Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Nat Immunol ; 22(10): 1327-1340, 2021 10.
Article in English | MEDLINE | ID: mdl-34556886

ABSTRACT

During the germinal center (GC) reaction, B cells undergo profound transcriptional, epigenetic and genomic architectural changes. How such changes are established remains unknown. Mapping chromatin accessibility during the humoral immune response, we show that OCT2 was the dominant transcription factor linked to differential accessibility of GC regulatory elements. Silent chromatin regions destined to become GC-specific super-enhancers (SEs) contained pre-positioned OCT2-binding sites in naive B cells (NBs). These preloaded SE 'seeds' featured spatial clustering of regulatory elements enriched in OCT2 DNA-binding motifs that became heavily loaded with OCT2 and its GC-specific coactivator OCAB in GC B cells (GCBs). SEs with high abundance of pre-positioned OCT2 binding preferentially formed long-range chromatin contacts in GCs, to support expression of GC-specifying factors. Gain in accessibility and architectural interactivity of these regions were dependent on recruitment of OCAB. Pre-positioning key regulators at SEs may represent a broadly used strategy for facilitating rapid cell fate transitions.


Subject(s)
Chromatin/immunology , Immunity, Humoral/immunology , Organic Cation Transporter 2/immunology , Protein Domains/immunology , Animals , B-Lymphocytes/immunology , Cell Differentiation/immunology , Epigenomics/methods , Female , Genomics/methods , Germinal Center/immunology , Male , Mice , Mice, Inbred C57BL , Transcription Factors/immunology
2.
Mol Cell ; 78(4): 765-778.e7, 2020 05 21.
Article in English | MEDLINE | ID: mdl-32298650

ABSTRACT

Increasing evidence suggests that tRNA levels are dynamically and specifically regulated in response to internal and external cues to modulate the cellular translational program. However, the molecular players and the mechanisms regulating the gene-specific expression of tRNAs are still unknown. Using an inducible auxin-degron system to rapidly deplete RPB1 (the largest subunit of RNA Pol II) in living cells, we identified Pol II as a direct gene-specific regulator of tRNA transcription. Our data suggest that Pol II transcription robustly interferes with Pol III function at specific tRNA genes. This activity was further found to be essential for MAF1-mediated repression of a large set of tRNA genes during serum starvation, indicating that repression of tRNA genes by Pol II is dynamically regulated. Hence, Pol II plays a direct and central role in the gene-specific regulation of tRNA expression.


Subject(s)
Gene Expression Regulation , RNA Polymerase III/metabolism , RNA Polymerase II/metabolism , RNA, Transfer/metabolism , Repressor Proteins/metabolism , Retinol-Binding Proteins, Cellular/metabolism , Transcription, Genetic , HeLa Cells , Humans , Protein Processing, Post-Translational , RNA Polymerase II/genetics , RNA Polymerase III/genetics , RNA, Transfer/genetics , Repressor Proteins/genetics , Retinol-Binding Proteins, Cellular/genetics
3.
Mol Cell ; 80(5): 845-861.e10, 2020 12 03.
Article in English | MEDLINE | ID: mdl-33232656

ABSTRACT

Locus control region (LCR) functions define cellular identity and have critical roles in diseases such as cancer, although the hierarchy of structural components and associated factors that drive functionality are incompletely understood. Here we show that OCA-B, a B cell-specific coactivator essential for germinal center (GC) formation, forms a ternary complex with the lymphoid-enriched OCT2 and GC-specific MEF2B transcription factors and that this complex occupies and activates an LCR that regulates the BCL6 proto-oncogene and is uniquely required by normal and malignant GC B cells. Mechanistically, through OCA-B-MED1 interactions, this complex is required for Mediator association with the BCL6 promoter. Densely tiled CRISPRi screening indicates that only LCR segments heavily bound by this ternary complex are essential for its function. Our results demonstrate how an intimately linked complex of lineage- and stage-specific factors converges on specific and highly essential enhancer elements to drive the function of a cell-type-defining LCR.


Subject(s)
B-Lymphocytes/immunology , Germinal Center/immunology , Locus Control Region/immunology , Animals , B-Lymphocytes/cytology , Cell Line, Tumor , Germinal Center/cytology , HEK293 Cells , Humans , MEF2 Transcription Factors/genetics , MEF2 Transcription Factors/immunology , Mice , Mice, Knockout , Organic Cation Transporter 2/genetics , Organic Cation Transporter 2/immunology , Proto-Oncogene Mas , Proto-Oncogene Proteins c-bcl-6/genetics , Proto-Oncogene Proteins c-bcl-6/immunology , Trans-Activators/genetics , Trans-Activators/immunology
4.
Genes Dev ; 34(11-12): 767-784, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32381628

ABSTRACT

Liver regeneration and metabolism are highly interconnected. Here, we show that hepatocyte-specific ablation of RNA polymerase II (Pol II)-associated Gdown1 leads to down-regulation of highly expressed genes involved in plasma protein synthesis and metabolism, a concomitant cell cycle re-entry associated with induction of cell cycle-related genes (including cyclin D1), and up-regulation of p21 through activation of p53 signaling. In the absence of p53, Gdown1-deficient hepatocytes show a severe dysregulation of cell cycle progression, with incomplete mitoses, and a premalignant-like transformation. Mechanistically, Gdown1 is associated with elongating Pol II on the highly expressed genes and its ablation leads to reduced Pol II recruitment to these genes, suggesting that Pol II redistribution may facilitate hepatocyte re-entry into the cell cycle. These results establish an important physiological function for a Pol II regulatory factor (Gdown1) in the maintenance of normal liver cell transcription through constraints on cell cycle re-entry of quiescent hepatocytes.


Subject(s)
Cell Cycle/genetics , Down-Regulation/genetics , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , Animals , Cell Proliferation/genetics , Gene Expression Regulation, Developmental/genetics , Gene Knockout Techniques , Genes, p53/genetics , Hepatocytes , Liver/cytology , Liver/metabolism , Signal Transduction/genetics
5.
Proc Natl Acad Sci U S A ; 118(6)2021 02 09.
Article in English | MEDLINE | ID: mdl-33542097

ABSTRACT

The chimeric transcription factor E2A-PBX1, containing the N-terminal activation domains of E2A fused to the C-terminal DNA-binding domain of PBX1, results in 5% of pediatric acute lymphoblastic leukemias (ALL). We recently have reported a mechanism for RUNX1-dependent recruitment of E2A-PBX1 to chromatin in pre-B leukemic cells; but the subsequent E2A-PBX1 functions through various coactivators and the general transcriptional machinery remain unclear. The Mediator complex plays a critical role in cell-specific gene activation by serving as a key coactivator for gene-specific transcription factors that facilitates their function through the RNA polymerase II transcriptional machinery, but whether Mediator contributes to aberrant expression of E2A-PBX1 target genes remains largely unexplored. Here we show that Mediator interacts directly with E2A-PBX1 through an interaction of the MED1 subunit with an E2A activation domain. Results of MED1 depletion by CRISPR/Cas9 further indicate that MED1 is specifically required for E2A-PBX1-dependent gene activation and leukemic cell growth. Integrated transcriptome and cistrome analyses identify pre-B cell receptor and cell cycle regulatory genes as direct cotargets of MED1 and E2A-PBX1. Notably, complementary biochemical analyses also demonstrate that recruitment of E2A-PBX1 to a target DNA template involves a direct interaction with DNA-bound RUNX1 that can be further stabilized by EBF1. These findings suggest that E2A-PBX1 interactions with RUNX1 and MED1/Mediator are of functional importance for both gene-specific transcriptional activation and maintenance of E2A-PBX1-driven leukemia. The MED1 dependency for E2A-PBX1-mediated gene activation and leukemogenesis may provide a potential therapeutic opportunity by targeting MED1 in E2A-PBX1+ pre-B leukemia.


Subject(s)
Carcinogenesis/genetics , Homeodomain Proteins/metabolism , Leukemia/genetics , Leukemia/pathology , Mediator Complex Subunit 1/metabolism , Oncogene Proteins, Fusion/metabolism , Transcription, Genetic , B-Lymphocytes/pathology , Carcinogenesis/pathology , Cell Cycle Checkpoints , Cell Proliferation/genetics , Cell Survival , Core Binding Factor Alpha 2 Subunit/metabolism , DNA, Neoplasm/metabolism , Down-Regulation/genetics , Gene Expression Regulation, Leukemic , Genes, Neoplasm , Humans , Protein Binding , Protein Stability
6.
Proc Natl Acad Sci U S A ; 111(4): 1355-60, 2014 Jan 28.
Article in English | MEDLINE | ID: mdl-24474760

ABSTRACT

O-linked N-acetylglucosamine (GlcNAc) transferase (OGT) is the only known enzyme that catalyzes the O-GlcNAcylation of proteins at the Ser or Thr side chain hydroxyl group. OGT participates in transcriptional and epigenetic regulation, and dysregulation of OGT has been implicated in diseases such as cancer. However, the underlying mechanism is largely unknown. Here we show that OGT is required for the trimethylation of histone 3 at K27 to form the product H3K27me3, a process catalyzed by the histone methyltransferase enhancer of zeste homolog 2 (EZH2) in the polycomb repressive complex 2 (PRC2). H3K27me3 is one of the most important histone modifications to mark the transcriptionally silenced chromatin. We found that the level of H3K27me3, but not other H3 methylation products, was greatly reduced upon OGT depletion. OGT knockdown specifically down-regulated the protein stability of EZH2, without altering the levels of H3K27 demethylases UTX and JMJD3, and disrupted the integrity of the PRC2 complex. Furthermore, the interaction of OGT and EZH2/PRC2 was detected by coimmunoprecipitation and cosedimentation experiments. Importantly, we identified that serine 75 is the site for EZH2 O-GlcNAcylation, and the EZH2 mutant S75A exhibited reduction in stability. Finally, microarray and ChIP analysis have characterized a specific subset of potential tumor suppressor genes subject to repression via the OGT-EZH2 axis. Together these results indicate that OGT-mediated O-GlcNAcylation at S75 stabilizes EZH2 and hence facilitates the formation of H3K27me3. The study not only uncovers a functional posttranslational modification of EZH2 but also reveals a unique epigenetic role of OGT in regulating histone methylation.


Subject(s)
Acetylglucosamine/metabolism , Polycomb Repressive Complex 2/metabolism , DNA Methylation , Down-Regulation , Enhancer of Zeste Homolog 2 Protein , Gene Knockdown Techniques , Genes, Tumor Suppressor , Humans , N-Acetylglucosaminyltransferases/genetics , N-Acetylglucosaminyltransferases/metabolism , Protein Stability
7.
Nat Commun ; 15(1): 2879, 2024 Apr 03.
Article in English | MEDLINE | ID: mdl-38570506

ABSTRACT

Despite regulating overlapping gene enhancers and pathways, CREBBP and KMT2D mutations recurrently co-occur in germinal center (GC) B cell-derived lymphomas, suggesting potential oncogenic cooperation. Herein, we report that combined haploinsufficiency of Crebbp and Kmt2d induces a more severe mouse lymphoma phenotype (vs either allele alone) and unexpectedly confers an immune evasive microenvironment manifesting as CD8+ T-cell exhaustion and reduced infiltration. This is linked to profound repression of immune synapse genes that mediate crosstalk with T-cells, resulting in aberrant GC B cell fate decisions. From the epigenetic perspective, we observe interaction and mutually dependent binding and function of CREBBP and KMT2D on chromatin. Their combined deficiency preferentially impairs activation of immune synapse-responsive super-enhancers, pointing to a particular dependency for both co-activators at these specialized regulatory elements. Together, our data provide an example where chromatin modifier mutations cooperatively shape and induce an immune-evasive microenvironment to facilitate lymphomagenesis.


Subject(s)
Lymphoma, Large B-Cell, Diffuse , Animals , Mice , B-Lymphocytes/metabolism , Chromatin/genetics , Chromatin/metabolism , Germinal Center/metabolism , Lymphoma, Large B-Cell, Diffuse/genetics , Mutation , Tumor Microenvironment/genetics
8.
Cell Res ; 33(2): 165-183, 2023 02.
Article in English | MEDLINE | ID: mdl-36646760

ABSTRACT

Estrogen-related receptors (ERRα/ß/γ) are orphan nuclear receptors that function in energy-demanding physiological processes, as well as in development and stem cell maintenance, but mechanisms underlying target gene activation by ERRs are largely unknown. Here, reconstituted biochemical assays that manifest ERR-dependent transcription have revealed two complementary mechanisms. On DNA templates, ERRs activate transcription with just the normal complement of general initiation factors through an interaction of the ERR DNA-binding domain with the p52 subunit of initiation factor TFIIH. On chromatin templates, activation by ERRs is dependent on AF2 domain interactions with the cell-specific coactivator PGC-1α, which in turn recruits the ubiquitous p300 and MED1/Mediator coactivators. This role of PGC-1α may also be fulfilled by other AF2-interacting coactivators like NCOA3, which is shown to recruit Mediator selectively to ERRß and ERRγ. Importantly, combined genetic and RNA-seq analyses establish that both the TFIIH and the AF2 interaction-dependent pathways are essential for ERRß/γ-selective gene expression and pluripotency maintenance in embryonic stem cells in which NCOA3 is a critical coactivator.


Subject(s)
Furylfuramide , Orphan Nuclear Receptors , DNA , Promoter Regions, Genetic , Transcriptional Activation , Receptors, Estrogen/metabolism
9.
bioRxiv ; 2023 Feb 13.
Article in English | MEDLINE | ID: mdl-36824887

ABSTRACT

Mutations affecting enhancer chromatin regulators CREBBP and KMT2D are highly co-occurrent in germinal center (GC)-derived lymphomas and other tumors, even though regulating similar pathways. Herein, we report that combined haploinsufficiency of Crebbp and Kmt2d (C+K) indeed accelerated lymphomagenesis. C+K haploinsufficiency induced GC hyperplasia by altering cell fate decisions, skewing B cells away from memory and plasma cell differentiation. C+K deficiency particularly impaired enhancer activation for immune synapse genes involved in exiting the GC reaction. This effect was especially severe at super-enhancers for immunoregulatory and differentiation genes. Mechanistically, CREBBP and KMT2D formed a complex, were highly co-localized on chromatin, and were required for each-other's stable recruitment to enhancers. Notably, C+K lymphomas in mice and humans manifested significantly reduced CD8 + T-cell abundance. Hence, deficiency of C+K cooperatively induced an immune evasive phenotype due at least in part to failure to activate key immune synapse super-enhancers, associated with altered immune cell fate decisions. SIGNIFICANCE: Although CREBBP and KMT2D have similar enhancer regulatory functions, they are paradoxically co-mutated in lymphomas. We show that their combined loss causes specific disruption of super-enhancers driving immune synapse genes. Importantly, this leads to reduction of CD8 cells in lymphomas, linking super-enhancer function to immune surveillance, with implications for immunotherapy resistance.

10.
J Biol Chem ; 286(41): 35843-35851, 2011 Oct 14.
Article in English | MEDLINE | ID: mdl-21852232

ABSTRACT

Global histone H1 phosphorylation correlates with cell cycle progression. However, the function of site-specific H1 variant phosphorylation remains unclear. Our mass spectrometry analysis revealed a novel N-terminal phosphorylation of the major H1 variant H1.4 at serine 35 (H1.4S35ph), which accumulates at mitosis immediately after H3 phosphorylation at serine 10. Protein kinase A (PKA) was found to be a kinase for H1.4S35. Importantly, Ser-35-phosphorylated H1.4 dissociates from mitotic chromatin. Moreover, H1.4S35A substitution mutant cannot efficiently rescue the mitotic defect following H1.4 depletion, and inhibition of PKA activity increases the mitotic chromatin compaction depending on H1.4. Our results not only indicate that PKA-mediated H1.4S35 phosphorylation dissociates H1.4 from mitotic chromatin but also suggest that this phosphorylation is necessary for specific mitotic functions.


Subject(s)
Chromatin/metabolism , Chromosomes, Human/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Histones/metabolism , Mitosis/physiology , Amino Acid Substitution , Chromatin/genetics , Chromosomes, Human/genetics , Cyclic AMP-Dependent Protein Kinases/genetics , HeLa Cells , Histones/genetics , Humans , Mutation, Missense , Phosphorylation/physiology , Serine/genetics , Serine/metabolism
11.
Cancer Discov ; 10(3): 440-459, 2020 03.
Article in English | MEDLINE | ID: mdl-31915197

ABSTRACT

CREBBP mutations are highly recurrent in B-cell lymphomas and either inactivate its histone acetyltransferase (HAT) domain or truncate the protein. Herein, we show that these two classes of mutations yield different degrees of disruption of the epigenome, with HAT mutations being more severe and associated with inferior clinical outcome. Genes perturbed by CREBBP mutation are direct targets of the BCL6-HDAC3 onco-repressor complex. Accordingly, we show that HDAC3-selective inhibitors reverse CREBBP-mutant aberrant epigenetic programming, resulting in: (i) growth inhibition of lymphoma cells through induction of BCL6 target genes such as CDKN1A and (ii) restoration of immune surveillance due to induction of BCL6-repressed IFN pathway and antigen-presenting genes. By reactivating these genes, exposure to HDAC3 inhibitors restored the ability of tumor-infiltrating lymphocytes to kill DLBCL cells in an MHC class I and II-dependent manner, and synergized with PD-L1 blockade in a syngeneic model in vivo. Hence, HDAC3 inhibition represents a novel mechanism-based immune epigenetic therapy for CREBBP-mutant lymphomas. SIGNIFICANCE: We have leveraged the molecular characterization of different types of CREBBP mutations to define a rational approach for targeting these mutations through selective inhibition of HDAC3. This represents an attractive therapeutic avenue for targeting synthetic vulnerabilities in CREBBP-mutant cells in tandem with promoting antitumor immunity.This article is highlighted in the In This Issue feature, p. 327.


Subject(s)
CREB-Binding Protein/genetics , Cyclin-Dependent Kinase Inhibitor p21/genetics , Histone Deacetylases/genetics , Lymphoma/genetics , Proto-Oncogene Proteins c-bcl-6/genetics , Animals , Antigen-Presenting Cells/drug effects , Antigen-Presenting Cells/immunology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , B7-H1 Antigen/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Epigenome/genetics , Epigenome/immunology , Genes, MHC Class I/immunology , Histocompatibility Antigens Class II/immunology , Histone Acetyltransferases/genetics , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/drug effects , Humans , Immune Checkpoint Inhibitors/pharmacology , Immune System/drug effects , Immune System/immunology , Interferons/genetics , Interferons/immunology , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Lymphoma/drug therapy , Lymphoma/immunology , Lymphoma/pathology , Mice , Mutation/genetics , Signal Transduction/drug effects
12.
Cell Rep ; 28(2): 472-485.e5, 2019 07 09.
Article in English | MEDLINE | ID: mdl-31291582

ABSTRACT

The NuRD complex contains both chromatin remodeling and histone deacetylase activities. Mice lacking the MTA2 subunit of NuRD show developmental defects in pro-B, pre-B, immature B, and marginal zone B cells, and abnormal germinal center B cell differentiation during immune responses. Mta2 inactivation also causes a derepression of Igll1 and VpreB1 genes in pre-B cells. Furthermore, MTA2/NuRD interacts directly with AIOLOS/IKAROS and shows a striking overlap with AIOLOS/IKAROS target genes in human pre-B cells, suggesting a functional inter-dependence between MTA2/NuRD and AIOLOS. Mechanistically, MTA2 deficiency in mice leads to increased H3K27 acetylation at both Igll1 and VpreB1 promoters. Gene profiling analyses also identify distinct MTA2-dependent transcription programs in pro-B and pre-B cells. In addition, we find a strong synergy between MTA2 and OCA-B in repressing Igll1 and VpreB1 at the pre-B cell stage, and in regulating both the pre-B to immature B transition and splenic B cell development.


Subject(s)
B-Lymphocytes/immunology , Mi-2 Nucleosome Remodeling and Deacetylase Complex/genetics , Precursor Cells, B-Lymphoid/immunology , Repressor Proteins/genetics , Trans-Activators/genetics , Animals , Humans , Mice
13.
Cancer Discov ; 7(1): 38-53, 2017 01.
Article in English | MEDLINE | ID: mdl-27733359

ABSTRACT

Somatic mutations in CREBBP occur frequently in B-cell lymphoma. Here, we show that loss of CREBBP facilitates the development of germinal center (GC)-derived lymphomas in mice. In both human and murine lymphomas, CREBBP loss-of-function resulted in focal depletion of enhancer H3K27 acetylation and aberrant transcriptional silencing of genes that regulate B-cell signaling and immune responses, including class II MHC. Mechanistically, CREBBP-regulated enhancers are counter-regulated by the BCL6 transcriptional repressor in a complex with SMRT and HDAC3, which we found to bind extensively to MHC class II loci. HDAC3 loss-of-function rescued repression of these enhancers and corresponding genes, including MHC class II, and more profoundly suppressed CREBBP-mutant lymphomas in vitro and in vivo Hence, CREBBP loss-of-function contributes to lymphomagenesis by enabling unopposed suppression of enhancers by BCL6/SMRT/HDAC3 complexes, suggesting HDAC3-targeted therapy as a precision approach for CREBBP-mutant lymphomas. SIGNIFICANCE: Our findings establish the tumor suppressor function of CREBBP in GC lymphomas in which CREBBP mutations disable acetylation and result in unopposed deacetylation by BCL6/SMRT/HDAC3 complexes at enhancers of B-cell signaling and immune response genes. Hence, inhibition of HDAC3 can restore the enhancer histone acetylation and may serve as a targeted therapy for CREBBP-mutant lymphomas. Cancer Discov; 7(1); 38-53. ©2016 AACR.See related commentary by Höpken, p. 14This article is highlighted in the In This Issue feature, p. 1.


Subject(s)
CREB-Binding Protein/genetics , Germinal Center/metabolism , Histone Deacetylases/genetics , Lymphoma, Large B-Cell, Diffuse/genetics , Mutation , Acetylation , Animals , CREB-Binding Protein/metabolism , Cell Line, Tumor , Enhancer Elements, Genetic , Gene Knockout Techniques , Histone Deacetylases/metabolism , Histones/metabolism , Humans , Lymphoma, Large B-Cell, Diffuse/metabolism , Mice , Neoplasm Transplantation , Nuclear Receptor Co-Repressor 2/genetics , Proto-Oncogene Proteins c-bcl-6/genetics , Transcription, Genetic
14.
Lipids ; 41(8): 797-803, 2006 Aug.
Article in English | MEDLINE | ID: mdl-17120934

ABSTRACT

Our ex vivo study revealed that BRE had significantly stronger ability to inhibit LDL oxidation than white rice extract (WRE). The purpose of this study was to investigate whether black rice extract (BRE) supplementation might ameliorate oxidative stress and enhance antioxidant enzyme activities in HepG2 cells and in C57BL/6 mice. In the cellular study, superoxide anions (O2*-) and reactive oxygen species (ROS) in the BRE group were significantly suppressed. The BRE group also showed significant increases in superoxide dismutase (SOD) and catalase (CAT) activities by 161.6% and 73.4%, respectively. The major components responsible for the free-radical-scavenging and antioxidative properties might be cyanidin-3-O-glucoside chloride and peonidin-3-O-glucuside chloride. In the animal study, male C57BL/6 mice were divided into three groups (control, BRE, and WRE). Plasma HDL-cholesterol was significantly higher, and thiobarbituric, acid-reactive substances were significantly lower in the BRE group, whereas plasma levels of total cholesterol and triglyceride were not affected by BRE supplementation. Increased hepatic SOD and CAT activities were observed in BRE-treated mice as compared to the control mice. However, no changes were detected for the protein expression of antioxidant enzymes by Western blot analysis. Our data suggest that antioxidative effects exerted by BRE are mediated through decreases in free-radical generation as well as increases in SOD and CAT activities both in vitro and in vivo.


Subject(s)
Antioxidants/physiology , Catalase/biosynthesis , Oryza/physiology , Superoxide Dismutase/biosynthesis , Animals , Cell Line , Enzyme Induction/physiology , Humans , Lipoproteins, LDL/metabolism , Male , Mice , Mice, Inbred C57BL , Oxidation-Reduction/drug effects , Plant Extracts/pharmacology
15.
Cell Res ; 21(4): 642-53, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21221131

ABSTRACT

Glucose-regulated protein 78 (GRP78), a key regulator of endoplasmic reticulum (ER) stress, facilitates cancer cell growth and viral replication. The mechanism leading to grp78 gene activation during viral infection is largely unknown. In this study, we show that the immediate-early 1 (IE1-72) protein of the human cytomegalovirus (HCMV) is essential for HCMV-mediated GRP78 activation. IE1-72 upregulated grp78 gene expression depending on the ATP-binding site, the zinc-finger domain and the putative leucine-zipper motif of IE1-72, as well as the ER stress response elements (ERSEs) on the grp78 promoter. The purified IE1-72 protein bound to the CCAAT box within ERSE in vitro, whereas deletion mutants of IE1-72 deficient in grp78 promoter stimulation failed to do so. Moreover, IE1-72 binding to the grp78 promoter in infected cells accompanied the recruitment of TATA box-binding protein-associated factor 1 (TAF1), a histone acetyltransferase, and the increased level of acetylated histone H4, an indicator of active-state chromatin. These results provide evidence that HCMV IE1-72 activates grp78 gene expression through direct promoter binding and modulation of the local chromatin structure, indicating an active viral mechanism of cellular chaperone induction for viral growth.


Subject(s)
Cytomegalovirus/metabolism , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/metabolism , Heat-Shock Proteins/metabolism , Immediate-Early Proteins/metabolism , Transcriptional Activation , Blotting, Western , CCAAT-Binding Factor/metabolism , Cell Line , Chromatin Immunoprecipitation , Cytomegalovirus/genetics , Endoplasmic Reticulum Chaperone BiP , Gene Expression , Heat-Shock Proteins/genetics , Histones/metabolism , Humans , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Polymerase Chain Reaction , Promoter Regions, Genetic , TATA-Binding Protein Associated Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL