Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 224
Filtrar
1.
Curr Mol Med ; 22(1): 50-66, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-33622224

RESUMEN

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.


Asunto(s)
Antivirales/farmacología , Vacunas contra la COVID-19 , COVID-19/prevención & control , SARS-CoV-2/química , Proteínas Virales/efectos de los fármacos , Proteínas Virales/inmunología , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Antivirales/inmunología , Anticuerpos Antivirales/uso terapéutico , Antígenos Virales/inmunología , COVID-19/inmunología , Diseño de Fármacos , Humanos , Inmunoterapia , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/inmunología , Glicoproteína de la Espiga del Coronavirus/inmunología , Desarrollo de Vacunas , Proteínas no Estructurales Virales/efectos de los fármacos , Proteínas no Estructurales Virales/inmunología , Proteínas no Estructurales Virales/fisiología , Proteínas Virales/fisiología , Proteínas Reguladoras y Accesorias Virales/efectos de los fármacos , Proteínas Reguladoras y Accesorias Virales/inmunología , Proteínas Reguladoras y Accesorias Virales/fisiología , Proteínas Estructurales Virales/efectos de los fármacos , Proteínas Estructurales Virales/inmunología , Proteínas Estructurales Virales/fisiología , Vacunas de ARNm , Tratamiento Farmacológico de COVID-19
2.
Theranostics ; 11(17): 8362-8378, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34373747

RESUMEN

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.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Transactivadores/metabolismo , Factores de Transcripción/metabolismo , Proteínas Reguladoras y Accesorias Virales/metabolismo , Adulto , Anciano , Animales , Quimera , China , ADN Viral/genética , Proteínas de Unión al ADN/fisiología , Femenino , Células Hep G2 , Hepatitis B/genética , Hepatitis B/metabolismo , Hepatitis B/virología , Virus de la Hepatitis B/genética , Virus de la Hepatitis B/patogenicidad , Hepatocitos/metabolismo , Humanos , Hígado/metabolismo , Masculino , Ratones , Persona de Mediana Edad , Proteína-Arginina N-Metiltransferasas/metabolismo , Transactivadores/fisiología , Factores de Transcripción/fisiología , Transcripción Genética/genética , Proteínas Reguladoras y Accesorias Virales/fisiología , Replicación Viral
3.
Mol Microbiol ; 116(3): 877-889, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34184328

RESUMEN

When subcloned into low-copy-number expression vectors, rumAB, encoding polVR391 (RumA'2 B), is best characterized as a potent mutator giving rise to high levels of spontaneous mutagenesis in vivo. This is in dramatic contrast to the poorly mutable phenotype when polVR391 is expressed from the native 88.5 kb R391, suggesting that R391 expresses cis-acting factors that suppress the expression and/or the activity of polVR391 . Indeed, we recently discovered that SetRR391 , an ortholog of λ cI repressor, is a transcriptional repressor of rumAB. Here, we report that CroSR391 , an ortholog of λ Cro, also serves as a potent transcriptional repressor of rumAB. Levels of RumA are dependent upon an interplay between SetRR391 and CroSR391 , with the greatest reduction of RumA protein levels observed in the absence of SetRR391 and the presence of CroSR391 . Under these conditions, CroSR391 completely abolishes the high levels of mutagenesis promoted by polVR391 expressed from low-copy-number plasmids. Furthermore, deletion of croSR391 on the native R391 results in a dramatic increase in mutagenesis, indicating that CroSR391 plays a major role in suppressing polVR391 mutagenesis in vivo. Inactivating mutations in CroSR391 therefore have the distinct possibility of increasing cellular mutagenesis that could lead to the evolution of antibiotic resistance of pathogenic bacteria harboring R391.


Asunto(s)
Bacteriófago lambda/fisiología , ADN Polimerasa Dirigida por ADN/metabolismo , Proteínas de Escherichia coli/metabolismo , Escherichia coli/metabolismo , Escherichia coli/virología , Metiltransferasas/metabolismo , Mutagénesis , Proteínas Represoras/fisiología , Proteínas Reguladoras y Accesorias Virales/fisiología , Regulación Bacteriana de la Expresión Génica , Genes Bacterianos , Respuesta SOS en Genética , Eliminación de Secuencia
4.
Bosn J Basic Med Sci ; 21(5): 515-527, 2021 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-33714258

RESUMEN

Coronavirus disease-19 (COVID-19) is an extremely infectious disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) that has become a major global health concern. The induction of a coordinated immune response is crucial to the elimination of any pathogenic infection. However, SARS-CoV-2 can modulate the host immune system to favor viral adaptation and persistence within the host. The virus can counteract type I interferon (IFN-I) production, attenuating IFN-I signaling pathway activation and disrupting antigen presentation. Simultaneously, SARS-CoV-2 infection can enhance apoptosis and the production of inflammatory mediators, which ultimately results in increased disease severity. SARS-CoV-2 produces an array of effector molecules, including nonstructural proteins (NSPs) and open-reading frames (ORFs) accessory proteins. We describe the complex molecular interplay of SARS-CoV-2 NSPs and accessory proteins with the host's signaling mediating immune evasion in the current review. In addition, the crucial role played by immunomodulation therapy to address immune evasion is discussed. Thus, the current review can provide new directions for the development of vaccines and specific therapies.


Asunto(s)
COVID-19/inmunología , Evasión Inmune/fisiología , Inmunidad Innata/fisiología , SARS-CoV-2/patogenicidad , Proteínas no Estructurales Virales/fisiología , Proteínas Reguladoras y Accesorias Virales/fisiología , Humanos
5.
Autophagy ; 17(11): 3461-3474, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-33509017

RESUMEN

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.


Asunto(s)
Autofagia/fisiología , Enzimas Desubicuitinizantes/fisiología , Herpesvirus Humano 4/fisiología , Proteína Sequestosoma-1/fisiología , Proteínas Reguladoras y Accesorias Virales/fisiología , Autofagia/genética , Enzimas Desubicuitinizantes/genética , Infecciones por Virus de Epstein-Barr/patología , Infecciones por Virus de Epstein-Barr/virología , Células HeLa , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/patogenicidad , Interacciones Microbiota-Huesped/genética , Interacciones Microbiota-Huesped/fisiología , Humanos , Proteína Huntingtina/genética , Proteína Huntingtina/metabolismo , Macroautofagia/genética , Macroautofagia/fisiología , Proteínas Asociadas a Microtúbulos/metabolismo , Mutación , Agregado de Proteínas/genética , Agregado de Proteínas/fisiología , Proteína Sequestosoma-1/genética , Transfección , Ubiquitinación , Proteínas Reguladoras y Accesorias Virales/genética
6.
Cancer Lett ; 492: 147-161, 2020 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-32827601

RESUMEN

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.


Asunto(s)
Carcinoma Hepatocelular/etiología , Centrosoma/fisiología , Inestabilidad Cromosómica , Péptidos y Proteínas de Señalización Intracelular/fisiología , Neoplasias Hepáticas/etiología , Proteínas de la Membrana/fisiología , Microtúbulos/fisiología , Transactivadores/fisiología , Proteínas Supresoras de Tumor/fisiología , Proteínas Reguladoras y Accesorias Virales/fisiología , Adulto , Aneuploidia , Células Hep G2 , Humanos , Masculino
7.
Proc Natl Acad Sci U S A ; 117(34): 20576-20585, 2020 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-32788352

RESUMEN

Temperate bacteriophages can enter one of two life cycles following infection of a sensitive host: the lysogenic or the lytic life cycle. The choice between the two alternative life cycles is dependent upon a tight regulation of promoters and their cognate regulatory proteins within the phage genome. We investigated the genetic switch of TP901-1, a bacteriophage of Lactococcus lactis, controlled by the CI repressor and the modulator of repression (MOR) antirepressor and their interactions with DNA. We determined the solution structure of MOR, and we solved the crystal structure of MOR in complex with the N-terminal domain of CI, revealing the structural basis of MOR inhibition of CI binding to the DNA operator sites. 15N NMR Carr-Purcell-Meiboom-Gill (CPMG) relaxation dispersion and rotating frame R1ρ measurements demonstrate that MOR displays molecular recognition dynamics on two different time scales involving a repacking of aromatic residues at the interface with CI. Mutations in the CI:MOR binding interface impair complex formation in vitro, and when introduced in vivo, the bacteriophage switch is unable to choose the lytic life cycle showing that the CI:MOR complex is essential for proper functioning of the genetic switch. On the basis of sequence alignments, we show that the structural features of the MOR:CI complex are likely conserved among a larger family of bacteriophages from human pathogens implicated in transfer of antibiotic resistance.


Asunto(s)
Bacteriófagos/fisiología , Lisogenia , Proteínas Represoras/fisiología , Proteínas Reguladoras y Accesorias Virales/fisiología , Genoma Bacteriano , Interacciones Huésped-Patógeno , Cinética , Lactococcus lactis/virología , Simulación de Dinámica Molecular , Regiones Operadoras Genéticas , Conformación Proteica , Proteínas Represoras/química , Proteínas Reguladoras y Accesorias Virales/química
8.
Viruses ; 12(7)2020 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-32664401

RESUMEN

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.


Asunto(s)
Carcinoma Hepatocelular/virología , Virus de la Hepatitis B/metabolismo , Hepatitis B/complicaciones , Neoplasias Hepáticas/virología , MicroARNs/metabolismo , Transactivadores/metabolismo , Proteínas Reguladoras y Accesorias Virales/metabolismo , Carcinoma Hepatocelular/inmunología , Carcinoma Hepatocelular/metabolismo , Hepatitis B/inmunología , Hepatitis B/metabolismo , Hepatitis B/virología , Virus de la Hepatitis B/inmunología , Humanos , Neoplasias Hepáticas/metabolismo , Transactivadores/inmunología , Transactivadores/fisiología , Proteínas Reguladoras y Accesorias Virales/inmunología , Proteínas Reguladoras y Accesorias Virales/fisiología
9.
J Virol ; 94(16)2020 07 30.
Artículo en Inglés | MEDLINE | ID: mdl-32493824

RESUMEN

Ebola virus (EBOV) inclusion bodies (IBs) are cytoplasmic sites of nucleocapsid formation and RNA replication, housing key steps in the virus life cycle that warrant further investigation. During infection, IBs display dynamic properties regarding their size and location. The contents of IBs also must transition prior to further viral maturation, assembly, and release, implying additional steps in IB function. Interestingly, the expression of the viral nucleoprotein (NP) alone is sufficient for the generation of IBs, indicating that it plays an important role in IB formation during infection. In addition to NP, other components of the nucleocapsid localize to IBs, including VP35, VP24, VP30, and the RNA polymerase L. We previously defined and solved the crystal structure of the C-terminal domain of NP (NP-Ct), but its role in virus replication remained unclear. Here, we show that NP-Ct is necessary for IB formation when NP is expressed alone. Interestingly, we find that NP-Ct is also required for the production of infectious virus-like particles (VLPs), and that defective VLPs with NP-Ct deletions are significantly reduced in viral RNA content. Furthermore, coexpression of the nucleocapsid component VP35 overcomes deletion of NP-Ct in triggering IB formation, demonstrating a functional interaction between the two proteins. Of all the EBOV proteins, only VP35 is able to overcome the defect in IB formation caused by the deletion of NP-Ct. This effect is mediated by a novel protein-protein interaction between VP35 and NP that controls both regulation of IB formation and RNA replication itself and that is mediated by a newly identified functional domain of NP, the central domain.IMPORTANCE Inclusion bodies (IBs) are cytoplasmic sites of RNA synthesis for a variety of negative-sense RNA viruses, including Ebola virus. In addition to housing important steps in the viral life cycle, IBs protect new viral RNA from innate immune attack and contain specific host proteins whose function is under study. A key viral factor in Ebola virus IB formation is the nucleoprotein, NP, which also is important in RNA encapsidation and synthesis. In this study, we have identified two domains of NP that control inclusion body formation. One of these, the central domain (CD), interacts with viral protein VP35 to control both inclusion body formation and RNA synthesis. The other is the NP C-terminal domain (NP-Ct), whose function has not previously been reported. These findings contribute to a model in which NP and its interactions with VP35 link the establishment of IBs to the synthesis of viral RNA.


Asunto(s)
Ebolavirus/metabolismo , Cuerpos de Inclusión Viral/metabolismo , Nucleoproteínas/fisiología , Línea Celular , Ebolavirus/patogenicidad , Genoma Viral/genética , Células HEK293 , Fiebre Hemorrágica Ebola/virología , Humanos , Cuerpos de Inclusión/metabolismo , Nucleocápside/metabolismo , Nucleocápside/fisiología , Proteínas de la Nucleocápside/metabolismo , Proteínas de la Nucleocápside/fisiología , Nucleoproteínas/metabolismo , ARN/biosíntesis , ARN Viral/genética , Factores de Transcripción/metabolismo , Proteínas Virales/metabolismo , Proteínas Reguladoras y Accesorias Virales/metabolismo , Proteínas Reguladoras y Accesorias Virales/fisiología , Virión/metabolismo , Replicación Viral/fisiología
11.
J Virol ; 94(13)2020 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-32321804

RESUMEN

Viral proteins pUL16 and pUL21 are required for efficient nuclear egress of herpes simplex virus 2 capsids. To better understand the role of these proteins in nuclear egress, we established whether nuclear egress complex (NEC) distribution and/or function was altered in the absence of either pUL16 or pUL21. NEC distribution in cells infected with pUL16-deficient viruses was indistinguishable from that observed in cells infected with wild-type viruses. In contrast, NEC distribution was aberrant in cells infected with pUL21-deficient virus and, instead, showed some similarity to the aberrant NEC distribution pattern observed in cells infected with pUs3-deficient virus. These results indicated that pUL16 plays a role in nuclear egress that is distinct from that of pUL21 and pUs3. Higher-resolution examination of nuclear envelope ultrastructure in cells infected with pUL21-deficient viruses by transmission electron microscopy showed different types of nuclear envelope perturbations, including some that were not observed in cells infected with pUs3 deficient virus. The formation of the nuclear envelope perturbations observed in pUL21-deficient virus infections was dependent on a functional NEC, revealing a novel role for pUL21 in regulating NEC activity. The results of comparisons of nuclear envelope ultrastructure in cells infected with viruses lacking pUs3, pUL16, or both pUs3 and pUL16 were consistent with a role for pUL16 in advance of primary capsid envelopment and shed new light on how pUs3 functions in nuclear egress.IMPORTANCE The membrane deformation activity of the herpesvirus nuclear egress complex (NEC) allows capsids to transit through both nuclear membranes into the cytoplasm. NEC activity must be precisely controlled during viral infection, and yet our knowledge of how NEC activity is controlled is incomplete. To determine how pUL16 and pUL21, two viral proteins required for nuclear egress of herpes simplex virus 2, function in nuclear egress, we examined how the lack of each protein impacted NEC distribution. These analyses revealed a function of pUL16 in nuclear egress distinct from that of pUL21, uncovered a novel role for pUL21 in regulating NEC activity, and shed new light on how a viral kinase, pUs3, regulates nuclear egress. Nuclear egress of capsids is required for all herpesviruses. A complete understanding of all aspects of nuclear egress, including how viral NEC activity is controlled, may yield strategies to disrupt this process and aid the development of herpes-specific antiviral therapies.


Asunto(s)
Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Virales/metabolismo , Proteínas Reguladoras y Accesorias Virales/metabolismo , Animales , Cápside/metabolismo , Proteínas de la Cápside/metabolismo , Núcleo Celular/virología , Chlorocebus aethiops , Fibroblastos , Células HeLa , Herpes Simple/virología , Infecciones por Herpesviridae/metabolismo , Herpesvirus Humano 1/fisiología , Herpesvirus Humano 2/fisiología , Humanos , Ratones , Membrana Nuclear/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Serina-Treonina Quinasas/fisiología , Simplexvirus/metabolismo , Simplexvirus/patogenicidad , Células Vero , Proteínas Virales/fisiología , Proteínas Reguladoras y Accesorias Virales/fisiología , Virión/metabolismo , Ensamble de Virus , Liberación del Virus/fisiología , Replicación Viral
12.
Gut ; 69(11): 2016-2024, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32114505

RESUMEN

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.


Asunto(s)
Carcinoma Hepatocelular/etiología , Virus de la Hepatitis B/fisiología , Hepatocitos/patología , Neoplasias Hepáticas/etiología , Transactivadores/fisiología , Proteínas Reguladoras y Accesorias Virales/fisiología , Replicación Viral/fisiología , Técnicas de Cultivo de Célula , ADN Circular , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Hepatocitos/metabolismo , Humanos , ARN Largo no Codificante/metabolismo
13.
Cell Microbiol ; 22(3): e13148, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31829498

RESUMEN

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.


Asunto(s)
ARN Helicasas DEAD-box/metabolismo , Virus de la Hepatitis B/fisiología , Hepatitis B/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas de Complejo Poro Nuclear/metabolismo , Transactivadores/fisiología , Proteínas Reguladoras y Accesorias Virales/fisiología , Animales , Línea Celular , Núcleo Celular/metabolismo , ARN Helicasas DEAD-box/genética , Replicación del ADN , ADN Viral , Regulación de la Expresión Génica , Células HEK293 , Células Hep G2 , Hepatitis B/genética , Hepatitis B/virología , Interacciones Microbiota-Huesped , Humanos , Ratones , Ratones Transgénicos , Proteínas de Neoplasias/genética , Regulación hacia Arriba , Replicación Viral
14.
J Virol ; 93(11)2019 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-30867316

RESUMEN

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.


Asunto(s)
Membrana Celular/metabolismo , Proteínas del Virus de la Inmunodeficiencia Humana/metabolismo , Macrófagos/metabolismo , Proteínas Reguladoras y Accesorias Virales/metabolismo , Antígenos CD/metabolismo , Antígeno 2 del Estroma de la Médula Ósea/metabolismo , Citoplasma/metabolismo , Proteínas Ligadas a GPI/genética , Proteínas Ligadas a GPI/metabolismo , Regulación Viral de la Expresión Génica/genética , Células HEK293 , Proteína p24 del Núcleo del VIH/metabolismo , Infecciones por VIH/metabolismo , Infecciones por VIH/virología , Seropositividad para VIH , VIH-1/inmunología , VIH-1/metabolismo , VIH-1/patogenicidad , Células HeLa , Proteínas del Virus de la Inmunodeficiencia Humana/fisiología , Humanos , Macrófagos/virología , Proteínas Reguladoras y Accesorias Virales/fisiología , Virión/metabolismo , Ensamble de Virus/fisiología , Liberación del Virus/fisiología
15.
PLoS Pathog ; 15(2): e1007596, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30785952

RESUMEN

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.


Asunto(s)
ARN Helicasas/metabolismo , Estabilidad del ARN/fisiología , Proteínas Reguladoras y Accesorias Virales/metabolismo , Línea Celular , Núcleo Celular , Proteínas Activadoras de GTPasa/metabolismo , Proteínas Activadoras de GTPasa/fisiología , Regulación Viral de la Expresión Génica/genética , Genes Virales/genética , Células HEK293 , Herpesvirus Humano 8/metabolismo , Herpesvirus Humano 8/patogenicidad , Humanos , Proteínas Nucleares , Proteína I de Unión a Poli(A)/metabolismo , Proteína I de Unión a Poli(A)/fisiología , Unión Proteica , ARN Helicasas/fisiología , Estabilidad del ARN/genética , ARN Nuclear/fisiología , ARN Viral , Proteínas de Unión al ARN , Factores de Transcripción , Proteínas Virales/genética , Proteínas Virales/metabolismo , Proteínas Reguladoras y Accesorias Virales/fisiología , Replicación Viral
17.
PLoS Pathog ; 14(8): e1007232, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30096191

RESUMEN

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.


Asunto(s)
Multimerización de Proteína , Proteínas Reguladoras y Accesorias Virales/química , Proteínas Reguladoras y Accesorias Virales/metabolismo , Dominio Catalítico , Cristalografía por Rayos X , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/metabolismo , Humanos , Modelos Moleculares , Simulación del Acoplamiento Molecular , Sistemas de Lectura Abierta , Multimerización de Proteína/genética , Estabilidad Proteica , Estructura Cuaternaria de Proteína , Proteínas Reguladoras y Accesorias Virales/genética , Proteínas Reguladoras y Accesorias Virales/fisiología
18.
Viruses ; 10(8)2018 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-30060558

RESUMEN

The porcine epidemic diarrhea virus (PEDV) is an important swine pathogen responsible for severe watery diarrhea, particularly in neonatal piglets. Despite extensive studies performed to elucidate the function of several viral proteins, the contribution of an accessory protein ORF3 in PEDV replication is still largely unknown. Here, we constructed expression plasmids as well as recombinant PEDV carrying myc-tagged ORF3 to assess their expression and subcellular localization in both transfected and infected cells. In PEDV-infected cells, ORF3 was predominantly localized in the cytoplasm, partially in the endoplasmic reticulum (ER) and the Golgi apparatus (Golgi). Interestingly, ORF3 with the N-terminal Flag tag was also detected on the cell surface concomitant with the spike (S) protein as determined by flow cytometry and confocal microscopy. ORF3 and S proteins were also co-localized at perinuclear compartments and in the vesicle-like structures in transfected and infected cells. We also demonstrated that both full-length and naturally truncated ORF3 proteins could interact with the S protein but with different binding affinity, which correlate with the ability of the protein to regulate virus replication in cell culture. Collectively, our results underscore the unprecedented role of the ORF3, which involves the interaction of ORF3 with S and, possibly, other structural protein during PEDV replication.


Asunto(s)
Virus de la Diarrea Epidémica Porcina/fisiología , Glicoproteína de la Espiga del Coronavirus/fisiología , Proteínas Reguladoras y Accesorias Virales/fisiología , Replicación Viral , Animales , Chlorocebus aethiops , Infecciones por Coronavirus/veterinaria , Infecciones por Coronavirus/virología , Citoplasma/química , Citoplasma/virología , Replicación del ADN , Diarrea/veterinaria , Diarrea/virología , Células HEK293 , Humanos , Microscopía Confocal , Plásmidos/genética , Virus de la Diarrea Epidémica Porcina/genética , Virus de la Diarrea Epidémica Porcina/patogenicidad , Unión Proteica , Glicoproteína de la Espiga del Coronavirus/genética , Porcinos , Enfermedades de los Porcinos/virología , Transfección , Células Vero , Proteínas Reguladoras y Accesorias Virales/genética
19.
Retrovirology ; 15(1): 6, 2018 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-29329537

RESUMEN

BACKGROUND: The HIV-1 accessory proteins Nef and Vpu alter cell surface levels of multiple host proteins to modify the immune response and increase viral persistence. Nef and Vpu can downregulate cell surface levels of the co-stimulatory molecule CD28, however the mechanism of this function has not been completely elucidated. RESULTS: Here, we provide evidence that Nef and Vpu decrease cell surface and total cellular levels of CD28. Moreover, using inhibitors we implicate the cellular degradation machinery in the downregulation of CD28. We shed light on the mechanisms of CD28 downregulation by implicating the Nef LL165 and DD175 motifs in decreasing cell surface CD28 and Nef DD175 in decreasing total cellular CD28. Moreover, the Vpu LV64 and S52/56 motifs were required for cell surface CD28 downregulation, while, unlike for CD4 downregulation, Vpu W22 was dispensable. The Vpu S52/56 motif was also critical for Vpu-mediated decreases in total CD28 protein level. Finally, the ability of Vpu to downregulate CD28 is conserved between multiple group M Vpu proteins and infection with viruses encoding or lacking Nef and Vpu have differential effects on activation upon stimulation. CONCLUSIONS: We report that Nef and Vpu downregulate cell surface and total cellular CD28 levels. We identified inhibitors and mutations within Nef and Vpu that disrupt downregulation, shedding light on the mechanisms utilized to downregulate CD28. The conservation and redundancy between the abilities of two HIV-1 proteins to downregulate CD28 highlight the importance of this function, which may contribute to the development of latently infected cells.


Asunto(s)
Antígenos CD28/genética , Linfocitos T CD4-Positivos/inmunología , Regulación hacia Abajo , Infecciones por VIH/inmunología , VIH-1/fisiología , Proteínas del Virus de la Inmunodeficiencia Humana/fisiología , Proteínas Reguladoras y Accesorias Virales/fisiología , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/fisiología , Secuencias de Aminoácidos/genética , Antígenos CD28/metabolismo , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD4-Positivos/virología , Membrana Celular/metabolismo , Células Cultivadas , Infecciones por VIH/metabolismo , Infecciones por VIH/virología , Interacciones Huésped-Patógeno , Proteínas del Virus de la Inmunodeficiencia Humana/química , Proteínas del Virus de la Inmunodeficiencia Humana/genética , Humanos , Activación de Linfocitos , Lisosomas/metabolismo , Mutación , Proteínas Reguladoras y Accesorias Virales/química , Proteínas Reguladoras y Accesorias Virales/genética , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/química , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/genética
20.
PLoS Pathog ; 14(1): e1006852, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29357390

RESUMEN

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.


Asunto(s)
Proteína 58 DEAD Box/metabolismo , Herpesviridae/enzimología , Interferones/farmacología , Factores de Transcripción/metabolismo , Proteínas de Motivos Tripartitos/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas Reguladoras y Accesorias Virales/fisiología , Núcleo Celular/metabolismo , Células Cultivadas , Células HEK293 , Células HeLa , Humanos , Receptores Inmunológicos , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Ubiquitina/metabolismo , Ubiquitinación , Replicación Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...