Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 78
Filtrar
Mais filtros

Intervalo de ano de publicação
1.
Cell ; 186(26): 5675-5676, 2023 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-38134873

RESUMO

Autoimmune cross-reaction of specific antibodies to Epstein-Barr virus (EBV) has been associated with development of multiple sclerosis (MS). In this issue of Cell, Vietzen et al. identify additional immunological regulatory mechanisms that protect against autoimmunity in healthy people but are reduced in MS cases. The results confirm the link between EBV and MS.


Assuntos
Infecções por Vírus Epstein-Barr , Esclerose Múltipla , Humanos , Herpesvirus Humano 4 , Infecções por Vírus Epstein-Barr/complicações
2.
J Virol ; 96(12): e0039422, 2022 06 22.
Artigo em Inglês | MEDLINE | ID: mdl-35612313

RESUMO

The main target cells for Epstein-Barr virus (EBV) infection and persistence are B lymphocytes, although T and NK cells can also become infected. In this paper, we characterize the EBV present in 21 pediatric and adult patients who were treated in France for a range of diseases that involve infection of T or NK cells. Of these 21 cases, 5 pediatric patients (21%) and 11 adult patients (52%) were of Caucasian origin. In about 30% of the cases, some of the EBV genomes contain a large deletion. The deletions are different in every patient but tend to cluster near the BART region of the viral genome. Detailed investigation of a family in which several members have persistent T or NK cell infection by EBV indicates that the virus genome deletions arise or are selected independently in each individual patient. Genome sequence polymorphisms in the EBV in these T or NK cell diseases reflect the geographic origin of the patient and not a distinct type of EBV (the 21 cases studied included examples of both type 1 and type 2 EBV infection). Using virus produced from type 1 or type 2 EBV genomes cloned in bacterial artificial chromosome (BAC) vectors, we demonstrate infection of T cells in cord blood from healthy donors. Our results are consistent with transient infection of some T cells being part of normal asymptomatic infection by EBV in young children. IMPORTANCE EBV contributes to several types of human cancer. Some cancers and nonmalignant lymphoproliferative diseases involving T or NK cells contain EBV. These diseases are relatively frequent in Japan and China and have been shown sometimes to have deletions in the EBV genome in the disease cells. We identify further examples of deletions within the EBV genome associated with T or NK cell diseases, and we provide evidence that the virus genomes with these deletions are most likely selected in the individual cases, rather than being transmitted between people during infection. We demonstrate EBV infection of cord blood T cells by highly characterized, cloned EBV genomes and suggest that transient infection of T cells may be part of normal asymptomatic infection by EBV in young children.


Assuntos
Infecções por Vírus Epstein-Barr , Deleção de Genes , Genoma Viral , Herpesvirus Humano 4 , Transtornos Linfoproliferativos , Adulto , Infecções Assintomáticas , Criança , Herpesvirus Humano 4/genética , Humanos , Células Matadoras Naturais/virologia , Transtornos Linfoproliferativos/virologia , Linfócitos T/virologia
3.
PLoS Pathog ; 17(8): e1009834, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34352044

RESUMO

Viruses suppress immune recognition through diverse mechanisms. Epstein-Barr Virus (EBV) establishes latent infection in memory B-lymphocytes and B-cell malignancies where it impacts B-cell immune function. We show here that EBV primary infection of naïve B-cells results in a robust down-regulation of HLA genes. We found that the viral encoded transcriptional regulatory factor EBNA2 bound to multiple regulatory regions in the HLA locus. Conditional expression of EBNA2 correlated with the down regulation of HLA class II transcription. EBNA2 down-regulation of HLA transcription was found to be dependent on CIITA, the major transcriptional activator of HLA class II gene transcription. We identified a major EBNA2 binding site downstream of the CIITA gene and upstream of DEXI, a dexamethasone inducible gene that is oriented head-to-head with CIITA gene transcripts. CRISPR/Cas9 deletion of the EBNA2 site upstream of DEXI attenuated CIITA transcriptional repression. EBNA2 caused an increase in DEXI transcription and a graded change in histone modifications with activation mark H3K27ac near the DEXI locus, and a loss of activation marks at the CIITA locus. A prominent CTCF binding site between CIITA and DEXI enhancers was mutated and further diminished the effects of EBNA2 on CIITA. Analysis of HiC data indicate that DEXI and CIITA enhancers are situated in different chromosome topological associated domains (TADs). These findings suggest that EBNA2 down regulates HLA-II genes through the down regulation of CIITA, and that this down regulation is an indirect consequence of EBNA2 enhancer formation at a neighboring TAD. We propose that enhancer competition between these neighboring chromosome domains represents a novel mechanism for gene regulation demonstrated by EBNA2.


Assuntos
Linfócitos B/virologia , Proteínas de Ligação a DNA/genética , Elementos Facilitadores Genéticos , Infecções por Vírus Epstein-Barr/virologia , Antígenos Nucleares do Vírus Epstein-Barr/metabolismo , Genes MHC da Classe II/fisiologia , Proteínas de Membrana/genética , Proteínas Nucleares/genética , Transativadores/genética , Proteínas Virais/metabolismo , Linfócitos B/metabolismo , Proteínas de Ligação a DNA/metabolismo , Infecções por Vírus Epstein-Barr/genética , Infecções por Vírus Epstein-Barr/metabolismo , Antígenos Nucleares do Vírus Epstein-Barr/genética , Regulação da Expressão Gênica , Herpesvirus Humano 4/fisiologia , Humanos , Proteínas de Membrana/metabolismo , Proteínas Nucleares/metabolismo , Regiões Promotoras Genéticas , Transativadores/metabolismo , Proteínas Virais/genética
4.
PLoS Pathog ; 16(2): e1008365, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-32059024

RESUMO

Humans are infected with two distinct strains (Type 1 (T1) and Type 2 (T2)) of Epstein-Barr virus (EBV) that differ substantially in their EBNA2 and EBNA 3A/B/C latency genes and the ability to transform B cells in vitro. While most T1 EBV strains contain the "prototype" form of the BZLF1 immediate-early promoter ("Zp-P"), all T2 strains contain the "Zp-V3" variant, which contains an NFAT binding motif and is activated much more strongly by B-cell receptor signalling. Whether B cells infected with T2 EBV are more lytic than cells infected with T1 EBV is unknown. Here we show that B cells infected with T2 EBV strains (AG876 and BL5) have much more lytic protein expression compared to B cells infected with T1 EBV strains (M81, Akata, and Mutu) in both a cord blood-humanized (CBH) mouse model and EBV-transformed lymphoblastoid cell lines (LCLs). Although T2 LCLs grow more slowly than T1 LCLs, both EBV types induce B-cell lymphomas in CBH mice. T1 EBV strains (M81 and Akata) containing Zp-V3 are less lytic than T2 EBV strains, suggesting that Zp-V3 is not sufficient to confer a lytic phenotype. Instead, we find that T2 LCLs express much higher levels of activated NFATc1 and NFATc2, and that cyclosporine (an NFAT inhibitor) and knockdown of NFATc2 attenuate constitutive lytic infection in T2 LCLs. Both NFATc1 and NFATc2 induce lytic EBV gene expression when combined with activated CAMKIV (which is activated by calcium signaling and activates MEF2D) in Burkitt Akata cells. Together, these results suggest that B cells infected with T2 EBV are more lytic due to increased activity of the cellular NFATc1/c2 transcription factors in addition to the universal presence of the Zp-V3 form of BZLF1 promoter.


Assuntos
Linfócitos B/metabolismo , Fatores de Transcrição NFATC/genética , Animais , Linfócitos B/virologia , Linhagem Celular , Proteínas de Ligação a DNA/metabolismo , Infecções por Vírus Epstein-Barr/genética , Antígenos Nucleares do Vírus Epstein-Barr , Expressão Gênica/genética , Regulação Viral da Expressão Gênica/genética , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/metabolismo , Herpesvirus Humano 4/patogenicidade , Humanos , Camundongos , Regiões Promotoras Genéticas/genética , Transativadores/genética , Transativadores/metabolismo , Fatores de Transcrição/metabolismo , Proteínas Virais/metabolismo , Ativação Viral , Latência Viral
5.
Br J Haematol ; 195(2): 249-255, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34431085

RESUMO

Chronic active Epstein-Barr virus (CAEBV) disease is a rare condition characterised by persistent EBV infection in previously healthy individuals. Defective EBV genomes were found in East Asian patients with CAEBV. In the present study, we sequenced 14 blood EBV samples from three UK patients with CAEBV, comparing the results with saliva CAEBV samples and other conditions. We observed EBV deletions in blood, some of which may disrupt viral replication, but not saliva in CAEBV. Deletions were lost overtime after successful treatment. These findings are compatible with CAEBV being associated with the evolution and persistence of EBV+ haematological clones that are lost on successful treatment.


Assuntos
Infecções por Vírus Epstein-Barr/sangue , Herpesvirus Humano 4/genética , Saliva/metabolismo , Deleção de Sequência/genética , Adolescente , Biomarcadores/análise , Estudos de Casos e Controles , Criança , Pré-Escolar , Doença Crônica , Vírus Defeituosos/genética , Infecções por Vírus Epstein-Barr/tratamento farmacológico , Infecções por Vírus Epstein-Barr/epidemiologia , Ásia Oriental/epidemiologia , Feminino , Humanos , Fatores Imunológicos/uso terapêutico , Masculino , Transplante de Células-Tronco de Sangue Periférico/métodos , Polimorfismo de Nucleotídeo Único/genética , Rituximab/uso terapêutico , Resultado do Tratamento , Replicação Viral/genética
6.
PLoS Pathog ; 15(7): e1007458, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31283782

RESUMO

Natural variation separates Epstein-Barr virus (EBV) into type 1 and type 2 strains. Type 2 EBV is less transforming in vitro due to sequence differences in the EBV transcription factor EBNA2. This correlates with reduced activation of the EBV oncogene LMP1 and some cell genes. Transcriptional activation by type 1 EBNA2 can be suppressed through the binding of two PXLXP motifs in its transactivation domain (TAD) to the dimeric coiled-coil MYND domain (CC-MYND) of the BS69 repressor protein (ZMYND11). We identified a third conserved PXLXP motif in type 2 EBNA2. We found that type 2 EBNA2 peptides containing this motif bound BS69CC-MYND efficiently and that the type 2 EBNA2TAD bound an additional BS69CC-MYND molecule. Full-length type 2 EBNA2 also bound BS69 more efficiently in pull-down assays. Molecular weight analysis and low-resolution structures obtained using small-angle X-ray scattering showed that three BS69CC-MYND dimers bound two molecules of type 2 EBNA2TAD, in line with the dimeric state of full-length EBNA2 in vivo. Importantly, mutation of the third BS69 binding motif in type 2 EBNA2 improved B-cell growth maintenance and the transcriptional activation of the LMP1 and CXCR7 genes. Our data indicate that increased association with BS69 restricts the function of type 2 EBNA2 as a transcriptional activator and driver of B cell growth and may contribute to reduced B-cell transformation by type 2 EBV.


Assuntos
Proteínas de Transporte/metabolismo , Antígenos Nucleares do Vírus Epstein-Barr/genética , Antígenos Nucleares do Vírus Epstein-Barr/metabolismo , Herpesvirus Humano 4/genética , Proteínas Virais/genética , Proteínas Virais/metabolismo , Sequência de Aminoácidos , Substituição de Aminoácidos , Linfócitos B/metabolismo , Linfócitos B/virologia , Sítios de Ligação/genética , Proteínas de Transporte/química , Proteínas de Transporte/genética , Proteínas de Ciclo Celular , Linhagem Celular , Transformação Celular Viral/genética , Transformação Celular Viral/fisiologia , Proteínas Correpressoras , Proteínas de Ligação a DNA , Antígenos Nucleares do Vírus Epstein-Barr/química , Genes Virais , Herpesvirus Humano 4/classificação , Herpesvirus Humano 4/patogenicidade , Interações entre Hospedeiro e Microrganismos/genética , Interações entre Hospedeiro e Microrganismos/fisiologia , Humanos , Modelos Moleculares , Mutação , Domínios e Motivos de Interação entre Proteínas , Estrutura Quaternária de Proteína , Transativadores/química , Transativadores/genética , Transativadores/metabolismo , Proteínas Virais/química
7.
Int J Equity Health ; 20(1): 254, 2021 12 13.
Artigo em Inglês | MEDLINE | ID: mdl-34903229

RESUMO

BACKGROUND: Air quality is monitored at a local level in the UK as part of the Local Air Quality Management (LAQM) system. If air quality objectives within an area are not achieved an Air Quality Management Area (AQMA) is declared and action plan developed. The efficacy of this system in reducing air pollution has increasingly come into question, however very little is known about its impact on health or health inequalities. We therefore investigated the effect of declaring an AQMA on emergency hospitalisations for respiratory conditions in the North West Coast region of England, and examined whether the effect differed between more compared to less deprived neighbourhoods. METHODS: This longitudinal controlled ecological study analysed neighbourhoods located within or touching the boundaries of AQMAs declared in the North West Coast region between 2006 and 2016. Each of these intervention neighbourhoods were matched with five control neighbourhoods which had never been located within/touching an AQMA boundary. Difference-in-differences methods were used to compare the change in hospitalisation rates in the intervention neighbourhoods to the change in hospitalisation rates in the matched control neighbourhoods, before and after the declaration of an AQMA. RESULTS: In total, 108 intervention neighbourhoods and 540 control neighbourhoods were analysed over the period 2005-2017, giving a total sample size of 8424 neighbourhood-years. Emergency hospitalisations for respiratory conditions decreased in the intervention neighbourhoods by 158 per 100,000 per year [95% CI 90 to 227] after an AQMA was declared relative to the control neighbourhoods. There was a larger decrease in hospitalisation rates following the declaration of an AQMA in more compared to less income deprived neighbourhoods. CONCLUSIONS: Our results suggest the LAQM system has contributed to a reduction in emergency hospitalisations for respiratory conditions, and may represent an effective strategy to reduce inequalities in health. These findings highlight the importance of measuring the success of air quality policies not just in terms of air pollution but also in terms of population health.


Assuntos
Poluição do Ar , Poluição do Ar/análise , Inglaterra/epidemiologia , Hospitalização , Humanos , Políticas , Características de Residência
8.
Nucleic Acids Res ; 47(6): 2807-2821, 2019 04 08.
Artigo em Inglês | MEDLINE | ID: mdl-30649516

RESUMO

Epstein-Barr virus proteins EBNA3A, EBNA3B and EBNA3C control hundreds of host genes after infection. Changes in epigenetic marks around EBNA3-regulated genes suggest that they exert transcriptional control in collaboration with epigenetic factors. The roles of polycomb repressive complex (PRC)2 subunit SUZ12 and of PRC1 subunit BMI1 were assessed for their importance in EBNA3-mediated repression and activation. ChIP-seq experiments for SUZ12 and BMI1 were performed to determine their global localization on chromatin and analysis offered further insight into polycomb protein distribution in differentiated cells. Their localization was compared to that of each EBNA3 to resolve longstanding questions about the EBNA3-polycomb relationship. SUZ12 did not co-localize with any EBNA3, whereas EBNA3C co-localized significantly and co-immunoprecipitated with BMI1. In cells expressing a conditional EBNA3C, BMI1 was sequestered to EBNA3C-binding sites after EBNA3C activation. When SUZ12 or BMI1 was knocked down in the same cells, SUZ12 did not contribute to EBNA3C-mediated regulation. Surprisingly, after BMI1 knockdown, EBNA3C repressed equally efficiently but host gene activation by EBNA3C was impaired. This overturns previous assumptions about BMI1/PRC1 functions during EBNA3C-mediated regulation, for the first time identifies directly a host factor involved in EBNA3-mediated activation and provides a new insight into how PRC1 can be involved in gene activation.


Assuntos
Antígenos Nucleares do Vírus Epstein-Barr/fisiologia , Interações Hospedeiro-Patógeno/genética , Complexo Repressor Polycomb 1/fisiologia , Ativação Transcricional , Infecções por Vírus Epstein-Barr/genética , Infecções por Vírus Epstein-Barr/metabolismo , Antígenos Nucleares do Vírus Epstein-Barr/metabolismo , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Células HEK293 , Herpesvirus Humano 4/fisiologia , Humanos , Complexo Repressor Polycomb 1/metabolismo , Ligação Proteica
9.
PLoS Pathog ; 14(7): e1007179, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-30052684

RESUMO

Latent Epstein-Barr virus (EBV) infection contributes to both B-cell and epithelial-cell malignancies. However, whether lytic EBV infection also contributes to tumors is unclear, although the association between malaria infection and Burkitt lymphomas (BLs) may involve excessive lytic EBV replication. A particular variant of the viral promoter (Zp) that controls lytic EBV reactivation is over-represented, relative to its frequency in non-malignant tissue, in EBV-positive nasopharyngeal carcinomas and AIDS-related lymphomas. To date, no functional differences between the prototype Zp (Zp-P) and the cancer-associated variant (Zp-V3) have been identified. Here we show that a single nucleotide difference between the Zp-V3 and Zp-P promoters creates a binding site for the cellular transcription factor, NFATc1, in the Zp-V3 (but not Zp-P) variant, and greatly enhances Zp activity and lytic viral reactivation in response to NFATc1-inducing stimuli such as B-cell receptor activation and ionomycin. Furthermore, we demonstrate that restoring this NFATc1-motif to the Zp-P variant in the context of the intact EBV B95.8 strain genome greatly enhances lytic viral reactivation in response to the NFATc1-activating agent, ionomycin, and this effect is blocked by the NFAT inhibitory agent, cyclosporine, as well as NFATc1 siRNA. We also show that the Zp-V3 variant is over-represented in EBV-positive BLs and gastric cancers, and in EBV-transformed B-cell lines derived from EBV-infected breast milk of Kenyan mothers that had malaria during pregnancy. These results demonstrate that the Zp-V3 enhances EBV lytic reactivation to physiologically-relevant stimuli, and suggest that increased lytic infection may contribute to the increased prevalence of this variant in EBV-associated malignancies.


Assuntos
Infecções por Vírus Epstein-Barr/genética , Transativadores/genética , Ativação Viral/genética , Variação Genética/genética , Herpesvirus Humano 4/genética , Humanos , Regiões Promotoras Genéticas/genética
10.
J Virol ; 92(22)2018 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-30111570

RESUMO

One hundred thirty-eight new Epstein-Barr virus (EBV) genome sequences have been determined. One hundred twenty-five of these and 116 from previous reports were combined to produce a multiple-sequence alignment of 241 EBV genomes, which we have used to analyze variation within the viral genome. The type 1/type 2 classification of EBV remains the major form of variation and is defined mostly by EBNA2 and EBNA3, but the type 2 single-nucleotide polymorphisms (SNPs) at the EBNA3 locus extend into the adjacent gp350 and gp42 genes, whose products mediate infection of B cells by EBV. A small insertion within the BART microRNA region of the genome was present in 21 EBV strains. EBV from saliva of U.S. patients with chronic active EBV infection aligned with the wild-type EBV genome with no evidence of WZhet rearrangements. The V3 polymorphism in the Zp promoter for BZLF1 was found to be frequent in nasopharyngeal carcinoma cases from both Hong Kong and Indonesia. Codon usage was found to differ between latent and lytic cycle EBV genes, and the main forms of variation of the EBNA1 protein have been identified.IMPORTANCE Epstein-Barr virus causes most cases of infectious mononucleosis and posttransplant lymphoproliferative disease. It contributes to several types of cancer, including Hodgkin's lymphoma, Burkitt's lymphoma, diffuse large B cell lymphoma, nasopharyngeal carcinoma, and gastric carcinoma. EBV genome variation is important because some of the diseases associated with EBV have very different incidences in different populations and geographic regions, and differences in the EBV genome might contribute to these diseases. Some specific EBV genome alterations that appear to be significant in EBV-associated cancers are already known, and current efforts to make an EBV vaccine and antiviral drugs should also take account of sequence differences in the proteins used as targets.


Assuntos
Linfoma de Burkitt/genética , Genoma Viral/genética , Herpesvirus Humano 4/genética , Mononucleose Infecciosa/genética , Polimorfismo de Nucleotídeo Único/genética , Neoplasias Gástricas/genética , Sequência de Bases , Antígenos Nucleares do Vírus Epstein-Barr/genética , Humanos , Regiões Promotoras Genéticas/genética , Alinhamento de Sequência , Análise de Sequência de DNA , Transativadores/genética , Proteínas Virais/genética
11.
J Virol ; 91(15)2017 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-28490597

RESUMO

Molluscum contagiosum virus (MCV), the only known extant human-adapted poxvirus, causes a long-duration infection characterized by skin lesions that typically display an absence of inflammation despite containing high titers of live virus. Despite this curious presentation, MCV is very poorly characterized in terms of host-pathogen interactions. The absence of inflammation around MCV lesions suggests the presence of potent inhibitors of human antiviral immunity and inflammation. However, only a small number of MCV immunomodulatory genes have been characterized in detail. It is likely that many more remain to be discovered, given the density of such sequences in other poxvirus genomes. NF-κB activation occurs in response to both virus-induced pattern recognition receptor (PRR) signaling and cellular activation by virus-induced proinflammatory cytokines like tumor necrosis factor and interleukin-1. Activated NF-κB drives cytokine and interferon gene expression, leading to inflammation and virus clearance. We report that MC005, which has no orthologs in other poxvirus genomes, is a novel inhibitor of PRR- and cytokine-stimulated NF-κB activation. MC005 inhibited NF-κB proximal to the IκB kinase (IKK) complex, and unbiased affinity purification revealed that MC005 interacts with the IKK subunit NEMO (NF-κB essential modulator). MC005 binding to NEMO prevents the conformational priming of the IKK complex that occurs when NEMO binds to ubiquitin chains during pathway activation. These data reveal a novel mechanism of poxvirus inhibition of human innate immunity, validate current dynamic models of NEMO-dependent IKK complex activation, and further clarify how the human-adapted poxvirus MCV can so effectively evade antiviral immunity and suppress inflammation to persist in human skin lesions.IMPORTANCE Poxviruses adapt to specific hosts over time, evolving and tailoring elegantly precise inhibitors of the rate-limiting steps within the signaling pathways that control innate immunity and inflammation. These inhibitors reveal new features of the antiviral response, clarify existing models of signaling regulation while offering potent new tools for approaching therapeutic intervention in autoimmunity and inflammatory disease. Molluscum contagiosum virus (MCV) is the only known extant poxvirus specifically adapted to human infection and appears adept at evading normal human antiviral responses, yet it remains poorly characterized. We report the identification of MCV protein MC005 as an inhibitor of the pathways leading to the activation of NF-κB, an essential regulator of innate immunity. Further, identification of the mechanism of inhibition of NF-κB by MC005 confirms current models of the complex way in which NF-κB is regulated and greatly expands our understanding of how MCV so effectively evades human immunity.


Assuntos
Interações Hospedeiro-Patógeno , Quinase I-kappa B/antagonistas & inibidores , Evasão da Resposta Imune , Vírus do Molusco Contagioso/patogenicidade , NF-kappa B/antagonistas & inibidores , Proteínas Virais/metabolismo , Animais , Linhagem Celular , Humanos
12.
J Virol ; 91(15)2017 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-28515295

RESUMO

Viral gene sequences from an enlarged set of about 200 Epstein-Barr virus (EBV) strains, including many primary isolates, have been used to investigate variation in key viral genetic regions, particularly LMP1, Zp, gp350, EBNA1, and the BART microRNA (miRNA) cluster 2. Determination of type 1 and type 2 EBV in saliva samples from people from a wide range of geographic and ethnic backgrounds demonstrates a small percentage of healthy white Caucasian British people carrying predominantly type 2 EBV. Linkage of Zp and gp350 variants to type 2 EBV is likely to be due to their genes being adjacent to the EBNA3 locus, which is one of the major determinants of the type 1/type 2 distinction. A novel classification of EBNA1 DNA binding domains, named QCIGP, results from phylogeny analysis of their protein sequences but is not linked to the type 1/type 2 classification. The BART cluster 2 miRNA region is classified into three major variants through single-nucleotide polymorphisms (SNPs) in the primary miRNA outside the mature miRNA sequences. These SNPs can result in altered levels of expression of some miRNAs from the BART variant frequently present in Chinese and Indonesian nasopharyngeal carcinoma (NPC) samples. The EBV genetic variants identified here provide a basis for future, more directed analysis of association of specific EBV variations with EBV biology and EBV-associated diseases.IMPORTANCE Incidence of diseases associated with EBV varies greatly in different parts of the world. Thus, relationships between EBV genome sequence variation and health, disease, geography, and ethnicity of the host may be important for understanding the role of EBV in diseases and for development of an effective EBV vaccine. This paper provides the most comprehensive analysis so far of variation in specific EBV genes relevant to these diseases and proposed EBV vaccines. By focusing on variation in LMP1, Zp, gp350, EBNA1, and the BART miRNA cluster 2, new relationships with the known type 1/type 2 strains are demonstrated, and a novel classification of EBNA1 and the BART miRNAs is proposed.


Assuntos
Infecções por Vírus Epstein-Barr/virologia , Variação Genética , Genótipo , Herpesvirus Humano 4/classificação , Herpesvirus Humano 4/genética , MicroRNAs/genética , Proteínas Virais/genética , Infecções por Vírus Epstein-Barr/epidemiologia , Etnicidade , Geografia , Herpesvirus Humano 4/isolamento & purificação , Humanos , Londres , Epidemiologia Molecular , Saliva/virologia , Estudantes , Estados Unidos , Voluntários
13.
Blood ; 138(22): 2155-2156, 2021 12 02.
Artigo em Inglês | MEDLINE | ID: mdl-34854879

Assuntos
Hepcidinas , Ferro
14.
Nucleic Acids Res ; 44(10): 4636-50, 2016 06 02.
Artigo em Inglês | MEDLINE | ID: mdl-26883634

RESUMO

In B cells infected by the cancer-associated Epstein-Barr virus (EBV), RUNX3 and RUNX1 transcription is manipulated to control cell growth. The EBV-encoded EBNA2 transcription factor (TF) activates RUNX3 transcription leading to RUNX3-mediated repression of the RUNX1 promoter and the relief of RUNX1-directed growth repression. We show that EBNA2 activates RUNX3 through a specific element within a -97 kb super-enhancer in a manner dependent on the expression of the Notch DNA-binding partner RBP-J. We also reveal that the EBV TFs EBNA3B and EBNA3C contribute to RUNX3 activation in EBV-infected cells by targeting the same element. Uncovering a counter-regulatory feed-forward step, we demonstrate EBNA2 activation of a RUNX1 super-enhancer (-139 to -250 kb) that results in low-level RUNX1 expression in cells refractory to RUNX1-mediated growth inhibition. EBNA2 activation of the RUNX1 super-enhancer is also dependent on RBP-J. Consistent with the context-dependent roles of EBNA3B and EBNA3C as activators or repressors, we find that these proteins negatively regulate the RUNX1 super-enhancer, curbing EBNA2 activation. Taken together our results reveal cell-type-specific exploitation of RUNX gene super-enhancers by multiple EBV TFs via the Notch pathway to fine tune RUNX3 and RUNX1 expression and manipulate B-cell growth.


Assuntos
Linfócitos B/virologia , Subunidades alfa de Fatores de Ligação ao Core/genética , Elementos Facilitadores Genéticos , Antígenos Nucleares do Vírus Epstein-Barr/metabolismo , Fatores de Transcrição/metabolismo , Ativação Transcricional , Linfócitos B/metabolismo , Linhagem Celular , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Subunidade alfa 3 de Fator de Ligação ao Core/genética , Humanos , Proteína de Ligação a Sequências Sinal de Recombinação J de Imunoglobina/metabolismo , Receptores Notch/metabolismo
15.
Adv Exp Med Biol ; 962: 283-298, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28299664

RESUMO

RUNX1 and RUNX3 are the main RUNX genes expressed in B lymphocytes. Both are expressed throughout B-cell development and play key roles at certain key developmental transitions. The tumour-associated Epstein-Barr virus (EBV) has potent B-cell transforming ability and manipulates RUNX3 and RUNX1 transcription through novel mechanisms to control B cell growth. In contrast to resting mature B cells where RUNX1 expression is high, in EBV-infected cells RUNX1 levels are low and RUNX3 levels are high. Downregulation of RUNX1 in these cells results from cross-regulation by RUNX3 and serves to relieve RUNX1-mediated growth repression. RUNX3 is upregulated by the EBV transcription factor (TF) EBNA2 and represses RUNX1 transcription through RUNX sites in the RUNX1 P1 promoter. Recent analysis revealed that EBNA2 activates RUNX3 transcription through an 18 kb upstream super-enhancer in a manner dependent on the EBNA2 and Notch DNA-binding partner RBP-J. This super-enhancer also directs RUNX3 activation by two further RBP-J-associated EBV TFs, EBNA3B and 3C. Counter-intuitively, EBNA2 also hijacks RBP-J to target a super-enhancer region upstream of RUNX1 to maintain some RUNX1 expression in certain cell backgrounds, although the dual functioning EBNA3B and 3C proteins limit this activation. Interestingly, the B-cell genome binding sites of EBV TFs overlap extensively with RUNX3 binding sites and show enrichment for RUNX motifs. Therefore in addition to B-cell growth manipulation through the long-range control of RUNX transcription, EBV may also use RUNX proteins as co-factors to deregulate the transcription of many B cell genes during immortalisation.


Assuntos
Linfócitos B/metabolismo , Subunidades alfa de Fatores de Ligação ao Core/metabolismo , Animais , Linfócitos B/virologia , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/metabolismo , Humanos , Ativação Linfocitária/genética , Transcrição Gênica/genética , Ativação Transcricional/genética , Proteínas Virais/metabolismo
16.
J Virol ; 89(8): 4624-35, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25673702

RESUMO

UNLABELLED: Respiratory syncytial virus (RSV) infects epithelial cells of the respiratory tract and is a major cause of bronchiolitis and pneumonia in children and the elderly. The virus assembles and buds through the plasma membrane, forming elongated membrane filaments, but details of how this happens remain obscure. Oligomerization of the matrix protein (M) is a key step in the process of assembly and infectious virus production. In addition, it was suggested to affect the conformation of the fusion protein, the major current target for RSV antivirals, in the mature virus. The structure and assembly of M are thus key parameters in the RSV antiviral development strategy. The structure of RSV M was previously published as a monomer. Other paramyxovirus M proteins have been shown to dimerize, and biochemical data suggest that RSV M also dimerizes. Here, using size exclusion chromatography-multiangle laser light scattering, we show that the protein is dimeric in solution. We also crystallized M in two crystal forms and show that it assembles into equivalent dimers in both lattices. Dimerization interface mutations destabilize the M dimer in vitro. To assess the biological relevance of dimerization, we used confocal imaging to show that dimerization interface mutants of M fail to assemble into viral filaments on the plasma membrane. Additionally, budding and release of virus-like particles are prevented in M mutants that fail to form filaments. Importantly, we show that M is biologically active as a dimer and that the switch from M dimers to higher-order oligomers triggers viral filament assembly and virus production. IMPORTANCE: Human respiratory syncytial virus (RSV) is the most frequent cause of infantile bronchiolitis and pneumonia. The enormous burden of RSV makes it a major unmet target for a vaccine and antiviral drug therapy. Oligomerization of the matrix protein is a key step in the process of assembly and production of infectious virus, but the molecular mechanism of RSV assembly is still poorly understood. Here we show that the RSV matrix protein forms dimers in solution and in crystals; the dimer is essential for formation of higher-order oligomers. Destabilizing the dimer interface resulted in the loss of RSV filament formation and a lack of budding of virus-like particles. Importantly, our findings can potentially lead to new structure-based RSV inhibitors targeting the assembly process.


Assuntos
Modelos Moleculares , Vírus Sinciciais Respiratórios/genética , Vírus Sinciciais Respiratórios/fisiologia , Proteínas da Matriz Viral/metabolismo , Montagem de Vírus/fisiologia , Replicação Viral/fisiologia , Western Blotting , Cromatografia em Gel , Cristalização , Dimerização , Eletroforese em Gel de Poliacrilamida , Humanos , Microscopia Confocal , Conformação Proteica , Vírus Sinciciais Respiratórios/metabolismo , Proteínas da Matriz Viral/genética
17.
J Virol ; 89(16): 8406-15, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26041281

RESUMO

Molluscum contagiosum virus (MCV) is unique in being the only known extant, human-adapted poxvirus, yet to date, it is very poorly characterized in terms of host-pathogen interactions. MCV causes persistent skin lesions filled with live virus, but these are generally immunologically silent, suggesting the presence of potent inhibitors of human antiviral immunity and inflammation. Fewer than five MCV immunomodulatory genes have been characterized in detail, but it is likely that many more remain to be discovered given the density of such sequences in all well-characterized poxviruses. Following virus infection, NF-B activation occurs in response to both pattern recognition receptor (PRR) signaling and cellular activation by virus-elicited proinflammatory cytokines, such as tumor necrosis factor (TNF). As such, NF-B activation is required for virus detection, antiviral signaling, inflammation, and clearance of viral infection. Hence, we screened a library of MCV genes for effects on TNF-stimulated NF-B activation. This revealed MC132, a unique protein with no orthologs in other poxviral genomes, as a novel inhibitor of NF-B. Interestingly, MC132 also inhibited PRR- and virus-activated NF-B, since MC132 interacted with the NF-B subunit p65 and caused p65 degradation. Unbiased affinity purification to identify host targets of MC132 revealed that MC132 acted by targeting NF-B p65 for ubiquitin-dependent proteasomal degradation by recruiting p65 to a host Cullin-5/Elongin B/Elongin C complex. These data reveal a novel mechanism for poxviral inhibition of human innate immunity and further clarify how the human-adapted poxvirus MCV can so effectively evade antiviral immunity to persist in skin lesions.


Assuntos
Tolerância Imunológica/imunologia , Imunidade Inata/imunologia , Vírus do Molusco Contagioso/imunologia , NF-kappa B/metabolismo , Proteínas Virais/imunologia , eIF-2 Quinase/metabolismo , Cromatografia Líquida , Ensaio de Imunoadsorção Enzimática , Humanos , Immunoblotting , Imunoprecipitação , Microscopia Confocal , Vírus do Molusco Contagioso/metabolismo , Oligonucleotídeos/genética , Proteólise , Interferência de RNA , Reação em Cadeia da Polimerase em Tempo Real , Receptores de Reconhecimento de Padrão/metabolismo , Espectrometria de Massas em Tandem , Proteínas Virais/metabolismo
18.
J Virol ; 89(22): 11711-4, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26339045

RESUMO

Functions of Epstein-Barr virus (EBV)-encoded RNAs (EBERs) were tested in lymphoblastoid cell lines containing EBER mutants of EBV. Binding of EBER1 to ribosomal protein L22 (RPL22) was confirmed. Deletion of EBER1 or EBER2 correlated with increased levels of cytoplasmic EBV LMP2 RNA and with small effects on specific cellular microRNA (miRNA) levels, but protein levels of LMP1 and LMP2A were not affected. Wild-type EBV and EBER deletion EBV had approximately equal abilities to infect immunodeficient mice reconstituted with a human hematopoietic system.


Assuntos
Herpesvirus Humano 4/genética , RNA Viral/genética , Proteínas da Matriz Viral/metabolismo , Animais , Linhagem Celular , Cisplatino/farmacologia , Humanos , Camundongos , Camundongos Knockout , Camundongos SCID , MicroRNAs/genética , RNA Viral/biossíntese , Proteínas de Ligação a RNA/metabolismo , Proteínas Ribossômicas/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Proteínas da Matriz Viral/genética
19.
J Virol ; 89(10): 5222-37, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25787276

RESUMO

UNLABELLED: Epstein-Barr virus (EBV) infects most of the world's population and is causally associated with several human cancers, but little is known about how EBV genetic variation might influence infection or EBV-associated disease. There are currently no published wild-type EBV genome sequences from a healthy individual and very few genomes from EBV-associated diseases. We have sequenced 71 geographically distinct EBV strains from cell lines, multiple types of primary tumor, and blood samples and the first EBV genome from the saliva of a healthy carrier. We show that the established genome map of EBV accurately represents all strains sequenced, but novel deletions are present in a few isolates. We have increased the number of type 2 EBV genomes sequenced from one to 12 and establish that the type 1/type 2 classification is a major feature of EBV genome variation, defined almost exclusively by variation of EBNA2 and EBNA3 genes, but geographic variation is also present. Single nucleotide polymorphism (SNP) density varies substantially across all known open reading frames and is highest in latency-associated genes. Some T-cell epitope sequences in EBNA3 genes show extensive variation across strains, and we identify codons under positive selection, both important considerations for the development of vaccines and T-cell therapy. We also provide new evidence for recombination between strains, which provides a further mechanism for the generation of diversity. Our results provide the first global view of EBV sequence variation and demonstrate an effective method for sequencing large numbers of genomes to further understand the genetics of EBV infection. IMPORTANCE: Most people in the world are infected by Epstein-Barr virus (EBV), and it causes several human diseases, which occur at very different rates in different parts of the world and are linked to host immune system variation. Natural variation in EBV DNA sequence may be important for normal infection and for causing disease. Here we used rapid, cost-effective sequencing to determine 71 new EBV sequences from different sample types and locations worldwide. We showed geographic variation in EBV genomes and identified the most variable parts of the genome. We identified protein sequences that seem to have been selected by the host immune system and detected variability in known immune epitopes. This gives the first overview of EBV genome variation, important for designing vaccines and immune therapy for EBV, and provides techniques to investigate relationships between viral sequence variation and EBV-associated diseases.


Assuntos
Infecções por Vírus Epstein-Barr/virologia , Variação Genética , Genoma Viral , Herpesvirus Humano 4/genética , Sequência de Aminoácidos , Antígenos Virais/genética , Portador Sadio/virologia , Linhagem Celular Tumoral , DNA Viral/genética , Epitopos de Linfócito T/genética , Antígenos Nucleares do Vírus Epstein-Barr/genética , Herpesvirus Humano 4/classificação , Herpesvirus Humano 4/isolamento & purificação , Humanos , Filogenia , Polimorfismo de Nucleotídeo Único , Recombinação Genética , Proteínas da Matriz Viral/genética
20.
Curr Top Microbiol Immunol ; 390(Pt 1): 45-69, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26424643

RESUMO

What is wild-type Epstein-Barr virus and are there genetic differences in EBV strains that contribute to some of the EBV-associated diseases? Recent progress in DNA sequencing has resulted in many new Epstein-Barr virus (EBV) genome sequences becoming available. EBV isolates worldwide can be grouped into type 1 and type 2, a classification based on the EBNA2 gene sequence. Type 1 transforms human B cells into lymphoblastoid cell lines much more efficiently than type 2 EBV and molecular mechanisms that may account for this difference in cell transformation are now becoming understood. Study of geographic variation of EBV strains independent of the type 1/type 2 classification and systematic investigation of the relationship between viral strains, infection and disease are now becoming possible. So we should consider more directly whether viral sequence variation might play a role in the incidence of some EBV-associated diseases.


Assuntos
Infecções por Vírus Epstein-Barr/virologia , Variação Genética , Herpesvirus Humano 4/genética , Animais , Genoma Viral , Herpesvirus Humano 4/isolamento & purificação , Herpesvirus Humano 4/metabolismo , Humanos , Proteínas Virais/genética , Proteínas Virais/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA