Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
PLoS Pathog ; 15(8): e1007980, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31461506

RESUMEN

Almost one third of herpesvirus proteins are expressed with late kinetics. Many of these late proteins serve crucial structural functions such as formation of virus particles, attachment to host cells and internalization. Recently, we and others identified a group of Epstein-Barr virus early proteins that form a pre-initiation complex (vPIC) dedicated to transcription of late genes. Currently, there is a fundamental gap in understanding the role of post-translational modifications in regulating assembly and function of the complex. Here, we used mass spectrometry to map potential phosphorylation sites in BGLF3, a core component of the vPIC module that connects the BcRF1 viral TATA box binding protein to other components of the complex. We identified threonine 42 (T42) in BGLF3 as a phosphoacceptor residue. T42 is conserved in BGLF3 orthologs encoded by other gamma herpesviruses. Abolishing phosphorylation at T42 markedly reduced expression of vPIC-dependent late genes and disrupted production of new virus particles, but had no effect on early gene expression, viral DNA replication, or expression of vPIC-independent late genes. We complemented failure of BGLF3(T42A) to activate late gene expression by ectopic expression of other components of vPIC. Only BFRF2 and BVLF1 were sufficient to suppress the defect in late gene expression associated with BGLF3(T42A). These results were corroborated by the ability of wild type BGLF3 but not BGLF3(T42A) to form a trimeric complex with BFRF2 and BVLF1. Our findings suggest that phosphorylation of BGLF3 at threonine 42 serves as a new checkpoint for subsequent formation of BFRF2:BGLF3:BVLF1; a trimeric subcomplex essential for transcription of late genes. Our findings provide evidence that post-translational modifications regulate the function of the vPIC nanomachine that initiates synthesis of late transcripts in herpesviruses.


Asunto(s)
Replicación del ADN , Regulación Viral de la Expresión Génica , Herpesvirus Humano 4/genética , Treonina/metabolismo , Transcripción Genética , Proteínas Virales/genética , Proteínas Virales/metabolismo , Secuencia de Aminoácidos , ADN Viral/genética , Células HEK293 , Humanos , Mutación , Fosforilación , Unión Proteica , Homología de Secuencia , Treonina/química , Treonina/genética , Proteínas Virales/química , Replicación Viral
2.
J Virol ; 93(17)2019 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-31189703

RESUMEN

Herpesviruses are ubiquitous, and infection by some, like Epstein-Barr virus (EBV), is nearly universal. To persist, EBV must periodically switch from a latent to a replicative/lytic phase. This productive phase is responsible for most herpesvirus-associated diseases. EBV encodes a latency-to-lytic switch protein which, upon activation, sets off a vectorially constrained cascade of gene expression that results in production of infectious virus. While triggering expression of the switch protein ZEBRA is essential to lytic cycle entry, sustaining its expression is equally important to avoid premature termination of the lytic cascade. We report that the viral protein kinase (vPK), encoded by a gene that is kinetically downstream of the lytic switch, sustains expression of ZEBRA, amplifies the lytic cascade, increasing virus production, and, importantly, prevents the abortive lytic cycle. We find that vPK, through a noncanonical site phosphorylation, activates the cellular phosphatidylinositol 3-kinase-related kinase ATM to cause phosphorylation of the heterochromatin enforcer KAP1/TRIM28 even in the absence of EBV genomes or other EBV proteins. Phosphorylation of KAP1 renders it unable to restrain ZEBRA, thereby further derepressing and sustaining its expression to culminate in virus production. This partnership with a host kinase and a transcriptional corepressor enables retrograde regulation by vPK of ZEBRA, an observation that is counter to the unidirectional regulation of gene expression reminiscent of most DNA viruses.IMPORTANCE Herpesviruses infect nearly all humans and persist quiescently for the life of the host. These viruses intermittently activate into the lytic phase to produce infectious virus, thereby causing disease. To ensure that lytic activation is not prematurely terminated, expression of the virally encoded lytic switch protein needs to be sustained. In studying Epstein-Barr virus, one of the most prevalent human herpesviruses that also causes cancer, we have discovered that a viral kinase activated by the viral lytic switch protein partners with a cellular kinase to deactivate a silencer of the lytic switch protein, thereby providing a positive feedback loop to ensure successful completion of the viral productive phase. Our findings highlight key nodes of interaction between the host and virus that could be exploited to treat lytic phase-associated diseases by terminating the lytic phase or kill cancer cells harboring herpesviruses by accelerating the completion of the lytic cascade.


Asunto(s)
Herpesvirus Humano 4/fisiología , Proteínas Serina-Treonina Quinasas/metabolismo , Transactivadores/genética , Proteínas Virales/metabolismo , Latencia del Virus , Línea Celular , Línea Celular Tumoral , Epigénesis Genética , Células HEK293 , Humanos , Fosforilación , Transactivadores/metabolismo , Proteína 28 que Contiene Motivos Tripartito/metabolismo
3.
J Virol ; 93(14)2019 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-31068430

RESUMEN

The pathogenesis of Epstein-Barr virus (EBV) infection, including development of lymphomas and carcinomas, is dependent on the ability of the virus to transit from latency to the lytic phase. This conversion, and ultimately disease development, depends on the molecular switch protein, ZEBRA, a viral bZIP transcription factor that initiates transcription from promoters of viral lytic genes. By binding to the origin of viral replication, ZEBRA is also an essential replication protein. Here, we identified a novel DNA-binding motif of ZEBRA, N terminal to the canonical bZIP domain. This RRTRK motif is important for high-affinity binding to DNA and is essential for recognizing the methylation state of viral promoters. Mutations in this motif lead to deficiencies in DNA binding, recognition of DNA methylation, lytic cycle DNA replication, and viral late gene expression. This work advances our understanding of ZEBRA-dependent activation of the viral lytic cascade.IMPORTANCE The binding of ZEBRA to methylated and unmethylated viral DNA triggers activation of the EBV lytic cycle, leading to viral replication and, in some patients, cancer development. Our work thoroughly examines how ZEBRA uses a previously unrecognized basic motif to bind nonmethylated and methylated DNA targets, leading to viral lytic activation. Our findings show that two different positively charged motifs, including the canonical BZIP domain and a newly identified RRTRK motif, contribute to the mechanism of DNA recognition by a viral AP-1 protein. This work contributes to the assessment of ZEBRA as a potential therapeutic target for antiviral and oncolytic treatments.


Asunto(s)
Metilación de ADN/fisiología , ADN Viral/metabolismo , Regulación Viral de la Expresión Génica/fisiología , Herpesvirus Humano 4/fisiología , Regiones Promotoras Genéticas/fisiología , Transactivadores/metabolismo , Activación Viral/fisiología , Secuencias de Aminoácidos , Línea Celular Tumoral , ADN Viral/genética , Células HEK293 , Humanos , Mutación , Dominios Proteicos , Transactivadores/genética
4.
J Virol ; 92(19)2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-30021895

RESUMEN

Epstein-Barr virus (EBV) ZEBRA protein activates the EBV lytic cycle. Cellular AP-1 proteins with alanine-to-serine [AP-1(A/S)] substitutions homologous to ZEBRA(S186) assume some functions of EBV ZEBRA. These AP-1(A/S) mutants bind methylated EBV DNA and activate expression of some EBV genes. Here, we compare expression of 67 viral genes induced by ZEBRA versus expression induced by AP-1(A/S) proteins. AP-1(A/S) activated 24 genes to high levels and 15 genes to intermediate levels; activation of 28 genes by AP-1(A/S) was severely impaired. We show that AP-1(A/S) proteins are defective at stimulating viral lytic DNA replication. The impairment of expression of many late genes compared to that of ZEBRA is likely due to the inability of AP-1(A/S) proteins to promote viral DNA replication. However, even in the absence of detectable viral DNA replication, AP-1(A/S) proteins stimulated expression of a subgroup of late genes that encode viral structural proteins and immune modulators. In response to ZEBRA, expression of this subgroup of late genes was inhibited by phosphonoacetic acid (PAA), which is a potent viral replication inhibitor. However, when the lytic cycle was activated by AP-1(A/S), PAA did not reduce expression of this subgroup of late genes. We also provide genetic evidence, using the BMRF1 knockout bacmid, that these genes are true late genes in response to ZEBRA. AP-1(A/S) binds to the promoter region of at least one of these late genes, BDLF3, encoding an immune modulator.IMPORTANCE Mutant c-Jun and c-Fos proteins selectively activate expression of EBV lytic genes, including a subgroup of viral late genes, in the absence of viral DNA replication. These findings indicate that newly synthesized viral DNA is not invariably required for viral late gene expression. While viral DNA replication may be obligatory for late gene expression driven by viral transcription factors, it does not limit the ability of cellular transcription factors to activate expression of some viral late genes. Our results show that expression of all late genes may not be strictly dependent on viral lytic DNA replication. The c-Fos A151S mutation has been identified in a human cancer. c-Fos A151S in combination with wild-type c-Jun activates the EBV lytic cycle. Our data provide proof of principle that mutant cellular transcription factors could cause aberrant regulation of viral lytic cycle gene expression and play important roles in EBV-associated diseases.


Asunto(s)
Antígenos Virales/genética , ADN Viral/genética , Herpesvirus Humano 4/genética , Interacciones Huésped-Patógeno , Glicoproteínas de Membrana/genética , Transactivadores/genética , Factor de Transcripción AP-1/genética , Proteínas Virales/genética , Sustitución de Aminoácidos , Antígenos Virales/inmunología , Antivirales/farmacología , Sitios de Unión , Línea Celular Tumoral , Metilación de ADN/efectos de los fármacos , ADN Viral/inmunología , Regulación de la Expresión Génica , Células HEK293 , Herpesvirus Humano 4/efectos de los fármacos , Herpesvirus Humano 4/inmunología , Humanos , Linfocitos/inmunología , Linfocitos/virología , Glicoproteínas de Membrana/inmunología , Mutación , Ácido Fosfonoacético/farmacología , Regiones Promotoras Genéticas , Unión Proteica , Transducción de Señal , Transactivadores/inmunología , Factor de Transcripción AP-1/inmunología , Proteínas Virales/inmunología , Replicación Viral/efectos de los fármacos
5.
J Immunol ; 198(8): 3029-3032, 2017 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-28264970

RESUMEN

Pregnant women have greater mortality and complications associated with viral infections compared with the general population, but the reason for the increased susceptibility is not well defined. Placenta type I IFN is an important immune modulator and protects the pregnancy. We hypothesized that loss of placental IFN affects the regulation of the maternal immune system, resulting in the differential response to infections observed in pregnancy. Pregnant mice lacking the IFN-α/ß receptor (IFNAR) became viremic and had higher mortality compared with nonpregnant animals. Notably, an embryo with functional IFN signaling alone was sufficient to rescue the pregnant IFNAR-/- dam from virus-associated demise. Placental IFN was also an important regulator of viral replication in placental tissue and significantly affected viral transmission to the fetus. These findings highlight the role of fetal/placental IFN in the modulation of viral infection in the mother and fetus.


Asunto(s)
Feto/inmunología , Infecciones por Herpesviridae/inmunología , Interferón Tipo I/inmunología , Placenta/inmunología , Complicaciones Infecciosas del Embarazo/inmunología , Animales , Femenino , Genotipo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Embarazo , Rhadinovirus/inmunología , Infecciones Tumorales por Virus/inmunología , Carga Viral
6.
PLoS Pathog ; 12(11): e1006008, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27855219

RESUMEN

Subversion of host immune surveillance is a crucial step in viral pathogenesis. Epstein-Barr virus (EBV) encodes two immune evasion gene products, BCRF1 (viral IL-10) and BPLF1 (deubiquitinase/deneddylase); both proteins suppress antiviral immune responses during primary infection. The BCRF1 and BPLF1 genes are expressed during the late phase of the lytic cycle, an essential but poorly understood phase of viral gene expression. Several late gene regulators recently identified in beta and gamma herpesviruses form a viral pre-initiation complex for transcription. Whether each of these late gene regulators is necessary for transcription of all late genes is not known. Here, studying viral gene expression in the absence and presence of siRNAs to individual components of the viral pre-initiation complex, we identified two distinct groups of late genes. One group includes late genes encoding the two immunoevasins, BCRF1 and BPLF1, and is transcribed independently of the viral pre-initiation complex. The second group primarily encodes viral structural proteins and is dependent on the viral pre-initiation complex. The protein kinase BGLF4 is the only known late gene regulator necessary for expression of both groups of late genes. ChIP-seq analysis showed that the transcription activator Rta associates with the promoters of eight late genes including genes encoding the viral immunoevasins. Our results demonstrate that late genes encoding immunomodulatory proteins are transcribed by a mechanism distinct from late genes encoding viral structural proteins. Understanding the mechanisms that specifically regulate expression of the late immunomodulatory proteins could aid the development of antiviral drugs that impair immune evasion by the oncogenic EB virus.


Asunto(s)
Infecciones por Virus de Epstein-Barr/genética , Regulación Viral de la Expresión Génica/fisiología , Herpesvirus Humano 4/genética , Evasión Inmune/genética , Proteínas Virales/biosíntesis , Proteínas Reguladoras y Accesorias Virales/biosíntesis , Western Blotting , Inmunoprecipitación de Cromatina , Infecciones por Virus de Epstein-Barr/inmunología , Técnicas de Silenciamiento del Gen , Células HEK293 , Herpesvirus Humano 4/inmunología , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Evasión Inmune/inmunología , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteínas Virales/genética , Proteínas Reguladoras y Accesorias Virales/genética , Proteínas Estructurales Virales/biosíntesis
7.
PLoS Pathog ; 10(8): e1004307, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25166506

RESUMEN

The mechanism regulating expression of late genes, encoding viral structural components, is an unresolved problem in the biology of DNA tumor viruses. Here we show that BGLF4, the only protein kinase encoded by Epstein-Barr virus (EBV), controls expression of late genes independent of its effect on viral DNA replication. Ectopic expression of BGLF4 in cells lacking the kinase gene stimulated the transcript levels of six late genes by 8- to 10-fold. Introduction of a BGLF4 mutant that eliminated its kinase activity did not stimulate late gene expression. In cells infected with wild-type EBV, siRNA to BGLF4 (siG4) markedly reduced late gene expression without compromising viral DNA replication. Synthesis of late products was restored upon expression of a form of BGLF4 resistant to the siRNA. Studying the EBV transcriptome using mRNA-seq during the late phase of the lytic cycle in the absence and presence of siG4 showed that BGLF4 controlled expression of 31 late genes. Analysis of the EBV transcriptome identified BGLF3 as a gene whose expression was reduced as a result of silencing BGLF4. Knockdown of BGLF3 markedly reduced late gene expression but had no effect on viral DNA replication or expression of BGLF4. Our findings reveal the presence of a late control locus encompassing BGLF3 and BGLF4 in the EBV genome, and provide evidence for the importance of both proteins in post-replication events that are necessary for expression of late genes.


Asunto(s)
Regulación Viral de la Expresión Génica/genética , Proteínas Serina-Treonina Quinasas/biosíntesis , Proteínas Serina-Treonina Quinasas/genética , Proteínas Virales/biosíntesis , Proteínas Virales/genética , Proteínas Estructurales Virales/biosíntesis , Proteínas Estructurales Virales/genética , Northern Blotting , Sitios Genéticos , Células HEK293 , Humanos , Reacción en Cadena en Tiempo Real de la Polimerasa , Transfección , Replicación Viral/genética
8.
Curr Top Microbiol Immunol ; 391: 237-61, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26428377

RESUMEN

Epstein-Barr virus, which mainly infects B cells and epithelial cells, has two modes of infection: latent and lytic. Epstein-Barr virus infection is predominantly latent; however, lytic infection is detected in healthy seropositive individuals and becomes more prominent in certain pathological conditions. Lytic infection is divided into several stages: early gene expression, DNA replication, late gene expression, assembly, and egress. This chapter summarizes the most recent progress made toward understanding the molecular mechanisms that regulate the different lytic stages leading to production of viral progeny. In addition, the chapter highlights the potential role of lytic infection in disease development and current attempts to purposely induce lytic infection as a therapeutic approach.


Asunto(s)
Infecciones por Virus de Epstein-Barr/virología , Herpesvirus Humano 4/fisiología , Activación Viral , Animales , Linfocitos B/virología , Regulación Viral de la Expresión Génica , Herpesvirus Humano 4/genética , Humanos , Latencia del Virus
9.
J Virol ; 87(1): 208-23, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23077295

RESUMEN

Two transcription factors, ZEBRA and Rta, switch Epstein-Barr virus (EBV) from the latent to the lytic state. While ZEBRA also plays an obligatory role as an activator of replication, it is not known whether Rta is directly required for replication. Rta is dispensable for amplification of an oriLyt-containing plasmid in a transient-replication assay. Here, we assessed the requirement for Rta in activation of viral DNA synthesis from the endogenous viral genome, a function that has not been established. Initially, we searched for a ZEBRA mutant that supports viral replication but not transcription. We found that Z(S186A), a mutant of ZEBRA unable to activate transcription of Rta or viral genes encoding replication proteins, is competent to bind to oriLyt and to function as an origin recognition protein. Ectopic expression of the six components of the EBV lytic replication machinery failed to rescue replication by Z(S186A). However, addition of Rta to Z(S186A) and the mixture of replication factors activated viral replication and late gene expression. Deletion mutagenesis of Rta indicated that the C-terminal 10 amino acids (aa) were essential for the function of Rta in replication. In vivo DNA binding studies revealed that Rta interacted with the enhancer region of oriLyt. In addition, expression of Rta and Z(S186A) together, but not individually, activated synthesis of the BHLF1 transcript, a lytic transcript required for the process of viral DNA replication. Our findings demonstrate that Rta plays an indispensable role in the process of lytic DNA replication.


Asunto(s)
Replicación del ADN , Herpesvirus Humano 4/fisiología , Proteínas Inmediatas-Precoces/metabolismo , Transactivadores/metabolismo , Replicación Viral , Línea Celular , Análisis Mutacional de ADN , ADN Viral/metabolismo , Eliminación de Gen , Herpesvirus Humano 4/genética , Humanos , Proteínas Inmediatas-Precoces/genética , Transactivadores/genética
10.
PLoS Pathog ; 6(8): e1001054, 2010 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-20808903

RESUMEN

ZEBRA is a site-specific DNA binding protein that functions as a transcriptional activator and as an origin binding protein. Both activities require that ZEBRA recognizes DNA motifs that are scattered along the viral genome. The mechanism by which ZEBRA discriminates between the origin of lytic replication and promoters of EBV early genes is not well understood. We explored the hypothesis that activation of replication requires stronger association between ZEBRA and DNA than does transcription. A ZEBRA mutant, Z(S173A), at a phosphorylation site and three point mutants in the DNA recognition domain of ZEBRA, namely Z(Y180E), Z(R187K) and Z(K188A), were similarly deficient at activating lytic DNA replication and expression of late gene expression but were competent to activate transcription of viral early lytic genes. These mutants all exhibited reduced capacity to interact with DNA as assessed by EMSA, ChIP and an in vivo biotinylated DNA pull-down assay. Over-expression of three virally encoded replication proteins, namely the primase (BSLF1), the single-stranded DNA-binding protein (BALF2) and the DNA polymerase processivity factor (BMRF1), partially rescued the replication defect in these mutants and enhanced ZEBRA's interaction with oriLyt. The findings demonstrate a functional role of replication proteins in stabilizing the association of ZEBRA with viral DNA. Enhanced binding of ZEBRA to oriLyt is crucial for lytic viral DNA replication.


Asunto(s)
Replicación del ADN/fisiología , Regulación Viral de la Expresión Génica/fisiología , Herpesvirus Humano 4/fisiología , Transactivadores/metabolismo , Replicación Viral/fisiología , Northern Blotting , Southern Blotting , Línea Celular , Inmunoprecipitación de Cromatina , Ensayo de Cambio de Movilidad Electroforética , Expresión Génica , Genes Virales , Humanos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección , Proteínas Virales/metabolismo
11.
J Virol ; 84(2): 993-1004, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19889776

RESUMEN

A fundamental problem in studying the latent-to-lytic switch of Epstein-Barr virus (EBV) and the viral lytic cycle itself is the lack of a culture system fully permissive to lytic cycle induction. Strategies to target EBV-positive tumors by inducing the viral lytic cycle with chemical agents are hindered by inefficient responses to stimuli. In vitro, even in the most susceptible cell lines, more than 50% of cells latently infected with EBV are refractory to induction of the lytic cycle. The mechanisms underlying the refractory state are not understood. We separated lytic from refractory Burkitt lymphoma-derived HH514-16 cells after treatment with an HDAC inhibitor, sodium butyrate. Both refractory- and lytic-cell populations responded to the inducing stimulus by hyperacetylation of histone H3. However, analysis of host cell gene expression showed that specific cellular transcripts Stat3, Fos, and interleukin-8 (IL-8) were preferentially upregulated in the refractory-cell population, while IL-6 was upregulated in the lytic population. STAT3 protein levels were also substantially increased in refractory cells relative to untreated or lytic cells. This increase in de novo expression resulted primarily in unphosphorylated STAT3. Examination of single cells revealed that high levels of STAT3 were strongly associated with the refractory state. The refractory state is manifest in a unique subpopulation of cells that exhibits different cellular responses than do lytic cells exposed to the same stimulus. Identifying characteristics of cells refractory to lytic induction relative to cells that undergo lytic activation will be an important step in developing a better understanding of the regulation of the EBV latent to lytic switch.


Asunto(s)
Linfocitos B , Herpesvirus Humano 4/efectos de los fármacos , Inhibidores de Histona Desacetilasas/farmacología , Factor de Transcripción STAT3/metabolismo , Regulación hacia Arriba , Activación Viral/efectos de los fármacos , Animales , Linfocitos B/efectos de los fármacos , Linfocitos B/virología , Butiratos/farmacología , Línea Celular Transformada , Línea Celular Tumoral , Herpesvirus Humano 4/fisiología , Humanos , Conejos , Latencia del Virus
12.
J Biol Chem ; 284(43): 29269-82, 2009 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-19690161

RESUMEN

Kaposi sarcoma-associated herpesvirus-encoded interleukin-6 (vIL-6) and its human cellular homologue (huIL-6) share similar biological functions. Our previous work showed that N-linked glycosylation was required for optimal function of vIL6 but not huIL-6 (1). Here we describe heterogeneity in the composition of the glycans of the two N-linked sites of vIL-6. The Asn-89 site of vIL-6, found to be required for optimal cytokine function, is composed of complex glycans. The Asn-78 site is composed of high mannose glycans, which are dispensable for cytokine function. N-Linked glycosylation at the Asn-89 site was required for intracellular production of functional vIL-6, but endoglycosidase-mediated removal of N-linked glycans from secreted vIL-6 did not impair protein function. With the use of a conformation-specific antibody and tryptic digestion assays, we showed that glycosylation at the Asn-89 site of vIL-6 affected protein conformation. Human IL-6, but not vIL-6, requires IL-6Ralpha for binding to gp130. We tested the hypothesis that the Asn-89 complex glycan of vIL-6 alone was sufficient to confer binding to gp130 independently of IL-6Ralpha. Two mutants of huIL-6, made to contain additional complex N-linked glycans in the region that interacts with IL-6Ralpha, did not confer binding to gp130 independently of IL-6Ralpha. Our findings support the conclusion that complex glycans on Asn-89 of vIL-6 specifically promote a protein conformation that allows the viral cytokine to bind gp130 independently of IL-6Ralpha.


Asunto(s)
Receptor gp130 de Citocinas/metabolismo , Herpesvirus Humano 8/metabolismo , Interleucina-6/metabolismo , Polisacáridos/metabolismo , Pliegue de Proteína , Receptores de Interleucina-6/metabolismo , Proteínas Virales/metabolismo , Anticuerpos Antivirales/farmacología , Línea Celular Tumoral , Receptor gp130 de Citocinas/genética , Glucosidasas/genética , Glucosidasas/metabolismo , Glicosilación , Herpesvirus Humano 8/genética , Humanos , Interleucina-6/genética , Mutación , Polisacáridos/genética , Unión Proteica/efectos de los fármacos , Unión Proteica/genética , Estructura Terciaria de Proteína/efectos de los fármacos , Receptores de Interleucina-6/genética , Proteínas Virales/genética
13.
J Exp Med ; 199(4): 503-14, 2004 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-14970177

RESUMEN

Kaposi's sarcoma-associated herpesvirus interleukin-6 (vIL-6) is a structural and functional homologue of the human cytokine IL-6 (hIL-6). hIL-6 and vIL-6 exhibit similar biological functions and both act via the gp130 receptor subunit to activate the Janus tyrosine kinase (JAK)1 and signal transducer and activator of transcription (STAT)1/3 pathway. Here we show that vIL-6 is N-linked glycosylated at N78 and N89 and demonstrate that N-linked glycosylation at site N89 of vIL-6 markedly enhances binding to gp130, signaling through the JAK1-STAT1/3 pathway and functions in a cytokine-dependent cell proliferation bioassay. Although hIL-6 is also N-glycosylated at N73 and multiply O-glycosylated, neither N-linked nor O-linked glycosylation is necessary for IL-6 receptor alpha-dependent binding to gp130 or signaling through JAK1-STAT1/3. As distinct from vIL-6, unglycosylated hIL-6 is as potent as glycosylated hIL-6 in stimulating B cell proliferation. These findings highlight distinct functional roles of N-linked glycosylation in viral and cellular IL-6.


Asunto(s)
Herpesvirus Humano 8/inmunología , Interleucina-6/inmunología , Linfocitos B/inmunología , División Celular/inmunología , Línea Celular , Línea Celular Tumoral , Clonación Molecular , Contactinas , Escherichia coli/genética , Glicosilación , Humanos , Interleucina-6/genética , Activación de Linfocitos/inmunología , Moléculas de Adhesión de Célula Nerviosa/inmunología , Fragmentos de Péptidos/química , Plásmidos , Proteínas Recombinantes/inmunología
14.
Am J Reprod Immunol ; 75(4): 451-60, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26892235

RESUMEN

PROBLEM: Preterm birth (PTB) affects approximately 12% of pregnancies and at least 50% of cases have no known risk factors. We hypothesize that subclinical viral infections of the placenta are a factor sensitizing women to intrauterine bacterial infection. Specifically, we propose that viral-induced placental IFN-ß inhibition results in a robust inflammatory response to low concentrations of bacteria. METHODS: Human trophoblast SW.71, C57BL/6, and interferon (IFN) receptor knockout animals were used to determine IFN function. Illumina and Bio-Rad microarrays identified pathways. RESULTS: Inhibiting the IFN-ß pathway resulted in a significant increase in inflammatory cytokines such as IL-1B in response to LPS. Twist was positively correlated with IFN-ß expression and STAT3 phosphorylation and overexpressing Twist reduced IL-1B. Treating IFNAR-/- mice with low-dose LPS at E15.5 caused preterm birth. CONCLUSION: IFN-ß was identified as a key modulator of placental inflammation and, importantly, is commonly affected by viruses. We propose dysregulation of IFN-ß is a major determinant for preterm birth associated with polymicrobial infection.


Asunto(s)
Interferón beta/inmunología , Complicaciones Infecciosas del Embarazo/inmunología , Nacimiento Prematuro/inmunología , Receptor de Interferón alfa y beta/inmunología , Trofoblastos/inmunología , Virosis/inmunología , Animales , Línea Celular Transformada , Femenino , Regulación de la Expresión Génica/inmunología , Humanos , Interferón beta/genética , Interleucina-1beta/genética , Interleucina-1beta/inmunología , Ratones , Ratones Noqueados , Fosforilación/genética , Fosforilación/inmunología , Embarazo , Complicaciones Infecciosas del Embarazo/genética , Complicaciones Infecciosas del Embarazo/patología , Nacimiento Prematuro/genética , Nacimiento Prematuro/microbiología , Nacimiento Prematuro/virología , Receptor de Interferón alfa y beta/genética , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/inmunología , Trofoblastos/microbiología , Trofoblastos/patología , Trofoblastos/virología , Virosis/genética , Virosis/patología
15.
PLoS One ; 9(4): e92593, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24705134

RESUMEN

Many viruses target cytoplasmic polyA binding protein (PABPC) to effect widespread inhibition of host gene expression, a process termed viral host-shutoff (vhs). During lytic replication of Epstein Barr Virus (EBV) we observed that PABPC was efficiently translocated from the cytoplasm to the nucleus. Translocated PABPC was diffusely distributed but was excluded from viral replication compartments. Vhs during EBV infection is regulated by the viral alkaline nuclease, BGLF5. Transfection of BGLF5 alone into BGLF5-KO cells or uninfected 293 cells promoted translocation of PAPBC that was distributed in clumps in the nucleus. ZEBRA, a viral bZIP protein, performs essential functions in the lytic program of EBV, including activation or repression of downstream viral genes. ZEBRA is also an essential replication protein that binds to viral oriLyt and interacts with other viral replication proteins. We report that ZEBRA also functions as a regulator of vhs. ZEBRA translocated PABPC to the nucleus, controlled the intranuclear distribution of PABPC, and caused global shutoff of host gene expression. Transfection of ZEBRA alone into 293 cells caused nuclear translocation of PABPC in the majority of cells in which ZEBRA was expressed. Co-transfection of ZEBRA with BGLF5 into BGLF5-KO cells or uninfected 293 cells rescued the diffuse intranuclear pattern of PABPC seen during lytic replication. ZEBRA mutants defective for DNA-binding were capable of regulating the intranuclear distribution of PABPC, and caused PABPC to co-localize with ZEBRA. One ZEBRA mutant, Z(S186E), was deficient in translocation yet was capable of altering the intranuclear distribution of PABPC. Therefore ZEBRA-mediated nuclear translocation of PABPC and regulation of intranuclear PABPC distribution are distinct events. Using a click chemistry-based assay for new protein synthesis, we show that ZEBRA and BGLF5 each function as viral host shutoff factors.


Asunto(s)
Núcleo Celular/metabolismo , Desoxirribonucleasas/fisiología , Herpesvirus Humano 4 , Proteína I de Unión a Poli(A)/metabolismo , Transactivadores/fisiología , Proteínas Virales/fisiología , Transporte Activo de Núcleo Celular , Núcleo Celular/virología , Células Cultivadas , Infecciones por Virus de Epstein-Barr/virología , Células HEK293 , Herpesvirus Humano 4/patogenicidad , Herpesvirus Humano 4/fisiología , Interacciones Huésped-Patógeno/genética , Humanos , Distribución Tisular , Replicación Viral/genética
17.
J Virol ; 81(7): 3303-16, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17215287

RESUMEN

The Epstein-Barr virus ZEBRA protein controls the viral lytic cycle. ZEBRA activates the transcription of viral genes required for replication. ZEBRA also binds to oriLyt and interacts with components of the viral replication machinery. The mechanism that differentiates the roles of ZEBRA in regulation of transcription and initiation of lytic replication is unknown. Here we show that S173, a residue in the regulatory domain, is obligatory for ZEBRA to function as an origin binding protein but is dispensable for its role as a transcriptional activator of early genes. Serine-to-alanine substitution of this residue, which prevents phosphorylation of S173, resulted in a threefold reduction in the DNA binding affinity of ZEBRA for oriLyt, as assessed by chromatin immunoprecipitation. An independent assay based on ZEBRA solubility demonstrated a marked defect in DNA binding by the Z(S173A) mutant. The phenotype of a phosphomimetic mutant, the Z(S173D) mutant, was similar to that of wild-type ZEBRA. Our findings suggest that phosphorylation of S173 promotes viral replication by enhancing ZEBRA's affinity for DNA. The results imply that stronger DNA binding is required for ZEBRA to activate replication than that required to activate transcription.


Asunto(s)
Replicación del ADN/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Regulación Viral de la Expresión Génica , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/metabolismo , Serina/metabolismo , Transactivadores/genética , Transactivadores/metabolismo , Proteínas Virales/genética , Proteínas Virales/metabolismo , Alanina/genética , Alanina/metabolismo , Desoxirribonucleasa I/metabolismo , Imitación Molecular , Mutación/genética , Fenotipo , Fosforilación , Elementos Reguladores de la Transcripción/genética , Serina/genética , Solubilidad , Factores de Tiempo
18.
J Infect Dis ; 195(4): 483-92, 2007 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-17230407

RESUMEN

Primary Epstein-Barr virus (EBV) infection is characterized by the presence of IgM antibodies to viral capsid antigen and the absence of antibodies to EB nuclear antigen. Here, using a flow cytometry-based assay, we investigated whether IgA antibodies are a marker for primary infection. Serum IgA antibodies in 15 individuals with primary EBV infection reacted with 15%-55.6% of HH514-16 Burkitt lymphoma cells expressing early lytic antigens (EAs), whereas IgA antibodies in serum samples from 15 healthy EBV-seropositive individuals reacted with 0.02%-2% of cells with EAs (P<.0001). IgA antibodies in primary infection were directed against the Bam Z Epstein-Barr replication activator (ZEBRA) (BZLF1) and diffuse EA (BMRF1) EAs. Thus, IgA antibodies to EBV EAs are produced during primary EBV infection and are likely to be stimulated as a result of lytic EBV replication in mucosal sites. Detection of IgA antibodies to EA may be developed into a diagnostic tool for primary EBV infection.


Asunto(s)
Anticuerpos Antivirales/sangre , Antígenos Virales/inmunología , Proteínas de Unión al ADN/inmunología , Infecciones por Virus de Epstein-Barr/inmunología , Herpesvirus Humano 4/inmunología , Inmunoglobulina A/sangre , Transactivadores/inmunología , Proteínas Virales/inmunología , Adolescente , Adulto , Biomarcadores , Línea Celular Tumoral , Niño , Preescolar , Citometría de Flujo , Humanos , Persona de Mediana Edad
19.
Adv Cancer Res ; 97: 81-109, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17419942

RESUMEN

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


Asunto(s)
Efecto Citopatogénico Viral/fisiología , Regulación Viral de la Expresión Génica/genética , Herpesvirus Humano 4/fisiología , Herpesvirus Humano 8/fisiología , Virus Oncogénicos/fisiología , Azacitidina/farmacología , Cicloheximida/farmacología , Efecto Citopatogénico Viral/efectos de los fármacos , Efecto Citopatogénico Viral/genética , Replicación del ADN , ADN Viral/biosíntesis , ADN Viral/genética , Infecciones por Virus de Epstein-Barr/virología , Regulación Viral de la Expresión Génica/efectos de los fármacos , Infecciones por Herpesviridae/virología , Herpesvirus Humano 4/efectos de los fármacos , Herpesvirus Humano 4/genética , Herpesvirus Humano 8/efectos de los fármacos , Herpesvirus Humano 8/genética , Humanos , Proteínas Inmediatas-Precoces/fisiología , Mutación , Fosforilación , Procesamiento Proteico-Postraduccional , Estructura Terciaria de Proteína , Transactivadores/química , Transactivadores/genética , Transactivadores/fisiología , Infecciones Tumorales por Virus/virología , Latencia del Virus/efectos de los fármacos , Latencia del Virus/genética
20.
J Biol Chem ; 281(6): 3085-95, 2006 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-16321978

RESUMEN

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


Asunto(s)
Proteínas de Unión al ADN/fisiología , Herpesvirus Humano 4/metabolismo , Transactivadores/fisiología , Proteínas Virales/fisiología , Sitios de Unión , Bioquímica/métodos , Línea Celular Tumoral , Movimiento Celular , Cisteína/química , Proteínas de Unión al ADN/química , Escherichia coli/metabolismo , Vectores Genéticos , Humanos , Inmunoprecipitación , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Sistema de Señalización de MAP Quinasas , Metionina/química , Mutagénesis , Péptidos/química , Fosforilación , Estructura Terciaria de Proteína , Serina/química , Treonina/química , Transactivadores/química , Transfección , Tripsina/química , Tirosina/química , Proteínas Virales/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA