Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
J Med Virol ; 95(3): e28659, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36905218

RESUMO

Kaposi sarcoma-associated herpesvirus (KSHV) is the etiological agent of several human diseases, including Kaposi sarcoma, primary effusion lymphoma, and a subset of multicentric Castleman's disease. KSHV uses its gene products to manipulate many aspects of the host responses during its life cycles. Among KSHV-encoded proteins, ORF45 is unique in both temporal and spatial expression: it is expressed as an immediate-early gene product and is an abundant tegument protein contained in the virion. ORF45 is specific to the gammaherpesvirinae subfamily but the homologs share only very limited homology and differ dramatically in protein length. In the past two decades, we and others have shown that ORF45 plays critical roles in immune evasion, viral replication, and virion assembly by targeting various host and viral factors. Herein, we summarize our current knowledge of ORF45 throughout the KSHV life cycle. We discuss the cellular processes targeted by ORF45 with emphasis on the modulation of host innate immune responses and rewiring the host signaling through impacting three major posttranslational modifications: phosphorylation, SUMOylation, and ubiquitination.


Assuntos
Herpesvirus Humano 8 , Proteínas Imediatamente Precoces , Sarcoma de Kaposi , Humanos , Linhagem Celular , Proteínas Imediatamente Precoces/genética , Proteínas Imediatamente Precoces/metabolismo , Replicação Viral/fisiologia
2.
J Biol Chem ; 299(3): 102986, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36754281

RESUMO

Dengue virus (DENV) is one of the most prevalent mosquito-transmitted human viruses that causes significant morbidity and mortality worldwide. To persist in the cell and consequently cause disease, DENV is evolved with mechanisms to suppress the induction of type I interferons by antagonizing cGAS-STING signaling. Using recombinant proteins and in vitro cleavage assays, we have shown that the DENV protease NS2B3 is capable of cleaving cGAS in the N-terminal region without disrupting the C-terminal catalytic center. This generates two major cleavage products: cleavage product N-terminal (CP-N) and cleavage product C-terminal (CP-C). We observed reduction in DNA-binding affinity of CP-C as compared to full-length cGAS. Reduction in DNA-binding affinity is also correlated with the decrease in enzymatic activity of CP-C. CP-N, on the other hand, has almost comparable DNA-binding ability as that of the full-length cGAS. In fact, CP-N competitively inhibits cyclic GMP-AMP production by both full-length cGAS and CP-C. We hypothesize that high DNA-binding affinity of CP-N enables it to sequester the DNA from CP-C and noncleaved full-length cGAS and thus reduces the rate of enzyme activation and cyclic GMP-AMP synthesis. Furthermore, we found that NS2B3 physically interacts with full-length cGAS and CP-C, laying the basis for their shuttling to and eventual degradation in the autophagosome. Overall, our study highlights a multifaceted and effective strategy by which an RNA virus antagonizes cGAS-STING signaling which may be useful for the design of antivirals targeting viral proteases.


Assuntos
Vírus da Dengue , Nucleotidiltransferases , Peptídeo Hidrolases , Humanos , Vírus da Dengue/enzimologia , Imunidade Inata , Nucleotidiltransferases/metabolismo , Peptídeo Hidrolases/metabolismo
3.
Proc Natl Acad Sci U S A ; 119(33): e2200285119, 2022 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-35939686

RESUMO

Kaposi's sarcoma-associated herpesvirus (KSHV) inhibitor of cyclic GMP-AMP synthase (cGAS) (KicGAS) encoded by ORF52 is a conserved major tegument protein of KSHV and the first reported viral inhibitor of cGAS. In our previous study, we found that KicGAS is highly oligomerized in solution and that oligomerization is required for its cooperative DNA binding and for inhibiting DNA-induced phase separation and activation of cGAS. However, how KicGAS oligomerizes remained unclear. Here, we present the crystal structure of KicGAS at 2.5 Å resolution, which reveals an "L"-shaped molecule with each arm of the L essentially formed by a single α helix (α1 and α2). Antiparallel dimerization of α2 helices from two KicGAS molecules leads to a unique "Z"-shaped dimer. Surprisingly, α1 is also a dimerization domain. It forms a parallel dimeric leucine zipper with the α1 from a neighboring dimer, leading to the formation of an infinite chain of KicGAS dimers. Residues involved in leucine zipper dimer formation are among the most conserved residues across ORF52 homologs of gammaherpesviruses. The self-oligomerization increases the valence and cooperativity of interaction with DNA. The resultant multivalent interaction is critical for the formation of liquid condensates with DNA and consequent sequestration of DNA from being sensed by cGAS, explaining its role in restricting cGAS activation. The structure presented here not only provides a mechanistic understanding of the function of KicGAS but also informs a molecular target for rational design of antivirals against KSHV and related viruses.


Assuntos
Herpesvirus Humano 8 , Nucleotidiltransferases , Proteínas Estruturais Virais , Herpesvirus Humano 8/metabolismo , Humanos , Nucleotidiltransferases/metabolismo , Domínios Proteicos , Multimerização Proteica , Proteínas Estruturais Virais/química
4.
Nat Immunol ; 23(6): 916-926, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35618833

RESUMO

At steady state, the NOD-like receptor (NLR)-containing pyrin domain (PYD) (NLRP)1 inflammasome is maintained in an auto-inhibitory complex by dipeptidyl peptidases 8 and 9 (DPP8 and DPP9) and is activated by pathogen-encoded proteases after infection. Here, we showed that the open reading frame (ORF)45 protein of the Kaposi's sarcoma-associated herpesvirus activated the human NLRP1 (hNLRP1) inflammasome in a non-protease-dependent manner, and we additionally showed that the Linker1 region of hNLRP1, situated between the PYD and NACHT domains, was required for the auto-inhibition and non-protease-dependent activation of hNLRP1. At steady state, the interaction between Linker1 and the UPA subdomain silenced the activation of hNLRP1 in auto-inhibitory complexes either containing DPP9 or not in a manner independent of DPP9. ORF45 binding to Linker1 displaced UPA from the Linker1-UPA complex and induced the release of the C-terminal domain of hNLRP1 for inflammasome assembly. The ORF45-dependent activation of the NLRP1 inflammasome was conserved in primates but was not observed for murine NLRP1b inflammasomes.


Assuntos
Herpesvirus Humano 8 , Inflamassomos , Proteínas Virais/metabolismo , Animais , Proteínas Adaptadoras de Sinalização CARD/metabolismo , Herpesvirus Humano 8/metabolismo , Humanos , Inflamassomos/metabolismo , Camundongos , Proteínas NLR/química , Proteínas NLR/metabolismo
5.
PLoS Pathog ; 18(4): e1010504, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35482828

RESUMO

RSK1, an essential cellular kinase for Kaposi's sarcoma-associated herpesvirus (KSHV) replication, is highly phosphorylated and SUMOylated during KSHV lytic cycle, which determine the substrate phosphorylation and specificity of RSK1, respectively. However, the SUMO E3 ligase responsible for attaching SUMO to RSK1 has not yet been identified. By genome-wide screening, we found that KSHV ORF45 is necessary and sufficient to enhance RSK1 SUMOylation. Mechanistically, KSHV ORF45 binds to SUMOs via two classic SUMO-interacting motifs (SIMs) and functions as a SIM-dependent SUMO E3 ligase for RSK1. Mutations on these ORF45 SIMs resulted in much lower lytic gene expressions, viral DNA replication, and mature progeny virus production. Interestingly, KSHV ORF45 controls RSK1 SUMOylation and phosphorylation via two separated functional regions: SIMs and amino acid 17-90, respectively, which do not affect each other. Similar to KSHV ORF45, ORF45 of Rhesus Macaque Rhadinovirus has only one SIM and also increases RSK1 SUMOylation in a SIM-dependent manner, while other ORF45 homologues do not have this function. Our work characterized ORF45 as a novel virus encoded SUMO E3 ligase, which is required for ORF45-RSK1 axis-mediated KSHV lytic gene expression.


Assuntos
Herpesvirus Humano 8 , Proteínas Imediatamente Precoces , Animais , Linhagem Celular , Replicação do DNA , DNA Viral , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/metabolismo , Proteínas Imediatamente Precoces/genética , Proteínas Imediatamente Precoces/metabolismo , Macaca mulatta/metabolismo , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo , Replicação Viral
6.
Nat Commun ; 13(1): 472, 2022 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-35078976

RESUMO

The Kaposi's sarcoma associated herpesvirus protein ORF45 binds the extracellular signal-regulated kinase (ERK) and the p90 Ribosomal S6 kinase (RSK). ORF45 was shown to be a kinase activator in cells but a kinase inhibitor in vitro, and its effects on the ERK-RSK complex are unknown. Here, we demonstrate that ORF45 binds ERK and RSK using optimized linear binding motifs. The crystal structure of the ORF45-ERK2 complex shows how kinase docking motifs recognize the activated form of ERK. The crystal structure of the ORF45-RSK2 complex reveals an AGC kinase docking system, for which we provide evidence that it is functional in the host. We find that ORF45 manipulates ERK-RSK signaling by favoring the formation of a complex, in which activated kinases are better protected from phosphatases and docking motif-independent RSK substrate phosphorylation is selectively up-regulated. As such, our data suggest that ORF45 interferes with the natural design of kinase docking systems in the host.


Assuntos
Cristalografia por Raios X/métodos , Herpesvirus Humano 8/metabolismo , Proteínas Imediatamente Precoces/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/química , Proteínas Quinases S6 Ribossômicas 90-kDa/química , Sarcoma de Kaposi/metabolismo , Linhagem Celular , Biologia Computacional , Herpesvirus Humano 8/química , Herpesvirus Humano 8/isolamento & purificação , Humanos , Proteínas Imediatamente Precoces/química , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Fosforilação , Proteínas Quinases S6 Ribossômicas 90-kDa/metabolismo , Sarcoma de Kaposi/patologia , Sarcoma de Kaposi/virologia , Transdução de Sinais
7.
PLoS Pathog ; 17(12): e1010123, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34871326

RESUMO

RSK1, a downstream kinase of the MAPK pathway, has been shown to regulate multiple cellular processes and is essential for lytic replication of a variety of viruses, including Kaposi's sarcoma-associated herpesvirus (KSHV). Besides phosphorylation, it is not known whether other post-translational modifications play an important role in regulating RSK1 function. We demonstrate that RSK1 undergoes robust SUMOylation during KSHV lytic replication at lysine residues K110, K335, and K421. SUMO modification does not alter RSK1 activation and kinase activity upon KSHV ORF45 co-expression, but affects RSK1 downstream substrate phosphorylation. Compared to wild-type RSK1, the overall phosphorylation level of RxRxxS*/T* motif is significantly declined in RSK1K110/335/421R expressing cells. Specifically, SUMOylation deficient RSK1 cannot efficiently phosphorylate eIF4B. Sequence analysis showed that eIF4B has one SUMO-interacting motif (SIM) between the amino acid position 166 and 170 (166IRVDV170), which mediates the association between eIF4B and RSK1 through SUMO-SIM interaction. These results indicate that SUMOylation regulates the phosphorylation of RSK1 downstream substrates, which is required for efficient KSHV lytic replication.


Assuntos
Herpesvirus Humano 8/fisiologia , Interações Hospedeiro-Patógeno/fisiologia , Proteínas Quinases S6 Ribossômicas 90-kDa/metabolismo , Sumoilação/fisiologia , Replicação Viral/fisiologia , Linhagem Celular , Humanos
8.
Viruses ; 13(9)2021 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-34578410

RESUMO

The Open Reading Frame 45 (ORF45) of Kaposi sarcoma-associated herpesvirus (KSHV) is a gammaherpesvirus-specific, immediate-early, tegument protein required for efficient viral replication and virion production. We have previously shown that ORF45 interacts with the conserved herpesviral protein ORF33 through the highly conserved C-terminal 19 amino acids (C19) of ORF45. Because the deletion of C19 abolished ORF33 accumulation and viral production, we reasoned that this interaction could be critical for viral production and explored as an antiviral target for gammaherpesviruses. In work described in this article, we characterize this interaction in further detail, first by revealing that this interaction is conserved among gammaherpesviruses, then by identifying residues in C19 critical for its interaction with and stabilization of ORF33. More importantly, we show that disruption of the interaction, either by mutating key residues (W403A or W405A) in C19 or by using competing cell penetration peptide TAT-C19, dramatically reduce the yield of KSHV progeny viruses. Our results not only reveal critical roles of this interaction to viral production but also provide a proof of concept for targeting the ORF33-ORF45 interaction as a novel antiviral strategy against KSHV and other gammaherpesviruses.


Assuntos
Proteínas do Capsídeo/fisiologia , Herpesvirus Humano 8/fisiologia , Proteínas Imediatamente Precoces/fisiologia , Replicação Viral/fisiologia , Domínio Catalítico , Sequência Conservada , Células HEK293 , Herpesvirus Humano 8/genética , Humanos , Mutagênese , Peptídeos/metabolismo , Estabilidade Proteica , Vírion/genética , Vírion/fisiologia , Replicação Viral/genética
9.
Nucleic Acids Res ; 49(16): 9389-9403, 2021 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-34387695

RESUMO

Cyclic GMP-AMP synthase (cGAS) is a key DNA sensor that detects aberrant cytosolic DNA arising from pathogen invasions or genotoxic stresses. Upon binding to DNA, cGAS is activated and catalyzes the synthesis of cyclic GMP-AMP (cGAMP), which induces potent antimicrobial and antitumor responses. Kaposi sarcoma-associated herpesvirus (KSHV) is a human DNA tumor virus that causes Kaposi sarcoma and several other malignancies. We previously reported that KSHV inhibitor of cGAS (KicGAS) encoded by ORF52, inhibits cGAS enzymatic activity, but the underlying mechanisms remained unclear. To define the inhibitory mechanisms, here we performed in-depth biochemical and functional characterizations of KicGAS, and mapped its functional domains. We found KicGAS self-oligomerizes and binds to double stranded DNA cooperatively. This self-oligomerization is essential for its DNA binding and cGAS inhibition. Interestingly, KicGAS forms liquid droplets upon binding to DNA, which requires collective multivalent interactions with DNA mediated by both structured and disordered domains coordinated through the self-oligomerization of KicGAS. We also observed that KicGAS inhibits the DNA-induced phase separation and activation of cGAS. Our findings reveal a novel mechanism by which DNA viruses target the host protein phase separation for suppression of the host sensing of viral nucleic acids.


Assuntos
Herpesvirus Humano 8/genética , Interações Hospedeiro-Patógeno/genética , Nucleotidiltransferases/genética , Sarcoma de Kaposi/genética , Citosol/enzimologia , Citosol/microbiologia , Quebras de DNA de Cadeia Dupla/efeitos dos fármacos , Dano ao DNA/genética , DNA Viral/genética , Proteínas de Ligação a DNA/genética , Herpesvirus Humano 8/patogenicidade , Humanos , Evasão da Resposta Imune/efeitos dos fármacos , Imunidade Inata/genética , Nucleotídeos Cíclicos/genética , Nucleotidiltransferases/antagonistas & inibidores , Sarcoma de Kaposi/tratamento farmacológico , Sarcoma de Kaposi/virologia , Proteínas Virais/genética
10.
Front Cell Infect Microbiol ; 11: 647992, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33791247

RESUMO

Sensing of viral constituents is the first and critical step in the host innate immune defense against viruses. In mammalian cells, there are a variety of pathogen recognition receptors (PRRs) that detect diverse pathogen-associated molecular patterns (PAMPs) including viral RNA and DNA. In the past decade, a number of host DNA sensors have been discovered and the underlying sensing mechanisms have been elucidated. Herpesviruses belong to a large family of enveloped DNA viruses. They are successful pathogens whose elaborate immune evasion mechanisms contribute to high prevalence of infection among their hosts. The three subfamilies of herpesviruses have all been found to employ diverse and overlapping strategies to interfere with host DNA sensing. These strategies include masking viral DNA or the DNA sensor, degradation of the DNA sensor, and post-transcriptional modification of the DNA sensor or its adaptor protein. In this review, we will discuss the current state of our knowledge on how human herpesviruses use these strategies to evade DNA-induced immune responses. Comprehensive understanding of herpesvirus immune-evasion mechanisms will aid in the development of vaccines and antivirals for herpesvirus-associated diseases.


Assuntos
Herpesviridae , Imunidade Inata , Animais , DNA Viral , Interações Hospedeiro-Patógeno , Humanos , Evasão da Resposta Imune , Moléculas com Motivos Associados a Patógenos
11.
Mol Ther Nucleic Acids ; 17: 175-184, 2019 Sep 06.
Artigo em Inglês | MEDLINE | ID: mdl-31265948

RESUMO

Nasopharyngeal carcinoma (NPC) is an invasive head-and-neck tumor with Epstein-Barr virus (EBV) as an important etiological cause. The EBV oncoprotein Latent membrane protein 1 (LMP1) can be trafficked into exosomes with unclear roles, and this trafficking is a potential problem in NPC control. MicroRNA-203 (miR-203) was found by us to be downregulated by LMP1, and it functions as a tumor suppressor in NPC. In this study, aspirin reversed the epithelial-mesenchymal transition (EMT) by promoting miR-203 expression in cells, and, remarkably, it repressed exosomal LMP1 (exo-LMP1) secretion from EBV-positive cells. Nuclear factor κB (NF-κB) activation was required for the exo-LMP1 production. The exo-LMP1 uptake influenced the EMT potential of EBV-negative recipient NPC cells. The exo-LMP1 level was upregulated in clinical NPC plasma samples. Aspirin treatment observably inhibited NPC lung metastasis in nude mice. The study revealed that aspirin is a promising drug for NPC therapy via its targeting of exo-LMP1 transfer and the regulatory effect of LMP1 on miR-203 expression. EBV can regulate its own tumorigenesis via the LMP1/NF-κB/exo-LMP1 axis, opening a new avenue for understanding the pathogenesis of this tumor virus. Our study also provides a rationale for the use of exo-LMP1 or exosomal miR-203 (exo-miR203) in EBV-targeted therapy by aspirin in invasive NPC.

12.
Nature ; 569(7758): 718-722, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-31118511

RESUMO

Nucleic acids from bacteria or viruses induce potent immune responses in infected cells1-4. The detection of pathogen-derived nucleic acids is a central strategy by which the host senses infection and initiates protective immune responses5,6. Cyclic GMP-AMP synthase (cGAS) is a double-stranded DNA sensor7,8. It catalyses the synthesis of cyclic GMP-AMP (cGAMP)9-12, which stimulates the induction of type I interferons through the STING-TBK1-IRF-3 signalling axis13-15. STING oligomerizes after binding of cGAMP, leading to the recruitment and activation of the TBK1 kinase8,16. The IRF-3 transcription factor is then recruited to the signalling complex and activated by TBK18,17-20. Phosphorylated IRF-3 translocates to the nucleus and initiates the expression of type I interferons21. However, the precise mechanisms that govern activation of STING by cGAMP and subsequent activation of TBK1 by STING remain unclear. Here we show that a conserved PLPLRT/SD motif within the C-terminal tail of STING mediates the recruitment and activation of TBK1. Crystal structures of TBK1 bound to STING reveal that the PLPLRT/SD motif binds to the dimer interface of TBK1. Cell-based studies confirm that the direct interaction between TBK1 and STING is essential for induction of IFNß after cGAMP stimulation. Moreover, we show that full-length STING oligomerizes after it binds cGAMP, and highlight this as an essential step in the activation of STING-mediated signalling. These findings provide a structural basis for the development of STING agonists and antagonists for the treatment of cancer and autoimmune disorders.


Assuntos
Motivos de Aminoácidos , Sequência Conservada , Proteínas de Membrana/química , Proteínas de Membrana/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Cristalografia por Raios X , Ativação Enzimática , Células HEK293 , Humanos , Interferon beta/metabolismo , Proteínas de Membrana/genética , Modelos Moleculares , Mutação , Nucleotídeos Cíclicos/metabolismo , Ligação Proteica , Transdução de Sinais
13.
J Virol ; 93(10)2019 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-30842327

RESUMO

The lytic replication of Kaposi's sarcoma-associated herpesvirus (KSHV) requires sustained extracellular signal-regulated kinase (ERK)-p90 ribosomal S6 kinase (RSK) activation, which is induced by an immediate early (IE) gene-encoded tegument protein called ORF45, to promote the late transcription and translation of viral lytic genes. An ORF45-null or single-point F66A mutation in ORF45 abolishes ORF45-RSK interaction and sustained ERK-RSK activation during lytic reactivation and subsequently results in a significant decrease in late lytic gene expression and virion production, indicating that ORF45-mediated RSK activation plays a critical role in KSHV lytic replication. Here, we demonstrate that a short ORF45-derived peptide in the RSK-binding region is sufficient for disrupting ORF45-RSK interaction, consequently suppressing lytic gene expression and virion production. We designed a nontoxic cell-permeable peptide derived from ORF45, TAT-10F10, which is composed of the ORF45 56 to 76 amino acid (aa) region and the HIV Tat protein transduction domain, and this peptide markedly inhibits KSHV lytic replication in iSLK.219 and BCBL1 cells. Importantly, this peptide enhances the inhibitory effect of rapamycin on KSHV-infected cells and decreases spontaneous and hypoxia-induced lytic replication in KSHV-positive lymphoma cells. These findings suggest that a small peptide that disrupts ORF45-RSK interaction might be a promising agent for controlling KSHV lytic infection and pathogenesis.IMPORTANCE ORF45-induced RSK activation plays an essential role in KSHV lytic replication, and ORF45-null or ORF45 F66A mutagenesis that abolishes sustained RSK activation and RSK inhibitors significantly decreases lytic replication, indicating that the ORF45-RSK association is a unique target for KSHV-related diseases. However, the side effects, low affinity, and poor efficacy of RSK modulators limit their clinical application. In this study, we developed a nontoxic cell-permeable ORF45-derived peptide from the RSK-binding region to disrupt ORF45-RSK associations and block ORF45-induced RSK activation without interfering with S6K1 activation. This peptide effectively suppresses spontaneous, hypoxia-induced, or chemically induced KSHV lytic replication and enhances the inhibitory effect of rapamycin on lytic replication and sensitivity to rapamycin in lytic KSHV-infected cells. Our results reveal that the ORF45-RSK signaling axis and KSHV lytic replication can be effectively targeted by a short peptide and provide a specific approach for treating KSHV lytic and persistent infection.


Assuntos
Herpesvirus Humano 8/efeitos dos fármacos , Proteínas Imediatamente Precoces/imunologia , Proteínas Quinases S6 Ribossômicas 90-kDa/metabolismo , Linhagem Celular , Regulação Viral da Expressão Gênica/genética , Genes Virais/genética , Células HEK293 , Infecções por Herpesviridae/genética , Herpesvirus Humano 8/patogenicidade , Humanos , Proteínas Imediatamente Precoces/genética , Proteínas Imediatamente Precoces/metabolismo , Ligação Proteica , Proteínas Quinases S6 Ribossômicas 90-kDa/imunologia , Vírion/metabolismo , Replicação Viral/efeitos dos fármacos , Replicação Viral/fisiologia
14.
Virol Sin ; 34(3): 253-261, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-30911896

RESUMO

Epstein-Barr virus (EBV) is an important human dsDNA virus, which has been shown to be associated with several malignancies including about 10% of gastric carcinomas. How EBV enters an epithelial cell has been an interesting project for investigation. "Cell-in-cell" infection was recently reported an efficient way for the entry of EBV into nasopharynx epithelial cells. The present approach was to explore the feasibility of this mode for EBV infection in gastric epithelial cells and the dynamic change of host inflammatory reaction. The EBV-positive lymphoblastic cells of Akata containing a GFP tag in the viral genome were co-cultured with the gastric epithelial cells (GES-1). The infection situation was observed under fluorescence and electron microscopies. Real-time quantitative PCR and Western-blotting assay were employed to detect the expression of a few specific cytokines and inflammatory factors. The results demonstrated that EBV could get into gastric epithelial cells by "cell-in-cell" infection but not fully successful due to the host fighting. IL-1ß, IL-6 and IL-8 played prominent roles in the cellular response to the infection. The activation of NF-κB and HSP70 was also required for the host antiviral response. The results imply that the gastric epithelial cells could powerfully resist the virus invader via cell-in-cell at the early stage through inflammatory and innate immune responses.


Assuntos
Formação de Célula em Célula , Células Epiteliais/virologia , Trato Gastrointestinal/virologia , Herpesvirus Humano 4/fisiologia , Linhagem Celular , Citocinas/imunologia , Células Epiteliais/imunologia , Fluorescência , Trato Gastrointestinal/citologia , Proteínas de Fluorescência Verde , Proteínas de Choque Térmico HSP72/metabolismo , Herpesvirus Humano 4/genética , Humanos , Imunidade Inata , Hibridização In Situ , Inflamação , Microscopia Eletrônica de Transmissão , NF-kappa B/metabolismo
15.
J Virol ; 93(7)2019 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-30651359

RESUMO

Kaposi's sarcoma-associated herpesvirus (KSHV; also called human herpesvirus 8 [HHV-8]), upon being reactivated, causes serious diseases in immunocompromised individuals. Its reactivation, especially how the cellular regulating mechanisms play roles in KSHV gene expression and viral DNA replication, is not fully understood. In searching for the cellular factors that regulate KSHV gene expression, we found that several histone deacetylases (HDACs) and sirtuins (SIRTs), including HDACs 2, 7, 8, and 11 and SIRTs 4 and 6, repress KSHV ori-Lyt promoter activity. Interestingly, the nuclear protein SIRT6 presents the greatest inhibitory effect on ori-Lyt promoter activity. A more detailed investigation revealed that SIRT6 exerts repressive effects on multiple promoters of KSHV. As a consequence of inhibiting the KSHV promoters, SIRT6 not only represses viral protein production but also inhibits viral DNA replication, as investigated in a KSHV-containing cell line, SLK-iBAC-gfpK52. Depletion of the SIRT6 protein using small interfering RNA could not directly reactivate KSHV from SLK-iBAC-gfpK52 cells but made the reactivation of KSHV by use of a small amount of the reactivator (doxycycline) more effective and enhanced viral DNA replication in the KSHV infection system. We performed DNA chromatin immunoprecipitation (ChIP) assays for SIRT6 in the SLK-iBAC-gfpK52 cell line to determine whether SIRT6 interacts with the KSHV genome in order to exhibit regulatory effects. Our results suggest that SIRT6 interacts with KSHV ori-Lyt and ORF50 promoters. Furthermore, the SIRT6-KSHV DNA interaction is significantly negated by reactivation. Therefore, we identified a cellular regulator, SIRT6, that represses KSHV replication by interacting with KSHV DNA and inhibiting viral gene expression.IMPORTANCE Kaposi's sarcoma-associated herpesvirus (KSHV) is a pathogen causing cancer in the immune-deficient population. The reactivation of KSHV from latency is important for it to be carcinogenic. Our finding that SIRT6 has inhibitory effects on KSHV reactivation by interacting with the viral genome and suppressing viral gene expression is important because it might lead to a strategy of interfering with KSHV reactivation. Overexpression of SIRT6 repressed the activities of several KSHV promoters, leading to reduced gene expression and DNA replication by KSHV in a KSHV bacterial artificial chromosome-containing cell line. Depletion of SIRT6 favored reactivation of KSHV from SLK-iBACV-gfpK52 cells. More importantly, we reveal that SIRT6 interacts with KSHV DNA. Whether the interaction of SIRT6 with KSHV DNA occurs at a global level will be further studied in the future.


Assuntos
Herpesvirus Humano 8/genética , Proteínas Imediatamente Precoces/genética , Sarcoma de Kaposi/metabolismo , Sarcoma de Kaposi/virologia , Sirtuínas/metabolismo , Transativadores/genética , Proteínas Virais/genética , Linhagem Celular , Linhagem Celular Tumoral , Replicação do DNA/genética , DNA Viral/genética , Regulação Viral da Expressão Gênica/genética , Células HEK293 , Humanos , Proteínas Nucleares/genética , Regiões Promotoras Genéticas/genética , RNA Interferente Pequeno/genética , Proteínas Repressoras/genética , Latência Viral/genética , Replicação Viral/genética
16.
Front Microbiol ; 10: 2879, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31921057

RESUMO

Epstein-Barr virus (EBV) nuclear antigen 1 (EBNA1)-mediated DNA episomal genome replication and persistence are essential for the viral pathogenesis. Cyclophilin A (CYPA) is upregulated in EBV-associated nasopharyngeal carcinoma (NPC) with unknown roles. In the present approach, cytosolic CYPA was found to be bound with EBNA1 into the nucleus. The amino acid 376-459 of the EBNA1 domain was important for the binding. CYPA depletion attenuated and ectopic CYPA expression improved EBNA1 expression in EBV-positive cells. The loss of viral copy number was also accelerated by CYPA consumption in daughter cells during culture passages. Mechanistically, CYPA mediated the connection of EBNA1 with oriP (origin of EBV DNA replication) and subsequent oriP transcription, which is a key step for the initiation of EBV genome replication. Moreover, CYPA overexpression markedly antagonized the connection of EBNA1 to Ubiquitin-specific protease 7 (USP7), which is a strong host barrier with a role of inhibiting EBV genome replication. The PPIase activity of CYPA was required for the promotion of oriP transcription and antagonism with USP7. The results revealed a strategy that EBV recruited a host factor to counteract the host defense, thus facilitating its own latent genome replication. This study provides a new insight into EBV pathogenesis and potential virus-targeted therapeutics in EBV-associated NPC, in which CYPA is upregulated at all stages.

17.
Virology ; 516: 55-70, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29329079

RESUMO

Epstein-Barr virus LMP1 is an oncoprotein required for immortalizing B lymphocytes and also plays important roles in transforming non-lymphoid tissue. The discovery of LMP1 protein interactions will likely generate targets to treat EBV-associated cancers. Here, we define the broader LMP1 interactome using the recently developed BioID method. Combined with mass spectrometry, we identified over 1000 proteins across seven independent experiments with direct or indirect relationships to LMP1. Pathway analysis suggests that a significant number of the proteins identified are involved in signal transduction and protein or vesicle trafficking. Interestingly, a large number of proteins thought to be important in the formation of exosomes and protein targeting were recognized as probable LMP1 interacting partners, including CD63, syntenin-1, ALIX, TSG101, HRS, CHMPs, and sorting nexins. Therefore, it is likely that LMP1 modifies protein trafficking and exosome biogenesis pathways. In support of this, knock-down of syntenin-1 and ALIX resulted in reduced exosomal LMP1.


Assuntos
Infecções por Vírus Epstein-Barr/metabolismo , Herpesvirus Humano 4/metabolismo , Proteínas da Matriz Viral/metabolismo , Biotina/metabolismo , Proteínas de Ligação ao Cálcio/genética , Proteínas de Ligação ao Cálcio/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Complexos Endossomais de Distribuição Requeridos para Transporte/genética , Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Infecções por Vírus Epstein-Barr/genética , Infecções por Vírus Epstein-Barr/virologia , Exossomos/metabolismo , Exossomos/virologia , Herpesvirus Humano 4/genética , Interações Hospedeiro-Patógeno , Humanos , Espectrometria de Massas , Ligação Proteica , Mapeamento de Interação de Proteínas , Transdução de Sinais , Sinteninas/genética , Sinteninas/metabolismo , Tetraspanina 30/genética , Tetraspanina 30/metabolismo , Proteínas da Matriz Viral/genética
19.
Virol Sin ; 32(5): 349-356, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-29116589

RESUMO

Herpesviruses are remarkable pathogens that have evolved multiple mechanisms to evade host immunity, ensuring their proliferation and egress. Among these mechanisms, herpesviruses utilize elaborate extracellular vesicles, including exosomes, for the intricate interplay between infected host and recipient cells. Herpesviruses incorporate genome expression products and direct cellular products into exosomal cargoes. These components alter the content and function of exosomes released from donor cells, thus affecting the downstream signalings of recipient cells. In this way, herpesviruses hijack exosomal pathways to ensure their survival and persistence, and exosomes are emerging as critical mediators for virus infection-associated intercellular communication and microenvironment alteration. In this review, the function and effects of exosomes in herpesvirus infection will be discussed, so that we will have a better understanding about the pathogenesis of herpesviruses.


Assuntos
Exossomos/metabolismo , Vesículas Extracelulares/metabolismo , Infecções por Herpesviridae/metabolismo , Animais , Exossomos/patologia , Infecções por Herpesviridae/patologia , Humanos , Transdução de Sinais/fisiologia
20.
J Virol ; 90(13): 5953-5964, 2016 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-27099309

RESUMO

UNLABELLED: Kaposi's sarcoma-associated herpesvirus (KSHV) is the causative agent of three human malignancies. KSHV ORF36 encodes a serine/threonine viral protein kinase, which is conserved throughout all herpesviruses. Although several studies have identified the viral and cellular substrates of conserved herpesvirus protein kinases (CHPKs), the precise functions of KSHV ORF36 during lytic replication remain elusive. Here, we report that ORF36 interacts with another lytic protein, ORF45, in a manner dependent on ORF36 kinase activity. We mapped the regions of ORF36 and ORF45 involved in the binding. Their association appears to be mediated by electrostatic interactions, since deletion of either the highly basic N terminus of ORF36 or an acidic patch of ORF45 abolished the binding. In addition, the dephosphorylation of ORF45 protein dramatically reduced its association with ORF36. Importantly, ORF45 enhances both the stability and kinase activity of ORF36. Consistent with previous studies of CHPK homologs, we detected ORF36 protein in extracellular virions. To investigate the roles of ORF36 in the context of KSHV lytic replication, we used bacterial artificial chromosome mutagenesis to engineer both ORF36-null and kinase-dead mutants. We found that ORF36-null/mutant virions are moderately defective in viral particle production and are further deficient in primary infection. In summary, our results uncover a functionally important interaction between ORF36 and ORF45 and indicate a significant role of ORF36 in the production of infectious progeny virions. IMPORTANCE: Kaposi's sarcoma-associated herpesvirus (KSHV) is a human tumor virus with a significant public health burden. KSHV ORF36 encodes a serine/threonine viral protein kinase, whose functions throughout the viral life cycle have not been elucidated. Here, we report that ORF36 interacts with another KSHV protein, ORF45. We mapped the regions of ORF36 and ORF45 involved in their association and further characterized the consequences of this interaction. We engineered ORF36 mutant viruses in order to investigate the functional roles of ORF36 in the context of KSHV lytic replication, and we confirmed that ORF36 is a component of KSHV virions. Moreover, we found that ORF36 mutants are defective in virion production and primary infection. In summary, we discovered and characterized a functionally important interaction between KSHV ORF36 and ORF45, and our results suggest a significant role of ORF36 in the production of infectious progeny virions, a process critical for KSHV pathogenesis.


Assuntos
Herpesvirus Humano 8/fisiologia , Proteínas Imediatamente Precoces/metabolismo , Proteínas Quinases/metabolismo , Replicação Viral , Linhagem Celular , Cromossomos Artificiais Bacterianos , Estabilidade Enzimática , Edição de Genes , Regulação Viral da Expressão Gênica , Células HEK293 , Herpesvirus Humano 8/enzimologia , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/patogenicidade , Humanos , Proteínas Imediatamente Precoces/genética , Mutagênese , Mutação , Fosforilação , Proteínas Quinases/genética , Eletricidade Estática , Vírion/química , Vírion/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA