Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
J Virol ; 83(20): 10694-709, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19656890

ABSTRACT

Epstein-Barr virus (EBV) can be reactivated from latency into the lytic cycle by many stimuli believed to operate by different mechanisms. Cell lines containing EBV differ in their responses to inducing stimuli, yet all stimuli require de novo protein synthesis (44). A crucial step preliminary to identifying these proteins and determining when they are required is to measure the duration of stimulus and response time needed for activation of expression of EBV BRLF1 and BZLF1, the earliest viral indicators of reactivation. Here we show, with four EBV-containing cell lines that respond to different inducing agents, that stimuli that are effective at reactivating EBV can be divided into two main groups. The histone deacetylase inhibitors sodium butyrate and trichostatin A require a relatively long period of exposure, from 2 to 4 h or longer. Phorbol esters, anti-immunoglobulin G (anti-IgG), and, surprisingly, 5-aza-2'-deoxycytidine require short exposures of 15 min or less. The cell/virus background influences the response time. Expression of the EBV BZLF1 and BRLF1 genes can be detected before 2 h in Akata cells treated with anti-IgG, but both long- and short-duration stimuli required 4 or more hr to activate BZLF1 and BRLF1 expression in HH514-16, Raji, or B95-8 cells. Thus, stimulus duration and response time are independent variables. Neither stimulus duration nor response time can be predicted by the number of cells activated into the lytic cycle. These experiments shed new light on the earliest events leading to lytic cycle reactivation of EBV.


Subject(s)
Antibodies, Anti-Idiotypic/pharmacology , Enzyme Inhibitors/pharmacology , Herpesvirus 4, Human/physiology , Histone Deacetylase Inhibitors , Lymphocytes/virology , Phorbol Esters/pharmacology , Virus Activation/drug effects , Cell Line , Herpesvirus 4, Human/metabolism , Histone Deacetylases/pharmacology , Humans , Immediate-Early Proteins/genetics , Immediate-Early Proteins/metabolism , Time Factors , Trans-Activators/genetics , Trans-Activators/metabolism , Viral Proteins/genetics , Viral Proteins/metabolism
2.
J Virol ; 82(10): 4706-19, 2008 May.
Article in English | MEDLINE | ID: mdl-18337569

ABSTRACT

Activation of the Epstein-Barr virus (EBV) lytic cycle is mediated through the combined actions of ZEBRA and Rta, the products of the viral BZLF1 and BRLF1 genes. During latency, these two genes are tightly repressed. Histone deacetylase inhibitors (HDACi) can activate viral lytic gene expression. Therefore, a widely held hypothesis is that Zp and Rp, the promoters for BZLF1 and BRLF1, are repressed by chromatin and that hyperacetylation of histone tails, by allowing the access of positively acting factors, leads to transcription of BZLF1 and BRLF1. To investigate this hypothesis, we used chromatin immunoprecipitation (ChIP) to examine the acetylation and phosphorylation states of histones H3 and H4 on Zp and Rp in three cell lines, Raji, B95-8, and HH514-16, which differ in their response to EBV lytic induction by HDACi. We studied the effects of three HDACi, sodium butyrate (NaB), trichostatin A (TSA), and valproic acid (VPA). We also examined the effects of tetradecanoyl phorbol acetate (TPA) and 5-aza-2'-deoxycytidine, a DNA methyltransferase inhibitor, on histone modification. In Raji cells, TPA and NaB act synergistically to activate the EBV lytic cycle and promote an increase in histone H3 and H4 acetylation and phosphorylation at Zp and Rp. Surprisingly, however, when Raji cells were treated with NaB or TSA, neither of which is sufficient to activate the lytic cycle, an increase of comparable magnitude of hyperacetylated and phosphorylated histone H3 at Zp and Rp was observed. In B95-8 cells, NaB inhibited lytic induction by TPA, yet NaB promoted hyperacetylation of H3 and H4. In HH514-16 cells, NaB and TSA strongly activated the EBV lytic cycle and caused hyperacetylation of histone H3 on Zp and Rp. However, when HH514-16 cells were treated with VPA, lytic cycle mRNAs or proteins were not induced, although histone H3 was hyperacetylated as measured by immunoblotting or by ChIP on Zp and Rp. Taken together, our data suggest that open chromatin at EBV BZLF1 and BRLF1 promoters is not sufficient to activate EBV lytic cycle gene expression.


Subject(s)
DNA, Viral/metabolism , Herpesvirus 4, Human/physiology , Histone Deacetylase Inhibitors , Histones/metabolism , Promoter Regions, Genetic , Virus Activation/drug effects , Virus Latency/physiology , Acetylation , Animals , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Butyrates/pharmacology , Callithrix , Cell Line , Chromatin Immunoprecipitation , DNA Modification Methylases/antagonists & inhibitors , Decitabine , Enzyme Inhibitors/pharmacology , Humans , Hydroxamic Acids/pharmacology , Phosphorylation , Protein Binding , Tetradecanoylphorbol Acetate/pharmacology , Valproic Acid/pharmacology
3.
Adv Cancer Res ; 97: 81-109, 2007.
Article in English | MEDLINE | ID: mdl-17419942

ABSTRACT

The seminal experiments of George and Eva Klein helped to define the two life cycles of Epstein-Barr Virus (EBV), namely latency and lytic or productive infection. Their laboratories described latent nuclear antigens expressed during latency and discovered several chemicals that activated the viral lytic cycle. The mechanism of the switch between latency and the lytic cycle of EBV and Kaposi's sarcoma-associated herpesvirus (KSHV) can be studied in cultured B cell lines. Lytic cycle activation of EBV is controlled by two viral transcription factors, ZEBRA and Rta. The homologue of Rta encoded in ORF50 is the lytic cycle activator of KSHV. Control of the lytic cycle can be divided into two distinct phases. Upstream events control expression of the virally encoded lytic cycle activator genes. Downstream events represent tasks carried out by the viral proteins in driving expression of lytic cycle genes and lytic viral DNA replication. In this chapter, we report three recent groups of experiments relating to upstream and downstream events. Azacytidine (AzaC) is a DNA methyltransferase inhibitor whose lytic cycle activation capacity was discovered by G. Klein and coworkers. We find that AzaC rapidly activates the EBV lytic cycle but does not detectably alter DNA methylation or histone acetylation on the promoters of the EBV lytic cycle activator genes. AzaC probably acts via a novel, yet to be elucidated, mechanism. The lytic cycle of both EBV and KSHV can be activated by sodium butyrate (NaB), a histone deacetylase inhibitor whose activity in disrupting latency was also discovered by G. Klein and coworkers. Activation of EBV by NaB requires protein synthesis; activation of KSHV is independent of protein synthesis. Thus, NaB works by a different pathway on the two closely related viruses. ZEBRA, the major downstream mediator of EBV lytic cycle activation is both a transcription activator and an essential replication protein. We show that phosphorylation of ZEBRA at its casein kinase 2 (CK2) site separates these two functions. Phosphorylation by CK2 is required for ZEBRA to activate lytic replication but not to induce expression of early lytic cycle genes. We discuss a number of unsolved mysteries about lytic cycle activation which should provide fertile territory for future research.


Subject(s)
Cytopathogenic Effect, Viral/physiology , Gene Expression Regulation, Viral/genetics , Herpesvirus 4, Human/physiology , Herpesvirus 8, Human/physiology , Oncogenic Viruses/physiology , Azacitidine/pharmacology , Cycloheximide/pharmacology , Cytopathogenic Effect, Viral/drug effects , Cytopathogenic Effect, Viral/genetics , DNA Replication , DNA, Viral/biosynthesis , DNA, Viral/genetics , Epstein-Barr Virus Infections/virology , Gene Expression Regulation, Viral/drug effects , Herpesviridae Infections/virology , Herpesvirus 4, Human/drug effects , Herpesvirus 4, Human/genetics , Herpesvirus 8, Human/drug effects , Herpesvirus 8, Human/genetics , Humans , Immediate-Early Proteins/physiology , Mutation , Phosphorylation , Protein Processing, Post-Translational , Protein Structure, Tertiary , Trans-Activators/chemistry , Trans-Activators/genetics , Trans-Activators/physiology , Tumor Virus Infections/virology , Virus Latency/drug effects , Virus Latency/genetics
4.
J Virol ; 80(18): 9115-33, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16940523

ABSTRACT

The ZEBRA protein of Epstein-Barr virus (EBV) drives the viral lytic cycle cascade. The capacity of ZEBRA to recognize specific DNA sequences resides in amino acids 178 to 194, a region in which 9 of 17 residues are either lysine or arginine. To define the basic domain residues essential for activity, a series of 46 single-amino-acid-substitution mutants were examined for their ability to bind ZIIIB DNA, a high-affinity ZEBRA binding site, and for their capacity to activate early and late EBV lytic cycle gene expression. DNA binding was obligatory for the protein to activate the lytic cascade. Nineteen mutants that failed to bind DNA were unable to disrupt latency. A single acidic replacement of a basic amino acid destroyed DNA binding and the biologic activity of the protein. Four mutants that bound weakly to DNA were defective at stimulating the expression of Rta, the essential first target of ZEBRA in lytic cycle activation. Four amino acids, R183, A185, C189, and R190, are likely to contact ZIIIB DNA specifically, since alanine or valine substitutions at these positions drastically weakened or eliminated DNA binding. Twenty-three mutants were proficient in binding to ZIIIB DNA. Some DNA binding-proficient mutants were refractory to supershift by BZ-1 monoclonal antibody (epitope amino acids 214 to 230), likely as the result of the increased solubility of the mutants. Mutants competent to bind DNA could be separated into four functional groups: the wild-type group (eight mutants), a group defective at activating Rta (five mutants, all with mutations at the S186 site), a group defective at activating EA-D (three mutants with the R179A, S186T, and K192A mutations), and a group specifically defective at activating late gene expression (seven mutants). Three late mutants, with a Y180A, Y180E, or K188A mutation, were defective at stimulating EBV DNA replication. This catalogue of point mutants reveals that basic domain amino acids play distinct functions in binding to DNA, in activating Rta, in stimulating early lytic gene expression, and in promoting viral DNA replication and viral late gene expression. These results are discussed in relationship to the recently solved crystal structure of ZEBRA bound to an AP-1 site.


Subject(s)
DNA Replication , DNA, Viral , DNA-Binding Proteins/chemistry , Gene Expression Regulation, Viral , Herpesvirus 4, Human/metabolism , Trans-Activators/chemistry , Viral Proteins/chemistry , Base Sequence , DNA/chemistry , DNA-Binding Proteins/metabolism , Epitopes/chemistry , Humans , Molecular Sequence Data , Mutation , Protein Binding , Protein Structure, Tertiary , Trans-Activators/metabolism , Viral Proteins/metabolism
5.
J Biol Chem ; 281(6): 3085-95, 2006 Feb 10.
Article in English | MEDLINE | ID: mdl-16321978

ABSTRACT

ZEBRA, the product of the Epstein-Barr virus gene bzlf1, and a member of the AP-1 subfamily of basic zipper (bZIP) transcription factors, is necessary and sufficient to disrupt viral latency and to initiate the viral lytic cycle. Two serine residues of ZEBRA, Ser167 and Ser173, are substrates for casein kinase 2 (CK2) and are constitutively phosphorylated in vivo. Phosphorylation of ZEBRA at its CK2 sites is required for proper temporal regulation of viral gene expression. Phosphopeptide analysis indicated that ZEBRA contains additional constitutive phosphorylation sites. Here we employed a co-migration strategy to map these sites in vivo. The cornerstone of this strategy was to correlate the migration of 32P- and 35S-labeled tryptic peptides of ZEBRA. The identity of the peptides was revealed by mutagenesis of methionine and cysteine residues present in each peptide. Phosphorylation sites within the peptide were identified by mutagenesis of serines and threonines. ZEBRA was shown to be phosphorylated at serine and threonine residues, but not tyrosine. Two previously unrecognized phosphorylation sites of ZEBRA were identified in the NH2-terminal region of the transactivation domain: a cluster of weak phosphorylation sites at Ser6, Thr7, and Ser8 and a strong phosphorylation site at Thr14. Thr14 was embedded in a MAP kinase consensus sequence and could be phosphorylated in vitro by JNK, despite the absence of a canonical JNK docking site. Thus ZEBRA is now known to be constitutively phosphorylated at three distinct sites.


Subject(s)
DNA-Binding Proteins/physiology , Herpesvirus 4, Human/metabolism , Trans-Activators/physiology , Viral Proteins/physiology , Binding Sites , Biochemistry/methods , Cell Line, Tumor , Cell Movement , Cysteine/chemistry , DNA-Binding Proteins/chemistry , Escherichia coli/metabolism , Genetic Vectors , Humans , Immunoprecipitation , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Signaling System , Methionine/chemistry , Mutagenesis , Peptides/chemistry , Phosphorylation , Protein Structure, Tertiary , Serine/chemistry , Threonine/chemistry , Trans-Activators/chemistry , Transfection , Trypsin/chemistry , Tyrosine/chemistry , Viral Proteins/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...