Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 68
Filter
Add more filters










Publication year range
1.
J Virol ; 77(24): 13267-74, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14645583

ABSTRACT

Epstein-Barr virus (EBV) has an accepted association with the epithelial malignancy nasopharyngeal carcinoma and has also been reported in other more controversial carcinoma settings. Evaluation of EBV association with epithelial carcinomas such as breast cancer would benefit from a better understanding of the outcome of EBV infection of these cells. Cell-free preparations of a green fluorescent protein-expressing virus, BX1, were used to infect breast cancer cell lines, which were then examined for EBV gene expression and viral genome copy number. Reverse transcription-PCR analyses revealed that the cells supported a mixture of latency II and lytic EBV gene expression. Lytic Zta and BMRF1 protein expression was detected by immunohistochemistry, and DNA PCR analyses estimated an EBV copy number of 300 to 600 genomes per infected cell. Evidence for lytic EBV expression was also found in breast tissue, where reverse transcription-PCR analyses detected lytic Zta transcripts in 7 of 10 breast carcinoma tissues and 4 of 10 normal tissues from the same patients. Scattered cells immunoreactive for Zta protein were also detectable in breast carcinoma. Quantitative real-time PCR analysis of EBV-positive breast carcinoma tissues suggested that less than 0.1% of the cells contained viral genomes. We suggest that sporadic lytic EBV infection may contribute to PCR-based detection of EBV in traditionally nonvirally associated epithelial malignancies.


Subject(s)
Breast Neoplasms/virology , Herpesvirus 4, Human/physiology , Viral Proteins , Virus Replication , Antigens, Viral/genetics , Antigens, Viral/metabolism , Breast/virology , Carcinoma/virology , DNA, Viral/analysis , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Epstein-Barr Virus Infections/virology , Female , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/isolation & purification , Herpesvirus 4, Human/pathogenicity , Humans , Polymerase Chain Reaction , Trans-Activators/genetics , Trans-Activators/metabolism , Tumor Cells, Cultured , Virus Latency
2.
Virology ; 288(1): 119-28, 2001 Sep 15.
Article in English | MEDLINE | ID: mdl-11543664

ABSTRACT

The Epstein-Barr virus (EBV) Mta protein is a posttranscriptional regulator of EBV lytic gene expression that affects RNA splicing and transport. Mta mediates cytoplasmic accumulation of unspliced EBV replication gene transcripts and shuttles between the nucleus and cytoplasm. Mta contains a recognized leucine-rich, putative nuclear export signal (NES) between aa 227 and 236. Deletion of this signal sequence eliminated shuttling, while mutation of the core LXL motif in the putative NES diminished but did not abolish the ability of Mta to shuttle from donor to recipient cells in a heterokaryon assay. A double mutation of the LXL motif plus an upstream VTL motif eliminated shuttling, suggesting that Mta may have two NES motifs. In confirmation of this, transfer of either the sequence encoding the leucine-rich aa 227-236 motif or that encoding the adjacent hydrophobic aa 218-227 sequence to a GFP-NLS-pyruvate kinase reporter protein conferred the property of cytoplasmic accumulation onto the heterologous protein. Cytoplasmic accumulation of both the aa 225-237 and 218-227 containing reporters was minimal in the presence of the inhibitor leptomycin B, indicating that both motifs mediated Crm-1-dependent export. Mutations in the NES signal sequences abolished the ability of Mta to mediate cytoplasmic accumulation of BALF2 replication gene transcripts. This included mutation of the LXL motif which still showed cytoplasmic shuttling, suggesting that the NES mutations might have additional effects on Mta function. Wild-type Mta co-immunoprecipitated with the splicing factor SC35 and colocalized with SC35 in transfected cells, modifying endogenous SC35 distribution within the nucleus to give more intense, rounded spots. Interestingly, the NES mutant proteins appeared to have altered interactions with the splicing complex, binding more tightly to SC35 in co-immunoprecipitation assays. These observations suggest a linkage between the splicing and export functions of Mta.


Subject(s)
Herpesvirus 4, Human/physiology , Phosphoproteins/metabolism , Trans-Activators/metabolism , Transcription, Genetic , Virus Replication , Amino Acid Sequence , Animals , Cell Line , Cell Nucleus/physiology , Cell Nucleus/virology , Chlorocebus aethiops , Cytoplasm/physiology , Cytoplasm/virology , Genes, Reporter , Green Fluorescent Proteins , HeLa Cells , Herpesvirus 4, Human/genetics , Humans , Leucine , Luminescent Proteins/genetics , Phosphoproteins/chemistry , Protein Transport , Pyruvate Kinase/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Sequence Deletion , Trans-Activators/chemistry , Transfection , Vero Cells , Viral Proteins/chemistry , Viral Proteins/metabolism
3.
J Virol ; 75(18): 8792-802, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11507224

ABSTRACT

Zta has a dual role in the Epstein-Barr virus (EBV) lytic cycle, acting as a key regulator of EBV lytic gene expression and also being essential for lytic viral DNA replication. Zta's replication function is mediated in part through interactions with the core viral replication proteins. We now show interaction between Zta and the helicase (BBLF4) and map the binding region to within amino acids (aa) 22 to 86 of the Zta activation domain. In immunofluorescence assays, green fluorescent protein (GFP)-tagged BBLF4 localized to the cytoplasm of transfected cells. Cotransfection of Zta resulted in translocation of BBLF4-GFP into the nucleus indicating interaction between these two proteins. However, Zta with a deletion of aa 24 to 86 was unable to mediate nuclear translocation of BBLF4-GFP. Results obtained with Zta variants carrying deletions across the aa 24 to 86 region indicated more than one contact site for BBLF4 within this domain, and this was reinforced by the behavior of the four-point mutant Zta (m22/26,74/75), which was severely impaired for BBLF4 interaction. Binding of BBLF4 to Zta was confirmed using GST affinity assays. In both cotransfection-replication assays and replication assays performed in EBV-positive P3HR1 cells, the Zta (m22/26,74/75) mutant was replication defective. In Zta-transfected D98-HR1 cells, replication compartments could be detected by immunofluorescence staining using anti-BMRF1 monoclonal antibody. Cells transfected with Zta variants that were defective for helicase binding still formed replication compartments, but Zta was excluded from these compartments. These experiments reveal a role for the Zta-helicase interaction in targeting Zta to sites of viral DNA replication.


Subject(s)
DNA Helicases/metabolism , DNA Replication , DNA-Binding Proteins/metabolism , Herpesvirus 4, Human/metabolism , Trans-Activators/metabolism , Viral Proteins , Virus Replication , DNA Helicases/genetics , DNA-Binding Proteins/genetics , Genes, Reporter , Green Fluorescent Proteins , HeLa Cells , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/physiology , Humans , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Trans-Activators/genetics , Transcriptional Activation
4.
Mol Cell Biol ; 21(18): 6222-32, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11509665

ABSTRACT

The CSL family protein CBF1 is a nuclear mediator of Notch signaling and has been predicted to contain an N-terminal nuclear localization signal in exon 4. Surprisingly, we found that CBF1 carrying mutations at codon 233 or 249 within exon 7 was restricted to the cytoplasm. In mammalian and yeast two-hybrid assays, these mutations were also associated with a loss of CBF1-mediated transcriptional repression and a severely impaired interaction with the corepressors SMRT and CIR. Overexpression of SMRT rescued the ability of mutant CBF1 to target to the nucleus of transfected cells and similarly rescued nuclear targeting of enhanced green fluorescent protein (EGFP)-CBF1 exons 6 to 9 CBF1(6-9)carrying the codon 233 or 249 mutations. Carboxy-terminally truncated SMRT with amino acids (aa) 1291 to 1495 deleted was unable to rescue the nuclear targeting of mutant EGFP-CBF1(6-9). In yeast two-hybrid assays, the SMRT aa 1291 to 1495 domain interacted with SKIP and SMRT aa 1291 to 1495 colocalized with SKIP within the nuclei of cotransfected cells. Comparison of the intracellular localization of CBF1(6-9) with that of CBF1(5-9) further supported the suggestion that nuclear targeting of CBF1 is dependent on the formation of a CBF1-SMRT-SKIP corepressor complex. These observations suggest that nuclear targeting of CBF1 is itself a component of CBF1-mediated gene regulation and that in the absence of signaling, CBF1 enters the nucleus precommitted to a transcriptional repression function. The activators NotchIC (the intracellular domain of Notch) and Epstein-Barr virus EBNA2 also mediated nuclear targeting of mutant CBF1, consistent with the competition model for activator versus corepressor binding to CBF1.


Subject(s)
DNA-Binding Proteins/metabolism , Nuclear Proteins , Repressor Proteins/metabolism , Signal Transduction , Cell Nucleus/metabolism , DNA-Binding Proteins/genetics , HeLa Cells , Humans , Immunoglobulin J Recombination Signal Sequence-Binding Protein , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mutation , Nuclear Receptor Co-Repressor 2 , Protein Binding , Receptors, Notch , Repressor Proteins/genetics , Transcriptional Activation
6.
J Virol ; 75(6): 2929-37, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11222718

ABSTRACT

Epstein-Barr virus (EBV) latency gene expression in lymphoblastoid cell lines is regulated by EBNA2. However, the factors regulating viral expression in EBV-associated tumors that do not express EBNA2 are poorly understood. In EBV-associated tumors, EBNA1 and frequently LMP1 are synthesized. We found that an alternative latent membrane protein 1 (LMP1) promoter, L1-TR, located within the terminal repeats is active in both nasopharyngeal carcinoma and Hodgkin's disease tissues. Examination of the L1-TR and the standard ED-L1 LMP1 promoters in electrophoretic mobility shift assays revealed that both promoters contain functional STAT binding sites. Further, both LMP1 promoters responded in reporter assays to activation of JAK-STAT signaling. Cotransfection of JAK1 or v-Src or treatment of cells with the cytokine interleukin-6 upregulated expression from ED-L1 and L1-TR reporter plasmids. Cotransfection of a dominant negative STAT3 beta revealed that STAT3 is likely to be the biologically relevant STAT for EBNA1 Qp and LMP1 L1-TR promoter regulation. In contrast, LMP1 expression from ED-L1 was not abrogated by STAT3 beta, indicating that the two LMP1 promoters are regulated by different STAT family members. Taken together with the previous demonstration of JAK-STAT activation of Qp driven EBNA1 expression, this places two of the EBV genes most commonly expressed in tumors under the control of the same signal transduction pathway. Immunohistochemical analyses of nasopharyngeal carcinoma tumors revealed that STAT3, STAT5, and STAT1 are constitutively activated in these tumors while STAT3 is constitutively activated in the malignant cells of Hodgkin's disease. We hypothesize that chronic or aberrant STAT activation may be both a necessary and predisposing event for EBV-driven tumorigenesis in immunocompetent individuals.


Subject(s)
DNA-Binding Proteins/metabolism , Gene Expression Regulation, Viral , Herpesvirus 4, Human/genetics , Hodgkin Disease/virology , Nasopharyngeal Neoplasms/virology , Trans-Activators/metabolism , Viral Matrix Proteins/metabolism , Cell Line , DNA-Binding Proteins/genetics , Herpesvirus 4, Human/metabolism , Herpesvirus 4, Human/pathogenicity , Hodgkin Disease/metabolism , Humans , Nasopharyngeal Neoplasms/metabolism , Promoter Regions, Genetic/genetics , Protein-Tyrosine Kinases/metabolism , STAT3 Transcription Factor , Signal Transduction , Trans-Activators/genetics , Viral Matrix Proteins/genetics , Virus Latency
7.
J Virol ; 75(6): 2946-56, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11222720

ABSTRACT

The Epstein-Barr virus (EBV) BamHI-A rightward transcripts (BARTs) are expressed in all EBV-associated tumors as well as in latently infected B cells in vivo and cultured B-cell lines. One of the BART family transcripts contains an open reading frame, RPMS1, that encodes a nuclear protein termed RPMS. Reverse transcription-PCR analysis revealed that BART transcripts with the splicing pattern that generates the RPMS1 open reading frame are commonly expressed in EBV-positive lymphoblastoid cell lines and are also detected in Hodgkin's disease tissues. Experiments undertaken to determine the function of RPMS revealed that RPMS interacts with both CBF1 and components of the CBF1-associated corepressor complex. RPMS interaction with CBF1 was demonstrated in a glutathione S-transferase (GST) affinity assay and by the ability of RPMS to alter the intracellular localization of a mutant CBF1. A Gal4-RPMS fusion protein mediated transcriptional repression, suggesting an additional interaction between RPMS and corepressor proteins. GST affinity assays revealed interaction between RPMS and the corepressor Sin3A and CIR. The RPMS-CIR interaction was further substantiated in mammalian two-hybrid, coimmunoprecipitation, and colocalization experiments. RPMS has been shown to interfere with NotchIC and EBNA2 activation of CBF1-containing promoters in reporter assays. Consistent with this function, immunofluorescence assays performed on cotransfected cells showed that there was colocalization of RPMS with NotchIC and with EBNA2 in intranuclear punctate speckles. The effect of RPMS on NotchIC function was further examined in a muscle cell differentiation assay where RPMS was found to partially reverse NotchIC-mediated inhibition of differentiation. The mechanism of RPMS action was examined in cotransfection and mammalian two-hybrid assays. The results revealed that RPMS blocked relief of CBF1-mediated repression and interfered with SKIP-CIR interactions. We conclude that RPMS acts as a negative regulator of EBNA2 and Notch activity through its interactions with the CBF1-associated corepressor complex.


Subject(s)
Deoxyribonuclease BamHI/metabolism , Epstein-Barr Virus Nuclear Antigens/metabolism , Gene Expression Regulation, Viral , Herpesvirus 4, Human/genetics , Membrane Proteins/metabolism , Neoplasm Proteins , RNA, Viral/genetics , RNA, Viral/metabolism , Repressor Proteins/metabolism , Viral Proteins , Cell Differentiation , Cell Line , Epstein-Barr Virus Nuclear Antigens/genetics , Herpesvirus 4, Human/metabolism , Humans , Membrane Proteins/genetics , Muscles/cytology , RNA, Messenger/metabolism , RNA, Viral/chemistry , Receptors, Notch , Repressor Proteins/genetics , Transcription, Genetic , Transfection
8.
J Virol ; 75(3): 1487-506, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11152521

ABSTRACT

Six predicted Kaposi's sarcoma virus herpesvirus (KSHV) proteins have homology with other well-characterized herpesvirus core DNA replication proteins and are expected to be essential for viral DNA synthesis. Intact Flag-tagged protein products from all six were produced from genomic expression vectors, although the ORF40/41 transcript encoding a primase-helicase component proved to be spliced with a 127-bp intron. The intracellular localization of these six KSHV replication proteins and the mechanism of their nuclear translocation were investigated. SSB (single-stranded DNA binding protein, ORF6) and PPF (polymerase processivity factor, ORF59) were found to be intrinsic nuclear proteins, whereas POL (polymerase, ORF9), which localized in the cytoplasm on its own, was translocated to the nucleus when cotransfected with PPF. PAF (primase-associated factor, ORF40/41), a component of the primase-helicase tripartite subcomplex together with PRI (primase, ORF56) and HEL (helicase, ORF44), required the presence of all five other replication proteins for efficient nuclear translocation. Surprisingly, even in the absence of a lytic cycle replication origin (ori-Lyt) and any known initiator or origin binding protein, the protein products of all six KSHV core replication genes cooperated in a transient cotransfection assay to form large globular shaped pseudo-replication compartments (pseudo-RC), which excluded cellular DNA. These pseudo-RC structures were confirmed to include POL, SSB, PRI, and PAF but did not contain any newly synthesized DNA. Similar to the human cytomegalovirus system, the peripheries of these KSHV pre-RC were also found to be surrounded by punctate PML oncogenic domains (PODs). Furthermore, by transient cotransfection, the six KSHV core replication machinery proteins successfully replicated a plasmid containing EBV ori-Lyt in the presence of the Epstein-Barr virus-encoded DNA binding initiator protein, ZTA. The KSHV-encoded K8 (ORF-K8) protein, which is a distant evolutionary homologue to ZTA, was incorporated into pseudo-RC structures formed by transient cotransfection with the six core KSHV replication genes. However, unlike ZTA, K8 displayed a punctate nuclear pattern both in transfected cells and at early stages of lytic infection and colocalized with the cellular PML proteins in PODs. Finally, K8 was also found to accumulate in functional viral RC, detected by incorporation of pulse-labeled bromodeoxyuridine into newly synthesized DNA in both tetradecanoyl phorbol acetate-induced JSC-1 primary effusion lymphoblasts and in KSHV lytically infected endothelial cells.


Subject(s)
Antifungal Agents , DNA Replication , DNA-Binding Proteins/physiology , Herpesvirus 8, Human/physiology , Neoplasm Proteins/physiology , Nuclear Proteins , Trans-Activators/physiology , Transcription Factors/physiology , Viral Proteins , Virus Assembly , Virus Replication , Amino Acid Sequence , Animals , Cell Nucleus/metabolism , Chlorocebus aethiops , Cloning, Molecular , Fungal Proteins/genetics , Fungal Proteins/physiology , Molecular Sequence Data , Open Reading Frames , Penicillium , Transfection , Tumor Suppressor Proteins , Vero Cells , Viral Core Proteins/physiology
9.
J Virol ; 74(20): 9637-45, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11000236

ABSTRACT

The human herpesvirus 8 (HHV-8) latency-associated nuclear antigen (LANA) is expressed in all latently HHV-8 infected cells and in HHV-8-associated tumors, including primary effusion lymphoma (PEL). To better understand the contribution of LANA to tumorigenesis and to the PEL phenotype, we performed a yeast two-hybrid screen which identified the corepressor protein SAP30 as a LANA binding protein. SAP30 is a constituent of a large multicomponent complex that brings histone deacetylases to the promoter. Glutathione S-transferase affinity assays confirmed interaction between LANA and SAP30 and also demonstrated interactions between LANA and two other members of the corepressor complex, mSin3A and CIR. The corepressors bound to the amino-terminal 340-amino-acid domain of LANA. In transient expression assays, this same domain of LANA mediated repression when targeted to a 5xGal4tk-CAT reporter as a GAL4-LANA fusion. PEL cells have the unusual feature that they are frequently dually infected with both HHV-8 and Epstein-Barr virus (EBV). We found that EBV EBNA-1 expression is downregulated in PEL cells at both the RNA and protein levels. In transient expression assays, LANA repressed activated expression from the EBV Qp and Cp latency promoters. Reduction of endogenous Qp activity could also be demonstrated in EBV-infected Rael cells transfected with a LANA expression plasmid. In contrast to the effect of LANA on EBV latency promoters, LANA activated expression from its own promoter. The data indicate that LANA can mediate transcriptional repression through recruitment of an mSin3 corepressor complex and further that LANA-mediated repression is likely to contribute to the low level of EBV latency gene expression seen in dually infected PEL cells.


Subject(s)
Antigens, Viral/physiology , Gene Expression Regulation, Viral , Herpesvirus 4, Human/genetics , Herpesvirus 8, Human/physiology , Histone Deacetylases/physiology , Nuclear Proteins/physiology , Repressor Proteins/physiology , Epstein-Barr Virus Nuclear Antigens/physiology , Histone Deacetylases/analysis , Humans , Nuclear Proteins/analysis
10.
J Virol ; 74(7): 3082-92, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10708423

ABSTRACT

CST (BART BARF0) family viral RNAs are expressed in several types of Epstein-Barr virus (EBV) infection, including EBV-associated cancers. Many different spliced forms of these RNAs have been described; here we have clarified the structures of some of the more abundant splicing patterns. We report the first cDNAs representing a full-length CST mRNA from a clone library and further characterize the transcription start. The relative abundance of splicing patterns and genomic analysis of the open reading frames (ORFs) suggest that, in addition to the much studied BARF0 ORF, there may be important products made from some of the upstream ORFs in the CST RNAs. Potential biological functions are identified for two of these. The product of the RPMS1 ORF is shown to be a nuclear protein that can bind to the CBF1 component of Notch signal transduction. RPMS1 can inhibit the transcription activation induced through CBF1 by NotchIC or EBNA-2. The protein product of another CST ORF, A73, is shown to be a cytoplasmic protein which can interact with the cell RACK1 protein. Since RACK1 modulates signaling from protein kinase C and Src tyrosine kinases, the results suggest a possible role for CST products in growth control, perhaps consistent with the abundant transcription of CST RNAs in cancers such as nasopharyngeal carcinoma.


Subject(s)
Herpesvirus 4, Human/genetics , RNA, Viral/genetics , Viral Proteins/genetics , Amino Acid Sequence , Base Sequence , DNA Primers , DNA, Complementary , Genome, Viral , HeLa Cells , Humans , Molecular Sequence Data , Open Reading Frames , Promoter Regions, Genetic , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid
11.
Mol Cell Biol ; 20(7): 2400-10, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10713164

ABSTRACT

Notch proteins are transmembrane receptors that mediate intercell communication and direct individual cell fate decisions. The activated intracellular form of Notch, NotchIC, translocates to the nucleus, where it targets the DNA binding protein CBF1. CBF1 mediates transcriptional repression through the recruitment of an SMRT-histone deacetylase-containing corepressor complex. We have examined the mechanism whereby NotchIC overcomes CBF1-mediated transcriptional repression. We identified SKIP (Ski-interacting protein) as a CBF1 binding protein in a yeast two-hybrid screen. Both CBF1 and SKIP are highly conserved evolutionarily, and the SKIP-CBF1 interaction is also conserved in assays using the Caenorhabditis elegans and Drosophila melanogaster SKIP homologs. Protein-protein interaction assays demonstrated interaction between SKIP and the corepressor SMRT. More surprisingly, SKIP also interacted with NotchIC. The SMRT and NotchIC interactions were mutually exclusive. In competition binding experiments SMRT displaced NotchIC from CBF1 and from SKIP. Contact with SKIP is required for biological activity of NotchIC. A mutation in the fourth ankyrin repeat that abolished Notch signal transduction did not affect interaction with CBF1 but abolished interaction with SKIP. Further, NotchIC was unable to block muscle cell differentiation in myoblasts expressing antisense SKIP. The results suggest a model in which NotchIC activates responsive promoters by competing with the SMRT-corepressor complex for contacts on both CBF1 and SKIP.


Subject(s)
Ankyrin Repeat/genetics , DNA-Binding Proteins/metabolism , Membrane Proteins/metabolism , Nuclear Proteins/metabolism , Amino Acid Sequence , Cell Differentiation , Cells, Cultured , DNA, Antisense , Humans , Immunoglobulin J Recombination Signal Sequence-Binding Protein , Molecular Sequence Data , Muscle Development , Mutation , Nuclear Receptor Co-Repressor 2 , Nuclear Receptor Coactivators , Receptors, Notch , Repressor Proteins/metabolism , Sequence Homology, Amino Acid , Transcription Factors
12.
J Virol ; 74(4): 1939-47, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10644367

ABSTRACT

EBNA2 is essential for Epstein-Barr virus (EBV) immortalization of B lymphocytes. EBNA2 functions as a transcriptional activator and targets responsive promoters through interaction with the cellular DNA binding protein CBF1. We have examined the mechanism whereby EBNA2 overcomes CBF1-mediated transcriptional repression. A yeast two-hybrid screen performed using CBF1 as the bait identified a protein, SKIP, which had not previously been recognized as a CBF1-associated protein. Protein-protein interaction assays demonstrated contacts between SKIP and the SMRT, CIR, Sin3A, and HDAC2 proteins of the CBF1 corepressor complex. Interestingly, EBNA2 also interacted with SKIP in glutathione S-transferase affinity and mammalian two-hybrid assays and colocalized with SKIP in immunofluorescence assays. Interaction with SKIP was not affected by mutation of EBNA2 conserved region 6, the CBF1 interaction region, but was abolished by mutation of conserved region 5. Mutation of conserved region 5 also severely impaired EBNA2 activation of a reporter containing CBF1 binding sites. Thus, interaction with both CBF1 and SKIP is necessary for efficient promoter activation by EBNA2. A model is presented in which EBNA2 competes with the SMRT-corepressor complex for contacts on SKIP and CBF1.


Subject(s)
DNA-Binding Proteins/metabolism , Epstein-Barr Virus Nuclear Antigens , Gene Expression Regulation , Nuclear Proteins/physiology , Promoter Regions, Genetic , Viral Proteins/metabolism , Amino Acid Sequence , Binding Sites , Cell Line, Transformed , DNA-Binding Proteins/genetics , HeLa Cells , Histone Deacetylase 2 , Histone Deacetylases/metabolism , Humans , Immunoglobulin J Recombination Signal Sequence-Binding Protein , Molecular Sequence Data , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Nuclear Receptor Co-Repressor 2 , Nuclear Receptor Coactivators , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Repressor Proteins/metabolism , Sin3 Histone Deacetylase and Corepressor Complex , Transcription Factors , Transfection , Viral Proteins/genetics
13.
Proc Natl Acad Sci U S A ; 96(16): 9339-44, 1999 Aug 03.
Article in English | MEDLINE | ID: mdl-10430944

ABSTRACT

In Epstein-Barr virus (EBV)-associated tumors in nonimmunocompromised patients, EBV gene expression is highly restricted. EBV-encoded nuclear antigen (EBNA)-1 is expressed, whereas the immunogenic and proliferative EBNAs are not. This pattern of EBNA expression is generated by usage of the BamHI-Q promoter (Qp). We have determined that the JAK/STAT pathway positively regulates Qp activity. In transient-transfection assays, a Qp-CAT reporter was activated by cotransfected JAK-1 and by treatment of cells with the cytokine IL-6. The ability of Qp to bind signal transducer and activator of transcription (STAT) proteins was directly demonstrated by electrophoretic mobility-shift assay, and mutation of potential STAT-binding sites reduced Qp responsiveness to Janus kinase (JAK)-1. Consistent with a role for STATs in Qp function, Qp using Burkitt's lymphoma Rael cells and cultured nasopharyngeal carcinoma (NPC) cells contained nuclear STAT protein. We investigated whether the inability to maintain EBV-positive NPC cell lines in culture was related to Qp activity. Passaging of the NPC cell line HK666 led to activation of expression of BZLF1, which encodes Zta and loss of Qp function. Transient expression assays linked Zta expression to the down-regulation of Qp. Cotransfection of Zta reduced Qp activity in reporter assays. This negative regulation required Zta DNA-binding activity. We provide evidence that Zta up-regulation of p53 leads to p53-mediated interference with JAK/STAT activation of Qp. The data imply that JAK/STAT signaling has a role in EBV-associated malignancies.


Subject(s)
DNA-Binding Proteins/metabolism , Epstein-Barr Virus Nuclear Antigens/genetics , Gene Expression Regulation, Viral , Herpesvirus 4, Human/genetics , Promoter Regions, Genetic , Protein-Tyrosine Kinases/metabolism , Trans-Activators/metabolism , B-Lymphocytes , Base Sequence , Burkitt Lymphoma , Cell Line , Chloramphenicol O-Acetyltransferase/genetics , Epstein-Barr Virus Nuclear Antigens/biosynthesis , HeLa Cells , Humans , Molecular Sequence Data , Mutagenesis, Site-Directed , Nasopharyngeal Neoplasms , Recombinant Fusion Proteins/biosynthesis , STAT1 Transcription Factor , STAT4 Transcription Factor , Signal Transduction , Transfection , Tumor Cells, Cultured , Virus Latency
14.
Blood ; 93(9): 3026-32, 1999 May 01.
Article in English | MEDLINE | ID: mdl-10216099

ABSTRACT

In addition to the Epstein-Barr virus (EBV) EBNA and LMP latency genes, there is a family of alternatively spliced BamHI-A rightward transcripts (BARTs). These latency transcripts are highly expressed in the EBV-associated malignancies nasopharyngeal carcinoma and Burkitt's lymphoma, and are expressed at lower levels in latently EBV-infected B-cell lines. The contribution of the BARTs to EBV biology or pathogenesis is unknown. Resting B cells have recently been recognized as a reservoir for EBV persistence in the peripheral blood. In these cells, EBV gene expression is tightly restricted and the only viral gene known to be consistently expressed is LMP2A. We used cell sorting and reverse-transcriptase polymerase chain reaction (RT-PCR) to examine whether BARTs are expressed in the restricted form of in vivo latency. Our results demonstrated that RNAs with splicing diagnostic for transcripts containing the BART RPMS1 and BARFO open-reading frames (ORFs) were expressed in CD19(+) but not in CD23(+) B cells isolated from peripheral blood of healthy individuals. The product of the proximal RPMS1 ORF has not previously been characterized. The RPMS1 ORF was shown to encode a 15-kD protein that localized to the nucleus of transfected cells. Expression of the BARTs in peripheral blood B cells suggests that the proteins encoded by these transcripts are likely to be important for maintenance of in vivo latency.


Subject(s)
B-Lymphocytes/virology , Gene Expression Regulation, Viral , Transcription, Genetic , Virus Latency/genetics , Antigens, CD/analysis , Antigens, CD19/analysis , B-Lymphocytes/immunology , Base Sequence , Burkitt Lymphoma/genetics , DNA Primers , Deoxyribonuclease BamHI , Epstein-Barr Virus Nuclear Antigens/genetics , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/physiology , Humans , Molecular Sequence Data , Open Reading Frames , Restriction Mapping , Reverse Transcriptase Polymerase Chain Reaction , Sequence Alignment , Sequence Homology, Nucleic Acid , Viral Matrix Proteins/genetics
15.
Proc Natl Acad Sci U S A ; 96(1): 23-8, 1999 Jan 05.
Article in English | MEDLINE | ID: mdl-9874765

ABSTRACT

CBF1 is a member of the CSL family of DNA binding factors, which mediate either transcriptional repression or transcriptional activation. CSL proteins play a central role in Notch signaling and in Epstein-Barr virus-induced immortalization. Notch is a transmembrane protein involved in cell-fate decisions, and the cytoplasmic domain of Notch (NotchIC) targets CBF1. The Epstein-Barr virus-immortalizing protein EBNA2 activates both cellular and viral gene expression by targeting CBF1 and mimicking NotchIC. We have examined the mechanism of CBF1-mediated repression and show that CBF1 binds to a unique corepressor, CBF1 interacting corepressor (CIR). A CIR homolog is encoded by Caenorhabditis elegans, indicating that CIR is evolutionarily conserved. Two CBF1 mutants that were unable to bind CIR did not function as repressors, suggesting that targeting of CIR to CBF1 is an important component of repression. When expressed as a Gal4 fusion protein, CIR repressed reporter gene expression. CIR binds to histone deacetylase and to SAP30 and serves as a linker between CBF1 and the histone deacetylase complex.


Subject(s)
DNA-Binding Proteins/metabolism , Histone Deacetylases/metabolism , Nuclear Proteins , Repressor Proteins/metabolism , Amino Acid Sequence , Animals , Binding Sites , Caenorhabditis elegans , Cell Nucleus/ultrastructure , Chlorocebus aethiops , Conserved Sequence , DNA-Binding Proteins/isolation & purification , Fluorescent Antibody Technique, Indirect , Humans , Immunoglobulin J Recombination Signal Sequence-Binding Protein , Molecular Sequence Data , Protein Binding , Recombinant Fusion Proteins/metabolism , Repressor Proteins/genetics , Repressor Proteins/isolation & purification , Sequence Homology, Amino Acid , Vero Cells
16.
J Virol ; 72(12): 9526-34, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9811685

ABSTRACT

The Epstein-Barr virus (EBV) Zta and Mta regulatory proteins were previously found to be required for efficient replication of oriLyt in cotransfection-replication assays, but the contribution of Mta to the replication process was unknown. We now demonstrate that Mta regulates replication gene expression. Using the polymerase processivity factor BMRF1 as an example, we found that in transfected cells, total BMRF1 mRNA levels were unaffected by Mta but that the amounts of cytoplasmic BMRF1 RNA and protein were greatly increased in the presence of Mta. Mta also increased cytoplasmic accumulation of the BALF2, BALF5, BSLF1, and BBLF4 replication gene mRNAs but did not affect cytoplasmic levels of BBLF2/3 mRNA. Thus, five of the six core replication genes require Mta for efficient accumulation of cytoplasmic RNA. The contribution of Mta to posttranscriptional RNA processing was examined. Examination of Mta localization in transfected cells by indirect immunofluorescence revealed that Mta colocalized with the splicing factor SC35. We also found that Mta has RNA binding activity. Glutathione S-transferase-Mta bound to BMRF1 and BMLF1 transcripts but not to a control cellular gene RNA. Mta contains a consensus leucine-rich nuclear export signal. Such signal sequences are characteristic of proteins that undergo nuclear export. Examination of Mta localization in a heterokaryon assay provided evidence that Mta shuttles between the nucleus and the cytoplasm. Our experiments indicate that Mta functions in RNA processing and transport and mediates cytoplasmic accumulation of a number of EBV early mRNAs.


Subject(s)
Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Viral/genetics , RNA, Viral/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Viral Proteins/genetics , Viral Proteins/metabolism , Amino Acid Sequence , Animals , Base Sequence , Biological Transport, Active , Cell Line , Cell Nucleus/metabolism , Cell Nucleus/virology , Chlorocebus aethiops , Cytoplasm/metabolism , Cytoplasm/virology , DNA Primers/genetics , Genes, Viral , Herpesvirus 4, Human/physiology , Humans , RNA Processing, Post-Transcriptional , Transfection , Vero Cells , Virus Replication/genetics
17.
J Virol ; 72(11): 8559-67, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9765394

ABSTRACT

The Epstein-Barr virus transactivator Zta triggers lytic gene expression and is essential for replication of the lytic origin, oriLyt. Previous analysis indicated that the Zta activation domain contributed a replication-specific function. We now show that the Zta activation domain interacts with components of the EBV helicase-primase complex. The three helicase-primase proteins BBLF4 (helicase), BSLF1 (primase), and BBLF2/3 (primase-associated factor) were expressed fused to the Myc epitope. When expression plasmids for BBLF4 or BBLF2/3 plus BSLF1 (primase subcomplex) were separately transfected, the proteins localized to the cytoplasm. Interaction between Zta and the components of the helicase-primase complex was tested by examining the ability of Zta to alter the intracellular localization of these proteins. Cotransfection of Zta with Myc-BBLF4 resulted in nuclear translocation of Myc-BBLF4; similarly, cotransfection of Zta with the primase subcomplex led to nuclear translocation of the Myc-BSLF1 and Myc-BBLF2/3 proteins. This relocalization provides evidence for an interaction between Zta and the helicase and Zta and the primase subcomplex. An affinity assay using glutathione S-transferase-Zta fusion proteins demonstrated that Myc-BBLF4 and Myc-BBLF2/3 plus BSLF1 bound to the Zta activation domain (amino acids 1 to 133). In the nuclear relocalization assay, the amino-terminal 25 amino acids of Zta were required for efficient interaction with the primase subcomplex but not for interaction with BBLF4. Evidence for interaction between oriLyt bound Zta and the helicase-primase complex was obtained in a superactivation assay using an oriLyt-chloramphenicol acetyltransferase (CAT) reporter. Zta activated expression from a CAT reporter containing the complete oriLyt region and regulated by the oriLyt BHLF1 promoter. Cotransfection of the helicase-primase proteins, one of which was fused to a heterologous activation domain, led to Zta-dependent superactivation of CAT expression. This assay also provided evidence for an interaction between the single-stranded DNA binding protein, BALF2, and the Zta-tethered helicase-primase complex. The helicase-primase interaction is consistent with a role for Zta in stabilizing the formation of an origin-bound replication complex.


Subject(s)
DNA Helicases/metabolism , DNA Primase/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/physiology , Trans-Activators/genetics , Trans-Activators/metabolism , Viral Proteins/genetics , Viral Proteins/metabolism , Animals , Antigens, Viral/metabolism , Base Sequence , Chloramphenicol O-Acetyltransferase/genetics , Chlorocebus aethiops , DNA Primers/genetics , DNA-Directed DNA Polymerase/metabolism , Genes, Reporter , Macromolecular Substances , Polymerase Chain Reaction , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Replication Origin , Sequence Deletion , Transcriptional Activation , Transfection , Vero Cells , Virus Replication
18.
Virology ; 236(1): 18-29, 1997 Sep 15.
Article in English | MEDLINE | ID: mdl-9299613

ABSTRACT

The Epstein-Barr virus (EBV) EBNA-1 protein has a central role in the maintenance of a latent EBV infection and is the only virus-encoded protein expressed in all EBV-associated tumors. EBNA-1 is required for replication of the episomal form of the latent viral genome and transactivates the latency C and LMP-1 promoters. The mechanisms by which EBNA-1 performs these functions are not known. Here we describe the cloning, expression, and characterization of a cellular protein, P32/TAP, which strongly interacts with EBNA-1. We show that P32/TAP is expressed at high levels in Raji cells and is synthesized as a proprotein of 282 amino acids (aa) that is posttranslationally processed by a two-step cleavage process to yield a mature protein of 209 aa. It has been previously reported that P32/TAP is expressed on the cell surface. Our transient expression assays detected full-length P32/TAP (1-282 aa) in the cytoplasm while mature P32/TAP protein localized to the nucleus. Three lines of evidence support P32/TAP interaction with EBNA-1. First, in the yeast two-hybrid system we mapped two interactive N-terminal regions of EBNA-1, aa 40-60 and aa 325-376, each of which contains arginine-glycine repeats. These regions interact with the C-terminal half of P32/TAP. Second, the full-length cytoplasmic P32/TAP protein can translocate nuclear EBNA-1 into the cytoplasm. Third, P32/TAP co-immunoprecipitated with EBNA-1. We have confirmed that a Gal4 fusion protein containing the C-terminal region of P32/TAP (aa 244-282) transactivates expression from a reporter containing upstream Gal4-binding sites. Deletion of the P32/TAP interactive regions of EBNA-1 severely diminished EBNA-1 transactivation of FRTKCAT in transient expression assays. Our data suggest that interaction with P32/TAP may contribute to EBNA-1-mediated transactivation.


Subject(s)
Epstein-Barr Virus Nuclear Antigens/metabolism , Herpesvirus 4, Human/physiology , Hyaluronan Receptors , Membrane Glycoproteins , Receptors, Complement/metabolism , Amino Acid Sequence , Animals , Arginine , Binding Sites , Carrier Proteins , Cell Line , Chloramphenicol O-Acetyltransferase/biosynthesis , Chlorocebus aethiops , Cloning, Molecular , Consensus Sequence , DNA Primers , DNA, Complementary , Epstein-Barr Virus Nuclear Antigens/biosynthesis , Epstein-Barr Virus Nuclear Antigens/chemistry , Gene Library , Genome, Viral , Glycine , Herpesvirus 4, Human/genetics , Humans , Lymphocytes/metabolism , Mice , Mitochondrial Proteins , Molecular Sequence Data , Polymerase Chain Reaction , Receptors, Complement/biosynthesis , Receptors, Complement/chemistry , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Saccharomyces cerevisiae , Sequence Alignment , Transfection
19.
J Virol ; 71(3): 1938-45, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9032325

ABSTRACT

EBNA2 is essential for immortalization of B cells by Epstein-Barr virus. EBNA2 is tethered to responsive promoters through a cellular factor, CBF1. CBF1 also binds to the activated form of mammalian Notch1, providing a linkage between EBNA2 function and Notch signalling. However, Notch2 is the predominant form expressed in spleen. The degree to which these Notch homologs are functionally convergent is not known. We present evidence that Notch2 also signals through CBF1. As is the case for Notch1, Notch2 interacted with the minimal repression domain of CBF1 and was targeted to CBF1 through the intracellular, subtransmembrane domain. Additional characterization suggested that the interaction domain of Notch may be bipartite. The intracellular domain of Notch2 (Notch2IC) located to the nucleus. This activated form of Notch2 transactivated expression of a target gene containing upstream CBF1 binding sites. The use of CBF1 mutants carrying amino acid substitutions in the transcriptional repression domain revealed that activation of gene expression by Notch2 is also based on masking of CBF1-mediated repression. Targeting of Notch1 and targeting of Notch2 were found to be identical and distinguishable from targeting by EBNA2. Mutation of CBF1 at codons 249 to 251 abolished interaction with both Notch proteins but not with EBNA2. In a biological examination of Notch2 function in muscle cells, Notch2IC activated endogenous HES-1 gene expression and blocked muscle cell differentiation. Overall, the data imply that at least a subset of the intracellular events following signalling in cells expressing Notch2 are common to those in Notch1-expressing cells. The concept that EBNA2 functions by mimicking Notch signalling is therefore viable whether cells are expressing Notch1 or Notch2.


Subject(s)
Avian Proteins , Cell Differentiation , Herpesvirus 4, Human/metabolism , Oncogene Proteins , Proto-Oncogene Proteins/metabolism , Receptors, Cell Surface/metabolism , Transcription Factors , Viral Proteins , Animals , Basic Helix-Loop-Helix Transcription Factors , Binding Sites , Biological Transport , Cell Line , Cell Nucleus/metabolism , Epstein-Barr Virus Nuclear Antigens/metabolism , Forkhead Transcription Factors , Gene Expression , Genes, Reporter , HeLa Cells , Homeodomain Proteins/genetics , Humans , Jurkat Cells , Luciferases/genetics , Membrane Proteins/metabolism , Mice , Proto-Oncogene Proteins/genetics , Rats , Receptor, Notch1 , Receptor, Notch2 , Receptors, Cell Surface/genetics , Signal Transduction/physiology , Structure-Activity Relationship , Transcription Factor HES-1 , Transcriptional Activation , Tumor Cells, Cultured , Up-Regulation
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...