Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 100
Filtrar
1.
Curr Mol Med ; 22(1): 50-66, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-33622224

RESUMO

The proteins of coronavirus are classified as non-structural, structural, and accessory. There are 16 non-structural viral proteins besides their precursors (1a and 1ab polyproteins). The non-structural proteins are named nsp1 to nsp16, and they act as enzymes, coenzymes, and binding proteins to facilitate the replication, transcription, and translation of the virus. The structural proteins are bound to the RNA in the nucleocapsid (N- protein) or to the lipid bilayer membrane of the viral envelope. The lipid bilayer proteins include the membrane protein (M), an envelope protein (E), and spike protein (S). Besides their role as structural proteins, they are essential for the host cells' binding and invasion. The SARS-CoV-2 contains six accessory proteins which participate in the viral replication, assembly and virus-host interactions. The SARS-CoV-2 accessory proteins are orf3a, orf6, orf7a, orf7b, orf8, and orf10. The functions of the SARS-CoV-2 are not well known, while the functions of their corresponding proteins in SARS-CoV are either well known or poorly studied. Recently, the Oxford University and Astrazeneca, Pfizer and BioNTech have made SARS-CoV-2 vaccines by targeting the spike protein gene. The US Food and Drug Administration (FDA) and the health authorities of the United Kingdom have approved and started conducting vaccinations using the Pfizer and BioNTech mRNA vaccine. Also, The FDA of the USA has approved the use of two monoclonal antibodies produced by Regeneron pharmaceuticals to target the spike protein for treating COVID-19. The SARS-CoV-2 proteins can be used for the diagnosis, as drug targets and in vaccination trials for COVID-19. In future COVID-19 research, more efforts should be made to elaborate the functions and structure of the SARS-CoV- 2 proteins so as to use them as targets for COVID-19 drugs and vaccines. Special attention should be paid to extensive research on the SARS-CoV-2 nsp3, orf8, and orf10.


Assuntos
Antivirais/farmacologia , Vacinas contra COVID-19 , COVID-19/prevenção & controle , SARS-CoV-2/química , Proteínas Virais/efeitos dos fármacos , Proteínas Virais/imunologia , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/uso terapêutico , Anticorpos Antivirais/imunologia , Anticorpos Antivirais/uso terapêutico , Antígenos Virais/imunologia , COVID-19/imunologia , Desenho de Fármacos , Humanos , Imunoterapia , SARS-CoV-2/efeitos dos fármacos , SARS-CoV-2/imunologia , Glicoproteína da Espícula de Coronavírus/imunologia , Desenvolvimento de Vacinas , Proteínas não Estruturais Virais/efeitos dos fármacos , Proteínas não Estruturais Virais/imunologia , Proteínas não Estruturais Virais/fisiologia , Proteínas Virais/fisiologia , Proteínas Virais Reguladoras e Acessórias/efeitos dos fármacos , Proteínas Virais Reguladoras e Acessórias/imunologia , Proteínas Virais Reguladoras e Acessórias/fisiologia , Proteínas Estruturais Virais/efeitos dos fármacos , Proteínas Estruturais Virais/imunologia , Proteínas Estruturais Virais/fisiologia , Vacinas de mRNA , Tratamento Farmacológico da COVID-19
2.
Theranostics ; 11(17): 8362-8378, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34373747

RESUMO

Rationale: Hepatitis B x protein (HBx) is required to initiate and maintain the replication of hepatitis B virus (HBV). Protein arginine methyltransferases 5 (PRMT5) negatively regulates HBV transcription. WD repeat domain 77 protein (WDR77) greatly enhances the methyltransferase activity of PRMT5. However, the role of WDR77 in the modulation of cccDNA transcription and HBV replication is poorly understood. In this study, we investigated the mechanism by which HBx modulated HBV replication involving WDR77 in the liver. Methods: A human liver-chimeric mouse model was established. Immunohistochemistry (IHC) staining, Western blot analysis, Southern blot analysis, Northern blot analysis, immunofluorescence assays, ELISA, RT-qPCR, CoIP assays, and ChIP assays were performed in human liver-chimeric mouse model, primary human hepatocytes (PHHs), HepG2-NTCP, dHepaRG and HepG2 cell lines. Results: HBV infection and HBx expression remarkably reduced the protein levels of WDR77 in human liver-chimeric mice and HepG2-NTCP cells. WDR77 restricted cccDNA transcription and HBV replication in PHHs and HepG2-NTCP cells. Mechanically, WDR77 enhanced PRMT5-triggered symmetric dimethylation of arginine 3 on H4 (H4R3me2s) on the cccDNA minichromosome to control cccDNA transcription. HBx drove the cellular DDB1-containing E3 ubiquitin ligase to degrade WDR77 through recruiting WDR77, leading to the disability of methyltransferase activity of PRMT5. Thus, HBx promoted HBV replication by driving a positive feedback loop of HBx-DDB1/WDR77/PRMT5/H4R3me2s/cccDNA/HBV/HBx in the liver. Conclusions: HBx attenuates the WDR77-mediated HBV repression by driving DDB1-induced WDR77 degradation in the liver. Our finding provides new insights into the mechanism by which HBx enhances HBV replication in the liver.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Transativadores/metabolismo , Fatores de Transcrição/metabolismo , Proteínas Virais Reguladoras e Acessórias/metabolismo , Adulto , Idoso , Animais , Quimera , China , DNA Viral/genética , Proteínas de Ligação a DNA/fisiologia , Feminino , Células Hep G2 , Hepatite B/genética , Hepatite B/metabolismo , Hepatite B/virologia , Vírus da Hepatite B/genética , Vírus da Hepatite B/patogenicidade , Hepatócitos/metabolismo , Humanos , Fígado/metabolismo , Masculino , Camundongos , Pessoa de Meia-Idade , Proteína-Arginina N-Metiltransferases/metabolismo , Transativadores/fisiologia , Fatores de Transcrição/fisiologia , Transcrição Gênica/genética , Proteínas Virais Reguladoras e Acessórias/fisiologia , Replicação Viral
3.
Autophagy ; 17(11): 3461-3474, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-33509017

RESUMO

Macroautophagy/autophagy plays an important role in the control of viral infections and viruses have evolved multiple strategies to interfere with autophagy to avoid destruction and promote their own replication and spread. Here we report that the deubiquitinase encoded in the N-terminal domain of the Epstein-Barr virus (EBV) large tegument protein, BPLF1, regulates selective autophagy. Mass spectrometry analysis identified several vesicular traffic and autophagy related proteins as BPLF1 interactors and potential substrates, suggesting that the viral protein targets this cellular defense during productive infection. Direct binding of BPLF1 to the autophagy receptor SQSTM1/p62 (sequestosome 1) was confirmed by co-immunoprecipitation of transfected BPLF1 and by in vitro affinity isolation of bacterially expressed proteins. Expression of the catalytically active BPLF1 was associated with decreased SQSTM1/p62 ubiquitination and failure to recruit LC3 to SQSTM1/p62-positive aggregates. Selective autophagy was inhibited as illustrated by the accumulation of large protein aggregates in BPLF1-positive cells co-transfected with an aggregate-prone HTT (huntingtin)-Q109 construct, and by a slower autophagy-dependent clearance of protein aggregates upon transfection of BPLF1 in cells expressing a tetracycline-regulated HTT-Q103. The inhibition of aggregate clearance was restored by overexpression of a SQSTM1/p62[E409A,K420R] mutant that does not require ubiquitination of Lys420 for cargo loading. These findings highlight a previously unrecognized role of the viral deubiquitinase in the regulation of selective autophagy, which may promote infection and the production of infectious virus.Abbreviations: BPLF1, BamH1 fragment left open reading frame-1; EBV, Epstein-Barr virus; GFP, green fluorescent protein; HTT, huntingtin; MAP1LC3/LC3, microtubule associated protein 1 light chain 3; PB1, Phox and Bem1 domain; PE, phosphatidylethanolamine; SQSTM1/p62, sequestosome 1; UBA, ubiquitin-associated domain.


Assuntos
Autofagia/fisiologia , Enzimas Desubiquitinantes/fisiologia , Herpesvirus Humano 4/fisiologia , Proteína Sequestossoma-1/fisiologia , Proteínas Virais Reguladoras e Acessórias/fisiologia , Autofagia/genética , Enzimas Desubiquitinantes/genética , Infecções por Vírus Epstein-Barr/patologia , Infecções por Vírus Epstein-Barr/virologia , Células HeLa , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/patogenicidade , Interações entre Hospedeiro e Microrganismos/genética , Interações entre Hospedeiro e Microrganismos/fisiologia , Humanos , Proteína Huntingtina/genética , Proteína Huntingtina/metabolismo , Macroautofagia/genética , Macroautofagia/fisiologia , Proteínas Associadas aos Microtúbulos/metabolismo , Mutação , Agregados Proteicos/genética , Agregados Proteicos/fisiologia , Proteína Sequestossoma-1/genética , Transfecção , Ubiquitinação , Proteínas Virais Reguladoras e Acessórias/genética
4.
Cancer Lett ; 492: 147-161, 2020 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-32827601

RESUMO

Liver cancer (hepatocellular carcinoma, HCC) is one of the most prevalent cancers worldwide. Several etiological factors of HCC, including hepatitis B or hepatitis C virus infection, liver cirrhosis and aflatoxin B1 intake has been identified. HBx, which is an oncogenic protein encoded by the hepatitis B virus, is strongly associated with hepatocarcinogenesis. Using stable HBx-expressing cell, we showed that HBx induced chromosome gain, with amplification of centrosomes numbers and deregulation of centrosome ultrastructure. To dissect the mechanism for chromosome instability, our result revealed that HBx contributed to a hyperactive centrosome-microtubule dynamics by accelerating microtubule nucleation and polymerization. Further investigations suggested that HBx interacted with a centrosome linker protein TAX1BP2, which has previously been shown to function as an intrinsic block of centrosome amplification and a tumour suppressor in HCC. Restoring TAX1BP2 was able to block HBx-mediated centrosome amplification and abolish the HBx-mediated centrosome aberration, thereby suppressing chromosome instability. Thus, we demonstrate here a mechanism by which HBx deregulates centrosome-microtubule dynamics through interacting with TAX1BP2, which underlines the possibility of restoration of TAX1BP2 to rescue cells from chromosome instability.


Assuntos
Carcinoma Hepatocelular/etiologia , Centrossomo/fisiologia , Instabilidade Cromossômica , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Neoplasias Hepáticas/etiologia , Proteínas de Membrana/fisiologia , Microtúbulos/fisiologia , Transativadores/fisiologia , Proteínas Supressoras de Tumor/fisiologia , Proteínas Virais Reguladoras e Acessórias/fisiologia , Adulto , Aneuploidia , Células Hep G2 , Humanos , Masculino
5.
Viruses ; 12(7)2020 07 10.
Artigo em Inglês | MEDLINE | ID: mdl-32664401

RESUMO

Currently, the treatment of hepatitis B virus (HBV)-associated hepatocellular carcinoma (HCC) [HBV-HCC] relies on blunt tools that are unable to offer effective therapy for later stage pathogenesis. The potential of miRNA to treat HBV-HCC offer a more targeted approach to managing this lethal carcinoma; however, the complexity of miRNA as an ancillary regulator of the immune system remains poorly understood. This review examines the overlapping roles of HBx-dysregulated miRNA in HBV-HCC and immune pathways and seeks to demonstrate that specific miRNA response in immune cells is not independent of their expression in hepatocytes. This interplay between the two pathways may provide us with the possibility of using candidate miRNA to manipulate this interaction as a potential therapeutic option.


Assuntos
Carcinoma Hepatocelular/virologia , Vírus da Hepatite B/metabolismo , Hepatite B/complicações , Neoplasias Hepáticas/virologia , MicroRNAs/metabolismo , Transativadores/metabolismo , Proteínas Virais Reguladoras e Acessórias/metabolismo , Carcinoma Hepatocelular/imunologia , Carcinoma Hepatocelular/metabolismo , Hepatite B/imunologia , Hepatite B/metabolismo , Hepatite B/virologia , Vírus da Hepatite B/imunologia , Humanos , Neoplasias Hepáticas/metabolismo , Transativadores/imunologia , Transativadores/fisiologia , Proteínas Virais Reguladoras e Acessórias/imunologia , Proteínas Virais Reguladoras e Acessórias/fisiologia
6.
Gut ; 69(11): 2016-2024, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32114505

RESUMO

OBJECTIVE: The HBV HBx regulatory protein is required for transcription from the covalently closed circular DNA (cccDNA) minichromosome and affects the epigenetic control of both viral and host cellular chromatin. DESIGN: We explored, in relevant cellular models of HBV replication, the functional consequences of HBx interaction with DLEU2, a long non-coding RNA (lncRNA) expressed in the liver and increased in human hepatocellular carcinoma (HCC), in the regulation of host target genes and the HBV cccDNA. RESULTS: We show that HBx binds the promoter region, enhances the transcription and induces the accumulation of DLEU2 in infected hepatocytes. We found that nuclear DLEU2 directly binds HBx and the histone methyltransferase enhancer of zeste homolog 2 (EZH2), the catalytic active subunit of the polycomb repressor complex 2 (PRC2) complex. Computational modelling and biochemical evidence suggest that HBx and EZH2 share two preferential binding sites in DLEU2 intron 1. HBx and DLEU2 co-recruitment on the cccDNA displaces EZH2 from the viral chromatin to boost transcription and viral replication. DLEU2-HBx association with target host promoters relieves EZH2 repression and leads to the transcriptional activation of a subset of EZH2/PRC2 target genes in HBV-infected cells and HBV-related HCCs. CONCLUSIONS: Our results highlight the ability of HBx to bind RNA to impact on the epigenetic control of both viral cccDNA and host genes and provide a new key to understand the role of DLEU2 and EZH2 overexpression in HBV-related HCCs and HBx contribution to hepatocytes transformation.


Assuntos
Carcinoma Hepatocelular/etiologia , Vírus da Hepatite B/fisiologia , Hepatócitos/patologia , Neoplasias Hepáticas/etiologia , Transativadores/fisiologia , Proteínas Virais Reguladoras e Acessórias/fisiologia , Replicação Viral/fisiologia , Técnicas de Cultura de Células , DNA Circular , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Hepatócitos/metabolismo , Humanos , RNA Longo não Codificante/metabolismo
7.
Cell Microbiol ; 22(3): e13148, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31829498

RESUMO

Hepatitis B virus (HBV) infection is a major cause of acute and chronic liver diseases. During the HBV life cycle, HBV hijacks various host factors to assist viral replication. In this research, we find that the HBV regulatory protein X (HBx) can induce the upregulation of DExH-box RNA helicase 9 (DHX9) expression by repressing proteasome-dependent degradation mediated by MDM2. Furthermore, we demonstrate that DHX9 contributes to viral DNA replication in dependence on its helicase activity and nuclear localization. In addition, the promotion of viral DNA replication by DHX9 is dependent on its interaction with Nup98. Our findings reveal that HBx-mediated DHX9 upregulation is essential for HBV DNA replication.


Assuntos
RNA Helicases DEAD-box/metabolismo , Vírus da Hepatite B/fisiologia , Hepatite B/metabolismo , Proteínas de Neoplasias/metabolismo , Complexo de Proteínas Formadoras de Poros Nucleares/metabolismo , Transativadores/fisiologia , Proteínas Virais Reguladoras e Acessórias/fisiologia , Animais , Linhagem Celular , Núcleo Celular/metabolismo , RNA Helicases DEAD-box/genética , Replicação do DNA , DNA Viral , Regulação da Expressão Gênica , Células HEK293 , Células Hep G2 , Hepatite B/genética , Hepatite B/virologia , Interações entre Hospedeiro e Microrganismos , Humanos , Camundongos , Camundongos Transgênicos , Proteínas de Neoplasias/genética , Regulação para Cima , Replicação Viral
8.
J Virol ; 93(11)2019 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-30867316

RESUMO

HIV-1 infection of macrophages leads to the sequestration of newly formed viruses in intracellular plasma membrane-connected structures termed virus-containing compartments (VCCs), where virions remain infectious and hidden from immune surveillance. The cellular restriction factor bone marrow stromal cell antigen 2 (BST2), which prevents HIV-1 dissemination by tethering budding viral particles at the plasma membrane, can be found in VCCs. The HIV-1 accessory protein Vpu counteracts the restriction factor BST2 by downregulating its expression and removing it from viral budding sites. Numerous studies described these Vpu countermeasures in CD4+ T cells or model cell lines, but the interplay between Vpu and BST2 in VCC formation and HIV-1 production in macrophages is less explored. Here, we show that Vpu expression in HIV-1-infected macrophages enhances viral release. This effect is related to Vpu's ability to circumvent BST2 antiviral activity. We show that in absence of Vpu, BST2 is enriched in VCCs and colocalizes with capsid p24, whereas Vpu expression significantly reduces the presence of BST2 in these compartments. Furthermore, our data reveal that BST2 is dispensable for the formation of VCCs and that Vpu expression impacts the volume of these compartments. This Vpu activity partly depends on BST2 expression and requires the integrity of the Vpu transmembrane domain, the dileucine-like motif E59XXXLV64 and phosphoserines 52 and 56 of Vpu. Altogether, these results highlight that Vpu controls the volume of VCCs and promotes HIV-1 release from infected macrophages.IMPORTANCE HIV-1 infection of macrophages leads to the sequestration of newly formed viruses in virus-containing compartments (VCCs), where virions remain infectious and hidden from immune surveillance. The restriction factor BST2, which prevents HIV-1 dissemination by tethering budding viral particles, can be found in VCCs. The HIV-1 Vpu protein counteracts BST2. This study explores the interplay between Vpu and BST2 in the viral protein functions on HIV-1 release and viral particle sequestration in VCCs in macrophages. The results show that Vpu controls the volume of VCCs and favors viral particle release. These Vpu functions partly depend on Vpu's ability to antagonize BST2. This study highlights that the transmembrane domain of Vpu and two motifs of the Vpu cytoplasmic domain are required for these functions. These motifs were notably involved in the control of the volume of VCCs by Vpu but were dispensable for the prevention of the specific accumulation of BST2 in these structures.


Assuntos
Membrana Celular/metabolismo , Proteínas do Vírus da Imunodeficiência Humana/metabolismo , Macrófagos/metabolismo , Proteínas Virais Reguladoras e Acessórias/metabolismo , Antígenos CD/metabolismo , Antígeno 2 do Estroma da Médula Óssea/metabolismo , Citoplasma/metabolismo , Proteínas Ligadas por GPI/genética , Proteínas Ligadas por GPI/metabolismo , Regulação Viral da Expressão Gênica/genética , Células HEK293 , Proteína do Núcleo p24 do HIV/metabolismo , Infecções por HIV/metabolismo , Infecções por HIV/virologia , Soropositividade para HIV , HIV-1/imunologia , HIV-1/metabolismo , HIV-1/patogenicidade , Células HeLa , Proteínas do Vírus da Imunodeficiência Humana/fisiologia , Humanos , Macrófagos/virologia , Proteínas Virais Reguladoras e Acessórias/fisiologia , Vírion/metabolismo , Montagem de Vírus/fisiologia , Liberação de Vírus/fisiologia
9.
PLoS Pathog ; 15(2): e1007596, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30785952

RESUMO

Nuclear RNAs are subject to a number of RNA decay pathways that serve quality control and regulatory functions. As a result, any virus that expresses its genes in the nucleus must have evolved mechanisms that avoid these pathways, but the how viruses evade nuclear RNA decay remains largely unknown. The multifunctional Kaposi's sarcoma-associated herpesvirus (KSHV) ORF57 (Mta) protein is required for the nuclear stability of viral transcripts. In the absence of ORF57, we show that viral transcripts are subject to degradation by two specific nuclear RNA decay pathways, PABPN1 and PAPα/γ-mediated RNA decay (PPD) in which decay factors are recruited through poly(A) tails, and an ARS2-mediated RNA decay pathway dependent on the 5' RNA cap. In transcription pulse chase assays, ORF57 appears to act primarily by inhibiting the ARS2-mediated RNA decay pathway. In the context of viral infection in cultured cells, inactivation of both decay pathways by RNAi is necessary for the restoration of ORF57-dependent viral genes produced from an ORF57-null bacmid. Mechanistically, we demonstrate that ORF57 protects viral transcripts by preventing the recruitment of the exosome co-factor hMTR4. In addition, our data suggest that ORF57 recruitment of ALYREF inhibits hMTR4 association with some viral RNAs, whereas other KSHV transcripts are stabilized by ORF57 in an ALYREF-independent fashion. In conclusion, our studies show that KSHV RNAs are subject to nuclear degradation by two specific host pathways, PPD and ARS2-mediated decay, and ORF57 protects viral transcripts from decay by inhibiting hMTR4 recruitment.


Assuntos
RNA Helicases/metabolismo , Estabilidade de RNA/fisiologia , Proteínas Virais Reguladoras e Acessórias/metabolismo , Linhagem Celular , Núcleo Celular , Proteínas Ativadoras de GTPase/metabolismo , Proteínas Ativadoras de GTPase/fisiologia , Regulação Viral da Expressão Gênica/genética , Genes Virais/genética , Células HEK293 , Herpesvirus Humano 8/metabolismo , Herpesvirus Humano 8/patogenicidade , Humanos , Proteínas Nucleares , Proteína I de Ligação a Poli(A)/metabolismo , Proteína I de Ligação a Poli(A)/fisiologia , Ligação Proteica , RNA Helicases/fisiologia , Estabilidade de RNA/genética , RNA Nuclear/fisiologia , RNA Viral , Proteínas de Ligação a RNA , Fatores de Transcrição , Proteínas Virais/genética , Proteínas Virais/metabolismo , Proteínas Virais Reguladoras e Acessórias/fisiologia , Replicação Viral
10.
PLoS Pathog ; 14(8): e1007232, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-30096191

RESUMO

Kaposi's sarcoma-associated herpesvirus (KSHV) is a γ-herpesvirus closely associated with Kaposi's sarcoma, primary effusion lymphoma and multicentric Castleman disease. Open reading frame 57 (ORF57), a viral early protein of KSHV promotes splicing, stability and translation of viral mRNA and is essential for viral lytic replication. Previous studies demonstrated that dimerization of ORF57 stabilizes the protein, which is critical for its function. However, the detailed structural basis of dimerization was not elucidated. In this study, we report the crystal structures of the C-terminal domain (CTD) of ORF57 (ORF57-CTD) in both dimer at 3.5 Å and monomer at 3.0 Å. Both structures reveal that ORF57-CTD binds a single zinc ion through the consensus zinc-binding motif at the bottom of each monomer. In addition, the N-terminal residues 167-222 of ORF57-CTD protrudes a long "arm" and holds the globular domains of the neighboring monomer, while the C-terminal residues 445-454 are locked into the globular domain in cis and the globular domains interact in trans. In vitro crosslinking and nuclear translocation assays showed that either deletion of the "arm" region or substitution of key residues at the globular interface led to severe dimer dissociation. Introduction of point mutation into the zinc-binding motif also led to sharp degradation of KSHV ORF57 and other herpesvirus homologues. These data indicate that the "arm" region, the residues at the globular interface and the zinc-binding motif are all equally important in ORF57 protein dimerization and stability. Consistently, KSHV recombinant virus with the disrupted zinc-binding motif by point mutation exhibited a significant reduction in the RNA level of ORF57 downstream genes ORF59 and K8.1 and infectious virus production. Taken together, this study illustrates the first structure of KSHV ORF57-CTD and provides new insights into the understanding of ORF57 protein dimerization and stability, which would shed light on the potential design of novel therapeutics against KSHV infection and related diseases.


Assuntos
Multimerização Proteica , Proteínas Virais Reguladoras e Acessórias/química , Proteínas Virais Reguladoras e Acessórias/metabolismo , Domínio Catalítico , Cristalografia por Raios X , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/metabolismo , Humanos , Modelos Moleculares , Simulação de Acoplamento Molecular , Fases de Leitura Aberta , Multimerização Proteica/genética , Estabilidade Proteica , Estrutura Quaternária de Proteína , Proteínas Virais Reguladoras e Acessórias/genética , Proteínas Virais Reguladoras e Acessórias/fisiologia
11.
PLoS Pathog ; 14(1): e1006852, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29357390

RESUMO

The N-terminal domains of the herpesvirus large tegument proteins encode a conserved cysteine protease with ubiquitin- and NEDD8-specific deconjugase activity. The proteins are expressed during the productive virus cycle and are incorporated into infectious virus particles, being delivered to the target cells upon primary infection. Members of this viral enzyme family were shown to regulate different aspects of the virus life cycle and the innate anti-viral response. However, only few substrates have been identified and the mechanisms of these effects remain largely unknown. In order to gain insights on the substrates and signaling pathways targeted by the viral enzymes, we have used co-immunoprecipitation and mass spectrometry to identify cellular proteins that interact with the Epstein-Barr virus encoded homologue BPLF1. Several members of the 14-3-3-family of scaffold proteins were found amongst the top hits of the BPLF1 interactome, suggesting that, through this interaction, BPLF1 may regulate a variety of cellular signaling pathways. Analysis of the shared protein-interaction network revealed that BPLF1 promotes the assembly of a tri-molecular complex including, in addition to 14-3-3, the ubiquitin ligase TRIM25 that participates in the innate immune response via ubiquitination of cytosolic pattern recognition receptor, RIG-I. The involvement of BPLF1 in the regulation of this signaling pathway was confirmed by inhibition of the type-I IFN responses in cells transfected with a catalytically active BPLF1 N-terminal domain or expressing the endogenous protein upon reactivation of the productive virus cycle. We found that the active viral enzyme promotes the dimerization and autoubiquitination of TRIM25. Upon triggering of the IFN response, RIG-I is recruited to the complex but ubiquitination is severely impaired, which functionally inactivates the RIG-I signalosome. The capacity to bind to and functionally inactivate the RIG-I signalosome is shared by the homologues encoded by other human herpesviruses.


Assuntos
Proteína DEAD-box 58/metabolismo , Herpesviridae/enzimologia , Interferons/farmacologia , Fatores de Transcrição/metabolismo , Proteínas com Motivo Tripartido/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Proteínas Virais Reguladoras e Acessórias/fisiologia , Núcleo Celular/metabolismo , Células Cultivadas , Células HEK293 , Células HeLa , Humanos , Receptores Imunológicos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Ubiquitina/metabolismo , Ubiquitinação , Replicação Viral
12.
J Virol ; 90(10): 5108-5118, 2016 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-26962215

RESUMO

UNLABELLED: Dendritic cells (DCs) are major targets of filovirus infection in vivo Previous studies have shown that the filoviruses Ebola virus (EBOV) and Marburg virus (MARV) suppress DC maturation in vitro Both viruses also encode innate immune evasion functions. The EBOV VP35 (eVP35) and the MARV VP35 (mVP35) proteins each can block RIG-I-like receptor signaling and alpha/beta interferon (IFN-α/ß) production. The EBOV VP24 (eVP24) and MARV VP40 (mVP40) proteins each inhibit the production of IFN-stimulated genes (ISGs) by blocking Jak-STAT signaling; however, this occurs by different mechanisms, with eVP24 blocking nuclear import of tyrosine-phosphorylated STAT1 and mVP40 blocking Jak1 function. MARV VP24 (mVP24) has been demonstrated to modulate host cell antioxidant responses. Previous studies demonstrated that eVP35 is sufficient to strongly impair primary human monocyte-derived DC (MDDC) responses upon stimulation induced through the RIG-I-like receptor pathways. We demonstrate that mVP35, like eVP35, suppresses not only IFN-α/ß production but also proinflammatory responses after stimulation of MDDCs with RIG-I activators. In contrast, eVP24 and mVP40, despite suppressing ISG production upon RIG-I activation, failed to block upregulation of maturation markers or T cell activation. mVP24, although able to stimulate expression of antioxidant response genes, had no measurable impact of DC function. These data are consistent with a model where filoviral VP35 proteins are the major suppressors of DC maturation during filovirus infection, whereas the filoviral VP24 proteins and mVP40 are insufficient to prevent DC maturation. IMPORTANCE: The ability to suppress the function of dendritic cells (DCs) likely contributes to the pathogenesis of disease caused by the filoviruses Ebola virus and Marburg virus. To clarify the basis for this DC suppression, we assessed the effect of filovirus proteins known to antagonize innate immune signaling pathways, including Ebola virus VP35 and VP24 and Marburg virus VP35, VP40, and VP24, on DC maturation and function. The data demonstrate that the VP35s from Ebola virus and Marburg virus are the major suppressors of DC maturation and that the effects on DCs of the remaining innate immune inhibitors are minor.


Assuntos
Células Dendríticas/fisiologia , Células Dendríticas/virologia , Ebolavirus/química , Marburgvirus/química , Vírus de RNA/fisiologia , Proteínas Virais/fisiologia , Proteínas Virais Reguladoras e Acessórias/fisiologia , Diferenciação Celular , Vírus da Encefalomiocardite/fisiologia , Interações Hospedeiro-Patógeno , Humanos , Interferon-alfa/genética , Interferon-alfa/imunologia , Interferon beta/genética , Interferon beta/imunologia , Interferon gama/metabolismo , Proteínas do Nucleocapsídeo , Nucleoproteínas/genética , Nucleoproteínas/fisiologia , Vírus Sendai/fisiologia , Transdução Genética , Proteínas do Core Viral/genética , Proteínas do Core Viral/fisiologia , Proteínas Virais/genética
13.
J Immunol Res ; 2015: 498054, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26557721

RESUMO

The human T-cell lymphotropic virus type 1 (HTLV-1) is a retrovirus associated with both proliferative and inflammatory disorders. This virus causes a persistent infection, mainly in CD4+ T lymphocyte. The ability to persist in the host is associated with the virus capacity to evade the immune response and to induce infected T-cell proliferation, once the HTLV-1 maintains the infection mainly by clonal expansion of infected cells. There are several evidences that ORF-I encoded proteins, such as p12 and p8, play an important role in this context. The present study will review the molecular mechanisms that HTLV-1 ORF-I encoded proteins have to induce dysregulation of intracellular signaling, in order to escape from immune response and to increase the infected T-cell proliferation rate. The work will also address the impact of ORF-I mutations on the human host and perspectives in this study field.


Assuntos
Vírus Linfotrópico T Tipo 1 Humano/fisiologia , Evasão da Resposta Imune , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Transdução de Sinais , Linfócitos T/imunologia , Linfócitos T/virologia , Proteínas Virais Reguladoras e Acessórias/fisiologia , Animais , Linfócitos T CD4-Positivos/imunologia , Proliferação de Células , Interações Hospedeiro-Patógeno , Vírus Linfotrópico T Tipo 1 Humano/genética , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Células Matadoras Naturais/imunologia , Ativação Linfocitária , Mutação , Linfócitos T Citotóxicos/imunologia , Proteínas Virais Reguladoras e Acessórias/genética
14.
Uirusu ; 65(1): 135-44, 2015.
Artigo em Japonês | MEDLINE | ID: mdl-26923968

RESUMO

Viroporins are small and hydrophobic viral proteins that form pores on host cell membranes, and their expression can increase the permeability of cellular membranes and the production of progeny virus particles. JC virus (JCV) is the causative agent of progressive multifocal leukoenchephalopathy (PML). We demonstrate that JCV Agno, which is the small and hydrophobic protein, andincreases the plasma membrane permeability and virion release, acts as a viroporin. We also demonstrate that an interaction of Agno with a host cellular protein regulates the viroporin activity of Agno. These findings indicate a new paradigm in virus-host interactions regulating viroporin activity and viral replication.


Assuntos
Permeabilidade da Membrana Celular , Vírus JC , Proteínas Virais Reguladoras e Acessórias/fisiologia , Interações Hospedeiro-Patógeno/fisiologia , Humanos , Vírus JC/genética , Vírus JC/patogenicidade , Vírus JC/fisiologia , Leucoencefalopatia Multifocal Progressiva/virologia , Lipídeos de Membrana/metabolismo , Proteínas Virais Reguladoras e Acessórias/química , Vírion/metabolismo , Replicação Viral
15.
Yakugaku Zasshi ; 133(10): 1103-11, 2013.
Artigo em Japonês | MEDLINE | ID: mdl-24088354

RESUMO

Human immunodeficiency virus (HIV) has no more than nine genes expressing approximately twenty proteins. When T lymphocytes and macrophages in a body are infected with HIV, these proteins work in turn at specific time and location, causing acquired immunodeficiency syndrome (AIDS), a disease yet to be overcome. Since the elucidation of molecular mechanism of HIV proteins should lead to remedy of AIDS, the author has been engaged in the study of HIV protein in the past decade. Described herein are viral protein X (Vpx), uniquely found in HIV-2, and its homologous protein Vpr found both in HIV-1 and -2. We found that Vpx enhances genome nuclear import in T lymphocytes, and is critical for reverse transcription of viral RNA in macrophages. This finding on the function in macrophages corrected long-term misleading belief. Furthermore, functional region mapping of Vpx was performed. In 2011, the protein SAMHD1 was identified as the host restriction factor counteracted by Vpx, by foreign researchers. After that, our independent study demonstrated the presence of SAMHD1-independent functions of Vpx in T cells, in addition to its SAMHD1-dependent functions in macrophages. Another topic of this review is Gag protein. Recently, it has reported by overseas researchers that PI(4,5)P2 (one of phosphoinositide) regulates Pr55(Gag) localization and assembly. In this study, we determined the binding affinity between N-terminal MA domain of Pr55(Gag) and various phosphoinositide derivatives using surface plasmon resonance. The results suggested that both negatively charged inositol phosphates and hydrophobic acyl chain are required for the MA binding.


Assuntos
HIV-1/genética , HIV-2/genética , Fosfatidilinositóis/fisiologia , Precursores de Proteínas/metabolismo , Transporte Proteico , Proteínas Virais Reguladoras e Acessórias , Produtos do Gene vpr do Vírus da Imunodeficiência Humana , Síndrome da Imunodeficiência Adquirida/virologia , Transporte Ativo do Núcleo Celular , Descoberta de Drogas , Genoma Viral , Macrófagos/virologia , Terapia de Alvo Molecular , Proteínas Monoméricas de Ligação ao GTP/fisiologia , Fosfatidilinositóis/química , Fosfatidilinositóis/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , RNA Viral/genética , Transcrição Reversa , Proteína 1 com Domínio SAM e Domínio HD , Ressonância de Plasmônio de Superfície , Linfócitos T/virologia , Proteínas Virais Reguladoras e Acessórias/genética , Proteínas Virais Reguladoras e Acessórias/fisiologia , Produtos do Gene vpr do Vírus da Imunodeficiência Humana/genética , Produtos do Gene vpr do Vírus da Imunodeficiência Humana/fisiologia
16.
J Biol Chem ; 288(28): 20683-91, 2013 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-23744077

RESUMO

Newly identified anti-HIV host factor, SAMHD1, restricts replication of lentiviruses such as HIV-1, HIV-2, and simian immunodeficiency virus in macrophages by enzymatically hydrolyzing and depleting cellular dNTPs, which are the substrates of viral DNA polymerases. HIV-2 and some simian immunodeficiency viruses express viral protein X (VPX), which counteracts SAMHD1 and elevates cellular dNTPs, enhancing viral replication in macrophages. Because nucleoside reverse transcriptase inhibitors (NRTIs), the most commonly used anti-HIV drugs, compete against cellular dNTPs for incorporation into proviral DNA, we tested whether SAMHD1 directly affects the efficacy of NRTIs in inhibiting HIV-1. We found that reduction of SAMHD1 levels with the use of virus-like particles expressing Vpx- and SAMHD1-specific shRNA subsequently elevates cellular dNTPs and significantly decreases HIV-1 sensitivity to various NRTIs in macrophages. However, virus-like particles +Vpx treatment of activated CD4(+) T cells only minimally reduced NRTI efficacy. Furthermore, with the use of HPLC, we could not detect SAMHD1-mediated hydrolysis of NRTI-triphosphates, verifying that the reduced sensitivity of HIV-1 to NRTIs upon SAMHD1 degradation is most likely caused by the elevation in cellular dNTPs.


Assuntos
Desoxirribonucleosídeos/metabolismo , HIV-1/efeitos dos fármacos , Proteínas Monoméricas de Ligação ao GTP/metabolismo , Inibidores da Transcriptase Reversa/farmacologia , Western Blotting , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD4-Positivos/virologia , Linhagem Celular , Células Cultivadas , Relação Dose-Resposta a Droga , HIV-1/genética , HIV-1/fisiologia , Interações Hospedeiro-Patógeno , Humanos , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Macrófagos/virologia , Proteínas Monoméricas de Ligação ao GTP/genética , Nevirapina/farmacologia , Interferência de RNA , Proteína 1 com Domínio SAM e Domínio HD , Proteínas Virais Reguladoras e Acessórias/genética , Proteínas Virais Reguladoras e Acessórias/fisiologia , Vírion/efeitos dos fármacos , Vírion/genética , Vírion/fisiologia , Replicação Viral/efeitos dos fármacos , Zidovudina/farmacologia
17.
Biochemistry ; 52(22): 3939-48, 2013 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-23651212

RESUMO

Nonenveloped viruses are generally released from the cell by the timely lysis of host cell membranes. SV40 has been used as a model virus for the study of the lytic nonenveloped virus life cycle. The expression of SV40 VP4 at later times during infection is concomitant with cell lysis. To investigate the role of VP4 in viral release and its mechanism of action, VP4 was expressed and purified from bacteria as a fusion protein for use in membrane disruption assays. Purified VP4 perforated membranes as demonstrated by the release of fluorescent markers encapsulated within large unilamellar vesicles or liposomes. Dynamic light scattering results revealed that VP4 treatment did not cause membrane lysis or change the size of the liposomes. Liposomes encapsulated with 4,4-difluoro-5,7-dimethyl-4-bora-3a,4a-diaza-3-indacene-labeled streptavidin were used to show that VP4 formed stable pores in membranes. These VP4 pores had an inner diameter of 1-5 nm. Asymmetrical liposomes containing pyrene-labeled lipids in the outer monolayer were employed to monitor transbilayer lipid diffusion. Consistent with VP4 forming toroidal pore structures in membranes, VP4 induced transbilayer lipid diffusion or lipid flip-flop. Altogether, these studies support a central role for VP4 acting as a viroporin in the disruption of cellular membranes to trigger SV40 viral release by forming toroidal pores that unite the outer and inner leaflets of membrane bilayers.


Assuntos
Membranas/efeitos dos fármacos , Vírus 40 dos Símios/metabolismo , Proteínas Virais Reguladoras e Acessórias/química , Proteínas Virais Reguladoras e Acessórias/fisiologia , Liberação de Vírus/fisiologia , Membrana Celular , Bicamadas Lipídicas/química , Lipossomos/química , Membranas/metabolismo
18.
J Virol ; 87(14): 7940-51, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23658451

RESUMO

Feline immunodeficiency virus (FIV) is a lentivirus that causes AIDS in domestic cats, similar to human immunodeficiency virus (HIV)/AIDS in humans. The FIV accessory protein Vif abrogates the inhibition of infection by cat APOBEC3 restriction factors. FIV also encodes a multifunctional OrfA accessory protein that has characteristics similar to HIV Tat, Vpu, Vpr, and Nef. To examine the role of vif and orfA accessory genes in FIV replication and pathogenicity, we generated chimeras between two FIV molecular clones with divergent disease potentials: a highly pathogenic isolate that replicates rapidly in vitro and is associated with significant immunopathology in vivo, FIV-C36 (referred to here as high-virulence FIV [HV-FIV]), and a less-pathogenic strain, FIV-PPR (referred to here as low-virulence FIV [LV-FIV]). Using PCR-driven overlap extension, we produced viruses in which vif, orfA, or both genes from virulent HV-FIV replaced equivalent genes in LV-FIV. The generation of these chimeras is more straightforward in FIV than in primate lentiviruses, since FIV accessory gene open reading frames have very little overlap with other genes. All three chimeric viruses exhibited increased replication kinetics in vitro compared to the replication kinetics of LV-FIV. Chimeras containing HV-Vif or Vif/OrfA had replication rates equivalent to those of the virulent HV-FIV parental virus. Furthermore, small interfering RNA knockdown of feline APOBEC3 genes resulted in equalization of replication rates between LV-FIV and LV-FIV encoding HV-FIV Vif. These findings demonstrate that Vif-APOBEC interactions play a key role in controlling the replication and pathogenicity of this immunodeficiency-inducing virus in its native host species and that accessory genes act as mediators of lentiviral strain-specific virulence.


Assuntos
Gatos/virologia , Citosina Desaminase/metabolismo , Produtos do Gene vif/metabolismo , Vírus da Imunodeficiência Felina/patogenicidade , Proteínas Virais Reguladoras e Acessórias/metabolismo , Replicação Viral/fisiologia , Análise de Variância , Animais , Linhagem Celular , Quimera/virologia , Primers do DNA/genética , Produtos do Gene vif/fisiologia , Células HEK293 , Humanos , Vírus da Imunodeficiência Felina/fisiologia , Reação em Cadeia da Polimerase , Interferência de RNA , Receptores OX40/metabolismo , Especificidade da Espécie , Proteínas Virais Reguladoras e Acessórias/fisiologia , Virulência
19.
Biochem J ; 452(2): 359-65, 2013 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-23557442

RESUMO

Activation of PKR (double-stranded-RNA-dependent protein kinase) by DNA plasmids decreases translation, and limits the amount of recombinant protein produced by transiently transfected HEK (human embryonic kidney)-293 cells. Co-expression with Ebola virus VP35 (virus protein 35), which blocked plasmid activation of PKR, substantially increased production of recombinant TPL-2 (tumour progression locus 2)-ABIN-2 [A20-binding inhibitor of NF-κB (nuclear factor κB) 2]-NF-κB1 p105 complex. VP35 also increased expression of other co-transfected proteins, suggesting that VP35 could be employed generally to boost recombinant protein production by HEK-293 cells.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/biossíntese , Ebolavirus/fisiologia , MAP Quinase Quinase Quinases/biossíntese , Subunidade p50 de NF-kappa B/biossíntese , Proteínas Proto-Oncogênicas/biossíntese , Regulação para Cima/genética , Proteínas Virais Reguladoras e Acessórias/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Ebolavirus/genética , Células HEK293 , Humanos , MAP Quinase Quinase Quinases/genética , Complexos Multiproteicos/biossíntese , Complexos Multiproteicos/genética , Subunidade p50 de NF-kappa B/genética , Proteínas Proto-Oncogênicas/genética , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética , Transfecção , Proteínas Virais Reguladoras e Acessórias/genética
20.
Uirusu ; 63(2): 165-74, 2013.
Artigo em Japonês | MEDLINE | ID: mdl-25366051

RESUMO

Human T-cell leukemia virus type 1 (HTLV-1) belongs to Delta Retorviridae, and induces a malignancy of CD4+CD25+ T-cells, adult T-cell leukemia (ATL), and several chronic inflammatory diseases, such as HTLV-1 associated myelopathy/tropical spastic paraparesis (HAM/TSP) and HTLV-1 uveitis. A nationwide survey of HTLV-1-infected subjects, which was recently conducted by Japanese government, revealed that the numbers of HTLV-1 carriers and patients with HTLV-1-associated diseases have not decreased much over the last two decades in Japan. In contrast, novel findings on HTLV-1 dynamics in vivo and molecular mechanisms of its pathogenesis are accumulating by detailed analysis of newly identified viral and cellular factors, novel technologies such as next-generation sequencing, and appropriate animal models for HTLV-1 research. In this review, we summarize the recent progress of HTLV-1 research.


Assuntos
Vírus Linfotrópico T Tipo 1 Humano , Animais , Fatores de Transcrição de Zíper de Leucina Básica/genética , Portador Sadio/epidemiologia , Genes pX/genética , Vírus Linfotrópico T Tipo 1 Humano/genética , Vírus Linfotrópico T Tipo 1 Humano/patogenicidade , Humanos , Imunoterapia , Japão/epidemiologia , Leucemia-Linfoma de Células T do Adulto/epidemiologia , Leucemia-Linfoma de Células T do Adulto/genética , Leucemia-Linfoma de Células T do Adulto/imunologia , Leucemia-Linfoma de Células T do Adulto/virologia , Análise de Sequência com Séries de Oligonucleotídeos , Paraparesia Espástica Tropical/epidemiologia , Paraparesia Espástica Tropical/virologia , RNA Viral , Proteínas dos Retroviridae , Uveíte/virologia , Proteínas Virais/genética , Proteínas Virais Reguladoras e Acessórias/genética , Proteínas Virais Reguladoras e Acessórias/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA