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










Publication year range
1.
Mol Cell ; 54(1): 67-79, 2014 Apr 10.
Article in English | MEDLINE | ID: mdl-24725595

ABSTRACT

In marmoset T cells transformed by Herpesvirus saimiri (HVS), a viral U-rich noncoding (nc) RNA, HSUR 1, specifically mediates degradation of host microRNA-27 (miR-27). High-throughput sequencing of RNA after crosslinking immunoprecipitation (HITS-CLIP) identified mRNAs targeted by miR-27 as enriched in the T cell receptor (TCR) signaling pathway, including GRB2. Accordingly, transfection of miR-27 into human T cells attenuates TCR-induced activation of mitogen-activated protein kinases (MAPKs) and induction of CD69. MiR-27 also robustly regulates SEMA7A and IFN-γ, key modulators and effectors of T cell function. Knockdown or ectopic expression of HSUR 1 alters levels of these proteins in virally transformed cells. Two other T-lymphotropic γ-herpesviruses, AlHV-1 and OvHV-2, do not produce a noncoding RNA to downregulate miR-27 but instead encode homologs of miR-27 target genes. Thus, oncogenic γ-herpesviruses have evolved diverse strategies to converge on common targets in host T cells.


Subject(s)
Herpesvirus 2, Saimiriine/metabolism , Lymphocyte Activation , MicroRNAs/metabolism , RNA, Untranslated/metabolism , RNA, Viral/metabolism , T-Lymphocytes/metabolism , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Antigens, Differentiation, T-Lymphocyte/metabolism , Base Sequence , Callithrix , Enzyme Activation , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , GRB2 Adaptor Protein/genetics , GRB2 Adaptor Protein/metabolism , Gene Expression Regulation , HEK293 Cells , Herpesvirus 2, Saimiriine/genetics , Herpesvirus 2, Saimiriine/pathogenicity , High-Throughput Nucleotide Sequencing , Host-Pathogen Interactions , Humans , Immunoprecipitation , Interferon-gamma/genetics , Interferon-gamma/metabolism , Jurkat Cells , Lectins, C-Type/metabolism , MicroRNAs/genetics , Mitogen-Activated Protein Kinases/metabolism , Molecular Sequence Data , RNA Stability , RNA, Untranslated/genetics , RNA, Viral/genetics , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Semaphorins/genetics , Semaphorins/metabolism , Sequence Analysis, RNA , Signal Transduction , T-Lymphocytes/immunology , T-Lymphocytes/virology , Time Factors , Transfection
2.
PLoS Pathog ; 10(2): e1003907, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24550725

ABSTRACT

The essential herpesvirus adaptor protein HVS ORF57, which has homologs in all other herpesviruses, promotes viral mRNA export by utilizing the cellular mRNA export machinery. ORF57 protein specifically recognizes viral mRNA transcripts, and binds to proteins of the cellular transcription-export (TREX) complex, in particular ALYREF. This interaction introduces viral mRNA to the NXF1 pathway, subsequently directing it to the nuclear pore for export to the cytoplasm. Here we have used a range of techniques to reveal the sites for direct contact between RNA and ORF57 in the absence and presence of ALYREF. A binding site within ORF57 was characterized which recognizes specific viral mRNA motifs. When ALYREF is present, part of this ORF57 RNA binding site, composed of an α-helix, binds preferentially to ALYREF. This competitively displaces viral RNA from the α-helix, but contact with RNA is still maintained by a flanking region. At the same time, the flexible N-terminal domain of ALYREF comes into contact with the viral RNA, which becomes engaged in an extensive network of synergistic interactions with both ALYREF and ORF57. Transfer of RNA to ALYREF in the ternary complex, and involvement of individual ORF57 residues in RNA recognition, were confirmed by UV cross-linking and mutagenesis. The atomic-resolution structure of the ORF57-ALYREF interface was determined, which noticeably differed from the homologous ICP27-ALYREF structure. Together, the data provides the first site-specific description of how viral mRNA is locked by a herpes viral adaptor protein in complex with cellular ALYREF, giving herpesvirus access to the cellular mRNA export machinery. The NMR strategy used may be more generally applicable to the study of fuzzy protein-protein-RNA complexes which involve flexible polypeptide regions.


Subject(s)
Herpesviridae Infections/metabolism , Host-Parasite Interactions/physiology , Nuclear Proteins/metabolism , RNA, Viral/metabolism , RNA-Binding Proteins/metabolism , Repressor Proteins/metabolism , Trans-Activators/metabolism , Transcription Factors/metabolism , Tumor Virus Infections/metabolism , Active Transport, Cell Nucleus/physiology , Herpesvirus 2, Saimiriine/chemistry , Herpesvirus 2, Saimiriine/metabolism , Herpesvirus 2, Saimiriine/pathogenicity , Humans , Nuclear Proteins/chemistry , Protein Structure, Quaternary , RNA Transport/physiology , RNA, Viral/analysis , RNA-Binding Proteins/chemistry , Repressor Proteins/chemistry , Trans-Activators/chemistry , Transcription Factors/chemistry
3.
Virus Res ; 165(2): 179-89, 2012 May.
Article in English | MEDLINE | ID: mdl-22374337

ABSTRACT

The potential of Herpesvirus saimiri (HVS) subgroups A, B and C and Herpesvirus ateles (HVA) to transform primary T cells to permanent growth in vitro is restricted by the primate host species and by viral variability represented by distinct viral oncoproteins. We now addressed the relation between the transforming potential of the different viruses and the signaling pathways activated by transiently expressed oncoproteins. Marmoset lymphocytes were transformed by all HVS subgroups as well as HVA, while transformation of human cells was restricted to HVS-C and, unexpectedly, HVA. NF-κB and Src-family kinase (SFK) activity was required for survival of all transformed lymphocytes. Accordingly, NF-κB was induced by oncoproteins of all viruses. In contrast, SFK-related signaling was detectable only for oncoproteins of HVS-C and HVA. Thus, the restricted transformation of human lymphocytes likely correlates with the specific SFK targeting by these oncoproteins. These results will enable further studies into novel SFK effector mechanisms relevant for T-cell proliferation.


Subject(s)
Herpesvirus 2, Saimiriine/pathogenicity , Lymphocyte Activation , Oncogene Proteins/metabolism , Rhadinovirus/pathogenicity , Signal Transduction , Animals , Callithrix , Cells, Cultured , Humans , Primates
4.
J Gen Virol ; 93(Pt 2): 330-340, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22012462

ABSTRACT

Herpesvirus saimiri (HVS), a T-lymphotropic monkey herpesvirus, induces fulminant T-cell lymphoma in non-natural primate hosts. In addition, it can immortalize human T-cells in vitro. HVS tyrosine kinase-interacting protein (Tip) is an essential viral gene required for T-cell transformation both in vitro and in vivo. In this study, we found that Tip interacts with the STAT6 transcription factor and induces phosphorylation of STAT6 in T-cells. The interaction with STAT6 requires the Tyr(127) residue and Lck-binding domain of Tip, which are indispensable for interleukin (IL)-2-independent T-cell transformation by HVS. It was also demonstrated that Tip induces nuclear translocation of STAT6, as well as activation of STAT6-dependent transcription in Jurkat T-cells. Interestingly, the phosphorylated STAT6 mainly colocalized with vesicles containing Tip within T-cells, but was barely detectable in the nucleus. However, nuclear translocation of phospho-STAT6 and transcriptional activation of STAT6 by IL-4 stimulation were not affected significantly in T-cells expressing Tip. Collectively, these findings suggest that the constitutive activation of STAT6 by Tip in T-cells may contribute to IL-2-independent T-cell transformation by HVS.


Subject(s)
Cell Transformation, Viral , Herpesvirus 2, Saimiriine/pathogenicity , Jurkat Cells/immunology , Jurkat Cells/virology , Phosphoproteins/metabolism , STAT6 Transcription Factor/metabolism , Viral Proteins/metabolism , Active Transport, Cell Nucleus , Humans , Protein Interaction Mapping , Transcription, Genetic
5.
Cold Spring Harb Protoc ; 2011(12): 1520-3, 2011 Dec 01.
Article in English | MEDLINE | ID: mdl-22135663

ABSTRACT

Herpesvirus saimiri (HVS) is capable of infecting a wide range of human cell types with high efficiency. The viral genome persists as high-copy-number, circular, nonintegrated episomes that segregate to progeny on cell division. This allows HVS-based vectors to transduce stably a dividing cell population and provide sustained transgene expression for an extended period of time both in vitro and in vivo. Moreover, the insertion of a bacterial artificial chromosome (BAC) cassette into the HVS genome simplifies the incorporation of large amounts of heterologous DNA for gene delivery. These properties offer characteristics similar to that of an artificial chromosome combined with an efficient delivery system. This protocol describes the use of an HVS recombinant virus expressing green fluorescent protein (GFP) (HVS-GFP) to assess the infectivity of a specific cell line.


Subject(s)
Genes, Reporter , Genetic Vectors , Green Fluorescent Proteins/metabolism , Herpesvirus 2, Saimiriine/pathogenicity , Staining and Labeling/methods , Virology/methods , Cell Line , Chromosomes, Artificial, Bacterial , Genetic Engineering , Genome, Viral , Green Fluorescent Proteins/genetics , Herpesvirus 2, Saimiriine/genetics , Humans , Recombination, Genetic , Transduction, Genetic
6.
J Med Virol ; 83(11): 1938-50, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21915869

ABSTRACT

Herpesvirus saimiri (HVS) causes acute lymphoma and leukemia upon experimental infection of various monkey species. HVS strain C488 is also capable of transforming human T-lymphocytes to stable growth in culture. The most susceptible species for oncogenesis are New World primates, in particular the cottontop tamarin (Saguinus oedipus). However, Old World monkeys such as macaques are the most used animal model for the close-to-human situation. The limited data on HVS infection in Old World monkeys prompted us to investigate susceptibility to infection and disease induction by HVS in macaques. After having established that rhesus macaques can be infected productively, and that rhesus T-cells can be transformed in vivo by HVS, we observed induction of lymphoma in all inoculated animals. Pre-existing humoral immunity in part of the rhesus colony capable of blocking HVS infection could be overcome by preselecting rhesus macaques for lack of this immunity of unknown origin. HVS infection of rhesus macaques as compared to that of New World monkeys has the advantages that disease progression is more prolonged, and larger blood volumes can be collected, which allows more extended analyses. Also, rhesus monkeys are the best immunologically and immunogenetically characterized primate species next to humans. This model could be useful for the evaluation of candidate tumor vaccines and to test novel approaches for cancer immunotherapy. In addition, HVS infection of macaques could eventually be useful as a surrogate model to address certain questions in rhadinovirus-induced human cancer such as effusion lymphoma or Kaposi's sarcoma.


Subject(s)
Cell Transformation, Viral , Disease Models, Animal , Herpesviridae Infections/pathology , Herpesvirus 2, Saimiriine/pathogenicity , Lymphoma/pathology , T-Lymphocytes/virology , Tumor Virus Infections/pathology , Animals , Female , Lymphoma/virology , Macaca mulatta , Male , Rhadinovirus/pathogenicity
7.
J Virol ; 85(20): 10627-38, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21849449

ABSTRACT

The mammalian retromer is an evolutionally conserved protein complex composed of a vacuolar protein sorting trimer (Vps 26/29/35) that participates in cargo recognition and a sorting nexin (SNX) dimer that binds to endosomal membranes. The retromer plays an important role in efficient retrograde transport for endosome-to-Golgi retrieval of the cation-independent mannose-6-phosphate receptor (CI-MPR), a receptor for lysosomal hydrolases, and other endosomal proteins. This ultimately contributes to the control of cell growth, cell adhesion, and cell migration. The herpesvirus saimiri (HVS) tyrosine kinase-interacting protein (Tip), required for the immortalization of primary T lymphocytes, targets cellular signaling molecules, including Lck tyrosine kinases and the p80 endosomal trafficking protein. Despite the pronounced effects of HVS Tip on T cell signal transduction, the details of its activity on T cell immortalization remain elusive. Here, we report that the amino-terminal conserved, glutamate-rich sequence of Tip specifically interacts with the retromer subunit Vps35 and that this interaction not only causes the redistribution of Vps35 from the early endosome to the lysosome but also drastically inhibits retromer activity, as measured by decreased levels of CI-MPR and lower activities of cellular lysosomal hydrolases. Physiologically, the inhibition of intracellular retromer activity by Tip is ultimately linked to the downregulation of CD4 surface expression and to the efficient in vitro immortalization of primary human T cells to interleukin-2 (IL-2)-independent permanent growth. Therefore, HVS Tip uniquely targets the retromer complex to impair the intracellular trafficking functions of infected cells, ultimately contributing to efficient T cell transformation.


Subject(s)
CD4-Positive T-Lymphocytes/virology , Cell Transformation, Viral , Herpesvirus 2, Saimiriine/pathogenicity , Phosphoproteins/metabolism , Vesicular Transport Proteins/metabolism , Viral Proteins/metabolism , Cell Line , Humans , Protein Interaction Mapping
8.
J Virol ; 85(11): 5456-64, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21430050

ABSTRACT

Herpesviruses establish latency in suitable host cells after primary infection and persist in their host organisms for life. Most of the viral genes are silenced during latency, also enabling the virus to escape from an immune response. This study addresses the control of viral gene silencing by epigenetic mechanisms, using Herpesvirus saimiri (HVS) as a model system. Strain C488 of this gamma-2-herpesvirus can transform human T cells to stable growth in vitro, and it persists in the nuclei of those latently infected T cells as a nonintegrating, circular, and histone-associated episome. The whole viral genome was probed for histone acetylation at high resolution by chromatin immunoprecipitation-on-chip (ChIP-on-chip) with a custom tiling microarray. Corresponding to their inactive status in human T cells, the lytic promoters consistently revealed a heterochromatic phenotype. In contrast, the left terminal region of the genome, which encodes the stably expressed oncogenes stpC and tip as well as the herpesvirus U RNAs, was associated with euchromatic histone acetylation marks representing "open" chromatin. Although HVS latency in human T lymphocytes is considered a stable and irreversible state, incubation with the histone deacetylase inhibitor trichostatin A resulted in changes reminiscent of the induction of early lytic replication. However, infectious viral particles were not produced, as the majority of cells went into apoptosis. These data show that epigenetic mechanisms are involved in both rhadinoviral latency and transition into lytic replication.


Subject(s)
Gene Expression Regulation, Viral , Herpesvirus 2, Saimiriine/pathogenicity , Histones/metabolism , Host-Pathogen Interactions , T-Lymphocytes/virology , Virus Latency , Acetylation , Cells, Cultured , Histone Deacetylase Inhibitors/metabolism , Humans
9.
J Biomed Biotechnol ; 2011: 785158, 2011.
Article in English | MEDLINE | ID: mdl-21197456

ABSTRACT

Herpesvirus saimiri (HVS) is a gamma herpesvirus with several properties that make it an amenable gene therapy vector; namely its large packaging capacity, its ability to persist as a nonintegrated episome, and its ability to infect numerous human cell types. We used RecA-mediated recombination to develop an HVS vector with a mutated virion protein. The heparan sulphate-binding region of HVS ORF51 was substituted for a peptide sequence which interacts with somatostatin receptors (SSTRs), overexpressed on hepatocellular carcinoma (HCC) cells. HVS mORF51 showed reduced infectivity in non-HCC human cell lines compared to wild-type virus. Strikingly, HVS mORF51 retained its ability to infect HCC cell lines efficiently. However, neutralisation assays suggest that HVS mORF51 has no enhanced binding to SSTRs. Therefore, mutation of the ORF51 glycoprotein has specifically targeted HVS to HCC cell lines by reducing the infectivity of other cell types; however, the mechanism for this targeting is unknown.


Subject(s)
Carcinoma, Hepatocellular/virology , Herpesvirus 2, Saimiriine/physiology , Liver Neoplasms/virology , Membrane Glycoproteins/genetics , Mutation , Viral Envelope Proteins/genetics , Amino Acid Sequence , Animals , Cell Line, Tumor , Cloning, Molecular , Electrophoresis, Agar Gel , Flow Cytometry , Genetic Vectors/genetics , Heparitin Sulfate/metabolism , Herpesvirus 2, Saimiriine/genetics , Herpesvirus 2, Saimiriine/pathogenicity , Humans , Membrane Glycoproteins/metabolism , Mice , Molecular Sequence Data , Neutralization Tests , Receptors, Somatostatin/metabolism , Viral Envelope Proteins/metabolism
10.
J Biotechnol ; 134(3-4): 287-96, 2008 Apr 30.
Article in English | MEDLINE | ID: mdl-18328588

ABSTRACT

Viral vectors have a number of obstacles to overcome for effective gene therapy, including immune stimulation, packaging potential and cell tropism. Herpesvirus saimiri (HVS) has many favourable traits including, a large packaging capability, wide cell tropism, and the ability to episomally persist as an artificial chromosome. To further develop HVS as a gene therapy vector we aim to produce a safe disabled HVS-based recombinant viral system for gene therapy applications. An HVS recombinant viral amplicon was constructed with a transgene packaging potential of 50 kb. The recombinant HVS genome was shown to be replication disabled and used to generate a stable cell line, OMKHVS Delta Bam, in which the modified genome persists as a non-integrated episome. To assess whether the modified genome could be packaged into a virus-like particle (HVSampVLP), OMKHVS Delta Bam was infected with replication competent virus or transfected with a defective helper virus. The resultant HVSampVLPs were able to infect SW480 tumour cells, delivering the recombinant disabled genome, which persisted as a non-integrated episome in the dividing cell population. This study forms the basis of a replication disabled HVS amplicon system for use in gene therapy applications.


Subject(s)
Defective Viruses/genetics , Genome, Viral , Herpesvirus 2, Saimiriine/genetics , Animals , Cell Line, Transformed , Cell Line, Tumor , Chromosomes, Artificial, Bacterial/metabolism , Defective Viruses/pathogenicity , Female , Gene Transfer Techniques , Helper Viruses/genetics , Herpesvirus 2, Saimiriine/pathogenicity , Recombination, Genetic , Transgenes/physiology , Virus Assembly/genetics
11.
J Virol ; 81(5): 2524-30, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17151105

ABSTRACT

Herpesvirus saimiri (HVS) subgroup C strains are able to growth transform human T lymphocytes in vitro. The stably persisting and nonintegrating HVS episome represents an optimal prerequisite for the investigation of the epigenetic state of latent herpesvirus genomes in vitro. Quantitative chromatin immunoprecipitation experiments using seven different histone acetylation- or methylation-specific antibodies revealed repressive marks at four lytic gene promoters and a variable pattern at the weakly transcribed LANA/orf73 promoter. The constitutive stpC/tip promoter regulating the viral oncoproteins and, more interestingly, the noncoding repetitive H-DNA elements flanking the coding region, showed a permissive chromatin structure. This study provides an appropriate model for the analysis of epigenetic herpesvirus genome modifications and their dynamics in T cells.


Subject(s)
Genome, Viral , Herpesvirus 2, Saimiriine/genetics , Histones/metabolism , T-Lymphocytes/virology , Acetylation , Base Sequence , Cell Transformation, Viral/genetics , Cell Transformation, Viral/physiology , DNA, Viral/genetics , Epigenesis, Genetic , Fetal Blood/cytology , Herpesvirus 2, Saimiriine/pathogenicity , Herpesvirus 2, Saimiriine/physiology , Histones/chemistry , Humans , In Vitro Techniques , Infant, Newborn , Methylation , Models, Genetic , Promoter Regions, Genetic , T-Lymphocytes/metabolism , Virus Latency/genetics , Virus Latency/physiology
12.
J Virol ; 78(18): 9814-9, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15331715

ABSTRACT

Herpesvirus saimiri group C strains are capable of transforming human and simian T-lymphocyte populations to permanent antigen-independent growth. Two viral oncoproteins, StpC and Tip, that are encoded by a single bicistronic mRNA, act in concert to mediate this phenotype. A closely related New World monkey herpesvirus, herpesvirus ateles, transcribes a single spliced mRNA at an equivalent genome locus. The encoded protein, Tio, has sequence homologies to both StpC and Tip. We inserted the tio sequence of herpesvirus ateles strain 73 into a recombinant herpesvirus saimiri C488 lacking its own stpC/tip oncogene. Simian as well as human T lymphocytes were growth transformed by the chimeric Tio-expressing viruses. Thus, a single herpesvirus protein appears to be responsible for the oncogenic effects of herpesvirus ateles.


Subject(s)
Herpesvirus 2, Saimiriine/pathogenicity , Oncogene Proteins, Viral/physiology , Phosphoproteins/physiology , Rhadinovirus/pathogenicity , T-Lymphocytes/virology , Viral Proteins/physiology , Animals , Base Sequence , Cell Transformation, Viral , Cells, Cultured , DNA, Viral/genetics , Herpesvirus 2, Saimiriine/genetics , Humans , Oncogene Proteins, Viral/genetics , Phosphoproteins/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Rhadinovirus/genetics , Saguinus , Viral Proteins/genetics
13.
Virology ; 326(1): 67-78, 2004 Aug 15.
Article in English | MEDLINE | ID: mdl-15262496

ABSTRACT

Herpesvirus saimiri (HVS) is a gamma(2)-herpesvirus sharing genomic colinearity and a high degree of functional homology with HHV-8. To begin exploring the correlates of HVS infectivity and neutralization, we designed and implemented a new reporter assay. Using this assay, we could demonstrate that HVS neutralizing antibodies are present at high levels in naturally infected squirrel monkeys and are strongly induced after pathogenic, experimental infection of common marmosets. Further, we demonstrated that viral entry is influenced by cellular glycosaminoglycans and that, similar to HHV-8, soluble heparin is capable of blocking infectivity. We next cloned and characterized the positional homologue of HHV-8 K8.1, HVS Orf51. N-glycosidase F treatment indicates that like K8.1, Orf51 is a glycoprotein. Found in the viral particle, it localizes to the endoplasmic reticulum of expressing cells. Like K8.1, Orf51 could bind to agarose-conjugated heparin, implicating this molecule in viral attachment to cells. These studies provide the groundwork for additional experiments into the role that this protein may be playing in viral pathogenicity, persistence, and cell tropism.


Subject(s)
Antibodies, Viral/blood , Glycoproteins/genetics , Herpesvirus 2, Saimiriine/genetics , Herpesvirus 2, Saimiriine/immunology , Membrane Glycoproteins/genetics , Viral Envelope Proteins/genetics , Amino Acid Sequence , Animals , Base Sequence , Binding Sites , Callithrix , Disease Models, Animal , Glycoproteins/immunology , Glycosaminoglycans/genetics , Glycosaminoglycans/metabolism , Heparin/pharmacology , Herpesviridae Infections/blood , Herpesviridae Infections/immunology , Herpesviridae Infections/veterinary , Herpesvirus 2, Saimiriine/pathogenicity , Membrane Glycoproteins/immunology , Molecular Sequence Data , Neutralization Tests/methods , Open Reading Frames/physiology , Rabbits , Saimiri , Tumor Virus Infections/blood , Tumor Virus Infections/immunology , Tumor Virus Infections/veterinary , Viral Envelope Proteins/immunology , Virulence/drug effects , Virus Replication
15.
Mol Cell Biol ; 24(12): 5369-82, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15169900

ABSTRACT

Because of its central regulatory role, T-cell receptor (TCR) signal transduction is a common target of viruses. We report here the identification of a small signaling protein, ORF5, of the T-lymphotropic tumor virus herpesvirus saimiri (HVS). ORF5 is predicted to contain 89 to 91 amino acids with an amino-terminal myristoylation site and six SH2 binding motifs, showing structural similarity to cellular LAT (linker for activation of T cells). Sequence analysis showed that, despite extensive sequence variation, the myristoylation site and SH2 binding motifs were completely conserved among 13 different ORF5 isolates. Upon TCR stimulation, ORF5 was efficiently tyrosine phosphorylated and subsequently interacted with cellular SH2-containing signaling proteins Lck, Fyn, SLP-76, and p85 through its tyrosine residues. ORF5 expression resulted in the marked augmentation of TCR signal transduction activity, evidenced by increased cellular tyrosine phosphorylation, intracellular calcium mobilization, CD69 surface expression, interleukin-2 production, and activation of the NF-AT, NF-kappa B, and AP-1 transcription factors. Despite its structural similarity to cellular LAT, however, ORF5 could only partially substitute for LAT function in TCR signal transduction. These results demonstrate that HVS utilizes a novel signaling protein, ORF5, to activate TCR signal transduction. This activation probably facilitates viral gene expression and, thereby, persistent infection.


Subject(s)
Herpesvirus 2, Saimiriine/pathogenicity , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/virology , Viral Proteins/physiology , Alleles , Amino Acid Sequence , Binding Sites , Cell Line , Genes, Viral , Herpesvirus 2, Saimiriine/genetics , Herpesvirus 2, Saimiriine/physiology , Humans , Jurkat Cells , Membrane Microdomains/virology , Molecular Sequence Data , Phosphorylation , Sequence Homology, Amino Acid , Signal Transduction , Tyrosine/chemistry , Viral Proteins/chemistry , Viral Proteins/genetics , src Homology Domains , src-Family Kinases/metabolism
16.
Virology ; 314(2): 471-87, 2003 Sep 30.
Article in English | MEDLINE | ID: mdl-14554077

ABSTRACT

Herpesvirus saimiri (HVS), the rhadinovirus prototype, is apathogenic in the persistently infected natural host, the squirrel monkey, but causes acute T cell leukemia in other New World primate species. In contrast to subgroups A and B, only strains of HVS subgroup C such as C488 are capable of transforming primary human T cells to stable antigen-independent growth in culture. Here, we report the complete 155-kb genome sequence of the transformation-competent HVS strain C488. The A+T-rich unique L-DNA of 113,027 bp encodes at least 77 open reading frames and 5 URNAs. In addition to the viral oncogenes stp and tip, only a few genes including the transactivator orf50 and the glycoprotein orf51 are highly divergent. In a series of new primary HVS isolates, the subgroup-specific divergence of the orf50/orf51 alleles was studied. In these new isolates, the orf50/orf51 alleles of the respective subgroup segregate with the stp and/or tip oncogene alleles, which are essential for transformation.


Subject(s)
Cell Transformation, Viral , Genome, Viral , Herpesvirus 2, Saimiriine/physiology , T-Lymphocytes/virology , Amino Acid Sequence , Animals , Cells, Cultured , Gene Expression Regulation, Viral , Herpesvirus 2, Saimiriine/pathogenicity , Humans , Molecular Sequence Data , Open Reading Frames/genetics , Saimiri/virology , Sequence Analysis, DNA , Viral Proteins/genetics
17.
Virology ; 294(1): 31-46, 2002 Mar 01.
Article in English | MEDLINE | ID: mdl-11886263

ABSTRACT

Efficiency of lymphoma induction by herpesvirus saimiri (HVS) isolates correlates with the genetically defined viral subgroups A, B, and C. To compare subgroup-specific effects, highly susceptible tamarins were infected with HVS strain A-11, B-SMHI, or C-488. All animals developed T-cell lymphomas indistinguishable with respect to clinical, pathological, and virological parameters. Ex vivo T-cell lines were established readily from the HVS C-488 animal, less efficiently in the presence of HVS A-11, and from only a single HVS B-SMHI sample. These cultivated cells revealed strain-specific biochemical characteristics. HVS A-11 strongly induced the expression of tyrosine kinase Lyn. HVS C-488 led to the activation of STAT3, which is most likely linked to the association of virus-encoded Tip with tyrosine kinase Lck. The lack of these activities in HVS B-SMHI-transformed cells may correlate with the reduced oncogenic phenotype of this virus in species other than tamarins.


Subject(s)
Herpesviridae Infections/virology , Herpesvirus 2, Saimiriine/pathogenicity , Lymphoma, T-Cell/virology , Tumor Virus Infections/virology , Animals , Cell Line, Transformed , DNA-Binding Proteins/metabolism , Herpesviridae Infections/pathology , Herpesvirus 2, Saimiriine/classification , Herpesvirus 2, Saimiriine/physiology , Lymphoma, T-Cell/pathology , Oncogene Proteins, Viral/genetics , Oncogene Proteins, Viral/metabolism , Phosphoproteins/metabolism , Phosphorylation , Protein-Tyrosine Kinases/metabolism , STAT3 Transcription Factor , Saguinus , T-Lymphocytes/virology , Trans-Activators/metabolism , Tumor Cells, Cultured , Tumor Virus Infections/pathology , Viral Proteins/metabolism
18.
DNA Cell Biol ; 20(2): 81-8, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11244565

ABSTRACT

New World primates develop T-cell lymphomas on infection with Herpesvirus saimiri. To investigate the oncogenic potential of the Tip gene of Herpesvirus saimiri strain C488, we tried to establish transgenic mice that should express Tip under control of a constitutive promoter. Although transgene-positive embryos were found, lines could not be established. However, using a system in which the transgene has to be activated by a Cre recombinase-mediated deletion, we were able to obtain several Tip transgenic lines. At high expression levels, the mice developed T-cell lymphomas. Thus, Tip can induce lymphomas and is therefore very likely responsible for the oncogenicity of Herpesvirus saimiri.


Subject(s)
Genes, Viral , Herpesvirus 2, Saimiriine/genetics , Lymphoma, T-Cell/genetics , Lymphoma, T-Cell/virology , Mice, Transgenic/genetics , Phosphoproteins/genetics , Viral Proteins/genetics , Viral Structural Proteins/genetics , Animals , Crosses, Genetic , Embryonic and Fetal Development/genetics , Herpesvirus 2, Saimiriine/pathogenicity , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , Lymphoma, T-Cell/mortality , Lymphoma, T-Cell/pathology , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Transgenic/virology , Phosphoproteins/biosynthesis , Phosphoproteins/metabolism , Phosphoproteins/physiology , Survival Analysis , Viral Proteins/biosynthesis , Viral Proteins/metabolism , Viral Proteins/physiology
19.
J Exp Med ; 193(5): 637-42, 2001 Mar 05.
Article in English | MEDLINE | ID: mdl-11238594

ABSTRACT

Cyclin D family members are cellular protooncogenes, and their viral homologues in the Kaposi's sarcoma-associated herpesvirus (KSHV, human herpesvirus type 8 [HHV-8]) and the closely related Herpesvirus saimiri have been implicated as putative cofactors of viral transformation and pathogenesis. KSHV is regularly found in Kaposi's sarcoma and in the primary effusion B cell lymphoma and Castleman's disease associated with immunosuppression and AIDS. H. saimiri strain C488 transforms human and marmoset T cells in vitro and causes polyclonal T cell lymphoma in New World monkeys. The viral cyclins stimulate cell cycle progression of quiescent fibroblasts, and they form active cyclin-dependent kinase (CDK)6 complexes of broad substrate specificity that can resist and downregulate cellular CDK inhibitors. This study shows that the viral cyclin of H. saimiri strain C488 is not required for viral replication, T cell transformation, and pathogenicity in New World primates.


Subject(s)
Cell Transformation, Viral , Cyclins/metabolism , Herpesvirus 2, Saimiriine/metabolism , Lymphoma, T-Cell/metabolism , Neoplasms, Experimental/metabolism , Animals , Aotidae , Callithrix , Cell Transformation, Viral/genetics , Cells, Cultured , Cyclin D , Cyclins/genetics , Gene Deletion , Gene Targeting , Herpesviridae Infections/metabolism , Herpesviridae Infections/pathology , Herpesvirus 2, Saimiriine/genetics , Herpesvirus 2, Saimiriine/pathogenicity , Humans , Kidney/cytology , Kidney/metabolism , Kidney/virology , Lymphocytes/cytology , Lymphocytes/metabolism , Lymphocytes/virology , Lymphoma, T-Cell/pathology , Lymphoma, T-Cell/virology , Neoplasms, Experimental/pathology , Neoplasms, Experimental/virology , Reverse Transcriptase Polymerase Chain Reaction , Saguinus , Tumor Virus Infections/metabolism , Tumor Virus Infections/pathology , Viral Proteins
20.
Eur J Biochem ; 267(12): 3413-21, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10848956

ABSTRACT

Protein tyrosine kinases (PTKs) are critically involved in signaling pathways that regulate cell growth, differentiation, activation, and transformation. It is not surprising, therefore, that viruses acquire effector molecules targeting these kinases to ensure their own replication and/or persistence. This review summarizes our current knowledge on Lck, a member of the Src family of PTK, and its viral interaction partners. Lck plays a key role in T lymphocyte activation and differentiation. It is associated with a variety of cell surface receptors and is critical for signal transduction from the T-cell antigen receptor (TCR). Consequently, Lck is targeted by regulatory proteins of T-lymphotropic viruses, especially by the Herpesvirus saimiri (HVS) tyrosine kinase interacting protein (Tip). This oncoprotein physically interacts with Lck in HVS transformed T cells and has an impact on its catalytic activity. However, while Tip inhibits Lck activity in stably expressing cell lines, opposite effects were observed in several in vitro systems. At least in part, this complex situation may be related to the bipartite nature of the interaction surface of the two proteins. Studies on the interrelationships between Lck and its viral partners contribute to the understanding of the mechanisms of T-cell growth regulation, in general, and of viral pathogenicity in particular. In addition, understanding the regulation of Lck activity by viral proteins may serve as a basis for the development of new drugs capable of modifying Lck activity in different pathological situations.


Subject(s)
Herpesvirus 2, Saimiriine/pathogenicity , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , Phosphoproteins/metabolism , T-Lymphocytes/metabolism , T-Lymphocytes/virology , Viral Proteins/metabolism , Animals , Cell Transformation, Viral , Cytokines/metabolism , Herpesvirus 2, Saimiriine/metabolism , Humans , Lymphocyte Activation , Phosphoproteins/genetics , Viral Proteins/genetics , src-Family Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL