Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 70
Filtrar
1.
Mamm Genome ; 33(2): 412-420, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-34491378

RESUMEN

Insights into interactions between viral factors and the cellular machinery usually lead to discoveries concerning host cell biology. Thus, the gene expression field has historically relied on viral model systems to discover mechanisms underlying different cellular processes. In recent years, the functional characterization of the small nuclear noncoding RNAs expressed by the oncogenic Herpesvirus saimiri, called HSURs, resulted in the discovery of two mechanisms for the regulation of gene expression. HSUR1 and HSUR2 associate with host microRNAs, which are small noncoding RNAs that broadly regulate gene expression by binding to messenger RNAs. HSUR1 provided the first example of a process known as target-directed miRNA degradation that operates in cells to regulate miRNA populations. HSUR2 functions as a miRNA adaptor, uncovering an entirely new, indirect mechanism by which miRNAs can inhibit mRNA function. Here, I review the path that led to these discoveries and their implications and postulate new exciting questions about the functions of these fascinating viral noncoding RNAs.


Asunto(s)
Herpesvirus Saimiriino 2 , MicroARNs , Virus , Herpesvirus Saimiriino 2/genética , Herpesvirus Saimiriino 2/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN no Traducido/genética , ARN no Traducido/metabolismo , Virus/genética , Virus/metabolismo
2.
Biochim Biophys Acta Mol Cell Res ; 1864(1): 1-11, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27751885

RESUMEN

Members of the herpesviral family use multiple strategies to hijack infected host cells and exploit cellular signaling for their pathogenesis and latent infection. Among the most intriguing weapons in the arsenal of pathogenic herpesviruses are the constitutively active virally-encoded G protein-coupled receptors (vGPCRs). Even though vGPCRs contribute to viral pathogenesis such as immune evasion and proliferative disorders, the molecular details of how vGPCRs continuously activate cellular signaling are largely unknown. Here, we report that the vGPCR of Herpesvirus saimiri (HVS), an oncogenic γ2-herpesvirus, constitutively activates T cells via a heteromeric interaction with cellular CXCR4. Constitutive T cell activation also occurs with expression of the vGPCR of Kaposi's sarcoma-associated herpesvirus (KSHV), but not the vGPCR of Epstein-Barr virus. Expression of HVS vGPCR down-regulated the surface expression of CXCR4 but did not induce the degradation of the chemokine receptor, suggesting that vGPCR/CXCR4 signaling continues in cytosolic compartments. The physical association of vGPCR with CXCR4 was demonstrated by proximity ligation assay as well as immunoprecipitation. Interestingly, the constitutive activation of T cells by HVS vGPCR is independent of proximal T cell receptor (TCR) signaling molecules, such as TCRß, Lck, and ZAP70, whereas CXCR4 silencing by shRNA abolished T cell activation by vGPCRs of HVS and KSHV. Furthermore, previously identified inactive vGPCR mutants failed to interact with CXCR4. These findings on the positive cooperativity of vGPCR with cellular CXCR4 in T cell activation extend our current understanding of the molecular mechanisms of vGPCR function and highlight the importance of heteromerization for GPCR activity.


Asunto(s)
Herpesvirus Saimiriino 2/metabolismo , Herpesvirus Humano 8/metabolismo , Receptores CXCR4/genética , Receptores de Quimiocina/genética , Linfocitos T/virología , Regulación de la Expresión Génica , Células HEK293 , Herpesvirus Saimiriino 2/genética , Herpesvirus Saimiriino 2/crecimiento & desarrollo , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/crecimiento & desarrollo , Herpesvirus Humano 4/metabolismo , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/crecimiento & desarrollo , Interacciones Huésped-Patógeno , Humanos , Activación de Linfocitos , Proteína Tirosina Quinasa p56(lck) Específica de Linfocito/genética , Proteína Tirosina Quinasa p56(lck) Específica de Linfocito/inmunología , Proteína Tirosina Quinasa p56(lck) Específica de Linfocito/metabolismo , Cultivo Primario de Células , Unión Proteica , Multimerización de Proteína , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Receptores de Antígenos de Linfocitos T alfa-beta/inmunología , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo , Receptores CXCR4/inmunología , Receptores CXCR4/metabolismo , Receptores de Quimiocina/inmunología , Receptores de Quimiocina/metabolismo , Transducción de Señal , Linfocitos T/inmunología , Linfocitos T/metabolismo , Proteína Tirosina Quinasa ZAP-70/genética , Proteína Tirosina Quinasa ZAP-70/inmunología , Proteína Tirosina Quinasa ZAP-70/metabolismo
3.
RNA ; 22(8): 1181-9, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27335146

RESUMEN

Herpesvirus saimiri, an oncogenic herpesvirus, during latency produces seven small nuclear RNAs, called the Herpesvirus saimiri U RNAs (HSUR1-7). HSUR1 mediates degradation of the host microRNA, miR-27, via a process that requires imperfect base-pairing. The decreased levels of miR-27 lead to prolonged T-cell activation and likely contribute to oncogenesis. To gain insight into HSUR1-mediated degradation of miR-27, we probed the in vivo secondary structure of HSUR1 and coupled this with bioinformatic structural analyses. The results suggest that HSUR1 adopts a conformation different than previously believed and that the region complementary to miR-27 lacks stable structure. To determine whether HSUR1 structural flexibility is important for its ability to mediate miR-27 degradation, we performed structurally informative mutagenic analyses of HSUR1. HSUR1 mutants in which the miR-27 binding site sequence is preserved, but sequestered in predicted helices, lose their ability to decrease miR-27 levels. These results indicate that the HSUR1 miR27-binding region must be available in a conformationally flexible segment for noncoding RNA function.


Asunto(s)
Genes Virales , Herpesvirus Saimiriino 2/metabolismo , MicroARNs/metabolismo , ARN Nuclear Pequeño/metabolismo , Animales , Callithrix , Herpesvirus Saimiriino 2/genética
4.
Genes Dev ; 29(14): 1552-64, 2015 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-26220997

RESUMEN

Herpesvirus saimiri (HVS) is an oncogenic γ-herpesvirus that produces microRNAs (miRNAs) by cotranscription of precursor miRNA (pre-miRNA) hairpins immediately downstream from viral small nuclear RNAs (snRNA). The host cell Integrator complex, which recognizes the snRNA 3' end processing signal (3' box), generates the 5' ends of HVS pre-miRNA hairpins. Here, we identify a novel 3' box-like sequence (miRNA 3' box) downstream from HVS pre-miRNAs that is essential for miRNA biogenesis. In vivo knockdown and rescue experiments confirmed that the 3' end processing of HVS pre-miRNAs also depends on Integrator activity. Interaction between Integrator and HVS primary miRNA (pri-miRNA) substrates that contain only the miRNA 3' box was confirmed by coimmunoprecipitation and an in situ proximity ligation assay (PLA) that we developed to localize specific transient RNA-protein interactions inside cells. Surprisingly, in contrast to snRNA 3' end processing, HVS pre-miRNA 3' end processing by Integrator can be uncoupled from transcription, enabling new approaches to study Integrator enzymology.


Asunto(s)
Herpesvirus Saimiriino 2/genética , MicroARNs/metabolismo , Procesamiento de Término de ARN 3'/fisiología , Técnicas de Silenciamiento del Gen , Células HEK293 , Células HeLa , Herpesvirus Saimiriino 2/metabolismo , Interacciones Huésped-Patógeno/genética , Humanos , Inmunoprecipitación , MicroARNs/química , MicroARNs/genética , Complejos Multiproteicos/genética , Complejos Multiproteicos/metabolismo , Procesamiento de Término de ARN 3'/genética , Precursores del ARN/genética , Precursores del ARN/metabolismo , ARN Nuclear Pequeño/metabolismo , Transcripción Genética
5.
Mol Cell ; 54(1): 67-79, 2014 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-24725595

RESUMEN

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.


Asunto(s)
Herpesvirus Saimiriino 2/metabolismo , Activación de Linfocitos , MicroARNs/metabolismo , ARN no Traducido/metabolismo , ARN Viral/metabolismo , Linfocitos T/metabolismo , Animales , Antígenos CD/genética , Antígenos CD/metabolismo , Antígenos de Diferenciación de Linfocitos T/metabolismo , Secuencia de Bases , Callithrix , Activación Enzimática , Proteínas Ligadas a GPI/genética , Proteínas Ligadas a GPI/metabolismo , Proteína Adaptadora GRB2/genética , Proteína Adaptadora GRB2/metabolismo , Regulación de la Expresión Génica , Células HEK293 , Herpesvirus Saimiriino 2/genética , Herpesvirus Saimiriino 2/patogenicidad , Secuenciación de Nucleótidos de Alto Rendimiento , Interacciones Huésped-Patógeno , Humanos , Inmunoprecipitación , Interferón gamma/genética , Interferón gamma/metabolismo , Células Jurkat , Lectinas Tipo C/metabolismo , MicroARNs/genética , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Datos de Secuencia Molecular , Estabilidad del ARN , ARN no Traducido/genética , ARN Viral/genética , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/metabolismo , Semaforinas/genética , Semaforinas/metabolismo , Análisis de Secuencia de ARN , Transducción de Señal , Linfocitos T/inmunología , Linfocitos T/virología , Factores de Tiempo , Transfección
6.
PLoS Pathog ; 10(2): e1003907, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24550725

RESUMEN

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.


Asunto(s)
Infecciones por Herpesviridae/metabolismo , Interacciones Huésped-Parásitos/fisiología , Proteínas Nucleares/metabolismo , ARN Viral/metabolismo , Proteínas de Unión al ARN/metabolismo , Proteínas Represoras/metabolismo , Transactivadores/metabolismo , Factores de Transcripción/metabolismo , Infecciones Tumorales por Virus/metabolismo , Transporte Activo de Núcleo Celular/fisiología , Herpesvirus Saimiriino 2/química , Herpesvirus Saimiriino 2/metabolismo , Herpesvirus Saimiriino 2/patogenicidad , Humanos , Proteínas Nucleares/química , Estructura Cuaternaria de Proteína , Transporte de ARN/fisiología , ARN Viral/análisis , Proteínas de Unión al ARN/química , Proteínas Represoras/química , Transactivadores/química , Factores de Transcripción/química
7.
RNA ; 18(12): 2166-73, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23097423

RESUMEN

RNase III enzymes are fundamental to the biogenesis of microRNAs (miRNAs) and small interfering RNAs (siRNAs) in all species studied. Although alternative miRNA pathways independent of Drosha or Dicer exist, each still requires one RNase III-type enzyme. Here, we describe two strategies that marry either RNase Z or the Integrator complex with the slicing activity of Argonaute2 to generate highly functional mature miRNAs. We provide stringent validation of their RNase III independence by demonstrating efficient miRNA biogenesis and activity in Drosha and Dicer knockout cells. These data provide proof-of-principle evidence for additional mechanistic possibilities for efficient generation of small regulatory RNAs, and represent novel silencing triggers that may be exploited for technical purposes.


Asunto(s)
MicroARNs/biosíntesis , Ribonucleasa III/metabolismo , Animales , Proteínas Argonautas/deficiencia , Proteínas Argonautas/genética , Proteínas Argonautas/metabolismo , Secuencia de Bases , Línea Celular , ARN Helicasas DEAD-box/deficiencia , ARN Helicasas DEAD-box/genética , ARN Helicasas DEAD-box/metabolismo , Endorribonucleasas/deficiencia , Endorribonucleasas/genética , Endorribonucleasas/metabolismo , Técnicas de Inactivación de Genes , Células HEK293 , Células HeLa , Herpesvirus Saimiriino 2/genética , Herpesvirus Saimiriino 2/metabolismo , Humanos , Ratones , MicroARNs/genética , ARN Nuclear Pequeño/genética , ARN Nuclear Pequeño/metabolismo , ARN Viral/genética , ARN Viral/metabolismo , Ribonucleasa III/deficiencia , Ribonucleasa III/genética
8.
J Virol ; 86(7): 3541-53, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22278248

RESUMEN

In recent studies, the nuclear domain 10 (ND10) components PML, Sp100, human Daxx (hDaxx), and ATRX were identified to be cellular restriction factors that are able to inhibit the replication of several herpesviruses. The antiviral function of ND10, however, is antagonized by viral effector proteins by a variety of strategies, including degradation of PML or relocalization of ND10 proteins. In this study, we analyzed the interplay between infection with herpesvirus saimiri (HVS), the prototypic rhadinovirus, and cellular defense by ND10. In contrast to other herpesviruses, we found that HVS specifically degraded the cellular ND10 component Sp100, whereas other factors like PML or hDaxx remained intact. We could further identify the ORF3 tegument protein of HVS, which shares homology with the cellular formylglycinamide ribotide amidotransferase (FGARAT) enzyme, to be the viral factor that induces the proteasomal degradation of Sp100. Interestingly, recent studies showed that the ORF3-homologous proteins ORF75c of murine gammaherpesvirus 68 and BNRF-1 of Epstein-Barr virus modulate the ND10 proteins PML and ATRX, respectively, suggesting that the ND10 targets of viral FGARAT-homologous proteins diversified during evolution. Furthermore, a virus with the ORF3 deletion was efficiently complemented in Sp100-depleted cells, indicating that Sp100 is able to inhibit HVS in the absence of antagonistic mechanisms. In contrast, we observed that PML, which was neither degraded nor redistributed after HVS infection, strongly restricted both wild-type HVS and virus with the ORF3 deletion. Thus, HVS may lack a factor that efficiently counteracts the repressive function of PML, which may foster latency as the outcome of infection.


Asunto(s)
Antígenos Nucleares/metabolismo , Autoantígenos/metabolismo , Infecciones por Herpesviridae/metabolismo , Herpesvirus Saimiriino 2/metabolismo , Proteínas Nucleares/inmunología , Sistemas de Lectura Abierta , Proteínas Virales/metabolismo , Antígenos Nucleares/genética , Autoantígenos/genética , Línea Celular , Regulación Viral de la Expresión Génica , Infecciones por Herpesviridae/genética , Infecciones por Herpesviridae/inmunología , Infecciones por Herpesviridae/virología , Herpesvirus Saimiriino 2/genética , Humanos , Inmunidad , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/genética , Unión Proteica , Proteínas Virales/genética
9.
Science ; 328(5985): 1563-6, 2010 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-20558719

RESUMEN

T cells transformed by Herpesvirus saimiri express seven viral U-rich noncoding RNAs of unknown function called HSURs. We noted that conserved sequences in HSURs 1 and 2 constitute potential binding sites for three host-cell microRNAs (miRNAs). Coimmunoprecipitation experiments confirmed that HSURs 1 and 2 interact with the predicted miRNAs in virally transformed T cells. The abundance of one of these miRNAs, miR-27, is dramatically lowered in transformed cells, with consequent effects on the expression of miR-27 target genes. Transient knockdown and ectopic expression of HSUR 1 demonstrate that it directs degradation of mature miR-27 in a sequence-specific and binding-dependent manner. This viral strategy illustrates use of a ncRNA to manipulate host-cell gene expression via the miRNA pathway.


Asunto(s)
Regulación hacia Abajo , Herpesvirus Saimiriino 2/genética , MicroARNs/metabolismo , Estabilidad del ARN , ARN no Traducido/metabolismo , ARN Viral/metabolismo , Animales , Emparejamiento Base , Sitios de Unión , Callithrix , Línea Celular Transformada , Transformación Celular Viral , Secuencia Conservada , Herpesvirus Saimiriino 2/metabolismo , Humanos , Células Jurkat , MicroARNs/química , MicroARNs/genética , ARN no Traducido/química , ARN Viral/química , Linfocitos T
10.
J Virol ; 83(19): 10299-304, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19640995

RESUMEN

Herpesvirus saimiri encodes a functional homolog of human regulator-of-complement-activation proteins named CCPH that inactivates complement by accelerating the decay of C3 convertases and by serving as a cofactor in factor I-mediated inactivation of their subunits C3b and C4b. Here, we map the functional domains of CCPH. We demonstrate that short consensus repeat 2 (SCR2) is the minimum domain essential for classical/lectin pathway C3 convertase decay-accelerating activity as well as for factor I cofactor activity for C3b and C4b. Thus, CCPH is the first example wherein a single SCR domain has been shown to display complement regulatory functions.


Asunto(s)
Proteínas del Sistema Complemento/química , Herpesvirus Saimiriino 2/metabolismo , Biotinilación , Complemento C3b/química , Complemento C4b/química , Vía Alternativa del Complemento , Electroforesis en Gel de Poliacrilamida , Eliminación de Gen , Humanos , Cinética , Lectinas/química , Ligandos , Mutación , Estructura Terciaria de Proteína , Proteínas Recombinantes/química
11.
J Gen Virol ; 90(Pt 3): 596-601, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19218204

RESUMEN

The herpesvirus saimiri (HVS) ORF57 protein binds viral RNA, enabling the efficient nuclear export of intronless viral mRNAs. However, it is not known how ORF57 recognizes these viral mRNAs. In this study, a systematic evolution of ligands by exponential enrichment (SELEX) approach was used to select RNA sequences that are preferentially bound by the ORF57 protein. Results identified a recurring motif, GAAGRG, within the majority of selected RNAs, which is also present in many late HVS mRNAs. RNA immunopreciptations demonstrated that disruption of this motif within a viral intronless RNA ablates ORF57 binding. These data suggest that the GAAGRG motif may be required within a HVS intronless mRNA for recognition by the ORF57 protein.


Asunto(s)
Herpesvirus Saimiriino 2/genética , ARN Mensajero/genética , Proteínas Represoras/metabolismo , Elementos de Respuesta/genética , Transactivadores/metabolismo , Animales , Secuencia de Bases , Biología Computacional/métodos , Herpesvirus Saimiriino 2/metabolismo , Datos de Secuencia Molecular , ARN Mensajero/química , ARN Mensajero/metabolismo , Técnica SELEX de Producción de Aptámeros
12.
J Biol Chem ; 284(1): 505-514, 2009 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-18990693

RESUMEN

Rhesus rhadinovirus (RRV) is currently the closest known, fully sequenced homolog of human Kaposi sarcoma-associated herpesvirus. Both these viruses encode complement inhibitors as follows: Kaposi sarcoma-associated herpesvirus-complement control protein (KCP) and RRV-complement control protein (RCP). Previously we characterized in detail the functional properties of KCP as a complement inhibitor. Here, we performed comparative analyses for two variants of RCP protein, encoded by RRV strains H26-95 and 17577. Both RCP variants and KCP inhibited human and rhesus complement when tested in hemolytic assays measuring all steps of activation via the classical and the alternative pathway. RCP variants from both RRV strains supported C3b and C4b degradation by factor I and decay acceleration of the classical C3 convertase, similar to KCP. Additionally, the 17577 RCP variant accelerated decay of the alternative C3 convertase, which was not seen for KCP. In contrast to KCP, RCP showed no affinity to heparin and is the first described complement inhibitor in which the binding site for C3b/C4b does not interact with heparin. Molecular modeling shows a structural disruption in the region of RCP that corresponds to the KCP-heparin-binding site. This makes RRV a superior model for future in vivo investigations of complement evasion, as RCP does not play a supportive role in viral attachment as KCP does.


Asunto(s)
Proteínas Inactivadoras de Complemento/metabolismo , Proteínas del Sistema Complemento/metabolismo , Herpesvirus Saimiriino 2/metabolismo , Glicoproteínas de Membrana/metabolismo , Modelos Moleculares , Proteínas Virales/metabolismo , Animales , Sitios de Unión , Células CHO , Proteínas Inactivadoras de Complemento/química , Proteínas Inactivadoras de Complemento/genética , Proteínas del Sistema Complemento/química , Proteínas del Sistema Complemento/genética , Cricetinae , Cricetulus , Heparina/química , Heparina/metabolismo , Herpesvirus Saimiriino 2/química , Herpesvirus Saimiriino 2/genética , Herpesvirus Humano 8/química , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/metabolismo , Humanos , Macaca mulatta , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/genética , Especificidad de la Especie , Relación Estructura-Actividad , Proteínas Virales/química , Proteínas Virales/genética , Acoplamiento Viral
13.
Front Biosci ; 13: 2928-38, 2008 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-17981766

RESUMEN

Herpesvirus saimiri (HVS) is the prototype gamma-2 herpesvirus and is a useful model to study the basic mechanisms of lytic replication in this herpesvirus subfamily. This review focuses upon the role of an essential lytic protein, ORF57, which is functionally conserved in all classes of herpesviruses. ORF57 is a multidomain, multifunctional protein responsible for both activation and repression of viral gene expression at a post-transcriptional level. ORF57-mediated repression of gene expression is determined by mRNA processing signals, in particular the presence of an intron within the target gene. This may also be linked to the ability of ORF57 to redistribute SC-35 and U2 splicing factors into specific nuclear domains. ORF57 also plays a pivotal role in transactivating viral gene expression by specifically mediating the nuclear export of HVS intronless transcripts. ORF57 has the ability to shuttle between the nucleus and the cytoplasm, bind viral RNA and recruit cellular nuclear export proteins, such as hTREX components and TAP, onto the viral mRNA. This enables the efficient nuclear export and cytoplasmic accumulation of virus intronless mRNA.


Asunto(s)
Núcleo Celular/virología , Regulación Viral de la Expresión Génica , Herpesvirus Saimiriino 2/metabolismo , Proteínas Represoras/fisiología , Transactivadores/fisiología , Proteínas Virales/fisiología , Transporte Activo de Núcleo Celular , Nucléolo Celular/metabolismo , Perfilación de la Expresión Génica , Modelos Biológicos , Unión Proteica , Procesamiento Postranscripcional del ARN , ARN Mensajero/metabolismo , ARN Viral/metabolismo , Proteínas Represoras/genética , Transactivadores/genética , Transcripción Genética , Proteínas Virales/genética , Proteínas Virales/metabolismo
14.
J Biochem Mol Biol ; 40(3): 341-8, 2007 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-17562285

RESUMEN

Herpesvirus saimiri (HVS), a member of the gamma-herpesvirus family, encodes an oncoprotein called Saimiri Transforming Protein (STP) which is required for lymphoma induction in non-human primates. Previous study has shown that STP-C, an oncoprotein of HVS, activates NF-kappaB signaling pathway. However, the detailed mechanism of STP-C-mediated NF-kappaB activation has not been reported yet. We first report that STP-C interacts with TRAF6 protein in vivo and in vitro and further investigation shows that Glu(12) residue of STP-C is critical for binding to TRAF6. Introduction of ubiquitin together with STP-C augments NF-kappaB activity compared to that of STP-C expression alone. STP-C expression further induces ubiquitination of endogenous TRAF6. In addition, either a deubiquitination enzyme, CYLD or a dominant negative E2-conjugation enzyme reduced NF-kappaB activity in spite of the presence of STP-C, supporting that the interaction between STP-C and TRAF6 induces ubiquitination of TRAF6. NF-kappaB activation by STP-C through the ubiquitinated TRAF6 causes the increased production of IL-8, an inflammatory chemokine and the enhanced expression of costimulatory molecule ICAM, which might ultimately contribute cellular transformation by the exposure of HVS-infected cells with inflammatory microenvironment and chronic activation.


Asunto(s)
Herpesvirus Saimiriino 2/metabolismo , FN-kappa B/metabolismo , Proteínas Oncogénicas Virales/metabolismo , Factor 6 Asociado a Receptor de TNF/metabolismo , Moléculas de Adhesión Celular/metabolismo , Línea Celular , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Glutatión Transferasa/genética , Glutatión Transferasa/metabolismo , Células HeLa , Humanos , Immunoblotting , Inmunoprecipitación , Interleucina-8/metabolismo , FN-kappa B/genética , Proteínas Oncogénicas Virales/genética , Unión Proteica , ARN Interferente Pequeño/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Factor 6 Asociado a Receptor de TNF/genética , Transfección , Ubiquitina/metabolismo
15.
Exp Mol Med ; 39(1): 56-64, 2007 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-17334229

RESUMEN

Herpesvirus saimiri (HVS), a member of the gamma-herpesvirus family, encodes an oncoprotein called Saimiri Transforming Protein (STP) which is required for lymphoma induction in non-human primates. However, a detailed mechanism of STP-A11-induced oncogenesis has not been revealed yet. We first report that STP-A11 oncoprotein interacts with TNF-alpha receptor-associated factor (TRAF) 6 in vivo and in vitro. Mutagenesis analysis of the TRAF6-binding motif (10)PQENDE(15) in STP-A11 reveals that Glu (E)(12) residue is critical for binding to TRAF6 and NF-kappaB activation. Interestingly, co-expression of E12A mutant, lack of TRAF6 binding, with cellular Src (Src) results in decreased transcriptional activity of Stat3 and AP-1, a novel target of STP-A11 compared to that of wild type. Furthermore, the presence of STP-A11 enhances the association of TRAF6 with Src and induces the translocation of both TRAF6 and Src to a nonionic detergent-insoluble fraction. Taken together, these studies suggest that STP-A11 oncoprotein up-regulates both NF-kappaB and AP-1 transcription activity through TRAF6, which would ultimately contribute cellular transformation.


Asunto(s)
Herpesvirus Saimiriino 2/metabolismo , FN-kappa B/metabolismo , Proteínas Oncogénicas Virales/metabolismo , Factor 6 Asociado a Receptor de TNF/metabolismo , Factor de Transcripción AP-1/metabolismo , Transcripción Genética , Línea Celular , Detergentes , Humanos , Iones , FN-kappa B/agonistas , Unión Proteica , Proteínas Proto-Oncogénicas pp60(c-src)/metabolismo , Factor de Transcripción STAT3/metabolismo , Solubilidad , Factor de Transcripción AP-1/agonistas
16.
Proc Natl Acad Sci U S A ; 103(41): 15190-5, 2006 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-17005724

RESUMEN

The nucleolus is the largest subnuclear structure and is plurifunctional in nature. Here, we demonstrate that nucleolar localization of a key herpesvirus regulatory protein is essential for its role in virus mRNA nuclear export. The herpesvirus saimiri ORF57 protein is a nucleocytoplasmic shuttle protein that is conserved in all herpesviruses and orchestrates the nuclear export of viral intronless mRNAs. We demonstrate that expression of the ORF57 protein induces nucleolar redistribution of human TREX (transcription/export) proteins that are involved in mRNA nuclear export. Moreover, we describe a previously unidentified nucleolar localization signal within ORF57 that is composed of two distinct nuclear localization signals. Intriguingly, point mutations that ablate ORF57 nucleolar localization lead to a failure of ORF57-mediated viral mRNA nuclear export. Furthermore, nucleolar retargeting of the ORF57 mutant was achieved by the incorporation of the HIV-1 Rev nucleolar localization signal, and analysis demonstrated that this modification was sufficient to restore viral mRNA nuclear export. This finding represents a unique and fundamental role for the nucleolus in nuclear export of viral mRNA.


Asunto(s)
Nucléolo Celular/metabolismo , Herpesvirus Saimiriino 2/genética , Intrones , ARN Mensajero/metabolismo , ARN Viral/metabolismo , Transporte Activo de Núcleo Celular/genética , Animales , Aotidae , Línea Celular , Células HeLa , Herpesvirus Saimiriino 2/metabolismo , Humanos , Intrones/genética , Proteínas Represoras/biosíntesis , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Transactivadores/biosíntesis , Transactivadores/genética , Transactivadores/metabolismo , Proteínas Virales/biosíntesis , Proteínas Virales/genética , Proteínas Virales/metabolismo
17.
Virology ; 352(2): 253-67, 2006 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-16780912

RESUMEN

Herpesvirus saimiri (HVS)-transformed human T cells become permissive for X4 and R5 strains of human immunodeficiency virus type 1 (HIV-1), evidence that HVS-encoded proteins associated with T cell transformation enhance HIV-1 replication. Analyzing the contribution of transformation-associated bicistronic HVS open reading frames (ORF) to HIV-1 replication revealed expression of the second ORF saimiri transformation-associated protein type C (StpC) conferred the permissive phenotype to T cells. In contrast, expression of the first HVS ORF tyrosine-kinase interacting protein (Tip) in the absence of StpC enhanced restriction of HIV-1 replication in T cell lines and peripheral blood mononuclear cells. Understanding the mechanism whereby Tip enhanced restriction of HIV-1 replication may uncover unique pathways that could be targeted therapeutically. Here we report that Tip restricts HIV-1 replication in a monocyte-derived cell line and restricts reactivation of replication of HIV-1 in a T cell line harboring provirus. In this report, we begin to unravel the molecular underpinnings of Tip-mediated restriction. Tip mediates both lymphocyte-cell-specific kinase (Lck)-dependent and -independent effects on HIV-1 replication. We also provide evidence that Tip-mediated restriction is in part due to inhibition of Tat transactivation of the HIV-1 long terminal repeat (LTR). Expression of Tip in T cells increased activation of Stat1 and Stat3, as well as activation of protein kinase RNA-dependent (PKR/p68) and interferon-gamma production. Taken together, these results provide evidence that Tip restricts HIV-1 replication and reactivation by inhibiting HIV-1 transcription while inducing an intercellular antiviral state. We propose that genetically engineered vectors driving Tip expression could provide a prototypic strategy for restricting HIV-1 replication and reactivation in diverse cell lineages.


Asunto(s)
Productos del Gen tat/antagonistas & inhibidores , VIH-1/patogenicidad , Herpesvirus Saimiriino 2/metabolismo , Fosfoproteínas/metabolismo , Proteínas Virales/metabolismo , Línea Celular , Efecto Citopatogénico Viral , Duplicado del Terminal Largo de VIH , VIH-1/inmunología , VIH-1/fisiología , Herpesvirus Saimiriino 2/genética , Humanos , Células Jurkat , Proteína Tirosina Quinasa p56(lck) Específica de Linfocito/metabolismo , Monocitos/inmunología , Monocitos/metabolismo , Monocitos/virología , Fosfoproteínas/genética , Factor de Transcripción STAT1/metabolismo , Factor de Transcripción STAT3/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Linfocitos T/virología , Transducción Genética , Transformación Genética , Células U937 , Proteínas Virales/genética , Replicación Viral , eIF-2 Quinasa/metabolismo , Productos del Gen tat del Virus de la Inmunodeficiencia Humana
18.
J Biol Chem ; 281(32): 23119-28, 2006 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-16760474

RESUMEN

Herpesvirus saimiri (HVS) is a lymphotropic virus that causes T-cell lymphomas in New World primates. It encodes a structural homolog of complement control proteins named complement control protein homolog (CCPH). Previously, CCPH has been shown to inhibit C3d deposition on target cells exposed to complement. Here we have studied the mechanism by which it inactivates complement. We have expressed the soluble form of CCPH in Escherichia coli, purified to homogeneity and compared its activity to vaccinia virus complement control protein (VCP) and human complement regulators factor H and soluble complement receptor 1. The expressed soluble form of CCPH bound to C3b (KD = 19.2 microm) as well as to C4b (KD = 0.8 microm) and accelerated the decay of the classical/lectin as well as alternative pathway C3-convertases. In addition, it also served as factor I cofactor and supported factor I-mediated inactivation of both C3b and C4b. Time course analysis indicated that although its rate of inactivation of C4b is comparable with VCP, it is 14-fold more potent than VCP in inactivating C3b. Site-directed mutagenesis revealed that Arg-118, which corresponds to Lys-120 of variola virus complement regulator SPICE (a residue critical for its enhanced C3b cofactor activity), contributes significantly in enhancing this activity. Thus, our data indicate that HVS encodes a potent complement inhibitor that allows HVS to evade the host complement attack.


Asunto(s)
Fibrinógeno/química , Herpesvirus Saimiriino 2/metabolismo , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/genética , Proteínas Virales/química , Proteínas Virales/genética , Arginina/química , Complemento C3b/química , Factor H de Complemento/química , Escherichia coli/metabolismo , Humanos , Cinética , Lectinas/química , Lisina/química , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Receptores de Complemento/química
19.
J Virol ; 80(1): 108-18, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16352535

RESUMEN

Lipid rafts are membrane microdomains that are proposed to function as platforms for both receptor signaling and trafficking. Our previous studies have demonstrated that Tip of herpesvirus saimiri (HVS), which is a T-lymphotropic tumor virus, is constitutively targeted to lipid rafts and interacts with cellular Lck tyrosine kinase and p80 WD repeat-containing endosomal protein. Through the interactions with Lck and p80, HVS Tip modulates diverse T-cell functions, which leads to the downregulation of T-cell receptor (TCR) and CD4 coreceptor surface expression, the inhibition of TCR signal transduction, and the activation of STAT3 transcription factor. In this study, we investigated the functional significance of Tip association with lipid rafts. We found that Tip expression remarkably increased lipid raft fractions in human T cells by enhancing the recruitment of lipid raft-resident proteins. Genetic analysis showed that the carboxyl-terminal transmembrane, but not p80 and Lck interaction, of Tip was required for the lipid raft localization and that lipid raft localization of Tip was necessary for the efficient downregulation of TCR and CD4 surface expression. Correlated with this, treatment with Filipin III, a lipid raft-disrupting agent, effectively reversed the downregulation of CD3 and CD4 surface expression induced by Tip. On the other hand, Tip mutants that were no longer present in lipid rafts were still capable of inhibiting TCR signaling and activating STAT3 transcription factor activity as efficiently as wild-type (wt) Tip. These results indicate that the association of Tip with lipid rafts is essential for the downregulation of TCR and CD4 surface expression but not for the inhibition of TCR signal transduction and the activation of STAT3 transcription factor. These results also suggest that the signaling and targeting activities of HVS Tip rely on functionally and genetically separable mechanisms, which may independently modulate T-cell function for viral persistence or pathogenesis.


Asunto(s)
Antígenos CD4/metabolismo , Herpesvirus Saimiriino 2/fisiología , Lípidos/fisiología , Receptores de Antígenos de Linfocitos T/fisiología , Transducción de Señal/fisiología , Línea Celular , Regulación hacia Abajo , Herpesvirus Saimiriino 2/genética , Herpesvirus Saimiriino 2/metabolismo
20.
Artículo en Inglés | MEDLINE | ID: mdl-17381320

RESUMEN

Some gammaherpesviruses encode nuclear noncoding RNAs (ncRNAs) that assemble with host proteins. Their conservation and abundance implies that they serve important functions for the virus. This paper focuses on our studies of three classes of nuclear noncoding herpesvirus RNAs. (1) EBERs 1 and 2 are expressed by Epstein-Barr virus in latent infection of human B lymphocytes. Recent studies revealed three sites on EBER1 that associate with ribosomal protein L22. In addition, heterokaryon assays have definitively shown that both EBERs are confined to the nucleus, arguing that their contribution to viral latency is purely nuclear. (2) HSURs 1-7 are U RNAs encoded by Herpesvirus saimiri, which causes aggressive T-cell leukemias and lymphomas. Comparison of monkey T cells transformed with wild-type or mutant virus lacking HSURs 1 and 2 revealed significant changes in host mRNAs implicated in T-cell signaling. (3) PAN is a 1-kb polyadenylated RNA that accumulates in the nucleus of Kaposi's sarcoma-associated herpesvirus lytically infected cells. A novel element, the ENE, is essential for its high accumulation. Recent results indicate that the ENE functions to counteract poly(A)-dependent RNA degradation, which we propose contributes to nuclear surveillance of mRNA transcripts in mammalian cells. Continuing studies of these viral RNAs will provide insights into both cellular and viral gene expression.


Asunto(s)
ARN Viral/genética , ARN Viral/metabolismo , Ribonucleoproteínas Nucleares Pequeñas/genética , Ribonucleoproteínas Nucleares Pequeñas/metabolismo , Animales , Linfocitos B/virología , Secuencia de Bases , Herpesvirus Saimiriino 2/genética , Herpesvirus Saimiriino 2/metabolismo , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/metabolismo , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/metabolismo , Humanos , Datos de Secuencia Molecular , Conformación de Ácido Nucleico , ARN Viral/química , Ribonucleoproteínas Nucleares Pequeñas/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA