Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Virol ; 97(5): e0058023, 2023 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-37166302

RESUMEN

Hepatitis B virus (HBV) infection affects hepatic metabolism. Serum metabolomics studies have suggested that HBV possibly hijacks the glycerol-3-phosphate (G3P) shuttle. In this study, the two glycerol-3-phosphate dehydrogenases (GPD1 and GPD2) in the G3P shuttle were analyzed for determining their role in HBV replication and the findings revealed that GPD2 and not GPD1 inhibited HBV replication. The knockdown of GPD2 expression upregulated HBV replication, while GPD2 overexpression reduced HBV replication. Moreover, the overexpression of GPD2 significantly reduced HBV replication in hydrodynamic injection-based mouse models. Mechanistically, this inhibitory effect is related to the GPD2-mediated degradation of HBx protein by recruiting the E3 ubiquitin ligase TRIM28 and not to the alterations in G3P metabolism. In conclusion, this study revealed GPD2, a key enzyme in the G3P shuttle, as a host restriction factor in HBV replication. IMPORTANCE The glycerol-3-phosphate (G3P) shuttle is important for the delivery of cytosolic reducing equivalents into mitochondria for oxidative phosphorylation. The study analyzed two key components of the G3P shuttle and identified GPD2 as a restriction factor in HBV replication. The findings revealed a novel mechanism of GPD2-mediated inhibition of HBV replication via the recruitment of TRIM28 for degrading HBx, and the HBx-GPD2 interaction could be another potential therapeutic target for anti-HBV drug development.


Asunto(s)
Glicerolfosfato Deshidrogenasa , Hepatitis B , Proteína 28 que Contiene Motivos Tripartito , Proteínas Reguladoras y Accesorias Virales , Animales , Ratones , Glicerol/metabolismo , Glicerolfosfato Deshidrogenasa/metabolismo , Hepatitis B/metabolismo , Virus de la Hepatitis B/fisiología , Mitocondrias/enzimología , Fosfatos/metabolismo , Proteína 28 que Contiene Motivos Tripartito/metabolismo , Proteínas Reguladoras y Accesorias Virales/genética , Proteínas Reguladoras y Accesorias Virales/metabolismo , Replicación Viral
2.
J Virol ; 96(13): e0058522, 2022 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-35862693

RESUMEN

The biogenesis of covalently closed circular DNA (cccDNA) from relaxed circular DNA (rcDNA) is essential for chronic hepatitis B virus (HBV) infection. Different host DNA repair proteins are involved in the conversion of rcDNA to cccDNA. Here, we reported that the DNA repair factor poly(ADP-ribose) polymerase 1 (PARP1) is engaged in HBV cccDNA formation. PARP1 depletion remarkably impaired HBV replication and cccDNA synthesis. Inhibition of PARP1 poly (ADP-ribosylation) activity by olaparib suppressed cccDNA synthesis both in vitro and in vivo. Specifically, the early stage of cccDNA reservoir establishment was more sensitive to olaparib, suggesting that PARP1 participated in de novo cccDNA formation. Furthermore, PARP1 was activated by recognizing the rcDNA-like lesions directly and combined with other DNA repair proteins. The results presented proposed that the DNA damage-sensing protein PARP1 and poly(ADP-ribosylation) modification play a key role in cccDNA formation, which might be the target for developing the anti-HBV drug. IMPORTANCE The biogenesis and eradication of HBV cccDNA have been a research priority in recent years. In this study, we identified the DNA repair factor PARP1 as a host factor required for the HBV de novo cccDNA formation. HBV infection caused PARylation through PARP1 in Huh7-NTCP cells, primary human hepatocytes, and human-liver chimeric mice. We found that PARP1 could directly bind to the rcDNA lesions and was activated, PARylating other DNA repair proteins. We address the importance of PARP1-mediated PARylation in HBV cccDNA formation, which is a potential therapeutic target for chronic hepatitis B.


Asunto(s)
ADN Circular , Hepatitis B , Poli(ADP-Ribosa) Polimerasa-1 , Animales , Reparación del ADN , ADN Circular/genética , ADN Circular/metabolismo , ADN Viral/genética , ADN Viral/metabolismo , Hepatitis B/virología , Virus de la Hepatitis B/genética , Humanos , Ratones , Poli(ADP-Ribosa) Polimerasa-1/genética , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Provirus/genética
3.
J Med Virol ; 95(3): e28610, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36840407

RESUMEN

Tick-borne encephalitis virus (TBEV) is the causative agent of a potentially fatal neurological infection in humans. Investigating virus-host interaction is important for understanding the pathogenesis of TBEV and developing effective antiviral drugs against this virus. Here, we report that mammalian ste20-like kinase 3 (MST3) is involved in the regulation of TBEV infection. The knockdown or knockout of MST3, but not other mammalian ste20-like kinase family members, inhibited TBEV replication. The knockdown of MST3 also significantly reduced TBEV replication in mouse primary astrocytes. Life cycle analysis indicated that MST3 remarkably impaired virion assembly efficiency and specific infectivity by respectively 59% and 95% in MST3-knockout cells. We further found that MST3 interacts with the viral proteins NS2A and prM; and MST3 enhances the interaction of NS2A-NS4A. Thus, MST3-NS2A complex plays a major role in recruiting prM-E heterodimers and NS4A and mediates the virion assembly. Additionally, we found that MST3 was biotinylated and combined with other proteins (e.g., ATG5, Sec24A, and SNX4) that are associated with the cellular membrane required for TBEV infection. Overall, our study revealed a novel function for MST3 in TBEV infection and identified as a novel host factor supporting TBEV assembly.


Asunto(s)
Virus de la Encefalitis Transmitidos por Garrapatas , Encefalitis Transmitida por Garrapatas , Animales , Ratones , Humanos , Virus de la Encefalitis Transmitidos por Garrapatas/genética , Proteínas Virales/metabolismo , Mamíferos/metabolismo , Proteínas de Transporte Vesicular
4.
J Biol Chem ; 297(6): 101380, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34740611

RESUMEN

Histone deacetylase 5 (HDAC5) has been reported to have a strong regulatory function in the proinflammatory response, but the mechanism is still unknown. Here, we identified HDAC5 as a positive regulator of NF-κB signaling in vivo. HDAC5-deficient mice exhibited enhanced survival in response to LPS challenge. Using LPS, TNFα, different kinds of viruses, hydrogen peroxide, or ultraviolet stimulation, we demonstrate that HDAC5-mediated regulation of NF-κB occurs in manners both dependent on and independent of IKK, an upstream kinase in the NF-κB signaling pathway. Deficiency in HDAC5 impaired the phosphorylation of IKKß, subsequent phosphorylation of the NF-κB inhibitor protein IκBα and NF-κB subunit p65. We also show that the phosphatase PP2A repressed transcriptional activation of NF-κB by decreasing phosphorylation of IKKß, p65, and IκBα. In vitro deacetylation experiments and site-directed mutagenesis experiments indicated that HDAC5 directly deacetylated PP2Ac at Lys136, which resulted in the deactivation of PP2A. Our data add mechanistic insight into the cross talk between epigenetic and posttranslational modifications regulating NF-κB signaling and protein phosphatase activation that mediate survival in response to inflammatory challenges.


Asunto(s)
Histona Desacetilasas/metabolismo , Proteína Fosfatasa 2/metabolismo , Transducción de Señal , Factor de Transcripción ReIA/metabolismo , Acetilación , Animales , Chlorocebus aethiops , Células HEK293 , Histona Desacetilasas/genética , Humanos , Ratones , Ratones Noqueados , Proteína Fosfatasa 2/genética , Células RAW 264.7 , Células THP-1 , Factor de Transcripción ReIA/genética , Células Vero
5.
J Virol ; 95(4)2021 01 28.
Artículo en Inglés | MEDLINE | ID: mdl-33208450

RESUMEN

Tick-borne encephalitis virus (TBEV), a major tick-borne viral pathogen of humans, is known to cause neurological diseases such as meningitis, encephalitis, and meningoencephalitis. However, the life cycle and pathogenesis of TBEV are not well understood. Here, we show that the knockdown or knockout of ADAM15 (a disintegrin and metalloproteinase 15), a host protein involved in neuroblastoma diseases, leads to TBEV replication and assembly defects. We characterized the disintegrin domain in ADAM15 and found that the ADAM15 subcellular localization was changed following TBEV infection. RNA interference (RNAi) screen analysis confirmed ADAM's nonredundant functions and identified a specific role for ADAM15 in TBEV infection. An RNA-sequencing analysis was also conducted to understand the causal link between TBEV infection and the cellular endomembrane network, namely, the generation of replication organelles promoting viral genome replication and virus production. Our data demonstrated that TBEV infection changes ADAM15 cellular localization, which contributes to membrane reorganization and viral replication.IMPORTANCE Tick populations are increasing, and their geographic ranges are expanding. Increases in tick-borne disease prevalence and transmission are important public health issues. Tick-borne encephalitis virus (TBEV) often results in meningitis, encephalitis, and meningoencephalitis. TBEV causes clinical disease in more than 20,000 humans in Europe and Asia per year. An increased incidence of TBE has been noted in Europe and Asia, as a consequence of climate and socioeconomic changes. The need to investigate the mechanism(s) of interaction between the virus and the host factors is apparent, as it will help us to understand the roles of host factors in the life cycle of TBEV. The significance of our research is in identifying the ADAM15 for TBEV replication, which will greatly enhance our understanding of TBEV life cycle and highlight a target for pharmaceutical consideration.


Asunto(s)
Proteínas ADAM/fisiología , Virus de la Encefalitis Transmitidos por Garrapatas/fisiología , Encefalitis Transmitida por Garrapatas/virología , Interacciones Microbiota-Huesped , Proteínas de la Membrana/fisiología , Animales , Chlorocebus aethiops , Cricetinae , Células HEK293 , Humanos , Células Vero , Replicación Viral
6.
Hepatology ; 72(5): 1569-1585, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32103509

RESUMEN

BACKGROUND AND AIMS: The regulation of hepatic very-low-density lipoprotein (VLDL) secretion is vital for lipid metabolism whose pathogenetic status is involved in fatty liver disease and dyslipidemia seen in hepatic steatosis. Accumulated evidence suggest that apolipoprotein E (ApoE) is closely related to hepatic VLDL secretion. Here, we report that the expression of patatin-like phospholipase domain containing protein 7 (PNPLA7) is strongly induced by hepatic steatosis and positively correlates with plasma triacylglycerol (TAG) levels in the human subjects, whereas the role of PNPLA7 in hepatic VLDL secretion is unknown. APPROACH AND RESULTS: Herein, with genetic manipulation in the mice, the deficiency of hepatic PNPLA7 expression resulted in reduced VLDL secretion accompanied by enhanced hepatic lipid accumulation and decreased hepatic ApoE expression. Furthermore, knockdown of PNPLA7 in the livers of the db/db mice also resulted in significant reduction in plasma TAG level but aggravated hepatic steatosis. Importantly, we observed that PNPLA7 interacted with ApoE and presumably at the site of endoplasmic reticulum. Mechanistically, we have shown that PNPLA7 could modulate polyubiquitination and proteasomal-mediated degradation of ApoE. Overexpressed ApoE restored the impaired VLDL-TAG metabolism in PNPLA7-knockdown primary hepatocytes. CONCLUSION: PNPLA7 plays a critical role in regulating hepatic VLDL secretion by modulating ApoE stability through its interaction with ApoE.


Asunto(s)
Apolipoproteínas E/metabolismo , Hígado Graso/metabolismo , Lipasa/metabolismo , Hígado/patología , Lisofosfolipasa/metabolismo , Animales , Apolipoproteínas E/genética , Línea Celular Tumoral , Retículo Endoplásmico/patología , Hígado Graso/sangre , Hígado Graso/diagnóstico , Hígado Graso/cirugía , Femenino , Técnicas de Silenciamiento del Gen , Células HEK293 , Humanos , Lipasa/genética , Metabolismo de los Lípidos , Lipoproteínas VLDL/sangre , Lipoproteínas VLDL/metabolismo , Hígado/cirugía , Lisofosfolipasa/genética , Masculino , Ratones , Ratones Noqueados para ApoE , Complejo de la Endopetidasa Proteasomal/metabolismo , Estabilidad Proteica , Proteolisis , Índice de Severidad de la Enfermedad , Triglicéridos/sangre , Triglicéridos/metabolismo , Ubiquitinación
7.
Clin Sci (Lond) ; 134(5): 529-542, 2020 03 13.
Artículo en Inglés | MEDLINE | ID: mdl-32100852

RESUMEN

Chronic hepatitis C virus (HCV) infection has a close association with type 2 diabetes mellitus. Although the mechanisms of insulin resistance in chronic hepatitis C (CHC) patients have been extensively studied, little attention has been given to the role of ß-cell function in HCV-associated diabetes. Here, we analysed ß-cell function in CHC patients and HCV-infected mouse model and found in addition to insulin resistance, impaired pancreatic ß-cell function occurred in CHC patients and HCV-infected C/OTg mice, not only in diabetic individuals but also in individuals with impaired fasting glucose levels. Both first-phase and second-phase insulin secretion were impaired, at least partially due to the reduction of exocytosis of secretory insulin-containing granules following HCV infection. Up-regulated p38δ in HCV-infected ß-cells resulted in inactivation of protein kinase D (PKD), which was responsible for impaired insulin secretory capacity of ß-cells. Thus, impaired insulin secretion due to HCV infection in ß-cells contributes to HCV-associated type 2 diabetes. These findings provided a new inspiration for the important prognostic and therapeutic implications in the management of CHC patients with impaired fasting glucose.


Asunto(s)
Exocitosis , Hepatitis C Crónica/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Proteína Quinasa 13 Activada por Mitógenos/metabolismo , Animales , Línea Celular Tumoral , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/metabolismo , Intolerancia a la Glucosa/sangre , Intolerancia a la Glucosa/metabolismo , Intolerancia a la Glucosa/virología , Hepatitis C Crónica/virología , Humanos , Insulina/metabolismo , Células Secretoras de Insulina/virología , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Ratones Transgénicos , Proteína Quinasa C/metabolismo
8.
J Virol ; 92(12)2018 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-29618641

RESUMEN

As a virus-encoded actin nucleation promoting factor (NPF), P78/83 induces actin polymerization to assist in Autographa californica multiple nucleopolyhedrovirus (AcMNPV) propagation. According to our previous study, although P78/83 actively undergoes ubiquitin-independent proteasomal degradation, AcMNPV encodes budded virus/occlusion derived virus (BV/ODV)-C42 (C42), which allows P78/83 to function as a stable NPF by inhibiting its degradation during viral infection. However, whether there are other viral proteins involved in regulating P78/83-induced actin polymerization has yet to be determined. In this study, we found that Ac102, an essential viral gene product previously reported to play a key role in mediating the nuclear accumulation of actin during AcMNPV infection, is a novel regulator of P78/83-induced actin polymerization. By characterizing an ac102 knockout bacmid, we demonstrated that Ac102 participates in regulating nuclear actin polymerization as well as the morphogenesis and distribution of capsid structures in the nucleus. These regulatory effects are heavily dependent on an interaction between Ac102 and C42. Further investigation revealed that Ac102 binds to C42 to suppress K48-linked ubiquitination of C42, which decreases C42 proteasomal degradation and consequently allows P78/83 to function as a stable NPF to induce actin polymerization. Thus, Ac102 and C42 form a regulatory cascade to control viral NPF activity, representing a sophisticated mechanism for AcMNPV to orchestrate actin polymerization in both a ubiquitin-dependent and ubiquitin-independent manner.IMPORTANCE Actin is one of the most functionally important proteins in eukaryotic cells. Morphologically, actin can be found in two forms: a monomeric form called globular actin (G-actin) and a polymeric form called filamentous actin (F-actin). G-actin can polymerize to form F-actin, and nucleation promoting factor (NPF) is the initiator of this process. Many viral pathogens harness the host actin polymerization machinery to assist in virus propagation. Autographa californica multiple nucleopolyhedrovirus (AcMNPV) induces actin polymerization in host cells. P78/83, a viral NPF, is responsible for this process. Previously, we identified that BV/ODV-C42 (C42) binds to P78/83 and protects it from degradation. In this report, we determined that another viral protein, Ac102, is involved in modulating C42 ubiquitination and, consequently, ensures P78/83 activity as an NPF to initiate actin polymerization. This regulatory cascade represents a novel mechanism by which a virus can harness the cellular actin cytoskeleton to assist in viral propagation.


Asunto(s)
Actinas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Complejo Mediador/metabolismo , Nucleopoliedrovirus/crecimiento & desarrollo , Replicación Viral/fisiología , Animales , Proteínas de la Cápside/metabolismo , Línea Celular , Núcleo Celular/virología , Técnicas de Inactivación de Genes , Péptidos y Proteínas de Señalización Intracelular/genética , Nucleopoliedrovirus/metabolismo , Polimerizacion , Células Sf9 , Spodoptera/virología , Ubiquitinación
9.
PLoS Pathog ; 12(11): e1005994, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27802336

RESUMEN

Actin, nucleation-promoting factors (NPFs), and the actin-related protein 2/3 complex (Arp2/3) are key elements of the cellular actin polymerization machinery. With nuclear actin polymerization implicated in ever-expanding biological processes and the discovery of the nuclear import mechanisms of actin and NPFs, determining Arp2/3 nucleo-cytoplasmic shuttling mechanism is important for understanding the function of nuclear actin. A unique feature of alphabaculovirus infection of insect cells is the robust nuclear accumulation of Arp2/3, which induces actin polymerization in the nucleus to assist in virus replication. We found that Ac34, a viral late gene product encoded by the alphabaculovirus Autographa californica multiple nucleopolyhedrovirus (AcMNPV), is involved in Arp2/3 nuclear accumulation during virus infection. Further assays revealed that the subcellular distribution of Arp2/3 under steady-state conditions is controlled by chromosomal maintenance 1 (CRM1)-dependent nuclear export. Upon AcMNPV infection, Ac34 inhibits CRM1 pathway and leads to Arp2/3 retention in the nucleus.


Asunto(s)
Complejo 2-3 Proteico Relacionado con la Actina/metabolismo , Transporte Activo de Núcleo Celular/fisiología , Carioferinas/metabolismo , Nucleopoliedrovirus/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Replicación Viral/fisiología , Animales , Núcleo Celular/metabolismo , Técnica del Anticuerpo Fluorescente , Inmunoprecipitación , Células Sf9 , Transfección , Proteínas Virales/metabolismo , Proteína Exportina 1
10.
J Gen Virol ; 98(6): 1410-1421, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28678687

RESUMEN

Ceruloplasmin (CP) is mainly synthesized by hepatocytes and plays an essential role in iron metabolism. Previous reports have shown that CP levels correlate negatively with disease progression in patients with chronic hepatitis B. However, the function of CP in the hepatitis B virus (HBV) life cycle and the mechanism underlying the above correlation remain unclear. Here, we report that CP can selectively inhibit the production of extracellular HBV virions without altering intracellular viral replication. HBV expression can also downregulate the expression of CP. Knockdown of CP using small interfering RNA significantly increased the level of extracellular HBV virions in both Huh7 and HepG2.2.15 cells, while overexpression of CP decreased this level. Mechanistically, CP could specifically interact with the HBV middle surface protein (MHB). Using an HBV replication-competent clone unable to express MHBs, we demonstrated that the overexpression of CP did not affect the production of extracellular HBV virions in the absence of MHBs. Furthermore, introduction of an MHB expression construct could rescue the impairment in virion production caused by CP. Taken together, our results suggest that CP may be an important host factor that targets MHBs during the envelopment and/or release of virions.


Asunto(s)
Ceruloplasmina/metabolismo , Antígenos de Superficie de la Hepatitis B/metabolismo , Virus de la Hepatitis B/crecimiento & desarrollo , Virus de la Hepatitis B/inmunología , Adulto , Línea Celular , Ceruloplasmina/análisis , Femenino , Hepatitis B Crónica/virología , Hepatocitos/virología , Humanos , Masculino , Persona de Mediana Edad , Mapeo de Interacción de Proteínas
11.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1862(7): 692-705, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28336330

RESUMEN

Lipid droplet (LD) accumulation in hepatocytes is a typical character of steatosis. Hepatitis C virus (HCV) infection, one of the risk factors related to steatosis, induced LD accumulation in cultured cells. However, the mechanisms of which HCV induce LD formation are not fully revealed. Previously we identified cytosolic phospholipase A2 gamma (PLA2G4C) as a host factor upregulated by HCV infection and involved in HCV replication. Here we further revealed that PLA2G4C plays an important role in LD biogenesis and refined the functional analysis of PLA2G4C in LD biogenesis and HCV assembly. LD formation upon fatty acid and HCV stimulation in PLA2G4C knockdown cells was impaired and could not be restored by complementation with PLA2G4A. PLA2G4C was tightly associated in the membrane with the domain around the amino acid residues 260-292, normally in ER but relocated into LDs upon oleate stimulation. Mutant PLA2G4C without enzymatic activity was not able to restore LD formation in PLA2G4C knockdown cells. Thus, both the membrane attachment and the enzymatic activity of PLA2G4C were required for its function in LD formation. The participation of PLA2G4C in LD formation is correlated with its involvement in HCV assembly. Finally, PLA2G4C overexpression itself led to LD formation in hepatic cells and enhanced LD accumulation in the liver of high-fat diet (HFD)-fed mice, suggesting its potential role in fatty liver disease.


Asunto(s)
Citosol/metabolismo , Fosfolipasas A2 Grupo IV/metabolismo , Gotas Lipídicas/metabolismo , Metabolismo de los Lípidos/fisiología , Aminoácidos/metabolismo , Animales , Línea Celular , Citosol/virología , Retículo Endoplásmico/metabolismo , Retículo Endoplásmico/virología , Ácidos Grasos/metabolismo , Células HEK293 , Hepacivirus/patogenicidad , Hepatitis C/metabolismo , Hepatocitos/metabolismo , Hepatocitos/virología , Humanos , Gotas Lipídicas/virología , Hígado/metabolismo , Hígado/virología , Masculino , Membranas/metabolismo , Membranas/virología , Ratones , Ratones Endogámicos C57BL
12.
J Virol ; 89(4): 2367-77, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25505071

RESUMEN

UNLABELLED: Several members of the phospholipase family have been reported to be involved in hepatitis C virus (HCV) replication. Here, we identified another phospholipase, phosphatidylserine-specific phospholipase A1 (PLA1A), as a host factor involved in HCV assembly. PLA1A was upregulated by HCV infection, and PLA1A knockdown significantly reduced J399EM (genotype 2a) HCV propagation at the assembly step but not the entry, RNA replication, and protein translation steps of the life cycle. Protein localization and interaction analysis further revealed a role of PLA1A in the interaction of NS2-E2 and NS2-NS5A, as the formation of the NS2-E2 and NS2-NS5A complexes was weakened in the absence of PLA1A. In addition, PLA1A stabilized the NS2/NS5A dotted structure during infection. These data suggest that PLA1A plays an important role in bridging the membrane-associated NS2-E2 complex and the NS5A-associated replication complex via its interaction with E2, NS2, and NS5A, which leads to a coordinating interaction between the structural and nonstructural proteins and facilitates viral assembly. IMPORTANCE: Hepatitis C virus (HCV) genomic replication is driven by the replication complex and occurs at the membranous web, while the lipid droplet is the organelle in which virion assembly is initiated. In this study, we identified phosphatidylserine-specific phospholipase A1 (PLA1A), a member of phospholipase A 1 family, as a novel host factor involved in the assembly process of HCV. PLA1A, which is induced by HCV infection at a late infection stage, interacts with HCV E2, NS2, and NS5A proteins and enhances and stabilizes the NS2-E2 and NS2-NS5A complex formation, which is essential for viral assembly. Thus, PLA1A is an important host factor which is involved in the initiation of the viral assembly in close proximity to Core-decorated lipid droplets through bringing together the HCV replication complex and envelope complex.


Asunto(s)
Hepacivirus/fisiología , Interacciones Huésped-Patógeno , Fosfolipasas A1/metabolismo , Multimerización de Proteína , Proteínas no Estructurales Virales/metabolismo , Ensamble de Virus , Línea Celular , Perfilación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos
13.
Virol Sin ; 2024 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-38950863

RESUMEN

The association between chronic HCV infection and type 2 diabetes mellitus (T2DM) has been established; however, there is limited research on ß-cell function particularly in the pre-diabetic population. Here, we evaluated indices of ß-cell function and insulin sensitivity across the spectrum from normal glucose tolerance to T2DM in individuals with and without chronic hepatitis C (CHC), and the effects of antiviral treatments on these variables. A total of 153 non-cirrhotic, non-fibrotic CHC patients with a BMI < 25 were enrolled in the study. Among them, 119 were successfully treated with either direct acting antiviral (DAA) drugs or pegylated interferon/ribavirin (IFN/RBV) anti-HCV therapy. Fasting state- and oral glucose tolerance test (OGTT)-derived indexes were used to evaluate ß-cell function and insulin sensitivity. Among all subjects, 19 (13%) had T2DM and 21% exhibited pre-diabetes including 8% isolated impaired fasting glucose (IFG) and 13% combined IFG and impaired glucose tolerance (IGT). Early and total insulin secretion adjusted for the degree of insulin resistance were decreased in prediabetic CHC patients compared to HCV-uninfected individuals. Viral eradication through DAA or IFN/RBV therapy demonstrated positive impacts on insulin sensitivity and ß-cell function in CHC patients who achieved sustained virologic response (SVR), regardless of fasting or OGTT state. These findings emphasize the role of HCV in the development of ß-cell dysfunction, while also suggesting that viral eradication can improve insulin secretion, reverse insulin resistance, and ameliorates glycemic control. These results have important implications for managing prediabetic CHC patients and could prevent diabetes-related clinical manifestations and complications.

14.
Nat Microbiol ; 9(4): 1075-1088, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38553607

RESUMEN

Although vaccines are available for SARS-CoV-2, antiviral drugs such as nirmatrelvir are still needed, particularly for individuals in whom vaccines are less effective, such as the immunocompromised, to prevent severe COVID-19. Here we report an α-ketoamide-based peptidomimetic inhibitor of the SARS-CoV-2 main protease (Mpro), designated RAY1216. Enzyme inhibition kinetic analysis shows that RAY1216 has an inhibition constant of 8.4 nM and suggests that it dissociates about 12 times slower from Mpro compared with nirmatrelvir. The crystal structure of the SARS-CoV-2 Mpro:RAY1216 complex shows that RAY1216 covalently binds to the catalytic Cys145 through the α-ketoamide group. In vitro and using human ACE2 transgenic mouse models, RAY1216 shows antiviral activities against SARS-CoV-2 variants comparable to those of nirmatrelvir. It also shows improved pharmacokinetics in mice and rats, suggesting that RAY1216 could be used without ritonavir, which is co-administered with nirmatrelvir. RAY1216 has been approved as a single-component drug named 'leritrelvir' for COVID-19 treatment in China.


Asunto(s)
COVID-19 , Vacunas , Humanos , Animales , Ratones , Ratas , SARS-CoV-2 , Tratamiento Farmacológico de COVID-19 , Cinética , Lactamas , Nitrilos , Ratones Transgénicos
15.
Virol Sin ; 38(5): 767-777, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37328107

RESUMEN

Tick-borne encephalitis virus (TBEV) is an important tick-borne pathogen that poses as a serious public health concern. The coverage and immunogenicity of the currently available vaccines against TBEV are relatively low; therefore, it is crucial to develop novel and effective vaccines against TBEV. The present study describes a novel strategy for the assembly of virus-like particles (VLPs) by co-expressing the structural (core/prM/E) and non-structural (NS2B/NS3Pro) proteins of TBEV. The efficacy of the VLPs was subsequently evaluated in C57BL/6 mice, and the resultant IgG serum could neutralize both Far-Eastern and European subtypes of TBEV. These findings indicated that the VLP-based vaccine elicited the production of cross-subtype reactive antibodies. The VLPs provided protection to mice lacking the type I interferon receptor (IFNAR-/-) against lethal TBEV challenge, with undetectable viral load in brain and intestinal tissues. Furthermore, the group that received the VLP vaccine did not exhibit significant pathological changes and the inflammatory factors were significantly suppressed compared to the control group. Immunization with the VLP vaccine induced the production of multiple-cytokine-producing antiviral CD4+ T cells in vivo, including TNF-α+, IL-2+, and IFN-γ+ T cells. Altogether, the findings suggest that noninfectious VLPs can serve as a potentially safe and effective vaccine candidate against diverse subtypes of TBEV.


Asunto(s)
Virus de la Encefalitis Transmitidos por Garrapatas , Encefalitis Transmitida por Garrapatas , Vacunas de Partículas Similares a Virus , Animales , Ratones , Virus de la Encefalitis Transmitidos por Garrapatas/genética , Vacunas de Partículas Similares a Virus/genética , Anticuerpos Antivirales , Encefalitis Transmitida por Garrapatas/prevención & control , Ratones Endogámicos C57BL
16.
Wei Sheng Wu Xue Bao ; 52(1): 52-9, 2012 Jan.
Artículo en Zh | MEDLINE | ID: mdl-22489460

RESUMEN

OBJECTIVE: We analyzed the effect of the stable expressed Hepatitis C virus core protein on PCK1 mRNA expression level and the molecular mechanisms involved in Huh7-lunet cells. METHODS: A retroviral vector mediated mammalian cell expression cell line of the HCV core protein was constructed. The mRNA and protein levels of PCK1, FOXO1 and PGC-1alpha were analyzed by Real-time PCR and luciferase assay in Huh7-lunet-core cells. RESULTS: HCV CORE upregulated the mRNA levels of PCK1 significantly. Both the mRNA and protein levels of FOXO1 were not affected in Huh7-lunet-core cells, whereas a decreased phosphorylation status of FOXO1 was exhibited. Moreover, activation of FOXO1 by HCV CORE was detected. Further, the mRNA level of PGC-1alpha was found to be significantly elevated in Huh7-lunet-core cells. CONCLUSION: Our results revealed for the first time that HCV core protein expression-mediated FOXO1 activation and the increased PGC-1alpha leaded to the elevation of PCK1 at the mRNA level, which suggesting the immoderate gluconeogenesis in HCV-infected hepatocytes. Our findings contributed to the understanding of the molecular mechanisms of HCV-related insulin resistance and provided potential new clues for the prevention and therapy of diabetes.


Asunto(s)
Factores de Transcripción Forkhead/fisiología , Proteínas de Choque Térmico/fisiología , Hepacivirus/fisiología , Péptidos y Proteínas de Señalización Intracelular/genética , Fosfoenolpiruvato Carboxiquinasa (GTP)/genética , Transducción de Señal , Factores de Transcripción/fisiología , Proteínas del Núcleo Viral/fisiología , Proteína Forkhead Box O1 , Humanos , Resistencia a la Insulina , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Transcripción Genética , Regulación hacia Arriba
17.
Antiviral Res ; 198: 105249, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35041910

RESUMEN

The binding of HBV polymerase (Pol) and the epsilon stem loop (ε) on the 5' terminal region of pgRNA is required for pgRNA packaging and HBV replication. Previous research has demonstrated that RNA binding motif protein 24 (RBM24) is involved in pgRNA packaging by mediating the interaction between HBV polymerase (Pol) and the ε element. Here, we demonstrate that RBM38 interacts with ε, pol, RBM24 and HBV core which mediate pgRNA packaging. RBM38 directly binds to the lower bulge of ε via RNA recognition submotifs (RNPs) and interacts with HBV Pol in an RNA-independent manner. RBM38 interacts with RBM24 and forms heterogeneous oligomers, which mediate Pol-ε binding and the formation of the Pol-RBM38/RBM24-ε complex. More important, RBM38 also binds to the HBV core via the C-terminal region (ARD domain), which facilitates the combination of Pol-ε with the HBV core protein. In conclusion, RBM38 facilitates the Pol-ε interaction and mediates Pol-ε in combining with the HBV core, triggering pgRNA packaging for reverse transcription and DNA synthesis. This study provides new insights into pgRNA encapsidation.


Asunto(s)
Virus de la Hepatitis B , ARN Viral , Virus de la Hepatitis B/genética , Virus de la Hepatitis B/metabolismo , Nucleocápside/metabolismo , ARN , ARN Viral/metabolismo , Motivos de Unión al ARN , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo
18.
Nat Microbiol ; 7(10): 1635-1649, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36151403

RESUMEN

Population antibody response is thought to be important in selection of virus variants. We report that SARS-CoV-2 infection elicits a population immune response that is mediated by a lineage of VH1-69 germline antibodies. A representative antibody R1-32 from this lineage was isolated. By cryo-EM, we show that it targets a semi-cryptic epitope in the spike receptor-binding domain. Binding to this non-ACE2 competing epitope results in spike destruction, thereby inhibiting virus entry. On the basis of epitope location, neutralization mechanism and analysis of antibody binding to spike variants, we propose that recurrent substitutions at 452 and 490 are associated with immune evasion of the identified population antibody response. These substitutions, including L452R (present in the Delta variant), disrupt interactions mediated by the VH1-69-specific hydrophobic HCDR2 to impair antibody-antigen association, enabling variants to escape. The first Omicron variants were sensitive to antibody R1-32 but subvariants that harbour L452R quickly emerged and spread. Our results provide insights into how SARS-CoV-2 variants emerge and evade host immune responses.


Asunto(s)
COVID-19 , SARS-CoV-2 , Anticuerpos Antivirales , Formación de Anticuerpos , Epítopos/genética , Humanos , Mutación , SARS-CoV-2/genética , Glicoproteína de la Espiga del Coronavirus/genética , Glicoproteína de la Espiga del Coronavirus/metabolismo
19.
Front Microbiol ; 12: 740464, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34803956

RESUMEN

Hepatitis viruses are primary causative agents of hepatitis and represent a major source of public health problems in the world. The host innate immune system forms the first line of defense against hepatitis viruses. Hepatitis viruses are sensed by specific pathogen recognition receptors (PRRs) that subsequently trigger the innate immune response and interferon (IFN) production. However, hepatitis viruses evade host immune surveillance via multiple strategies, which help compromise the innate immune response and create a favorable environment for viral replication. Therefore, this article reviews published findings regarding host innate immune sensing and response against hepatitis viruses. Furthermore, we also focus on how hepatitis viruses abrogate the antiviral effects of the host innate immune system.

20.
Protein Cell ; 12(4): 261-278, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-32772249

RESUMEN

TANK-binding kinase 1 (TBK1), a core kinase of antiviral pathways, activates the production of interferons (IFNs). It has been reported that deacetylation activates TBK1; however, the precise mechanism still remains to be uncovered. We show here that during the early stage of viral infection, the acetylation of TBK1 was increased, and the acetylation of TBK1 at Lys241 enhanced the recruitment of IRF3 to TBK1. HDAC3 directly deacetylated TBK1 at Lys241 and Lys692, which resulted in the activation of TBK1. Deacetylation at Lys241 and Lys692 was critical for the kinase activity and dimerization of TBK1 respectively. Using knockout cell lines and transgenic mice, we confirmed that a HDAC3 null mutant exhibited enhanced susceptibility to viral challenge via impaired production of type I IFNs. Furthermore, activated TBK1 phosphorylated HDAC3, which promoted the deacetylation activity of HDAC3 and formed a feedback loop. In this study, we illustrated the roles the acetylated and deacetylated forms of TBK1 play in antiviral innate responses and clarified the post-translational modulations involved in the interaction between TBK1 and HDAC3.


Asunto(s)
Histona Desacetilasas/inmunología , Proteínas Serina-Treonina Quinasas/inmunología , Virosis/inmunología , Animales , Chlorocebus aethiops , Células HEK293 , Histona Desacetilasas/genética , Humanos , Ratones , Ratones Transgénicos , Proteínas Serina-Treonina Quinasas/genética , Células RAW 264.7 , Células THP-1 , Células Vero , Virosis/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA