Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Cancers (Basel) ; 10(3)2018 Mar 17.
Article in English | MEDLINE | ID: mdl-29562599

ABSTRACT

Epstein-Barr Virus (EBV) BamHI-A rightward frame 1 (BARF1) protein is considered a viral oncogene in epithelial cells and has immune-modulating properties. During viral lytic replication BARF1 is expressed as an early gene, regulated by the immediate early EBV protein R. However, in viral latency BARF1 is exclusively expressed in epithelial tumors such as nasopharyngeal (NPC) and gastric carcinoma (GC) but not in lymphomas, indicating that activation of the BARF1 promoter is cell type specific. Undifferentiated NPC is characterized by high expression of ΔNp63 isoforms of the epithelial differentiation marker p63, a member of the p53 family of transcription factors. Transcription factor binding site analysis indicated potential p53 family binding sites within the BARF1 promoter region. This study investigated ability of various p53 family members to transactivate the BARF1 promoter. Using BARF1 promoter luciferase reporter constructs we demonstrate that only p63 isoform ΔNp63α is capable of transactivating the BARF1 promoter, but not the TAp63 isoforms, p53 or p73. Direct promoter binding of ΔNp63α was confirmed by Chromatin Immune Precipitation (ChIP) analysis. Deletion mutants of the BARF1 promoter revealed multiple ΔNp63 response elements to be responsible for BARF1 promoter transactivation. However, ΔNp63α alone was not sufficient to induce BARF1 in tumor cells harboring full EBV genomes, indicating that additional cofactors might be required for full BARF1 regulation. In conclusion, in EBV positive NPC and GC, BARF1 expression might be induced by the epithelial differentiation marker ΔNp63α, explaining BARF1 expression in the absence of lytic reactivation.

2.
J Virol ; 87(18): 10126-38, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23843639

ABSTRACT

All eight human herpesviruses have a conserved herpesvirus protein kinase (CHPK) that is important for the lytic phase of the viral life cycle. In this study, we show that heat shock protein 90 (Hsp90) interacts directly with each of the eight CHPKs, and we demonstrate that an Hsp90 inhibitor drug, 17-dimethylaminoethylamino-17-demethoxygeldanamycin (17-DMAG), decreases expression of all eight CHPKs in transfected HeLa cells. 17-DMAG also decreases expression the of the endogenous Epstein-Barr virus protein kinase (EBV PK, encoded by the BGLF4 gene) in lytically infected EBV-positive cells and inhibits phosphorylation of several different known EBV PK target proteins. Furthermore, 17-DMAG treatment abrogates expression of the human cytomegalovirus (HCMV) kinase UL97 in HCMV-infected human fibroblasts. Importantly, 17-DMAG treatment decreased the EBV titer approximately 100-fold in lytically infected AGS-Akata cells without causing significant cellular toxicity during the same time frame. Increased EBV PK expression in 17-DMAG-treated AGS-Akata cells did not restore EBV titers, suggesting that 17-DMAG simultaneously targets multiple viral and/or cellular proteins required for efficient viral replication. These results suggest that Hsp90 inhibitors, including 17-DMAG, may be a promising group of drugs that could have profound antiviral effects on herpesviruses.


Subject(s)
Antiviral Agents/metabolism , Benzoquinones/metabolism , Enzyme Inhibitors/metabolism , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Herpesvirus 4, Human/physiology , Lactams, Macrocyclic/metabolism , Protein Kinases/metabolism , Virus Replication/drug effects , Cell Line , Epithelial Cells/drug effects , Epithelial Cells/virology , Fibroblasts/drug effects , Fibroblasts/virology , Herpesvirus 4, Human/drug effects , Humans , Protein Interaction Mapping , Viral Load , Virus Cultivation
3.
J Virol ; 87(14): 8053-63, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23678172

ABSTRACT

The latent-to-lytic switch of Epstein-Barr virus (EBV) is mediated by the immediate early protein BZLF1 (Z). However, the cellular factors regulating this process remain incompletely characterized. In this report, we show that the B-cell-specific transcription factor Pax5 helps to promote viral latency in B cells by blocking Z function. Although Z was previously shown to directly interact with Pax5 and inhibit its activity, the effect of Pax5 on Z function has not been investigated. Here, we demonstrate that Pax5 inhibits Z-mediated lytic viral gene expression and the release of infectious viral particles in latently infected epithelial cell lines. Conversely, we found that shRNA-mediated knockdown of endogenous Pax5 in a Burkitt lymphoma B-cell line leads to viral reactivation. Furthermore, we show that Pax5 reduces Z activation of early lytic viral promoters in reporter gene assays and inhibits Z binding to lytic viral promoters in vivo. We confirm that Pax5 and Z directly interact and show that this interaction requires the carboxy-terminal DNA-binding/dimerization domain of Z and the amino-terminal DNA-binding domain of Pax5. A Pax5 DNA-binding mutant (V26G/P80R) that interacts with Z retains the ability to inhibit Z function, whereas a Pax5 mutant (Δ106-110) that is deficient for interaction with Z does not inhibit Z-mediated lytic viral reactivation. Since the B-cell-specific transcription factor Oct-2 also directly interacts with Z and inhibits its function, these results suggest that EBV uses multiple redundant mechanisms to establish and maintain viral latency in B cells.


Subject(s)
Gene Expression Regulation, Viral/physiology , Herpesvirus 4, Human/physiology , PAX5 Transcription Factor/metabolism , Trans-Activators/metabolism , Virus Latency/physiology , Blotting, Western , Cell Line, Tumor , Chromatin Immunoprecipitation , DNA Primers/genetics , Electrophoretic Mobility Shift Assay , Genetic Vectors/genetics , Humans , Lentivirus , Plasmids/genetics , Reverse Transcriptase Polymerase Chain Reaction
4.
J Virol ; 87(2): 935-50, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23135711

ABSTRACT

The Epstein-Barr virus (EBV) immediate-early proteins BZLF1 and BRLF1 can both induce lytic EBV reactivation when overexpressed in latently infected cells. Although EBV genome methylation is required for BZLF1-mediated activation of lytic gene expression, the effect of viral genome methylation on BRLF1-mediated viral reactivation has not been well studied. Here, we have compared the effect of viral DNA methylation on BZLF1- versus BRLF1-mediated activation of lytic EBV gene transcription and viral genome replication. We show that most early lytic viral promoters are preferentially activated by BZLF1 in the methylated form, while methylation decreases the ability of BRLF1 to activate most early lytic promoters, as well as the BLRF2 late viral promoter. Moreover, methylation of bacmid constructs containing the EBV genome enhances BZLF1-mediated, but decreases BRLF1-mediated, early lytic gene expression. Methylation of viral promoter DNA does not affect BRLF1 binding to a variety of different CpG-containing BRLF1 binding motifs (RREs) in vitro or in vivo. However, BRLF1 preferentially induces H3K9 histone acetylation of unmethylated promoters in vivo. The methylated and unmethylated forms of an oriLyt-containing plasmid replicate with similar efficiency when transfected into EBV-positive cells that express the essential viral replication proteins in trans. Most importantly, we demonstrate that lytic viral gene expression and replication can be induced by BRLF1, but not BZLF1, expression in an EBV-positive telomerase-immortalized epithelial cell line (NOKs-Akata) in which lytic viral gene promoters remain largely unmethylated. These results suggest that the unmethylated form of the EBV genome can undergo viral reactivation and replication in a BRLF1-dependent manner.


Subject(s)
DNA, Viral/metabolism , Gene Expression Regulation, Viral , Genome, Viral , Herpesvirus 4, Human/physiology , Immediate-Early Proteins/metabolism , Trans-Activators/metabolism , Virus Activation , DNA Methylation , Herpesvirus 4, Human/genetics , Humans , Virus Replication
5.
J Virol ; 86(24): 13360-70, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23015717

ABSTRACT

The Epstein-Barr virus (EBV) latent-to-lytic switch is mediated by the viral proteins BZLF1 (Z), BRLF1 (R), and BRRF1 (Na). Since we previously showed that DNA-damaging agents (including chemotherapy and irradiation) can induce EBV lytic reactivation and recently demonstrated that wild-type p53 contributes to lytic reactivation, we investigated the role of the ATM kinase during EBV reactivation. ATM phosphorylates and activates p53, as well as numerous other substrates involved in the cellular DNA damage response. Using an ATM inhibitor (KU55933), we found that ATM activity is required for efficient induction of EBV lytic gene expression by a variety of different stimuli, including a histone deacetylase (HDAC) inhibitor, the transforming growth factor ß (TGF-ß) cytokine, a demethylating agent (5-azacytidine), B cell receptor engagement with anti-IgG antibody, hydrogen peroxide, and the proteosome inhibitor bortezomib. In EBV-infected AGS (gastric) cells, knockdown of ATM, or p53, expression inhibits EBV reactivation. Conversely, treatment of these cells with nutlin-3 (which activates p53 and ATM) robustly induces lytic reactivation in a p53- and ATM-dependent manner. The ability of the EBV R and Na proteins to induce lytic reactivation in EBV-infected AGS cells is ATM dependent. However, overexpression of Z induces lytic gene expression in the presence or absence of ATM activity. Our results suggest that ATM enhances Z promoter activity in the context of the intact EBV genome and that p53 contributes to the ATM effect. Nevertheless, since we found that ATM inhibitors also reduce lytic reactivation in Burkitt lymphoma cells that have no p53, additional ATM substrates must also contribute to the ATM effect.


Subject(s)
Cell Cycle Proteins/metabolism , DNA-Binding Proteins/metabolism , Herpesvirus 4, Human/physiology , Phosphatidylinositol 3-Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/metabolism , Virus Activation , Ataxia Telangiectasia Mutated Proteins , Base Sequence , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/genetics , Cell Line , DNA Primers , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , Humans , Imidazoles/metabolism , Morpholines/pharmacology , Phosphatidylinositol 3-Kinases/genetics , Phosphoinositide-3 Kinase Inhibitors , Piperazines/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Pyrones/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Tumor Suppressor Protein p53/physiology , Tumor Suppressor Proteins/antagonists & inhibitors , Tumor Suppressor Proteins/genetics
6.
J Virol ; 86(4): 2011-20, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22171252

ABSTRACT

The human cytomegalovirus tegument protein UL69 has been shown to be required for efficient viral replication at low multiplicities of infection. Several functions have been associated with UL69, including its ability to regulate cell cycle progression, translation, and the export of viral transcripts from the nucleus to the cytoplasm. However, it remains unclear which, if any, of these activities contribute to the phenotype observed with the UL69 deletion mutant. UL69 has been shown to interact with the cellular protein SPT6. The functional significance of this interaction has never been examined in the context of an infection. To address this, we generated UL69 mutant viruses that were unable to interact with SPT6 and determined what effect these mutations had on virus replication. Abolishing UL69's ability to interact with the SPT6 protein inhibited virus replication to levels indistinguishable from those observed following infection with the UL69 deletion mutant. Surprisingly, abolishing UL69's interaction with SPT6 also resulted in the impairment of UL69 shuttling activity. Finally, we demonstrate that inhibition of SPT6 expression by short hairpin RNA (shRNA) knockdown inhibits wild-type virus replication. Taken together, our results demonstrate that UL69's ability to interact with SPT6 plays a critical role in viral replication.


Subject(s)
Cytomegalovirus Infections/metabolism , Cytomegalovirus/physiology , Transcription Factors/metabolism , Virus Replication , Cell Line , Cytomegalovirus/genetics , Cytomegalovirus Infections/genetics , Cytomegalovirus Infections/virology , Gene Expression Regulation, Viral , Humans , Protein Binding , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors/genetics
7.
J Virol ; 85(17): 8940-53, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21697476

ABSTRACT

The Epstein-Barr virus (EBV) latent-to-lytic switch is an essential part of the viral life cycle, but the cellular factors that promote viral reactivation are not well defined. In this report, we demonstrate that the cellular transcription factor Oct-1 cooperates with the EBV immediate-early protein BRLF1 (R, Rta) to induce lytic viral reactivation. We show that cotransfected Oct-1 enhances the ability of BRLF1 to activate lytic gene expression in 293 cells stably infected with a BRLF1-defective EBV mutant (BRLF1-stop) and that Oct-1 increases BRLF1-mediated activation of lytic EBV promoters in reporter gene assays. We find that Oct-1 interacts directly with BRLF1 in vitro and that a mutant BRLF1 protein (the M140A mutant) attenuated for the ability to interact with Oct-1 in vitro is also resistant to Oct-1-mediated transcriptional enhancement in 293 BRLF1-stop cells. Furthermore, we show that cotransfected Oct-1 augments BRLF1 binding to a variety of lytic EBV promoters in chromatin immunoprecipitation (ChIP) assays (including the BZLF1, BMRF1, and SM promoters) and that BRLF1 tethers Oct-1 to lytic EBV promoters. In addition, we demonstrate that an Oct-1 mutant defective in DNA binding (the S335D mutant) still retains the ability to enhance BRLF1 transcriptional effects. Finally, we show that knockdown of endogenous Oct-1 expression reduces the level of constitutive lytic EBV gene expression in both EBV-positive B-cell and EBV-positive epithelial cell lines. These results suggest that Oct-1 acts as a positive regulator of EBV lytic gene expression and that this effect is at least partially mediated through its interaction with the viral protein BRLF1.


Subject(s)
Herpesvirus 4, Human/pathogenicity , Host-Pathogen Interactions , Immediate-Early Proteins/metabolism , Octamer Transcription Factor-1/metabolism , Trans-Activators/metabolism , Virus Activation , Virus Latency , Amino Acid Substitution/genetics , Cell Line , Chromatin Immunoprecipitation , DNA, Viral/metabolism , Humans , Immediate-Early Proteins/genetics , Mutant Proteins/genetics , Mutant Proteins/metabolism , Mutation, Missense , Promoter Regions, Genetic , Protein Binding , Protein Interaction Mapping , Trans-Activators/genetics
8.
J Virol ; 85(9): 4318-29, 2011 May.
Article in English | MEDLINE | ID: mdl-21325409

ABSTRACT

The Epstein-Barr virus (EBV) BRRF1 lytic gene product (Na) is encoded within the same immediate-early region as the BZLF1 (Z) and BRLF1(R) gene products, but its role during EBV infection has not been well defined. We previously showed that Na cooperates with the R protein to induce lytic gene expression in latently infected EBV-positive 293 cells, and in some EBV-negative cell lines it can activate the Z promoter in reporter gene assays. Here we show that overexpression of Na alone is sufficient to induce lytic gene expression in several different latently infected epithelial cell lines (Hone-Akata, CNE2-Akata, and AGS-Akata), while knockdown of endogenous Na expression reduces lytic gene expression. Consistent with its ability to interact with tumor necrosis factor receptor-associated factor 2 (TRAF2) in a yeast two-hybrid assay, we demonstrate that Na interacts with TRAF2 in cells. Furthermore, we show that TRAF2 is required for Na induction of lytic gene expression, that Na induces Jun N-terminal protein kinase (JNK) activation in a TRAF2-dependent manner, and that a JNK inhibitor abolishes the ability of Na to disrupt viral latency. Additionally, we show that Na and the tumor suppressor protein p53 cooperate to induce lytic gene expression in epithelial cells (including the C666-1 nasopharyngeal carcinoma cell line), although Na does not appear to affect p53 function. Together these data suggest that Na plays an important role in regulating the switch between latent and lytic infection in epithelial cells and that this effect requires both the TRAF2 and p53 cellular proteins.


Subject(s)
Herpesvirus 4, Human/growth & development , Herpesvirus 4, Human/pathogenicity , Protein Interaction Mapping , TNF Receptor-Associated Factor 2/metabolism , Tumor Suppressor Protein p53/metabolism , Cell Line , Cell Survival , Humans , Protein Binding
9.
J Virol ; 84(18): 9649-54, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20610707

ABSTRACT

The human cytomegalovirus (HCMV) tegument protein UL69 is important for efficient viral replication at low multiplicities of infection. Several molecular mechanisms by which UL69 contributes to HCMV replication have been proposed, including UL69's ability to interact with the mRNA export factors UAP56 and URH49 to facilitate the shuttling of viral mRNAs from the nuclei of infected cells. Using a UL69 viral mutant that is unable to bind UAP56 and URH49, we demonstrated that UL69's interaction with UAP56 or URH49 does not contribute to the growth phenotype associated with the UL69 deletion mutant.


Subject(s)
Cytomegalovirus/physiology , DEAD-box RNA Helicases/metabolism , Host-Pathogen Interactions , Trans-Activators/metabolism , Virus Replication , Animals , Cell Line , Gene Deletion , Humans , Mice , Mutant Proteins/metabolism , Protein Binding , Trans-Activators/genetics
10.
J Virol ; 84(9): 4524-33, 2010 May.
Article in English | MEDLINE | ID: mdl-20147387

ABSTRACT

The Epstein-Barr virus (EBV)-encoded viral protein kinase, EBV-PK (the BGLF4 gene product), is required for efficient nuclear viral egress in 293 cells. However, since EBV-PK phosphorylates a number of different viral and cellular proteins (including lamin A/C), the relative importance of each target during lytic viral replication remains unclear. We show here that an EBV PK mutant (PKmut; containing stop codons at residues 1 and 5 in EBV-PK) is highly defective for release of infectious virus from 293 cells but not 293T cells. Furthermore, the phenotype of the PKmut in 293 cells is substantially reversed by expression of the simian virus 40 (SV40) large (T) and small (t) T antigens. Efficient rescue requires the presence of both SV40 T/t proteins. We show that 293T cells have a much higher level of constitutive lamin A/C phosphorylation than do 293 cells over residues (S22 and S392) that promote phosphorylation-dependent nuclear disassembly and that both large T and small t contribute to enhanced lamin A/C phosphorylation. Finally, we demonstrate that knockdown of lamin A/C expression using small interfering RNA also rescues the PKmut phenotype in 293 cells. These results suggest that essential roles of EBV-PK during lytic viral replication include the phosphorylation and dispersion of lamin A/C.


Subject(s)
Antigens, Polyomavirus Transforming/biosynthesis , Herpesvirus 4, Human/growth & development , Host-Pathogen Interactions , Lamin Type A/antagonists & inhibitors , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/physiology , Viral Proteins/physiology , Cell Line , Gene Knockdown Techniques , Herpesvirus 4, Human/genetics , Humans , Lamin Type A/metabolism , Phosphorylation , RNA, Small Interfering/genetics , Simian virus 40
11.
Proc Natl Acad Sci U S A ; 107(7): 3146-51, 2010 Feb 16.
Article in English | MEDLINE | ID: mdl-20133771

ABSTRACT

EBV causes infectious mononucleosis and is associated with certain malignancies. EBV nuclear antigen 1 (EBNA1) mediates EBV genome replication, partition, and transcription, and is essential for persistence of the viral genome in host cells. Here we demonstrate that Hsp90 inhibitors decrease EBNA1 expression and translation, and that this effect requires the Gly-Ala repeat domain of EBNA1. Hsp90 inhibitors induce the death of established, EBV-transformed lymphoblastoid cell lines at doses nontoxic to normal cells, and this effect is substantially reversed when lymphoblastoid cell lines are stably infected with a retrovirus expressing a functional EBNA1 mutant lacking the Gly-Ala repeats. Hsp90 inhibitors prevent EBV transformation of primary B cells, and strongly inhibit the growth of EBV-induced lymphoproliferative disease in SCID mice. These results suggest that Hsp90 inhibitors may be particularly effective for treating EBV-induced diseases requiring the continued presence of the viral genome.


Subject(s)
Benzoquinones/pharmacology , Epstein-Barr Virus Nuclear Antigens/metabolism , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Herpesvirus 4, Human , Lactams, Macrocyclic/pharmacology , Lymphoproliferative Disorders/drug therapy , Animals , Apoptosis/drug effects , Benzoquinones/therapeutic use , Cell Line, Tumor , DNA Primers/genetics , Dipeptides/genetics , Gene Expression Regulation, Viral/drug effects , Humans , Immunoblotting , Immunoprecipitation , Lactams, Macrocyclic/therapeutic use , Lymphoproliferative Disorders/virology , Mice , Mice, SCID , Reverse Transcriptase Polymerase Chain Reaction , Virus Replication/drug effects
12.
J Virol ; 84(9): 4383-94, 2010 May.
Article in English | MEDLINE | ID: mdl-20181712

ABSTRACT

The Epstein-Barr virus (EBV) immediate-early protein BZLF1 (Z) mediates the switch between latent and lytic EBV infection. Z not only activates early lytic viral gene transcription but also plays a direct role in lytic viral genome replication. Although a small fraction of Z is known to be sumoylated, the effects of this posttranslational modification on various different Z functions have not been well defined. In this report, we show that only the lysine at amino acid residue 12 is required for the sumoylation of Z, and that Z can be sumoylated by SUMO isoforms 1, 2, and 3. We also demonstrate that the sumo-defective Z mutants ZK12A and ZK12R have enhanced transcriptional activity. The sumoylated and nonsumoylated forms of Z were found to have a similar cellular location, both being localized primarily within the nuclear matrix. The Z sumo-defective mutants were, however, partially defective for disrupting promyelocytic leukemia (PML) bodies compared to the ability of wild-type Z. In addition, we show that lytic viral genome replication does not require the sumoylation of Z, although a Z mutant altered at both amino acids 12 and 13 is replication defective. Furthermore, we show that the sumoylation of Z is greatly increased (from less than 1 to about 11%) in lytically induced 293 cells infected with an EBV mutant virus deleted for the EBV-encoded protein kinase (EBV-PK) compared to that of 293 cells infected with wild-type EBV, and that the overexpression of EBV-PK leads to the reduced sumoylation of Z in EBV-negative cells. Our results suggest that the sumoylation of Z helps to promote viral latency, and that EBV-PK inhibits Z sumoylation during viral reactivation.


Subject(s)
Gene Expression Regulation, Viral , Herpesvirus 4, Human/physiology , Protein Kinases/metabolism , Protein Processing, Post-Translational , Trans-Activators/metabolism , Transcription, Genetic , Viral Proteins/metabolism , B-Lymphocytes/virology , Cell Line , Epithelial Cells/virology , Humans , Small Ubiquitin-Related Modifier Proteins/metabolism , Virus Activation , Virus Latency , Virus Replication
13.
J Virol ; 84(9): 4534-42, 2010 May.
Article in English | MEDLINE | ID: mdl-20181711

ABSTRACT

Ganciclovir (GCV) and acyclovir (ACV) are guanine nucleoside analogues that inhibit lytic herpesvirus replication. GCV and ACV must be monophosphorylated by virally encoded enzymes to be converted into nucleotides and incorporated into viral DNA. However, whether GCV and/or ACV phosphorylation in Epstein-Barr virus (EBV)-infected cells is mediated primarily by the EBV-encoded protein kinase (EBV-PK), the EBV-encoded thymidine kinase (EBV-TK), or both is controversial. To examine this question, we constructed EBV mutants containing stop codons in either the EBV-PK or EBV-TK open reading frame and selected for stable 293T clones latently infected with wild-type EBV or each of the mutant viruses. Cells were induced to the lytic form of viral replication with a BZLF1 expression vector in the presence and absence of various doses of GCV and ACV, and infectious viral titers were determined by a green Raji cell assay. As expected, virus production in wild-type EBV-infected 293T cells was inhibited by both GCV (50% inhibitory concentration [IC(50)] = 1.5 microM) and ACV (IC(50) = 4.1 microM). However, the EBV-PK mutant (which replicates as well as the wild-type (WT) virus in 293T cells) was resistant to both GCV (IC(50) = 19.6 microM) and ACV (IC(50) = 36.4 microM). Expression of the EBV-PK protein in trans restored GCV and ACV sensitivity in cells infected with the PK mutant virus. In contrast, in 293T cells infected with the TK mutant virus, viral replication remained sensitive to both GCV (IC(50) = 1.2 microM) and ACV (IC(50) = 2.8 microM), although susceptibility to the thymine nucleoside analogue, bromodeoxyuridine, was reduced. Thus, EBV-PK but not EBV-TK mediates ACV and GCV susceptibilities.


Subject(s)
Acyclovir/pharmacology , Antiviral Agents/pharmacology , Ganciclovir/pharmacology , Herpesvirus 4, Human/drug effects , Protein Serine-Threonine Kinases/metabolism , Thymidine Kinase/metabolism , Viral Proteins/metabolism , Acyclovir/metabolism , Antiviral Agents/metabolism , Cell Line , Codon, Nonsense , Ganciclovir/metabolism , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/growth & development , Humans , Inhibitory Concentration 50 , Mutagenesis, Site-Directed , Phosphorylation , Protein Serine-Threonine Kinases/deficiency , Thymidine Kinase/deficiency , Viral Proteins/genetics , Virus Replication/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...