | ID: mdl-12244176

ABSTRACT

IL-4 is a potent, pleiotropic cytokine that, in general, directs cellular activation, differentiation, and rescue from apoptosis. However, in mast cells, IL-4 induces the down-regulation of activation receptors and promotes cell death. Mast cells have been shown to transduce IL-4 signals through a unique C-terminally truncated isoform of STAT6. In this study, we examine the mechanism through which STAT6 is processed to generate this isoform. We demonstrate that STAT6 processing in mast cells is initiated by IL-4-induced phosphorylation and nuclear translocation of full-length STAT6 and subsequent cleavage by a nuclear serine-family protease. The location of the protease in the nucleus ensures that the truncated STAT6 has preferential access to bind DNA. IL-4-responsive target genes in mast cells are identified by chromatin immunoprecipitation of STAT6, including the IL-4 gene itself. These results suggest a molecular explanation for the suppressive effects of IL-4 on STAT6-regulated genes in mast cells.


Subject(s)
Interleukin-4/physiology , Mast Cells/immunology , Mast Cells/metabolism , Protein Processing, Post-Translational/immunology , Trans-Activators/metabolism , Active Transport, Cell Nucleus/immunology , Animals , Cell Line, Transformed , Cell Nucleus/enzymology , Cell Nucleus/immunology , Cell Nucleus/metabolism , Interleukin-4/genetics , Interleukin-4/metabolism , Mast Cells/enzymology , Mice , Mice, Inbred BALB C , Mice, Knockout , Phosphorylation , Promoter Regions, Genetic/immunology , Protein Binding/genetics , Protein Binding/immunology , Protein Isoforms/biosynthesis , Protein Isoforms/metabolism , STAT6 Transcription Factor , Serine Endopeptidases/metabolism , Signal Transduction/immunology , Trans-Activators/biosynthesis , Tumor Cells, Cultured , Tyrosine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...