Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nat Microbiol ; 9(4): 1075-1088, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38553607

RESUMO

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.


Assuntos
COVID-19 , Vacinas , Humanos , Animais , Camundongos , Ratos , SARS-CoV-2 , Tratamento Farmacológico da COVID-19 , Cinética , Lactamas , Nitrilas , Camundongos Transgênicos
2.
Virol Sin ; 38(5): 767-777, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37328107

RESUMO

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.


Assuntos
Vírus da Encefalite Transmitidos por Carrapatos , Encefalite Transmitida por Carrapatos , Vacinas de Partículas Semelhantes a Vírus , Animais , Camundongos , Vírus da Encefalite Transmitidos por Carrapatos/genética , Vacinas de Partículas Semelhantes a Vírus/genética , Anticorpos Antivirais , Encefalite Transmitida por Carrapatos/prevenção & controle , Camundongos Endogâmicos C57BL
3.
J Virol ; 97(5): e0058023, 2023 05 31.
Artigo em Inglês | MEDLINE | ID: mdl-37166302

RESUMO

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.


Assuntos
Glicerolfosfato Desidrogenase , Hepatite B , Proteína 28 com Motivo Tripartido , Proteínas Virais Reguladoras e Acessórias , Animais , Camundongos , Glicerol/metabolismo , Glicerolfosfato Desidrogenase/metabolismo , Hepatite B/metabolismo , Vírus da Hepatite B/fisiologia , Mitocôndrias/enzimologia , Fosfatos/metabolismo , Proteína 28 com Motivo Tripartido/metabolismo , Proteínas Virais Reguladoras e Acessórias/genética , Proteínas Virais Reguladoras e Acessórias/metabolismo , Replicação Viral
4.
J Med Virol ; 95(3): e28610, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36840407

RESUMO

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.


Assuntos
Vírus da Encefalite Transmitidos por Carrapatos , Encefalite Transmitida por Carrapatos , Animais , Camundongos , Humanos , Vírus da Encefalite Transmitidos por Carrapatos/genética , Proteínas Virais/metabolismo , Mamíferos/metabolismo , Proteínas de Transporte Vesicular
5.
Nat Microbiol ; 7(10): 1635-1649, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36151403

RESUMO

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.


Assuntos
COVID-19 , SARS-CoV-2 , Anticorpos Antivirais , Formação de Anticorpos , Epitopos/genética , Humanos , Mutação , SARS-CoV-2/genética , Glicoproteína da Espícula de Coronavírus/genética , Glicoproteína da Espícula de Coronavírus/metabolismo
6.
J Virol ; 96(13): e0058522, 2022 07 13.
Artigo em Inglês | MEDLINE | ID: mdl-35862693

RESUMO

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.


Assuntos
DNA Circular , Hepatite B , Poli(ADP-Ribose) Polimerase-1 , Animais , Reparo do DNA , DNA Circular/genética , DNA Circular/metabolismo , DNA Viral/genética , DNA Viral/metabolismo , Hepatite B/virologia , Vírus da Hepatite B/genética , Humanos , Camundongos , Poli(ADP-Ribose) Polimerase-1/genética , Poli(ADP-Ribose) Polimerase-1/metabolismo , Provírus/genética
7.
Antiviral Res ; 198: 105249, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35041910

RESUMO

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.


Assuntos
Vírus da Hepatite B , RNA Viral , Vírus da Hepatite B/genética , Vírus da Hepatite B/metabolismo , Nucleocapsídeo/metabolismo , RNA , RNA Viral/metabolismo , Motivos de Ligação ao RNA , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo
8.
J Biol Chem ; 297(6): 101380, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34740611

RESUMO

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.


Assuntos
Histona Desacetilases/metabolismo , Proteína Fosfatase 2/metabolismo , Transdução de Sinais , Fator de Transcrição RelA/metabolismo , Acetilação , Animais , Chlorocebus aethiops , Células HEK293 , Histona Desacetilases/genética , Humanos , Camundongos , Camundongos Knockout , Proteína Fosfatase 2/genética , Células RAW 264.7 , Células THP-1 , Fator de Transcrição RelA/genética , Células Vero
9.
Front Microbiol ; 12: 740464, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34803956

RESUMO

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.

10.
Cell Metab ; 33(8): 1655-1670.e8, 2021 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-34015269

RESUMO

How amphipathic phospholipids are shuttled between the membrane bilayer remains an essential but elusive process, particularly at the endoplasmic reticulum (ER). One prominent phospholipid shuttling process concerns the biogenesis of APOB-containing lipoproteins within the ER lumen, which may require bulk trans-bilayer movement of phospholipids from the cytoplasmic leaflet of the ER bilayer. Here, we show that TMEM41B, present in the lipoprotein export machinery, encodes a previously conceptualized ER lipid scramblase mediating trans-bilayer shuttling of bulk phospholipids. Loss of hepatic TMEM41B eliminates plasma lipids, due to complete absence of mature lipoproteins within the ER, but paradoxically also activates lipid production. Mechanistically, scramblase deficiency triggers unique ER morphological changes and unsuppressed activation of SREBPs, which potently promotes lipid synthesis despite stalled secretion. Together, this response induces full-blown nonalcoholic hepatosteatosis in the TMEM41B-deficient mice within weeks. Collectively, our data uncovered a fundamental mechanism safe-guarding ER function and integrity, dysfunction of which disrupts lipid homeostasis.


Assuntos
Retículo Endoplasmático , Fosfolipídeos , Animais , Retículo Endoplasmático/metabolismo , Homeostase , Lipogênese , Lipoproteínas/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Fosfolipídeos/metabolismo
11.
J Virol ; 95(4)2021 01 28.
Artigo em Inglês | MEDLINE | ID: mdl-33208450

RESUMO

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.


Assuntos
Proteínas ADAM/fisiologia , Vírus da Encefalite Transmitidos por Carrapatos/fisiologia , Encefalite Transmitida por Carrapatos/virologia , Interações entre Hospedeiro e Microrganismos , Proteínas de Membrana/fisiologia , Animais , Chlorocebus aethiops , Cricetinae , Células HEK293 , Humanos , Células Vero , Replicação Viral
12.
Protein Cell ; 12(4): 261-278, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-32772249

RESUMO

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.


Assuntos
Histona Desacetilases/imunologia , Proteínas Serina-Treonina Quinases/imunologia , Viroses/imunologia , Animais , Chlorocebus aethiops , Células HEK293 , Histona Desacetilases/genética , Humanos , Camundongos , Camundongos Transgênicos , Proteínas Serina-Treonina Quinases/genética , Células RAW 264.7 , Células THP-1 , Células Vero , Viroses/genética
13.
J Mol Cell Biol ; 12(12): 933-945, 2020 07 27.
Artigo em Inglês | MEDLINE | ID: mdl-32770227

RESUMO

Protein modification by small ubiquitin-like modifier (SUMO) is an important regulatory mechanism for multiple cellular processes. Although the canonical pathway involving the ubiquitylation or phosphorylation of IκBα has been well characterized, little is known about the sumoylation of IκBα in the control of NF-κB activity. Here, we find that histone deacetylase 4 (HDAC4) negatively regulates tumor necrosis factor-alpha- or lipopolysaccharide-triggered NF-κB activation. HDAC4 belongs to the SUMO E3 ligase family and can directly sumoylate IκBα. The cytoplasm location of HDAC4 is essential for IκBα sumoylation. The Cys292 of HDAC4 is a key site for its SUMO E3 ligase activity. The sumoylation of IκBα prevents its polyubiquitination and degradation because these two modifications occur both at the Lys21. Our findings reveal a previously undiscovered role for HDAC4 in the inflammatory response as a SUMO E3 ligase for IκBα sumoylation. Our work provides insight into mechanisms ensuring optimal mediation of the NF-κB pathway.


Assuntos
Histona Desacetilases/metabolismo , Inibidor de NF-kappaB alfa/metabolismo , NF-kappa B/metabolismo , Proteínas Repressoras/metabolismo , Transdução de Sinais/genética , Sumoilação/genética , Animais , Células HEK293 , Histona Desacetilases/genética , Humanos , Lipopolissacarídeos/farmacologia , Camundongos , Células RAW 264.7 , Proteínas Recombinantes/farmacologia , Proteínas Repressoras/genética , Transdução de Sinais/efeitos dos fármacos , Transfecção , Fator de Necrose Tumoral alfa/farmacologia
14.
Sci Rep ; 10(1): 8989, 2020 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-32461635

RESUMO

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

15.
Emerg Microbes Infect ; 9(1): 714-726, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-32196427

RESUMO

Tick-borne encephalitis virus (TBEV) accounts for approximately 10,000 annual cases of severe encephalitis in Europe and Asia and causes encephalitis in humans. In this study, we demonstrate TBEV appears to activate the interferon (IFN)-ß dependent on RIG-I/MDA5. Both the IFN-ß accumulation and the IFN stimulated genes (ISGs) transcription greatly delay. Further studies reveal that TBEV NS4A could block the phosphorylation and dimerization of STAT1/STAT2 to affect type I and II IFN-mediated STAT signalling. Additional data indicate that the residue at K132 of TBEV NS4A could be modified by ubiquitination and this modification is necessary for the interaction of NS4A with STAT1. Dynamic ubiquitination of the NS4 protein during TBEV infection might account for delayed activation of the ISGs. These results define the TBEV NS4A as an antagonist of the IFN response, by demonstrating a correlation between the association and STAT interference. Our findings provide a foundation for further understanding how TBEV evade innate immunity and a potential viral target for intervention.


Assuntos
Vírus da Encefalite Transmitidos por Carrapatos/fisiologia , Interferon Tipo I/antagonistas & inibidores , Proteínas não Estruturais Virais/metabolismo , Linhagem Celular , Proteína DEAD-box 58/metabolismo , Vírus da Encefalite Transmitidos por Carrapatos/metabolismo , Humanos , Fatores Reguladores de Interferon/antagonistas & inibidores , Fatores Reguladores de Interferon/genética , Fatores Reguladores de Interferon/metabolismo , Interferon Tipo I/metabolismo , Helicase IFIH1 Induzida por Interferon/metabolismo , Fator Gênico 3 Estimulado por Interferon, Subunidade gama/metabolismo , Interferon beta/genética , Interferon beta/metabolismo , Lisina/metabolismo , Fosforilação , Domínios e Motivos de Interação entre Proteínas , Multimerização Proteica , Receptores Imunológicos , Fator de Transcrição STAT1/metabolismo , Fator de Transcrição STAT2/metabolismo , Transdução de Sinais , Ubiquitinação , Regulação para Cima , Domínios de Homologia de src
16.
Hepatology ; 72(5): 1569-1585, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32103509

RESUMO

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.


Assuntos
Apolipoproteínas E/metabolismo , Fígado Gorduroso/metabolismo , Lipase/metabolismo , Fígado/patologia , Lisofosfolipase/metabolismo , Animais , Apolipoproteínas E/genética , Linhagem Celular Tumoral , Retículo Endoplasmático/patologia , Fígado Gorduroso/sangue , Fígado Gorduroso/diagnóstico , Fígado Gorduroso/cirurgia , Feminino , Técnicas de Silenciamento de Genes , Células HEK293 , Humanos , Lipase/genética , Metabolismo dos Lipídeos , Lipoproteínas VLDL/sangue , Lipoproteínas VLDL/metabolismo , Fígado/cirurgia , Lisofosfolipase/genética , Masculino , Camundongos , Camundongos Knockout para ApoE , Complexo de Endopeptidases do Proteassoma/metabolismo , Estabilidade Proteica , Proteólise , Índice de Gravidade de Doença , Triglicerídeos/sangue , Triglicerídeos/metabolismo , Ubiquitinação
17.
Clin Sci (Lond) ; 134(5): 529-542, 2020 03 13.
Artigo em Inglês | MEDLINE | ID: mdl-32100852

RESUMO

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.


Assuntos
Exocitose , Hepatite C Crônica/metabolismo , Secreção de Insulina , Células Secretoras de Insulina/metabolismo , Proteína Quinase 13 Ativada por Mitógeno/metabolismo , Animais , Linhagem Celular Tumoral , Diabetes Mellitus Tipo 2/sangue , Diabetes Mellitus Tipo 2/metabolismo , Intolerância à Glucose/sangue , Intolerância à Glucose/metabolismo , Intolerância à Glucose/virologia , Hepatite C Crônica/virologia , Humanos , Insulina/metabolismo , Células Secretoras de Insulina/virologia , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Camundongos Transgênicos , Proteína Quinase C/metabolismo
18.
ACS Chem Biol ; 15(5): 1232-1241, 2020 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-31972076

RESUMO

Upon sensing pathogen-associated patterns and secreting interferons (IFNs) into the environment, host cells perceive extracellular type I IFNs by the IFNα/ß receptors IFNAR1 and IFNAR2 to stimulate downstream innate immune signaling cascades. Through the use of chemical probes, we demonstrated that IFNAR2 facilitates hepatitis C virus (HCV) entry. Silencing of IFNAR2 significantly attenuated HCV proliferation. IFNAR2 binds infectious HCV virions through a direct interaction of its D2 domain with the C-terminal end of apolipoprotein E (apoE) on the viral envelope and facilitates virus entry into host cells. The antibody against the IFNAR2 D2 domain attenuates IFNAR2-apoE interaction and impairs HCV infection. The recombinant IFNAR2 protein and the chemical probe potently inhibit major HCV genotypes in various human liver cells in vitro. Moreover, the impact of a chemical probe on HCV genotype 2a is also documented in immune-compromised humanized transgenic mice. Our results not only expand the understanding of the biology of HCV entry and the virus-host relationship but also reveal a new target for the development of anti-HCV entry inhibitors.


Assuntos
Antivirais/metabolismo , Hepacivirus/metabolismo , Hepatite C/metabolismo , Receptor de Interferon alfa e beta/metabolismo , Internalização do Vírus/efeitos dos fármacos , Animais , Apolipoproteínas E/metabolismo , Desenho de Fármacos , Genótipo , Hepatócitos/citologia , Hepatócitos/metabolismo , Humanos , Camundongos Transgênicos , Ligação Proteica , Proteínas Recombinantes/metabolismo , Transdução de Sinais , Envelope Viral/metabolismo
19.
Antiviral Res ; 172: 104619, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31600533

RESUMO

Hepatitis B virus (HBV) infection remains an important public health problem worldwide. Covalently closed circular DNA (cccDNA) exhibits as an individual minichromosome and is the molecular basis of HBV infection persistence and antiviral treatment failure. In the current study, we demonstrated that histone deacetylase 11 (HDAC11) inhibits HBV transcription and replication in HBV-transfected Huh7 cells. By using an HBV in vitro infection system, HDAC11 was found to affect the transcriptional activity of cccDNA but did not affect cccDNA production. Chromatin immunoprecipitation (ChIP) assays were utilized to analyze the epigenetic modifications of cccDNA. The results show that HDAC11 specifically reduced the acetylation level of cccDNA-bound histone H3 but did not affect that of histone H4. Furthermore, HDAC11 overexpression decreased the levels of cccDNA-bound acetylated H3K9 (H3K9ac) and H3K27 (H3K27ac). In conclusion, HDAC11 restricts HBV replication through epigenetic repression of cccDNA transcription. These findings reveal the novel role of HDAC11 in HBV infection, further broadening our knowledge regarding the functions of HDAC11 and the roles of HDACs in the epigenetic regulation of HBV cccDNA.


Assuntos
Repressão Epigenética , Vírus da Hepatite B/genética , Histona Desacetilases/metabolismo , Replicação Viral/genética , Linhagem Celular , DNA Circular/metabolismo , DNA Viral/metabolismo , Epigênese Genética , Hepatite B/metabolismo , Hepatite B/virologia , Histonas/metabolismo , Humanos , Transcrição Gênica
20.
Metabolism ; 100: 153954, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31400386

RESUMO

BACKGROUND: Chronic hepatitis C virus (HCV) infection can predispose the host to metabolic abnormalities. The orphan nuclear receptor small heterodimer partner (SHP; NR0B2) has been identified as a key transcriptional regulatory factor of genes involved in diverse metabolic pathways. The protective effects of SHP against HCV-induced hepatic fibrosis have been reported. However, the exact mechanisms of its role on metabolism are largely unknown. We investigated the role of hepatic SHP in regulating glucose and lipid homeostasis, particularly in the metabolic stress response caused by HCV infection. MATERIALS AND METHODS: Gluconeogenesis and lipogenesis levels and SHP expression were measured in HCV-infected cells, as well as in liver samples from HCV-infected patients and persistently HCV-infected mice. RESULTS: We demonstrated that SHP is involved in gluconeogenesis via the acetylation of the Forkhead box O (FoxO) family transcription factor FoxO1, which is mediated by histone deacetylase 9 (HDAC9). Meanwhile, SHP regulates lipogenesis in the liver via suppressing the induction of sterol regulatory element-binding protein-1c (SREBP-1c) expression by the SUMOylation of Liver X receptor α (LXRα) at the SREBP-1c promoter. In particular, SHP can be strongly reduced upon stimulation, such as by HCV infection. The SHP expression levels were decreased in the livers from the CHC patients and persistently HCV-infected mice, and a negative correlation was observed between the SHP expression levels and gluconeogenic or lipogenic activities, emphasizing the clinical relevance of these results. CONCLUSIONS: Our results suggest that SHP is involved in HCV-induced abnormal glucose and lipid homeostasis and that SHP could be a major target for therapeutic interventions targeting HCV-associated metabolic diseases.


Assuntos
Glucose/metabolismo , Hepatite C Crônica/metabolismo , Homeostase , Metabolismo dos Lipídeos , Receptores Citoplasmáticos e Nucleares/metabolismo , Acetilação , Animais , Linhagem Celular Tumoral , Gluconeogênese , Humanos , Lipogênese , Camundongos , Camundongos Transgênicos , Regiões Promotoras Genéticas , Proteína de Ligação a Elemento Regulador de Esterol 1/genética , Proteína de Ligação a Elemento Regulador de Esterol 1/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...