Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 182
Filtrar
1.
J Virol ; 98(2): e0189923, 2024 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-38294245

RESUMO

After Epstein-Barr virus (EBV) genome replication and encapsidation in the nucleus, nucleocapsids are translocated into the cytoplasm for subsequent tegumentation and maturation. The EBV BGLF4 kinase, which induces partial disassembly of the nuclear lamina, and the nuclear egress complex BFRF1/BFLF2 coordinately facilitate the nuclear egress of nucleocapsids. Here, we demonstrate that within EBV reactivated epithelial cells, viral capsids, tegument proteins, and glycoproteins are clustered in the juxtanuclear concave region, accompanied by redistributed cytoplasmic organelles and the cytoskeleton regulator IQ-domain GTPase-activation protein 1 (IQGAP1), close to the microtubule-organizing center (MTOC). The assembly compartment (AC) structure was diminished in BGLF4-knockdown TW01-EBV cells and BGLF4-knockout bacmid-carrying TW01 cells, suggesting that the formation of AC structure is BGLF4-dependent. Notably, glycoprotein gp350/220 was observed by confocal imaging to be distributed in the perinuclear concave region and surrounded by the endoplasmic reticulum (ER) membrane marker calnexin, indicating that the AC may be located within a globular structure derived from ER membranes, adjacent to the outer nuclear membrane. Moreover, the viral capsid protein BcLF1 and tegument protein BBLF1 were co-localized with IQGAP1 near the cytoplasmic membrane in the late stage of replication. Knockdown of IQGAP1 did not affect the AC formation but decreased virion release from both TW01-EBV and Akata+ cells, suggesting IQGAP1-mediated trafficking regulates EBV virion release. The data presented here show that BGLF4 is required for cytoskeletal rearrangement, coordination with the redistribution of cytoplasmic organelles and IQGAP1 for virus maturation, and subsequent IQGAP1-dependent virion release.IMPORTANCEEBV genome is replicated and encapsidated in the nucleus, and the resultant nucleocapsids are translocated to the cytoplasm for subsequent virion maturation. We show that a cytoplasmic AC, containing viral proteins, markers of the endoplasmic reticulum, Golgi, and endosomes, is formed in the juxtanuclear region of epithelial and B cells during EBV reactivation. The viral BGLF4 kinase contributes to the formation of the AC. The cellular protein IQGAP1 is also recruited to the AC and partially co-localizes with the virus capsid protein BcLF1 and tegument protein BBLF1 in EBV-reactivated cells, dependent on the BGLF4-induced cytoskeletal rearrangement. In addition, virion release was attenuated in IQGAP1-knockdown epithelial and B cells after reactivation, suggesting that IQGAP1-mediated trafficking may regulate the efficiency of virus maturation and release.


Assuntos
Citoplasma , Herpesvirus Humano 4 , Proteínas Serina-Treonina Quinases , Proteínas Virais , Vírion , Montagem de Vírus , Liberação de Vírus , Proteínas Ativadoras de ras GTPase , Humanos , Proteínas do Capsídeo/metabolismo , Citoplasma/metabolismo , Citoplasma/virologia , Infecções por Vírus Epstein-Barr/metabolismo , Infecções por Vírus Epstein-Barr/virologia , Herpesvirus Humano 4/química , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/crescimento & desenvolvimento , Herpesvirus Humano 4/metabolismo , Proteínas de Membrana/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Ativadoras de ras GTPase/metabolismo , Proteínas Virais/metabolismo , Vírion/química , Vírion/crescimento & desenvolvimento , Vírion/metabolismo , Montagem de Vírus/fisiologia , Retículo Endoplasmático/metabolismo , Endossomos/metabolismo , Complexo de Golgi/metabolismo
2.
J Virol ; 96(17): e0094922, 2022 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-36037477

RESUMO

Epstein-Barr nuclear antigen 1 (EBNA1) is a multifunctional viral-encoded DNA-binding protein essential for Epstein-Barr virus (EBV) DNA replication and episome maintenance. EBNA1 binds to two functionally distinct elements at the viral origin of plasmid replication (oriP), termed the dyad symmetry (DS) element, required for replication initiation and the family of repeats (FR) required for episome maintenance. Here, we determined the cryo-electron microscopy (cryo-EM) structure of the EBNA1 DNA binding domain (DBD) from amino acids (aa) 459 to 614 and its interaction with two tandem sites at the DS and FR. We found that EBNA1 induces a strong DNA bending angle in the DS, while the FR is more linear. The N-terminal arm of the DBD (aa 444 to 468) makes extensive contact with DNA as it wraps around the minor groove, with some conformational variation among EBNA1 monomers. Mutation of variable-contact residues K460 and K461 had only minor effects on DNA binding but had abrogated oriP-dependent DNA replication. We also observed that the AT-rich intervening DNA between EBNA1 binding sites in the FR can be occupied by the EBNA1 AT hook, N-terminal domain (NTD) aa 1 to 90 to form a Zn-dependent stable complex with EBNA1 DBD on a 2×FR DNA template. We propose a model showing EBNA1 DBD and NTD cobinding at the FR and suggest that this may contribute to the oligomerization of viral episomes important for maintenance during latent infection. IMPORTANCE EBV latent infection is causally linked to diverse cancers and autoimmune disorders. EBNA1 is the viral-encoded DNA binding protein required for episomal maintenance during latent infection and is consistently expressed in all EBV tumors. The interaction of EBNA1 with different genetic elements confers different viral functions, such as replication initiation at DS and chromosome tethering at FR. Here, we used cryo-EM to determine the structure of the EBNA1 DNA-binding domain (DBD) bound to two tandem sites at the DS and at the FR. We also show that the NTD of EBNA1 can interact with the AT-rich DNA sequence between tandem EBNA1 DBD binding sites in the FR. These results provide new information on the mechanism of EBNA1 DNA binding at DS and FR and suggest a higher-order oligomeric structure of EBNA1 bound to FR. Our findings have implications for targeting EBNA1 in EBV-associated disease.


Assuntos
Antígenos Nucleares do Vírus Epstein-Barr/química , Herpesvirus Humano 4/química , Origem de Replicação , Sítios de Ligação , Microscopia Crioeletrônica , Replicação do DNA , Proteínas de Ligação a DNA/metabolismo , Infecções por Vírus Epstein-Barr , Antígenos Nucleares do Vírus Epstein-Barr/metabolismo , Antígenos Nucleares do Vírus Epstein-Barr/ultraestrutura , Herpesvirus Humano 4/metabolismo , Humanos , Infecção Latente , Plasmídeos , Replicação Viral
3.
Viruses ; 13(12)2021 11 28.
Artigo em Inglês | MEDLINE | ID: mdl-34960650

RESUMO

Epstein-Barr virus (EBV) is the first reported oncogenic virus and infects more than 90% of adults worldwide. EBV can establish a latent infection in B lymphocytes which is essential for persistence and transmission. Glycoprotein gp42 is an indispensable member of the triggering complex for EBV entry into a B cell. The N-terminal region of gp42 plays a key role in binding to gH/gL and triggering subsequent membrane fusion. However, no antibody has been reported to recognize this region and the immunogenicity of gp42 N-domain remains unknown. In the present study, we have generated a panel of nine mAbs against the gp42 N-terminal region (six mAbs to gp42-44-61aa and three mAbs to gp42-67-81aa). These mAbs show excellent binding activity and recognize different key residues locating on the gp42 N-domain. Among the nine mAbs, 4H7, 4H8 and 11G10 cross-react with rhLCV-gp42 while other mAbs specifically recognize EBV-gp42. Our newly obtained mAbs provide a useful tool for investigating the gp42 function and viral infection mechanism of γ-Herpesvirus. Furthermore, we assess the immunogenicity of the gp42 N-terminal region using the HBc149 particle as a carrier protein. The chimeric VLPs can induce high antibody titers and elicit neutralizing humoral responses to block EBV infection. More rational and effective designs are required to promote the gp42-N terminal region to become an epitope-based vaccine.


Assuntos
Anticorpos Antivirais/imunologia , Infecções por Vírus Epstein-Barr/imunologia , Glicoproteínas/química , Glicoproteínas/imunologia , Herpesvirus Humano 4/imunologia , Proteínas Virais/química , Proteínas Virais/imunologia , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Linfócitos B/imunologia , Linfócitos B/virologia , Mapeamento de Epitopos , Infecções por Vírus Epstein-Barr/virologia , Glicoproteínas/genética , Herpesvirus Humano 4/química , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/fisiologia , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Proteínas Virais/genética , Internalização do Vírus
4.
Commun Biol ; 4(1): 967, 2021 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-34400762

RESUMO

Epstein-Barr virus nuclear antigen 2 (EBNA2) is a transactivator of viral and cellular gene expression, which plays a critical role in the Epstein-Barr virus-associated diseases. It was reported that EBNA2 regulates gene expression by reorganizing chromatin and manipulating epigenetics. Recent studies showed that liquid-liquid phase separation plays an essential role in epigenetic and transcriptional regulation. Here we show that EBNA2 reorganized chromatin topology to form accessible chromatin domains (ACDs) of the host genome by phase separation. The N-terminal region of EBNA2, which is necessary for phase separation, is sufficient to induce ACDs. The C-terminal domain of EBNA2 promotes the acetylation of accessible chromatin regions by recruiting histone acetylase p300 to ACDs. According to these observations, we proposed a model of EBNA2 reorganizing chromatin topology for its acetylation through phase separation to explain the mechanism of EBNA2 hijacking the host genome by controlling its epigenetics.


Assuntos
Cromatina/química , Epigênese Genética , Herpesvirus Humano 4/genética , Proteínas Virais/química , Células HEK293 , Herpesvirus Humano 4/química , Humanos , Proteínas Virais/genética
5.
Elife ; 102021 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-33554858

RESUMO

Genome packaging in large double-stranded DNA viruses requires a powerful molecular motor to force the viral genome into nascent capsids, which involves essential accessory factors that are poorly understood. Here, we present structures of two such accessory factors from the oncogenic herpesviruses Kaposi's sarcoma-associated herpesvirus (KSHV; ORF68) and Epstein-Barr virus (EBV; BFLF1). These homologous proteins form highly similar homopentameric rings with a positively charged central channel that binds double-stranded DNA. Mutation of individual positively charged residues within but not outside the channel ablates DNA binding, and in the context of KSHV infection, these mutants fail to package the viral genome or produce progeny virions. Thus, we propose a model in which ORF68 facilitates the transfer of newly replicated viral genomes to the packaging motor.


Assuntos
Herpesvirus Humano 4/fisiologia , Herpesvirus Humano 8/fisiologia , Proteínas Virais/química , Proteínas Virais/metabolismo , Infecções por Vírus Epstein-Barr/virologia , Genoma Viral , Células HEK293 , Infecções por Herpesviridae/virologia , Herpesvirus Humano 4/química , Herpesvirus Humano 4/genética , Herpesvirus Humano 8/química , Herpesvirus Humano 8/genética , Humanos , Empacotamento do Genoma Viral , Proteínas Virais/genética , Replicação Viral
6.
J Immunol ; 205(12): 3291-3299, 2020 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-33188075

RESUMO

Immunosuppressants are associated with serious and often life-threatening adverse effects. To optimize immunotherapy, a tool that measures the immune reserve is necessary. We validated that a cell-based assay that measures TNF-α production by CD14+16+ intermediate monocytes following stimulation with EBV peptides has high sensitivity for the detection of over-immunosuppression (OIS) events. To develop a sequential, two-step assay with high specificity, we used PBMCs from kidney recipients (n = 87). Patients were classified as cases or controls, according to the occurrence of opportunistic infection, recurring bacterial infections, or de novo neoplasia. Patients who tested positive in the first step were randomly allocated to a training or a testing set for the development of the second step. In the discovery phase, an assay based on the examination of early mature B (eBm5) cells was able to discriminate OIS patients from controls with a specificity of 88%. The testing set also revealed a specificity of 88%. The interassay coefficient of variability between the experiments was 6.1%. Stratified analyses showed good diagnostic accuracy across tertiles of age and time posttransplant. In the adjusted model, the risk of OIS was more than 12 times higher in patients classified as positive than in those who tested negative (adjusted hazard ratio, 12.2; 95% confidence interval: 4.3-34.6). This sequential cell-based assay, which examines the monocyte and eBm5 cell response to EBV peptides, may be useful for identifying OIS in immunosuppressed patients.


Assuntos
Bioensaio , Herpesvirus Humano 4/química , Hospedeiro Imunocomprometido , Terapia de Imunossupressão/efeitos adversos , Transplante de Rim , Monócitos/imunologia , Peptídeos/química , Proteínas Virais/química , Adulto , Idoso , Feminino , Herpesvirus Humano 4/imunologia , Humanos , Masculino , Pessoa de Meia-Idade , Peptídeos/imunologia , Valor Preditivo dos Testes , Estudos Prospectivos , Proteínas Virais/imunologia
7.
Nat Commun ; 11(1): 5964, 2020 11 24.
Artigo em Inglês | MEDLINE | ID: mdl-33235207

RESUMO

The human γ-herpesviruses Kaposi sarcoma associated herpesvirus (KSHV) and Epstein-Barr virus (EBV) are associated with many human malignancies. Viral glycoprotein H (gH) and glycoprotein L (gL) are crucial for the cell tropism by binding to specific receptors. Recently, EphA2 was identified as the specific entry receptor for both KSHV and EBV. Here, we characterized the crystal structures of KSHV gHgL or EBV gHgL in complex with the ligand binding domain (LBD) of EphA2. Both KSHV and EBV gHgL bind to the channel and peripheral regions of LBD primarily using gL. Extensive interactions with more contacts contribute to the higher affinity of KSHV gHgL to LBD than that of EBV gHgL. These binding characteristics were verified using cell-based fusion assays with mutations in key EphA2 residues. Our experiments suggest that multiple animal γ-herpesviruses could use EphA2 as an entry receptor, implying a potential threat to human health.


Assuntos
Gammaherpesvirinae/química , Receptor EphA2/química , Proteínas do Envelope Viral/química , Cristalografia por Raios X/métodos , Herpesvirus Humano 4/química , Herpesvirus Humano 8/química , Interações entre Hospedeiro e Microrganismos , Glicoproteínas de Membrana/química , Chaperonas Moleculares/química , Ligação Proteica , Proteínas Virais/química , Proteínas Virais/metabolismo , Internalização do Vírus
8.
Nat Commun ; 11(1): 5405, 2020 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-33106493

RESUMO

Epstein-Barr virus (EBV) is a γ-herpesvirus associated with the occurrence of several human malignancies. BBRF2 and BSRF1 are two EBV tegument proteins that have been suggested to form a hetero-complex and mediate viral envelopment, but the molecular basis of their interaction and the functional mechanism of this complex remains unknown. Here, we present crystal structures of BBRF2 alone and in complex with BSRF1. BBRF2 has a compact globular architecture featuring a central ß-sheet that is surrounded by 10 helices, it represents a novel fold distinct from other known protein structures. The central portion of BSRF1 folds into two tightly associated antiparallel α-helices, forming a composite four-helix bundle with two α-helices from BBRF2 via a massive hydrophobic network. In vitro, a BSRF1-derived peptide binds to BBRF2 and reduces the number of viral genome copies in EBV-positive cells. Exogenous BBRF2 and BSRF1 co-localize at the Golgi apparatus. Furthermore, BBRF2 binds capsid and capsid-associated proteins, whereas BSRF1 associates with glycoproteins. These findings indicate that the BBRF2-BSRF1 complex tethers EBV nucleocapsids to the glycoprotein-enriched Golgi membrane, facilitating secondary envelopment.


Assuntos
Herpesvirus Humano 4/metabolismo , Capsídeo/química , Capsídeo/metabolismo , Infecções por Vírus Epstein-Barr/virologia , Genoma Viral , Herpesvirus Humano 4/química , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/crescimento & desenvolvimento , Humanos , Ligação Proteica , Conformação Proteica em alfa-Hélice
9.
Cell Res ; 30(10): 873-884, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32620850

RESUMO

Epstein-Barr virus (EBV) is the primary cause of infectious mononucleosis and has been shown to be closely associated with various malignancies. Here, we present a complete atomic model of EBV, including the icosahedral capsid, the dodecameric portal and the capsid-associated tegument complex (CATC). Our in situ portal from the tegumented capsid adopts a closed conformation with its channel valve holding the terminal viral DNA and with its crown region firmly engaged by three layers of ring-like dsDNA, which, together with the penton flexibility, effectively alleviates the capsid inner pressure placed on the portal cap. In contrast, the CATCs, through binding to the flexible penton vertices in a stoichiometric manner, accurately increase the inner capsid pressure to facilitate the pressure-driven genome delivery. Together, our results provide important insights into the mechanism by which the EBV capsid, portal, packaged genome and the CATCs coordinately achieve a pressure balance to simultaneously benefit both viral genome retention and ejection.


Assuntos
Capsídeo/ultraestrutura , Herpesvirus Humano 4/ultraestrutura , Substâncias Macromoleculares/química , Modelos Moleculares , Capsídeo/química , Proteínas do Capsídeo/química , Microscopia Crioeletrônica , DNA Viral/química , Herpesvirus Humano 4/química
10.
BioDrugs ; 34(4): 435-462, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32301049

RESUMO

To interfere with cell function, many scientists rely on methods that target DNA or RNA due to the ease with which they can be applied. Proteins are usually the final executors of function but are targeted only indirectly by these methods. Recent advances in targeted degradation of proteins based on proteolysis-targeting chimaeras (PROTACs), ubiquibodies, deGradFP (degrade Green Fluorescent Protein) and other approaches have demonstrated the potential of interfering directly at the protein level for research and therapy. Proteins can be targeted directly and very specifically by antibodies, but using antibodies inside cells has so far been considered to be challenging. However, it is possible to deliver antibodies or other proteins into the cytosol using standard laboratory equipment. Physical methods such as electroporation have been demonstrated to be efficient and validated thoroughly over time. The expression of intracellular antibodies (intrabodies) inside cells is another way to interfere with intracellular targets at the protein level. Methodological strategies to target the inside of cells with antibodies, including delivered antibodies and expressed antibodies, as well as applications in the research areas of neurobiology, viral infections and oncology, are reviewed here. Antibodies have already been used to interfere with a wide range of intracellular targets. Disease-related targets included proteins associated with neurodegenerative diseases such as Parkinson's disease (α-synuclein), Alzheimer's disease (amyloid-ß) or Huntington's disease (mutant huntingtin [mHtt]). The applications of intrabodies in the context of viral infections include targeting proteins associated with HIV (e.g. HIV1-TAT, Rev, Vif, gp41, gp120, gp160) and different oncoviruses such as human papillomavirus (HPV), hepatitis B virus (HBV), hepatitis C virus (HCV) and Epstein-Barr virus, and they have been used to interfere with various targets related to different processes in cancer, including oncogenic pathways, proliferation, cell cycle, apoptosis, metastasis, angiogenesis or neo-antigens (e.g. p53, human epidermal growth factor receptor-2 [HER2], signal transducer and activator of transcription 3 [STAT3], RAS-related RHO-GTPase B (RHOB), cortactin, vascular endothelial growth factor receptor 2 [VEGFR2], Ras, Bcr-Abl). Interfering at the protein level allows questions to be addressed that may remain unanswered using alternative methods. This review addresses why direct targeting of proteins allows unique insights, what is currently feasible in vitro, and how this relates to potential therapeutic applications.


Assuntos
Anticorpos/química , Infecções por Vírus Epstein-Barr , Herpesvirus Humano 4/química , Neurobiologia , Anticorpos/administração & dosagem , Humanos , Fator A de Crescimento do Endotélio Vascular/química , Fator A de Crescimento do Endotélio Vascular/metabolismo
11.
Immunol Lett ; 217: 15-24, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31689443

RESUMO

The aetiology of multiple sclerosis (MS) is as yet poorly understood. Multiple mechanisms in different disease stages are responsible for immunopathology in MS. HLA Class II DR2b (DRB1*1501 ß, DRA1*0101 α) is the strongest genetic risk factor for MS. Remnants of ancient retroviruses in the human genome, termed human endogenous retroviruses (HERV), and Epstein-Barr virus (EBV) infection are also associated with MS. In silico analyses of human endogenous retroviral envelope (HERV env) proteins and three myelin proteins that are principal targets of an autoimmune response in MS showed sequence similarities between potential TH epitopes within pairs of viral and myelin peptides predicted to bind HLA DR2b. This led to the proposal that such molecular mimicry may potentially trigger MS. HLA DR2b binding characteristics of previously identified peptides from the three myelin proteins and HERV env proteins as well as additional in silico predicted peptides from other encephalitogenic brain proteins and EBV proteins were studied to further investigate molecular mimicry. Peptides containing potential TH epitopes from the myelin oligodendrocyte glycoprotein and HERV env previously predicted to bind HLA DR2b as well as other pertinent potential HLA DR2b-restricted TH epitopes were confirmed to bind HLA DR2b molecules. Molecular modelling of HLA DR2b in complex with high affinity peptides derived from MOG and HERV env proteins showed that their binding could occur in a similar manner to a HLA DR2b-binding peptide containing a known TH epitope. A structurally related pair of peptides predicted to bind HLA DR2b from the EBV protein EBNA1 and ß synuclein, a brain protein implicated in MS, were also shown to similarly bind HLA DR2b. The findings justify investigating CD4+ T cell responses to the identified peptides.


Assuntos
Retrovirus Endógenos/química , Produtos do Gene env/química , Cadeias beta de HLA-DR/química , Herpesvirus Humano 4/química , Esclerose Múltipla/genética , Proteína Básica da Mielina/química , Glicoproteína Mielina-Oligodendrócito/química , beta-Sinucleína/química , Sequência de Aminoácidos/genética , Retrovirus Endógenos/genética , Epitopos/química , Produtos do Gene env/genética , Cadeias beta de HLA-DR/genética , Herpesvirus Humano 4/genética , Humanos , Modelos Moleculares , Mimetismo Molecular , Esclerose Múltipla/etiologia , Esclerose Múltipla/imunologia , Proteína Básica da Mielina/genética , Proteína Proteolipídica de Mielina/genética , Proteína Proteolipídica de Mielina/metabolismo , Glicoproteína Mielina-Oligodendrócito/genética , Ligação Proteica , Fatores de Risco , Linfócitos T/química , Linfócitos T/imunologia , beta-Sinucleína/genética , beta-Sinucleína/metabolismo
12.
Analyst ; 145(1): 52-60, 2019 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-31764916

RESUMO

Surface plasmon resonance (SPR) nanowire array chips with a microfluidic system are an effective detection method for a rapid test device. This study investigated a capped gold nanowire array and a microfluidic test platform to provide a fundamental understanding of the kinetic binding of SPR nanowires and the surface gold refractive index. The device sensitivity of the SPR nanowire array was 485 nm RIU-1 and the detection limit was 4.1 × 10-5 RIU. Moreover, a kinetic binding analysis also indicated that a peak shift resulted from a specific hybridization of the target molecule with the immobilized probe on the gold nanostructures. The peak shift (red-shift) value of latent membrane protein 1 (LMP1) DNA was 2.21 nm. The results demonstrated that this new method had high sensitivity to detect amplified DNA products without labeling or complex sample treatment. The SPR nanowire chip can detect the PCR products at lower cycle numbers compared to gel electrophoresis due to probe and DNA specificity. Furthermore, the mechanisms of SPR nanowire array fabrication and the detection of the LMP1 gene were studied. The findings can assist in improving the biosensing of DNA-amplified products and in developing rapid detection devices with a small-footprint nanostructured SPR chip.


Assuntos
DNA Viral/análise , Dispositivos Lab-On-A-Chip , Técnicas Analíticas Microfluídicas/métodos , Nanofios/química , Ressonância de Plasmônio de Superfície/métodos , Proteínas da Matriz Viral/genética , Sequência de Bases , Técnicas Biossensoriais/métodos , Sondas de DNA/química , Sondas de DNA/genética , DNA Viral/genética , Ouro/química , Herpesvirus Humano 4/química , Ácidos Nucleicos Imobilizados/química , Ácidos Nucleicos Imobilizados/genética , Limite de Detecção , Técnicas Analíticas Microfluídicas/instrumentação , Hibridização de Ácido Nucleico
13.
Biosens Bioelectron ; 143: 111610, 2019 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-31445386

RESUMO

A label-free and efficient electrochemical biosensor was developed for the ultrasensitive detection of EBV-related DNA by combing AgDNCs@DNA/AgNCs nanocomposites with noncanonical lambda exonuclease (λ exo)-assisted target recycling (LNTR). The conjugates of AgDNCs, DNA/AgNCs and probe DNA (pDNA-AgDNCs@DNA/AgNCs conjugates) worked as not only ideal nanocarriers but also efficient electrochemical tags. LNTR didn't require phosphorylated substrates and could be triggered specifically by target DNA, leading to the recycling use of target DNA and the liberation of plentiful linker probes (LP). Subsequently, the LP hybridized with the capture probes on the electrode and then bond to pDNA-AgDNCs@DNA/AgNCs conjugates, generating a sensitive electric signal directly. What's more, the signal amplification effects of DNA/AgNCs and LNTR were investigated. Under the optimal conditions, the proposed method exhibited a wide linear range of 1 fM to 1 nM and the detection limit down to 0.38 fM. In addition, the developed biosensing method exhibited excellent specificity and was successfully applied to detect target DNA in complex biological matrix. The proposed biosensor without extra bio-labels may provide a promising platform in bioanalysis and biochemical research.


Assuntos
Técnicas Biossensoriais , DNA Viral/isolamento & purificação , Técnicas Eletroquímicas , Herpesvirus Humano 4/isolamento & purificação , DNA Viral/química , Exonucleases/química , Herpesvirus Humano 4/química , Humanos , Nanocompostos/química
14.
RNA Biol ; 15(11): 1410-1419, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30339041

RESUMO

MicroRNAs (miRNAs) are small RNA molecules that post-transcriptionally regulate gene expression through silencing of complementary target mRNAs. miRNAs are involved in many biological processes, including cell proliferation, differentiation, cell signaling and cellular defense responses to infection. Strategies that allow for strong and stable suppression of specific microRNA activity are needed to study miRNA functions and to develop therapeutic intervention strategies aimed at interfering with miRNA activity in vivo. One of these classes of miRNA inhibitors are Tough Decoys (TuD) RNAs, which comprise of an imperfect RNA hairpin structure that harbors two opposing miRNA binding sites. Upon developing TuDs targeting Epstein-Barr virus miRNAs, we observed a strong variation in inhibitory potential between different TuD RNAs targeting the same miRNA. We show that the composition of the 'bulge' sequence in the miRNA binding sites has a strong impact on the inhibitory potency of the TuD. Our data implies that miRNA inhibition correlates with the thermodynamic properties of the TuD and that design aimed at lowering the TuD opening energy increases TuD potency. Our study provides specific guidelines for the design and construction of potent decoy-based miRNA inhibitors, which may be used for future therapeutic intervention strategies.


Assuntos
MicroRNAs/genética , Conformação de Ácido Nucleico , RNA/genética , Sítios de Ligação , Herpesvirus Humano 4/química , Herpesvirus Humano 4/genética , Humanos , MicroRNAs/antagonistas & inibidores , MicroRNAs/química , RNA/química , RNA Interferente Pequeno/química , RNA Interferente Pequeno/genética , Termodinâmica
15.
J Virol ; 92(18)2018 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-29976663

RESUMO

Epstein Barr virus (EBV) is a potentially oncogenic gammaherpesvirus that establishes a chronic, latent infection in memory B cells. The EBV genome persists in infected host cells as a chromatinized episome and is subject to chromatin-mediated regulation. Binding of the host insulator protein CTCF to the EBV genome has an established role in maintaining viral latency type. CTCF is posttranslationally modified by the host enzyme PARP1. PARP1, or poly(ADP-ribose) polymerase 1, catalyzes the transfer of a poly(ADP-ribose) (PAR) moiety from NAD+ onto acceptor proteins, including itself, histone proteins, and CTCF. PARylation of CTCF by PARP1 can affect CTCF's insulator activity, DNA binding capacity, and ability to form chromatin loops. Both PARP1 and CTCF have been implicated in the regulation of EBV latency and lytic reactivation. Thus, we predicted that pharmacological inhibition with PARP1 inhibitors would affect EBV latency type through a chromatin-specific mechanism. Here, we show that PARP1 and CTCF colocalize at specific sites throughout the EBV genome and provide evidence to suggest that PARP1 acts to stabilize CTCF binding and maintain the open chromatin landscape at the active Cp promoter during type III latency. Further, PARP1 activity is important in maintaining latency type-specific viral gene expression. The data presented here provide a rationale for the use of PARP inhibitors in the treatment of EBV-associated cancers exhibiting type III latency and ultimately could contribute to an EBV-specific treatment strategy for AIDS-related or posttransplant lymphomas.IMPORTANCE EBV is a human gammaherpesvirus that infects more than 95% of individuals worldwide. Upon infection, EBV circularizes as an episome and establishes a chronic, latent infection in B cells. In doing so, the virus utilizes host cell machinery to regulate and maintain the viral genome. In otherwise healthy individuals, EBV infection is typically nonpathological; however, latent infection is potentially oncogenic and is responsible for 1% of human cancers. During latent infection, EBV expresses specific sets of proteins according to the given latency type, each of which is associated with specific types of cancers. For example, type III latency, in which the virus expresses its full repertoire of latent proteins, is characteristic of AIDS-associated and posttransplant lymphomas associated with EBV infection. Understanding how viral latency type is regulated at the chromatin level may reveal potential targets for EBV-specific pharmacological intervention in EBV-associated cancers.


Assuntos
Fator de Ligação a CCCTC/genética , Herpesvirus Humano 4/fisiologia , Poli(ADP-Ribose) Polimerase-1/genética , Latência Viral/genética , Fator de Ligação a CCCTC/metabolismo , Linhagem Celular Tumoral , Cromatina/genética , Cromatina/metabolismo , Epigenômica , Regulação Viral da Expressão Gênica , Genoma Viral , Herpesvirus Humano 4/química , Herpesvirus Humano 4/genética , Humanos , Poli(ADP-Ribose) Polimerase-1/antagonistas & inibidores , Poli(ADP-Ribose) Polimerase-1/efeitos dos fármacos , Leucemia-Linfoma Linfoblástico de Células Precursoras/virologia , Regiões Promotoras Genéticas , Transcrição Gênica , Latência Viral/efeitos dos fármacos
16.
Bull Exp Biol Med ; 163(6): 766-771, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-29063322

RESUMO

Intracellular fragments of latent phase protein LMP1 of Epstein-Barr virus, denoted as CTAR1/2/3, can trigger a variety of cell cascades and contribute to the transforming potential of the virus. Generation of recombinant proteins CTAR1/2/3 is expected to yield more ample data on functional and immunogenic characteristics of LMP1. We created genetic constructs for prokaryotic expression of LMP1 CTAR fragments and selected optimal conditions for their production and purification. Using a new library of LMP1 CTAR fragments, we carried out epitope mapping of a diagnostic anti-LMP1 antibody S12. Analysis of polyclonal serum antibodies from mice immunized with full-length LMP1 confirmed immunogenicity of CTAR elements comparable with that of full-length protein.


Assuntos
Anticorpos Monoclonais/química , Anticorpos Antivirais/química , Fragmentos de Peptídeos/imunologia , Proteínas da Matriz Viral/imunologia , Latência Viral/imunologia , Animais , Anticorpos Monoclonais/biossíntese , Anticorpos Monoclonais/isolamento & purificação , Anticorpos Antivirais/biossíntese , Anticorpos Antivirais/isolamento & purificação , Clonagem Molecular , Mapeamento de Epitopos/métodos , Epitopos/genética , Epitopos/imunologia , Escherichia coli/genética , Escherichia coli/metabolismo , Expressão Gênica , Vetores Genéticos/química , Vetores Genéticos/metabolismo , Herpesvirus Humano 4/química , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/imunologia , Humanos , Imunização , Camundongos , Fragmentos de Peptídeos/genética , Biblioteca de Peptídeos , Domínios Proteicos , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Proteínas da Matriz Viral/genética
17.
Protein Sci ; 26(6): 1231-1235, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28383161

RESUMO

Epstein-Barr virus EBNA-LP protein is a transcriptional coactivator of EBNA2. Efficient nuclear localization of EBNA-LP is essential for cooperation with EBNA2. Here, we report the crystal structure of the nuclear import adaptor importin-α1 bound to the nuclear localization signal (NLS) of EBNA-LP that shows EBNA-LP residues 44-RRVRRR-49 binding to the major NLS-binding site at the P0-P5 positions. In contrast to previously characterized classical NLSs that invariably have a basic residue [either lysine (in the vast majority of cases) or arginine] at the P2 position, the EBNA-LP NLS is unique in that it has valine at the P2 position. The loss of the critical P2 lysine (or arginine) is compensated by arginine at the P0 position in the EBNA-LP NLS.


Assuntos
Herpesvirus Humano 4/química , Sinais de Localização Nuclear/química , Proteínas Virais/química , alfa Carioferinas/química , Cristalografia por Raios X , Herpesvirus Humano 4/metabolismo , Humanos , Sinais de Localização Nuclear/metabolismo , Estrutura Quaternária de Proteína , Proteínas Virais/metabolismo , alfa Carioferinas/metabolismo
18.
Nat Commun ; 7: 13557, 2016 12 08.
Artigo em Inglês | MEDLINE | ID: mdl-27929061

RESUMO

Herpesvirus entry into host cells is mediated by multiple virally encoded receptor binding and membrane fusion glycoproteins. Despite their importance in host cell tropism and associated disease pathology, the underlying and essential interactions between these viral glycoproteins remain poorly understood. For Epstein-Barr virus (EBV), gHgL/gp42 complexes bind HLA class II to activate membrane fusion with B cells, but gp42 inhibits fusion and entry into epithelial cells. To clarify the mechanism by which gp42 controls the cell specificity of EBV infection, here we determined the structure of gHgL/gp42 complex bound to an anti-gHgL antibody (E1D1). The critical regulator of EBV tropism is the gp42 N-terminal domain, which tethers the HLA-binding domain to gHgL by wrapping around the exterior of three gH domains. Both the gp42 N-terminal domain and E1D1 selectively inhibit epithelial-cell fusion; however, they engage distinct surfaces of gHgL. These observations clarify key determinants of EBV host cell tropism.


Assuntos
Herpesvirus Humano 4/fisiologia , Proteínas do Envelope Viral/fisiologia , Tropismo Viral , Animais , Células CHO , Cricetulus , Células Epiteliais/virologia , Herpesvirus Humano 4/química , Mutação , Conformação Proteica , Proteínas do Envelope Viral/química
19.
Biologicals ; 44(5): 423-33, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27461128

RESUMO

Variability in viral load measurements using nucleic acid amplification techniques (NAT) has a significant impact on the management of Epstein-Barr virus (EBV)-associated diseases, and has highlighted a need for standardisation of these measurements. The aim of this collaborative study was to evaluate the suitability of a range of candidate reference materials to harmonise EBV viral load measurements in a wide range of NAT assays. Candidate materials included lyophilised and liquid whole virus preparations of the EBV B95-8 strain, and preparations of Namalwa and Raji cells. Variability between the individual laboratory mean estimates for each candidate was 2.5 log10 copies/mL. The agreement between laboratories was improved when the potency of each candidate was expressed relative to the lyophilised B95-8 preparation. The results of the study indicate the suitability of this candidate as the 1st WHO International Standard for EBV for NAT. It was established in October 2011 by the WHO's Expert Committee on Biological Standardisation with an assigned potency of 5 × 10(6) International Units (IU) (NIBSC code 09/260). It is intended to be used for the calibration of secondary reference materials, used in EBV NAT assays, in IU, thereby improving the comparability of patient viral load measurements.


Assuntos
DNA Viral/química , DNA Viral/normas , Herpesvirus Humano 4/química , Técnicas de Amplificação de Ácido Nucleico/normas , Animais , Linhagem Celular , Humanos , Camundongos , Técnicas de Amplificação de Ácido Nucleico/métodos
20.
J Comput Aided Mol Des ; 30(4): 285-303, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27048620

RESUMO

The Epstein-Barr Nuclear Antigen 1 (EBNA1) is a critical protein encoded by the Epstein-Barr Virus (EBV). During latent infection, EBNA1 is essential for DNA replication and transcription initiation of viral and cellular genes and is necessary to immortalize primary B-lymphocytes. Nonetheless, the concept of EBNA1 as drug target is novel. Two EBNA1 crystal structures are publicly available and the first small-molecule EBNA1 inhibitors were recently discovered. However, no systematic studies have been reported on the structural details of EBNA1 "druggable" binding sites. We conducted computational identification and structural characterization of EBNA1 binding pockets, likely to accommodate ligand molecules (i.e. "druggable" binding sites). Then, we validated our predictions by docking against a set of compounds previously tested in vitro for EBNA1 inhibition (PubChem AID-2381). Finally, we supported assessments of pocket druggability by performing induced fit docking and molecular dynamics simulations paired with binding affinity predictions by Molecular Mechanics Generalized Born Surface Area calculations for a number of hits belonging to druggable binding sites. Our results establish EBNA1 as a target for drug discovery, and provide the computational evidence that active AID-2381 hits disrupt EBNA1:DNA binding upon interacting at individual sites. Lastly, structural properties of top scoring hits are proposed to support the rational design of the next generation of EBNA1 inhibitors.


Assuntos
Descoberta de Drogas , Infecções por Vírus Epstein-Barr/tratamento farmacológico , Antígenos Nucleares do Vírus Epstein-Barr/química , Herpesvirus Humano 4/efeitos dos fármacos , Linfócitos B/efeitos dos fármacos , Linfócitos B/virologia , Sítios de Ligação , Simulação por Computador , Cristalografia por Raios X , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/efeitos dos fármacos , Infecções por Vírus Epstein-Barr/virologia , Antígenos Nucleares do Vírus Epstein-Barr/uso terapêutico , Herpesvirus Humano 4/química , Herpesvirus Humano 4/patogenicidade , Humanos , Ligantes , Conformação Proteica/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...