Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Hepatology ; 72(2): 518-534, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-31758709

RESUMO

BACKGROUND AND AIMS: Major vault protein (MVP) is up-regulated during infections with hepatitis B virus (HBV) and hepatitis C virus (HCV). Here, we found that MVP deficiency inhibited hepatocellular carcinoma (HCC) development induced by diethylnitrosamine, hepatitis B X protein, and HCV core. APPROACH AND RESULTS: Forced MVP expression was sufficient to induce HCC in mice. Mechanistic studies demonstrate that the ubiquitin ligase human double minute 2 (HDM2) forms mutual exclusive complexes either with interferon regulatory factor 2 (IRF2) or with p53. In the presence of MVP, HDM2 is liberated from IRF2, leading to the ubiquitination of the tumor suppressor p53. Mouse xenograft models showed that HBV and HCV promote carcinogenesis through MVP induction, resulting in a loss of p53 mediated by HDM2. Analyses of clinical samples from chronic hepatitis B, liver cirrhosis, and HCC revealed that MVP up-regulation correlates with several hallmarks of malignancy and associates with poor overall survival. CONCLUSIONS: Taken together, through the sequestration of IRF2, MVP promotes an HDM2-dependent loss of p53 that promotes HCC development.


Assuntos
Carcinoma Hepatocelular/etiologia , Fator Regulador 2 de Interferon/fisiologia , Neoplasias Hepáticas/etiologia , Proteína Supressora de Tumor p53/fisiologia , Partículas de Ribonucleoproteínas em Forma de Abóbada/fisiologia , Animais , Humanos , Camundongos
2.
J Immunol ; 203(4): 1001-1011, 2019 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-31235549

RESUMO

Intracellular viral RNAs are recognized by the RIG-I-like receptors (RLRs), which signal through the mitochondrial antiviral signaling protein MAVS. MAVS recruits and activates TBK1 kinase, which further phosphorylates and activates the transcription factor IRF3, leading to the induction of type I IFN and downstream antiviral genes. We identified human nucleus accumbens-associated 1 (NAC1), a member of the BTB/POZ family, as a bridge for MAVS and TBK1 that positively regulates the RLR-mediated induction of type I IFN. Overexpression or knockdown of NAC1 could, respectively, enhance or impair Sendai virus-triggered activation of TBK1 and IRF3, as well as induction of IFN-ß. NAC1 also significantly boosted host antiviral responses against multiple RNA viruses. NAC1 was able to interact with MAVS and TBK1 upon viral infection. The BTB/POZ domain (aa 1-133) of NAC1 interacted with MAVS, and the remainder of NAC1 bound to TBK1. Furthermore, NAC1 could promote the recruitment of TBK1 to MAVS. In contrast, knockdown of NAC1 attenuated the interaction between TBK1 and MAVS. Collectively, our study characterizes NAC1 as an important component of RLR-mediated innate immune responses and uncovers a previously unrecognized function of the BTB/POZ family proteins.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/imunologia , Proteínas de Neoplasias/imunologia , Proteínas Serina-Treonina Quinases/imunologia , Infecções por Vírus de RNA/imunologia , Proteínas Repressoras/imunologia , Transdução de Sinais/imunologia , Linhagem Celular , Humanos , Imunidade Inata/imunologia , Vírus de RNA/imunologia
3.
J Virol ; 93(7)2019 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-30651365

RESUMO

The influenza virus nonstructural protein 1 (NS1) is a nonstructural protein that plays a major role in antagonizing host interferon responses during infection. However, a clear role for the NS1 protein in epigenetic modification has not been established. In this study, NS1 was found to regulate the expression of some key regulators of JAK-STAT signaling by inhibiting the DNA methylation of their promoters. Furthermore, DNA methyltransferase 3B (DNMT3B) is responsible for this process. Upon investigating the mechanisms underlying this event, NS1 was found to interact with DNMT3B but not DNMT3A, leading to the dissociation of DNMT3B from the promoters of the corresponding genes. In addition, the interaction between NS1 and DNMT3B changed the localization of DNMT3B from the nucleus to the cytosol, resulting in K48-linked ubiquitination and degradation of DNMT3B in the cytosol. We conclude that NS1 interacts with DNMT3B and changes its localization to mediate K48-linked polyubiquitination, subsequently contributing to the modulation of the expression of JAK-STAT signaling suppressors.IMPORTANCE The nonstructural protein 1 (NS1) of the influenza A virus (IAV) is a multifunctional protein that counters cellular antiviral activities and is a virulence factor. However, the involvement of NS1 in DNA methylation during IAV infection has not been established. Here, we reveal that the NS1 protein binds the cellular DNMT3B DNA methyltransferase, thereby inhibiting the methylation of the promoters of genes encoding suppressors of JAK-STAT signaling. As a result, these suppressor genes are induced, and JAK-STAT signaling is inhibited. Furthermore, we demonstrate that the NS1 protein transports DNMT3B to the cytoplasm for ubiquitination and degradation. Thus, we identify the NS1 protein as a potential trigger of the epigenetic deregulation of JAK-STAT signaling suppressors and illustrate a novel mechanism underlying the regulation of host immunity during IAV infection.


Assuntos
Citoplasma/virologia , DNA (Citosina-5-)-Metiltransferases/metabolismo , Epigênese Genética/genética , Interações Hospedeiro-Patógeno/genética , Vírus da Influenza A/genética , Ubiquitinação/genética , Proteínas não Estruturais Virais/genética , Células A549 , Animais , Linhagem Celular , Linhagem Celular Tumoral , Citoplasma/metabolismo , Metilação de DNA/genética , Cães , Células HEK293 , Humanos , Influenza Humana/metabolismo , Influenza Humana/virologia , Células Madin Darby de Rim Canino , Camundongos , Infecções por Orthomyxoviridae/metabolismo , Infecções por Orthomyxoviridae/virologia , Regiões Promotoras Genéticas/genética , Células RAW 264.7 , Transdução de Sinais/fisiologia , Proteínas não Estruturais Virais/metabolismo , Replicação Viral/genética , DNA Metiltransferase 3B
4.
J Immunol ; 201(1): 145-156, 2018 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-29777028

RESUMO

Enterovirus 71 (EV71) induces significantly elevated levels of cytokines and chemokines, leading to local or systemic inflammation and severe complications. As shown in our previous study, microRNA (miR) 302c regulates influenza A virus-induced IFN expression by targeting NF-κB-inducing kinase. However, little is known about the role of the miR-302 cluster in EV71-mediated proinflammatory responses. In this study, we found that the miR-302 cluster controls EV71-induced cytokine expression. Further studies demonstrated that karyopherin α2 (KPNA2) is a direct target of the miR-302 cluster. Interestingly, we also found that EV71 infection upregulates KPNA2 expression by downregulating miR-302 cluster expression. Upon investigating the mechanisms behind this event, we found that KPNA2 intracellularly associates with JNK1/JNK2 and p38, leading to translocation of those transcription factors from the cytosol into the nucleus. In EV71-infected patients, miR-302 cluster expression was downregulated and KPNA2 expression was upregulated compared with controls, and their expression levels were closely correlated. Taken together, our work establishes a link between the miR-302/ KPNA2 axis and EV71-induced cytokine expression and represents a promising target for future antiviral therapy.


Assuntos
Citocinas/metabolismo , Enterovirus Humano A/imunologia , Imunidade Inata/imunologia , MicroRNAs/metabolismo , alfa Carioferinas/metabolismo , Transporte Ativo do Núcleo Celular/fisiologia , Linhagem Celular Tumoral , Células HEK293 , Humanos , MicroRNAs/biossíntese , MicroRNAs/genética , Proteína Quinase 8 Ativada por Mitógeno/metabolismo , Proteína Quinase 9 Ativada por Mitógeno/metabolismo , Células THP-1 , Fator de Transcrição RelA/metabolismo , alfa Carioferinas/biossíntese , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
5.
J Virol ; 92(9)2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29467312

RESUMO

Fibronectin (FN) is a high-molecular-weight extracellular matrix protein that contains the RGDS motif, which is required to bind to integrins. Synthetic RGDS peptides have been reported to compete with FN to bind to the cell surface and inhibit the function of FN. Here, we identified that synthetic RGDS peptides significantly inhibit human enterovirus 71 (EV71) infection in cell cultures. In addition, mice treated with RGDS peptides and infected with EV71 had a significantly higher survival rate and a lower viral load than the control group. Because RGDS peptides affect the function of FN, we questioned whether FN may play a role in virus infection. Our study indicates that overexpression of FN enhanced EV71 infection. In contrast, knockout of FN significantly reduced viral yield and decreased the viral binding to host cells. Furthermore, EV71 entry, rather than intracellular viral replication, was blocked by FN inhibitor pretreatment. Next, we found that FN could interact with the EV71 capsid protein VP1, and further truncated-mutation assays indicated that the D2 domain of FN could interact with the N-terminal fragment of VP1. Taken together, our results demonstrate that the host factor FN binds to EV71 particles and facilitates EV71 entry, providing a potential therapy target for EV71 infection.IMPORTANCE Hand, foot, and mouth disease outbreaks have occurred frequently in recent years, sometimes causing severe neurological complications and even death in infants and young children worldwide. Unfortunately, no effective antiviral drugs are available for human enterovirus 71 (EV71), one of the viruses that cause hand, foot, and mouth disease. The infection process and the host factors involved remain unknown, although several receptors have been identified. In this study, we found that the host factor fibronectin (FN) facilitated EV71 replication by interacting with EV71 particles and further mediated their entry. The RGDS peptide, an FN inhibitor, significantly inhibited EV71 replication in both RD cells and mice. In conclusion, our research identified a new host factor involved in EV71 infection, providing a new potential antiviral target for EV71 treatment.


Assuntos
Enterovirus Humano A/metabolismo , Infecções por Enterovirus/patologia , Fibronectinas/metabolismo , Internalização do Vírus , Animais , Sistemas CRISPR-Cas , Linhagem Celular , Enterovirus Humano A/genética , Infecções por Enterovirus/virologia , Fibronectinas/genética , Técnicas de Inativação de Genes , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Replicação Viral/fisiologia
6.
J Immunol ; 198(9): 3690-3704, 2017 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-28356387

RESUMO

The innate immune response is critical for host defense and must be tightly controlled, but the molecular mechanisms responsible for its negative regulation are not yet completely understood. In this study, we report that transporter 1, ATP-binding cassette, subfamily B (TAP1), a virus-inducible endoplasmic reticulum-associated protein, negatively regulated the virus-triggered immune response. In this study, we observed upregulated expression of TAP1 following virus infection in human lung epithelial cells (A549), THP-1 monocytes, HeLa cells, and Vero cells. The overexpression of TAP1 enhanced virus replication by inhibiting the virus-triggered activation of NF-κB signaling and the production of IFNs, IFN-stimulated genes, and proinflammatory cytokines. TAP1 depletion had the opposite effect. In response to virus infection, TAP1 interacted with the TGF-ß-activated kinase (TAK)1 complex and impaired the phosphorylation of TAK1, subsequently suppressing the phosphorylation of the IκB kinase complex and NF-κB inhibitor α (IκBα) as well as NF-κB nuclear translocation. Our findings collectively suggest that TAP1 plays a novel role in the negative regulation of virus-triggered NF-κB signaling and the innate immune response by targeting the TAK1 complex.


Assuntos
Membro 2 da Subfamília B de Transportadores de Cassetes de Ligação de ATP/metabolismo , Vírus da Influenza A/fisiologia , Influenza Humana/imunologia , MAP Quinase Quinase Quinases/metabolismo , Mucosa Respiratória/fisiologia , Membro 2 da Subfamília B de Transportadores de Cassetes de Ligação de ATP/genética , Células HEK293 , Células HeLa , Homeostase , Humanos , Imunidade Inata , Interferons/genética , Interferons/metabolismo , NF-kappa B/metabolismo , RNA Interferente Pequeno/genética , Mucosa Respiratória/virologia , Transdução de Sinais , Replicação Viral
7.
J Biol Chem ; 292(52): 21291-21303, 2017 12 29.
Artigo em Inglês | MEDLINE | ID: mdl-29046356

RESUMO

During influenza A virus (IAV) infection, cytokine storms play a vital and critical role in clinical outcomes. We have previously reported that microRNA (miR)-302c regulates IAV-induced IFN expression by targeting the 3'-UTR of nuclear factor κB (NF-κB)-inducing kinase. In the current study, we found that miR-302a, another member of the miR-302 cluster, controls the IAV-induced cytokine storm. According to results from cell-based and knockout mouse models, IAV induces a cytokine storm via interferon regulatory factor-5 (IRF-5). We also found that IAV infection up-regulates IRF-5 expression and that IRF-5 in turn promotes IAV replication. Furthermore, we observed that IRF-5 is a direct target of miR-302a, which down-regulated IRF-5 expression by binding its 3'-UTR. Moreover, IAV increased IRF-5 expression by down-regulating miR-302a expression. Interestingly, miR-302a inhibited IAV replication. In IAV-infected patients, miR-302a expression was down-regulated, whereas IRF-5 expression was up-regulated. Taken together, our work uncovers and defines a signaling pathway implicated in an IAV-induced cytokine storm.


Assuntos
Vírus da Influenza A Subtipo H1N1/fisiologia , Fatores Reguladores de Interferon/biossíntese , MicroRNAs/imunologia , Células A549 , Animais , Modelos Animais de Doenças , Cães , Regulação para Baixo , Células HEK293 , Interações Hospedeiro-Patógeno , Humanos , Evasão da Resposta Imune , Vírus da Influenza A Subtipo H1N1/genética , Vírus da Influenza A Subtipo H1N1/imunologia , Vírus da Influenza A Subtipo H1N1/metabolismo , Influenza Humana/genética , Influenza Humana/imunologia , Influenza Humana/metabolismo , Influenza Humana/virologia , Fatores Reguladores de Interferon/genética , Fatores Reguladores de Interferon/imunologia , Fatores Reguladores de Interferon/metabolismo , Células Madin Darby de Rim Canino , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/genética , MicroRNAs/metabolismo , NF-kappa B/imunologia , NF-kappa B/metabolismo , Infecções por Orthomyxoviridae/genética , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/metabolismo , Infecções por Orthomyxoviridae/virologia , Transdução de Sinais , Replicação Viral
8.
J Immunol ; 196(6): 2753-66, 2016 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-26843330

RESUMO

Pathogen invasion triggers robust antiviral cytokine production via different transcription factor signaling pathways. We have previously demonstrated that major vault protein (MVP) induces type I IFN production during viral infection; however, little is known about the role of MVP in proinflammatory responses. In this study, we found in vitro that expression of MVP, IL-6, and IL-8 was inducible upon dsRNA stimulation or viral infection. Moreover, MVP was essential for the induction of IL-6 and IL-8, as impaired expression of IL-6 and IL-8 in MVP-deficient human PBMCs, human lung epithelial cells (A549), and THP-1 monocytes, as well as in murine splenocytes, peritoneal macrophages, and PBMCs from MVP-knockout (MVP(-/-)) mice, was observed. Upon investigation of the underlying mechanisms, we demonstrated that MVP acted in synergy with AP-1 (c-Fos) and CCAAT/enhancer binding protein (C/EBP)ß-liver-enriched transcriptional activating protein to activate the IL6 and IL8 promoters. Introduction of mutations into the AP-1 and C/EBPß binding sites on the IL6 and IL8 promoters resulted in the loss of synergistic activation with MVP. Furthermore, we found that MVP interacted with both c-Fos and C/EBPß. The interactions promoted nuclear translocation and recruitment of these transcription factors to IL6 and IL8 promoter regions. In the MVP(-/-) mouse model, significantly decreased expression of early antiviral cytokines resulted in higher viral titer in the lung, higher mortality, and heavier lung damage after infection with lethal influenza A virus. Taken together, our findings help to delineate a novel role of MVP in host proinflammatory response.


Assuntos
Células Epiteliais/imunologia , Inflamação/imunologia , Vírus da Influenza A/imunologia , Leucócitos Mononucleares/imunologia , Infecções por Orthomyxoviridae/imunologia , Partículas de Ribonucleoproteínas em Forma de Abóbada/metabolismo , Animais , Proteína beta Intensificadora de Ligação a CCAAT/genética , Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Citocinas/genética , Citocinas/metabolismo , Feminino , Células HEK293 , Humanos , Imunidade/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação/genética , RNA de Cadeia Dupla/imunologia , RNA Interferente Pequeno/genética , Fator de Transcrição AP-1/genética , Fator de Transcrição AP-1/metabolismo , Partículas de Ribonucleoproteínas em Forma de Abóbada/genética
9.
J Hepatol ; 62(5): 1015-23, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25481566

RESUMO

BACKGROUND & AIMS: We previously demonstrated that major vault protein (MVP) is a novel virus-induced host factor and its expression upregulates type-I interferon production, leading to cellular antiviral response. However, it remains unclear whether the antiviral function of MVP is impaired during hepatitis B virus (HBV) infection and what mechanisms are involved. Therefore, the aim of this study was to assess whether HBV can alter MVP expression despite the lack of type-I IFN induction and shed light on the underlying mechanisms HBV utilizes to evade host innate immune response. METHODS: The ability of HBV surface and e antigens to inhibit MVP signaling in interferon induction pathways was evaluated by co-immunoprecipitation, immunofluorescence, quantitative RT-PCR, Western blot and reporter assays. RESULTS: In our current study, we found high levels of MVP in peripheral blood mononuclear cells, sera, and liver tissue from HBV-infected patients relative to healthy individuals. We determined that MVP intracellularly associates with MyD88, an adapter protein involved in virus-triggered induction of type-I IFN. Protein truncation analysis revealed that the middle domain of MVP (amino acid residues 310-620) was essential for MyD88 binding. Conversely, HBV inhibited MVP-induced type-I IFN production by suppressing MVP/MyD88 interaction. HBV antigens, both HBsAg and HBeAg, suppressed this interaction by competitively binding to the essential MyD88 binding region of MVP and limiting downstream IFN signaling. CONCLUSIONS: MVP is a virus-induced protein capable of binding with MyD88 leading to type-I IFN production. HBV may evade an immune response by disrupting this interaction and limiting type-I IFN antiviral activity.


Assuntos
Antígenos de Superfície da Hepatite B/metabolismo , Antígenos E da Hepatite B/metabolismo , Vírus da Hepatite B/fisiologia , Hepatite B , Interferon Tipo I/biossíntese , Fator 88 de Diferenciação Mieloide/metabolismo , Partículas de Ribonucleoproteínas em Forma de Abóbada/metabolismo , Hepatite B/imunologia , Hepatite B/virologia , Interações Hospedeiro-Patógeno/imunologia , Humanos , Imunidade Inata/imunologia , Fígado/imunologia , Transdução de Sinais/imunologia
10.
Hepatology ; 56(1): 57-66, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22318991

RESUMO

UNLABELLED: Major vault protein (MVP) is the major constituent of vaults and is involved in multidrug resistance, nucleocytoplasmic transport, and cell signaling. However, little is known about the role of MVP during viral infections. In this study, high levels of MVP were found in peripheral blood mononuclear cells, sera, and liver tissue from patients infected with hepatitis C virus (HCV) relative to healthy individuals. HCV infections resulted in elevated levels of MVP messenger RNA (mRNA) and protein expression in the hepatocyte cell lines Huh7.5.1 and Huh7. Further studies demonstrated that the nuclear factor kappa B (NF-κB) and Sp1 pathways are involved in the induction of MVP expression by HCV. Interestingly, MVP expression suppressed HCV replication and protein synthesis by way of induction of type-I interferon mRNA expression and protein secretion. Upon investigating the mechanisms behind this event, we found that MVP enhanced the expression of interferon regulatory factor 7 (IRF7), but not IRF3. Translocation of activated IRF7 and NF-κB from the cytosol to the nucleus was involved in this process. Furthermore, vesicular stomatitis virus, influenza A virus, and enterovirus 71 also induced MVP production, and MVP in turn hampered viral replication and production. CONCLUSION: MVP is a novel virus-induced host factor and its expression up-regulates type-I interferon production, leading to cellular antiviral responses.


Assuntos
Hepacivirus/genética , Hepatite C Crônica/genética , Interferon Tipo I/genética , Partículas de Ribonucleoproteínas em Forma de Abóbada/genética , Replicação Viral/genética , Estudos de Casos e Controles , Células Cultivadas , Feminino , Regulação Viral da Expressão Gênica , Marcadores Genéticos/genética , Hepacivirus/metabolismo , Hepatite C Crônica/sangue , Hepatite C Crônica/fisiopatologia , Hepatócitos/metabolismo , Hepatócitos/virologia , Fator Proteico 1 do Hospedeiro/genética , Fator Proteico 1 do Hospedeiro/metabolismo , Humanos , Interferon Tipo I/metabolismo , Leucócitos Mononucleares/fisiologia , Masculino , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase em Tempo Real/métodos , Valores de Referência , Sensibilidade e Especificidade , Transdução de Sinais/genética , Regulação para Cima , Partículas de Ribonucleoproteínas em Forma de Abóbada/metabolismo
11.
Autophagy ; 19(6): 1764-1780, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36472478

RESUMO

Macroautophagy/autophagy, a stress-responsive cellular survival mechanism, plays important and context-dependent roles in cancer, and its inhibition has been implicated as a promising cancer therapeutic approach. The detailed mechanisms underlying the function of autophagy in cancer have not been fully understood. In this study, we show that autophagy inhibition promotes both the efficacy of chemotherapy for the treatment of glioblastoma (GBM) and therapy-induced senescence of GBM cells. As a specific cell fate characterized by permanent cell cycle arrest, senescence is also associated with the expression of a panel of specific secreted protein factors known as senescence-associated secretory phenotype (SASP). Intriguingly, we found that autophagy inhibition not only quantitatively enhanced GBM cell senescence but also qualitatively altered the spectrum of SASP. The altered SASP had increased potent activity to induce paracrine senescence of neighboring GBM cells, to skew macrophage polarization toward the anti-tumor M1 state, and to block the recruitment of pro-tumor neutrophils to GBM tumor tissues. Taken together, this study reveals novel functional communication between autophagy and senescence and suggests cancer therapeutic approaches harnessing autophagy blockage in inducing senescence-mediated antitumor immunity.


Assuntos
Autofagia , Glioblastoma , Humanos , Glioblastoma/tratamento farmacológico , Glioblastoma/patologia , Senescência Celular/fisiologia
12.
Nat Commun ; 12(1): 98, 2021 01 04.
Artigo em Inglês | MEDLINE | ID: mdl-33397935

RESUMO

Glucose metabolism and innate immunity evolved side-by-side. It is unclear if and how the two systems interact with each other during hepatitis B virus (HBV) infections and, if so, which mechanisms are involved. Here, we report that HBV activates glycolysis to impede retinoic acid-inducible gene I (RIG-I)-induced interferon production. We demonstrate that HBV sequesters MAVS from RIG-I by forming a ternary complex including hexokinase (HK). Using a series of pharmacological and genetic approaches, we provide in vitro and in vivo evidence indicating that HBV suppresses RLR signaling via lactate dehydrogenase-A-dependent lactate production. Lactate directly binds MAVS preventing its aggregation and mitochondrial localization during HBV infection. Therefore, we show that HK2 and glycolysis-derived lactate have important functions in the immune escape of HBV and that energy metabolism regulates innate immunity during HBV infection.


Assuntos
Vírus da Hepatite B/fisiologia , Imunidade Inata , Metaboloma , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Anaerobiose , Animais , Células Cultivadas , Proteína DEAD-box 58/metabolismo , Glucose/metabolismo , Glicólise , Células Hep G2 , Hepatócitos/metabolismo , Hepatócitos/patologia , Hepatócitos/virologia , Humanos , Evasão da Resposta Imune , Interferons/metabolismo , Ácido Láctico/metabolismo , Camundongos Endogâmicos C57BL , Modelos Biológicos , Transdução de Sinais , Vírion/metabolismo
13.
Nanotechnology ; 21(10): 105101, 2010 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-20154371

RESUMO

Recently there has been a rapid expansion of the development of bioinorganic hybrid systems for safe drug delivery. Layered double hydroxides (LDH), a variety of available inorganic matrix, possess great promise for this purpose. In this study, an oxidative stress biomarker system, including measurement of reactive oxygen species, glutathione content, endogenous nitric oxide, carbonyl content in proteins, DNA strand breaks and DNA-protein crosslinks, was designed to evaluate the biocompatibility of different concentrations of nano-Zn/Al-LDH with a Hela cell line. The drug delivery activity of the LDH-folic-acid complex was also assessed. The resulting data clearly demonstrated that nano-LDH could be applied as a relatively safe drug vehicle with good delivery activity, but with the caveat that the effects of high dosages observed here should not be ignored when attempting to maximize therapeutic activity by increasing LDH concentration.


Assuntos
Portadores de Fármacos/toxicidade , Hidróxidos/toxicidade , Apoptose/efeitos dos fármacos , Ensaio Cometa , Células HeLa , Humanos , Microscopia Eletrônica de Varredura , Nanotecnologia , Espécies Reativas de Oxigênio/metabolismo , Espectroscopia de Infravermelho com Transformada de Fourier
14.
Sci Adv ; 6(16): eaaz7086, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32494619

RESUMO

In this study, we demonstrated an essential function of the hexosamine biosynthesis pathway (HBP)-associated O-linked ß-N-acetylglucosamine (O-GlcNAc) signaling in influenza A virus (IAV)-induced cytokine storm. O-GlcNAc transferase (OGT), a key enzyme for protein O-GlcNAcylation, mediated IAV-induced cytokine production. Upon investigating the mechanisms driving this event, we determined that IAV induced OGT to bind to interferon regulatory factor-5 (IRF5), leading to O-GlcNAcylation of IRF5 on serine-430. O-GlcNAcylation of IRF5 is required for K63-linked ubiquitination of IRF5 and subsequent cytokine production. Analysis of clinical samples revealed that IRF5 is O-GlcNAcylated, and higher levels of proinflammatory cytokines correlated with higher levels of blood glucose in IAV-infected patients. We identified a molecular mechanism by which HBP-mediated O-GlcNAcylation regulates IRF5 function during IAV infection, highlighting the importance of glucose metabolism in IAV-induced cytokine storm.


Assuntos
Vírus da Influenza A , Síndrome da Liberação de Citocina , Citocinas , Hexosaminas , Humanos , Fatores Reguladores de Interferon , N-Acetilglucosaminiltransferases
15.
J Innate Immun ; 9(4): 419-435, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28376501

RESUMO

Guanylate binding protein (GBP) 5 belongs to the GBP family, which is involved in important cellular processes, including signal transduction, translation, vesicle trafficking, and exocytosis. Structurally, GBPs display a high degree of homology and share highly conserved GTP-binding or hydrolysis domains. GBP5 was reported to be a critical cellular factor in inflammasome assembly. However, little is known about its role in the host antiviral innate immune response. In this study, we found that GBP5 expression was significantly elevated in influenza patients and influenza A virus-infected A549 human lung epithelial cells. The overexpression of GBP5 inhibited virus replication by enhancing the expression of virus-induced interferon (IFN) and IFN-related effectors. Knockdown of GBP5 had the opposite effect. Moreover, GBP5 enhanced endogenous IFN expression by interacting with the NF-κB-essential modulator complex and stimulating NF-κB signaling. Additionally, the expression of proinflammatory factors, such as IL-6, IL-8, tumor necrosis factor-α, cyclooxygenase-2, and inducible nitric oxide synthase, was also activated by GBP5. Taken together, our results reveal that GBP5 inhibited virus replication through the activation of IFN signaling and proinflammatory factors.


Assuntos
Proteínas de Ligação ao GTP/genética , Vírus da Influenza A/fisiologia , Influenza Humana/imunologia , Interferons/metabolismo , Mucosa Respiratória/imunologia , Células A549 , Ciclo-Oxigenase 2/metabolismo , Citocinas/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Células HEK293 , Humanos , Imunidade Inata , Inflamassomos/metabolismo , Mediadores da Inflamação/metabolismo , NF-kappa B/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , RNA Interferente Pequeno/genética , Mucosa Respiratória/virologia , Transdução de Sinais , Replicação Viral
16.
Sci Rep ; 6: 33557, 2016 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-27629939

RESUMO

RNA virus infections are detected by the RIG-I family of receptors, which signal through the adaptor molecule mitochondrial antiviral signaling (MAVS). MAVS then recruits the adaptor's tumor necrosis factor receptor-associated factor (TRAF) 3 and TRAF6, which in turn activate IRF3 and NF-κB, respectively, to induce interferons (IFNs) and inflammatory responses. Here we show that the biotin-containing enzyme methylcrotonoyl-CoA carboxylase 1 (MCCC1) enhances virus-induced, MAVS-mediated IFN and inflammatory cytokine expression through the NF-κB signaling pathway. MCCC1 knockdown strongly inhibits induction of IFNs and inflammatory cytokines. Furthermore, MCCC1 shows extensive antiviral activity toward RNA viruses, including influenza A virus, human enterovirus 71, and vesicular stomatitis virus. Here, we have elucidated the mechanism underlying MCCC1-mediated inhibition of viral replication. MCCC1 interacts with MAVS and components of the MAVS signalosome and contributes to enhanced production of type I IFNs and pro-inflammatory cytokines by promoting phosphorylation of the IκB kinase (IKK) complex and NF-κB inhibitor-α (IκBα), as well as NF-κB nuclear translocation. This process leads to activation of IFNs and cytokine expression and subsequent activation of IFN-stimulated genes, including double-stranded RNA-dependent protein kinase PKR and myxovirus resistance protein 1. These findings demonstrate that MCCC1 plays an essential role in virus-triggered, MAVS-mediated activation of NF-κB signaling.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Carbono-Carbono Ligases/metabolismo , Proteína DEAD-box 58/metabolismo , NF-kappa B/metabolismo , Transdução de Sinais , Células A549 , Antivirais/metabolismo , Núcleo Celular/metabolismo , Citocinas/metabolismo , Técnicas de Silenciamento de Genes , Humanos , Mediadores da Inflamação/metabolismo , Interferon Tipo I/metabolismo , Inibidor de NF-kappaB alfa , Fosforilação , Transporte Proteico , RNA Interferente Pequeno/metabolismo , Fator 6 Associado a Receptor de TNF/metabolismo
17.
PLoS One ; 11(3): e0152721, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27023403

RESUMO

Fibronectin (FN) is a high molecular weight extracellular matrix protein that functions in cell adhesion, growth, migration, and embryonic development. However, little is known about the role of FN during viral infection. In the present study, we found significantly higher levels of FN in sera, and liver tissues from hepatitis B virus (HBV) patients relative to healthy individuals. HBV expression enhanced FN mRNA and protein levels in the hepatic cell lines Huh7 and HepG2. HBV infection of susceptible HepG2-sodium taurocholate co-transporting polypeptide cells also increased FN expression. We also found that transcriptional factor specificity protein 1 was involved in the induction of FN by HBV. Knockdown of FN expression significantly inhibited HBV DNA replication and protein synthesis through activating endogenous IFN-α production. In addition, FN interacted with the transforming growth factor ß-activated protein kinase 1 (TAK1) and TAK1-binding protein complex and attenuated interferon signaling by inhibiting TAK1 phosphorylation. Furthermore, the nuclear translocation of NF-κB/p65 was found to be inhibited by FN. We also observed that FN promoted HBV enhancers to support HBV expression. These results suggest novel functions of endogenous FN involved in immune evasion and maintenance of HBV replication.


Assuntos
Fibronectinas/metabolismo , Vírus da Hepatite B/metabolismo , Replicação Viral , Adulto , Elementos Facilitadores Genéticos/genética , Feminino , Fibronectinas/genética , Células HEK293 , Células Hep G2 , Vírus da Hepatite B/efeitos dos fármacos , Hepatite B Crônica/virologia , Humanos , Interferon-alfa/farmacologia , MAP Quinase Quinase Quinases/metabolismo , Masculino , Pessoa de Meia-Idade , NF-kappa B/metabolismo , Fosforilação/efeitos dos fármacos , Ligação Proteica/efeitos dos fármacos , Fator de Transcrição Sp1/metabolismo , Transcrição Gênica/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos
18.
Sci Rep ; 6: 22002, 2016 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-26906558

RESUMO

When retinoic acid-inducible gene 1 protein (RIG-I)-like receptors sense viral dsRNA in the cytosol, RIG-I and melanoma differentiation-associated gene 5 (MDA5) are recruited to the mitochondria to interact with mitochondrial antiviral signaling protein (MAVS) and initiate antiviral immune responses. In this study, we demonstrate that the biotin-containing enzyme pyruvate carboxylase (PC) plays an essential role in the virus-triggered activation of nuclear factor kappa B (NF-κB) signaling mediated by MAVS. PC contributes to the enhanced production of type I interferons (IFNs) and pro-inflammatory cytokines, and PC knockdown inhibits the virus-triggered innate immune response. In addition, PC shows extensive antiviral activity against RNA viruses, including influenza A virus (IAV), human enterovirus 71 (EV71), and vesicular stomatitis virus (VSV). Furthermore, PC mediates antiviral action by targeting the MAVS signalosome and induces IFNs and pro-inflammatory cytokines by promoting phosphorylation of NF-κB inhibitor-α (IκBα) and the IκB kinase (IKK) complex, as well as NF-κB nuclear translocation, which leads to activation of interferon-stimulated genes (ISGs), including double-stranded RNA-dependent protein kinase (PKR) and myxovirus resistance protein 1 (Mx1). Our findings suggest that PC is an important player in host antiviral signaling.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/imunologia , Proteína DEAD-box 58/imunologia , Enterovirus Humano A/imunologia , Hepatócitos/imunologia , Vírus da Influenza A Subtipo H3N2/imunologia , Piruvato Carboxilase/imunologia , Vesiculovirus/imunologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Linhagem Celular Tumoral , Citocinas/genética , Citocinas/imunologia , Proteína DEAD-box 58/genética , Enterovirus Humano A/genética , Regulação da Expressão Gênica , Genes Reporter , Células HEK293 , Hepatócitos/virologia , Humanos , Imunidade Inata , Vírus da Influenza A Subtipo H3N2/genética , Interferon Tipo I/genética , Interferon Tipo I/imunologia , Helicase IFIH1 Induzida por Interferon/genética , Helicase IFIH1 Induzida por Interferon/imunologia , Luciferases/genética , Luciferases/imunologia , Inibidor de NF-kappaB alfa/genética , Inibidor de NF-kappaB alfa/imunologia , NF-kappa B/genética , NF-kappa B/imunologia , Piruvato Carboxilase/antagonistas & inibidores , Piruvato Carboxilase/genética , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/imunologia , RNA Viral/genética , RNA Viral/imunologia , Receptores Imunológicos , Transdução de Sinais , Vesiculovirus/genética , eIF-2 Quinase/genética , eIF-2 Quinase/imunologia
19.
Sci Adv ; 2(4): e1501535, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27051880

RESUMO

G-quadruplex (G4) is one of the most important secondary structures in nucleic acids. Until recently, G4 RNAs have not been reported in any ribovirus, such as the hepatitis C virus. Our bioinformatics analysis reveals highly conserved guanine-rich consensus sequences within the core gene of hepatitis C despite the high genetic variability of this ribovirus; we further show using various methods that such consensus sequences can fold into unimolecular G4 RNA structures, both in vitro and under physiological conditions. Furthermore, we provide direct evidences that small molecules specifically targeting G4 can stabilize this structure to reduce RNA replication and inhibit protein translation of intracellular hepatitis C. Ultimately, the stabilization of G4 RNA in the genome of hepatitis C represents a promising new strategy for anti-hepatitis C drug development.


Assuntos
Quadruplex G , Hepacivirus/efeitos dos fármacos , Hepatite C/virologia , Proteínas do Core Viral/química , Sequência Conservada , Terapia Genética , Genoma Viral , Hepatite C/tratamento farmacológico , Humanos , Conformação de Ácido Nucleico , RNA Viral/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas/farmacologia , Proteínas do Core Viral/antagonistas & inibidores , Proteínas do Core Viral/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA