Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters










Publication year range
1.
Cell Death Dis ; 6: e1876, 2015 Sep 03.
Article in English | MEDLINE | ID: mdl-26335716

ABSTRACT

Autophagy, an important degradation system involved in maintaining cellular homeostasis, serves also to eliminate pathogens and process their fragments for presentation to the immune system. Several viruses have been shown to interact with the host autophagic machinery to suppress or make use of this cellular catabolic pathway to enhance their survival and replication. Epstein Barr virus (EBV) is a γ-herpes virus associated with a number of malignancies of epithelial and lymphoid origin in which establishes a predominantly latent infection. Latent EBV can periodically reactivate to produce infectious particles that allow the virus to spread and can lead to the death of the infected cell. In this study, we analyzed the relationship between autophagy and EBV reactivation in Burkitt's lymphoma cells. By monitoring autophagy markers and EBV lytic genes expression, we demonstrate that autophagy is enhanced in the early phases of EBV lytic activation but decreases thereafter concomitantly with increased levels of EBV lytic proteins. In a cell line defective for late antigens expression, we found an inverse correlation between EBV early antigens expression and autophagosomes formation, suggesting that early after activation, the virus is able to suppress autophagy. We report here for the first time that inhibition of autophagy by Bafilomycin A1 or shRNA knockdown of Beclin1 gene, highly incremented EBV lytic genes expression as well as intracellular viral DNA and viral progeny yield. Taken together, these findings indicate that EBV activation induces the autophagic response, which is soon inhibited by the expression of EBV early lytic products. Moreover, our findings open the possibility that pharmacological inhibitors of autophagy may be used to enhance oncolytic viral therapy of EBV-related lymphomas.


Subject(s)
Autophagy/genetics , Burkitt Lymphoma/genetics , Virus Replication/genetics , Cell Line, Tumor , DNA Replication , Gene Expression , Humans , Immunoblotting
2.
J Virol ; 88(9): 4721-35, 2014 May.
Article in English | MEDLINE | ID: mdl-24522914

ABSTRACT

UNLABELLED: The impact of Epstein-Barr virus (EBV) on human health is substantial, but vaccines that prevent primary EBV infections or treat EBV-associated diseases are not yet available. The Epstein-Barr nuclear antigen 1 (EBNA-1) is an important target for vaccination because it is the only protein expressed in all EBV-associated malignancies. We have designed and tested two therapeutic EBV vaccines that target the rhesus (rh) lymphocryptovirus (LCV) EBNA-1 to determine if ongoing T cell responses during persistent rhLCV infection in rhesus macaques can be expanded upon vaccination. Vaccines were based on two serotypes of E1-deleted simian adenovirus and were administered in a prime-boost regimen. To further modulate the response, rhEBNA-1 was fused to herpes simplex virus glycoprotein D (HSV-gD), which acts to block an inhibitory signaling pathway during T cell activation. We found that vaccines expressing rhEBNA-1 with or without functional HSV-gD led to expansion of rhEBNA-1-specific CD8(+) and CD4(+) T cells in 33% and 83% of the vaccinated animals, respectively. Additional animals developed significant changes within T cell subsets without changes in total numbers. Vaccination did not increase T cell responses to rhBZLF-1, an immediate early lytic phase antigen of rhLCV, thus indicating that increases of rhEBNA-1-specific responses were a direct result of vaccination. Vaccine-induced rhEBNA-1-specific T cells were highly functional and produced various combinations of cytokines as well as the cytolytic molecule granzyme B. These results serve as an important proof of principle that functional EBNA-1-specific T cells can be expanded by vaccination. IMPORTANCE: EBV is a common human pathogen that establishes a persistent infection through latency in B cells, where it occasionally reactivates. EBV infection is typically benign and is well controlled by the host adaptive immune system; however, it is considered carcinogenic due to its strong association with lymphoid and epithelial cell malignancies. Latent EBNA-1 is a promising target for a therapeutic vaccine, as it is the only antigen expressed in all EBV-associated malignancies. The goal was to determine if rhEBNA-1-specific T cells could be expanded upon vaccination of infected animals. Results were obtained with vaccines that target EBNA-1 of rhLCV, a virus closely related to EBV. We found that vaccination led to expansion of rhEBNA-1 immune cells that exhibited functions fit for controlling viral infection. This confirms that rhEBNA-1 is a suitable target for therapeutic vaccines. Future work should aim to generate more-robust T cell responses through modified vaccines.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Herpesviridae Infections/veterinary , Herpesvirus Vaccines/immunology , Lymphocryptovirus/immunology , Viral Proteins/immunology , Adenoviruses, Simian/genetics , Animals , Drug Carriers , Female , Genetic Vectors , Herpesviridae Infections/immunology , Herpesvirus Vaccines/administration & dosage , Herpesvirus Vaccines/genetics , Lymphocryptovirus/genetics , Macaca mulatta , Vaccination/methods , Viral Proteins/genetics
3.
J Virol ; 87(15): 8351-62, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23698300

ABSTRACT

Epstein-Barr virus (EBV) infection leads to lifelong viral persistence through its latency in B cells. EBV-specific T cells control reactivations and prevent the development of EBV-associated malignancies in most healthy carriers, but infection can sometimes cause chronic disease and malignant transformation. Epstein-Barr nuclear antigen 1 (EBNA-1) is the only viral protein consistently expressed during all forms of latency and in all EBV-associated malignancies and is a promising target for a therapeutic vaccine. Here, we studied the EBNA-1-specific immune response using the EBV-homologous rhesus lymphocryptovirus (rhLCV) infection in rhesus macaques. We assessed the frequency, phenotype, and cytokine production profiles of rhLCV EBNA-1 (rhEBNA-1)-specific T cells in 15 rhesus macaques and compared them to the lytic antigen of rhLCV BZLF-1 (rhBZLF-1). We were able to detect rhEBNA-1-specific CD4(+) and/or CD8(+) T cells in 14 of the 15 animals screened. In comparison, all 15 animals had detectable rhBZLF-1 responses. Most peptide-specific CD4(+) T cells exhibited a resting phenotype of central memory (TCM), while peptide-specific CD8(+) T cells showed a more activated phenotype, belonging mainly to the effector cell subset. By comparing our results to the human EBV immune response, we demonstrate that the rhLCV model is a valid system for studying chronic EBV infection and for the preclinical development of therapeutic vaccines.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Epstein-Barr Virus Nuclear Antigens/immunology , Herpesviridae Infections/immunology , Lymphocryptovirus/immunology , Trans-Activators/immunology , Tumor Virus Infections/immunology , Animals , Cytokines/metabolism , Disease Models, Animal , Female , Herpesviridae Infections/virology , Macaca mulatta , Tumor Virus Infections/virology
4.
J Virol ; 75(21): 10334-47, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11581402

ABSTRACT

Epstein-Barr virus (EBV) lytic cycle transcription and DNA replication require the transcriptional activation function of the viral immediate-early protein Zta. We describe a series of alanine substitution mutations in the Zta activation domain that reveal two functional motifs based on amino acid composition. Alanine substitution of single or paired hydrophobic aromatic amino acid residues resulted in modest transcription activation defects, while combining four substitutions of aromatic residues (F22/F26/W74/F75) led to more severe transcription defects. Substitution of acidic amino acid residue E27, D35, or E54 caused severe transcription defects on most viral promoters. Promoter- and cell-specific defects were observed for some substitution mutants. Aromatic residues were required for Zta interaction with TFIIA-TFIID and the CREB-binding protein (CBP) and for stimulation of CBP histone acetyltransferase activity in vitro. In contrast, acidic amino acid substitution mutants interacted with TFIIA-TFIID and CBP indistinguishably from the wild type. The nuclear domain 10 (ND10) protein SP100 was dispersed by most Zta mutants, but acidic residue mutations led to reduced, while aromatic substitution mutants led to increased SP100 nuclear staining. Acidic residue substitution mutants had more pronounced defects in transcription activation of endogenous viral genes in latently infected cells and for viral replication, as measured by the production of infectious virus. One mutant, K12/F13, was incapable of stimulating EBV lytic replication but had only modest transcription defects. These results indicate that Zta stimulates viral reactivation through two nonredundant structural motifs, one of which interacts with general transcription factors and coactivators, and the other has an essential but as yet not understood function in lytic transcription.


Subject(s)
DNA-Binding Proteins/chemistry , Herpesvirus 4, Human/physiology , Saccharomyces cerevisiae Proteins , Trans-Activators/chemistry , Viral Proteins , Virus Activation , Acetyltransferases/metabolism , CREB-Binding Protein , DNA Replication , DNA-Binding Proteins/genetics , DNA-Binding Proteins/physiology , Histone Acetyltransferases , Nuclear Proteins/metabolism , Promoter Regions, Genetic , Trans-Activators/genetics , Trans-Activators/metabolism , Trans-Activators/physiology , Transcription Factor TFIIA , Transcription Factor TFIID , Transcription Factors/genetics , Transcription Factors, TFII/genetics , Transcriptional Activation , Virus Replication
5.
J Biol Chem ; 276(19): 15886-92, 2001 May 11.
Article in English | MEDLINE | ID: mdl-11278496

ABSTRACT

Transcription factor IIA (TFIIA) is a positive acting general factor that contacts the TATA-binding protein (TBP) and mediates an activator-induced conformational change in the transcription factor IID (TFIID) complex. Previously, we have found that phosphorylation of yeast TFIIA stimulates TFIIA.TBP.TATA complex formation and transcription activation in vivo. We now show that human TFIIA is phosphorylated in vivo on serine residues that are partially conserved between yeast and human TFIIA large subunits. Alanine substitution mutation of serine residues 316 and 321 in TFIIA alphabeta reduced TFIIA phosphorylation significantly in vivo. Additional alanine substitutions at serines 280 and 281 reduced phosphorylation to undetectable levels. Mutation of all four serine residues reduced the ability of TFIIA to stimulate transcription in transient transfection assays with various activators and promoters, indicating that TFIIA phosphorylation is required globally for optimal function. In vitro, holo-TFIID and TBP-associated factor 250 (TAF(II)250) phosphorylated TFIIA on the beta subunit. Mutation of the four serines required for in vivo phosphorylation eliminated TFIID and TAF(II)250 phosphorylation in vitro. The NH(2)-terminal kinase domain of TAF(II)250 was sufficient for TFIIA phosphorylation, and this activity was inhibited by full-length retinoblastoma protein but not by a retinoblastoma protein mutant defective for TAF(II)250 interaction or tumor suppressor activity. TFIIA phosphorylation had little effect on the TFIIA.TBP.TATA complex in electrophoretic mobility shift assay. However, phosphorylation of TFIIA containing a gamma subunit Y65A mutation strongly stimulated TFIIA.TBP.TATA complex formation. TFIIA-gammaY65A is defective for binding to the beta-sheet domain of TBP identified in the crystal structure. These results suggest that TFIIA phosphorylation is important for strengthening the TFIIA.TBP contact or creating a second contact between TFIIA and TBP that was not visible in the crystal structure.


Subject(s)
DNA-Binding Proteins/metabolism , Nuclear Proteins/metabolism , Serine , TATA-Binding Protein Associated Factors , Trans-Activators/metabolism , Transcription Factors/metabolism , Amino Acid Sequence , Cell Line , Histone Acetyltransferases , Humans , Molecular Sequence Data , Mutagenesis, Site-Directed , Phosphorylation , Protein Subunits , Recombinant Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , TATA-Box Binding Protein , Transcription Factor TFIIA , Transcription Factor TFIID , Transcription Factors/chemistry , Transcription Factors/genetics , Transcription Factors, TFII/metabolism , Transcription, Genetic , Transfection
6.
Mol Cell Biol ; 21(2): 476-87, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11134336

ABSTRACT

The transcriptional coactivator CREB binding protein (CBP) possesses intrinsic histone acetyltransferase (HAT) activity that is important for gene regulation. CBP binds to and cooperates with numerous nuclear factors to stimulate transcription, but it is unclear if these factors modulate CBP HAT activity. Our previous work showed that CBP interacts with the Epstein-Barr virus-encoded basic region zipper (b-zip) protein, Zta, and augments its transcriptional activity. Here we report that Zta strongly enhances CBP-mediated acetylation of nucleosomal histones. Zta stimulated the HAT activity of CBP that had been partially purified or immunoprecipitated from mammalian cells as well as from affinity-purified, baculovirus expressed CBP. Stimulation of nucleosome acetylation required the CBP HAT domain, the Zta DNA binding and transcription activation domain, and nucleosomal DNA. In addition to Zta, we found that two other b-zip proteins, NF-E2 and C/EBPalpha, strongly stimulated nucleosomal HAT activity. In contrast, several CBP-binding proteins, including phospho-CREB, JUN/FOS, GATA-1, Pit-1, and EKLF, failed to stimulate HAT activity. These results demonstrate that a subset of transcriptional activators enhance the nucleosome-directed HAT activity of CBP and suggest that nuclear factors may regulate transcription by altering substrate recognition and/or the enzymatic activity of chromatin modifying coactivators.


Subject(s)
Acetyltransferases/metabolism , DNA-Binding Proteins/metabolism , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Nucleosomes/enzymology , Saccharomyces cerevisiae Proteins , Trans-Activators/chemistry , Trans-Activators/metabolism , Viral Proteins , Acetylation , Acetyltransferases/chemistry , Animals , Binding Sites , CCAAT-Enhancer-Binding Protein-alpha/metabolism , CREB-Binding Protein , Catalytic Domain , Cell Line , DNA/chemistry , DNA/genetics , DNA/metabolism , DNA-Binding Proteins/chemistry , Enzyme Activation , Erythroid-Specific DNA-Binding Factors , G-Box Binding Factors , GATA1 Transcription Factor , Histone Acetyltransferases , Histones/metabolism , Humans , NF-E2 Transcription Factor , NF-E2 Transcription Factor, p45 Subunit , Nucleosomes/genetics , Nucleosomes/metabolism , Protein Structure, Tertiary , Substrate Specificity , Transcription Factors/metabolism , Transcriptional Activation , Transfection
7.
J Virol ; 74(24): 11800-10, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11090180

ABSTRACT

Nuclear domains called ND10 (nuclear domain 10) are discrete nuclear protein aggregations characterized by a set of interferon-upregulated proteins including Sp100 and PML, where papova-, adeno-, and herpesviruses begin their transcription and DNA replication. Both the alpha- and betaherpesvirus subfamilies disrupt ND10 upon infection by dispersing and/or destroying ND10-associated proteins. We studied the effect of the gammaherpesvirus Epstein-Barr virus (EBV) on ND10 and its spatial distribution in the nucleus of cells during latency and lytic reactivation. In latently infected Burkitt's lymphoma, lymphoblastoid, and D98/HR1 cells, ND10 were intact, as judged by immunofluorescence localization of PML, Sp100, NDP55, and Daxx. Fluorescent in situ hybridization revealed no association between viral episomes and ND10 during latency, implying that the maintenance replication of EBV, which depends on host cell proliferation, occurs independent of ND10. As in mitosis, the EBV genomes were attached to interphase chromosomes, suggesting that they are unable to move freely within the interchromosomal space and thus unable to associate with the interchromosomally located ND10 or other nuclear domains. Upon lytic activation, ND10 became dispersed in cells expressing lytic proteins. Redistribution of ND10 proteins occurred sequentially at different stages of the lytic cycle, with Sp100, Daxx, and NDP55 dispersed before and PML dispersed after the onset of lytic replication. ND10 remnants were retained until the early stages of lytic replication, and replicating EBV genomes were frequently found beside this nuclear domain; the number of replication domains was usually lower than the average latent virus frequency. Thus, latency does not require or induce interaction of EBV with ND10 for transcription and replication, whereas lytic replication triggers dispersion of ND10 proteins and occurs in close association with PML aggregates. The required movement of chromosome-attached latent EBV episomes to ND10 after reactivation from latency might include physical release of the chromosome-bound episomes. Only episomes contacting ND10 after such a release might be able to begin lytic replication.


Subject(s)
Herpesvirus 4, Human/physiology , Neoplasm Proteins/physiology , Nuclear Proteins/physiology , Transcription Factors/physiology , Virus Replication , Cell Line , Humans , Promyelocytic Leukemia Protein , Tumor Suppressor Proteins , Virus Latency
8.
J Biol Chem ; 275(1): 122-8, 2000 Jan 07.
Article in English | MEDLINE | ID: mdl-10617594

ABSTRACT

The general transcription factor IIA (TFIIA) stimulates RNA polymerase II-specific transcription by stabilizing the association of the TATA-binding protein (TBP) with promoter DNA, inhibiting repressors of TBP, and facilitating activator-dependent conformational changes in the preinitiation complex. TFIIA is encoded by two genes (alphabeta and gamma) that are highly conserved between human and yeast. Here, we report the molecular cloning of a novel human gene that shares significant sequence similarity to the evolutionarily conserved amino- and carboxyl-terminal domains of TFIIAalphabeta. The TFIIA-related protein (TFIIAtau) was cloned from a testis-specific cDNA library, and its mRNA is expressed predominantly in testis tissue as determined by expressed sequence tag data base analysis and Northern blotting analysis. The TFIIA complex reconstituted with the testis-specific subunit, TFIIA (tau+gamma), formed the TFIIA-TBP-TATA DNA (T-A) and TFIIA-TFIIB-TBP-TATA DNA (TAB) complexes indistinguishably from TFIIA (alphabeta+gamma). TFIIA (tau+gamma) supported basal and activated transcription for most activators in reactions reconstituted with TFIIA-depleted nuclear extracts. However, TFIIA (tau+gamma) was reduced relative to TFIIA (alphabeta+gamma) for stimulating transcription with at least one activator, suggesting that these two forms of TFIIA have activator specificity. These results suggest that TFIIAtau may be important for testis-specific transcription regulation.


Subject(s)
DNA-Binding Proteins/metabolism , Testis/metabolism , Transcription Factors/metabolism , Transcriptional Activation , Amino Acid Sequence , Chromosome Mapping , Chromosomes, Human, Pair 2 , DNA, Complementary/genetics , Databases, Factual , Expressed Sequence Tags , Humans , Male , Molecular Sequence Data , Protein Binding , RNA, Messenger/isolation & purification , Sequence Homology, Amino Acid , TATA-Box Binding Protein , Tissue Distribution , Transcription Factor TFIIA , Transcription Factors/genetics
9.
Mol Cell Biol ; 20(2): 634-47, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10611242

ABSTRACT

SAGA is a 1.8-MDa yeast protein complex that is composed of several distinct classes of transcription-related factors, including the adaptor/acetyltransferase Gcn5, Spt proteins, and a subset of TBP-associated factors. Our results indicate that mutations that completely disrupt SAGA (deletions of SPT7 or SPT20) strongly reduce transcriptional activation at the HIS3 and TRP3 genes and that Gcn5 is required for normal HIS3 transcriptional start site selection. Surprisingly, mutations in Spt proteins involved in the SAGA-TBP interaction (Spt3 and Spt8) cause derepression of HIS3 and TRP3 transcription in the uninduced state. Consistent with this finding, wild-type SAGA inhibits TBP binding to the HIS3 promoter in vitro, while SAGA lacking Spt3 or Spt8 is not inhibitory. We detected two distinct forms of SAGA in cell extracts and, strikingly, one lacks Spt8. Conditions that induce HIS3 and TRP3 transcription result in an altered balance between these complexes strongly in favor of the form without Spt8. These results suggest that the composition of SAGA may be dynamic in vivo and may be regulated through dissociable inhibitory subunits.


Subject(s)
DNA-Binding Proteins/antagonists & inhibitors , Fungal Proteins/metabolism , Multienzyme Complexes/metabolism , Promoter Regions, Genetic/genetics , Protein Kinases/metabolism , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae/genetics , Transcription Factors/antagonists & inhibitors , Acetyltransferases/metabolism , Anthranilate Synthase/genetics , DNA Footprinting , DNA, Fungal/genetics , DNA, Fungal/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Fungal Proteins/genetics , Gene Deletion , Gene Expression Regulation, Fungal , Genes, Fungal/genetics , Genes, Fungal/physiology , Histone Acetyltransferases , Hydro-Lyases/genetics , Indole-3-Glycerol-Phosphate Synthase/genetics , Models, Genetic , Multienzyme Complexes/chemistry , Multienzyme Complexes/genetics , Protein Kinases/genetics , Repressor Proteins/metabolism , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/growth & development , TATA Box/genetics , TATA-Box Binding Protein , Trans-Activators/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic/genetics
10.
Mol Cell Biol ; 19(11): 7610-20, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10523649

ABSTRACT

The TATA binding protein (TBP) plays a central role in eukaryotic and archael transcription initiation. We describe the isolation of a novel 23-kDa human protein that displays 41% identity to TBP and is expressed in most human tissue. Recombinant TBP-related protein (TRP) displayed barely detectable binding to consensus TATA box sequences but bound with slightly higher affinities to nonconsensus TATA sequences. TRP did not substitute for TBP in transcription reactions in vitro. However, addition of TRP potently inhibited basal and activated transcription from multiple promoters in vitro and in vivo. General transcription factors TFIIA and TFIIB bound glutathione S-transferase-TRP in solution but failed to stimulate TRP binding to DNA. Preincubation of TRP with TFIIA inhibited TBP-TFIIA-DNA complex formation and addition of TFIIA overcame TRP-mediated transcription repression. TRP transcriptional repression activity was specifically reduced by mutations in TRP that disrupt the TFIIA binding surface but not by mutations that disrupt the TFIIB or DNA binding surface of TRP. These results suggest that TFIIA is a primary target of TRP transcription inhibition and that TRP may modulate transcription by a novel mechanism involving the partial mimicry of TBP functions.


Subject(s)
DNA-Binding Proteins/metabolism , Gene Expression Regulation , Promoter Regions, Genetic , Repressor Proteins/metabolism , Transcription Factors/metabolism , Transcription, Genetic , Amino Acid Sequence , Binding Sites , Cloning, Molecular , Conserved Sequence , DNA, Complementary/genetics , DNA-Binding Proteins/genetics , Evolution, Molecular , Glutathione Transferase/genetics , Humans , Molecular Sequence Data , Nucleic Acid Synthesis Inhibitors , Protein Binding , Repetitive Sequences, Amino Acid , Repressor Proteins/genetics , Sequence Homology, Amino Acid , TATA Box Binding Protein-Like Proteins , TATA-Box Binding Protein , Tissue Distribution , Transcription Factor TFIIA , Transcription Factor TFIIB , Transcription Factors/genetics
11.
Mol Cell Biol ; 19(4): 2846-52, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10082550

ABSTRACT

Posttranslational modification of general transcription factors may be an important mechanism for global gene regulation. The general transcription factor IIA (TFIIA) binds to the TATA binding protein (TBP) and is essential for high-level transcription mediated by various activators. Modulation of the TFIIA-TBP interaction is a likely target of transcriptional regulation. We report here that Toa1, the large subunit of yeast TFIIA, is phosphorylated in vivo and that this phosphorylation stabilizes the TFIIA-TBP-DNA complex and is required for high-level transcription. Alanine substitution of serine residues 220, 225, and 232 completely eliminated in vivo phosphorylation of Toa1, although no single amino acid substitution of these serine residues eliminated phosphorylation in vivo. Phosphorylated TFIIA was 30-fold more efficient in forming a stable complex with TBP and TATA DNA. Dephosphorylation of yeast-derived TFIIA reduced DNA binding activity, and recombinant TFIIA could be stimulated by in vitro phosphorylation with casein kinase II. Yeast strains expressing the toa1 S220/225/232A showed reduced high-level transcriptional activity at the URA1, URA3, and HIS3 promoters but were viable. However, S220/225/232A was synthetically lethal when combined with an alanine substitution mutation at W285, which disrupts the TFIIA-TBP interface. Phosphorylation of TFIIA could therefore be an important mechanism of transcription modulation, since it stimulates TFIIA-TBP association, enhances high-level transcription, and contributes to yeast viability.


Subject(s)
DNA-Binding Proteins/metabolism , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae/physiology , TATA Box , Transcription Factors/metabolism , Transcription, Genetic , Fungal Proteins/genetics , Gene Expression Regulation , Genes, Fungal , Hydro-Lyases/genetics , Models, Genetic , Phosphorylation , Protein Binding , Protein Processing, Post-Translational , TATA-Box Binding Protein , Transcription Factor TFIIA
12.
Mol Cell Biol ; 19(3): 1617-26, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10022850

ABSTRACT

Latent Epstein-Barr virus (EBV) is maintained as a nucleosome-covered episome that can be transcriptionally activated by overexpression of the viral immediate-early protein, Zta. We show here that reactivation of latent EBV by Zta can be significantly enhanced by coexpression of the cellular coactivators CREB binding protein (CBP) and p300. A stable complex containing both Zta and CBP could be isolated from lytically stimulated, but not latently infected RAJI nuclear extracts. Zta-mediated viral reactivation and transcriptional activation were both significantly inhibited by coexpression of the E1A 12S protein but not by an N-terminal deletion mutation of E1A (E1ADelta2-36), which fails to bind CBP. Zta bound directly to two related cysteine- and histidine-rich domains of CBP, referred to as C/H1 and C/H3. These domains both interacted specifically with the transcriptional activation domain of Zta in an electrophoretic mobility shift assay. Interestingly, we found that the C/H3 domain was a potent dominant negative inhibitor of Zta transcriptional activation function. In contrast, an amino-terminal fragment containing the C/H1 domain was sufficient for coactivation of Zta transcription and viral reactivation function. Thus, CBP can stimulate the transcription of latent EBV in a histone acetyltransferase-independent manner mediated by the CBP amino-terminal C/H1-containing domain. We propose that CBP may regulate aspects of EBV latency and reactivation by integrating cellular signals mediated by competitive interactions between C/H1, C/H3, and the Zta activation domain.


Subject(s)
DNA-Binding Proteins/metabolism , Herpesvirus 4, Human/physiology , Nuclear Proteins/metabolism , Trans-Activators/metabolism , Transcriptional Activation , Viral Proteins/metabolism , Virus Latency/physiology , Acetyltransferases/biosynthesis , Acetyltransferases/genetics , Animals , Binding Sites , CREB-Binding Protein , Cell Cycle Proteins/biosynthesis , Cell Cycle Proteins/genetics , DNA-Binding Proteins/genetics , E1A-Associated p300 Protein , Gene Expression Regulation, Viral , HeLa Cells , Herpesvirus 4, Human/growth & development , Histone Acetyltransferases , Humans , Mice , Nuclear Proteins/biosynthesis , Nuclear Proteins/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Trans-Activators/biosynthesis , Trans-Activators/genetics , Transcription Factors , Viral Proteins/genetics , Virus Activation , p300-CBP Transcription Factors
13.
J Biol Chem ; 273(23): 14293-300, 1998 Jun 05.
Article in English | MEDLINE | ID: mdl-9603936

ABSTRACT

The interaction of the general transcription factor (TF) IIA with TFIID is required for transcription activation in vitro. TFIID consists of the TATA-binding protein (TBP) and TBP associated factors (TAFIIs). TFIIA binds directly to TBP and stabilizes its interaction with TATA-containing DNA. In this work, we present evidence that TAFIIs inhibit TBP-DNA and TBP-TFIIA binding, and that TFIIA stimulates transcription, in part, by overcoming this TAFII-mediated inhibition of TBP-DNA binding. TFIIA mutants modestly compromised for interaction with TBP were found to be significantly more defective in forming complexes with TFIID. Subtle changes in the stability or conformation of the TFIIA-TBP complex resulted in a failure of TFIIA to overcome TAFII-mediated inhibition of TBP-DNA binding and transcription function. Inhibition of TBP-DNA binding by TAFIIs could be partially relieved by limited proteolysis of TFIID. Proteolysis significantly stimulated TFIIA-TFIID-TATA binding in both electrophoresis mobility shift assay and DNase I footprinting but had little effect on complexes formed with TBP. Recombinant TAFII250 inhibits TBP-DNA binding, whereas preincubation of TFIIA with TBP prevents this inhibition. Thus, TFIIA competes with TAFII250 for access to TBP and alters the TATA binding properties of the resulting complex. Transcriptional activation by Zta was enhanced by temperature shift inactivation of TAFII250 in the ts13 cell line, suggesting that TAFII250 has transcriptional inhibitory activity in vivo. Together, these results suggest that TAFIIs may regulate transcription initiation by inhibiting TBP-TFIIA and TBP-DNA complex formation.


Subject(s)
DNA-Binding Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Viral Proteins , DNA/metabolism , DNA Footprinting , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , Endopeptidase K/metabolism , Gene Expression Regulation/genetics , Humans , Mutation/genetics , Recombinant Proteins/metabolism , TATA Box/genetics , TATA Box Binding Protein-Like Proteins , TATA-Box Binding Protein , Trans-Activators/genetics , Transcription Factor TFIIA , Transcription Factor TFIID , Transcription Factors, TFII/metabolism , Transcriptional Activation/physiology
14.
Mol Cell Biol ; 18(5): 2559-70, 1998 May.
Article in English | MEDLINE | ID: mdl-9566876

ABSTRACT

The general transcription factor IIA (TFIIA) interacts with the TATA binding protein (TBP) and promoter DNA to mediate transcription activation in vitro. To determine if this interaction is generally required for activation of all class II genes in vivo, we have constructed substitution mutations in yeast TFIIA which compromise its ability to bind TBP. Substitution mutations in the small subunit of TFIIA (Toa2) at residue Y69 or W76 significantly impaired the ability of TFIIA to stimulate TBP-promoter binding in vitro. Gene replacement of wild-type TOA2 with a W76E or Y69A/W76A mutant was lethal in Saccharomyces cerevisiae, while the Y69F/W76F mutant exhibited extremely slow growth at 30 degrees C. Both the Y69A and W76A mutants were conditionally lethal at higher temperatures. Light microscopy indicated that viable toa2 mutant strains accumulate as equal-size dumbbells and multibudded clumps. Transcription of the cell cycle-regulatory genes CLB1, CLB2, CLN1, and CTS1 was significantly reduced in the toa2 mutant strains, while the noncycling genes PMA1 and ENO2 were only modestly affected, suggesting that these toa2 mutant alleles disrupt cell cycle progression. The differential effect of these toa2 mutants on gene transcription was examined for a number of other genes. toa2 mutant strains supported high levels of CUP1, PHO5, TRP3, and GAL1 gene activation, but the constitutive expression of DED1 was significantly reduced. Activator-induced start site expression for HIS3, GAL80, URA1, and URA3 promoters was defective in toa2 mutant strains, suggesting that the TFIIA-TBP complex is important for promoters which require an activator-dependent start site selection from constitutive to regulated expression. We present evidence to indicate that transcription defects in toa2 mutants can be both activator and promoter dependent. These results suggest that the association of TFIIA with TBP regulates activator-induced start site selection and cell cycle progression in S. cerevisiae.


Subject(s)
Cell Cycle/genetics , DNA-Binding Proteins/metabolism , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae/genetics , Transcription Factors/metabolism , DNA Mutational Analysis , Gene Expression Regulation, Fungal , Mutagenesis, Site-Directed , Phenotype , Promoter Regions, Genetic , Protein Binding , Saccharomyces cerevisiae/growth & development , TATA Box , TATA-Box Binding Protein , Transcription Factor TFIIA , Transcription Factors/genetics , Transcription, Genetic , Transcriptional Activation
15.
Mol Cell Biol ; 17(11): 6624-32, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9343426

ABSTRACT

Different mechanisms of transcriptional activation may be required for distinct classes of promoters and cellular conditions. The Epstein-Barr virus (EBV)-encoded transcriptional activator Zta recruits the general transcription factors IID (TFIID) and IIA (TFIIA) to promoter DNA and induces a TATA box-binding protein (TBP)-associated factor-dependent footprint downstream of the transcriptional initiation site. In this study, we investigated the functional significance of TFIID-TFIIA (D-A complex) recruitment by Zta. Alanine substitution mutations in the Zta activation domain which eliminate the ability of Zta to stimulate the D-A complex were examined. These Zta mutants were defective in the ability to activate transcription from an EBV-derived promoter (BHLF1) but activated a highly responsive synthetic promoter (Z7E4T). Both the number of activator binding sites and the core promoter region contribute to the requirement for D-A complex recruitment. These functionally distinct core promoters had significant differences in affinity for TBP and TFIID binding. The D-A complex-recruiting activity of Zta was found to be important for promoter selection in the presence of a competitor template. Conditions which limit TFIID binding to the TATA element or compromise the ability of TFIIA to bind TBP required activator stimulation of the D-A complex. These results indicate that D-A complex recruitment is one of at least two activation pathways utilized by Zta and is the essential pathway for a subset of promoters and conditions which limit TFIID binding to the TATA element.


Subject(s)
DNA-Binding Proteins/metabolism , Promoter Regions, Genetic , Trans-Activators/metabolism , Transcription Factors, TFII/metabolism , Transcription Factors/metabolism , Viral Proteins/metabolism , DNA-Binding Proteins/genetics , Gene Expression Regulation , Mutation , Phenotype , Protein Binding , TATA Box , TATA-Box Binding Protein , Trans-Activators/genetics , Transcription Factor TFIIA , Transcription Factor TFIID , Transcription, Genetic , Viral Proteins/genetics
16.
Methods ; 12(3): 217-23, 1997 Jul.
Article in English | MEDLINE | ID: mdl-9237166

ABSTRACT

The general transcription factors TFIID and TFIIA are critical for the recognition of promoter start sites and mediate the stimulatory effect of some transcriptional activators. The regulation of TFIID binding to promoter DNA by activators and coactivators can be studied using a modified gel electrophoresis mobility shift assay (EMSA). TFIID is a multiprotein complex that consists of the TATA binding protein (TBP) and TBP associated factors (TAFs). TBP is a sequence-specific DNA binding protein that binds in the minor groove and introduces an energetically unfavorable bending angle of 100 degrees in the DNA. The activated preinitiation complex consists of TAFs, TBP, TFIIA, multiple activators, and approximately 200 bp of promoter DNA. The large mass and DNA distortions of the preinitiation complex preclude the use of conventional low ionic strength polyacrylamide gel EMSA for analysis. These large complexes can be analyzed by EMSA in agarose gels that contain magnesium ion. The Mg-agarose EMSA is a simple system useful for resolution of large multiprotein complexes that may introduce distortions in linear DNA. Important parameters are discussed so that this technique can be generally applied to other model activators.


Subject(s)
Transcription Factors, TFII/chemistry , Transcription Factors, TFII/physiology , Transcriptional Activation , Cell Line , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/physiology , Electrophoresis, Agar Gel/methods , Magnesium , Nucleic Acid Probes , Promoter Regions, Genetic , Protein Binding , Trans-Activators/chemistry , Trans-Activators/physiology , Transcription Factor TFIIA , Transcription Factor TFIID , Transcription Factors/chemistry , Transcription Factors/physiology , Viral Proteins/chemistry , Viral Proteins/physiology
17.
Virology ; 239(2): 340-51, 1997 Dec 22.
Article in English | MEDLINE | ID: mdl-9434725

ABSTRACT

Epstein-Barr virus (EBV) replicates as a stable multicopy episome in latently infected mammalian cells. Latent cycle DNA replication requires only two viral elements, the cis-acting origin of plasmid replication (oriP) and the trans-acting origin binding protein (EBNA1). EBNA1 binds multiple recognition sites in oriP, but has not other enzymatic activities associated with replication functions. To identify human cellular proteins that mediate EBNA1 function, we designed a one-hybrid assay in yeast to select for proteins that bind to EBNA1 when bound to criP in vivo. A human cDNA encoding the Rch1/hSRP1 alpha/ importin alpha protein was isolated and shown to bind to full-length EBNA1, but not to an amino terminal deletion mutant of EBNA1 when bound to oriP in yeast. The interaction of EBNA1 with Rch1 was confirmed biochemically by coimmunoprecipitation from nuclear extracts and by direct binding of recombinant proteins in vitro. Internal deletion mutations in EBNA1 which compromised DNA replication activity were similarly reduced for binding to Rch1. Mutations with no effect on DNA replication activity were similarly unaffected for Rch1 binding. Rch1/importin alpha has been shown to bind to the nuclear localization sequence (NLS) of several proteins and stimulate nuclear import. A substitution mutation in the EBNA1 nuclear localization sequence reduced Rch1 binding, but had no effect on DNA replication function, indicating that Rch1 binding affinity does not correspond precisely with replication activity. Nevertheless, the identification of a stable interaction between Rch1 and EBNA1 at the origin of viral DNA replication raises the intriguing possibility that Rch1 contributes to the nuclear functions of EBNA1.


Subject(s)
Carrier Proteins/metabolism , Cell Nucleus/metabolism , DNA Replication , DNA, Viral/metabolism , Epstein-Barr Virus Nuclear Antigens/metabolism , Herpesvirus 4, Human/physiology , Nuclear Proteins/metabolism , Amino Acid Sequence , Biological Transport , Cells, Cultured , DNA, Complementary/genetics , Epstein-Barr Virus Nuclear Antigens/genetics , Herpesvirus 4, Human/genetics , Humans , Molecular Sequence Data , Protein Binding , Recombinant Fusion Proteins/metabolism , Regulatory Sequences, Nucleic Acid , Replication Origin , alpha Karyopherins
18.
J Virol ; 70(12): 8340-7, 1996 Dec.
Article in English | MEDLINE | ID: mdl-8970953

ABSTRACT

The Zta transactivator is crucial for both Epstein-Barr virus (EBV) lytic gene expression and lytic DNA replication. We have used a cotransfection-replication assay to examine the effect of mutations in the Zta activation domain (amino acids [aa] 1 to 167) on Zta replication activity. Deletion of Zta aa 25 to 86, which are critical for transcriptional activation of ori-Lyt, or aa 93 to 141 did not adversely affect replication of an ori-Lyt-containing target plasmid. However, removal of aa 2 to 25 (delta2-25) abolished replication activity. Within this subdomain, deletion of aa 2 to 10 (delta2-10) or mutation of codons 18 and 19 (m18/19) or 22 and 26 (m22/26) did not affect replication competency, while deletion of codons 13 to 19 (delta13-19) or mutation at codons 12 and 13 (m12/13) impaired Zta replication function. Each of the replication-negative Zta variants was capable of transactivating expression from both BHLF1 promoter-chloramphenicol acetyltransferase constructions and the BMRF1 promoter on endogenous EBV genomes in Raji cells with efficiency comparable to that of the wild-type polypeptide. Thus, a replication contribution of Zta was functionally separable from its transactivation activity and was supplied by the N-terminal region encompassing aa 11 to 25. Replication by a subset of the impaired Zta mutants was partially rescued upon the addition of Rta to the replication assay. The contribution of Rta mapped to domain II of the Rta activation domain and was specific for this region. A chimeric Rta-EBNA-2 transactivation domain fusion, which retains the DNA-binding and transactivation properties associated with wild-type Rta, failed to rescue replication-deficient Zta. Our data suggest that Rta may act as an ancillary replication factor in EBV ori-Lyt DNA synthesis by stabilizing Zta-replisome interactions.


Subject(s)
DNA Replication , DNA-Binding Proteins/genetics , Herpesvirus 4, Human/genetics , Replication Origin , Trans-Activators/genetics , Viral Proteins/genetics , Animals , Binding Sites , Chlorocebus aethiops , Gene Expression Regulation, Viral , Immediate-Early Proteins/genetics , Sequence Deletion , Structure-Activity Relationship , Transcription Factors/genetics , Tumor Cells, Cultured , Vero Cells
19.
EMBO J ; 15(24): 7079-87, 1996 Dec 16.
Article in English | MEDLINE | ID: mdl-9003783

ABSTRACT

Transcriptional repression of eukaryotic genes is essential for many cellular and developmental processes, yet the precise mechanisms of repression remain poorly understood. The Dorsal Switch Protein (DSP1) was identified in a genetic screen for activities which convert Dorsal into a transcriptional repressor. DSP1 shares structural homology with the HMG-1/2 family and inhibits activation by the rel transcription factors Dorsal and NF-kappaB in transfection studies. Here we investigate the mechanism of transcriptional repression by DSP1. We found that DSP1 protein can act as a potent transcriptional repressor for multiple activator families in vitro and in transfection studies. DSP1 bound directly to the TATA binding protein (TBP), and formed a stable ternary complex with TBP bound to DNA. DSP1 preferentially disrupted the DNA binding of TBP complexes containing TFIIA and displaced TFIIA from binding to TBP. Consistent with the inhibition of TFIIA-bound complexes, DSP1 was shown to inhibit activated but not basal transcription reactions in vitro. The ability of DSP1 to interact with TBP and to repress transcription was mapped to the carboxy-terminal domain which contains two HMG boxes. Our results support the model that DSP1 represses activated transcription by interfering with the binding of TFIIA, a general transcription factor implicated in activated transcription pathways.


Subject(s)
DNA-Binding Proteins/metabolism , Drosophila Proteins , Nuclear Proteins/metabolism , Phosphoproteins/metabolism , Repressor Proteins/metabolism , Transcription Factors/metabolism , Animals , Drosophila/embryology , Humans , Protein Binding , TATA Box , TATA-Box Binding Protein , Transcription Factor TFIIA , Transcription, Genetic
20.
Mol Cell Biol ; 16(5): 2110-8, 1996 May.
Article in English | MEDLINE | ID: mdl-8628277

ABSTRACT

The regulation of RNA polymerase II-mediated transcription involves both direct and indirect interactions among regulatory proteins and the general transcription factors (GTFs) that assemble at TATA-containing promoters. Here we show that the oncogenic transcription factors Fos and Jun make direct physical contacts with three proteins of the basal transcription apparatus, TFIIE-34 (TFIIE-beta), TFIIF-30 (RAP30), and TFIIF-74 (RAP74). The interactions among the activator proteins and these three GTFs were not detected with other transcription factors, including some bZIP protein family members. Both coimmunoprecipitation and protein blotting experiments demonstrated that the interactions were strongly favored by dimerization of Fos and Jun and that they involved the basic region and basic region-proximal domain of both proteins. Mutations within the DNA-binding domains of Fos and Jun abolished binding to GTFs, although the presence of DNA was not required for the association. Surprisingly, only a single basic region in the context of a protein dimer was sufficient for the interaction. Squelching of AP-1-dependent transcription in vitro by an excess of Fos-Jun dimers was relieved by the addition of TFIIE, indicating that it is a direct functional target of Fos and Jun. These results suggest that dimerization induces a conformational alteration in the basic region of Fos and Jun that promotes an association with TFIIE-34 and TFIIF, thus contributing to transcription initiation.


Subject(s)
Proto-Oncogene Proteins c-fos/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Transcription Factors, TFII , Transcription Factors/metabolism , Transcription, Genetic , Animals , Blotting, Western , Cell Line , Cloning, Molecular , Gene Expression Regulation , Humans , Mutagenesis , Plasmids , Protein Multimerization , Proto-Oncogene Proteins c-fos/biosynthesis , Proto-Oncogene Proteins c-jun/biosynthesis , RNA Polymerase II/metabolism , Rats , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/metabolism , Sequence Tagged Sites , Transcription Factor AP-1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...