Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 88
Filter
1.
Genes Dev ; 2024 Aug 21.
Article in English | MEDLINE | ID: mdl-39168636

ABSTRACT

Despite recent advances in therapeutic treatments, multiple myeloma (MM) remains an incurable malignancy. Epigenetic factors contribute to the initiation, progression, relapse, and clonal heterogeneity in MM, but our knowledge on epigenetic mechanisms underlying MM development is far from complete. The SAGA complex serves as a coactivator in transcription and catalyzes acetylation and deubiquitylation. Analyses of data sets in the Cancer Dependency Map Project revealed that many SAGA components are selective dependencies in MM. To define SAGA-specific functions, we focused on ADA2B, the only subunit in the lysine acetyltransferase (KAT) module that specifically functions in SAGA. Integration of RNA sequencing (RNA-seq), assay for transposase-accessible chromatin with sequencing (ATAC-seq), and cleavage under targets and release using nuclease assay (CUT&RUN) results identified pathways directly regulated by ADA2B including MTORC1 signaling and oncogenic programs driven by MYC, E2F, and MM-specific MAF. We discovered that ADA2B is recruited to MAF and MYC gene targets, and that MAF shares a majority of its targets with MYC in MM cells. Furthermore, we found that the SANT domain of ADA2B is required for interaction with both GCN5 and PCAF acetyltransferases, incorporation into SAGA, and ADA2B protein stability. Our findings uncover previously unknown SAGA KAT module-dependent mechanisms controlling MM cell growth, revealing a vulnerability that might be exploited for future development of MM therapy.

2.
Mol Cell ; 82(4): 716-727, 2022 02 17.
Article in English | MEDLINE | ID: mdl-35016034

ABSTRACT

Protein acetylation is conserved across phylogeny and has been recognized as one of the most prominent post-translational modifications since its discovery nearly 60 years ago. Histone acetylation is an active mark characteristic of open chromatin, but acetylation on specific lysine residues and histone variants occurs in different biological contexts and can confer various outcomes. The significance of acetylation events is indicated by the associations of lysine acetyltransferases, deacetylases, and acetyl-lysine readers with developmental disorders and pathologies. Recent advances have uncovered new roles of acetylation regulators in chromatin-centric events, which emphasize the complexity of these functional networks. In this review, we discuss mechanisms and dynamics of acetylation in chromatin organization and DNA-templated processes, including gene transcription and DNA repair and replication.


Subject(s)
Chromatin Assembly and Disassembly , Chromatin/metabolism , Histones/metabolism , Protein Processing, Post-Translational , Acetylation , Animals , Chromatin/genetics , DNA Repair , DNA Replication , Genomic Instability , Histones/genetics , Humans , Lysine , Transcription, Genetic
3.
Cell ; 159(3): 558-71, 2014 Oct 23.
Article in English | MEDLINE | ID: mdl-25417107

ABSTRACT

The recognition of modified histones by "reader" proteins constitutes a key mechanism regulating gene expression in the chromatin context. Compared with the great variety of readers for histone methylation, few protein modules that recognize histone acetylation are known. Here, we show that the AF9 YEATS domain binds strongly to histone H3K9 acetylation and, to a lesser extent, H3K27 and H3K18 acetylation. Crystal structural studies revealed that AF9 YEATS adopts an eight-stranded immunoglobin fold and utilizes a serine-lined aromatic "sandwiching" cage for acetyllysine readout, representing a novel recognition mechanism that is distinct from that of known acetyllysine readers. ChIP-seq experiments revealed a strong colocalization of AF9 and H3K9 acetylation genome-wide, which is important for the chromatin recruitment of the H3K79 methyltransferase DOT1L. Together, our studies identified the evolutionarily conserved YEATS domain as a novel acetyllysine-binding module and established a direct link between histone acetylation and DOT1L-mediated H3K79 methylation in transcription control.


Subject(s)
Histone Code , Methyltransferases/chemistry , Methyltransferases/metabolism , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Acetylation , Amino Acid Sequence , Gene Expression Regulation , Histone Acetyltransferases/chemistry , Histone Acetyltransferases/metabolism , Histone-Lysine N-Methyltransferase , Histones/metabolism , Humans , Methylation , Models, Molecular , Molecular Sequence Data , Protein Processing, Post-Translational , Protein Structure, Tertiary , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Sequence Alignment , Transcription, Genetic
4.
Cell ; 152(4): 685-9, 2013 Feb 14.
Article in English | MEDLINE | ID: mdl-23375745

ABSTRACT

Signal transduction pathways converge upon sequence-specific DNA binding factors to reprogram gene expression. Transcription factors, in turn, team up with chromatin modifying activities. However, chromatin is not simply an endpoint for signaling pathways. Histone modifications relay signals to other proteins to trigger more immediate responses than can be achieved through altered gene transcription, which might be especially important to time-urgent processes such as the execution of cell-cycle check points, chromosome segregation, or exit from mitosis. In addition, histone-modifying enzymes often have multiple nonhistone substrates, and coordination of activity toward different targets might direct signals both to and from chromatin.


Subject(s)
Chromatin/metabolism , Signal Transduction , Animals , Chromatin Assembly and Disassembly , Histones/metabolism , Humans , Methylation , Ubiquitination
5.
Cell ; 146(5): 709-19, 2011 Sep 02.
Article in English | MEDLINE | ID: mdl-21884933

ABSTRACT

Histone H3K4 trimethylation by the Set1/MLL family of proteins provides a hallmark for transcriptional activity from yeast to humans. In S. cerevisiae, H3K4 methylation is mediated by the Set1-containing COMPASS complex and is regulated in trans by prior ubiquitination of histone H2BK123. All of the events that regulate H2BK123ub and H3K4me are thought to occur at gene promoters. Here we report that this pathway is indispensable for methylation of the only other known substrate of Set1, K233 in Dam1, at kinetochores. Deletion of RAD6, BRE1, or Paf1 complex members abolishes Dam1 methylation, as does mutation of H2BK123. Our results demonstrate that Set1-mediated methylation is regulated by a general pathway regardless of substrate that is composed of transcriptional regulatory factors functioning independently of transcription. Moreover, our data identify a node of regulatory crosstalk in trans between a histone modification and modification on a nonhistone protein, demonstrating that changing chromatin states can signal functional changes in other essential cellular proteins and machineries.


Subject(s)
Cell Cycle Proteins/metabolism , Chromatin/metabolism , Histones/metabolism , Kinetochores/metabolism , Microtubule-Associated Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Signal Transduction , Animals , Histone-Lysine N-Methyltransferase/metabolism , Humans , Methylation , Mitosis , Neoplasm Proteins/metabolism , Nuclear Proteins/metabolism , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/metabolism , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitination
6.
Mol Cell ; 62(4): 558-71, 2016 05 19.
Article in English | MEDLINE | ID: mdl-27132940

ABSTRACT

Histone H2B monoubiquitination (H2Bub1) is centrally involved in gene regulation. The deubiquitination module (DUBm) of the SAGA complex is a major regulator of global H2Bub1 levels, and components of this DUBm are linked to both neurodegenerative diseases and cancer. Unexpectedly, we find that ablation of USP22, the enzymatic center of the DUBm, leads to a reduction, rather than an increase, in global H2bub1 levels. In contrast, depletion of non-enzymatic components, ATXN7L3 or ENY2, results in increased H2Bub1. These observations led us to discover two H2Bub1 DUBs, USP27X and USP51, which function independently of SAGA and compete with USP22 for ATXN7L3 and ENY2 for activity. Like USP22, USP51 and USP27X are required for normal cell proliferation, and their depletion suppresses tumor growth. Our results reveal that ATXN7L3 and ENY2 orchestrate activities of multiple deubiquitinating enzymes and that imbalances in these activities likely potentiate human diseases including cancer.


Subject(s)
Breast Neoplasms/enzymology , Cell Proliferation , Deubiquitinating Enzymes/metabolism , Histones/metabolism , Transcription Factors/metabolism , Tumor Burden , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Deubiquitinating Enzymes/genetics , Endopeptidases/genetics , Endopeptidases/metabolism , Female , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Genotype , HEK293 Cells , Humans , MCF-7 Cells , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Phenotype , RNA Interference , Signal Transduction , Thiolester Hydrolases/genetics , Thiolester Hydrolases/metabolism , Time Factors , Transcription Factors/genetics , Transfection , Ubiquitin Thiolesterase , Ubiquitin-Specific Proteases/genetics , Ubiquitin-Specific Proteases/metabolism , Ubiquitination
7.
Genes Dev ; 30(10): 1187-97, 2016 05 15.
Article in English | MEDLINE | ID: mdl-27198228

ABSTRACT

Histone H3 methylation on Lys4 (H3K4me) is associated with active gene transcription in all eukaryotes. In Saccharomyces cerevisiae, Set1 is the sole lysine methyltransferase required for mono-, di-, and trimethylation of this site. Although H3K4me3 is linked to gene expression, whether H3K4 methylation regulates other cellular processes, such as mitosis, is less clear. Here we show that both Set1 and H3K4 mutants display a benomyl resistance phenotype that requires components of the spindle assembly checkpoint (SAC), including Bub3 and Mad2. These proteins inhibit Cdc20, an activator of the anaphase-promoting complex/cyclosome (APC/C). Mutations in Cdc20 that block Mad2 interactions suppress the benomyl resistance of both set1 and H3K4 mutant cells. Furthermore, the HORMA domain in Mad2 directly binds H3, identifying a new histone H3 "reader" motif. Mad2 undergoes a conformational change important for execution of the SAC. We found that the closed (active) conformation of both yeast and human Mad2 is capable of binding methylated H3K4, but, in contrast, the open (inactive) Mad2 conformation limits interaction with methylated H3. Collectively, our data indicate that interactions between Mad2 and H3K4 regulate resolution of the SAC by limiting closed Mad2 availability for Cdc20 inhibition.


Subject(s)
Histones/metabolism , M Phase Cell Cycle Checkpoints/genetics , Mad2 Proteins/metabolism , Benomyl/pharmacology , Cdc20 Proteins/genetics , Cdc20 Proteins/metabolism , Drug Resistance/genetics , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Histones/genetics , Humans , M Phase Cell Cycle Checkpoints/drug effects , Methylation , Mutation , Protein Binding/genetics , Protein Conformation , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Spindle Apparatus/genetics , Spindle Apparatus/pathology , Transcriptional Activation/drug effects , Transcriptional Activation/physiology , Tubulin Modulators/pharmacology
8.
Genes Dev ; 29(8): 803-16, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25877919

ABSTRACT

Embryonic stem cells are maintained in a self-renewing and pluripotent state by multiple regulatory pathways. Pluripotent-specific transcriptional networks are sequentially reactivated as somatic cells reprogram to achieve pluripotency. How epigenetic regulators modulate this process and contribute to somatic cell reprogramming is not clear. Here we performed a functional RNAi screen to identify the earliest epigenetic regulators required for reprogramming. We identified components of the SAGA histone acetyltransferase complex, in particular Gcn5, as critical regulators of reprogramming initiation. Furthermore, we showed in mouse pluripotent stem cells that Gcn5 strongly associates with Myc and that, upon initiation of somatic reprogramming, Gcn5 and Myc form a positive feed-forward loop that activates a distinct alternative splicing network and the early acquisition of pluripotency-associated splicing events. These studies expose a Myc-SAGA pathway that drives expression of an essential alternative splicing regulatory network during somatic cell reprogramming.


Subject(s)
Alternative Splicing , Cellular Reprogramming/genetics , Epigenomics , Histone Acetyltransferases/metabolism , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Animals , Cell Differentiation , Cell Movement/genetics , Cells, Cultured , Embryonic Stem Cells , Gene Expression Regulation, Developmental , Histone Acetyltransferases/genetics , Mice , Pluripotent Stem Cells , RNA Interference , RNA Processing, Post-Transcriptional/genetics
9.
Development ; 146(4)2019 02 22.
Article in English | MEDLINE | ID: mdl-30718289

ABSTRACT

USP22, a component of the SAGA complex, is overexpressed in highly aggressive cancers, but the normal functions of this deubiquitinase are not well defined. We determined that loss of USP22 in mice results in embryonic lethality due to defects in extra-embryonic placental tissues and failure to establish proper vascular interactions with the maternal circulatory system. These phenotypes arise from abnormal gene expression patterns that reflect defective kinase signaling, including TGFß and several receptor tyrosine kinase pathways. USP22 deletion in endothelial cells and pericytes that are induced from embryonic stem cells also hinders these signaling cascades, with detrimental effects on cell survival and differentiation as well as on the ability to form vessels. Our findings provide new insights into the functions of USP22 during development that may offer clues to its role in disease states.


Subject(s)
Endopeptidases/metabolism , Gene Expression Regulation, Developmental , Placenta/metabolism , Signal Transduction , Animals , Cardiovascular System/metabolism , Cell Differentiation , Cell Survival , Chorioallantoic Membrane/metabolism , Ear, Inner/embryology , Embryonic Stem Cells/metabolism , Endothelial Cells/metabolism , Female , Gene Expression , Gene Expression Profiling , Mice , Phenotype , Pregnancy , Protein Processing, Post-Translational , Time Factors , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta1/metabolism , Ubiquitin Thiolesterase
10.
Exp Dermatol ; 31(3): 330-340, 2022 03.
Article in English | MEDLINE | ID: mdl-34657330

ABSTRACT

Loss of function mutations in HOXC13 have been associated with Ectodermal Dysplasia-9, Hair/Nail Type (ECTD9) in consanguineous families, characterized by sparse to complete absence of hair and nail dystrophy. Here we characterize the spontaneous mouse mutation Naked (N) as a terminal truncation in the Hoxc13 (homeobox C13) gene. Similar to previous reports for homozygous Hoxc13 knock-out (KO) mice, homozygous N/N mice exhibit generalized alopecia with abnormal nails and a short lifespan. However, in contrast to Hoxc13 heterozygous KO mice, N/+ mice show generalized or partial alopecia, associated with loss of hair fibres, along with normal lifespan and fertility. Our data point to a lack of nonsense-mediated Hoxc13 transcript decay and the presence of the truncated mutant protein in N/N and N/+ hair follicles, thus suggesting a dominant-negative mutation. To our knowledge, this is the first report of a semi-dominant and potentially dominant-negative mutation affecting Hoxc13/HOXC13. Furthermore, recreating the N mutant allele in mice using CRISPR/Cas9-mediated genome editing resulted in the same spectrum of deficiencies as those associated with the spontaneous Naked mutation, thus confirming that N is indeed a Hoxc13 mutant allele. Considering the low viability of the Hoxc13 KO mice, the Naked mutation provides an attractive new model for studying ECTD9 disease mechanisms.


Subject(s)
Ectodermal Dysplasia , Nail Diseases , Alopecia/genetics , Animals , Codon, Nonsense , Ectodermal Dysplasia/genetics , Genes, Homeobox , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Mice , Mutation , Nail Diseases/genetics , Transcription Factors/genetics
11.
Genome Res ; 28(2): 159-170, 2018 02.
Article in English | MEDLINE | ID: mdl-29273624

ABSTRACT

Noncoding transcription is a defining feature of active enhancers, linking transcription factor (TF) binding to the molecular mechanisms controlling gene expression. To determine the relationship between enhancer activity and biological outcomes in breast cancers, we profiled the transcriptomes (using GRO-seq and RNA-seq) and epigenomes (using ChIP-seq) of 11 different human breast cancer cell lines representing five major molecular subtypes of breast cancer, as well as two immortalized ("normal") human breast cell lines. In addition, we developed a robust and unbiased computational pipeline that simultaneously identifies putative subtype-specific enhancers and their cognate TFs by integrating the magnitude of enhancer transcription, TF mRNA expression levels, TF motif P-values, and enrichment of H3K4me1 and H3K27ac. When applied across the 13 different cell lines noted above, the Total Functional Score of Enhancer Elements (TFSEE) identified key breast cancer subtype-specific TFs that act at transcribed enhancers to dictate gene expression patterns determining growth outcomes, including Forkhead TFs, FOSL1, and PLAG1. FOSL1, a Fos family TF, (1) is highly enriched at the enhancers of triple negative breast cancer (TNBC) cells, (2) acts as a key regulator of the proliferation and viability of TNBC cells, but not Luminal A cells, and (3) is associated with a poor prognosis in TNBC breast cancer patients. Taken together, our results validate our enhancer identification pipeline and reveal that enhancers transcribed in breast cancer cells direct critical gene regulatory networks that promote pathogenesis.


Subject(s)
Carcinogenesis/genetics , Enhancer Elements, Genetic/genetics , Transcriptome/genetics , Triple Negative Breast Neoplasms/genetics , Adult , Cell Line, Tumor , Cell Proliferation/genetics , Cell Survival/genetics , Female , Gene Expression Regulation, Neoplastic/genetics , Gene Regulatory Networks/genetics , Histones/genetics , Humans , Middle Aged , RNA, Messenger/genetics , Transcription Factors/genetics , Triple Negative Breast Neoplasms/classification , Triple Negative Breast Neoplasms/pathology
12.
Nat Rev Genet ; 15(2): 93-106, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24366184

ABSTRACT

Cellular differentiation is, by definition, epigenetic. Genome-wide profiling of pluripotent cells and differentiated cells suggests global chromatin remodelling during differentiation, which results in a progressive transition from a fairly open chromatin configuration to a more compact state. Genetic studies in mouse models show major roles for a variety of histone modifiers and chromatin remodellers in key developmental transitions, such as the segregation of embryonic and extra-embryonic lineages in blastocyst stage embryos, the formation of the three germ layers during gastrulation and the differentiation of adult stem cells. Furthermore, rather than merely stabilizing the gene expression changes that are driven by developmental transcription factors, there is emerging evidence that chromatin regulators have multifaceted roles in cell fate decisions.


Subject(s)
Chromatin Assembly and Disassembly/genetics , Chromatin/genetics , Gene Expression Regulation , Histones/metabolism , Animals , Cell Differentiation/genetics , Cell Lineage/genetics , Chromatin/metabolism , Humans , Models, Genetic , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism
13.
J Biol Chem ; 293(19): 7476-7485, 2018 05 11.
Article in English | MEDLINE | ID: mdl-29523690

ABSTRACT

Proteins with domains that recognize and bind post-translational modifications (PTMs) of histones are collectively termed epigenetic readers. Numerous interactions between specific reader protein domains and histone PTMs and their regulatory outcomes have been reported, but little is known about how reader proteins may in turn be modulated by these interactions. Tripartite motif-containing protein 24 (TRIM24) is a histone reader aberrantly expressed in multiple cancers. Here, our investigation revealed functional cross-talk between histone acetylation and TRIM24 SUMOylation. Binding of TRIM24 to chromatin via its tandem PHD-bromodomain, which recognizes unmethylated lysine 4 and acetylated lysine 23 of histone H3 (H3K4me0/K23ac), led to TRIM24 SUMOylation at lysine residues 723 and 741. Inactivation of the bromodomain, either by mutation or with a small-molecule inhibitor, IACS-9571, abolished TRIM24 SUMOylation. Conversely, inhibition of histone deacetylation markedly increased TRIM24's interaction with chromatin and its SUMOylation. Of note, gene expression profiling of MCF7 cells expressing WT versus SUMO-deficient TRIM24 identified cell adhesion as the major pathway regulated by the cross-talk between chromatin acetylation and TRIM24 SUMOylation. In conclusion, our findings establish a new link between histone H3 acetylation and SUMOylation of the reader protein TRIM24, a functional connection that may bear on TRIM24's oncogenic function and may inform future studies of PTM cross-talk between histones and epigenetic regulators.


Subject(s)
Carrier Proteins/metabolism , Cell Adhesion , Chromatin/metabolism , Sumoylation , Acetylation , Carrier Proteins/chemistry , Epigenesis, Genetic , HEK293 Cells , Histones/metabolism , Humans , MCF-7 Cells , Oncogenes , Protein Processing, Post-Translational
14.
J Immunol ; 198(10): 3927-3938, 2017 05 15.
Article in English | MEDLINE | ID: mdl-28424240

ABSTRACT

Histone acetyltransferases (HATs) regulate inducible transcription in multiple cellular processes and during inflammatory and immune response. However, the functions of general control nonrepressed-protein 5 (Gcn5), an evolutionarily conserved HAT from yeast to human, in immune regulation remain unappreciated. In this study, we conditionally deleted Gcn5 (encoded by the Kat2a gene) specifically in T lymphocytes by crossing floxed Gcn5 and Lck-Cre mice, and demonstrated that Gcn5 plays important roles in multiple stages of T cell functions including development, clonal expansion, and differentiation. Loss of Gcn5 functions impaired T cell proliferation, IL-2 production, and Th1/Th17, but not Th2 and regulatory T cell differentiation. Gcn5 is recruited onto the il-2 promoter by interacting with the NFAT in T cells upon TCR stimulation. Interestingly, instead of directly acetylating NFAT, Gcn5 catalyzes histone H3 lysine H9 acetylation to promote IL-2 production. T cell-specific suppression of Gcn5 partially protected mice from myelin oligodendrocyte glycoprotein-induced experimental autoimmune encephalomyelitis, an experimental model for human multiple sclerosis. Our study reveals previously unknown physiological functions for Gcn5 and a molecular mechanism underlying these functions in regulating T cell immunity. Hence Gcn5 may be an important new target for autoimmune disease therapy.


Subject(s)
Histone Acetyltransferases/metabolism , Lymphocyte Activation , T-Lymphocytes/immunology , Animals , Cell Differentiation , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Gene Expression Regulation , Histone Acetyltransferases/deficiency , Histone Acetyltransferases/genetics , Interleukin-2/deficiency , Interleukin-2/genetics , Interleukin-2/immunology , Mice , NFATC Transcription Factors/genetics , Promoter Regions, Genetic , Protein Binding , Protein Processing, Post-Translational , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/physiology , Th1 Cells/immunology , Th1 Cells/physiology , Th2 Cells/immunology , Th2 Cells/physiology
15.
J Neurosci ; 37(10): 2565-2579, 2017 03 08.
Article in English | MEDLINE | ID: mdl-28154153

ABSTRACT

Diencephalic defects underlie an array of neurological diseases. Previous studies have suggested that retinoic acid (RA) signaling is involved in diencephalic development at late stages of embryonic development, but its roles and mechanisms of action during early neural development are still unclear. Here we demonstrate that mice lacking enzymatic activity of the acetyltransferase GCN5 ((Gcn5hat/hat )), which were previously characterized with respect to their exencephalic phenotype, exhibit significant diencephalic expansion, decreased diencephalic RA signaling, and increased diencephalic WNT and SHH signaling. Using a variety of molecular biology techniques in both cultured neuroepithelial cells treated with a GCN5 inhibitor and forebrain tissue from (Gcn5hat/hat ) embryos, we demonstrate that GCN5, RARα/γ, and the poorly characterized protein TACC1 form a complex in the nucleus that binds specific retinoic acid response elements in the absence of RA. Furthermore, RA triggers GCN5-mediated acetylation of TACC1, which results in dissociation of TACC1 from retinoic acid response elements and leads to transcriptional activation of RA target genes. Intriguingly, RA signaling defects caused by in vitro inhibition of GCN5 can be rescued through RA-dependent mechanisms that require RARß. Last, we demonstrate that the diencephalic expansion and transcriptional defects seen in (Gcn5hat/hat ) mutants can be rescued with gestational RA supplementation, supporting a direct link between GCN5, TACC1, and RA signaling in the developing diencephalon. Together, our studies identify a novel, nonhistone substrate for GCN5 whose modification regulates a previously undescribed, tissue-specific mechanism of RA signaling that is required to restrict diencephalic size during early forebrain development.SIGNIFICANCE STATEMENT Changes in diencephalic size and shape, as well as SNPs associated with retinoic acid (RA) signaling-associated genes, have been linked to neuropsychiatric disorders. However, the mechanisms that regulate diencephalic morphogenesis and the involvement of RA signaling in this process are poorly understood. Here we demonstrate a novel role of the acetyltransferase GCN5 in a previously undescribed mechanism of RA signaling in the developing forebrain that is required to maintain the appropriate size of the diencephalon. Together, our experiments identify a novel nonhistone substrate of GCN5, highlight an essential role for both GCN5 and RA signaling in early diencephalic development, and elucidate a novel molecular regulatory mechanism for RA signaling that is specific to the developing forebrain.


Subject(s)
Diencephalon/anatomy & histology , Diencephalon/metabolism , Signal Transduction/physiology , Tretinoin/metabolism , p300-CBP Transcription Factors/metabolism , Animals , Diencephalon/embryology , Enzyme Activation , Female , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Organ Size/physiology
16.
BMC Genomics ; 19(1): 150, 2018 02 20.
Article in English | MEDLINE | ID: mdl-29458327

ABSTRACT

BACKGROUND: Epigenetic regulators are frequently mutated or aberrantly expressed in a variety of cancers, leading to altered transcription states that result in changes in cell identity, behavior, and response to therapy. RESULTS: To define alterations in epigenetic landscapes in breast cancers, we profiled the distributions of 8 key histone modifications by ChIP-Seq, as well as primary (GRO-seq) and steady state (RNA-Seq) transcriptomes, across 13 distinct cell lines that represent 5 molecular subtypes of breast cancer and immortalized human mammary epithelial cells. DISCUSSION: Using combinatorial patterns of distinct histone modification signals, we defined subtype-specific chromatin signatures to nominate potential biomarkers. This approach identified AFAP1-AS1 as a triple negative breast cancer-specific gene associated with cell proliferation and epithelial-mesenchymal-transition. In addition, our chromatin mapping data in basal TNBC cell lines are consistent with gene expression patterns in TCGA that indicate decreased activity of the androgen receptor pathway but increased activity of the vitamin D biosynthesis pathway. CONCLUSIONS: Together, these datasets provide a comprehensive resource for histone modification profiles that define epigenetic landscapes and reveal key chromatin signatures in breast cancer cell line subtypes with potential to identify novel and actionable targets for treatment.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Histones/metabolism , Biomarkers, Tumor , Breast Neoplasms/pathology , Cell Line, Tumor , Chromatin/genetics , Chromatin/metabolism , Female , Gene Expression Profiling , Humans , Transcriptome
17.
EMBO J ; 33(17): 1912-27, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-25024434

ABSTRACT

Neuronal histone acetylation has been linked to memory consolidation, and targeting histone acetylation has emerged as a promising therapeutic strategy for neuropsychiatric diseases. However, the role of histone-modifying enzymes in the adult brain is still far from being understood. Here we use RNA sequencing to screen the levels of all known histone acetyltransferases (HATs) in the hippocampal CA1 region and find that K-acetyltransferase 2a (Kat2a)--a HAT that has not been studied for its role in memory function so far--shows highest expression. Mice that lack Kat2a show impaired hippocampal synaptic plasticity and long-term memory consolidation. We furthermore show that Kat2a regulates a highly interconnected hippocampal gene expression network linked to neuroactive receptor signaling via a mechanism that involves nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB). In conclusion, our data establish Kat2a as a novel and essential regulator of hippocampal memory consolidation.


Subject(s)
Gene Expression Regulation , Gene Regulatory Networks , Histone Acetyltransferases/metabolism , Memory , Animals , CA1 Region, Hippocampal/enzymology , Gene Expression Profiling , Histone Acetyltransferases/genetics , Mice , Mice, Knockout
18.
J Allergy Clin Immunol ; 140(6): 1651-1659.e1, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28192143

ABSTRACT

BACKGROUND: Leucine-rich repeat containing 8A (LRRC8A) is an ubiquitously expressed transmembrane protein with 17 leucine-rich repeats (LRRs) at its C-terminal end and is an essential component of the volume-regulated anion channel (VRAC), which controls cellular volume. A heterozygous mutation in LRRC8A that truncates the 2 terminal LRRs was reported in a patient with agammaglobulinemia and absent B cells and was demonstrated to exert a dominant negative effect on T- and B-cell development in mice. Lrrc8a-/- mice have severely defective T-cell development and function. It is not known whether the T- and B-cell defects caused by LRRC8A deficiency are caused by loss of VRAC activity. OBJECTIVE: We sought to determine whether VRAC activity is required for normal T-cell development and function. METHODS: VRAC activity was examined by using patch-clamp analysis. Flow cytometry was used to examine T-cell development. T-cell proliferation, cytokine secretion, and antibody titers were measured by using standard techniques. RESULTS: We demonstrate that the spontaneous mouse mutant ébouriffé (ebo/ebo) harbors a homozygous 2-bp frameshift mutation in Lrrc8a that truncates the 15 terminal LRRs of LRRC8A. The Lrrc8aebo mutation does not affect protein expression but drastically diminishes VRAC activity in T cells. ebo/ebo mice share features with Lrrc8a-/- mice that include curly hair, infertility, reduced longevity, and kidney abnormalities. However, in contrast to Lrrc8a-/- mice, ebo/ebo mice have normal T-cell development and function and intact antibody response to T-dependent antigen. CONCLUSION: LRRC8A-dependent VRAC activity is dispensable for T-cell development and function.


Subject(s)
Agammaglobulinemia/genetics , Anion Transport Proteins/metabolism , B-Lymphocytes/physiology , Membrane Proteins/metabolism , T-Lymphocytes/physiology , Animals , Antibodies/blood , Cell Differentiation , Cell Proliferation , Cell Size , Cells, Cultured , Humans , Ion Transport/genetics , Lymphocyte Activation , Membrane Proteins/genetics , Mice , Mice, Knockout , Mice, Mutant Strains , Sequence Deletion/genetics
20.
Mol Cell ; 35(3): 352-64, 2009 Aug 14.
Article in English | MEDLINE | ID: mdl-19683498

ABSTRACT

Histone acetyltransferases (HATs) play important roles in gene regulation and DNA repair by influencing the accessibility of chromatin to transcription factors and repair proteins. Here, we show that deletion of Gcn5 leads to telomere dysfunction in mouse and human cells. Biochemical studies reveal that depletion of Gcn5 or ubiquitin-specific protease 22 (Usp22), which is another bona fide component of the Gcn5-containing SAGA complex, increases ubiquitination and turnover of TRF1, a primary component of the telomeric shelterin complex. Inhibition of the proteasome or overexpression of USP22 opposes this effect. The USP22 deubiquitinating module requires association with SAGA complexes for activity, and we find that depletion of Gcn5 compromises this association in mammalian cells. Thus, our results indicate that Gcn5 regulates TRF1 levels through effects on Usp22 activity and SAGA integrity.


Subject(s)
Telomere-Binding Proteins/metabolism , Telomere/metabolism , Telomeric Repeat Binding Protein 1/metabolism , Thiolester Hydrolases/metabolism , p300-CBP Transcription Factors/physiology , Animals , Cells, Cultured , Chromosome Aberrations , DNA Breaks, Double-Stranded , DNA Repair/genetics , Gene Deletion , Humans , Mice , Models, Biological , Proteasome Inhibitors , Protein Stability , Shelterin Complex , Telomere-Binding Proteins/genetics , Telomeric Repeat Binding Protein 1/genetics , Thiolester Hydrolases/genetics , Ubiquitin Thiolesterase , p300-CBP Transcription Factors/genetics , p300-CBP Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL