Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Mol Cell ; 76(3): 500-515.e8, 2019 11 07.
Article in English | MEDLINE | ID: mdl-31422874

ABSTRACT

Diet-induced obesity can be caused by impaired thermogenesis of beige adipocytes, the brown-like adipocytes in white adipose tissue (WAT). Promoting brown-like features in WAT has been an attractive therapeutic approach for obesity. However, the mechanism underlying beige adipocyte formation is largely unknown. N-α-acetyltransferase 10 protein (Naa10p) catalyzes N-α-acetylation of nascent proteins, and overexpression of human Naa10p is linked to cancer development. Here, we report that both conventional and adipose-specific Naa10p deletions in mice result in increased energy expenditure, thermogenesis, and beige adipocyte differentiation. Mechanistically, Naa10p acetylates the N terminus of Pgc1α, which prevents Pgc1α from interacting with Pparγ to activate key genes, such as Ucp1, involved in beige adipocyte function. Consistently, fat tissues of obese human individuals show higher NAA10 expression. Thus, Naa10p-mediated N-terminal acetylation of Pgc1α downregulates thermogenic gene expression, making inhibition of Naa10p enzymatic activity a potential strategy for treating obesity.


Subject(s)
Adipocytes, Beige/enzymology , Adipose Tissue, Beige/enzymology , N-Terminal Acetyltransferase A/metabolism , N-Terminal Acetyltransferase E/metabolism , Obesity/enzymology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Protein Processing, Post-Translational , Thermogenesis , Acetylation , Adipose Tissue, Beige/physiopathology , Adiposity , Adolescent , Adult , Aged , Animals , Case-Control Studies , Diet, High-Fat , Disease Models, Animal , Energy Metabolism , Female , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , N-Terminal Acetyltransferase A/deficiency , N-Terminal Acetyltransferase A/genetics , N-Terminal Acetyltransferase E/deficiency , N-Terminal Acetyltransferase E/genetics , NIH 3T3 Cells , Obesity/genetics , Obesity/physiopathology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Phenotype , Signal Transduction , Young Adult
2.
Mol Cell ; 68(1): 89-103.e7, 2017 Oct 05.
Article in English | MEDLINE | ID: mdl-28943313

ABSTRACT

Genomic imprinting is an allelic gene expression phenomenon primarily controlled by allele-specific DNA methylation at the imprinting control region (ICR), but the underlying mechanism remains largely unclear. N-α-acetyltransferase 10 protein (Naa10p) catalyzes N-α-acetylation of nascent proteins, and mutation of human Naa10p is linked to severe developmental delays. Here we report that Naa10-null mice display partial embryonic lethality, growth retardation, brain disorders, and maternal effect lethality, phenotypes commonly observed in defective genomic imprinting. Genome-wide analyses further revealed global DNA hypomethylation and enriched dysregulation of imprinted genes in Naa10p-knockout embryos and embryonic stem cells. Mechanistically, Naa10p facilitates binding of DNA methyltransferase 1 (Dnmt1) to DNA substrates, including the ICRs of the imprinted allele during S phase. Moreover, the lethal Ogden syndrome-associated mutation of human Naa10p disrupts its binding to the ICR of H19 and Dnmt1 recruitment. Our study thus links Naa10p mutation-associated Ogden syndrome to defective DNA methylation and genomic imprinting.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/genetics , Developmental Disabilities/genetics , Epigenesis, Genetic , Genomic Imprinting , N-Terminal Acetyltransferase A/genetics , N-Terminal Acetyltransferase E/genetics , RNA, Long Noncoding/genetics , Animals , DNA/genetics , DNA/metabolism , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation , Developmental Disabilities/metabolism , Developmental Disabilities/pathology , Disease Models, Animal , Embryo, Mammalian , Female , Gene Deletion , Genes, Lethal , Genome-Wide Association Study , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mouse Embryonic Stem Cells/metabolism , Mouse Embryonic Stem Cells/pathology , N-Terminal Acetyltransferase A/deficiency , N-Terminal Acetyltransferase E/deficiency , Protein Binding , RNA, Long Noncoding/metabolism , S Phase/genetics
3.
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
4.
Cancer Res ; 73(15): 4711-21, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23722541

ABSTRACT

The retinoblastoma binding protein RBP2 (KDM5A) is a histone demethylase that promotes gastric cancer cell growth and is enriched in drug-resistant lung cancer cells. In tumor-prone mice lacking the tumor suppressor gene RB or MEN1, genetic ablation of RBP2 can suppress tumor initiation, but the pathogenic breadth and mechanistic aspects of this effect relative to human tumors have not been defined. Here, we approached this question in the context of lung cancer. RBP2 was overexpressed in human lung cancer tissues where its depletion impaired cell proliferation, motility, migration, invasion, and metastasis. RBP2 oncogenicity relied on its demethylase and DNA-binding activities. RBP2 upregulated expression of cyclins D1 and E1 while suppressing the expression of cyclin-dependent kinase inhibitor p27 (CDKN1B), each contributing to RBP2-mediated cell proliferation. Expression microarray analyses revealed that RBP2 promoted expression of integrin-ß1 (ITGB1), which is implicated in lung cancer metastasis. Mechanistic investigations established that RBP2 bound directly to the p27, cyclin D1, and ITGB1 promoters and that exogenous expression of cyclin D1, cyclin E1, or ITGB1 was sufficient to rescue proliferation or migration/invasion, respectively. Taken together, our results establish an oncogenic role for RBP2 in lung tumorigenesis and progression and uncover novel RBP2 targets mediating this role.


Subject(s)
Carcinoma, Non-Small-Cell Lung/enzymology , Cell Transformation, Neoplastic/metabolism , Lung Neoplasms/enzymology , Neoplasm Invasiveness/pathology , Retinol-Binding Proteins, Cellular/metabolism , Animals , Blotting, Western , Carcinoma, Non-Small-Cell Lung/pathology , Cell Proliferation , Cells, Cultured , Chromatin Immunoprecipitation , Disease Progression , Female , Gene Knockdown Techniques , Humans , Lung Neoplasms/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Metastasis , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction , Xenograft Model Antitumor Assays
5.
Cell Rep ; 2(3): 568-79, 2012 Sep 27.
Article in English | MEDLINE | ID: mdl-22999938

ABSTRACT

Tumor suppressor gene silencing through cytosine methylation contributes to cancer formation. Whether DNA demethylation enzymes counteract this oncogenic effect is unknown. Here, we show that TET1, a dioxygenase involved in cytosine demethylation, is downregulated in prostate and breast cancer tissues. TET1 depletion facilitates cell invasion, tumor growth, and cancer metastasis in prostate xenograft models and correlates with poor survival rates in breast cancer patients. Consistently, enforced expression of TET1 reduces cell invasion and breast xenograft tumor formation. Mechanistically, TET1 suppresses cell invasion through its dioxygenase and DNA binding activities. Furthermore, TET1 maintains the expression of tissue inhibitors of metalloproteinase (TIMP) family proteins 2 and 3 by inhibiting their DNA methylation. Concurrent low expression of TET1 and TIMP2 or TIMP3 correlates with advanced node status in clinical samples. Together, these results illustrate a mechanism by which TET1 suppresses tumor development and invasion partly through downregulation of critical gene methylation.


Subject(s)
Breast Neoplasms/metabolism , DNA-Binding Proteins/biosynthesis , Gene Expression Regulation, Neoplastic , Prostatic Neoplasms/metabolism , Proto-Oncogene Proteins/biosynthesis , Tissue Inhibitor of Metalloproteinase-2/biosynthesis , Tissue Inhibitor of Metalloproteinase-3/biosynthesis , Tumor Suppressor Proteins/biosynthesis , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , DNA Methylation/genetics , DNA, Neoplasm/genetics , DNA, Neoplasm/metabolism , DNA-Binding Proteins/genetics , Dioxygenases/genetics , Dioxygenases/metabolism , Down-Regulation/genetics , Female , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Mixed Function Oxygenases , Neoplasm Invasiveness , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins/genetics , Tissue Inhibitor of Metalloproteinase-2/genetics , Tissue Inhibitor of Metalloproteinase-3/genetics , Tumor Suppressor Proteins/genetics
6.
PLoS One ; 7(4): e35128, 2012.
Article in English | MEDLINE | ID: mdl-22514714

ABSTRACT

Microenvironment plays an important role in cancer development. We have reported that the cancer-associated stromal cells exhibit phenotypic and functional changes compared to stromal cells neighboring to normal tissues. However, the molecular mechanisms as well as the maintenance of these changes remain elusive. Here we showed that through co-culture with breast cancer cells for at least three to four passages, breast normal tissue-associated fibroblasts (NAFs) gained persistent activity for promoting cancer cell invasion, partly via up-regulating ADAM metallopeptidase with thrombospondin type 1 motif, 1 (ADAMTS1). Furthermore, we demonstrated that the DNA methylation pattern in the ADAMTS1 promoter has no alteration. Instead, the loss of EZH2 binding to the ADAMTS1 promoter and the resulting decrease of promoter-associated histone H3K27 methylation may account for the up-regulation of ADAMTS1. Importantly, the lack of EZH2 binding and the H3K27 methylation on the ADAMTS1 promoter were sustained in cancer cell-precocultured NAFs after removal of cancer cells. These results suggest that cancer cells are capable of inducing stromal fibroblasts to secrete ADAMTS1 persistently for their invasion and the effect is epigenetically inheritable.


Subject(s)
ADAM Proteins/metabolism , Epigenesis, Genetic/genetics , Fibroblasts/metabolism , ADAM Proteins/genetics , ADAMTS1 Protein , Blotting, Western , Cell Line, Tumor , Cells, Cultured , Chromatin Immunoprecipitation , Coculture Techniques , Enzyme-Linked Immunosorbent Assay , Female , Humans , Immunoprecipitation , Neoplasm Metastasis/genetics , Proteoglycans/genetics , Proteoglycans/metabolism , Real-Time Polymerase Chain Reaction , Vesicular Transport Proteins/genetics , Vesicular Transport Proteins/metabolism
7.
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
8.
Cell Res ; 21(8): 1230-47, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21445097

ABSTRACT

Chromatin assembly factor 1 (CAF1) consisting of p150, p60 and p48 is known to assemble histones onto newly synthesized DNA and thus maintain the chromatin structure. Here, we show that CAF1 expression was induced in human cytomegalovirus (HCMV)-infected cells, concomitantly with global chromatin decondensation. This apparent conflict was thought to result, in part, from CAF1 mislocalization to compartments of HCMV DNA synthesis through binding of its largest subunit p150 to viral immediate-early protein 2 (IE2). p150 interaction with p60 and IE2 facilitated HCMV DNA synthesis. The IE2Q548R mutation, previously reported to result in impaired HCMV growth with unknown mechanism, disrupted IE2/p150 and IE2/histones association in our study. Moreover, IE2 interaction with histones partly depends on p150, and the HCMV-induced chromatin decondensation was reduced in cells ectopically expressing the p150 mutant defective in IE2 binding. These results not only indicate that CAF1 was hijacked by IE2 to facilitate the replication of the HCMV genome, suggesting chromatin assembly plays an important role in herpesviral DNA synthesis, but also provide a model of the virus-induced chromatin instability through CAF1.


Subject(s)
Chromatin Assembly Factor-1/metabolism , Cytomegalovirus/metabolism , Host-Pathogen Interactions , Immediate-Early Proteins/metabolism , Trans-Activators/metabolism , Amino Acid Substitution , Cell Line , Chromatin/metabolism , Chromatin Assembly Factor-1/analysis , Chromatin Assembly and Disassembly , DNA, Viral/metabolism , Histones/metabolism , Humans , Immediate-Early Proteins/analysis , Immediate-Early Proteins/genetics , Protein Binding , Protein Subunits/analysis , Protein Subunits/metabolism , Trans-Activators/analysis , Trans-Activators/genetics , Virus Replication
9.
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
10.
J Clin Invest ; 120(8): 2920-30, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20592467

ABSTRACT

Hypermethylation-mediated tumor suppressor gene silencing plays a crucial role in tumorigenesis. Understanding its underlying mechanism is essential for cancer treatment. Previous studies on human N-alpha-acetyltransferase 10, NatA catalytic subunit (hNaa10p; also known as human arrest-defective 1 [hARD1]), have generated conflicting results with regard to its role in tumorigenesis. Here we provide multiple lines of evidence indicating that it is oncogenic. We have shown that hNaa10p overexpression correlated with poor survival of human lung cancer patients. In vitro, enforced expression of hNaa10p was sufficient to cause cellular transformation, and siRNA-mediated depletion of hNaa10p impaired cancer cell proliferation in colony assays and xenograft studies. The oncogenic potential of hNaa10p depended on its interaction with DNA methyltransferase 1 (DNMT1). Mechanistically, hNaa10p positively regulated DNMT1 enzymatic activity by facilitating its binding to DNA in vitro and its recruitment to promoters of tumor suppressor genes, such as E-cadherin, in vivo. Consistent with this, interaction between hNaa10p and DNMT1 was required for E-cadherin silencing through promoter CpG methylation, and E-cadherin repression contributed to the oncogenic effects of hNaa10p. Together, our data not only establish hNaa10p as an oncoprotein, but also reveal that it contributes to oncogenesis through modulation of DNMT1 function.


Subject(s)
Acetyltransferases/physiology , DNA (Cytosine-5-)-Methyltransferases/physiology , Gene Silencing , Genes, Tumor Suppressor , Lung Neoplasms/etiology , Acetyltransferases/genetics , Animals , Cadherins/genetics , DNA (Cytosine-5-)-Methyltransferase 1 , DNA Methylation , Humans , Male , Mice , N-Terminal Acetyltransferase A , N-Terminal Acetyltransferase E , NIH 3T3 Cells , Promoter Regions, Genetic , RNA, Messenger/analysis
11.
Cancer Res ; 70(14): 5807-17, 2010 Jul 15.
Article in English | MEDLINE | ID: mdl-20570896

ABSTRACT

Overexpression of DNA 5'-cytosine-methyltransferases (DNMT), which are enzymes that methylate the cytosine residue of CpGs, is involved in many cancers. However, the mechanism of DNMT overexpression remains unclear. Here, we showed that wild-type p53 negatively regulated DNMT1 expression by forming a complex with specificity protein 1 (Sp1) protein and chromatin modifiers on the DNMT1 promoter. However, the stoichiometry between p53 and Sp1 determined whether Sp1 acts as a transcription activator or corepressor. Low level of exogenous Sp1 enhanced the repressive activity of endogenous p53 on the DNMT1 promoter whereas high level of Sp1 upregulated DNMT1 gene expression level in A549 (p53 wild-type) cells. In H1299 (p53 null) cells, exogenous Sp1 induced DNMT1 expression in a dose-dependent manner. We also discovered a new mechanism whereby high level of Sp1, via its COOH-terminal domain, induced interaction between p53 and MDM2, resulting in degradation of p53 by MDM2-mediated ubiquitination. Clinical data from 102 lung cancer patients indicated that overexpression of DNMT1 was associated with p53 mutation (P = 0.014) and high expression of Sp1 protein (P = 0.006). In addition, patients with overexpression of both DNMT1 and Sp1 proteins showed poor prognosis (P = 0.037). Our cell and clinical data provided compelling evidence that deregulation of DNMT1 is associated with gain of transcriptional activation of Sp1 and/or loss of repression of p53. DNMT1 overexpression results in epigenetic alteration of multiple tumor suppressor genes and ultimately leads to lung tumorigenesis and poor prognosis.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/biosynthesis , Lung Neoplasms/enzymology , Sp1 Transcription Factor/metabolism , Tumor Suppressor Protein p53/metabolism , 5-Methylcytosine/metabolism , Binding Sites , Cell Line, Tumor , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/genetics , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Humans , Immunoprecipitation , Lung Neoplasms/genetics , Point Mutation , Promoter Regions, Genetic , Proto-Oncogene Proteins c-mdm2/metabolism , Sp1 Transcription Factor/genetics , Transcription, Genetic , Tumor Suppressor Protein p53/genetics , Ubiquitination
12.
J Biol Chem ; 284(21): 14040-9, 2009 May 22.
Article in English | MEDLINE | ID: mdl-19324875

ABSTRACT

Chromatin assembly factor 1 contains three subunits, p150, p60, and p48. It is essential for coupling nucleosome assembly to newly synthesized DNA. Whether chromatin assembly factor 1 subunits have functions beyond escorting histones, which depends on the complex formation of p150 and p60, has been an issue of great interest. This study reveals a novel role of p150, but not p60, in gene-specific transcriptional activation. We found that p150 transcriptionally activated an essential viral promoter, the major immediate early promoter (MIEP) of the human cytomegalovirus, independently of p60. Knocking down p150 decreased the MIEP function in both transfected and virally infected cells. The chromatin immunoprecipitation analysis and the in vitro protein-DNA binding assay demonstrated that p150 used its KER domain to associate with the MIEP from -593 to -574 bp. The N-terminal 244 residues were also found essential for p150-mediated MIEP activation, likely through recruiting the acetyltransferase p300 to acetylate local histones. Domain swapping experiments further showed that the KER and the N terminus of p150 acted as an independent DNA binding and transcriptional activation domain, respectively. Because p60 did not seem involved in the reaction, together these results indicate for the first time that p150 directly activates transcription, independently of its histone deposition function.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , DNA-Binding Proteins/metabolism , Immediate-Early Proteins/genetics , Protein Subunits/metabolism , Transcriptional Activation/genetics , Base Pairing , Cell Line, Tumor , Chromatin Assembly Factor-1 , Chromosomal Proteins, Non-Histone/chemistry , Cytomegalovirus/genetics , DNA-Binding Proteins/chemistry , Gene Expression Regulation, Viral , Humans , Promoter Regions, Genetic/genetics , Protein Binding , Protein Structure, Tertiary , Transcription Factors , Transfection , p300-CBP Transcription Factors/metabolism
13.
Nat Struct Mol Biol ; 15(4): 419-21, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18270511

ABSTRACT

The histone H3 lysine 4 demethylase RBP2 contains a DNA binding domain, the AT-rich interaction domain (ARID). We solved the structure of ARID by NMR, identified its DNA binding motif (CCGCCC) and characterized the binding contacts. Immunofluorescence and luciferase assays indicated that ARID is required for RBP2 demethylase activity in cells and that DNA recognition is essential to regulate transcription.


Subject(s)
DNA/metabolism , Retinol-Binding Proteins, Cellular/metabolism , Fluorescent Antibody Technique , Humans , Nuclear Magnetic Resonance, Biomolecular , Promoter Regions, Genetic , Protein Binding , Protein Serine-Threonine Kinases/genetics , Retinol-Binding Proteins, Cellular/chemistry , Structure-Activity Relationship , Transcription Factors
14.
J Immunol ; 177(9): 6199-206, 2006 Nov 01.
Article in English | MEDLINE | ID: mdl-17056549

ABSTRACT

Human CMV (HCMV) is a widespread human pathogen that causes blindness by inducing retinitis in AIDS patients. Previously, we showed that viral immediate early 2 (IE2) protein may allow HCMV to evade the immune control by killing the Fas receptor-positive T lymphocytes attracted to the infected retina with increased secretion of Fas ligand (FasL). In this study, we further demonstrate that the secreted FasL also kills uninfected Fas-rich bystander retinal cells and that IE2 simultaneously protects the infected cells from undergoing apoptotic death, in part, by activating the expression of cellular FLIP (c-FLIP), an antiapoptotic molecule that blocks the direct downstream executer caspase 8 of the FasL/Fas pathway. c-FLIP induction requires the N-terminal 98 residues of IE2 and the c-FLIP promoter region spanning nucleotides -978 to -696. In vivo association of IE2 to this region, IE2-specific c-FLIP activation, and decrease of FasL-up-regulated activities of caspases 8 and 3 were all demonstrated in HCMV-infected human retinal cells. Moreover, c-FLIP up-regulation by IE2 appeared to involve PI3K and might also render cells resistant to TRAIL-mediated death. Finally, enhanced c-FLIP signals were immunohistochemically detected in IE-positive cells in the HCMV-infected lesions of the human retina. Taken together, these data demonstrate specific activation of c-FLIP by HCMV IE2 and indicate a novel role for c-FLIP in the pathogenesis of HCMV retinitis.


Subject(s)
Apoptosis/genetics , CASP8 and FADD-Like Apoptosis Regulating Protein/genetics , Cytomegalovirus Retinitis/genetics , Immediate-Early Proteins/metabolism , Trans-Activators/metabolism , Transcriptional Activation , CASP8 and FADD-Like Apoptosis Regulating Protein/analysis , Cells, Cultured , Cytomegalovirus Retinitis/metabolism , Fas Ligand Protein/metabolism , Humans , Immediate-Early Proteins/analysis , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Promoter Regions, Genetic , Retina/chemistry , Retina/metabolism , Retina/virology , Sequence Deletion , Trans-Activators/analysis , Up-Regulation
15.
Mol Cell Biol ; 25(23): 10442-53, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16287857

ABSTRACT

Steroidogenic factor 1 (SF-1) is a nuclear receptor essential for steroidogenic gene expression, but how its activity is regulated is unclear. Here we demonstrate that p300 plays an important role in regulating SF-1 function. SF-1 was acetylated in vitro and in vivo by p300 at the KQQKK motif in the Ftz-F1 (Fushi-tarazu factor 1) box adjacent to its DNA-binding domain. Mutation of the KQQKK motif reduced the DNA-binding activity and p300-dependent activation of SF-1. When stimulated with cyclic AMP (cAMP), adrenocortical Y1 cells expressed more p300, leading to additional SF-1 association with p300 and increased SF-1 acetylation and DNA binding. It also increased SF-1 colocalization with p300 in nuclear foci. Collectively, these results indicate that SF-1 transcriptional activity is regulated by p300 in response to the cAMP signaling pathway by way of increased acetylation, DNA binding, and recruitment to nuclear foci.


Subject(s)
Cyclic AMP/metabolism , DNA/metabolism , Homeodomain Proteins/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Transcription Factors/metabolism , p300-CBP Transcription Factors/metabolism , Acetylation , Amino Acid Sequence , Animals , Cell Line , Cell Nucleus/genetics , Cell Nucleus/metabolism , Homeodomain Proteins/chemistry , Homeodomain Proteins/genetics , Humans , Mice , Molecular Sequence Data , Mutation/genetics , Protein Binding , Protein Transport , Receptors, Cytoplasmic and Nuclear/chemistry , Receptors, Cytoplasmic and Nuclear/genetics , Sequence Alignment , Sequence Homology, Amino Acid , Steroidogenic Factor 1 , Transcription Factors/chemistry , Transcription Factors/genetics , p300-CBP Transcription Factors/genetics
16.
EMBO J ; 23(11): 2269-80, 2004 Jun 02.
Article in English | MEDLINE | ID: mdl-15141169

ABSTRACT

Targeting of cellular histone acetyltransferases (HATs) by viral proteins is important in the development of virus-associated diseases. The immediate-early 2 protein (IE2) of human cytomegalovirus (HCMV) binds to the tumor suppressor, p53, and inactivates its functions by unknown mechanisms. Here, we show that IE2 binds to the HAT domain of the p53 coactivators, p300 and CREB-binding protein (CBP), and blocks their acetyltransferase activity on both histones and p53. The minimal HAT inactivation region on IE2 involves the N-terminal 98 amino acids. The in vivo DNA binding of p53 and local histone acetylation on p53-dependent promoters are all reduced by IE2, but not by mutant IE2 proteins that lack the HAT inhibition region. Furthermore, the p53 acetylation site mutant, K320/373/382R, retains both DNA binding and promoter transactivation activity in vivo and these effects are repressed by IE2 as well. Together with the finding that only wild-type IE2 exerts an antiapoptotic effect, our results suggest that HCMV IE2 downregulates p53-dependent gene activation by inhibiting p300/CBP-mediated local histone acetylation and that IE2 may have oncogenic activity.


Subject(s)
Cytomegalovirus/metabolism , Immediate-Early Proteins/metabolism , Tumor Suppressor Protein p53/metabolism , Acetyltransferases , Amino Acid Sequence , Annexin A5/metabolism , Apoptosis , Blotting, Western , Cell Line, Tumor , Chromatin Immunoprecipitation , Cyclic AMP Response Element-Binding Protein/chemistry , Cyclic AMP Response Element-Binding Protein/metabolism , Cytomegalovirus/genetics , Histone Acetyltransferases , Humans , Immediate-Early Proteins/chemistry , Immediate-Early Proteins/genetics , Lung Neoplasms/pathology , Mutagenesis, Site-Directed , Mutation , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Plasmids , Precipitin Tests , Promoter Regions, Genetic , Protein Binding , Protein Structure, Tertiary , Trans-Activators/chemistry , Trans-Activators/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptional Activation
17.
Virology ; 319(1): 60-70, 2004 Feb 05.
Article in English | MEDLINE | ID: mdl-14967488

ABSTRACT

Hepatitis delta virus (HDV) is a single-stranded RNA virus that encodes two viral nucleocapsid proteins named small and large form hepatitis delta antigen (S-HDAg and L-HDAg). The S-HDAg is essential for viral RNA replication while the L-HDAg is required for viral assembly. In this study, we demonstrated that HDAg are acetylated proteins. Metabolic labeling with [(3)H]acetate revealed that both forms of HDAg could be acetylated in vivo. The histone acetyltransferase (HAT) domain of cellular acetyltransferase p300 could acetylate the full-length and the N-terminal 88 amino acids of S-HDAg in vitro. By mass spectrometric analysis of the modified protein, Lys-72 of S-HDAg was identified as one of the acetylation sites. Substitution of Lys-72 to Arg caused the mutant S-HDAg to redistribute from the nucleus to the cytoplasm. The mutant reduced viral RNA accumulation and resulted in the earlier appearance of L-HDAg. These results demonstrated that HDAg is an acetylated protein and mutation of HDAg at Lys-72 modulates HDAg subcellular localization and may participate in viral RNA nucleocytoplasmic shuttling and replication.


Subject(s)
Hepatitis delta Antigens/chemistry , Hepatitis delta Antigens/metabolism , Lysine/metabolism , Acetylation , Acetyltransferases/metabolism , Cell Cycle Proteins/metabolism , Cell Line , Cell Nucleus/virology , Cytoplasm/virology , Hepatitis Delta Virus/immunology , Hepatitis Delta Virus/physiology , Hepatitis delta Antigens/analysis , Hepatitis delta Antigens/genetics , Histone Acetyltransferases , Humans , Lysine/genetics , Mutation , RNA, Viral/biosynthesis , Transcription Factors , Virus Replication , p300-CBP Transcription Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...