Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
1.
PLoS Pathog ; 19(6): e1011443, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37327222

RESUMEN

The host always employs various ways to defend against viral infection and spread. However, viruses have evolved their own effective strategies, such as inhibition of RNA translation of the antiviral effectors, to destroy the host's defense barriers. Protein synthesis, commonly controlled by the α-subunit of eukaryotic translation initiation factor 2 (eIF2α), is a basic cellular biological process among all species. In response to viral infection, in addition to inducing the transcription of antiviral cytokines by innate immunity, infected cells also inhibit the RNA translation of antiviral factors by activating the protein kinase R (PKR)-eIF2α signaling pathway. Regulation of innate immunity has been well studied; however, regulation of the PKR-eIF2α signaling pathway remains unclear. In this study, we found that the E3 ligase TRIM21 negatively regulates the PKR-eIF2α signaling pathway. Mechanistically, TRIM21 interacts with the PKR phosphatase PP1α and promotes K6-linked polyubiquitination of PP1α. Ubiquitinated PP1α augments its interaction with PKR, causing PKR dephosphorylation and subsequent translational inhibition release. Furthermore, TRIM21 can constitutively restrict viral infection by reversing PKR-dependent translational inhibition of various previously known and unknown antiviral factors. Our study highlights a previously undiscovered role of TRIM21 in regulating translation, which will provide new insights into the host antiviral response and novel targets for the treatment of translation-associated diseases in the clinic.


Asunto(s)
ARN , Virosis , Humanos , ARN/metabolismo , eIF-2 Quinasa/metabolismo , Procesamiento Proteico-Postraduccional , Fosforilación , Antivirales , Factor 2 Eucariótico de Iniciación/genética , Factor 2 Eucariótico de Iniciación/metabolismo , Replicación Viral
2.
J Virol ; 97(12): e0151323, 2023 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-38032198

RESUMEN

IMPORTANCE: The precise regulation of the innate immune response is essential for the maintenance of homeostasis. MAVS and STING play key roles in immune signaling pathways activated by RNA and DNA viruses, respectively. Here, we showed that DHCR24 impaired the antiviral response by targeting MAVS and STING. Notably, DHCR24 interacts with MAVS and STING and inhibits TRIM21-triggered K27-linked ubiquitination of MAVS and AMFR-triggered K27-linked ubiquitination of STING, restraining the activation of MAVS and STING, respectively. Together, this study elucidates how one cholesterol key enzyme orchestrates two antiviral signal transduction pathways.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Inmunidad Innata , Proteínas de la Membrana , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH , Animales , Humanos , Ratones , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Hidroxiesteroides , Proteínas de la Membrana/metabolismo , Oxidorreductasas , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH/genética , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH/metabolismo , Ubiquitinación , Línea Celular
3.
J Immunol ; 209(10): 1987-1998, 2022 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-36426955

RESUMEN

Pyroptosis is a form of regulated cell death mediated by the gasdermin protein family. During virus infection, cell pyroptosis restricts viral replication. The mechanisms of the tripartite motif (TRIM) protein family and IFN-stimulated genes (ISGs) against viruses have been studied. The role of TRIMs and ISGs in pyroptosis remains unclear. In this study, we show that TRIM21 interacts with ISG12a in viral infection and facilitates its translocation into the mitochondria by promoting its ubiquitination, thereby causing caspase 3 activation. Gasdermin E (GSDME) is specifically cleaved by caspase 3 upon viral infection, releasing the GSDME N-terminal domain, perforating the cell membrane, and causing cell pyroptosis. Our study uncovers a new mechanism of TRIM21 and ISG12a in regulating virus-induced cell pyroptosis.


Asunto(s)
Piroptosis , Virus , Piroptosis/fisiología , Caspasa 3/metabolismo , Ubiquitinación , Muerte Celular , Proteínas de Motivos Tripartitos/metabolismo
4.
J Virol ; 96(7): e0000122, 2022 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-35254105

RESUMEN

The induction of interferons (IFNs) plays an important role in the elimination of invading pathogens. Heat shock binding protein 21 (HBP21), first known as a molecular chaperone of HSP70, is involved in tumor development. Heat shock binding proteins have been shown to regulate diverse biological processes, such as cell cycle, kinetochore localization, transcription, and cilium formation. Their role in antimicrobial immunity remains unknown. Here, we found that HBP21 drives a positive feedback loop to promote IRF3-mediated IFN production triggered by viral infection. HBP21 deficiency significantly impaired the virus-induced production of IFN and resulted in greater susceptibility to viral infection both in vitro and in vivo. Mechanistically, HBP21 interacted with IRF3 and promoted the formation of a TBK1-IRF3 complex. Moreover, HBP21 abolished the interaction between PP2A and IRF3 to repress the dephosphorylation of IRF3. Analysis of HBP21 protein structure further confirmed that HBP21 promotes the activation of IRF3 by depressing the dephosphorylation of IRF3 by PP2A. Further study demonstrated that virus-induced phosphorylation of Ser85 and Ser153 of HBP21 itself is important for the phosphorylation and dimerization of IRF3. Our study identifies HBP21 as a new positive regulator of innate antiviral response, which adds novel insight into activation of IRF3 controlled by multiple networks that specify behavior of tumors and immunity. IMPORTANCE The innate immune system is the first-line host defense against microbial pathogen invasion. The physiological functions of molecular chaperones, involving cell differentiation, migration, proliferation and inflammation, have been intensively studied. HBP21 as a molecular chaperone is critical for tumor development. Tumor is related to immunity. Whether HBP21 regulates immunity remains unknown. Here, we found that HBP21 promotes innate immunity response by dual regulation of IRF3. HBP21 interacts with IRF3 and promotes the formation of a TBK1-IRF3 complex. Moreover, HBP21 disturbs the interaction between PP2A and IRF3 to depress the dephosphorylation of IRF3. Analysis of HBP21 protein structure confirms that HBP21 promotes the activation of IRF3 by blocking the dephosphorylation of IRF3 by PP2A. Interestingly, virus-induced Ser85 and Ser153 phosphorylation of HBP21 is important for IRF3 activation. Our findings add to the known novel immunological functions of molecular chaperones and provide new insights into the regulation of innate immunity.


Asunto(s)
Inmunidad Innata , Chaperonas Moleculares , Virosis , Humanos , Inmunidad Innata/genética , Inmunidad Innata/inmunología , Factor 3 Regulador del Interferón/metabolismo , Chaperonas Moleculares/metabolismo , Fosforilación , Virosis/inmunología
5.
J Virol ; 96(6): e0217521, 2022 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-35107381

RESUMEN

REC8 meiotic recombination protein (REC8) is a member of structural maintenance of chromosome (SMC) protein partners, which play an important role in meiosis, antitumor activity, and sperm formation. As the adaptor proteins of RIG-I-like receptor (RLR) signaling and cyclic GMP-AMP synthase (cGAS)-DNA signaling, the activity and stability of MAVS (mitochondrial antiviral signaling protein; also known as VISA, Cardif, and IPS-1) and STING (stimulator of interferon genes; also known as MITA) are critical for innate immunity. Here, we report that REC8 interacts with MAVS and STING and inhibits their ubiquitination and subsequent degradation, thereby promoting innate antiviral signaling. REC8 is upregulated through the JAK-STAT signaling pathway during viral infection. Knockdown of REC8 impairs the innate immune responses against vesicular stomatitis virus (VSV), Newcastle disease virus (NDV), and herpes simplex virus (HSV). Mechanistically, during infection with viruses, the SUMOylated REC8 is transferred from the nucleus to the cytoplasm and then interacts with MAVS and STING to inhibit their K48-linked ubiquitination triggered by RNF5. Moreover, REC8 promotes the recruitment of TBK1 to MAVS and STING. Thus, REC8 functions as a positive modulator of innate immunity. Our work highlights a previously undocumented role of meiosis-associated protein REC8 in regulating innate immunity. IMPORTANCE The innate immune response is crucial for the host to resist the invasion of viruses and other pathogens. STING and MAVS play a critical role in the innate immune response to DNA and RNA viral infection, respectively. In this study, REC8 promoted the innate immune response by targeting STING and MAVS. Notably, REC8 interacts with MAVS and STING in the cytoplasm and inhibits K48-linked ubiquitination of MAVS and STING triggered by RNF5, stabilizing MAVS and STING protein to promote innate immunity and gradually inhibiting viral infection. Our study provides a new insight for the study of antiviral innate immunity.


Asunto(s)
Proteínas de Ciclo Celular , Inmunidad Innata , Virosis , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Antivirales , Proteínas de Ciclo Celular/inmunología , Proteínas de la Membrana/metabolismo , Virus de la Enfermedad de Newcastle , Simplexvirus , Ubiquitinación , Virus de la Estomatitis Vesicular Indiana , Virosis/inmunología
6.
Apoptosis ; 27(7-8): 590-605, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35717659

RESUMEN

This study aimed to investigate the role and regulatory mechanism of RNF126 in nasopharyngeal carcinoma. Firstly, the expression and prognosis of RNF126 were analyzed by TCGA database. The expression of RNF126 was further verified by NPC tissue samples and cells. An ectopic xenograft model was constructed to verify the regulatory role of RNF126 in NPC tumor progression. The regulatory effect of RNF126 on macrophage polarization and migration was verified by co-culture of tumor cells and THP-1 cells. The role of RNF126 in tumor exosomes involved in intercellular communication was further verified by nanoparticle tracking technology, western blotting and immunofluorescence assays. QRT-PCR, half-life assay and WB assay were used to verify the regulatory effect of RNF126 on PTEN ubiquitination and PI3K/AKT pathway. Finally, an in vivo assay was used to verify the regulation of exosomes on tumor growth and metastasis. In summary, we found for the first time that tumor-derived exosomal PTEN degrades PTEN through ubiquitination to regulate the tumor immune microenvironment and promote NPC growth and metastasis. These results provide the basis for the screening of early markers of NPC and targeted therapy.


Asunto(s)
Exosomas , MicroARNs , Neoplasias Nasofaríngeas , Apoptosis , Línea Celular Tumoral , Proliferación Celular , Exosomas/genética , Exosomas/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , MicroARNs/metabolismo , Carcinoma Nasofaríngeo/genética , Carcinoma Nasofaríngeo/metabolismo , Carcinoma Nasofaríngeo/patología , Neoplasias Nasofaríngeas/genética , Neoplasias Nasofaríngeas/metabolismo , Neoplasias Nasofaríngeas/patología , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/metabolismo , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Microambiente Tumoral/genética , Ubiquitina-Proteína Ligasas , Ubiquitinación
7.
J Am Chem Soc ; 143(46): 19317-19329, 2021 11 24.
Artículo en Inglés | MEDLINE | ID: mdl-34762804

RESUMEN

GFP-like fluorescent proteins and their molecular mimics have revolutionized bioimaging research, but their emissions are largely limited in the visible to far-red region, hampering the in vivo applications in intact animals. Herein, we structurally modulate GFP-like chromophores using a donor-acceptor-acceptor (D-A-A') molecular configuration to discover a set of novel fluorogenic derivatives with infrared-shifted spectra. These chromophores can be fluorescently elicited by their specific interaction with G-quadruplex (G4), a unique noncanonical nucleic acid secondary structure, via inhibition of the chromophores' twisted-intramolecular charge transfer. This feature allows us to create, for the first time, FP mimics with tunable emission in the near-infrared (NIR) region (Emmax = 664-705 nm), namely, infrared G-quadruplex mimics of FPs (igMFP). Compared with their FP counterparts, igMFPs exhibit remarkably higher quantum yields, larger Stokes shift, and better photostability. In a proof-of-concept application using pathogen-related G4s as the target, we exploited igMFPs to directly visualize native hepatitis C virus (HCV) RNA genome in living cells via their in situ formation by the chromophore-bound viral G4 structure in the HCV core gene. Furthermore, igMFPs are capable of high contrast HCV RNA imaging in living mice bearing a HCV RNA-presenting mini-organ, providing the first application of FP mimics in whole-animal imaging.


Asunto(s)
Fluorescencia , Colorantes Fluorescentes/química , Proteínas Luminiscentes/química , Ácidos Nucleicos/química , ARN Viral/análisis , Animales , Línea Celular Tumoral , Colorantes Fluorescentes/síntesis química , Hepacivirus/genética , Humanos , Rayos Infrarrojos , Proteínas Luminiscentes/síntesis química , Ratones , ARN Viral/genética , Espectrometría de Fluorescencia
8.
J Virol ; 94(22)2020 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-32878885

RESUMEN

Innate immunity is an essential way for host cells to resist viral infection through the production of interferons (IFNs) and proinflammatory cytokines. Interferon regulatory factor 3 (IRF3) plays a critical role in the innate immune response to viral infection. However, the role of IRF1 in innate immunity remains largely unknown. In this study, we found that IRF1 is upregulated through the IFN/JAK/STAT signaling pathway upon viral infection. The silencing of IRF1 attenuates the innate immune response to viral infection. IRF1 interacts with IRF3 and augments the activation of IRF3 by blocking the interaction between IRF3 and protein phosphatase 2A (PP2A). The DNA binding domain (DBD) of IRF1 is the key functional domain for its interaction with IRF3. Overall, our study reveals a novel mechanism by which IRF1 promotes the innate immune response to viral infection by enhancing the activation of IRF3, thereby inhibiting viral infection.IMPORTANCE The activation of innate immunity is essential for host cells to restrict the spread of invading viruses and other pathogens. IRF3 plays a critical role in the innate immune response to RNA viral infection. However, whether IRF1 plays a role in innate immunity is unclear. In this study, we demonstrated that IRF1 promotes the innate immune response to viral infection. IRF1 is induced by viral infection. Notably, IRF1 targets and augments the phosphorylation of IRF3 by blocking the interaction between IRF3 and PP2A, leading to the upregulation of innate immunity. Collectively, the results of our study provide new insight into the regulatory mechanism of IFN signaling and uncover the role of IRF1 in the positive regulation of the innate immune response to viral infection.


Asunto(s)
Inmunidad Innata/inmunología , Factor 1 Regulador del Interferón/inmunología , Factor 3 Regulador del Interferón/metabolismo , Virosis/inmunología , Línea Celular , Proteínas de Unión al ADN , Células HEK293 , Humanos , Factor 1 Regulador del Interferón/metabolismo , Fosforilación , Infecciones por Virus ARN/inmunología , Virus ARN , Transducción de Señal/inmunología
9.
Neurol Sci ; 42(9): 3857-3863, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-33523317

RESUMEN

Intravenous immunoglobulin (IVIg) has been used for neuromyelitis optica spectrum disorder (NMOSD) patients to prevent relapses in several studies. However, efficacy of the rescue treatment of IVIG was just assessed in a small sample research. The aim of this study is to investigate the efficacy of IVIG in NMOSD as a rescue treatment and whether it could reduce the relapse rate. We retrospectively reviewed patients with NMOSD in the First and Second Affiliated Hospital of Wenzhou Medical University. Clinical parameters were extracted from the medical records, such as expanded disability scale score (EDSS) and time to next relapse. Thirty-one events of 20 NMOSD patients were included in the intravenous methylprednisolone (IVMT) + IVIG group and 72 events of 39 patients in the IVMT group. IVMT therapy combined with IVIG could improve the neurological disability when discharged (p < 0.001), whereas patients first attacked did not show a similar trend. Patients who were treated with IVMT + IVIG (17.39 ± 2.75 months) show a longer time to next relapse compared to patients who were treated with IVMT (9.50 ± 0.79 months) (log rank test p = 0.002), especially in relapsed patients or anti-aquaporin-4 antibody (AQP4-Ab) seropositive patients. IVIG might be helpful for NMOSD patients as the rescue treatment and might bring a longer remission, especially for patients with relapse and AQP4-ab seropositive patients.


Asunto(s)
Inmunoglobulinas Intravenosas , Neuromielitis Óptica , Acuaporina 4 , Autoanticuerpos , Humanos , Inmunoglobulinas Intravenosas/uso terapéutico , Metilprednisolona/uso terapéutico , Neuromielitis Óptica/tratamiento farmacológico , Recurrencia , Estudios Retrospectivos
10.
J Am Chem Soc ; 141(13): 5182-5191, 2019 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-30860368

RESUMEN

RNA viruses represent a major global health threat, and the visualization of their RNA genome in infected cells is essential for virological research and clinical diagnosis. Due to the lack of chemical toolkits for the live-cell imaging of viral RNA genomes, especially native viral genomes without labeling and genetic modification, studies on native virus infection at the single-live-cell level are challenging. Herein, taking hepatitis C virus (HCV) as a representative RNA virus, we propose that the innate noncanonical G-quadruplex (G4) structure of viral RNA can serve as a specific imaging target and report a new benzothiazole-based G4-targeted fluorescence light-up probe, ThT-NE, for the direct visualization of the native RNA genome of HCV in living host cells. We demonstrate the use of the ThT-NE probe for several previously intractable applications, including the sensitive detection of individual virus-infected cells by small-molecule staining, real-time monitoring of the subcellular distribution of the viral RNA genome in live cells, and continuous live-cell tracking of the infection and propagation of clinically isolated native HCV. The fluorogenic-probe-based viral RNA light-up system opens up a promising chemical strategy for cutting-edge live-cell viral analysis, providing a potentially powerful tool for viral biology, medical diagnosis, and drug development.


Asunto(s)
Colorantes Fluorescentes/análisis , Genoma Viral/genética , Hepacivirus/genética , Hepacivirus/aislamiento & purificación , Hepatitis C/patología , Hepatitis C/virología , Imagen Óptica , ARN Viral/análisis , Línea Celular Tumoral , Supervivencia Celular , Colorantes Fluorescentes/síntesis química , Colorantes Fluorescentes/química , G-Cuádruplex , Hepatitis C/diagnóstico por imagen , Humanos , Estructura Molecular , ARN Viral/genética
11.
J Virol ; 92(14)2018 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-29743353

RESUMEN

Human innate immunity responds to viral infection by activating the production of interferons (IFNs) and proinflammatory cytokines. The mitochondrial adaptor molecule MAVS plays a critical role in innate immune response to viral infection. In this study, we show that TRIM21 (tripartite motif-containing protein 21) interacts with MAVS to positively regulate innate immunity. Under viral infection, TRIM21 is upregulated through the IFN/JAK/STAT signaling pathway. Knockdown of TRIM21 dramatically impairs innate immune response to viral infection. Moreover, TRIM21 interacts with MAVS and catalyzes its K27-linked polyubiquitination, thereby promoting the recruitment of TBK1 to MAVS. Specifically, the PRY-SPRY domain of TRIM21 is the key domain for its interaction with MAVS, while the RING domain of TRIM21 facilitates the polyubiquitination chains of MAVS. In addition, the MAVS-mediated innate immune response is enhanced by both the PRY-SPRY and RING domains of TRIM21. Mutation analyses of all the lysine residues of MAVS further revealed that Lys325 of MAVS is catalyzed by TRIM21 for the K27-linked polyubiquitination. Overall, this study reveals a novel mechanism by which TRIM21 promotes the K27-linked polyubiquitination of MAVS to positively regulate innate immune response, thereby inhibiting viral infection.IMPORTANCE Activation of innate immunity is essential for host cells to restrict the spread of invading viruses and other pathogens. MAVS plays a critical role in innate immune response to RNA viral infection. In this study, we demonstrated that TRIM21 targets MAVS to positively regulate innate immunity. Notably, TRIM21 targets and catalyzes K27-linked polyubiquitination of MAVS and then promotes the recruitment of TBK1 to MAVS, leading to upregulation of innate immunity. Our study outlines a novel mechanism by which the IFN signaling pathway blocks RNA virus to escape immune elimination.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Inmunidad Innata/inmunología , Proteínas Serina-Treonina Quinasas/metabolismo , Infecciones por Virus ARN/inmunología , Virus ARN/inmunología , Ribonucleoproteínas/metabolismo , Ubiquitina/metabolismo , Interacciones Huésped-Patógeno , Humanos , Lisina/química , Infecciones por Virus ARN/metabolismo , Infecciones por Virus ARN/virología , Transducción de Señal , Células Tumorales Cultivadas , Ubiquitinación
12.
J Virol ; 92(17)2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-29899107

RESUMEN

Emerging evidence indicates that long noncoding RNAs (lncRNAs) regulate various biological processes, especially innate and adaptive immunity. However, the relationship between lncRNAs and the interferon (IFN) pathway remains largely unknown. Here, we report that lncRNA ITPRIP-1 (lncITPRIP-1) is involved in viral infection and plays a crucial role in the virus-triggered IFN signaling pathway through the targeting of melanoma differentiation-associated gene 5 (MDA5). LncITPRIP-1 can be induced by viral infection, which is not entirely dependent on the IFN signal. Besides, there is no coding potential found in the lncITPRIP-1 transcript. LncITPRIP-1 binds to the C terminus of MDA5, and it possesses the ability to boost the oligomerization of both the full length and the 2 caspase activation and recruitment domains of MDA5 in a K63-linked polyubiquitination-independent manner. Amazingly, we also found that MDA5 can suppress hepatitis C virus (HCV) replication independently of IFN signaling through its C-terminal-deficient domain bound to viral RNA, in which lncITPRIP-1 plays a role as an assistant. In addition, the expression of lncITPRIP-1 is highly consistent with MDA5 expression, indicating that lncITPRIP-1 may function as a cofactor of MDA5. All the data suggest that lncITPRIP-1 enhances the innate immune response to viral infection through the promotion of oligomerization and activation of MDA5. Our study discovers the first lncRNA ITPRIP-1 involved in MDA5 activation.IMPORTANCE Hepatitis C virus infection is a global health issue, and there is still no available vaccine, which makes it urgent to reveal the underlying mechanisms of HCV and host factors. Although RIG-I has been recognized as the leading cytoplasmic sensor against HCV for a long time, recent findings that MDA5 regulates the IFN response to HCV have emerged. Our work validates the significant role of MDA5 in IFN signaling and HCV infection and proposes the first lncRNA inhibiting HCV replication by promoting the activation of MDA5 and mediating the association between MDA5 and HCV RNA, the study of which may shed light on the MDA5 function and treatment for hepatitis C patients. Our suggested model of how lncITPRIP-1 orchestrates signal transduction for IFN production illustrates the essential role of lncRNAs in virus elimination.


Asunto(s)
Inmunidad Innata/fisiología , Helicasa Inducida por Interferón IFIH1/genética , Interferones/inmunología , Proteínas de la Membrana/fisiología , ARN Largo no Codificante/fisiología , Transducción de Señal/inmunología , Línea Celular , Regulación de la Expresión Génica/inmunología , Hepacivirus/genética , Hepacivirus/inmunología , Hepatitis C/inmunología , Hepatitis C/virología , Interacciones Huésped-Patógeno/inmunología , Humanos , Inmunidad Innata/inmunología , Helicasa Inducida por Interferón IFIH1/fisiología , Interferones/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/inmunología , ARN Viral/genética , Transducción de Señal/genética
13.
J Virol ; 91(23)2017 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-28956771

RESUMEN

Activation of innate immunity is essential for host cells to restrict the spread of invading viruses and other pathogens. However, attenuation or termination of signaling is also necessary for preventing immune-mediated tissue damage and spontaneous autoimmunity. Here, we identify nucleotide binding oligomerization domain (NOD)-like receptor X1 (NLRX1) as a negative regulator of the mitochondrial antiviral signaling protein (MAVS)-mediated signaling pathway during hepatitis C virus (HCV) infection. The depletion of NLRX1 enhances the HCV-triggered activation of interferon (IFN) signaling and causes the suppression of HCV propagation in hepatocytes. NLRX1, a HCV-inducible protein, interacts with MAVS and mediates the K48-linked polyubiquitination and subsequent degradation of MAVS via the proteasomal pathway. Moreover, poly(rC) binding protein 2 (PCBP2) interacts with NLRX1 to participate in the NLRX1-induced degradation of MAVS and the inhibition of antiviral responses during HCV infection. Mutagenic analyses further revealed that the NOD of NLRX1 is essential for NLRX1 to interact with PCBP2 and subsequently induce MAVS degradation. Our study unlocks a key mechanism of the fine-tuning of innate immunity by which NLRX1 restrains the retinoic acid-inducible gene I-like receptor (RLR)-MAVS signaling cascade by recruiting PCBP2 to MAVS for inducing MAVS degradation through the proteasomal pathway. NLRX1, a negative regulator of innate immunity, is a pivotal host factor for HCV to establish persistent infection.IMPORTANCE Innate immunity needs to be tightly regulated to maximize the antiviral response and minimize immune-mediated pathology, but the underlying mechanisms are poorly understood. In this study, we report that NLRX1 is a proviral host factor for HCV infection and functions as a negative regulator of the HCV-triggered innate immune response. NLRX1 recruits PCBP2 to MAVS and induces the K48-linked polyubiquitination and degradation of MAVS, leading to the negative regulation of the IFN signaling pathway and promoting HCV infection. Overall, this study provides intriguing insights into how innate immunity is regulated during viral infection.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Hepacivirus/inmunología , Hepatitis C/inmunología , Proteínas Mitocondriales/química , Proteínas Mitocondriales/metabolismo , Proteínas de Unión al ARN/metabolismo , Sitios de Unión , Línea Celular , Células HEK293 , Hepacivirus/fisiología , Humanos , Inmunidad Innata , Proteínas Mitocondriales/genética , Mutación , Unión Proteica , Dominios Proteicos , Proteolisis , Transducción de Señal , Replicación Viral
14.
J Virol ; 90(15): 6832-45, 2016 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-27194766

RESUMEN

UNLABELLED: Interferons (IFNs) restrict various kinds of viral infection via induction of hundreds of IFN-stimulated genes (ISGs), while the functions of the majority of ISGs are broadly unclear. Here, we show that a high-IFN-inducible gene, ISG12a (also known as IFI27), exhibits a nonapoptotic antiviral effect on hepatitis C virus (HCV) infection. Viral NS5A protein is targeted specifically by ISG12a, which mediates NS5A degradation via a ubiquitination-dependent proteasomal pathway. K374R mutation in NS5A domain III abrogates ISG12a-induced ubiquitination and degradation of NS5A. S-phase kinase-associated protein 2 (SKP2) is identified as an ubiquitin E3 ligase for NS5A. ISG12a functions as a crucial adaptor that promotes SKP2 to interact with and degrade viral protein. Moreover, the antiviral effect of ISG12a is dependent on the E3 ligase activity of SKP2. These findings uncover an intriguing mechanism by which ISG12a restricts viral infection and provide clues for understanding the actions of innate immunity. IMPORTANCE: Upon virus invasion, IFNs induce numerous ISGs to control viral spread, while the functions of the majority of ISGs are broadly unclear. The present study shows a novel antiviral mechanism of ISGs and elucidated that ISG12a recruits an E3 ligase, SKP2, for ubiquitination and degradation of viral protein and restricts viral infection. These findings provide important insights into exploring the working principles of innate immunity.


Asunto(s)
Antivirales/farmacología , Hepacivirus/efectos de los fármacos , Hepatitis C/prevención & control , Proteínas de la Membrana/farmacología , Transducción de Señal/efectos de los fármacos , Ubiquitina/metabolismo , Proteínas no Estructurales Virales/metabolismo , Células Cultivadas , Hepatitis C/metabolismo , Hepatitis C/virología , Hepatocitos/citología , Hepatocitos/efectos de los fármacos , Hepatocitos/virología , Humanos , Proteolisis , Ubiquitinación
15.
Neurol Sci ; 38(10): 1859-1863, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28779361

RESUMEN

Neutrophil to lymphocyte ratio (NLR) was introduced to assess the activity in autoimmune diseases. Neuromyelitis optica spectrum disorder (NMOSD) has been defined as a chronic inflammatory disease with a course of relapse-remission. Therefore, the relationship between NLR and NMOSD was assessed in this article. Data of NMOSD patients was extracted. NLR is calculated as the absolute count of neutrophil divided by the absolute count of lymphocytes. Correlations between NLR and characteristics of NMOSD patients were evaluated. Effect of treatments on NLR was also analyzed. Increased level of NLR was observed in patients with NMOSD compared healthy individuals (p < 0.001); moreover, patients who were experiencing acute attack had a higher level of NLR compared with those who in remission (p < 0.001). NLR was correlated with RDW (r = 0.288, p = 0.021), ΔEDSS (r = 0.301, p = 0.016). NLR may be a helpful marker to assess the disease activity of NMOSD. Meanwhile, NLR may reflect the aggravated degree of neurological disability.


Asunto(s)
Linfocitos , Neuromielitis Óptica/sangre , Neutrófilos , Adulto , Femenino , Humanos , Terapia de Inmunosupresión , Recuento de Leucocitos , Masculino , Neuromielitis Óptica/terapia
16.
Proc Natl Acad Sci U S A ; 111(13): E1264-73, 2014 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-24616513

RESUMEN

The absence of a robust cell culture system for hepatitis B virus (HBV) and hepatitis C virus (HCV) infection has limited the analysis of the virus lifecycle and drug discovery. We have established a hepatoma cell line, HLCZ01, the first cell line, to the authors' knowledge, supporting the entire lifecycle of both HBV and HCV. HBV surface antigen (HBsAg)-positive particles can be observed in the supernatant and the lumen of the endoplasmic reticulum of the cells via electron microscopy. Interestingly, HBV and HCV clinical isolates propagate in HLCZ01 cells. Both viruses replicate in the cells without evidence of overt interference. HBV and HCV entry are blocked by antibodies against HBsAg and human CD81, respectively, and the replication of HBV and HCV is inhibited by antivirals. HLCZ01 cells mount an innate immune response to virus infection. The cell line provides a powerful tool for exploring the mechanisms of virus entry and replication and the interaction between host and virus, facilitating the development of novel antiviral agents and vaccines.


Asunto(s)
Carcinoma Hepatocelular/virología , Hepacivirus/fisiología , Virus de la Hepatitis B/fisiología , Neoplasias Hepáticas/virología , Replicación Viral/fisiología , Animales , Antivirales/farmacología , Antivirales/uso terapéutico , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Coinfección/tratamiento farmacológico , Coinfección/patología , Coinfección/virología , Hepacivirus/efectos de los fármacos , Hepacivirus/crecimiento & desarrollo , Hepatitis B/tratamiento farmacológico , Hepatitis B/virología , Virus de la Hepatitis B/efectos de los fármacos , Virus de la Hepatitis B/crecimiento & desarrollo , Virus de la Hepatitis B/aislamiento & purificación , Hepatitis C/tratamiento farmacológico , Hepatitis C/virología , Humanos , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/patología , Masculino , Ratones , Ratones Endogámicos NOD , Replicación Viral/efectos de los fármacos
17.
Int J Neurosci ; 127(8): 735-744, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27680606

RESUMEN

Monoclonal-antibody has been used for patients with autoimmune disorders for several years, and efficacy and safety were appreciated for these patients. Neuromyelitis optica specturm disorder (NMOSD) has been defined as an autoimmune demyelination disorder of the central nervous system (CNS) with a course of relapse-remission. Treatment of prevention is important for patients with NMOSD because of the increased disability after several attacks. Multiple factors were involved in the pathogenesis of NMOSD. Currently, targeting specific factor was favored in the research into the treatment for NMOSD. Previous studies reported the efficacy and tolerance in NMOSD for drugs such as rituximab, tocilizumab, and eculizumab. The aim of this article is to review the current monoclonal therapies for NMOSD patients, and also future alternative options.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/uso terapéutico , Neuromielitis Óptica/tratamiento farmacológico , Neuromielitis Óptica/inmunología , Anticuerpos Monoclonales/metabolismo , Anticuerpos Monoclonales Humanizados/inmunología , Anticuerpos Monoclonales Humanizados/metabolismo , Anticuerpos Monoclonales Humanizados/uso terapéutico , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/inmunología , Barrera Hematoencefálica/metabolismo , Predicción , Humanos , Inmunoterapia/métodos , Inmunoterapia/tendencias , Neuromielitis Óptica/metabolismo
18.
Int J Neurosci ; 127(6): 508-515, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27323840

RESUMEN

BACKGROUND: Azathioprine is widely used for neuromyelitis optica spectrum disorder (NMOSD) patients, while a consensus of timing to receive azathioprine has not been proposed. OBJECTIVE: The objective of this paper was to evaluate the efficacy of early access of azathioprine in NMOSD patients. METHODS: We conducted a retrospective review of NMOSD patients based on medical records. Included patients were divided into three groups: group IVMT + AZA, group AZA after IVMT and group IVMT. Time to next relapse was adopted as the endpoint. RESULTS: Patients from group IVMT + AZA had a longer duration of remission compared with patients from group AZA after IVMT ( p = 0.025) and group IVMT ( p < 0.001), and longer duration showed in the group AZA after IVMT when compared with group IVMT ( p = 0.005). We found that older age of initial attack was a risk factor for NMOSD patients (HR: 1.235; p = 0.022), and younger age of receiving treatment was a protect factor (HR: 0.804; p = 0.023). Partial patients have used azathioprine before this study in group IVMT + AZA, result showed there was no significance between the patients who had or had not used azathioprine ( p = 0.299). CONCLUSION: Azathioprine could prolong the duration of remission after treatment, especially given within two weeks after attack. Patients who received azathioprine combined with glucocorticoids had a preferable effect than glucocorticoids alone in the remission.


Asunto(s)
Azatioprina/uso terapéutico , Inmunosupresores/uso terapéutico , Neuromielitis Óptica/tratamiento farmacológico , Enfermedad Aguda , Adulto , Anciano , Antiinflamatorios/administración & dosificación , Femenino , Humanos , Inyecciones Intravenosas , Masculino , Metilprednisolona/administración & dosificación , Persona de Mediana Edad , Estudios Retrospectivos , Estadísticas no Paramétricas , Análisis de Supervivencia , Adulto Joven
19.
J Virol ; 90(5): 2332-44, 2015 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-26656705

RESUMEN

UNLABELLED: High-mobility group box 1 (HMGB1) protein is a highly conserved nuclear protein involved in multiple human diseases, including infectious diseases, immune disorders, metabolic disorders, and cancer. HMGB1 is comprised of two tandem HMG boxes (the A box and the B box) containing DNA-binding domains and an acidic C-terminal peptide. It has been reported that HMGB1 enhances viral replication by binding to viral proteins. However, its role in hepatitis C virus (HCV) replication is unknown. Here, we show that HMGB1 promoted HCV replication but had no effect on HCV translation. RNA immunoprecipitation experiments indicated that the positive strand, not the negative strand, of HCV RNA interacted with HMGB1. HCV infection triggered HMGB1 protein translocation from the nucleus to the cytoplasm, in which it interacted with the HCV genome. Moreover, the A box of HMGB1 is the pivotal domain to interact with stem-loop 4 (SL4) of the HCV 5' untranslated region. Deletion of the HMGB1 A box abrogated the enhancement of HCV replication by HMGB1. Our data suggested that HMGB1 serves as a proviral factor of HCV to facilitate viral replication in hepatocytes by interaction with the HCV genome. IMPORTANCE: Hepatitis C virus (HCV) is a major global health threat, affecting more than 170 million people infection worldwide. These patients are at high risk of developing severe liver diseases such as chronic hepatitis, cirrhosis, and hepatocellular carcinoma. Currently, no vaccine is available. Many host factors may be implicated in the pathogenesis of HCV-related diseases. In this study, we found a novel HCV RNA-binding protein, HMGB1, that promotes HCV RNA replication. Moreover, SL4 in the 5' untranslated region of the HCV genome is the key region for HMGB1 binding, and the A box of HMGB1 protein is the functional domain to interact with HCV RNA and enhance viral replication. HMGB1 appears to play an important role in HCV-related diseases, and further investigation is warranted to elucidate the specific actions of HMGB1 in HCV pathogenesis.


Asunto(s)
Regiones no Traducidas 5' , Proteína HMGB1/metabolismo , Hepacivirus/fisiología , Interacciones Huésped-Patógeno , ARN Viral/metabolismo , Replicación Viral , Línea Celular , Hepatocitos/virología , Humanos , Unión Proteica
20.
J Virol ; 88(4): 1990-9, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24307579

RESUMEN

Hepatitis C virus (HCV) core protein is essential for virus assembly. HCV core protein was expressed and purified. Aptamers against core protein were raised through the selective evolution of ligands by the exponential enrichment approach. Detection of HCV infection by core aptamers and the antiviral activities of aptamers were characterized. The mechanism of their anti-HCV activity was determined. The data showed that selected aptamers against core specifically recognize the recombinant core protein but also can detect serum samples from hepatitis C patients. Aptamers have no effect on HCV RNA replication in the infectious cell culture system. However, the aptamers inhibit the production of infectious virus particles. Beta interferon (IFN-ß) and interferon-stimulated genes (ISGs) are not induced in virally infected hepatocytes by aptamers. Domains I and II of core protein are involved in the inhibition of infectious virus production by the aptamers. V31A within core is the major resistance mutation identified. Further study shows that the aptamers disrupt the localization of core with lipid droplets and NS5A and perturb the association of core protein with viral RNA. The data suggest that aptamers against HCV core protein inhibit infectious virus production by disrupting the localization of core with lipid droplets and NS5A and preventing the association of core protein with viral RNA. The aptamers for core protein may be used to understand the mechanisms of virus assembly. Core-specific aptamers may hold promise for development as early diagnostic reagents and potential therapeutic agents for chronic hepatitis C.


Asunto(s)
Anticuerpos Monoclonales/metabolismo , Aptámeros de Nucleótidos/farmacología , Hepacivirus/genética , Hepacivirus/metabolismo , Proteínas del Núcleo Viral/metabolismo , Virión/efectos de los fármacos , Animales , Aptámeros de Nucleótidos/genética , Aptámeros de Nucleótidos/metabolismo , Western Blotting , Línea Celular , Cartilla de ADN/genética , Ensayo de Inmunoadsorción Enzimática , Técnica del Anticuerpo Fluorescente , Biblioteca de Genes , Hepacivirus/efectos de los fármacos , Humanos , Inmunoprecipitación , Ratones , Plásmidos/genética , Unión Proteica/efectos de los fármacos , Reacción en Cadena en Tiempo Real de la Polimerasa , Técnica SELEX de Producción de Aptámeros , Proteínas del Núcleo Viral/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA